51
|
Blanco-Ania D, Heus R, Rutjes FPJT. Privileged heterocycles: bioactivity and synthesis of 1,9-diazaspiro[5.5]undecane-containing compounds. Chem Heterocycl Compd (N Y) 2017; 53:827-845. [PMID: 32214420 PMCID: PMC7088121 DOI: 10.1007/s10593-017-2133-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 08/24/2017] [Indexed: 12/01/2022]
Abstract
This review discusses the biological activity and synthesis of 1,9-diazaspiro[5.5]undecanes, including those ring-fused with arenes and heteroarenes and/or containing a carbonyl group at position 2. These compounds could be used for the treatment of obesity, pain, as well as various immune system, cell signaling, cardiovascular, and psychotic disorders.
Collapse
Affiliation(s)
- Daniel Blanco-Ania
- Radboud University, Institute for Molecules and Materials, Heyendaalseweg 135, 6526 AJ Nijmegen, the Netherlands
| | - Rik Heus
- Radboud University, Institute for Molecules and Materials, Heyendaalseweg 135, 6526 AJ Nijmegen, the Netherlands
| | - Floris P J T Rutjes
- Radboud University, Institute for Molecules and Materials, Heyendaalseweg 135, 6526 AJ Nijmegen, the Netherlands
| |
Collapse
|
52
|
Albasanz JL, Santana S, Guzman-Sanchez F, León D, Burgos JS, Martín M. 2-Methyl-6-(phenylethynyl)pyridine Hydrochloride Modulates Metabotropic Glutamate 5 Receptors Endogenously Expressed in Zebrafish Brain. ACS Chem Neurosci 2016; 7:1690-1697. [PMID: 27635438 DOI: 10.1021/acschemneuro.6b00213] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Due to phylogenetic proximity to the human, zebrafish has been recognized as a reliable model to study Alzheimer's disease (AD) and other central nervous system disorders. Furthermore, metabotropic glutamate receptors have been previously reported to be impaired in brain from AD patients. Metabotropic glutamate 5 (mGlu5) receptors are G-protein coupled receptors proposed as potential targets for therapy of different neurodegenerative disorders. Thus, MPEP (2-methyl-6-(phenylethynyl)pyridine hydrochloride), a selective noncompetitive mGlu5 receptor antagonist, has been suggested for pharmacological treatment of AD. The aim of the present work was to quantify mGlu5 receptors in brain from zebrafish and to study the possible modulation of these receptors by MPEP treatment. To this end, radioligand binding assay and open field test were used. Results showed a slightly higher presence of mGlu5 receptors in brain from male than in that from female zebrafish. However, a significant increase of mGlu5 receptor in male without variation in female was observed after MPEP treatment. This gender specific response was also observed in locomotor behavior, being significantly decreased only in male zebrafish. These results confirm the presence of mGlu5 receptors in brain from zebrafish and their gender specific modulation by selective antagonist treatment and suggest a role of these receptors on locomotor activity, which is affected in many disorders. In addition, our data point to zebrafish as a useful model to study mGlu receptor function in both healthy and pathological conditions.
Collapse
Affiliation(s)
- José Luis Albasanz
- Departamento de Química Inorgánica,
Orgánica y Bioquímica, Facultad de Medicina de Ciudad
Real/Facultad de Ciencias y Tecnologías Químicas, Universidad de Castilla-La Mancha, Centro Regional de Investigaciones Biomédicas (CRIB), Avenida Camilo José Cela 10, 13071 Ciudad Real, Spain
| | | | | | - David León
- Departamento de Química Inorgánica,
Orgánica y Bioquímica, Facultad de Medicina de Ciudad
Real/Facultad de Ciencias y Tecnologías Químicas, Universidad de Castilla-La Mancha, Centro Regional de Investigaciones Biomédicas (CRIB), Avenida Camilo José Cela 10, 13071 Ciudad Real, Spain
| | | | - Mairena Martín
- Departamento de Química Inorgánica,
Orgánica y Bioquímica, Facultad de Medicina de Ciudad
Real/Facultad de Ciencias y Tecnologías Químicas, Universidad de Castilla-La Mancha, Centro Regional de Investigaciones Biomédicas (CRIB), Avenida Camilo José Cela 10, 13071 Ciudad Real, Spain
| |
Collapse
|
53
|
Abstract
INTRODUCTION Treatment of patients suffering from major depression could be highly challenging for psychiatrists. Intractability as well as relapse is commonly seen among these patients, leading to functional impairment and poor quality of life. The present review discusses some of the novel investigational drugs that are under pre-clinical or clinical phases in the treatment of major depression. Areas covered: Molecules belonging to different classes such as triple reuptake inhibitors, opioid receptors, ionotropic and metabotropic glutamate receptors, and neurotrophin in the treatment of major depression are covered in this article. Expert opinion: Although the historical discovery of earlier antidepressant molecules (iproniazid and imipramine) is through serendipitous discovery, the present research focuses on discovering novel molecules based on our current pathophysiological knowledge of the disease condition. The fast-acting antidepressant property of N-methyl-d-aspartate (NMDA) receptor molecules, including ketamine is an exciting area of research. Other drug molecules such as amitifadine (triple reuptake inhibitor), ALKS-5461 (kappa receptor antagonist and mu opioidergic receptor agonist), rapastinel (NMDA glutamatergic receptor modulator) are under Phase-III clinical trials and could be approved in the near future for the treatment of major depression.
Collapse
Affiliation(s)
- Ashish Dhir
- a Department of Neurology, School of Medicine , University of California Davis , Sacramento , CA , USA
| |
Collapse
|
54
|
Gumru S, Aricioglu F. Antipsychotics: Neurobiological Bases for a Therapeutic Approach. ACTA ACUST UNITED AC 2016. [DOI: 10.5455/bcp.20130320010604] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
- Salih Gumru
- Marmara University, School of Pharmacy Department of Pharmacology and Psychopharmacology Research Unit, Istanbul-Turkey
| | - Feyza Aricioglu
- Marmara University, School of Pharmacy Department of Pharmacology and Psychopharmacology Research Unit, Istanbul-Turkey
| |
Collapse
|
55
|
Zhang Z, Bassam B, Thomas AG, Williams M, Liu J, Nance E, Rojas C, Slusher BS, Kannan S. Maternal inflammation leads to impaired glutamate homeostasis and up-regulation of glutamate carboxypeptidase II in activated microglia in the fetal/newborn rabbit brain. Neurobiol Dis 2016; 94:116-28. [PMID: 27326668 PMCID: PMC5394739 DOI: 10.1016/j.nbd.2016.06.010] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 06/05/2016] [Accepted: 06/16/2016] [Indexed: 12/12/2022] Open
Abstract
Astrocyte dysfunction and excessive activation of glutamatergic systems have been implicated in a number of neurologic disorders, including periventricular leukomalacia (PVL) and cerebral palsy (CP). However, the role of chorioamnionitis on glutamate homeostasis in the fetal and neonatal brains is not clearly understood. We have previously shown that intrauterine endotoxin administration results in intense microglial 'activation' and increased pro-inflammatory cytokines in the periventricular region (PVR) of the neonatal rabbit brain. In this study, we assessed the effect of maternal inflammation on key components of the glutamate pathway and its relationship to astrocyte and microglial activation in the fetal and neonatal New Zealand white rabbit brain. We found that intrauterine endotoxin exposure at gestational day 28 (G28) induced acute and prolonged glutamate elevation in the PVR of fetal (G29, 1day post-injury) and postnatal day 1 (PND1, 3days post-injury) brains along with prominent morphological changes in the astrocytes (soma hypertrophy and retracted processes) in the white matter tracts. There was a significant increase in glutaminase and N-Methyl-d-Aspartate receptor (NMDAR) NR2 subunit expression along with decreased glial L-glutamate transporter 1 (GLT-1) in the PVR at G29, that would promote acute dysregulation of glutamate homeostasis. This was accompanied with significantly decreased TGF-β1 at PND1 in CP kits indicating ongoing neuroinflammation. We also show for the first time that glutamate carboxypeptidase II (GCPII) was significantly increased in the activated microglia at the periventricular white matter area in both G29 and PND1 CP kits. This was confirmed by in vitro studies demonstrating that LPS activated primary microglia markedly upregulate GCPII enzymatic activity. These results suggest that maternal intrauterine endotoxin exposure results in early onset and long-lasting dysregulation of glutamate homeostasis, which may be mediated by impaired astrocyte function and GCPII upregulation in activated microglia.
Collapse
Affiliation(s)
- Zhi Zhang
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins School of Medicine, 1800 Orleans St, Baltimore, MD 21287, USA
| | - Bassam Bassam
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins School of Medicine, 1800 Orleans St, Baltimore, MD 21287, USA
| | - Ajit G Thomas
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, 1800 Orleans St, Baltimore, MD 21287, USA
| | - Monica Williams
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins School of Medicine, 1800 Orleans St, Baltimore, MD 21287, USA
| | - Jinhuan Liu
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins School of Medicine, 1800 Orleans St, Baltimore, MD 21287, USA
| | - Elizabeth Nance
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins School of Medicine, 1800 Orleans St, Baltimore, MD 21287, USA
| | - Camilo Rojas
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, 1800 Orleans St, Baltimore, MD 21287, USA
| | - Barbara S Slusher
- Neurology, Johns Hopkins School of Medicine, 1800 Orleans St, Baltimore, MD 21287, USA; Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, 1800 Orleans St, Baltimore, MD 21287, USA
| | - Sujatha Kannan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins School of Medicine, 1800 Orleans St, Baltimore, MD 21287, USA.
| |
Collapse
|
56
|
Valentinova K, Mameli M. mGluR-LTD at Excitatory and Inhibitory Synapses in the Lateral Habenula Tunes Neuronal Output. Cell Rep 2016; 16:2298-307. [PMID: 27545888 PMCID: PMC5009114 DOI: 10.1016/j.celrep.2016.07.064] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 06/27/2016] [Accepted: 07/25/2016] [Indexed: 12/21/2022] Open
Abstract
Excitatory and inhibitory transmission onto lateral habenula (LHb) neurons is instrumental for the expression of positive and negative motivational states. However, insights into the molecular mechanisms modulating synaptic transmission and the repercussions for neuronal activity within the LHb remain elusive. Here, we report that, in mice, activation of group I metabotropic glutamate receptors triggers long-term depression at excitatory (eLTD) and inhibitory (iLTD) synapses in the LHb. mGluR-eLTD and iLTD rely on mGluR1 and PKC signaling. However, mGluR-dependent adaptations of excitatory and inhibitory synaptic transmission differ in their expression mechanisms. mGluR-eLTD occurs via an endocannabinoid receptor-dependent decrease in glutamate release. Conversely, mGluR-iLTD occurs postsynaptically through PKC-dependent reduction of β2-containing GABAA-R function. Finally, mGluR-dependent plasticity of excitation or inhibition decides the direction of neuronal firing, providing a synaptic mechanism to bidirectionally control LHb output. We propose mGluR-LTD as a cellular substrate that underlies LHb-dependent encoding of opposing motivational states.
Collapse
Affiliation(s)
- Kristina Valentinova
- Institut du Fer à Moulin, 75005 Paris, France; Inserm, UMR-S 839, 75005 Paris, France; Université Pierre et Marie Curie, 75005 Paris, France
| | - Manuel Mameli
- Institut du Fer à Moulin, 75005 Paris, France; Inserm, UMR-S 839, 75005 Paris, France; Université Pierre et Marie Curie, 75005 Paris, France.
| |
Collapse
|
57
|
Tian X, An L, Gao LY, Bai JP, Wang J, Meng WH, Ren TS, Zhao QC. Compound MQA, a Caffeoylquinic Acid Derivative, Protects Against NMDA-Induced Neurotoxicity and Potential Mechanisms In Vitro. CNS Neurosci Ther 2016; 21:575-84. [PMID: 26096046 DOI: 10.1111/cns.12408] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Revised: 04/16/2015] [Accepted: 04/22/2015] [Indexed: 11/27/2022] Open
Abstract
AIMS Compound MQA (1,5-O-dicaffeoyl-3-O-[4-malic acid methyl ester]-quinic acid) is a natural derivative of caffeoylquinic acid isolated from Arctium lappa L. roots. However, we know little about the effects of MQA on the central nervous system. This study aims to investigate the neuroprotective effects and underlying mechanisms of MQA against the neurotoxicity of N-methyl-d-aspartate (NMDA). METHODS AND RESULTS Pretreatment with MQA attenuated the loss of cell viability after SH-SY5Y cells treated with 1 mM NMDA for 30 min by MTT assay. Hoechst 33342 and Annexin V-PI double staining showed that MQA inhibited NMDA-induced apoptosis. In addition to preventing Ca(2+) influx, the potential mechanisms are associated with increases in the Bcl-2/Bax ratio, attenuation of cytochrome c release, caspase-3, caspase-9 activities, and expressions. Also, MQA inhibited NMDA-induced phosphorylation of ERK1/2, p38, and JNK1/2. Furthermore, deactivation of CREB, AKT, and GSK-3β, upregulation of GluN2B-containing NMDA receptors (NMDARs), and downregulation of GluN2A-containing NMDARs were significantly reversed by MQA treatment. Computational docking simulation indicates that MQA possesses a well affinity for NMDARs. CONCLUSION The protective effects of MQA against NMDA-induced cell injury may be mediated by blocking NMDARs. The potential mechanisms are related with mitochondrial apoptosis, ERK-CREB, AKT/GSK-3β, p38, and JNK1/2 pathway.
Collapse
Affiliation(s)
- Xing Tian
- Department of Pharmacy, General Hospital of Shenyang Military Area Command, Shenyang, China.,Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, China
| | - Li An
- Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, China
| | - Ling-Yue Gao
- Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, China
| | - Jun-Peng Bai
- Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, China
| | - Jian Wang
- Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, China
| | - Wei-Hong Meng
- Department of Pharmacy, General Hospital of Shenyang Military Area Command, Shenyang, China
| | - Tian-Shu Ren
- Department of Pharmacy, General Hospital of Shenyang Military Area Command, Shenyang, China
| | - Qing-Chun Zhao
- Department of Pharmacy, General Hospital of Shenyang Military Area Command, Shenyang, China
| |
Collapse
|
58
|
Domin H, Przykaza Ł, Jantas D, Kozniewska E, Boguszewski PM, Śmiałowska M. Neuroprotective potential of the group III mGlu receptor agonist ACPT-I in animal models of ischemic stroke: In vitro and in vivo studies. Neuropharmacology 2016; 102:276-94. [DOI: 10.1016/j.neuropharm.2015.11.025] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 11/07/2015] [Accepted: 11/24/2015] [Indexed: 01/21/2023]
|
59
|
Webster CI, Caram-Salas N, Haqqani AS, Thom G, Brown L, Rennie K, Yogi A, Costain W, Brunette E, Stanimirovic DB. Brain penetration, target engagement, and disposition of the blood-brain barrier-crossing bispecific antibody antagonist of metabotropic glutamate receptor type 1. FASEB J 2016; 30:1927-40. [PMID: 26839377 DOI: 10.1096/fj.201500078] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 01/22/2016] [Indexed: 12/13/2022]
Abstract
Receptor mediated transcytosis harnessing the cellular uptake and transport of natural ligands across the blood-brain barrier (BBB) has been identified as a means for antibody delivery to the CNS. In this study, we characterized bispecific antibodies in which a BBB-crossing antibody fragment FC5 was used as a BBB carrier. Cargo antibodies were either a high-affinity, selective antibody antagonist of the metabotropic glutamate receptor-1 (BBB-mGluR1), a widely abundant CNS target, or an IgG that does not bind the CNS target (BBB-NiP). Both BBB-NiP and BBB-mGluR1 demonstrated a similar 20-fold enhanced rate of transcytosis across an in vitro BBB model compared with mGluR1 IgG fused to a control antibody fragment. All 3 bispecific antibodies exhibited identical pharmacokinetics in vivo Comparative assessment of BBB-NiP and BBB-mGluR1 revealed that, whereas their serum pharmacokinetics and BBB penetration were identical, their central disposition (brain levels) and elimination (cerebrospinal fluid levels) were widely different, due to central target-mediated removal of the mGluR1-engaging antibody. Central mGluR1 target engagement after systemic administration was demonstrated by a dose-dependent inhibition of mGluR-1-mediated thermal hyperalgesia and by colocalization of the antibody with thalamic neurons involved in mGluR1-mediated pain processing. We demonstrate the feasibility of targeting central G-protein-coupled receptors using a BBB-crossing bispecific antibody approach and emerging principles that govern brain distribution and disposition of these antibodies. These data will be important for designing safe and selective CNS antibody therapeutics.-Webster, C. I., Caram-Salas, N., Haqqani, A. S., Thom, G., Brown, L., Rennie, K., Yogi, A., Costain, W., Brunette, E., Stanimirovic, D. B. Brain penetration, target engagement, and disposition of the blood-brain barrier-crossing bispecific antibody antagonist of metabotropic glutamate receptor type 1.
Collapse
Affiliation(s)
- Carl I Webster
- Antibody Discovery and Protein Engineering, MedImmune, Cambridge, United Kingdom;
| | - Nadia Caram-Salas
- Human Health Therapeutics Portfolio, National Research Council of Canada, Ottawa, Ontario, Canada
| | - Arsalan S Haqqani
- Human Health Therapeutics Portfolio, National Research Council of Canada, Ottawa, Ontario, Canada
| | - George Thom
- Antibody Discovery and Protein Engineering, MedImmune, Cambridge, United Kingdom
| | - Lee Brown
- Translational Sciences, MedImmune, Cambridge, United Kingdom; and
| | - Kerry Rennie
- Human Health Therapeutics Portfolio, National Research Council of Canada, Ottawa, Ontario, Canada
| | - Alvaro Yogi
- Human Health Therapeutics Portfolio, National Research Council of Canada, Ottawa, Ontario, Canada
| | - Willard Costain
- Human Health Therapeutics Portfolio, National Research Council of Canada, Ottawa, Ontario, Canada
| | - Eric Brunette
- Human Health Therapeutics Portfolio, National Research Council of Canada, Ottawa, Ontario, Canada
| | - Danica B Stanimirovic
- Human Health Therapeutics Portfolio, National Research Council of Canada, Ottawa, Ontario, Canada
| |
Collapse
|
60
|
Marciniak M, Chruścicka B, Lech T, Burnat G, Pilc A. Expression of group III metabotropic glutamate receptors in the reproductive system of male mice. Reprod Fertil Dev 2016; 28:369-74. [DOI: 10.1071/rd14132] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Accepted: 06/12/2014] [Indexed: 01/29/2023] Open
Abstract
Although the presence of metabotropic glutamate (mGlu) receptors in the central nervous system is well documented, they have recently been found in peripheral and non-neuronal tissues. In the present study we investigated the expression of group III mGlu receptors in the reproductive system of male mice. Reverse transcription–polymerase chain reaction analysis revealed the presence of mGlu6, mGlu7 and mGlu8 (but not mGlu4) receptor transcripts in testes and epididymides from adult mice. In addition, expression of mGlu6 (Grm6) and mGlu8 receptor (Grm8) mRNA was detected in spermatozoa isolated from the vas deferens. The vas deferens was found to contain only mGlu7 receptor (Grm7) mRNA, which was particularly intense in 21-day-old male mice. In penile homogenates, only the mGlu7 receptor signal was detected. Genetic ablation of the mGlu7 receptor in males led to fertility disorders manifested by decreased insemination capability as well as deterioration of sperm parameters, particularly sperm motility, vitality, sperm membrane integrity and morphology, with a simultaneous increase in sperm concentration. These results indicate that constitutively expressed mGlu receptors in the male reproductive system may play an important role in ejaculation and/or erection processes, as well as in the formation and maturation of spermatozoa.
Collapse
|
61
|
Hicyilmaz H, Vural H, Delibas N, Sutcu R, Gultekin F, Yilmaz N. The effects of walnut supplementation on hippocampal NMDA receptor subunits NR2A and NR2B of rats. Nutr Neurosci 2015. [DOI: 10.1080/1028415x.2015.1106165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
62
|
Arsenault D, Coulombe K, Zhu A, Gong C, Kil KE, Choi JK, Poutiainen P, Brownell AL. Loss of Metabotropic Glutamate Receptor 5 Function on Peripheral Benzodiazepine Receptor in Mice Prenatally Exposed to LPS. PLoS One 2015; 10:e0142093. [PMID: 26536027 PMCID: PMC4633140 DOI: 10.1371/journal.pone.0142093] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 10/16/2015] [Indexed: 11/18/2022] Open
Abstract
Parental microglial induced neuroinflammation, triggered by bacterial- or viral infections, can induce neuropsychiatric disorders like schizophrenia and autism to offspring in animal models. Recent investigations suggest that microglia, the resident immune cells of the brain, provides a link between neurotransmission, immune cell activation, brain inflammation and neuronal dysfunction seen with the offspring. Relatively little is known about how reduction of brain inflammation and restoration of glial function are associated with diminution of brain degeneration and behavioral deficits in offspring. Increased mGluR5 expression and the long-lasting excitotoxic effects of the neurotoxin during brain development are associated with the glial dysfunctions. We investigated the relationship of mGluR5 and PBR and how they regulate glial function and inflammatory processes in mice prenatally exposed to LPS (120μg/kg, between gestational days 15 and 17), an inflammatory model of a psychiatric disorder. Using PET imaging, we showed that pharmacological activation of mGluR5 during 5 weeks reduced expression of classic inflammation marker PBR in many brain areas and that this molecular association was not present in LPS-exposed offspring. The post-mortem analysis revealed that the down regulation of PBR was mediated through activation of mGluR5 in astrocytes. In addition, we demonstrated that this interaction is defective in a mouse model of the psychiatric deficit offering a novel insight of mGluR5 involvement to brain related disorders and PBR related imaging studies. In conclusion, mGluR5 driven glutamatergic activity regulates astrocytic functions associated with PBR (cholesterol transport, neurosteroidogenesis, glial phenotype) during maturation and could be associated with neuropsychiatric disorders in offspring.
Collapse
Affiliation(s)
- Dany Arsenault
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, United States of America
| | - Katherine Coulombe
- Centre de Recherche du CHU de Québec, Axe Neurosciences, Faculté de Pharmacie, Université Laval, Québec, QC, Canada
| | - Aijun Zhu
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, United States of America
| | - Chunyu Gong
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, United States of America
| | - Kun-Eek Kil
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, United States of America
| | - Ji-Kyung Choi
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, United States of America
| | - Pekka Poutiainen
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, United States of America
| | - Anna-Liisa Brownell
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
63
|
Thiazole: a promising heterocycle for the development of potent CNS active agents. Eur J Med Chem 2014; 92:1-34. [PMID: 25544146 DOI: 10.1016/j.ejmech.2014.12.031] [Citation(s) in RCA: 112] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Revised: 11/10/2014] [Accepted: 12/18/2014] [Indexed: 01/15/2023]
Abstract
Thiazole is a valuable scaffold in the field of medicinal chemistry and has accounted to display a variety of biological activities. Thiazole and its derivatives have attracted continuing interest to design various novel CNS active agents. In the past few decades, thiazoles have been widely used to develop a variety of therapeutic agents against numerous CNS targets. Thiazole containing drug molecules are currently being used in treatment of various CNS disorders and a number of thiazole derivatives are also presently in clinical trials. A lot of research has been carried out on thiazole and their analogues, which has proved their efficacy to overcome several CNS disorders in rodent as well as primate models. The aim of present review is to highlights diverse CNS activities displayed by thiazole and their derivatives. SAR of this nucleus has also been well discussed. This review covers the recent updates present in literature and will surely provide a greater insight for the designing and development of potent thiazole based CNS active agents in future.
Collapse
|
64
|
Laukkanen V, Kärkkäinen O, Kupila J, Kautiainen H, Tiihonen J, Storvik M. Increased metabotropic glutamate 2/3 receptor binding in the perigenual anterior cingulate cortex of Cloninger type 2 alcoholics: a whole-hemisphere autoradiography study. Alcohol Alcohol 2014; 50:62-7. [PMID: 25425009 DOI: 10.1093/alcalc/agu081] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
AIMS Metabotropic glutamate receptors 2 and 3 (mGluR2/3) contribute to control the level of glutamate in the synapse. In rodents, mGluR2/3 agonists attenuate the reinstatement of alcohol-seeking behavior. Linking possible alterations of the mGluR2/3 system to the etiology and type of alcoholism could provide valuable information for the development of novel mGluR2/3 function modulating therapies in addiction treatment. To date, mGluR2/3 binding density has not been studied in human alcoholics. We aimed to investigate the possible differences in mGluR2/3 binding between Cloninger type 1 anxiety-prone and type 2 impulsive alcoholics and controls. METHODS We performed a post-mortem whole-hemisphere autoradiography to study the mGluR2/3 binding density of 9 type 1 alcoholics, 8 type 2 alcoholics and 10 controls. [(3)H]LY341495, a potent group II metabotropic glutamate receptor antagonist, was used as the radio-ligand with l-glutamate as a displacer. RESULTS [(3)H]LY341495 binding density was statistically significantly increased (P = 0.046) in the perigenual anterior cingulate cortex (pACC) of type 2 alcoholics when compared with controls. In other brain areas, no significant difference between the groups was found. CONCLUSION This preliminary study suggests that impulsive type 2 alcoholics might have alterations in the mGluR2/3 function in the pACC, a brain area presumed to be involved in the control of drug-seeking behaviors and self-control.
Collapse
Affiliation(s)
- Virpi Laukkanen
- Department of Forensic Psychiatry, University of Eastern Finland, Niuvanniemi Hospital, Niuvankuja 65, FI-70240 Kuopio, Finland Department of Psychiatry, Kuopio University Hospital, PO Box 1777, FI-70211 Kuopio, Finland
| | - Olli Kärkkäinen
- Department of Pharmacology and Toxicology, University of Eastern Finland, PO Box 1627, FI-70211 Kuopio, Finland
| | - Jukka Kupila
- Department of Forensic Psychiatry, University of Eastern Finland, Niuvanniemi Hospital, Niuvankuja 65, FI-70240 Kuopio, Finland
| | - Hannu Kautiainen
- Unit of Primary Health Care, Helsinki University Central Hospital, FI-00014 Helsinki, Finland Department of General Practice, University of Helsinki, FI-00014 Helsinki, Finland
| | - Jari Tiihonen
- Department of Forensic Psychiatry, University of Eastern Finland, Niuvanniemi Hospital, Niuvankuja 65, FI-70240 Kuopio, Finland Department of Clinical Neuroscience, Karolinska Institutet, Karolinska Sjukhuset, 17176 Stockholm, Sweden
| | - Markus Storvik
- Department of Pharmacology and Toxicology, University of Eastern Finland, PO Box 1627, FI-70211 Kuopio, Finland
| |
Collapse
|
65
|
Amalric M. Targeting metabotropic glutamate receptors (mGluRs) in Parkinson's disease. Curr Opin Pharmacol 2014; 20:29-34. [PMID: 25462289 DOI: 10.1016/j.coph.2014.11.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Revised: 11/03/2014] [Accepted: 11/03/2014] [Indexed: 12/28/2022]
Abstract
The interplay between dopamine and glutamate in the basal ganglia regulate critical aspects of motor and cognitive behavior. Metabotropic glutamate (mGlu) receptors are key modulators of glutamatergic dysfunction in Parkinson's disease (PD). Preclinical evidence demonstrate that group I mGlu receptor antagonism and groups II and III mGlu receptor activation improve motor symptomatology of PD and decrease l-DOPA-induced dyskinesia by regulating excitatory and inhibitory transmission in the basal ganglia. Emotional and cognitive deficits are also observed in PD. Treatment of these symptoms is challenging and underscore the need for novel effective and well tolerated pharmacological treatments. This article will thus review the currently available knowledge regarding the therapeutic potential of targeting mGlu receptors to restore motor and nonmotor symptoms of PD.
Collapse
Affiliation(s)
- Marianne Amalric
- Aix-Marseille University, CNRS UMR 7291, Laboratoire de Neurosciences Cognitives (LNC), FR3C 3512, 13331 Marseille, France.
| |
Collapse
|
66
|
Moha ou Maati H, Bourcier-Lucas C, Veyssiere J, Kanzari A, Heurteaux C, Borsotto M, Haddjeri N, Lucas G. The peptidic antidepressant spadin interacts with prefrontal 5-HT4 and mGluR2 receptors in the control of serotonergic function. Brain Struct Funct 2014; 221:21-37. [DOI: 10.1007/s00429-014-0890-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Accepted: 09/11/2014] [Indexed: 10/24/2022]
|
67
|
Yu C, Wang J. Neuroprotective effect of penehyclidine hydrochloride on focal cerebral ischemia-reperfusion injury. Neural Regen Res 2014; 8:622-32. [PMID: 25206707 PMCID: PMC4145985 DOI: 10.3969/j.issn.1673-5374.2013.07.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2012] [Accepted: 02/07/2013] [Indexed: 12/30/2022] Open
Abstract
Penehyclidine hydrochloride can promote microcirculation and reduce vascular permeability. However, the role of penehyclidine hydrochloride in cerebral ischemia-reperfusion injury remains unclear. In this study, in vivo middle cerebral artery occlusion models were established in experimental rats, and penehyclidine hydrochloride pretreatment was given via intravenous injection prior to model establishment. Tetrazolium chloride, terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate-biotin nick end labeling and immunohistochemical staining showed that, penehyclidine hydrochloride pretreatment markedly attenuated neuronal histopathological changes in the cortex, hippocampus and striatum, reduced infarction size, increased the expression level of Bcl-2, decreased the expression level of caspase-3, and inhibited neuronal apoptosis in rats with cerebral ischemia-reperfusion injury. Xanthine oxidase and thiobarbituric acid chromogenic results showed that penehyclidine hydrochloride upregulated the activity of superoxide dismutase and downregulated the concentration of malondialdehyde in the ischemic cerebral cortex and hippocampus, as well as reduced the concentration of extracellular excitatory amino acids in rats with cerebral ischemia-reperfusion injury. In addition, penehyclidine hydrochloride inhibited the expression level of the NR1 subunit in hippocampal nerve cells in vitro following oxygen-glucose deprivation, as detected by PCR. Experimental findings indicate that penehyclidine hydrochloride attenuates neuronal apoptosis and oxidative stress injury after focal cerebral ischemia-reperfusion, thus exerting a neuroprotective effect.
Collapse
Affiliation(s)
- Cuicui Yu
- Department of Anesthesiology, First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning Province, China ; Department of Anesthesiology, Affiliated Yuhuangding Hospital, Medical College of Qingdao University, Yantai 264000, Shandong Province, China
| | - Junke Wang
- Department of Anesthesiology, First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| |
Collapse
|
68
|
Lentivirus mediated siRNA against GluN2B subunit of NMDA receptor reduces nociception in a rat model of neuropathic pain. BIOMED RESEARCH INTERNATIONAL 2014; 2014:871637. [PMID: 25243192 PMCID: PMC4163390 DOI: 10.1155/2014/871637] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 07/08/2014] [Accepted: 08/07/2014] [Indexed: 11/17/2022]
Abstract
Although neuropathic pain (NP) is still not fully understood by scientists and clinicians alike, studies suggest that N-methyl-D-aspartate (NMDA) receptors play an important role in the induction and maintenance of NP. A promising treatment for NP is through the downregulation of NMDA subunit GluN2B by RNA interference; however, naked siRNA (small interference RNA) is not effective in long-term treatments. In order to concoct a viable prolonged treatment for NP, Lv-siGluN2B (lentivirus carrying siRNA targeting GluN2B subunit) was prepared and the antinociception effects were observed in chronic constriction injury (CCI) rats in the present study. Results showed that Lv-siGluN2B was transduced into spinal cord cells after intrathecal injections and effectively reduced the nociception induced by sciatic nerve ligation while inhibiting the mRNA and protein expression of GluN2B. This antinociception effect lasted approximately 7 weeks. These findings suggest that GluN2B subunit could be a target for NP treatment and Lv-siGluN2B represents a new potential option for long-term treatment of NP.
Collapse
|
69
|
Lopatina NG, Vaido AI, Zachepilo TG, Kamyshev NG. Metabotropic receptor of the group I of the 5th subtype (ImGluR5) in honeybee associative olfactory learning. J EVOL BIOCHEM PHYS+ 2014. [DOI: 10.1134/s0022093014030077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
70
|
Arsenault D, Zhu A, Gong C, Kil KE, Kura S, Choi JK, Brownell AL. Hypo-anxious phenotype of adolescent offspring prenatally exposed to LPS is associated with reduced mGluR5 expression in hippocampus. ACTA ACUST UNITED AC 2014; 3:202-211. [PMID: 25419490 DOI: 10.4236/ojmp.2014.33022] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Many studies have reported long-term modulation of metabotropic glutamate receptor 5 (mGluR5) by inflammatory processes and a pharmacological modulation of mGluR5 is known to regulate anxiety level. However, it is not known if non-pharmacological modulation of mGluR5 by inflammation impaired the unconditional level of anxiety. In this study, we investigated this relation in LPS prenatal immune challenge (120μg/kg, 3x i.p. injection in late gestation), a developmental model of neuroinflammation in which some studies have reported hypo-anxious phenotype. Using positron emission tomographic imaging (PET) approaches, we have demonstrated a decrease in the binding potential of [18F]fluoro-5-(2-pyridinylethynyl)benzonitrile ([18F]FPEB, a radioligand for mGluR5) in hippocampus of adolescent offspring prenatally exposed to LPS, without significant change in the binding of [11C]peripheral benzodiazepine receptor 28 ([11C]PBR28), an inflammatory marker. In addition, dark-light box emergence test revealed a lower level of anxiety in LPS-exposed offspring and this behavioural phenotype was associated with the binding potential of [18F]FPEB in hippocampus. These results confirm that neuroinflammation during developmental phase modulates the physiology of mGluR5 and this alteration can be associated with behavioural phenotype related to anxiety. In addition, this study supports a hypotheses that mGluR5 could be used as a diagnostic target in anxiety.
Collapse
Affiliation(s)
- Dany Arsenault
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, 149 13 Street, Charlestown, Massachusetts 02129, USA
| | - Aijun Zhu
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, 149 13 Street, Charlestown, Massachusetts 02129, USA
| | - Chunyu Gong
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, 149 13 Street, Charlestown, Massachusetts 02129, USA
| | - Kun-Eek Kil
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, 149 13 Street, Charlestown, Massachusetts 02129, USA
| | - Sreekanth Kura
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, 149 13 Street, Charlestown, Massachusetts 02129, USA
| | - Ji-Kyung Choi
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, 149 13 Street, Charlestown, Massachusetts 02129, USA
| | - Anna-Liisa Brownell
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, 149 13 Street, Charlestown, Massachusetts 02129, USA
| |
Collapse
|
71
|
Ambrosi G, Cerri S, Blandini F. A further update on the role of excitotoxicity in the pathogenesis of Parkinson’s disease. J Neural Transm (Vienna) 2014; 121:849-59. [DOI: 10.1007/s00702-013-1149-z] [Citation(s) in RCA: 131] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Accepted: 12/19/2013] [Indexed: 11/30/2022]
|
72
|
Schumann G, Binder EB, Holte A, de Kloet ER, Oedegaard KJ, Robbins TW, Walker-Tilley TR, Bitter I, Brown VJ, Buitelaar J, Ciccocioppo R, Cools R, Escera C, Fleischhacker W, Flor H, Frith CD, Heinz A, Johnsen E, Kirschbaum C, Klingberg T, Lesch KP, Lewis S, Maier W, Mann K, Martinot JL, Meyer-Lindenberg A, Müller CP, Müller WE, Nutt DJ, Persico A, Perugi G, Pessiglione M, Preuss UW, Roiser JP, Rossini PM, Rybakowski JK, Sandi C, Stephan KE, Undurraga J, Vieta E, van der Wee N, Wykes T, Haro JM, Wittchen HU. Stratified medicine for mental disorders. Eur Neuropsychopharmacol 2014; 24:5-50. [PMID: 24176673 DOI: 10.1016/j.euroneuro.2013.09.010] [Citation(s) in RCA: 115] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Revised: 09/09/2013] [Accepted: 09/26/2013] [Indexed: 12/13/2022]
Abstract
There is recognition that biomedical research into the causes of mental disorders and their treatment needs to adopt new approaches to research. Novel biomedical techniques have advanced our understanding of how the brain develops and is shaped by behaviour and environment. This has led to the advent of stratified medicine, which translates advances in basic research by targeting aetiological mechanisms underlying mental disorder. The resulting increase in diagnostic precision and targeted treatments may provide a window of opportunity to address the large public health burden, and individual suffering associated with mental disorders. While mental health and mental disorders have significant representation in the "health, demographic change and wellbeing" challenge identified in Horizon 2020, the framework programme for research and innovation of the European Commission (2014-2020), and in national funding agencies, clear advice on a potential strategy for mental health research investment is needed. The development of such a strategy is supported by the EC-funded "Roadmap for Mental Health Research" (ROAMER) which will provide recommendations for a European mental health research strategy integrating the areas of biomedicine, psychology, public health well being, research integration and structuring, and stakeholder participation. Leading experts on biomedical research on mental disorders have provided an assessment of the state of the art in core psychopathological domains, including arousal and stress regulation, affect, cognition social processes, comorbidity and pharmacotherapy. They have identified major advances and promising methods and pointed out gaps to be addressed in order to achieve the promise of a stratified medicine for mental disorders.
Collapse
Affiliation(s)
- Gunter Schumann
- MRC-Social Genetic Developmental Psychiatry Centre, Institute of Psychiatry, King's College London, PO80, 16 De Crespigny Park, London SE5 8AF, UK.
| | | | - Arne Holte
- Norwegian Institute of Public Health, Oslo, Norway
| | - E Ronald de Kloet
- Department of Endocrinology and Metabolism, Leiden University Medical Centre and Medical Pharmacology, LACDR, Leiden University, The Netherlands
| | - Ketil J Oedegaard
- Department of Clinical Medicine, Section of Psychiatry, University of Bergen and Psychiatric division, Health Bergen, Norway
| | - Trevor W Robbins
- Behavioural and Clinical Neuroscience Institute and Department of Psychology, Cambridge University, Cambridge, UK
| | - Tom R Walker-Tilley
- MRC-Social Genetic Developmental Psychiatry Centre, Institute of Psychiatry, King's College London, PO80, 16 De Crespigny Park, London SE5 8AF, UK
| | - Istvan Bitter
- Department of Psychiatry and Psychotherapy, Semmelweis University, Budapest, Hungary
| | - Verity J Brown
- Department of Psychology, University of St Andrews, St Andrews, UK
| | - Jan Buitelaar
- Department of Cognitive Neuroscience, University Medical Center, St Radboud and Karakter Child and Adolescent Psychiatry University Center, Nijmegen, The Netherlands
| | - Roberto Ciccocioppo
- Department of Experimental Medicine and Public Health, University of Camerino, Camerino, Macerata, Italy
| | | | - Carles Escera
- Department of Psychiatry and Clinical Psychobiology, University of Barcelona, Barcelona, Spain
| | - Wolfgang Fleischhacker
- Department of Psychiatry and Psychotherapy, Medical University Innsbruck, Innsbruck, Austria
| | - Herta Flor
- Department of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Chris D Frith
- Wellcome Trust Centre for Neuroimaging, University College London, London, UK
| | - Andreas Heinz
- Berlin School of Mind and Brain, Bernstein Center for Computational Neuroscience (BCCN), Clinic for Psychiatry and Psychotherapy, Charité - Universitätsmedizin, Berlin, Germany
| | - Erik Johnsen
- Department of Clinical Medicine, Section of Psychiatry, University of Bergen and Psychiatric division, Health Bergen, Norway
| | - Clemens Kirschbaum
- Technische Universität Dresden, Department of Psychology, Dresden, Germany
| | | | - Klaus-Peter Lesch
- Division of Molecular Psychiatry, Laboratory of Translational Neuroscience, University of Würzburg, Würzburg, Germany and Department of Neuroscience, School of Mental Health and Neuroscience (MHENS), Maastricht University, Maastricht, The Netherlands
| | - Shon Lewis
- University of Manchester, Manchester, UK
| | - Wolfgang Maier
- Department of Psychiatry and Psychotherapy, University of Bonn, Bonn, Germany
| | - Karl Mann
- Department of Addictive Behaviour and Addiction Medicine, Central Institute of Mental Health, Mannheim, Germany
| | - Jean-Luc Martinot
- Institut National de la Santé et de la Recherche Médicale, INSERM CEA Unit 1000 "Imaging & Psychiatry", University Paris Sud, Orsay; AP-HP Department of Adolescent Psychopathology and Medicine, Maison de Solenn, University Paris Descartes, Paris, France
| | - Andreas Meyer-Lindenberg
- Department of Cognitive and Clinical Neuroscience, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Christian P Müller
- Psychiatric University Hospital, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Walter E Müller
- Department of Pharmacology, Biocenter Niederursel, University of Frankfurt, Frankfurt, Germany
| | - David J Nutt
- Neuropsychopharmacology Unit, Division of Brain Sciences, Imperial College, London, UK
| | - Antonio Persico
- Child and Adolescent Neuropsychiatry Unit & Laboratory of Molecular Psychiatry and Neurogenetics, University Campus Bio-Medico, Rome, Italy
| | - Giulio Perugi
- Department of Psychiatry, University of Pisa, Pisa, Italy
| | - Mathias Pessiglione
- Institut du Cerveau et de la Moelle épinière (ICM), Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Ulrich W Preuss
- Department of Psychiatry, Psychotherapy and Psychosomatics, Martin-Luther-University of Halle-Wittenberg, Halle/Saale, Germany
| | - Jonathan P Roiser
- Institute of Cognitive Neuroscience, University College London, London, UK
| | - Paolo M Rossini
- Department of Geriatrics, Neuroscience & Orthopaedics, Catholic University of Sacred Heart, Policlinico A. Gemelli, Rome, Italy
| | - Janusz K Rybakowski
- Department of Adult Psychiatry, Poznan University of Medical Sciences, Poznan, Poland
| | - Carmen Sandi
- Laboratory of Behavioural Genetics, Brain Mind Institute, EPFL, Lausanne, Switzerland
| | - Klaas E Stephan
- Translational Neuromodeling Unit (TNU), Institute for Biomedical Engineering, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Juan Undurraga
- Translational Neuromodeling Unit (TNU), Institute for Biomedical Engineering, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Eduard Vieta
- Bipolar Disorders Programme, Institute of Neuroscience, Hospital Clínic Barcelona, IDIBAPS, CIBERSAM, University of Barcelona, Barcelona, Catalonia, Spain
| | - Nic van der Wee
- Leiden Institute for Brain and Cogntion/Psychiatric Neuroimaging, Dept. of Psychiatry, Leiden University Medical Center, The Netherlands
| | - Til Wykes
- Department of Psychology, Institute of Psychiatry, King's College London, UK
| | - Josep Maria Haro
- Parc Sanitari Sant Joan de Déu, University of Barcelona, CIBERSAM, Barcelona, Spain
| | - Hans Ulrich Wittchen
- Institute of Clinical Psychology and Psychotherapy, TU Dresden, Dresden, Germany
| |
Collapse
|
73
|
Williams CJ, Dexter DT. Neuroprotective and symptomatic effects of targeting group III mGlu receptors in neurodegenerative disease. J Neurochem 2013; 129:4-20. [PMID: 24224472 DOI: 10.1111/jnc.12608] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2013] [Revised: 10/28/2013] [Accepted: 11/06/2013] [Indexed: 12/21/2022]
Abstract
Neurodegenerative disorders possess common pathological mechanisms, such as protein aggregation, inflammation, oxidative stress (OS) and excitotoxicity, raising the possibility of shared therapeutic targets. As a result of the selective cellular and regional expression of group III metabotropic glutamate (mGlu) receptors, drugs targeting such receptors have demonstrated both neuroprotective properties and symptomatic improvements in several models of neurodegeneration. In recent years, the discovery and development of subtype-selective ligands for the group III mGlu receptors has gained pace, allowing further research into the functions of these receptors and revealing their roles in health and disease. Activation of this class of receptors results in neuroprotection, with a variety of underlying mechanisms implicated. Group III mGlu receptor stimulation prevents excitotoxicity by inhibiting glutamate release from neurons and microglia and increasing glutamate uptake by astrocytes. It also attenuates the neuroinflammatory response by reducing glial reactivity and encourages neurotrophic phenotypes. This article will review the current literature with regard to the neuroprotective and symptomatic effects of group III mGlu receptor activation and discuss their promise as therapeutic targets in neurodegenerative disease. We review the neuroprotective and symptomatic effects of targeting group III mGlu receptors in neurodegenerative disease: Excess extracellular glutamate causes overactivation of NMDA receptors resulting in excitotoxicity. Externalization of phosphatidylserine stimulates phagocytosis of neurons by activated microglia, which contribute to damage through glutamate and pro-inflammatory factor release. Reactive astrocytes produce cytotoxic factors enhancing neuronal cell death. Activation of group III mGlu receptors by glutamate and/or mGlu receptor ligands results in inhibition of glutamate release from presynaptic terminals and microglia, reducing excitotoxicity. Astrocytic glutamate uptake is increased and microglia produce neurotrophic factors.
Collapse
Affiliation(s)
- Claire J Williams
- Parkinson's Disease Research Group, Centre for Neuroinflammation and Neurodegeneration, Division of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, London, UK
| | | |
Collapse
|
74
|
Dexter DT, Jenner P. Parkinson disease: from pathology to molecular disease mechanisms. Free Radic Biol Med 2013; 62:132-144. [PMID: 23380027 DOI: 10.1016/j.freeradbiomed.2013.01.018] [Citation(s) in RCA: 469] [Impact Index Per Article: 39.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2012] [Revised: 01/17/2013] [Accepted: 01/18/2013] [Indexed: 12/21/2022]
Abstract
Parkinson disease (PD) is a complex neurodegenerative disorder with both motor and nonmotor symptoms owing to a spreading process of neuronal loss in the brain. At present, only symptomatic treatment exists and nothing can be done to halt the degenerative process, as its cause remains unclear. Risk factors such as aging, genetic susceptibility, and environmental factors all play a role in the onset of the pathogenic process but how these interlink to cause neuronal loss is not known. There have been major advances in the understanding of mechanisms that contribute to nigral dopaminergic cell death, including mitochondrial dysfunction, oxidative stress, altered protein handling, and inflammation. However, it is not known if the same processes are responsible for neuronal loss in nondopaminergic brain regions. Many of the known mechanisms of cell death are mirrored in toxin-based models of PD, but neuronal loss is rapid and not progressive and limited to dopaminergic cells, and drugs that protect against toxin-induced cell death have not translated into neuroprotective therapies in humans. Gene mutations identified in rare familial forms of PD encode proteins whose functions overlap widely with the known molecular pathways in sporadic disease and these have again expanded our knowledge of the neurodegenerative process but again have so far failed to yield effective models of sporadic disease when translated into animals. We seem to be missing some key parts of the jigsaw, the trigger event starting many years earlier in the disease process, and what we are looking at now is merely part of a downstream process that is the end stage of neuronal death.
Collapse
Affiliation(s)
- David T Dexter
- Parkinson's Disease Research Group, Centre for Neuroinflammation & Neurodegeneration, Division of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Peter Jenner
- Neurodegenerative Diseases Research Group, Institute of Pharmaceutical Science, School of Biomedical Sciences, King's College London, London SE1 9NH, UK.
| |
Collapse
|
75
|
Gasparini F, Di Paolo T, Gomez-Mancilla B. Metabotropic glutamate receptors for Parkinson's disease therapy. PARKINSON'S DISEASE 2013; 2013:196028. [PMID: 23853735 PMCID: PMC3703788 DOI: 10.1155/2013/196028] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2013] [Accepted: 05/29/2013] [Indexed: 12/21/2022]
Abstract
Excessive glutamatergic signalling within the basal ganglia is implicated in the progression of Parkinson's disease (PD) and inthe emergence of dyskinesia associated with long-term treatment with L-DOPA. There is considerable research focus on the discovery and development of compounds that modulate glutamatergic signalling via glutamate receptors, as treatments for PD and L-DOPA-induced dyskinesia (LID). Although initial preclinical studies with ionotropic glutamate receptor antagonists showed antiparkinsonian and antidyskinetic activity, their clinical use was limited due to psychiatric adverse effects, with the exception of amantadine, a weak N-methyl-d-aspartate (NMDA) antagonist, currently used to reduce dyskinesia in PD patients. Metabotropic receptor (mGlu receptor) modulators were considered to have a more favourable side-effect profile, and several agents have been studied in preclinical models of PD. The most promising results have been seen clinically with selective antagonists of mGlu5 receptor and preclinically with selective positive allosteric modulators of mGlu4 receptor. The growing understanding of glutamate receptor crosstalk also raises the possibility of more precise modulation of glutamatergic transmission, which may lead to the development of more effective agents for PD.
Collapse
Affiliation(s)
- Fabrizio Gasparini
- Novartis Pharma AG, Novartis Institutes for BioMedical Research Basel, Forum 1, Novartis Campus, 4056 Basel, Switzerland
| | - Thérèse Di Paolo
- Neuroscience Research Unit, Centre Hospitalier Universitaire de Québec, CHUL, Quebec City, QC, Canada G1V 4G2
- Faculty of Pharmacy, Laval University, Quebec City, QC, Canada G1K 7P4
| | - Baltazar Gomez-Mancilla
- Novartis Pharma AG, Novartis Institutes for BioMedical Research Basel, Forum 1, Novartis Campus, 4056 Basel, Switzerland
| |
Collapse
|
76
|
Trabanco AA, Cid JM. mGluR2 positive allosteric modulators: a patent review (2009 - present). Expert Opin Ther Pat 2013; 23:629-47. [PMID: 23452205 DOI: 10.1517/13543776.2013.777043] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION The mGlu2 receptor, which belongs to the group II subfamily of metabotropic glutamate receptors (mGlu) along with the mGlu3 receptor, has proven to be of particular importance in neuropharmacology. Preferentially expressed on presynaptic nerve terminals, the mGlu2 receptor negatively modulates glutamate and GABA release and is widely distributed in the brain. High levels of mGlu2 receptors are seen in brain areas such as prefrontal cortex, hippocampus and amygdala where glutamate hyperfunction may be implicated in disorders and diseases such as anxiety and schizophrenia. Given the promise offered by mGlu2/3 receptor activation, there is increased interest in identifying small molecules which activate the receptor. A preferred approach is via positive allosteric modulators (PAMs) which bind at an alternative site to agonists. AREAS COVERED This review covers the patent applications which were published between April 2009 and December 2012 on PAMs of the mGlu2, and it is a continuation of an earlier review published in this journal. EXPERT OPINION Advances in medicinal chemistry and pharmacology have set the stage in the field of mGlu2 receptor PAMs. Compounds currently advancing in clinical trials will soon establish the therapeutic potential of this allosteric approach.
Collapse
Affiliation(s)
- Andrés A Trabanco
- Janssen Research and Development, Neuroscience Medicinal Chemistry Department, Toledo, Spain.
| | | |
Collapse
|