1
|
Huang L, Xiao W, Wang Y, Li J, Gong J, Tu E, Long L, Xiao B, Yan X, Wan L. Metabotropic glutamate receptors (mGluRs) in epileptogenesis: an update on abnormal mGluRs signaling and its therapeutic implications. Neural Regen Res 2024; 19:360-368. [PMID: 37488891 PMCID: PMC10503602 DOI: 10.4103/1673-5374.379018] [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: 01/24/2023] [Revised: 04/07/2023] [Accepted: 05/22/2023] [Indexed: 07/26/2023] Open
Abstract
Epilepsy is a neurological disorder characterized by high morbidity, high recurrence, and drug resistance. Enhanced signaling through the excitatory neurotransmitter glutamate is intricately associated with epilepsy. Metabotropic glutamate receptors (mGluRs) are G protein-coupled receptors activated by glutamate and are key regulators of neuronal and synaptic plasticity. Dysregulated mGluR signaling has been associated with various neurological disorders, and numerous studies have shown a close relationship between mGluRs expression/activity and the development of epilepsy. In this review, we first introduce the three groups of mGluRs and their associated signaling pathways. Then, we detail how these receptors influence epilepsy by describing the signaling cascades triggered by their activation and their neuroprotective or detrimental roles in epileptogenesis. In addition, strategies for pharmacological manipulation of these receptors during the treatment of epilepsy in experimental studies is also summarized. We hope that this review will provide a foundation for future studies on the development of mGluR-targeted antiepileptic drugs.
Collapse
Affiliation(s)
- Leyi Huang
- Department of Anatomy and Neurobiology, Central South University Xiangya Medical School, Changsha, Hunan Province, China
| | - Wenjie Xiao
- Department of Anatomy and Neurobiology, Central South University Xiangya Medical School, Changsha, Hunan Province, China
| | - Yan Wang
- Department of Anatomy and Neurobiology, Central South University Xiangya Medical School, Changsha, Hunan Province, China
| | - Juan Li
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Jiaoe Gong
- Department of Neurology, Hunan Children’s Hospital, Changsha, Hunan Province, China
| | - Ewen Tu
- Department of Neurology, Brain Hospital of Hunan Province, Changsha, Hunan Province, China
| | - Lili Long
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Bo Xiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Xiaoxin Yan
- Department of Anatomy and Neurobiology, Central South University Xiangya Medical School, Changsha, Hunan Province, China
| | - Lily Wan
- Department of Anatomy and Neurobiology, Central South University Xiangya Medical School, Changsha, Hunan Province, China
| |
Collapse
|
2
|
Torazza C, Provenzano F, Gallia E, Cerminara M, Balbi M, Bonifacino T, Tessitore S, Ravera S, Usai C, Musante I, Puliti A, Van Den Bosch L, Jafar-nejad P, Rigo F, Milanese M, Bonanno G. Genetic Downregulation of the Metabotropic Glutamate Receptor Type 5 Dampens the Reactive and Neurotoxic Phenotype of Adult ALS Astrocytes. Cells 2023; 12:1952. [PMID: 37566031 PMCID: PMC10416852 DOI: 10.3390/cells12151952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/24/2023] [Accepted: 07/25/2023] [Indexed: 08/12/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by progressive degeneration of motor neurons (MNs). Astrocytes display a toxic phenotype in ALS, which results in MN damage. Glutamate (Glu)-mediated excitotoxicity and group I metabotropic glutamate receptors (mGluRs) play a pathological role in the disease progression. We previously demonstrated that in vivo genetic ablation or pharmacological modulation of mGluR5 reduced astrocyte activation and MN death, prolonged survival and ameliorated the clinical progression in the SOD1G93A mouse model of ALS. This study aimed to investigate in vitro the effects of mGluR5 downregulation on the reactive spinal cord astrocytes cultured from adult late symptomatic SOD1G93A mice. We observed that mGluR5 downregulation in SOD1G93A astrocytes diminished the cytosolic Ca2+ overload under resting conditions and after mGluR5 simulation and reduced the expression of the reactive glial markers GFAP, S100β and vimentin. In vitro exposure to an anti-mGluR5 antisense oligonucleotide or to the negative allosteric modulator CTEP also ameliorated the altered reactive astrocyte phenotype. Downregulating mGluR5 in SOD1G93A mice reduced the synthesis and release of the pro-inflammatory cytokines IL-1β, IL-6 and TNF-α and ameliorated the cellular bioenergetic profile by improving the diminished oxygen consumption and ATP synthesis and by lowering the excessive lactate dehydrogenase activity. Most relevantly, mGluR5 downregulation hampered the neurotoxicity of SOD1G93A astrocytes co-cultured with spinal cord MNs. We conclude that selective reduction in mGluR5 expression in SOD1G93A astrocytes positively modulates the astrocyte reactive phenotype and neurotoxicity towards MNs, further supporting mGluR5 as a promising therapeutic target in ALS.
Collapse
Affiliation(s)
- Carola Torazza
- Department of Pharmacy (DIFAR), University of Genoa, Viale Cembrano 4, 16148 Genova, Italy; (C.T.); (F.P.); (E.G.); (M.B.); (T.B.); (S.T.); (G.B.)
| | - Francesca Provenzano
- Department of Pharmacy (DIFAR), University of Genoa, Viale Cembrano 4, 16148 Genova, Italy; (C.T.); (F.P.); (E.G.); (M.B.); (T.B.); (S.T.); (G.B.)
| | - Elena Gallia
- Department of Pharmacy (DIFAR), University of Genoa, Viale Cembrano 4, 16148 Genova, Italy; (C.T.); (F.P.); (E.G.); (M.B.); (T.B.); (S.T.); (G.B.)
| | - Maria Cerminara
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Largo Paolo Daneo, 16132 Genoa, Italy; (M.C.); (A.P.)
- UOC Genetica Medica, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy;
| | - Matilde Balbi
- Department of Pharmacy (DIFAR), University of Genoa, Viale Cembrano 4, 16148 Genova, Italy; (C.T.); (F.P.); (E.G.); (M.B.); (T.B.); (S.T.); (G.B.)
| | - Tiziana Bonifacino
- Department of Pharmacy (DIFAR), University of Genoa, Viale Cembrano 4, 16148 Genova, Italy; (C.T.); (F.P.); (E.G.); (M.B.); (T.B.); (S.T.); (G.B.)
- Inter-University Center for the Promotion of the 3Rs Principles in Teaching & Research (Centro 3R), 56122 Pisa, Italy
| | - Sara Tessitore
- Department of Pharmacy (DIFAR), University of Genoa, Viale Cembrano 4, 16148 Genova, Italy; (C.T.); (F.P.); (E.G.); (M.B.); (T.B.); (S.T.); (G.B.)
| | - Silvia Ravera
- Department of Experimental Medicine (DIMES), University of Genoa, Via Alberti L.B. 2, 16132 Genova, Italy;
| | - Cesare Usai
- Institute of Biophysics, National Research Council (CNR), Via De Marini 6, 16149 Genoa, Italy;
| | - Ilaria Musante
- UOC Genetica Medica, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy;
| | - Aldamaria Puliti
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Largo Paolo Daneo, 16132 Genoa, Italy; (M.C.); (A.P.)
- UOC Genetica Medica, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy;
| | - Ludo Van Den Bosch
- Department of Neurosciences, Experimental Neurology, and Leuven Brain Institute, KU Leuven-University of Leuven, 3000 Leuven, Belgium;
- VIB-Center for Brain & Disease Research, Laboratory of Neurobiology, 3000 Leuven, Belgium
| | | | - Frank Rigo
- Ionis Pharmaceuticals, Carlsbad, CA 92010, USA; (P.J.-n.); (F.R.)
| | - Marco Milanese
- Department of Pharmacy (DIFAR), University of Genoa, Viale Cembrano 4, 16148 Genova, Italy; (C.T.); (F.P.); (E.G.); (M.B.); (T.B.); (S.T.); (G.B.)
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy
| | - Giambattista Bonanno
- Department of Pharmacy (DIFAR), University of Genoa, Viale Cembrano 4, 16148 Genova, Italy; (C.T.); (F.P.); (E.G.); (M.B.); (T.B.); (S.T.); (G.B.)
| |
Collapse
|
3
|
Sivakumar S, Ghasemi M, Schachter SC. Targeting NMDA Receptor Complex in Management of Epilepsy. Pharmaceuticals (Basel) 2022; 15:ph15101297. [PMID: 36297409 PMCID: PMC9609646 DOI: 10.3390/ph15101297] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 10/17/2022] [Accepted: 10/20/2022] [Indexed: 11/05/2022] Open
Abstract
N-methyl-D-aspartate receptors (NMDARs) are widely distributed in the central nervous system (CNS) and play critical roles in neuronal excitability in the CNS. Both clinical and preclinical studies have revealed that the abnormal expression or function of these receptors can underlie the pathophysiology of seizure disorders and epilepsy. Accordingly, NMDAR modulators have been shown to exert anticonvulsive effects in various preclinical models of seizures, as well as in patients with epilepsy. In this review, we provide an update on the pathologic role of NMDARs in epilepsy and an overview of the NMDAR antagonists that have been evaluated as anticonvulsive agents in clinical studies, as well as in preclinical seizure models.
Collapse
Affiliation(s)
- Shravan Sivakumar
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| | - Mehdi Ghasemi
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
- Correspondence: (M.G.); (S.C.S.)
| | - Steven C. Schachter
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02114, USA
- Consortia for Improving Medicine with Innovation & Technology (CIMIT), Boston, MA 02114, USA
- Correspondence: (M.G.); (S.C.S.)
| |
Collapse
|
4
|
Proskurina EY, Zaitsev AV. Regulation of Potassium and Chloride Concentrations in Nervous Tissue as a Method of Anticonvulsant Therapy. J EVOL BIOCHEM PHYS+ 2022. [DOI: 10.1134/s0022093022050015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Abstract
Under some pathological conditions, such as pharmacoresistant
epilepsy, status epilepticus or certain forms of genetic abnormalities,
spiking activity of GABAergic interneurons may enhance excitation
processes in neuronal circuits and provoke the generation of ictal
discharges. As a result, anticonvulsants acting on the GABAergic
system may be ineffective or even increase seizure activity. This
paradoxical effect of the inhibitory system is due to ionic imbalances
in nervous tissue. This review addresses the mechanisms of ictal
discharge initiation in neuronal networks due to the imbalance of
chloride and potassium ions, as well as possible ways to regulate
ionic concentrations. Both the enhancement (or attenuation) of the
activity of certain neuronal ion transporters and ion pumps and
their additional expression via gene therapy can be effective in
suppressing seizure activity caused by ionic imbalances. The Na+–K+-pump,
NKCC1 and KCC2 cotransporters are important for maintaining proper
K+ and Cl– concentrations
in nervous tissue, having been repeatedly considered as pharmacological
targets for antiepileptic exposures. Further progress in this direction
is hampered by the lack of sufficiently selective pharmacological
tools and methods for providing effective drug delivery to the epileptic
focus. The use of the gene therapy techniques, such as overexpressing
of the KCC2 transporter in the epileptic focus, seems to be a more promising
approach. Another possible direction could be the use of optogenetic
tools, namely specially designed light-activated ion pumps or ion
channels. In this case, photon energy can be used to create the
required gradients of chloride and potassium ions, although these
methods also have significant limitations which complicate their
rapid introduction into medicine.
Collapse
|
5
|
Kovalenko AA, Zakharova MV, Schwarz AP, Dyomina AV, Zubareva OE, Zaitsev AV. Changes in Metabotropic Glutamate Receptor Gene Expression in Rat Brain in a Lithium-Pilocarpine Model of Temporal Lobe Epilepsy. Int J Mol Sci 2022; 23:ijms23052752. [PMID: 35269897 PMCID: PMC8910969 DOI: 10.3390/ijms23052752] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 02/27/2022] [Accepted: 02/27/2022] [Indexed: 11/16/2022] Open
Abstract
Preventing epileptogenesis in people at risk is an unmet medical need. Metabotropic glutamate receptors (mGluRs) are promising targets for such therapy. However, drugs acting on mGluRs are not used in the clinic due to limited knowledge of the involvement of mGluRs in epileptogenesis. This study aimed to analyze the changes in gene expression of mGluR subtypes (1-5, 7, 8) in various rat brain regions in the latent and chronic phases of a lithium-pilocarpine model of epilepsy. For this study, multiplex test systems were selected and optimized to analyze mGluR gene expression using RT-qPCR. Region- and phase-specific changes in expression were revealed. During the latent phase, mGluR5 mRNA levels were increased in the dorsal and ventral hippocampus, and expression of group III genes was decreased in the hippocampus and temporal cortex, which could contribute to epileptogenesis. Most of the changes in expression detected in the latent stage were absent in the chronic stage, but mGluR8 mRNA production remained reduced in the hippocampus. Moreover, we found that gene expression of group II mGluRs was altered only in the chronic phase. The study deepened our understanding of the mechanisms of epileptogenesis and suggested that agonists of group III mGluRs are the most promising targets for preventing epilepsy.
Collapse
|
6
|
Lam J, DuBois JM, Rowley J, Rousset OG, González-Otárula KA, Soucy JP, Massarweh G, Hall JA, Guiot MC, Zimmermann M, Minuzzi L, Rosa-Neto P, Kobayashi E. In vivo hippocampal cornu ammonis 1-3 glutamatergic abnormalities are associated with temporal lobe epilepsy surgery outcomes. Epilepsia 2021; 62:1559-1568. [PMID: 34060082 DOI: 10.1111/epi.16952] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 05/11/2021] [Accepted: 05/12/2021] [Indexed: 11/30/2022]
Abstract
OBJECTIVE Previous positron emission tomography (PET) studies using [11 C]ABP688 show reduced metabotropic glutamate receptor type 5 (mGluR5) allosteric binding site availability in the epileptogenic hippocampus of mesial temporal lobe epilepsy (MTLE) patients. However, the link between mGluR5 abnormalities and postsurgical outcomes remains unclear. Here, we test whether reduced PET [11 C]ABP688 binding in cornu ammonis (CA) sectors more vulnerable to glutamatergic excitotoxicity relates to surgical outcomes. METHODS We obtained magnetic resonance imaging (MRI) and [11 C]ABP688-PET from 31 unilateral MTLE patients and 30 healthy controls. MRI hippocampal subfields were segmented using FreeSurfer. To respect the lower PET special resolution, MRI-derived anatomical subfields were combined into CA1-3, CA4/dentate gyrus, and Subiculum. Partial volume corrected [11 C]ABP688 nondisplaceable binding potential (BPND ) values were averaged across each subfield, and Z-scores were calculated. Subfield [11 C]ABP688-BPND was compared between seizure-free and non-seizure-free patients. In addition, we also assessed subfield volumes and [18 F]fluorodeoxyglucose (FDG) uptake in each clinical group. RESULTS MTLE [11 C]ABP688-BPND was reduced in ipsilateral (epileptogenic) CA1-3 and CA4/dentate-gyrus (p < .001, 95% confidence interval [CI] = .29-.51) compared to controls, with no difference in Subiculum. [11 C]ABP688-BPND and subfield volumes were compared between seizure-free (Engel IA, n = 13) and non-seizure-free patients (Engel IC-III, n = 10). In ipsilateral CA1-3 only, [11 C]ABP688-BPND was lower in seizure-free patients than in non-seizure-free patients (p = .012, 95% CI = 1.46-11.0) independently of volume. A subset analysis of 12 patients with [11 C]ABP688-PET+[18 F]FDG-PET showed no between-group significant difference in [18 F]FDG uptake, whereas CA1-3 [11 C]ABP688-BPND remained significantly lower in the seven of 12 seizure-free patients (p = .03, 95% CI = -3.13 to -.21). SIGNIFICANCE Reduced mGluR5 allosteric site availability in hippocampal CA1-3, measured in vivo by [11 C]ABP688-PET, is associated with postsurgery seizure freedom independent of atrophy or hypometabolism. Information derived from hippocampal CA1-3 [11 C]ABP688-PET is a promising imaging biomarker potentially impactful in surgical decisions for MRI-negative/PET-negative MTLE patients.
Collapse
Affiliation(s)
- Jack Lam
- Department of Neurology and Neurosurgery, Montréal Neurological Institute, McGill University, Montréal, Québec, Canada
| | - Jonathan M DuBois
- Department of Neurology and Neurosurgery, Montréal Neurological Institute, McGill University, Montréal, Québec, Canada
| | - Jared Rowley
- Translational Neuroimaging Laboratory, McGill University, Montréal, Québec, Canada
| | - Olivier G Rousset
- Division of Nuclear Medicine and Molecular Imaging, Johns Hopkins University, Baltimore, Maryland, USA
| | - Karina A González-Otárula
- Department of Neurology and Neurosurgery, Montréal Neurological Institute, McGill University, Montréal, Québec, Canada
| | - Jean-Paul Soucy
- Department of Neurology and Neurosurgery, Montréal Neurological Institute, McGill University, Montréal, Québec, Canada.,PET Unit, McConnell Brain Imaging Centre, McGill University, Montréal, Québec, Canada
| | - Gassan Massarweh
- PET Unit, McConnell Brain Imaging Centre, McGill University, Montréal, Québec, Canada
| | - Jeffery A Hall
- Department of Neurology and Neurosurgery, Montréal Neurological Institute, McGill University, Montréal, Québec, Canada
| | - Marie-Christine Guiot
- Department of Pathology, Montreal Neurological Institute, McGill University, Montréal, Québec, Canada
| | - Maria Zimmermann
- Department of Neurology and Neurosurgery, Montréal Neurological Institute, McGill University, Montréal, Québec, Canada
| | - Luciano Minuzzi
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, Ontario, Canada
| | - Pedro Rosa-Neto
- Department of Neurology and Neurosurgery, Montréal Neurological Institute, McGill University, Montréal, Québec, Canada.,Translational Neuroimaging Laboratory, McGill University, Montréal, Québec, Canada.,PET Unit, McConnell Brain Imaging Centre, McGill University, Montréal, Québec, Canada
| | - Eliane Kobayashi
- Department of Neurology and Neurosurgery, Montréal Neurological Institute, McGill University, Montréal, Québec, Canada
| |
Collapse
|
7
|
Valian N, Sorayya M, Asadi S, Sherafati F, Ershad A, Savaheli S, Ahmadiani A. Preconditioning by ultra-low dose of tramadol reduces the severity of tramadol-induced seizure: Contribution of glutamate receptors. Biomed Pharmacother 2021; 133:111031. [PMID: 33249277 DOI: 10.1016/j.biopha.2020.111031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 11/07/2020] [Accepted: 11/15/2020] [Indexed: 12/15/2022] Open
Abstract
Tramadol, a weak agonist of mu-opioid receptors, causes seizure via several mechanisms. Preconditioning has been purposed to reduce the epileptic seizures in animal models of epilepsy. The preconditioning effect of tramadol on seizure is not studied yet. This study was designed to evaluate the preconditioning effect of ultra-low dose of tramadol on the seizures induced by tramadol at high dose. Furthermore, regarding the critical role of glutamate signaling in the pathogenesis of epilepsy, the effect of preconditioning on some glutamate signaling elements was also examined. Male Wistar rats received tramadol (2 mg/kg, i.p) or normal saline (1 mL/kg, i.p) in preconditioning and control groups, respectively. After 4 days, the challenging tramadol dose (150 mg/kg) was injected to all rats. Epileptic behaviors were recorded during 50 min. The expression of Norbin (as a regulator of metabotropic glutamate receptor 5), Calponin3 (as a regulator of excitatory synaptic markers), NR1 (NMDA receptor subunit 1) and GluR1 (AMPA receptor subunit 1) was measured in hippocampus, prefrontal cortex (PFC) and amygdala. Preconditioning decreased the number and duration of tremors and tonic-clonic seizures. Norbin, Calponin3, NR1 and GluR1 expression were decreased in hippocampus, and preconditioning had no effect on them. In contrast, it increased Norbin expression in PFC and amygdala, and attenuated NR1 and GluR1 upregulation following tramadol at high dose. These findings indicated that preconditioning by ultra-low dose of tramadol protected the animals against seizures following high dose of tramadol mediated, at least in part, by Norbin up regulation, and NR1 and GluR1 down regulation.
Collapse
Affiliation(s)
- Neda Valian
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Marziyeh Sorayya
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sareh Asadi
- Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Sherafati
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Ershad
- Department of Clinical Toxicology, Loghman Hakim Educational Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sara Savaheli
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Abolhassan Ahmadiani
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
8
|
Marafi D, Mitani T, Isikay S, Hertecant J, Almannai M, Manickam K, Abou Jamra R, El-Hattab AW, Rajah J, Fatih JM, Du H, Karaca E, Bayram Y, Punetha J, Rosenfeld JA, Jhangiani SN, Boerwinkle E, Akdemir ZC, Erdin S, Hunter JV, Gibbs RA, Pehlivan D, Posey JE, Lupski JR. Biallelic GRM7 variants cause epilepsy, microcephaly, and cerebral atrophy. Ann Clin Transl Neurol 2020; 7:610-627. [PMID: 32286009 PMCID: PMC7261753 DOI: 10.1002/acn3.51003] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 02/10/2020] [Accepted: 02/11/2020] [Indexed: 01/29/2023] Open
Abstract
Objective Defects in ion channels and neurotransmitter receptors are implicated in developmental and epileptic encephalopathy (DEE). Metabotropic glutamate receptor 7 (mGluR7), encoded by GRM7, is a presynaptic G‐protein‐coupled glutamate receptor critical for synaptic transmission. We previously proposed GRM7 as a candidate disease gene in two families with neurodevelopmental disorders (NDDs). One additional family has been published since. Here, we describe three additional families with GRM7 biallelic variants and deeply characterize the associated clinical neurological and electrophysiological phenotype and molecular data in 11 affected individuals from six unrelated families. Methods Exome sequencing and family‐based rare variant analyses on a cohort of 220 consanguineous families with NDDs revealed three families with GRM7 biallelic variants; three additional families were identified through literature search and collaboration with a clinical molecular laboratory. Results We compared the observed clinical features and variants of 11 affected individuals from the six unrelated families. Identified novel deleterious variants included two homozygous missense variants (c.2671G>A:p.Glu891Lys and c.1973G>A:p.Arg685Gln) and one homozygous stop‐gain variant (c.1975C>T:p.Arg659Ter). Developmental delay, neonatal‐ or infantile‐onset epilepsy, and microcephaly were universal. Three individuals had hypothalamic–pituitary–axis dysfunction without pituitary structural abnormality. Neuroimaging showed cerebral atrophy and hypomyelination in a majority of cases. Two siblings demonstrated progressive loss of myelination by 2 years in both and an acquired microcephaly pattern in one. Five individuals died in early or late childhood. Conclusion Detailed clinical characterization of 11 individuals from six unrelated families demonstrates that rare biallelic GRM7 pathogenic variants can cause DEEs, microcephaly, hypomyelination, and cerebral atrophy.
Collapse
Affiliation(s)
- Dana Marafi
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, 77030.,Department of Pediatrics, Faculty of Medicine, Kuwait University, P.O. Box 24923, 13110, Safat, Kuwait
| | - Tadahiro Mitani
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, 77030
| | - Sedat Isikay
- Department of Physiotherapy and Rehabilitation, School of Health Sciences, Hasan Kalyoncu University, Gaziantep, 27000, Turkey
| | - Jozef Hertecant
- Pediatric Metabolic and Genetics Division, Tawam Hospital, Al Ain, Abu Dhabi, United Arab Emirates
| | - Mohammed Almannai
- Section of Medical Genetics, Children's Hospital, King Fahad Medical City, Riyadh, 11525, Saudi Arabia
| | - Kandamurugu Manickam
- Division of Genetic and Genomic Medicine, Nationwide Children's Hospital, Columbus, Ohio
| | - Rami Abou Jamra
- Institute of Human Genetics, University Medical Center Leipzig, 04103, Leipzig, Germany
| | - Ayman W El-Hattab
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Jaishen Rajah
- Sheikh Khalifa Medical City (SKMC), P.O. Box: 51900, Abu Dhabi, United Arab Emirates
| | - Jawid M Fatih
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, 77030
| | - Haowei Du
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, 77030
| | - Ender Karaca
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, 77030
| | - Yavuz Bayram
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, 77030
| | - Jaya Punetha
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, 77030
| | - Jill A Rosenfeld
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, 77030
| | - Shalini N Jhangiani
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, 77030
| | - Eric Boerwinkle
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, 77030.,Human Genetics Center, University of Texas Health Science Center at Houston, Houston, Texas
| | - Zeynep C Akdemir
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, 77030
| | - Serkan Erdin
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Jill V Hunter
- Texas Children's Hospital, Houston, Texas, 77030.,Department of Radiology, Baylor College of Medicine, Houston, Texas, 77030
| | - Richard A Gibbs
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, 77030.,Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, 77030
| | - Davut Pehlivan
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, 77030.,Texas Children's Hospital, Houston, Texas, 77030.,Section of Pediatric Neurology and Developmental Neuroscience, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, 77030
| | - Jennifer E Posey
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, 77030
| | - James R Lupski
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, 77030.,Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, 77030.,Texas Children's Hospital, Houston, Texas, 77030.,Department of Pediatrics, Baylor College of Medicine, Houston, Texas, 77030
| |
Collapse
|
9
|
Alese OO, Rakgantsho C, Mkhize NV, Zulu S, Mabandla MV. Prolonged febrile seizure history exacerbates seizure severity in a pentylenetetrazole rat model of epilepsy. Brain Res Bull 2019; 155:137-144. [PMID: 31837458 DOI: 10.1016/j.brainresbull.2019.11.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 11/07/2019] [Accepted: 11/30/2019] [Indexed: 12/29/2022]
Abstract
Epilepsy is a debilitating neurological illness that affects all aspect of an individual life. Despite advancement in research there is little reduction in the incidence of this disease. Prolonged febrile seizure (PFS) has been linked to epilepsy however, the pathophysiology of this is still not clear. We therefore looked at the effect of PFS on the development of epilepsy in a pentylenetetrazole (PTZ) rat model of epilepsy. A total of 42 male Sprague-Dawley rats were used for the experiment. On post-natal day (PND) 14, PFS was induced in 14 rats. This was followed by the induction of epilepsy in the 14 PFS animal and 14 animals from the remaining 28 rats by an initial injection of PTZ at a dose of 60 mg/kg on day one followed by 35 mg/kg on alternate day until kindle. We looked at the effect of PFS on the onset and the stage of convulsion at kindle. We also observed it effect on the hippocampal glial fibrillary acidic protein (GFAP), synaptophysin and metabotropic glutamate receptor 3 (mGluR3) expression measured with immunofluorescence, LI Cor Tissue florescence and immunohistochemistry respectively. Our study showed that PFS reduced seizure threshold by decreasing the time it took animals to kindle and also increased the stage of convulsion. The hippocampal GFAP, synaptophysin and mGluR3 expressions where upregulated in PTZ rats with PFS history when compared to PTZ rats alone.These findings indicated that PFS may increase the severity of epilepsy and alter brain expression of GFAP, synaptophysin and mGluR3 proteins.
Collapse
Affiliation(s)
- Oluwole Ojo Alese
- Department of Human Physiology, College of Health Sciences, University of Kwazulu-Natal, South Africa.
| | - Cleopatra Rakgantsho
- Department of Human Physiology, College of Health Sciences, University of Kwazulu-Natal, South Africa
| | - Nombuso V Mkhize
- Department of Human Physiology, College of Health Sciences, University of Kwazulu-Natal, South Africa
| | - Simo Zulu
- Department of Human Physiology, College of Health Sciences, University of Kwazulu-Natal, South Africa
| | - Musa V Mabandla
- Department of Human Physiology, College of Health Sciences, University of Kwazulu-Natal, South Africa
| |
Collapse
|
10
|
Celli R, Santolini I, Van Luijtelaar G, Ngomba RT, Bruno V, Nicoletti F. Targeting metabotropic glutamate receptors in the treatment of epilepsy: rationale and current status. Expert Opin Ther Targets 2019; 23:341-351. [PMID: 30801204 DOI: 10.1080/14728222.2019.1586885] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
INTRODUCTION Several drugs targeting the GABAergic system are used in the treatment of epilepsy, but only one drug targeting glutamate receptors is on the market. This is surprising because an imbalance between excitatory and inhibitory neurotransmission lies at the core of the pathophysiology of epilepsy. One possible explanation is that drug development has been directed towards the synthesis of molecules that inhibit the activity of ionotropic glutamate receptors. These receptors mediate fast excitatory synaptic transmission in the central nervous system (CNS) and their blockade may cause severe adverse effects such as sedation, cognitive impairment, and psychotomimetic effects. Metabotropic glutamate (mGlu) receptors are more promising drug targets because these receptors modulate synaptic transmission rather than mediate it. Areas covered: We review the current evidence that links mGlu receptor subtypes to the pathophysiology and experimental treatment of convulsive and absence seizures. Expert opinion: While mGlu5 receptor negative allosteric modulators have the potential to be protective against convulsive seizures and hyperactivity-induced neurodegeneration, drugs that enhance mGlu5 and mGlu7 receptor function may have beneficial effects in the treatment of absence epilepsy. Evidence related to the other mGlu receptor subtypes is more fragmentary; further investigations are required for an improved understanding of their role in the generation and propagation of seizures.
Collapse
Affiliation(s)
| | | | | | | | - Valeria Bruno
- a IRCCS NEUROMED , Pozzilli , Italy.,d Departments of Physiology and Pharmacology , University Sapienza , Rome , Italy
| | - Ferdinando Nicoletti
- a IRCCS NEUROMED , Pozzilli , Italy.,d Departments of Physiology and Pharmacology , University Sapienza , Rome , Italy
| |
Collapse
|
11
|
Lam J, DuBois JM, Rowley J, González-Otárula KA, Soucy JP, Massarweh G, Hall JA, Guiot MC, Rosa-Neto P, Kobayashi E. In vivo metabotropic glutamate receptor type 5 abnormalities localize the epileptogenic zone in mesial temporal lobe epilepsy. Ann Neurol 2019; 85:218-228. [PMID: 30597619 DOI: 10.1002/ana.25404] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 12/22/2018] [Accepted: 12/24/2018] [Indexed: 01/31/2023]
Abstract
OBJECTIVE Surgical specimens from patients with mesial temporal lobe epilepsy (MTLE) show abnormalities in tissue concentrations of metabotropic glutamate receptor type 5 (mGluR5). To clarify whether these abnormalities are specific to the epileptogenic zone (EZ), we characterized in vivo whole-brain mGluR5 availability in MTLE patients using positron emission tomography (PET) and [11 C]ABP688, a radioligand that binds specifically to the mGluR5 allosteric site. METHODS Thirty-one unilateral MTLE patients and 30 healthy controls underwent [11 C]ABP688 PET. We compared partial volume corrected [11 C]ABP688 nondisplaceable binding potentials (BPND ) between groups using region-of-interest and whole-brain voxelwise analyses. [18 F]Fluorodeoxyglucose (FDG) PET was acquired in 15 patients, for whom we calculated asymmetry indices of [11 C]ABP688 BPND and [18 F]FDG uptake to compare lateralization and localization differences. RESULTS [11 C]ABP688 BPND was focally reduced in the epileptogenic hippocampal head and amygdala (p < 0.001). Patients with hippocampal atrophy showed more extensive abnormalities, including the ipsilateral temporal neocortex (p = 0.006). [11 C]ABP688 BPND showed interhemispheric differences of higher magnitude and discriminated the epileptogenic structures more accurately when compared to [18 F]FDG uptake, which showed more widespread hypometabolism. Among 23 of 25 operated patients with >1 year of follow-up, 13 were seizure-free (Engel Ia) and showed significantly lower [11 C]ABP688 BPND in the ipsilateral entorhinal cortex. INTERPRETATION [11 C]ABP688 PET provides a focal biomarker for the EZ in MTLE with higher spatial accuracy compared to [18 F]FDG PET. Focally reduced mGluR5 availability in the EZ might reflect receptor internalization or conformational changes in response to excessive extracellular glutamate, supporting a potential role for mGluR5 as therapeutic target in human MTLE. Ann Neurol 2019; 1-11 ANN NEUROL 2019;85:218-228.
Collapse
Affiliation(s)
- Jack Lam
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Jonathan M DuBois
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Jared Rowley
- Translational Neuroimaging Laboratory, McGill University, Montreal, Quebec, Canada
| | - Karina A González-Otárula
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Jean-Paul Soucy
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada.,PET Unit, McConnell Brain Imaging Centre, McGill University, Montreal, Quebec, Canada
| | - Gassan Massarweh
- PET Unit, McConnell Brain Imaging Centre, McGill University, Montreal, Quebec, Canada
| | - Jeffery A Hall
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Marie-Christine Guiot
- Department of Pathology, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Pedro Rosa-Neto
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada.,Translational Neuroimaging Laboratory, McGill University, Montreal, Quebec, Canada.,PET Unit, McConnell Brain Imaging Centre, McGill University, Montreal, Quebec, Canada
| | - Eliane Kobayashi
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
12
|
Hussain T, Kil H, Hattiangady B, Lee J, Kodali M, Shuai B, Attaluri S, Takata Y, Shen J, Abba MC, Shetty AK, Aldaz CM. Wwox deletion leads to reduced GABA-ergic inhibitory interneuron numbers and activation of microglia and astrocytes in mouse hippocampus. Neurobiol Dis 2018; 121:163-176. [PMID: 30290271 DOI: 10.1016/j.nbd.2018.09.026] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 09/18/2018] [Accepted: 09/30/2018] [Indexed: 02/07/2023] Open
Abstract
The association of WW domain-containing oxidoreductase WWOX gene loss of function with central nervous system (CNS) related pathologies is well documented. These include spinocerebellar ataxia, epilepsy and mental retardation (SCAR12, OMIM: 614322) and early infantile epileptic encephalopathy (EIEE28, OMIM: 616211) syndromes. However, there is complete lack of understanding of the pathophysiological mechanisms at play. In this study, using a Wwox knockout (Wwox KO) mouse model (2 weeks old, both sexes) and stereological studies we observe that Wwox deletion leads to a significant reduction in the number of hippocampal GABA-ergic (γ-aminobutyric acid) interneurons. Wwox KO mice displayed significantly reduced numbers of calcium-binding protein parvalbumin (PV) and neuropeptide Y (NPY) expressing interneurons in different subfields of the hippocampus in comparison to Wwox wild-type (WT) mice. We also detected decreased levels of Glutamic Acid Decarboxylase protein isoforms GAD65/67 expression in Wwox null hippocampi suggesting lower levels of GABA synthesis. In addition, Wwox deficiency was associated with signs of neuroinflammation such as evidence of activated microglia, astrogliosis, and overexpression of inflammatory cytokines Tnf-a and Il6. We also performed comparative transcriptome-wide expression analyses of neural stem cells grown as neurospheres from hippocampi of Wwox KO and WT mice thus identifying 283 genes significantly dysregulated in their expression. Functional annotation of transcriptome profiling differences identified 'neurological disease' and 'CNS development related functions' to be significantly enriched. Several epilepsy-related genes were found differentially expressed in Wwox KO neurospheres. This study provides the first genotype-phenotype observations as well as potential mechanistic clues associated with Wwox loss of function in the brain.
Collapse
Affiliation(s)
- Tabish Hussain
- Department of Epigenetics and Molecular Carcinogenesis, Science Park, The University of Texas MD Anderson Cancer Center, Smithville, TX, United States
| | - Hyunsuk Kil
- Department of Epigenetics and Molecular Carcinogenesis, Science Park, The University of Texas MD Anderson Cancer Center, Smithville, TX, United States
| | - Bharathi Hattiangady
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, Temple and College Station, TX, United States; Research Service, Olin E. Teague Veterans' Medical Center, CTVHCS, Temple, TX, United States
| | - Jaeho Lee
- Department of Epigenetics and Molecular Carcinogenesis, Science Park, The University of Texas MD Anderson Cancer Center, Smithville, TX, United States
| | - Maheedhar Kodali
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, Temple and College Station, TX, United States; Research Service, Olin E. Teague Veterans' Medical Center, CTVHCS, Temple, TX, United States
| | - Bing Shuai
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, Temple and College Station, TX, United States; Research Service, Olin E. Teague Veterans' Medical Center, CTVHCS, Temple, TX, United States
| | - Sahithi Attaluri
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, Temple and College Station, TX, United States; Research Service, Olin E. Teague Veterans' Medical Center, CTVHCS, Temple, TX, United States
| | - Yoko Takata
- Department of Epigenetics and Molecular Carcinogenesis, Science Park, The University of Texas MD Anderson Cancer Center, Smithville, TX, United States
| | - Jianjun Shen
- Department of Epigenetics and Molecular Carcinogenesis, Science Park, The University of Texas MD Anderson Cancer Center, Smithville, TX, United States
| | - Martin C Abba
- CINIBA, School of Medicine, UNLP, La Plata, Argentina
| | - Ashok K Shetty
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, Temple and College Station, TX, United States; Research Service, Olin E. Teague Veterans' Medical Center, CTVHCS, Temple, TX, United States
| | - C Marcelo Aldaz
- Department of Epigenetics and Molecular Carcinogenesis, Science Park, The University of Texas MD Anderson Cancer Center, Smithville, TX, United States.
| |
Collapse
|
13
|
Vergouts M, Doyen PJ, Peeters M, Opsomer R, Hermans E. Constitutive downregulation protein kinase C epsilon in hSOD1 G93A astrocytes influences mGluR5 signaling and the regulation of glutamate uptake. Glia 2017; 66:749-761. [PMID: 29266405 DOI: 10.1002/glia.23279] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 11/19/2017] [Accepted: 11/24/2017] [Indexed: 01/06/2023]
Abstract
Accumulating evidence indicates that motor neuron degeneration in amyotrophic lateral sclerosis (ALS) is a non-cell-autonomous process and that impaired glutamate clearance by astrocytes, leading to excitotoxicity, could participate in progression of the disease. In astrocytes derived from an animal model of ALS (hSOD1G93A rats), activation of type 5 metabotropic glutamate receptor (mGluR5) fails to increase glutamate uptake, impeding a putative dynamic neuroprotective mechanism involving astrocytes. Using astrocyte cultures from hSOD1G93A rats, we have demonstrated that the typical Ca2+ oscillations associated with mGluR5 activation were reduced, and that the majority of cells responded with a sustained elevation of intracellular Ca2+ concentration. Since the expression of protein kinase C epsilon isoform (PKCɛ) has been found to be considerably reduced in astrocytes from hSOD1G93A rats, the consequences of manipulating its activity and expression on mGluR5 signaling and on the regulation of glutamate uptake have been examined. Increasing PKCɛ expression was found to restore Ca2+ oscillations induced by mGluR5 activation in hSOD1G93A -expressing astrocytes. This was also associated with an increase in glutamate uptake capacity in response to mGluR5 activation. Conversely, reducing PKCɛ expression in astrocytes from wild-type animals with specific PKCɛ-shRNAs was found to alter the mGluR5 associated oscillatory signaling profile, and consistently reduced the regulation of the glutamate uptake-mediated by mGluR5 activation. These results suggest that PKCɛ is required to generate Ca2+ oscillations following mGluR5 activation, which support the regulation of astrocytic glutamate uptake. Reduced expression of astrocytic PKCɛ could impair this neuroprotective process and participate in the progression of ALS.
Collapse
Affiliation(s)
- Maxime Vergouts
- Institute of Neuroscience, Université catholique de Louvain, Avenue Hippocrate B1.54.10, Brussels, 1200, Belgium
| | - Pierre J Doyen
- Institute of Neuroscience, Université catholique de Louvain, Avenue Hippocrate B1.54.10, Brussels, 1200, Belgium
| | - Michael Peeters
- De Duve Institute, Université catholique de Louvain, Avenue Hippocrate VIRO B1.74.07, Brussels, 1200, Belgium
| | - Remi Opsomer
- Alzheimer Dementia Group, Institute of Neuroscience, Université catholique de Louvain, Avenue Mounier B1.53.02, Brussels, 1200, Belgium
| | - Emmanuel Hermans
- Institute of Neuroscience, Université catholique de Louvain, Avenue Hippocrate B1.54.10, Brussels, 1200, Belgium
| |
Collapse
|
14
|
DePaula-Silva AB, Hanak TJ, Libbey JE, Fujinami RS. Theiler's murine encephalomyelitis virus infection of SJL/J and C57BL/6J mice: Models for multiple sclerosis and epilepsy. J Neuroimmunol 2017; 308:30-42. [PMID: 28237622 DOI: 10.1016/j.jneuroim.2017.02.012] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Revised: 02/01/2017] [Accepted: 02/10/2017] [Indexed: 10/20/2022]
Abstract
Mouse models are great tools to study the mechanisms of disease development. Theiler's murine encephalomyelitis virus is used in two distinct viral infection mouse models to study the human diseases multiple sclerosis (MS) and epilepsy. Intracerebral (i.c.) infection of the SJL/J mouse strain results in persistent viral infection of the central nervous system and a MS-like disease, while i.c. infection of the C57BL/6J mouse strain results in acute seizures and epilepsy. Our understanding of how the immune system contributes to the development of two disparate diseases caused by the same virus is presented.
Collapse
Affiliation(s)
- Ana Beatriz DePaula-Silva
- Department of Pathology, University of Utah School of Medicine, 15 North Medical Drive East, 2600 EEJMRB, Salt Lake City, UT 84112, USA
| | - Tyler J Hanak
- Department of Pathology, University of Utah School of Medicine, 15 North Medical Drive East, 2600 EEJMRB, Salt Lake City, UT 84112, USA
| | - Jane E Libbey
- Department of Pathology, University of Utah School of Medicine, 15 North Medical Drive East, 2600 EEJMRB, Salt Lake City, UT 84112, USA
| | - Robert S Fujinami
- Department of Pathology, University of Utah School of Medicine, 15 North Medical Drive East, 2600 EEJMRB, Salt Lake City, UT 84112, USA.
| |
Collapse
|
15
|
Malik R, Mehta P, Srivastava S, Choudhary BS, Sharma M. Structure-based screening, ADMET profiling, and molecular dynamic studies on mGlu2 receptor for identification of newer antiepileptic agents. J Biomol Struct Dyn 2016; 35:3433-3448. [DOI: 10.1080/07391102.2016.1257440] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- Ruchi Malik
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, NH-8, Bandarsindri, Kishangarh, Ajmer, Rajasthan 305817, India
| | - Pakhuri Mehta
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, NH-8, Bandarsindri, Kishangarh, Ajmer, Rajasthan 305817, India
| | - Shubham Srivastava
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, NH-8, Bandarsindri, Kishangarh, Ajmer, Rajasthan 305817, India
| | - Bhanwar Singh Choudhary
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, NH-8, Bandarsindri, Kishangarh, Ajmer, Rajasthan 305817, India
| | - Manish Sharma
- School of Pharmacy, Maharishi Markandeshwar University, Sadopur, Ambala, Haryana 134007, India
| |
Collapse
|
16
|
Proton magnetic resonance spectroscopy in juvenile myoclonic epilepsy: A systematic review and meta-analysis. Epilepsy Res 2016; 121:33-8. [DOI: 10.1016/j.eplepsyres.2016.01.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Revised: 12/07/2015] [Accepted: 01/24/2016] [Indexed: 12/30/2022]
|
17
|
Rosa-Falero C, Torres-Rodríguez S, Jordán C, Licier R, Santiago Y, Toledo Z, Santiago M, Serrano K, Sosa J, Ortiz JG. Citrus aurantium increases seizure latency to PTZ induced seizures in zebrafish thru NMDA and mGluR's I and II. Front Pharmacol 2015; 5:284. [PMID: 25762932 PMCID: PMC4327740 DOI: 10.3389/fphar.2014.00284] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 12/03/2014] [Indexed: 12/02/2022] Open
Abstract
Epilepsy is a serious neurological condition and pharmacotherapy is not effective for all patients and causes serious adverse effects and pharmacokinetic and pharmacodynamic interactions. Natural products and ethnobotanical resources can help develop new therapeutic options for conditions like epilepsy. In Puerto Rico, ethnobotanical resources highlight the anxiolytic properties of a tea like preparation made from the leaves of the Citrus aurantium tree or bitter orange. Studies performed with essential oils from the peel of the fruit have shown to increase seizure latency to pentylenetetrazole (PTZ) and maximal electroshock seizure in mice. We characterized the extract composition, and used a model of PTZ induces seizures in the zebrafish and a receptor-ligand binding assay to determine if this preparation has anticonvulsant properties and its mechanism of action. We determined that the aqueous extract made from the leaves of the C. aurantium tree contains hesperidin, neohesperidin, and neohesperidin dihydrochalcone. Using our zebrafish model, we determined that exposure to the C. aurantium 28 mg/mL extract in aquarium water increases seizure latency by 119% compared to controls. We ruled out a mechanism involving GABAA receptors using the selective antagonist gabazine. We used two approaches to study the role of glutamate in the mechanism of the C. aurantium extract. The ligand binding assay revealed C. aurantium extracts at concentrations of 0.42 to 5.6 mg/mL significantly reduced [3H]Glu binding indicating an interaction with glutamate receptors, in particular with NMDA receptors and mGluR II. This interaction was confirmed with our animal model using selective receptor antagonists and we identified an interaction with mGluR I, not observed in the ligand binding experiment. These study provide evidence of the anticonvulsant properties of the aqueous extract made from the leaves of the C. aurantium tree and a mechanism involving NMDA and mGluR's I and II.
Collapse
Affiliation(s)
- Coral Rosa-Falero
- Neuropharmacology Laboratory, Pharmacology and Toxicology Department, University of Puerto Rico-Medical Sciences Campus San Juan, PR, USA
| | - Stephanie Torres-Rodríguez
- Neuropharmacology Laboratory, Pharmacology and Toxicology Department, University of Puerto Rico-Medical Sciences Campus San Juan, PR, USA
| | - Claudia Jordán
- Neuropharmacology Laboratory, Pharmacology and Toxicology Department, University of Puerto Rico-Medical Sciences Campus San Juan, PR, USA
| | - Rígel Licier
- Neuropharmacology Laboratory, Pharmacology and Toxicology Department, University of Puerto Rico-Medical Sciences Campus San Juan, PR, USA
| | - Yolimar Santiago
- Neuropharmacology Laboratory, Pharmacology and Toxicology Department, University of Puerto Rico-Medical Sciences Campus San Juan, PR, USA
| | - Zuleyma Toledo
- Neuropharmacology Laboratory, Pharmacology and Toxicology Department, University of Puerto Rico-Medical Sciences Campus San Juan, PR, USA
| | - Marely Santiago
- Neuropharmacology Laboratory, Pharmacology and Toxicology Department, University of Puerto Rico-Medical Sciences Campus San Juan, PR, USA
| | - Kiara Serrano
- Neuropharmacology Laboratory, Pharmacology and Toxicology Department, University of Puerto Rico-Medical Sciences Campus San Juan, PR, USA
| | - Jeffrey Sosa
- School of Science and Technology, Universidad del Este Carolina, PR, USA
| | - José G Ortiz
- Neuropharmacology Laboratory, Pharmacology and Toxicology Department, University of Puerto Rico-Medical Sciences Campus San Juan, PR, USA
| |
Collapse
|
18
|
Abstract
Metabotropic glutamate receptors (mGluRs) are found throughout thalamus and cortex and are clearly important to circuit behavior in both structures, and so considering only participation of ionotropic glutamate receptors (e.g., [R,S]-α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid [AMPA] and N-methyl-d-aspartate receptors [NMDA] receptors) in glutamatergic processing would be an unfortunate oversimplification. These mGluRs are found both postsynaptically, on target cells of glutamatergic afferents, and presynaptically, on various synaptic terminals themselves, and when activated, they produce prolonged effects lasting at least hundreds of msec to several sec and perhaps longer. Two main types exist: activation of group I mGluRs causes postsynaptic depolarization, and group II, hyperpolarization. Both types are implicated in synaptic plasticity, both short term and long term. Their evident importance in functioning of thalamus and cortex makes it critical to develop a better understanding of how these receptors are normally activated, especially because they also seem implicated in a wide range of neurological and cognitive pathologies.
Collapse
|
19
|
In vivo imaging of mGluR5 changes during epileptogenesis using [11C]ABP688 PET in pilocarpine-induced epilepsy rat model. PLoS One 2014; 9:e92765. [PMID: 24663806 PMCID: PMC3963947 DOI: 10.1371/journal.pone.0092765] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 02/26/2014] [Indexed: 11/28/2022] Open
Abstract
Introduction Metabotropic glutamate receptor 5 (mGluR5) that regulates glutamatergic neurotransmission contributes to pathophysiology of epilepsy. In this study, we monitored the changes of mGluR5 in vivo using [11C]ABP688 PET during the epileptogenesis in a pilocarpine-induced epilepsy rat model. Methods In vivo mGluR5 images were acquired using [11C]ABP688 microPET/CT in pilocarpine-induced chronic epilepsy rat models and controls. We also acquired microPET/CT at acute, subacute as well as chronic periods after status epilepticus. Non-displaceable binding potential (BPND) of [11C]ABP688 was calculated using simplified reference tissue model in a voxel-based manner. mGluR5 BPND of the rat brains of epilepsy models and controls were compared. Results Status epilepticus developed after pilocarpine administration and was followed by recurrent spontaneous seizures for more than 3 weeks. In chronic epilepsy rat model, BPND in hippocampus and amygdala was reduced on a voxel-based analysis. Temporal changes of mGluR5 BPND was also found. In acute period after status epilepticus, mGluR5 BPND was reduced in the whole brain. BPND of caudate-putamen was restored in subacute period, while BPND of the rest of the brain was still lower. In chronic period, global BPND was normalized except in hippocampus and amygdala. Conclusions In vivo imaging of mGluR5 using [11C]ABP688 microPET/CT could successfully reveal the regional changes of mGluR5 binding potential of the rat brain in a pilocarpine-induced epilepsy model. The temporal and spatial changes in mGluR5 availability suggest [11C]ABP688 PET imaging in epilepsy provide abnormal glutamatergic network during epileptogenesis.
Collapse
|
20
|
Michalovicz LT, Konat GW. Peripherally restricted acute phase response to a viral mimic alters hippocampal gene expression. Metab Brain Dis 2014; 29:75-86. [PMID: 24363211 PMCID: PMC4343041 DOI: 10.1007/s11011-013-9471-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Accepted: 12/13/2013] [Indexed: 11/26/2022]
Abstract
We have previously shown that peripherally restricted acute phase response (APR) elicited by intraperitoneal (i.p.) injection of a viral mimic, polyinosinic-polycytidylic acid (PIC), renders the brain hypersusceptible to excitotoxic insult as seen from profoundly exacerbated kainic acid (KA)-induced seizures. In the present study, we found that this hypersusceptibility was protracted for up to 72 h. RT-PCR profiling of hippocampal gene expression revealed rapid upregulation of 23 genes encoding cytokines, chemokines and chemokine receptors generally within 6 h after PIC challenge. The expression of most of these genes decreased by 24 h. However, two chemokine genes, i.e., Ccl19 and Cxcl13 genes, as well as two chemokine receptor genes, Ccr1 and Ccr7, remained upregulated for 72 h suggesting their possible involvement in the induction and sustenance of seizure hypersusceptibility. Also, 12 genes encoding proteins related to glutamatergic and GABAergic neurotransmission featured initial upregulation or downregulation followed by gradual normalization. The upregulation of the Gabrr3 gene remained upregulated at 72 h, congruent with its plausible role in the hypersusceptible phenotype. Moreover, the expression of ten microRNAs (miRs) was rapidly affected by PIC challenge, but their levels generally exhibited oscillating profiles over the time course of seizure hypersusceptibility. These results indicate that protracted seizure susceptibility following peripheral APR is associated with a robust polygenic response in the hippocampus.
Collapse
|
21
|
Harvey B, Siok C, Kiss T, Volfson D, Grimwood S, Shaffer C, Hajós M. Neurophysiological signals as potential translatable biomarkers for modulation of metabotropic glutamate 5 receptors. Neuropharmacology 2013; 75:19-30. [DOI: 10.1016/j.neuropharm.2013.06.020] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Revised: 06/12/2013] [Accepted: 06/17/2013] [Indexed: 01/17/2023]
|
22
|
Van Liefferinge J, Massie A, Portelli J, Di Giovanni G, Smolders I. Are vesicular neurotransmitter transporters potential treatment targets for temporal lobe epilepsy? Front Cell Neurosci 2013; 7:139. [PMID: 24009559 PMCID: PMC3757300 DOI: 10.3389/fncel.2013.00139] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Accepted: 08/11/2013] [Indexed: 12/18/2022] Open
Abstract
The vesicular neurotransmitter transporters (VNTs) are small proteins responsible for packing synaptic vesicles with neurotransmitters thereby determining the amount of neurotransmitter released per vesicle through fusion in both neurons and glial cells. Each transporter subtype was classically seen as a specific neuronal marker of the respective nerve cells containing that particular neurotransmitter or structurally related neurotransmitters. More recently, however, it has become apparent that common neurotransmitters can also act as co-transmitters, adding complexity to neurotransmitter release and suggesting intriguing roles for VNTs therein. We will first describe the current knowledge on vesicular glutamate transporters (VGLUT1/2/3), the vesicular excitatory amino acid transporter (VEAT), the vesicular nucleotide transporter (VNUT), vesicular monoamine transporters (VMAT1/2), the vesicular acetylcholine transporter (VAChT) and the vesicular γ-aminobutyric acid (GABA) transporter (VGAT) in the brain. We will focus on evidence regarding transgenic mice with disruptions in VNTs in different models of seizures and epilepsy. We will also describe the known alterations and reorganizations in the expression levels of these VNTs in rodent models for temporal lobe epilepsy (TLE) and in human tissue resected for epilepsy surgery. Finally, we will discuss perspectives on opportunities and challenges for VNTs as targets for possible future epilepsy therapies.
Collapse
|
23
|
Abstract
Benign epilepsy with centrotemporal spikes, early-onset childhood occipital epilepsy (Panayiotopoulos syndrome [PS]) and late-onset childhood occipital epilepsy (Gastaut type [LOCE-G]) are the principal pediatric focal epilepsy syndromes. They share major common characteristics: the appearance and resolution of electroclinical features are age related, there is a strong genetic predisposition, the clinical course is often mild with infrequent and easy to control seizures, interictal epileptiform activity is disproportionately abundant when compared with the clinical correlate, and tends to potentiate and generalize during sleep. In this review, we outline the relevant pathophysiology underlying this electroclinical spectrum. Then, the initial description of individual syndromes is followed by a summary of overlapping features and intermediate presentations that question the boundaries between these entities and provide the basis for the concept of a childhood seizure susceptibility syndrome. Additionally, we outline the main features of the related epileptic encephalopathies. An outlook on potential future lines of research completes this review.
Collapse
|
24
|
Eugenin EA, Basilio D, Sáez JC, Orellana JA, Raine CS, Bukauskas F, Bennett MVL, Berman JW. The role of gap junction channels during physiologic and pathologic conditions of the human central nervous system. J Neuroimmune Pharmacol 2012; 7:499-518. [PMID: 22438035 PMCID: PMC3638201 DOI: 10.1007/s11481-012-9352-5] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Accepted: 02/28/2012] [Indexed: 12/15/2022]
Abstract
Gap junctions (GJs) are expressed in most cell types of the nervous system, including neuronal stem cells, neurons, astrocytes, oligodendrocytes, cells of the blood brain barrier (endothelial cells and astrocytes) and under inflammatory conditions in microglia/macrophages. GJs connect cells by the docking of two hemichannels, one from each cell with each hemichannel being formed by 6 proteins named connexins (Cx). Unapposed hemichannels (uHC) also can be open on the surface of the cells allowing the release of different intracellular factors to the extracellular space. GJs provide a mechanism of cell-to-cell communication between adjacent cells that enables the direct exchange of intracellular messengers, such as calcium, nucleotides, IP(3), and diverse metabolites, as well as electrical signals that ultimately coordinate tissue homeostasis, proliferation, differentiation, metabolism, cell survival and death. Despite their essential functions in physiological conditions, relatively little is known about the role of GJs and uHC in human diseases, especially within the nervous system. The focus of this review is to summarize recent findings related to the role of GJs and uHC in physiologic and pathologic conditions of the central nervous system.
Collapse
Affiliation(s)
- Eliseo A Eugenin
- Department of Pathology, F727, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY 10461, USA.
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Lyddon R, Navarrett S, Dracheva S. Ionotropic glutamate receptor mRNA editing in the prefrontal cortex: no alterations in schizophrenia or bipolar disorder. J Psychiatry Neurosci 2012; 37:267-72. [PMID: 22469055 PMCID: PMC3380098 DOI: 10.1503/jpn.110107] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Dysfunction of glutamate neurotransmission has been implicated in the pathology of schizophrenia and bipolar disorder, and one mechanism by which glutamate signalling can be altered is through RNA editing of ionotropic glutamate receptors (iGluRs). The objectives of the present study were to evaluate the editing status of iGluRs in the human prefrontal cortex, determine whether iGluR editing is associated with psychiatric disease or suicide and evaluate a potential association between editing and alternative splicing in the α-amino-3-hydroxy-5-methylisoxazole-4-propionate (AMPA) iGluR subunits' pre-mRNA. METHODS We studied specimens derived from patients with antemortem diagnoses of bipolar disorder (n = 31) or schizophrenia (n = 34) who died by suicide or other causes, and from psychiatrically healthy controls (n = 34) who died from causes other than suicide. The RNA editing at all 8 editing sites within AMPA (GluA2-4 subunits) and kainate (GluK1-2 subunits) iGluRs was analyzed using a novel real-time quantitative polymerase chain reaction assay. RESULTS No differences in editing were detected among schizophrenia, bipolar or control groups or between suicide completers and patients who died from causes other than suicide. The editing efficiency was significantly higher in the flop than in the flip splicoforms of GluA3-4 AMPA subunits (all p < 0.001). LIMITATIONS The study is limited by the near absence of specimens from medicationnaive psychiatric patients and considerable variation in medication regimens among individuals, both of which introduce considerable uncertainty into the analysis of potential medication effects. CONCLUSION We found that iGluR RNA editing status was not associated with bipolar disorder, schizophrenia or suicide. Differences in editing between flip and flop splicoforms suggest that glutamate sensitivity of receptors containing GluA3 and/or GluA4 flop subunits is moderated as a result of increased editing.
Collapse
Affiliation(s)
| | | | - Stella Dracheva
- Correspondence to: S. Dracheva, Psychiatry Research (4F-02), Bronx VA Medical Center, 130 West Kingsbridge Rd., Bronx NY 10468;
| |
Collapse
|
26
|
Synergistic actions of metabotropic acetylcholine and glutamate receptors on the excitability of hippocampal CA1 pyramidal neurons. J Neurosci 2012; 32:6081-91. [PMID: 22553015 DOI: 10.1523/jneurosci.6519-11.2012] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
A variety of neurotransmitters are responsible for regulating neural activity during different behavioral states. Unique responses to combinations of neurotransmitters provide a powerful mechanism by which neural networks could be differentially activated during a broad range of behaviors. Here, we show, using whole-cell recordings in rat hippocampal slices, that group I metabotropic glutamate receptors (mGluRs) and muscarinic acetylcholine receptors (mAChRs) synergistically increase the excitability of hippocampal CA1 pyramidal neurons by converting the post-burst afterhyperpolarization to an afterdepolarization via a rapidly reversible upregulation of Ca(v)2.3 R-type calcium channels. Coactivation of mAChRs and mGluRs also induced a long-lasting enhancement of the responses mediated by each receptor type. These results suggest that cooperative signaling via mAChRs and group I mGluRs could provide a mechanism by which cognitive processes may be modulated by conjoint activation of two separate neurotransmitter systems.
Collapse
|
27
|
Hagerman R, Lauterborn J, Au J, Berry-Kravis E. Fragile X syndrome and targeted treatment trials. Results Probl Cell Differ 2012; 54:297-335. [PMID: 22009360 PMCID: PMC4114775 DOI: 10.1007/978-3-642-21649-7_17] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Work in recent years has revealed an abundance of possible new treatment targets for fragile X syndrome (FXS). The use of animal models, including the fragile X knockout mouse which manifests a phenotype very similar to FXS in humans, has resulted in great strides in this direction of research. The lack of Fragile X Mental Retardation Protein (FMRP) in FXS causes dysregulation and usually overexpression of a number of its target genes, which can cause imbalances of neurotransmission and deficits in synaptic plasticity. The use of metabotropic glutamate receptor (mGluR) blockers and gamma amino-butyric acid (GABA) agonists have been shown to be efficacious in reversing cellular and behavioral phenotypes, and restoring proper brain connectivity in the mouse and fly models. Proposed new pharmacological treatments and educational interventions are discussed in this chapter. In combination, these various targeted treatments show promising preliminary results in mitigating or even reversing the neurobiological abnormalities caused by loss of FMRP, with possible translational applications to other neurodevelopmental disorders including autism.
Collapse
Affiliation(s)
- Randi Hagerman
- Department of Pediatrics, University of California, Sacramento, CA, USA.
| | | | | | | |
Collapse
|
28
|
Developmental regulation of group I metabotropic glutamate receptors in the premature brain and their protective role in a rodent model of periventricular leukomalacia. ACTA ACUST UNITED AC 2011; 6:277-88. [PMID: 22169210 DOI: 10.1017/s1740925x11000111] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Cerebral white matter injury in premature infants, known as periventricular leukomalacia (PVL), is common after hypoxia-ischemia (HI). While ionotropic glutamate receptors (iGluRs) can mediate immature white matter injury, we have previously shown that excitotoxic injury to premyelinating oligodendrocytes (preOLs) in vitro can be attenuated by group I metabotropic glutamate receptor (mGluR) agonists. Thus, we evaluated mGluR expression in developing white matter in rat and human brain, and tested the protective efficacy of a central nervous system (CNS)-penetrating mGluR agonist on injury to developing oligodendrocytes (OLs) in vivo. Group I mGluRs (mGluR1 and mGluR5) were strongly expressed on OLs in neonatal rodent cerebral white matter throughout normal development, with highest expression early in development on preOLs. Specifically at P6, mGluR1 and mGLuR5 were most highly expressed on GalC-positive OLs compared to neurons, axons, astrocytes and microglia. Systemic administration of (1S,3R) 1-aminocyclopentane-trans-1,3,-dicarboxylic acid (ACPD) significantly attenuated the loss of myelin basic protein in the white matter following HI in P6 rats. Assessment of postmortem human tissue showed both mGluR1 and mGluR5 localized on immature OLs in white matter throughout development, with mGluR5 highest in the preterm period. These data indicate group I mGluRs are highly expressed on OLs during the peak period of vulnerability to HI and modulation of mGluRs is protective in a rodent model of PVL. Group I mGluRs may represent important therapeutic targets for protection from HI-mediated white matter injury.
Collapse
|
29
|
Cosgrove KE, Galván EJ, Barrionuevo G, Meriney SD. mGluRs modulate strength and timing of excitatory transmission in hippocampal area CA3. Mol Neurobiol 2011; 44:93-101. [PMID: 21559753 DOI: 10.1007/s12035-011-8187-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2011] [Accepted: 04/28/2011] [Indexed: 01/25/2023]
Abstract
Excitatory transmission within hippocampal area CA3 stems from three major glutamatergic pathways: the perforant path formed by axons of layer II stellate cells in the entorhinal cortex, the mossy fiber axons originating from the dentate gyrus granule cells, and the recurrent axon collaterals of CA3 pyramidal cells. The synaptic communication of each of these pathways is modulated by metabotropic glutamate receptors that fine-tune the signal by affecting both the timing and strength of the connection. Within area CA3 of the hippocampus, group I mGluRs (mGluR1 and mGluR5) are expressed postsynaptically, whereas group II (mGluR2 and mGluR3) and III mGluRs (mGluR4, mGluR7, and mGluR8) are expressed presynaptically. Receptors from each group have been demonstrated to be required for different forms of pre- and postsynaptic long-term plasticity and also have been implicated in regulating short-term plasticity. A recent observation has demonstrated that a presynaptically expressed mGluR can affect the timing of action potentials elicited in the postsynaptic target. Interestingly, mGluRs can be distributed in a target-specific manner, such that synaptic input from one presynaptic neuron can be modulated by different receptors at each of its postsynaptic targets. Consequently, mGluRs provide a mechanism for synaptic specialization of glutamatergic transmission in the hippocampus. This review will highlight the variability in mGluR modulation of excitatory transmission within area CA3 with an emphasis on how these receptors contribute to the strength and timing of network activity within pyramidal cells and interneurons.
Collapse
Affiliation(s)
- Kathleen E Cosgrove
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | | | | | | |
Collapse
|
30
|
Sheffler DJ, Gregory KJ, Rook JM, Conn PJ. Allosteric modulation of metabotropic glutamate receptors. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2011; 62:37-77. [PMID: 21907906 DOI: 10.1016/b978-0-12-385952-5.00010-5] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The development of receptor subtype-selective ligands by targeting allosteric sites of G protein-coupled receptors (GPCRs) has proven highly successful in recent years. One GPCR family that has greatly benefited from this approach is the metabotropic glutamate receptors (mGlus). These family C GPCRs participate in the neuromodulatory actions of glutamate throughout the CNS, where they play a number of key roles in regulating synaptic transmission and neuronal excitability. A large number of mGlu subtype-selective allosteric modulators have been identified, the majority of which are thought to bind within the transmembrane regions of the receptor. These modulators can either enhance or inhibit mGlu functional responses and, together with mGlu knockout mice, have furthered the establishment of the physiologic roles of many mGlu subtypes. Numerous pharmacological and receptor mutagenesis studies have been aimed at providing a greater mechanistic understanding of the interaction of mGlu allosteric modulators with the receptor, which have revealed evidence for common allosteric binding sites across multiple mGlu subtypes and the presence for multiple allosteric sites within a single mGlu subtype. Recent data have also revealed that mGlu allosteric modulators can display functional selectivity toward particular signal transduction cascades downstream of an individual mGlu subtype. Studies continue to validate the therapeutic utility of mGlu allosteric modulators as a potential therapeutic approach for a number of disorders including anxiety, schizophrenia, Parkinson's disease, and Fragile X syndrome.
Collapse
Affiliation(s)
- Douglas J Sheffler
- Department of Pharmacology, Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | | | | | |
Collapse
|
31
|
Deng PY, Xiao Z, Lei S. Distinct modes of modulation of GABAergic transmission by Group I metabotropic glutamate receptors in rat entorhinal cortex. Hippocampus 2010; 20:980-93. [PMID: 19739246 DOI: 10.1002/hipo.20697] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Activation of metabotropic glutamate receptors (mGluRs) modulates synaptic transmission, whereas the roles of mGluRs in GABAergic transmission in the entorhinal cortex (EC) are elusive. Here, we examined the effects of mGluRs on GABAergic transmission onto the principal neurons in the superficial layers of the EC. Bath application of DHPG, a selective Group I mGluR agonist, increased the frequency and amplitude of spontaneous IPSCs (sIPSCs) whereas application of DCG-IV, an agonist for Group II mGluRs or L-AP4, an agonist for Group III mGluRs failed to change significantly sIPSC frequency and amplitude. Bath application of DHPG failed to change significantly the frequency and amplitude of miniature IPSCs (mIPSCs) recorded in the presence of tetradotoxin but significantly reduced the amplitude of IPSCs evoked by extracellular field stimulation or in synaptically connected interneuron-pyramidal neuron pairs in layer III of the EC. DHPG increased the frequency but reduced the amplitude of APs recorded from entorhinal interneurons. Bath application of DHPG generated membrane depolarization and increased the input resistance of GABAergic interneurons. DHPG-mediated depolarization of GABAergic interneurons was mediated by inhibition of background K(+) channels which are insensitive to extracellular Cs(+), TEA, 4-AP, and Ba(2+). DHPG-induced facilitation of sIPSCs was mediated by mGluR(5) and required the function of Galphaq but was independent of phospholipase C activity. Elevation of synaptic glutamate concentration by bath application of glutamate transporter inhibitors significantly increased sIPSC frequency and amplitude demonstrating a physiological role of mGluRs in GABAergic transmission. Our results provide a cellular and molecular mechanism to explain the physiological and pathological roles of mGluRs in the EC.
Collapse
Affiliation(s)
- Pan-Yue Deng
- Department of Pharmacology, Physiology and Therapeutics, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota 58203, USA
| | | | | |
Collapse
|
32
|
Park JY, Remy S, Varela J, Cooper DC, Chung S, Kang HW, Lee JH, Spruston N. A post-burst after depolarization is mediated by group i metabotropic glutamate receptor-dependent upregulation of Ca(v)2.3 R-type calcium channels in CA1 pyramidal neurons. PLoS Biol 2010; 8:e1000534. [PMID: 21103408 PMCID: PMC2982802 DOI: 10.1371/journal.pbio.1000534] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2010] [Accepted: 09/21/2010] [Indexed: 12/20/2022] Open
Abstract
The excitability of hippocampal pyramidal neurons is regulated by activation of metabotropic glutamate receptors, an effect that is mediated by modulation of R-type calcium channels. Activation of group I metabotropic glutamate receptors (subtypes mGluR1 and mGluR5) regulates neural activity in a variety of ways. In CA1 pyramidal neurons, activation of group I mGluRs eliminates the post-burst afterhyperpolarization (AHP) and produces an afterdepolarization (ADP) in its place. Here we show that upregulation of Cav2.3 R-type calcium channels is responsible for a component of the ADP lasting several hundred milliseconds. This medium-duration ADP is rapidly and reversibly induced by activation of mGluR5 and requires activation of phospholipase C (PLC) and release of calcium from internal stores. Effects of mGluR activation on subthreshold membrane potential changes are negligible but are large following action potential firing. Furthermore, the medium ADP exhibits a biphasic activity dependence consisting of short-term facilitation and longer-term inhibition. These findings suggest that mGluRs may dramatically alter the firing of CA1 pyramidal neurons via a complex, activity-dependent modulation of Cav2.3 R-type channels that are activated during spiking at physiologically relevant rates and patterns. The hippocampus is an essential structure in the brain for the formation of new declarative memories. Understanding the cellular basis of memory formation, storage, and recall in the hippocampus requires a knowledge of the properties of the relevant neurons and how they are modulated by activity in the neural circuit. For many years, we have known that various chemical neurotransmitters can modulate the electrical excitability of neurons in the hippocampus. Here, we report new experiments to reveal how the chemical neurotransmitter glutamate increases neuronal excitability. The effect we study is the conversion of the afterhyperpolarization (a cellular consequence of firing an action potential) to an afterdepolarization. We identified the metabotropic glutamate receptors involved in this conversion (receptors called mGluR1 and mGluR5) as well as the final target of modulation (R-type calcium channels composed of Cav2.3 subunits), which cause the neurons to exhibit altered excitability in the presence of glutamate. We also determined some of the intermediate steps between activation of the glutamate receptors and modulation of the calcium channels responsible for the change in excitability, offering further mechanistic insight into how synaptic transmission can regulate cellular and network activity.
Collapse
Affiliation(s)
- Jin-Yong Park
- Department of Neurobiology and Physiology, Northwestern University, Evanston, Illinois, United States of America
| | - Stefan Remy
- Department of Neurobiology and Physiology, Northwestern University, Evanston, Illinois, United States of America
| | - Juan Varela
- Department of Neurobiology and Physiology, Northwestern University, Evanston, Illinois, United States of America
| | - Donald C. Cooper
- Department of Neurobiology and Physiology, Northwestern University, Evanston, Illinois, United States of America
| | - Sungkwon Chung
- Department of Neurobiology and Physiology, Northwestern University, Evanston, Illinois, United States of America
| | - Ho-Won Kang
- Department of Life Science and Basic Science Institute for Cell Damage Control, Sogang University, Seoul, Korea
| | - Jung-Ha Lee
- Department of Life Science and Basic Science Institute for Cell Damage Control, Sogang University, Seoul, Korea
| | - Nelson Spruston
- Department of Neurobiology and Physiology, Northwestern University, Evanston, Illinois, United States of America
- * E-mail:
| |
Collapse
|
33
|
Gregory KJ, Dong EN, Meiler J, Conn PJ. Allosteric modulation of metabotropic glutamate receptors: structural insights and therapeutic potential. Neuropharmacology 2010; 60:66-81. [PMID: 20637216 DOI: 10.1016/j.neuropharm.2010.07.007] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2010] [Revised: 06/28/2010] [Accepted: 07/06/2010] [Indexed: 10/19/2022]
Abstract
Allosteric modulation of G protein-coupled receptors (GPCRs) represents a novel approach to the development of probes and therapeutics that is expected to enable subtype-specific regulation of central nervous system target receptors. The metabotropic glutamate receptors (mGlus) are class C GPCRs that play important neuromodulatory roles throughout the brain, as such they are attractive targets for therapeutic intervention for a number of psychiatric and neurological disorders including anxiety, depression, Fragile X Syndrome, Parkinson's disease and schizophrenia. Over the last fifteen years, selective allosteric modulators have been identified for many members of the mGlu family. The vast majority of these allosteric modulators are thought to bind within the transmembrane-spanning domains of the receptors to enhance or inhibit functional responses. A combination of mutagenesis-based studies and pharmacological approaches are beginning to provide a better understanding of mGlu allosteric sites. Collectively, when mapped onto a homology model of the different mGlu subtypes based on the β(2)-adrenergic receptor, the previous mutagenesis studies suggest commonalities in the location of allosteric sites across different members of the mGlu family. In addition, there is evidence for multiple allosteric binding pockets within the transmembrane region that can interact to modulate one another. In the absence of a class C GPCR crystal structure, this approach has shown promise with respect to the interpretation of mutagenesis data and understanding structure-activity relationships of allosteric modulator pharmacophores.
Collapse
Affiliation(s)
- Karen J Gregory
- Department of Pharmacology, Vanderbilt Program in Drug Discovery, Vanderbilt University Medical Center, Nashville, TN 37232-0697, USA.
| | | | | | | |
Collapse
|
34
|
The expression of astroglial glutamate transporters in patients with focal cortical dysplasia: an immunohistochemical study. Acta Neurochir (Wien) 2010; 152:845-53. [PMID: 19859653 DOI: 10.1007/s00701-009-0548-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2009] [Accepted: 10/12/2009] [Indexed: 10/20/2022]
Abstract
PURPOSE An abnormal increase in the extracellular glutamate is thought to play a crucial role in the initiation, spread, and maintenance of seizure activity.In normal conditions, the majority of this excess glutamate is cleared via glial glutamate transporters (EAAT-1 and EAAT-2). We aimed to examine the immunohistochemical expression of these transporters in the dysplastic tissues of patients with focal cortical dysplasia (FCD). METHODS The parafin-embedded dysplastic tissues of 33 patients who were operated on due to medically intractable epilepsy and histopathologically diagnosed with FCD between 2001 and 2006 were stained immunohistochemically with appropriate antibodies, and the distribution and intensity of immunoreactivity (IR) of EAAT-1 and EAAT-2 were examined.The findings were compared with the histologically normal tissues of five patients who underwent temporal lobectomy for epilepsy surgery and 10 fresh postmortem cases. RESULTS In the majority of the patients, the EAAT-1 and EAAT-2 IR were decreased, their astrocytic expression were lower, and the pattern of distribution were more diffused when compared to the control groups.Analyzing these findings according to the types of FCD revealed that as the severity of the dysplasia increased, the IR and astrocytic expression of both transporters are decreased and their distribution tend to be more "diffused." CONCLUSION The results of this study suggest a relationship between the decreased glutamate transporter expressions in dysplastic tissues which,in turn, may cause increased extracellular concentrations of glutamate and FCD pathophysiology.Further studies with larger patient populations,investigating the expression of glutamate transporters at mRNA and protein levels, are required to clarify their roles in the pathophysiology of FCD.
Collapse
|
35
|
Abstract
Epileptogenesis is defined as the process of developing epilepsy-a disorder characterized by recurrent seizures-following an initial insult. Seizure incidence during the human lifespan is at its highest in infancy and childhood. Animal models of epilepsy and human tissue studies suggest that epileptogenesis involves a cascade of molecular, cellular and neuronal network alterations. Within minutes to days following the initial insult, there are acute early changes in neuronal networks, which include rapid alterations to ion channel kinetics as a result of membrane depolarization, post-translational modifications to existing functional proteins, and activation of immediate early genes. Subacute changes occur over hours to weeks, and include transcriptional events, neuronal death and activation of inflammatory cascades. The chronic changes that follow over weeks to months include anatomical changes, such as neurogenesis, mossy fiber sprouting, network reorganization, and gliosis. These epileptogenic processes are developmentally regulated and might contribute to differences in epileptogenesis between adult and developing brains. Here we review the factors responsible for enhanced seizure susceptibility in the developing brain, and consider age-specific mechanisms of epileptogenesis. An understanding of these factors could yield potential therapeutic targets for the prevention of epileptogenesis and also provide biomarkers for identifying patients at risk of developing epilepsy or for monitoring disease progression.
Collapse
|
36
|
Watanabe Y, Kaida Y, Takechi K, Kamei C. Anticonvulsant Effect of (RS)-1-Aminoindan-1,5-dicarboxylic Acid on Pentetrazol-Induced Kindled Seizures in Mice. Biol Pharm Bull 2010; 33:647-52. [DOI: 10.1248/bpb.33.647] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Yusuke Watanabe
- Department of Medicinal Pharmacology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences
| | - Yuko Kaida
- Department of Medicinal Pharmacology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences
| | - Kenshi Takechi
- Department of Medicinal Pharmacology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences
| | - Chiaki Kamei
- Department of Medicinal Pharmacology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences
| |
Collapse
|
37
|
|
38
|
Pałucha-Poniewiera A, Wierońska JM, Brański P, Stachowicz K, Chaki S, Pilc A. On the mechanism of the antidepressant-like action of group II mGlu receptor antagonist, MGS0039. Psychopharmacology (Berl) 2010; 212:523-35. [PMID: 20703449 PMCID: PMC2981731 DOI: 10.1007/s00213-010-1978-5] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2010] [Accepted: 07/25/2010] [Indexed: 10/27/2022]
Abstract
RATIONALE Several studies have suggested that modulation of the glutamatergic system could be a new, efficient way to achieve antidepressant activity. Behavioral data showed that group II mGlu receptor antagonists (i.e., (1R, 2R, 3R, 5R, 6R)-2-amino-3-(3,4-dichlorobenzyloxy)-6-fluorobicyclo[3.1.0]hexane-2,6-dicarboxylic acid (MGS0039) and (2S)-2-amino-2-[(1S,2S)-2-carboxycycloprop-1-yl]-3-(xan th-9-yl) propanoic acid (LY341495)) elicited antidepressant activity in several animal models of depression in rats and/or mice. Although the antidepressant-like activity of MGS0039 and LY341495 is well documented, the mechanism of the antidepressant action of these compounds is still not clear. OBJECTIVES The aim of the present study was to specify the role of the serotonergic system in the mechanism of the antidepressant-like activity of group II mGlu receptor ligands by using the tail suspension test (TST) in mice; the role of AMPA receptors was also investigated. Furthermore, the possible antidepressant-like action of MGS0039 using the olfactory bulbectomy (OB) model of depression in rats was investigated. RESULTS The results of the TST studies showed that antidepressant-like action of group II mGlu receptor antagonists does not depend on serotonergic system activation. However, the AMPA receptor seems to play a key role in the antidepressant-like action of these compounds. Moreover, we have shown that repeated administration of MGS0039 attenuated OB-related deficits, confirming antidepressant-like activity of the tested compound. CONCLUSIONS The results suggest that the blockade of group II mGlu receptors may be effective in the treatment of depression. Moreover, we have found that the mechanism of action of group II mGlu receptor antagonists differs from that of typical antidepressants, such as SSRIs.
Collapse
Affiliation(s)
- Agnieszka Pałucha-Poniewiera
- Department of Neurobiology, Institute of Pharmacology Polish Academy of Sciences, Smętna 12, 31-343, Kraków, Poland.
| | - Joanna M. Wierońska
- Department of Neurobiology, Institute of Pharmacology Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland
| | - Piotr Brański
- Department of Neurobiology, Institute of Pharmacology Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland
| | - Katarzyna Stachowicz
- Department of Neurobiology, Institute of Pharmacology Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland
| | - Shigeyuki Chaki
- Discovery Pharmacology, Molecular Function and Pharmacology Laboratories, Taisho Pharmaceutical Co., Ltd, 1-403 Yoshino-cho, Kita-ku, Saitama, 331-9530 Japan
| | - Andrzej Pilc
- Department of Neurobiology, Institute of Pharmacology Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland ,Collegium Medicum, Faculty of Health Sciences, Jagiellonian University, Kraków, Poland
| |
Collapse
|
39
|
Abstract
Calpain is a ubiquitous protease found in different tissue types and in many organisms including mammals. It generally does not destroy its large variety of substrates, but more commonly disrupts their function. In neurons, many of its substrates become dysregulated as a result of cleavage of their regulatory domain by this protease, leading to altered signaling between cells. In glutamatergic synaptic transmission, direct targets of calpain include all of the major glutamate receptors: NMDA receptors, AMPA receptors and mGluR. By cleaving these receptors and associated intracellular proteins, calpain may regulate the physiology at glutamatergic synapses. As a result, calpain-mediated cleavage in neurons might not only be involved in pathological events like excitotoxicity, but may also have neuroprotective effects and roles in physiological synaptic transmission.
Collapse
Affiliation(s)
- Shachee Doshi
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | | |
Collapse
|
40
|
Szaniszlo P, German P, Hajas G, Saenz DN, Kruzel M, Boldogh I. New insights into clinical trial for Colostrinin in Alzheimer's disease. J Nutr Health Aging 2009; 13:235-41. [PMID: 19262960 DOI: 10.1007/s12603-009-0065-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
BACKGROUND The pathomechanism of Alzheimer's disease (AD) is multifactorial although the most popular hypotheses are centered on the effects of the misfolded, aggregated protein, amyloid beta (Abeta) and on Tau hyperphosphorylation. OBJECTIVES Double blinded clinical trials were planned to demonstrate the effect of Colostrinin (CLN) on instrumental daily activities of AD patients. The potential molecular mechanisms by which CLN mediates its effects were investigated by gene expression profiling. METHODS RNAs isolated from CLN-treated cells were analyzed by high-density oligonucleotide arrays. Network and pathway analyses were performed using the Ingenuity Pathway Analysis software. RESULTS The Full Sample Analysis at week 15 showed a stabilizing effect of CLN on cognitive function in ADAS-cog (p = 0.02) and on daily function in IADL (p = 0.02). The overall patient response was also in favor of CLN (p = 0.03). Patients graded as mild on entry also showed a superior response of ADAS-cog compared to more advanced cases (p = 0.01). Data derived from microarray network analysis show that CLN elicits highly complex and multiphasic changes in the cells' transcriptome. Importantly, transcriptomal analysis showed that CLN alters gene expression of molecular networks implicated in Abeta precursor protein synthesis, Tau phosphorylation and increased levels of enzymes that proteolitically eliminate Abeta. In addition, CLN enhanced the defense against oxidative stress and decreased expression of inflammatory chemokines and cytokines, thereby attenuating inflammatory processes that precede Alzheimer's and other neurological diseases. CONCLUSION Together these data suggest that CLN has promising potential for clinical use in prevention and therapy of Alzheimer's and other age-associated central nervous system diseases.
Collapse
Affiliation(s)
- P Szaniszlo
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, Galveston, Texas 77555, USA
| | | | | | | | | | | |
Collapse
|
41
|
Byrnes KR, Loane DJ, Faden AI. Metabotropic glutamate receptors as targets for multipotential treatment of neurological disorders. Neurotherapeutics 2009; 6:94-107. [PMID: 19110202 PMCID: PMC2634659 DOI: 10.1016/j.nurt.2008.10.038] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Glutamate is a major excitatory neurotransmitter in the CNS that is involved in numerous cellular functions, including cell death and survival. Metabotropic glutamate receptors (mGluR) are G-protein coupled receptors that have been classified into three groups on the basis of signal transduction pathways and pharmacological profiles. Group I, II, and III mGluRs are found on cell types within and peripheral to the CNS, including neurons, microglia, astrocytes, oligodendrocytes, T- and B-cell lymphocytes, osteoblasts, hepatocytes, and endothelial cells, among others. These receptors have a number of effects on cells that can influence outcome after trauma, including reducing neuronal and oligodendroglial cell death, inflammation, and endothelial permeability. Thus, mGluRs are a promising multipotential therapeutic approach. Because the pathology of CNS trauma and neurodegeneration is multifactorial (including, for example, oxidative stress, mitochondrial breakdown, and inflammation), therapies that serve to modulate multiple pathophysiological pathways may prove more effective than those directed at a single target. This review examines the multipotential therapeutic utility of mGluR modulation in acute and chronic injury and neurodegeneration.
Collapse
Affiliation(s)
- Kimberly R Byrnes
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC 20057, USA.
| | | | | |
Collapse
|
42
|
Szaniszlo P, German P, Hajas G, Saenz DN, Woodberry MW, Kruzel ML, Boldogh I. Effects of Colostrinin on gene expression-transcriptomal network analysis. Int Immunopharmacol 2008; 9:181-93. [PMID: 19015048 DOI: 10.1016/j.intimp.2008.10.022] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2008] [Revised: 10/23/2008] [Accepted: 10/27/2008] [Indexed: 11/26/2022]
Abstract
Colostrinin (CLN) is a uniform mixture of low-molecular weight proline-rich polypeptides isolated from the mother's first milk, colostrum. Exposure of cells to CLN decreases intracellular levels of reactive oxygen species by regulating glutathione metabolism and modulating activities of antioxidant enzymes and mitochondrial function. It also inhibits beta amyloid-induced apoptosis and induces neurite outgrowth of pheochromocytoma cells. Administration of CLN to Alzheimer's disease patients has resulted in a stabilizing effect on cognitive function. We analyzed CLN-induced gene expression changes using high-density oligonucleotide arrays and transcriptomal network analysis. We found that CLN elicited highly complex and multiphasic changes in the gene expression profile of treated cells. CLN treatment affected a total of 58 molecular networks, 27 of which contained at least 10 differentially expressed genes. Here we present CLN-modulated gene networks as potential underlying molecular mechanisms leading to the reported effects of CLN on cellular oxidative state, chemokine and cytokine production, and cell differentiation, as well as on pathological processes like allergy, asthma, Alzheimer's, and other neurological diseases. Based on our results, we also predict possible modulatory effects of CLN on adipocytokine gene networks that play a crucial role in the pathobiology of diabetes, cardiovascular disorders, obesity, and inflammation. Taken together, CLN-altered gene expression networks presented here provide the molecular basis for previously described biological phenomena and predict potential fields of application for CLN in the prevention and treatment of diseases.
Collapse
Affiliation(s)
- Peter Szaniszlo
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas 77555, USA
| | | | | | | | | | | | | |
Collapse
|
43
|
Maciejak P, Lehner M, Turzyńska D, Szyndler J, Bidziński A, Taracha E, Sobolewska A, Walkowiak J, Skórzewska A, Wisłowska A, Hamed A, Płaźnik A. The opposite role of hippocampal mGluR1 in fear conditioning in kindled and non-kindled rats. Brain Res 2008; 1187:184-93. [DOI: 10.1016/j.brainres.2007.10.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2007] [Revised: 10/10/2007] [Accepted: 10/11/2007] [Indexed: 11/24/2022]
|
44
|
Rousseaux CG. A Review of Glutamate Receptors II: Pathophysiology and Pathology. J Toxicol Pathol 2008. [DOI: 10.1293/tox.21.133] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Affiliation(s)
- Colin G. Rousseaux
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, University of Ottawa
| |
Collapse
|
45
|
Palucha A, Pilc A. Metabotropic glutamate receptor ligands as possible anxiolytic and antidepressant drugs. Pharmacol Ther 2007; 115:116-47. [PMID: 17582504 DOI: 10.1016/j.pharmthera.2007.04.007] [Citation(s) in RCA: 175] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2007] [Accepted: 04/25/2007] [Indexed: 11/18/2022]
Abstract
Depression and anxiety represent a major problem. However, the current treatment of both groups of diseases is not satisfactory. As the glutamatergic system may play an important role in pathophysiology of both depression and anxiety, we decided to discuss the recent data on possible anxiolytic and/or antidepressant effects of metabotropic glutamate (mGlu) receptor ligands. Preclinical data indicated that antagonists of group I mGlu receptors, particularly antagonists of mGlu5 receptors, produced both anxiolytic-like and antidepressant-like effects. Clinical data also demonstrated that mGlu5 receptor antagonist, fenobam, was an active anxiolytic drug. The anxiolytic effects exerted by mGlu5 receptor antagonists are profound, comparable with or stronger than those of benzodiazepines. However, the problem with the psychotomimetic activity of mGlu5 receptor antagonists and their possible influence on memory has to be further investigated. Among all mGlu receptor ligands, group II mGlu receptor agonists seem to be the drugs with the most promising therapeutic potential and a good safety profile. Animal studies showed anxiolytic-like effects of group II mGlu receptor agonists. Currently, group II mGlu receptor agonists are in phase III clinical trials for potential treatment of anxiety disorders. On the other hand, data has been accumulated, indicating that antagonists of group II mGlu receptors have an antidepressant potential. Group III mGlu receptor ligands represent the least investigated group of mGlu receptors. However, preclinical data also indicates that ligands of these receptors, both agonists and antagonists, may have an anxiolytic-like and antidepressant-like potential.
Collapse
Affiliation(s)
- Agnieszka Palucha
- Institute of Pharmacology, Polish Academy of Sciences, Smetna 12, 31-343 Kraków, Poland
| | | |
Collapse
|
46
|
Bach P, Isaac M, Slassi A. Metabotropic glutamate receptor 5 modulators and their potential therapeutic applications. Expert Opin Ther Pat 2007. [DOI: 10.1517/13543776.17.4.371] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
47
|
Abstract
Chemical Biology is a relatively new field, and as such is not yet simply or succinctly defined. It includes such a wide range of fundamental problems that this commentary could only include just a few snapshots of potential areas of interest. Overarching themes and selected recent successes and ideas in chemical biology are described to illustrate broadly the scope of the field, but should not be taken as exhaustive. The Chemical Biology Section of Chemistry Central Journal is pleased to receive manuscripts describing research into all and any aspects of the subject.
Collapse
Affiliation(s)
- Elizabeth L Ostler
- School of Pharmacy and Biomolecular Sciences, University of Brighton, Brighton, UK.
| |
Collapse
|