1
|
Sharbafshaaer M, Cirillo G, Esposito F, Tedeschi G, Trojsi F. Harnessing Brain Plasticity: The Therapeutic Power of Repetitive Transcranial Magnetic Stimulation (rTMS) and Theta Burst Stimulation (TBS) in Neurotransmitter Modulation, Receptor Dynamics, and Neuroimaging for Neurological Innovations. Biomedicines 2024; 12:2506. [PMID: 39595072 PMCID: PMC11592033 DOI: 10.3390/biomedicines12112506] [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: 08/31/2024] [Revised: 10/27/2024] [Accepted: 10/29/2024] [Indexed: 11/28/2024] Open
Abstract
Transcranial magnetic stimulation (TMS) methods have become exciting techniques for altering brain activity and improving synaptic plasticity, earning recognition as valuable non-medicine treatments for a wide range of neurological disorders. Among these methods, repetitive TMS (rTMS) and theta-burst stimulation (TBS) show significant promise in improving outcomes for adults with complex neurological and neurodegenerative conditions, such as Alzheimer's disease, stroke, Parkinson's disease, etc. However, optimizing their effects remains a challenge due to variability in how patients respond and a limited understanding of how these techniques interact with crucial neurotransmitter systems. This narrative review explores the mechanisms of rTMS and TBS, which enhance neuroplasticity and functional improvement. We specifically focus on their effects on GABAergic and glutamatergic pathways and how they interact with key receptors like N-Methyl-D-Aspartate (NMDA) and AMPA receptors, which play essential roles in processes like long-term potentiation (LTP) and long-term depression (LTD). Additionally, we investigate how rTMS and TBS impact neuroplasticity and functional connectivity, particularly concerning brain-derived neurotrophic factor (BDNF) and tropomyosin-related kinase receptor type B (TrkB). Here, we highlight the significant potential of this research to expand our understanding of neuroplasticity and better treatment outcomes for patients. Through clarifying the neurobiology mechanisms behind rTMS and TBS with neuroimaging findings, we aim to develop more effective, personalized treatment plans that effectively address the challenges posed by neurological disorders and ultimately enhance the quality of neurorehabilitation services and provide future directions for patients' care.
Collapse
Affiliation(s)
- Minoo Sharbafshaaer
- First Division of Neurology, Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (F.E.); (G.T.); (F.T.)
| | - Giovanni Cirillo
- Division of Human Anatomy, Neuronal Networks Morphology & Systems Biology Lab, Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli, 80138 Naples, Italy;
| | - Fabrizio Esposito
- First Division of Neurology, Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (F.E.); (G.T.); (F.T.)
| | - Gioacchino Tedeschi
- First Division of Neurology, Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (F.E.); (G.T.); (F.T.)
| | - Francesca Trojsi
- First Division of Neurology, Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (F.E.); (G.T.); (F.T.)
| |
Collapse
|
2
|
Jin D, Chen H, Zhou MH, Chen SR, Pan HL. mGluR5 from Primary Sensory Neurons Promotes Opioid-Induced Hyperalgesia and Tolerance by Interacting with and Potentiating Synaptic NMDA Receptors. J Neurosci 2023; 43:5593-5607. [PMID: 37451981 PMCID: PMC10401648 DOI: 10.1523/jneurosci.0601-23.2023] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 06/29/2023] [Accepted: 07/04/2023] [Indexed: 07/18/2023] Open
Abstract
Aberrant activation of presynaptic NMDARs in the spinal dorsal horn is integral to opioid-induced hyperalgesia and analgesic tolerance. However, the signaling mechanisms responsible for opioid-induced NMDAR hyperactivity remain poorly identified. Here, we show that repeated treatment with morphine or fentanyl reduced monomeric mGluR5 protein levels in the dorsal root ganglion (DRG) but increased levels of mGluR5 monomers and homodimers in the spinal cord in mice and rats of both sexes. Coimmunoprecipitation analysis revealed that monomeric and dimeric mGluR5 in the spinal cord, but not monomeric mGluR5 in the DRG, directly interacted with GluN1. By contrast, mGluR5 did not interact with μ-opioid receptors in the DRG or spinal cord. Repeated morphine treatment markedly increased the mGluR5-GluN1 interaction and protein levels of mGluR5 and GluN1 in spinal synaptosomes. The mGluR5 antagonist MPEP reversed morphine treatment-augmented mGluR5-GluN1 interactions, GluN1 synaptic expression, and dorsal root-evoked monosynaptic EPSCs of dorsal horn neurons. Furthermore, CRISPR-Cas9-induced conditional mGluR5 knockdown in DRG neurons normalized mGluR5 levels in spinal synaptosomes and NMDAR-mediated EPSCs of dorsal horn neurons increased by morphine treatment. Correspondingly, intrathecal injection of MPEP or conditional mGluR5 knockdown in DRG neurons not only potentiated the acute analgesic effect of morphine but also attenuated morphine treatment-induced hyperalgesia and tolerance. Together, our findings suggest that opioid treatment promotes mGluR5 trafficking from primary sensory neurons to the spinal dorsal horn. Through dimerization and direct interaction with NMDARs, presynaptic mGluR5 potentiates and/or stabilizes NMDAR synaptic expression and activity at primary afferent central terminals, thereby maintaining opioid-induced hyperalgesia and tolerance.SIGNIFICANCE STATEMENT Opioids are essential analgesics for managing severe pain caused by cancer, surgery, and tissue injury. However, these drugs paradoxically induce pain hypersensitivity and tolerance, which can cause rapid dose escalation and even overdose mortality. This study demonstrates, for the first time, that opioids promote trafficking of mGluR5, a G protein-coupled glutamate receptor, from peripheral sensory neurons to the spinal cord; there, mGluR5 proteins dimerize and physically interact with NMDARs to augment their synaptic expression and activity. Through dynamic interactions, the two distinct glutamate receptors mutually amplify and sustain nociceptive input from peripheral sensory neurons to the spinal cord. Thus, inhibiting mGluR5 activity or disrupting mGluR5-NMDAR interactions could reduce opioid-induced hyperalgesia and tolerance and potentiate opioid analgesic efficacy.
Collapse
Affiliation(s)
- Daozhong Jin
- Center for Neuroscience and Pain Research, Department of Anesthesiology and Perioperative Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas 77030
| | - Hong Chen
- Center for Neuroscience and Pain Research, Department of Anesthesiology and Perioperative Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas 77030
| | - Meng-Hua Zhou
- Center for Neuroscience and Pain Research, Department of Anesthesiology and Perioperative Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas 77030
| | - Shao-Rui Chen
- Center for Neuroscience and Pain Research, Department of Anesthesiology and Perioperative Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas 77030
| | - Hui-Lin Pan
- Center for Neuroscience and Pain Research, Department of Anesthesiology and Perioperative Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas 77030
| |
Collapse
|
3
|
Rodrigues NC, Silva-Cruz A, Caulino-Rocha A, Bento-Oliveira A, Alexandre Ribeiro J, Cunha-Reis D. Hippocampal CA1 theta burst-induced LTP from weaning to adulthood: Cellular and molecular mechanisms in young male rats revisited. Eur J Neurosci 2021; 54:5272-5292. [PMID: 34251729 DOI: 10.1111/ejn.15390] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 07/08/2021] [Accepted: 07/08/2021] [Indexed: 01/05/2023]
Abstract
Long-term potentiation (LTP) is a highly studied cellular process, yet determining the transduction and gamma aminobutyric acid (GABAergic) pathways that are the essential versus modulatory for LTP elicited by theta burst stimulation (TBS) in the hippocampal Cornu Ammonis 1 (CA1) area is still elusive, due to the use of different TBS intensities, patterns or different rodent/cellular models. We now characterised the developmental maturation and the transduction and GABAergic pathways required for mild TBS-induced LTP in hippocampal CA1 area in male rats. LTP induced by TBS (5x4) (five bursts of four pulses delivered at 100 Hz) lasted for up to 3 h and was increasingly larger from weaning to adulthood. Stronger TBS patterns - TBS (15x4) or three TBS (15x4) separated by 6 min induced nearly maximal LTP not being the best choice to study the value of LTP-enhancing drugs. LTP induced by TBS (5x4) in young adults was fully dependent on N-methyl D-aspartate (NMDA) receptor and calmodulin-dependent protein kinase II (CaMKII) activity but independent of protein kinase A (PKA) or protein kinase C (PKC) activity. Furthermore, it was partially dependent on GABAB receptor activation and was potentiated by GABAA receptor blockade and less by GAT-1 transporter blockade. AMPA GluA1 phosphorylation on Ser831 (CaMKII target) but not GluA1 Ser845 (PKA target) was essential for LTP expression. The phosphorylation of the Kv4.2 channel was observed at Ser438 (CaMKII target) but not at Thr602 or Thr607 (ERK/MAPK pathway target). This suggests that cellular kinases like PKA, PKC, or kinases of the ERK/MAPK family although important modulators of TBS (5x4)-induced LTP may not be essential for its expression in the CA1 area of the hippocampus.
Collapse
Affiliation(s)
| | - Armando Silva-Cruz
- Instituto de Medicina Molecular, Unidade de Neurociências, Lisbon, Portugal
| | - Ana Caulino-Rocha
- Departamento de Química e Bioquímica, Faculty of Sciences, Universidade de Lisboa, Lisbon, Portugal.,BioISI-Biosystems & Integrative Sciences Institute, Faculdade de Ciências, Universidade de Lisboa, Lisbon, Portugal
| | - Andreia Bento-Oliveira
- Departamento de Química e Bioquímica, Faculty of Sciences, Universidade de Lisboa, Lisbon, Portugal.,BioISI-Biosystems & Integrative Sciences Institute, Faculdade de Ciências, Universidade de Lisboa, Lisbon, Portugal
| | - Joaquim Alexandre Ribeiro
- Instituto de Medicina Molecular, Unidade de Neurociências, Lisbon, Portugal.,Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Diana Cunha-Reis
- Instituto de Medicina Molecular, Unidade de Neurociências, Lisbon, Portugal.,Departamento de Química e Bioquímica, Faculty of Sciences, Universidade de Lisboa, Lisbon, Portugal.,BioISI-Biosystems & Integrative Sciences Institute, Faculdade de Ciências, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
4
|
RGS14 Regulation of Post-Synaptic Signaling and Spine Plasticity in Brain. Int J Mol Sci 2021; 22:ijms22136823. [PMID: 34201943 PMCID: PMC8268017 DOI: 10.3390/ijms22136823] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 06/12/2021] [Accepted: 06/14/2021] [Indexed: 02/06/2023] Open
Abstract
The regulator of G-protein signaling 14 (RGS14) is a multifunctional signaling protein that regulates post synaptic plasticity in neurons. RGS14 is expressed in the brain regions essential for learning, memory, emotion, and stimulus-induced behaviors, including the basal ganglia, limbic system, and cortex. Behaviorally, RGS14 regulates spatial and object memory, female-specific responses to cued fear conditioning, and environmental- and psychostimulant-induced locomotion. At the cellular level, RGS14 acts as a scaffolding protein that integrates G protein, Ras/ERK, and calcium/calmodulin signaling pathways essential for spine plasticity and cell signaling, allowing RGS14 to naturally suppress long-term potentiation (LTP) and structural plasticity in hippocampal area CA2 pyramidal cells. Recent proteomics findings indicate that RGS14 also engages the actomyosin system in the brain, perhaps to impact spine morphogenesis. Of note, RGS14 is also a nucleocytoplasmic shuttling protein, where its role in the nucleus remains uncertain. Balanced nuclear import/export and dendritic spine localization are likely essential for RGS14 neuronal functions as a regulator of synaptic plasticity. Supporting this idea, human genetic variants disrupting RGS14 localization also disrupt RGS14’s effects on plasticity. This review will focus on the known and unexplored roles of RGS14 in cell signaling, physiology, disease and behavior.
Collapse
|
5
|
Key J, Mueller AK, Gispert S, Matschke L, Wittig I, Corti O, Münch C, Decher N, Auburger G. Ubiquitylome profiling of Parkin-null brain reveals dysregulation of calcium homeostasis factors ATP1A2, Hippocalcin and GNA11, reflected by altered firing of noradrenergic neurons. Neurobiol Dis 2019; 127:114-130. [PMID: 30763678 DOI: 10.1016/j.nbd.2019.02.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 12/05/2018] [Accepted: 02/08/2019] [Indexed: 02/06/2023] Open
Abstract
Parkinson's disease (PD) is the second most frequent neurodegenerative disorder in the old population. Among its monogenic variants, a frequent cause is a mutation in the Parkin gene (Prkn). Deficient function of Parkin triggers ubiquitous mitochondrial dysfunction and inflammation in the brain, but it remains unclear how selective neural circuits become vulnerable and finally undergo atrophy. We attempted to go beyond previous work, mostly done in peripheral tumor cells, which identified protein targets of Parkin activity, an ubiquitin E3 ligase. Thus, we now used aged Parkin-knockout (KO) mouse brain for a global quantification of ubiquitylated peptides by mass spectrometry (MS). This approach confirmed the most abundant substrate to be VDAC3, a mitochondrial outer membrane porin that modulates calcium flux, while uncovering also >3-fold dysregulations for neuron-specific factors. Ubiquitylation decreases were prominent for Hippocalcin (HPCA), Calmodulin (CALM1/CALML3), Pyruvate Kinase (PKM2), sodium/potassium-transporting ATPases (ATP1A1/2/3/4), the Rab27A-GTPase activating protein alpha (TBC1D10A) and an ubiquitin ligase adapter (DDB1), while strong increases occurred for calcium transporter ATP2C1 and G-protein subunits G(i)/G(o)/G(Tr). Quantitative immunoblots validated elevated abundance for the electrogenic pump ATP1A2, for HPCA as neuron-specific calcium sensor, which stimulates guanylate cyclases and modifies axonal slow afterhyperpolarization (sAHP), and for the calcium-sensing G-protein GNA11. We assessed if compensatory molecular regulations become insufficient over time, leading to functional deficits. Patch clamp experiments in acute Parkin-KO brain slices indeed revealed alterations of the electrophysiological properties in aged noradrenergic locus coeruleus (LC) neurons. LC neurons of aged Parkin-KO brain showed an acceleration of the spontaneous pacemaker frequency, a reduction in sAHP and shortening of action potential duration, without modulation of KCNQ potassium currents. These findings indicate altered calcium-dependent excitability in a PARK2 model of PD, mediated by diminished turnover of potential Parkin targets such as ATP1A2 and HPCA. The data also identified further novel Parkin substrate candidates like SIRT2, OTUD7B and CUL5. Our elucidation of neuron-specific mechanisms of PD pathogenesis helps to explain the known exceptional susceptibility of noradrenergic and dopaminergic projections to alterations of calcium homeostasis and its mitochondrial buffering.
Collapse
Affiliation(s)
- J Key
- Exp. Neurology, Goethe University Medical School, 60590 Frankfurt am Main, Germany
| | - A K Mueller
- Institute for Physiology and Pathophysiology, Vegetative Physiology and Marburg Center for Mind, Brain and Behavior - MCMBB; Clinic for Neurology, Philipps-University Marburg, 35037 Marburg, Germany
| | - S Gispert
- Exp. Neurology, Goethe University Medical School, 60590 Frankfurt am Main, Germany
| | - L Matschke
- Institute for Physiology and Pathophysiology, Vegetative Physiology and Marburg Center for Mind, Brain and Behavior - MCMBB; Clinic for Neurology, Philipps-University Marburg, 35037 Marburg, Germany
| | - I Wittig
- Functional Proteomics, SFB 815 Core Unit, Goethe University Medical School, 60590 Frankfurt am Main, Germany
| | - O Corti
- Institut du Cerveau et de la Moelle épinière, ICM, Paris, F-75013, France; Inserm, U1127, Paris, F-75013, France; CNRS, UMR 7225, Paris, F-75013, France; Sorbonne Universités, Paris, F-75013, France
| | - C Münch
- Institute of Biochemistry II, Goethe University Medical School, 60590 Frankfurt am Main, Germany
| | - N Decher
- Institute for Physiology and Pathophysiology, Vegetative Physiology and Marburg Center for Mind, Brain and Behavior - MCMBB; Clinic for Neurology, Philipps-University Marburg, 35037 Marburg, Germany.
| | - G Auburger
- Exp. Neurology, Goethe University Medical School, 60590 Frankfurt am Main, Germany.
| |
Collapse
|
6
|
Hippocampal Protein Kinase C Signaling Mediates the Short-Term Memory Impairment Induced by Delta9-Tetrahydrocannabinol. Neuropsychopharmacology 2018; 43:1021-1031. [PMID: 28816239 PMCID: PMC5854793 DOI: 10.1038/npp.2017.175] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 08/02/2017] [Accepted: 08/09/2017] [Indexed: 01/17/2023]
Abstract
Cannabis affects cognitive performance through the activation of the endocannabinoid system, and the molecular mechanisms involved in this process are poorly understood. Using the novel object-recognition memory test in mice, we found that the main psychoactive component of cannabis, delta9-tetrahydrocannabinol (THC), alters short-term object-recognition memory specifically involving protein kinase C (PKC)-dependent signaling. Indeed, the systemic or intra-hippocampal pre-treatment with the PKC inhibitors prevented the short-term, but not the long-term, memory impairment induced by THC. In contrast, systemic pre-treatment with mammalian target of rapamycin complex 1 inhibitors, known to block the amnesic-like effects of THC on long-term memory, did not modify such a short-term cognitive deficit. Immunoblot analysis revealed a transient increase in PKC signaling activity in the hippocampus after THC treatment. Thus, THC administration induced the phosphorylation of a specific Ser residue in the hydrophobic-motif at the C-terminal tail of several PKC isoforms. This significant immunoreactive band that paralleled cognitive performance did not match in size with the major PKC isoforms expressed in the hippocampus except for PKCθ. Moreover, THC transiently enhanced the phosphorylation of the postsynaptic calmodulin-binding protein neurogranin in a PKC dependent manner. These data demonstrate that THC alters short-term object-recognition memory through hippocampal PKC/neurogranin signaling.
Collapse
|
7
|
Akiba C, Nakajima M, Miyajima M, Ogino I, Miura M, Inoue R, Nakamura E, Kanai F, Tada N, Kunichika M, Yoshida M, Nishimura K, Kondo A, Sugano H, Arai H. Leucine-rich α2-glycoprotein overexpression in the brain contributes to memory impairment. Neurobiol Aging 2017; 60:11-19. [PMID: 28917663 DOI: 10.1016/j.neurobiolaging.2017.08.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 07/28/2017] [Accepted: 08/15/2017] [Indexed: 12/19/2022]
Abstract
We previously reported increase in leucine-rich α2-glycoprotein (LRG) concentration in cerebrospinal fluid is associated with cognitive decline in humans. To investigate relationship between LRG expression in the brain and memory impairment, we analyzed transgenic mice overexpressing LRG in the brain (LRG-Tg) focusing on hippocampus. Immunostaining and Western blotting revealed age-related increase in LRG expression in hippocampal neurons in 8-, 24-, and 48-week-old controls and LRG-Tg. Y-maze and Morris water maze tests indicated retained spatial memory in 8- and 24-week-old LRG-Tg, while deteriorated in 48-week-old LRG-Tg compared with age-matched controls. Field excitatory postsynaptic potentials declined with age in LRG-Tg compared with controls at 8, 24, and 48 weeks. Paired-pulse ratio decreased with age in LRG-Tg, while increased in controls. As a result, long-term potentiation was retained in 8- and 24-week-old LRG-Tg, whereas diminished in 48-week-old LRG-Tg compared with age-matched controls. Electron microscopy observations revealed fewer synaptic vesicles and junctions in LRG-Tg compared with age-matched controls, which became significant with age. Hippocampal LRG overexpression contributes to synaptic dysfunction, which leads to memory impairment with advance of age.
Collapse
Affiliation(s)
- Chihiro Akiba
- Department of Neurosurgery, Juntendo University Graduate School of Medicine, Tokyo, Japan.
| | - Madoka Nakajima
- Department of Neurosurgery, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Masakazu Miyajima
- Department of Neurosurgery, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Ikuko Ogino
- Department of Neurosurgery, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Masami Miura
- Neurophysiology Research Group, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Ritsuko Inoue
- Neurophysiology Research Group, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Eri Nakamura
- Laboratory of Disease Model Research, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Fumio Kanai
- Laboratory of Disease Model Research, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Norihiro Tada
- Laboratory of Disease Model Research, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Miyuki Kunichika
- Laboratory of Morpheme Analysis Imaging Research, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Mitsutaka Yoshida
- Laboratory of Morpheme Analysis Imaging Research, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Kinya Nishimura
- Neurophysiology Research Group, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan; Department of Anesthesiology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Akihide Kondo
- Department of Neurosurgery, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hidenori Sugano
- Department of Neurosurgery, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hajime Arai
- Department of Neurosurgery, Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
8
|
Chen A, Hu WW, Jiang XL, Potegal M, Li H. Molecular mechanisms of group I metabotropic glutamate receptor mediated LTP and LTD in basolateral amygdala in vitro. Psychopharmacology (Berl) 2017; 234:681-694. [PMID: 28028604 DOI: 10.1007/s00213-016-4503-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 12/08/2016] [Indexed: 11/30/2022]
Abstract
The roles of group I metabotropic glutamate receptors, metabotropic glutamate receptor 1 (mGluR1) and mGluR5, in regulating synaptic plasticity and metaplasticity in the basolateral amygdala (BLA) remain unclear. The present study examined mGluR1- and mGluR5-mediated synaptic plasticity in the BLA and their respective signaling mechanisms. Bath application of the group I mGluR agonist, 3,5-dihydroxyphenylglycine (DHPG) (20 μM), directly suppressed basal fEPSPs (84.5 ± 6.3% of the baseline). The suppressive effect persisted for at least 30 min after washout; it was abolished by the mGluR1 antagonist 7-(hydroxyimino)cyclopropa[b]chromen-1a-carboxylate ethyl ester (CPCCOEt) but was unaffected by the mGluR5 antagonist 2-methyl-6- (phenylethynyl)-pyridine (MPEP). Interestingly, application of DHPG (at both 2 and 20 μM), regardless of the presence of CPCCOEt, could transform single theta burst stimulation (TBS)-induced short-term synaptic potentiation into a long-term potentiation (LTP). Such a facilitating effect could be blocked by the mGluR5 antagonist MPEP. Blockade of phospholipase C (PLC), the downstream enzyme of group I mGluR, with U73122, prevented both mGluR1- and mGluR5-mediated effects on synaptic plasticity. Nevertheless, blockade of protein kinase C (PKC), the downstream enzyme of PLC, with chelerythrine (5 μM) only prevented the transforming effect of DHPG on TBS-induced LTP and did not affect DHPG-induced long-term depression (LTD). These results suggest that mGluR1 activation induced LTD via a PLC-dependent and PKC-independent mechanism, while the priming action of mGluR5 receptor on the BLA LTP is both PLC and PKC dependent. The BLA metaplasticity mediated by mGluR1 and mGluR5 may provide signal switching mechanisms mediating learning and memory with emotional significance.
Collapse
Affiliation(s)
- A Chen
- Department of Physiology, Fujian Medical University, Fuzhou, People's Republic of China
| | - W W Hu
- Department of Physiology, Fujian Medical University, Fuzhou, People's Republic of China
| | - X L Jiang
- Department of Psychiatry, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, 20814-4799, USA
| | - M Potegal
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, 55414, USA
| | - H Li
- Department of Psychiatry, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, 20814-4799, USA.
| |
Collapse
|
9
|
Majewski L, Kuznicki J. SOCE in neurons: Signaling or just refilling? BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:1940-52. [DOI: 10.1016/j.bbamcr.2015.01.019] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2014] [Revised: 01/22/2015] [Accepted: 01/26/2015] [Indexed: 01/14/2023]
|
10
|
Nicotinic and muscarinic agonists and acetylcholinesterase inhibitors stimulate a common pathway to enhance GluN2B-NMDAR responses. Proc Natl Acad Sci U S A 2014; 111:12538-43. [PMID: 25114227 DOI: 10.1073/pnas.1408805111] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Nicotinic and muscarinic ACh receptor agonists and acetylcholinesterase inhibitors (AChEIs) can enhance cognitive function. However, it is unknown whether a common signaling pathway is involved in the effect. Here, we show that in vivo administration of nicotine, AChEIs, and an m1 muscarinic (m1) agonist increase glutamate receptor, ionotropic, N-methyl D-aspartate 2B (GluN2B)-containing NMDA receptor (NR2B-NMDAR) responses, a necessary component in memory formation, in hippocampal CA1 pyramidal cells, and that coadministration of the m1 antagonist pirenzepine prevents the effect of cholinergic drugs. These observations suggest that the effect of nicotine is secondary to increased release of ACh via the activation of nicotinic ACh receptors (nAChRs) and involves m1 receptor activation through ACh. In vitro activation of m1 receptors causes the selective enhancement of NR2B-NMDAR responses in CA1 pyramidal cells, and in vivo exposure to cholinergic drugs occludes the in vitro effect. Furthermore, in vivo exposure to cholinergic drugs suppresses the potentiating effect of Src on NMDAR responses in vitro. These results suggest that exposure to cholinergic drugs maximally stimulates the m1/guanine nucleotide-binding protein subunit alpha q/PKC/proline-rich tyrosine kinase 2/Src signaling pathway for the potentiation of NMDAR responses in vivo, occluding the in vitro effects of m1 activation and Src. Thus, our results indicate not only that nAChRs, ACh, and m1 receptors are on the same pathway involving Src signaling but also that NR2B-NMDARs are a point of convergence of cholinergic and glutamatergic pathways involved in learning and memory.
Collapse
|
11
|
Heraud-Farlow JE, Sharangdhar T, Li X, Pfeifer P, Tauber S, Orozco D, Hörmann A, Thomas S, Bakosova A, Farlow AR, Edbauer D, Lipshitz HD, Morris QD, Bilban M, Doyle M, Kiebler MA. Staufen2 regulates neuronal target RNAs. Cell Rep 2013; 5:1511-8. [PMID: 24360961 DOI: 10.1016/j.celrep.2013.11.039] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Revised: 11/04/2013] [Accepted: 11/21/2013] [Indexed: 11/30/2022] Open
Abstract
RNA-binding proteins play crucial roles in directing RNA translation to neuronal synapses. Staufen2 (Stau2) has been implicated in both dendritic RNA localization and synaptic plasticity in mammalian neurons. Here, we report the identification of functionally relevant Stau2 target mRNAs in neurons. The majority of Stau2-copurifying mRNAs expressed in the hippocampus are present in neuronal processes, further implicating Stau2 in dendritic mRNA regulation. Stau2 targets are enriched for secondary structures similar to those identified in the 3' UTRs of Drosophila Staufen targets. Next, we show that Stau2 regulates steady-state levels of many neuronal RNAs and that its targets are predominantly downregulated in Stau2-deficient neurons. Detailed analysis confirms that Stau2 stabilizes the expression of one synaptic signaling component, the regulator of G protein signaling 4 (Rgs4) mRNA, via its 3' UTR. This study defines the global impact of Stau2 on mRNAs in neurons, revealing a role in stabilization of the levels of synaptic targets.
Collapse
Affiliation(s)
- Jacki E Heraud-Farlow
- Department of Neuronal Cell Biology, Center for Brain Research, 1090 Vienna, Austria; Department of Chromosome Biology, Max F. Perutz Laboratories, University of Vienna, 1030 Vienna, Austria
| | - Tejaswini Sharangdhar
- Department of Anatomy and Cell Biology, Ludwig-Maximilians-University, 80336 Munich, Germany
| | - Xiao Li
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada; The Donnelly Centre, University of Toronto, Toronto, ON M5S 1E3, Canada
| | - Philipp Pfeifer
- Department of Neuronal Cell Biology, Center for Brain Research, 1090 Vienna, Austria
| | - Stefanie Tauber
- Department of Laboratory Medicine and Core Facility Genomics, Medical University of Vienna, 1090 Vienna, Austria
| | - Denise Orozco
- Adolf Butenandt Institute, Ludwig-Maximilians-University, 80336 Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), 80336 Munich, Germany
| | - Alexandra Hörmann
- Department of Neuronal Cell Biology, Center for Brain Research, 1090 Vienna, Austria
| | - Sabine Thomas
- Department of Neuronal Cell Biology, Center for Brain Research, 1090 Vienna, Austria; Department of Anatomy and Cell Biology, Ludwig-Maximilians-University, 80336 Munich, Germany
| | - Anetta Bakosova
- Department of Neuronal Cell Biology, Center for Brain Research, 1090 Vienna, Austria
| | - Ashley R Farlow
- Gregor Mendel Institute of Molecular Plant Biology, Austrian Academy of Sciences, 1030 Vienna, Austria
| | - Dieter Edbauer
- Adolf Butenandt Institute, Ludwig-Maximilians-University, 80336 Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), 80336 Munich, Germany; Munich Cluster of Systems Neurology (SyNergy), 80336 Munich, Germany
| | - Howard D Lipshitz
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Quaid D Morris
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada; The Donnelly Centre, University of Toronto, Toronto, ON M5S 1E3, Canada
| | - Martin Bilban
- Department of Laboratory Medicine and Core Facility Genomics, Medical University of Vienna, 1090 Vienna, Austria
| | - Michael Doyle
- Department of Neuronal Cell Biology, Center for Brain Research, 1090 Vienna, Austria.
| | - Michael A Kiebler
- Department of Neuronal Cell Biology, Center for Brain Research, 1090 Vienna, Austria; Department of Anatomy and Cell Biology, Ludwig-Maximilians-University, 80336 Munich, Germany.
| |
Collapse
|
12
|
Doolen S, Blake CB, Smith BN, Taylor BK. Peripheral nerve injury increases glutamate-evoked calcium mobilization in adult spinal cord neurons. Mol Pain 2012; 8:56. [PMID: 22839304 PMCID: PMC3490774 DOI: 10.1186/1744-8069-8-56] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Accepted: 07/05/2012] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Central sensitization in the spinal cord requires glutamate receptor activation and intracellular Ca2+ mobilization. We used Fura-2 AM bulk loading of mouse slices together with wide-field Ca2+ imaging to measure glutamate-evoked increases in extracellular Ca2+ to test the hypotheses that: 1. Exogenous application of glutamate causes Ca2+ mobilization in a preponderance of dorsal horn neurons within spinal cord slices taken from adult mice; 2. Glutamate-evoked Ca2+ mobilization is associated with spontaneous and/or evoked action potentials; 3. Glutamate acts at glutamate receptor subtypes to evoked Ca2+ transients; and 4. The magnitude of glutamate-evoked Ca2+ responses increases in the setting of peripheral neuropathic pain. RESULTS Bath-applied glutamate robustly increased [Ca2+]i in 14.4 ± 2.6 cells per dorsal horn within a 440 x 330 um field-of-view, with an average time-to-peak of 27 s and decay of 112 s. Repeated application produced sequential responses of similar magnitude, indicating the absence of sensitization, desensitization or tachyphylaxis. Ca2+ transients were glutamate concentration-dependent with a Kd = 0.64 mM. Ca2+ responses predominantly occurred on neurons since: 1) Over 95% of glutamate-responsive cells did not label with the astrocyte marker, SR-101; 2) 62% of fura-2 AM loaded cells exhibited spontaneous action potentials; 3) 75% of cells that responded to locally-applied glutamate with a rise in [Ca2+]i also showed a significant increase in AP frequency upon a subsequent glutamate exposure; 4) In experiments using simultaneous on-cell recordings and Ca2+ imaging, glutamate elicited a Ca2+ response and an increase in AP frequency. AMPA/kainate (CNQX)- and AMPA (GYKI 52466)-selective receptor antagonists significantly attenuated glutamate-evoked increases in [Ca2+]i, while NMDA (AP-5), kainate (UBP-301) and class I mGluRs (AIDA) did not. Compared to sham controls, peripheral nerve injury significantly decreased mechanical paw withdrawal threshold and increased glutamate-evoked Ca2+ signals. CONCLUSIONS Bulk-loading fura-2 AM into spinal cord slices is a successful means for determining glutamate-evoked Ca2+ mobilization in naïve adult dorsal horn neurons. AMPA receptors mediate the majority of these responses. Peripheral neuropathic injury potentiates Ca2+ signaling in dorsal horn.
Collapse
Affiliation(s)
- Suzanne Doolen
- Department of Physiology, School of Medicine, University of Kentucky Medical Center, Lexington, KY 40536, USA
| | | | | | | |
Collapse
|
13
|
Kato HK, Kassai H, Watabe AM, Aiba A, Manabe T. Functional coupling of the metabotropic glutamate receptor, InsP3 receptor and L-type Ca2+ channel in mouse CA1 pyramidal cells. J Physiol 2012; 590:3019-34. [PMID: 22586220 DOI: 10.1113/jphysiol.2012.232942] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Activity-dependent regulation of calcium dynamics in neuronal cells can play significant roles in the modulation of many cellular processes such as intracellular signalling, neuronal activity and synaptic plasticity. Among many calcium influx pathways into neurons, the voltage-dependent calcium channel (VDCC) is the major source of calcium influx, but its modulation by synaptic activity has still been under debate. While the metabotropic glutamate receptor (mGluR) is supposed to modulate L-type VDCCs (L-VDCCs), its reported actions include both facilitation and suppression, probably reflecting the uncertainty of both the molecular targets of the mGluR agonists and the source of the recorded calcium signal in previous reports. In this study, using subtype-specific knockout mice, we have shown that mGluR5 induces facilitation of the depolarization-evoked calcium current. This facilitation was not accompanied by the change in single-channel properties of the VDCC itself; instead, it required the activation of calcium-induced calcium release (CICR) that was triggered by VDCC opening, suggesting that the opening of CICR-coupled cation channels was essential for the facilitation. This facilitation was blocked or reduced by the inhibitors of both L-VDCCs and InsP3 receptors (InsP3Rs). Furthermore, L-VDCCs and mGluR5 were shown to form a complex by coimmunoprecipitation, suggesting that the specific functional coupling between mGluR5, InsP3Rs and L-VDCCs played a pivotal role in the calcium-current facilitation. Finally, we showed that mGluR5 enhanced VDCC-dependent long-term potentiation (LTP) of synaptic transmission. Our study has identified a novel mechanism of the interaction between the mGluR and calcium signalling, and suggested a contribution of mGluR5 to synaptic plasticity.
Collapse
Affiliation(s)
- Hiroyuki K Kato
- Division of Neuronal Network, Department of Basic Medical Sciences, Institute of Medical Science, University of Tokyo, Tokyo, 108-8639, Japan
| | | | | | | | | |
Collapse
|
14
|
Navarrete M, Araque A. Endocannabinoids potentiate synaptic transmission through stimulation of astrocytes. Neuron 2010; 68:113-26. [PMID: 20920795 DOI: 10.1016/j.neuron.2010.08.043] [Citation(s) in RCA: 347] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/17/2010] [Indexed: 12/31/2022]
Abstract
Endocannabinoids and their receptor CB1 play key roles in brain function. Astrocytes express CB1Rs that are activated by endocannabinoids released by neurons. However, the consequences of the endocannabinoid-mediated neuron-astrocyte signaling on synaptic transmission are unknown. We show that endocannabinoids released by hippocampal pyramidal neurons increase the probability of transmitter release at CA3-CA1 synapses. This synaptic potentiation is due to CB1R-induced Ca(2+) elevations in astrocytes, which stimulate the release of glutamate that activates presynaptic metabotropic glutamate receptors. While endocannabinoids induce synaptic depression in the stimulated neuron by direct activation of presynaptic CB1Rs, they indirectly lead to synaptic potentiation in relatively more distant neurons by activation of CB1Rs in astrocytes. Hence, astrocyte calcium signal evoked by endogenous stimuli (neuron-released endocannabinoids) modulates synaptic transmission. Therefore, astrocytes respond to endocannabinoids that then potentiate synaptic transmission, indicating that astrocytes are actively involved in brain physiology.
Collapse
Affiliation(s)
- Marta Navarrete
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | | |
Collapse
|
15
|
Opazo F, Schulz JB, Falkenburger BH. PKC links Gq-coupled receptors to DAT-mediated dopamine release. J Neurochem 2010; 114:587-96. [DOI: 10.1111/j.1471-4159.2010.06788.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
16
|
Thathiah A, De Strooper B. G protein-coupled receptors, cholinergic dysfunction, and Abeta toxicity in Alzheimer's disease. Sci Signal 2009; 2:re8. [PMID: 19843960 DOI: 10.1126/scisignal.293re8] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
The beta-amyloid (Abeta) peptide is associated with the pathogenesis of Alzheimer's disease (AD). Evidence gathered over the last two decades suggests that the gradual accumulation of soluble and insoluble Abeta peptide species triggers a cascade of events that leads to the clinical manifestation of AD. Abeta accumulation has also been associated with the cholinergic dysfunction observed in AD, which is characterized by diminished acetylcholine release and impaired coupling of the muscarinic acetylcholine receptors (mAChRs) to heterotrimeric GTP-binding proteins (G proteins). Although the mechanism of Abeta-mediated toxicity is not clearly understood, evidence shows that Abeta accumulation has an effect on the oligomerization of the angiotensin II (AngII) AT(2) (angiotensin type 2) receptor and sequestration of the Galpha(q/11) family of G proteins. Sequestration of Galpha(q/11) results in dysfunctional coupling and signaling between M(1) mAChR and Galpha(q/11) and accompanies neurodegeneration, tau phosphorylation, and neuronal loss in an AD transgenic mouse model. Collectively, these results provide a putative link among Abeta toxicity, AT(2) receptor oligomerization, and disruption of the signaling pathway through M(1) mAChR and Galpha(q/11) and potentially contribute to our understanding of the cholinergic deficit observed in AD.
Collapse
|
17
|
Abstract
The age of an experimental animal can be a critical variable, yet age matters are often overlooked within neuroscience. Many studies make use of young animals, without considering possible differences between immature and mature subjects. This is especially problematic when attempting to model traits or diseases that do not emerge until adulthood. In this commentary we discuss the reasons for this apparent bias in age of experimental animals, and illustrate the problem with a systematic review of published articles on long-term potentiation. Additionally, we review the developmental stages of a rat and discuss the difficulty of using the weight of an animal as a predictor of its age. Finally, we provide original data from our laboratory and review published data to emphasize that development is an ongoing process that does not end with puberty. Developmental changes can be quantitative in nature, involving gradual changes, rapid switches, or inverted U-shaped curves. Changes can also be qualitative. Thus, phenomena that appear to be unitary may be governed by different mechanisms at different ages. We conclude that selection of the age of the animals may be critically important in the design and interpretation of neurobiological studies.
Collapse
Affiliation(s)
- James Edgar McCutcheon
- Department of Cellular and Molecular Pharmacology, Rosalind Franklin University of Medicine and Science, The Chicago Medical School, 3333 Green Bay Road, North Chicago, IL 60064, USA
| | | |
Collapse
|
18
|
AbdAlla S, Lother H, el Missiry A, Sergeev P, Langer A, el Faramawy Y, Quitterer U. Dominant Negative AT2 Receptor Oligomers Induce G-protein Arrest and Symptoms of Neurodegeneration. J Biol Chem 2009; 284:6566-74. [DOI: 10.1074/jbc.m808277200] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
19
|
AbdAlla S, Lother H, el Missiry A, Langer A, Sergeev P, el Faramawy Y, Quitterer U. Angiotensin II AT2 receptor oligomers mediate G-protein dysfunction in an animal model of Alzheimer disease. J Biol Chem 2008; 284:6554-65. [PMID: 19074441 DOI: 10.1074/jbc.m807746200] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Progressive neurodegeneration and decline of cognitive functions are major hallmarks of Alzheimer disease (AD). Neurodegeneration in AD correlates with dysfunction of diverse signal transduction mechanisms, such as the G-protein-stimulated phosphoinositide hydrolysis mediated by Galphaq/11. We report here that impaired Galphaq/11-stimulated signaling in brains of AD patients and mice correlated with the appearance of cross-linked oligomeric angiotensin II AT2 receptors sequestering Galphaq/11. Amyloid beta (Abeta) was causal to AT2 oligomerization, because cerebral microinjection of Abeta triggered AT2 oligomerization in the hippocampus of mice in a dose-dependent manner. Abeta induced AT2 oligomerization by a two-step process of oxidative and transglutaminase-dependent cross-linking. The induction of AT2 oligomers in a transgenic mouse model with AD-like symptoms was associated with Galphaq/11 dysfunction and enhanced neurodegeneration. Vice versa, stereotactic inhibition of AT2 oligomers by RNA interference prevented the impairment of Galphaq/11 and delayed Tau phosphorylation. Thus, Abeta induces the formation of cross-linked AT2 oligomers that contribute to the dysfunction of Galphaq/11 in an animal model of Alzheimer disease.
Collapse
Affiliation(s)
- Said AbdAlla
- Heinrich-Pette-Institute, Martinistrasse 52, D-20251 Hamburg, Germany
| | | | | | | | | | | | | |
Collapse
|
20
|
Miura M, Masuda M, Aosaki T. Roles of micro-opioid receptors in GABAergic synaptic transmission in the striosome and matrix compartments of the striatum. Mol Neurobiol 2008; 37:104-15. [PMID: 18473190 DOI: 10.1007/s12035-008-8023-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2008] [Accepted: 04/17/2008] [Indexed: 10/22/2022]
Abstract
The striatum is divided into two compartments, the striosomes and extrastriosomal matrix, which differ in several cytochemical markers, input-output connections, and time of neurogenesis. Since it is thought that limbic, reward-related information and executive aspects of behavioral information may be differentially processed in the striosomes and matrix, respectively, intercompartmental communication should be of critical importance to proper functioning of the basal ganglia-thalamocortical circuits. Cholinergic interneurons are in a suitable position for this communication since they are preferentially located in the striosome-matrix boundaries and are known to elicit a conditioned pause response during sensorimotor learning. Recently, micro-opioid receptor (MOR) activation was found to presynaptically suppress the amplitude of GABAergic inhibitory postsynaptic currents in striosomal cells but not in matrix cells. Disinhibition of cells in the striosomes is further enhanced by inactivation of the protein kinase C cascade. We discuss in this review the possibility that MOR activation in the striosomes affects the activity of cholinergic interneurons and thus leads to changes in synaptic efficacy in the striatum.
Collapse
Affiliation(s)
- Masami Miura
- Neural Circuits Dynamics Research Group, Tokyo Metropolitan Institute of Gerontology, 35-2, Sakae-cho, Itabashi-ku, Tokyo 173-0015, Japan
| | | | | |
Collapse
|
21
|
Neyman S, Manahan-Vaughan D. Metabotropic glutamate receptor 1 (mGluR1) and 5 (mGluR5) regulate late phases of LTP and LTD in the hippocampal CA1 region in vitro. Eur J Neurosci 2008; 27:1345-52. [PMID: 18364018 PMCID: PMC2327219 DOI: 10.1111/j.1460-9568.2008.06109.x] [Citation(s) in RCA: 119] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The group I metabotropic glutamate receptors, mGluR1 and mGluR5, exhibit differences in their regulation of synaptic plasticity, suggesting that these receptors may subserve separate functional roles in information storage. In addition, although effects in vivo are consistently described, conflicting reports of the involvement of mGluRs in hippocampal synaptic plasticity in vitro exist. We therefore addressed the involvement of mGluR1 and mGluR5 in long-term potentiation (LTP) and long-term depression (LTD) in the hippocampal CA1 region of adult male rats in vitro. The mGluR1 antagonist (S)-(+)-α-amino-4-carboxy-2-methylbenzene-acetic acid (LY367385) impaired both induction and late phases of both LTP and LTD, when applied before high-frequency tetanization (HFT; 100 Hz) or low-frequency stimulation (LFS; 1 Hz), respectively. Application after either HFT or LFS had no effect. The mGluR5 antagonist 2-methyl-6-(phenylethynyl)pyridine (MPEP), when given before HFT, inhibited both the induction and late phases of LTP. When given after HFT, late LTP was inhibited. MPEP, given prior to LFS, impaired LTD induction, although stable LTD was still expressed. Application after LFS significantly impaired late phases of LTD. Activation of protein synthesis may comprise a key mechanism underlying the group I mGluR contribution to synaptic plasticity. The mGluR5 agonist (R,S)-2-chloro-5-hydroxyphenylglycine (CHPG) converted short-term depression into LTD. Effects were prevented by application of the protein synthesis inhibitor anisomycin, suggesting that protein synthesis is triggered by group I mGluR activation to enable persistency of synaptic plasticity. Taken together, these data support the notion that both mGluR1 and mGluR5 are critically involved in bidirectional synaptic plasticity in the CA1 region and may enable functional differences in information encoding through LTP and LTD.
Collapse
Affiliation(s)
- Sergey Neyman
- Institute for Physiology of the Charité, Synaptic Plasticity Research Group, Humboldt University, Berlin, Germany
| | | |
Collapse
|
22
|
Jenkins TC, Andrews JB, Meyer-Bernstein EL. Daily oscillation of phospholipase C beta4 in the mouse suprachiasmatic nucleus. Brain Res 2007; 1178:83-91. [PMID: 17920566 DOI: 10.1016/j.brainres.2007.07.098] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2007] [Revised: 07/06/2007] [Accepted: 07/08/2007] [Indexed: 11/26/2022]
Abstract
An endogenous biological clock located in the hypothalamic suprachiasmatic nucleus (SCN) regulates the timing of an organism's physiology and behavior. A variety of receptors are found on SCN pacemaker cells which permit the clock mechanism to respond to extra- and intra-SCN chemical messengers. A subset of these receptors is coupled to G-proteins, which when bound, lead to the activation of a variety of intracellular signaling cascades. One common signaling pathway employs the phosphotidylinositol-specific phospholipase C enzyme to increase intracellular calcium levels. A specific isoform of this enzyme, phospholipase C beta4, is of particular interest to circadian biologists because in its absence, mice display a circadian phenotype. Moreover, it has been shown to be associated with receptor types that are involved in clock resetting. Despite compelling data that this enzyme could be a critical component of an intracellular signaling pathway in the SCN, no study to date has investigated the possible oscillation of phospholipase C in any mammalian tissue. In the present study, we analyzed the temporal variation in the number of phospholipase C beta4 immunoreactive cells in the SCN. Herein, we show that PLCbeta4 levels oscillate in the SCN of mice housed in a light:dark photoperiod. Protein levels reached a significant peak during the early night and a trough during the day. The oscillation was considerably damped in the SCN of mice housed in constant dark conditions indicating the cycle is photoperiod-dependent. These data are critical to understanding the temporal regulation of a variety of inputs to the mammalian central circadian clock.
Collapse
Affiliation(s)
- Travis C Jenkins
- Department of Biology, College of Charleston, 66 George Street, Charleston, SC 29424, USA
| | | | | |
Collapse
|
23
|
Fowler MA, Sidiropoulou K, Ozkan ED, Phillips CW, Cooper DC. Corticolimbic expression of TRPC4 and TRPC5 channels in the rodent brain. PLoS One 2007; 2:e573. [PMID: 17593972 PMCID: PMC1892805 DOI: 10.1371/journal.pone.0000573] [Citation(s) in RCA: 135] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2007] [Accepted: 05/30/2007] [Indexed: 11/19/2022] Open
Abstract
The canonical transient receptor potential (TRPC) channels are a family of non-selective cation channels that are activated by increases in intracellular Ca(2+) and G(q)/phospholipase C-coupled receptors. We used quantitative real-time PCR, in situ hybridization, immunoblots and patch-clamp recording from several brain regions to examine the expression of the predominant TRPC channels in the rodent brain. Quantitative real-time PCR of the seven TRPC channels in the rodent brain revealed that TRPC4 and TRPC5 channels were the predominant TRPC subtypes in the adult rat brain. In situ hybridization histochemistry and immunoblotting further resolved a dense corticolimbic expression of the TRPC4 and TRPC5 channels. Total protein expression of HIP TRPC4 and 5 proteins increased throughout development and peaked late in adulthood (6-9 weeks). In adults, TRPC4 expression was high throughout the frontal cortex, lateral septum (LS), pyramidal cell layer of the hippocampus (HIP), dentate gyrus (DG), and ventral subiculum (vSUB). TRPC5 was highly expressed in the frontal cortex, pyramidal cell layer of the HIP, DG, and hypothalamus. Detailed examination of frontal cortical layer mRNA expression indicated TRPC4 mRNA is distributed throughout layers 2-6 of the prefrontal cortex (PFC), motor cortex (MCx), and somatosensory cortex (SCx). TRPC5 mRNA expression was concentrated specifically in the deep layers 5/6 and superficial layers 2/3 of the PFC and anterior cingulate. Patch-clamp recording indicated a strong metabotropic glutamate-activated cation current-mediated depolarization that was dependent on intracellular Ca(2+)and inhibited by protein kinase C in brain regions associated with dense TRPC4 or 5 expression and absent in regions lacking TRPC4 and 5 expression. Overall, the dense corticolimbic expression pattern suggests that these Gq/PLC coupled nonselective cation channels may be involved in learning, memory, and goal-directed behaviors.
Collapse
Affiliation(s)
- Melissa A. Fowler
- Psychiatry Department, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Kyriaki Sidiropoulou
- Computational Biology Lab, Institute of Molecular Biology and Biotechnology (IMBB)-Foundation for Research and Technology (FORTH), Vassilika Vouton, Heraklio, Greece
| | - Emin D. Ozkan
- Psychiatry Department, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Christopher W. Phillips
- Psychiatry Department, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Donald C. Cooper
- Psychiatry Department, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
24
|
Zhu PJ, Lovinger DM. Persistent Synaptic Activity Produces Long-Lasting Enhancement of Endocannabinoid Modulation and Alters Long-Term Synaptic Plasticity. J Neurophysiol 2007; 97:4386-9. [PMID: 17392410 DOI: 10.1152/jn.01228.2006] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Learning and memory are thought to involve activity-dependent changes in synaptic efficacy such as long-term potentiation (LTP) and long-term depression (LTD). Recent studies have indicated that endocannabinoid-dependent modulation of inhibitory transmission facilitates induction of hippocampal LTP and that endocannabinoids play a key role in certain forms of LTD. Here, we show that repetitive low-frequency synaptic stimulation (LFS) produces persistent up-regulation of endocannabinoid signaling at hippocampal CA1 GABAergic synapses. This LFS also produces LTD of inhibitory synapses and facilitates LTP at excitatory, glutamatergic synapses. These endocannabinoid-mediated plastic changes could contribute to information storage within the brain.
Collapse
Affiliation(s)
- Ping Jun Zhu
- Lab. for Integrative Neuroscience, NIH/NIAAA, 5625 Fishers Lane, Rm. TS-28, Bethesda, MD 20892-9411, USA
| | | |
Collapse
|
25
|
Desai NS, Casimiro TM, Gruber SM, Vanderklish PW. Early Postnatal Plasticity in Neocortex of Fmr1 Knockout Mice. J Neurophysiol 2006; 96:1734-45. [PMID: 16823030 DOI: 10.1152/jn.00221.2006] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Fragile X syndrome is produced by a defect in a single X-linked gene, called Fmr1, and is characterized by abnormal dendritic spine morphologies with spines that are longer and thinner in neocortex than those from age-matched controls. Studies using Fmr1 knockout mice indicate that spine abnormalities are especially pronounced in the first month of life, suggesting that altered developmental plasticity underlies some of the behavioral phenotypes associated with the syndrome. To address this issue, we used intracellular recordings in neocortical slices from early postnatal mice to examine the effects of Fmr1 disruption on two forms of plasticity active during development. One of these, long-term potentiation of intrinsic excitability, is intrinsic in expression and requires mGluR5 activation. The other, spike timing-dependent plasticity, is synaptic in expression and requires N-methyl-d-aspartate receptor activation. While intrinsic plasticity was normal in the knockout mice, synaptic plasticity was altered in an unusual and striking way: long-term depression was robust but long-term potentiation was entirely absent. These findings underscore the ideas that Fmr1 has highly selective effects on plasticity and that abnormal postnatal development is an important component of the disorder.
Collapse
MESH Headings
- Action Potentials/genetics
- Action Potentials/physiology
- Animals
- Animals, Newborn/genetics
- Animals, Newborn/physiology
- Fragile X Mental Retardation Protein/genetics
- Fragile X Mental Retardation Protein/physiology
- Fragile X Syndrome/genetics
- Fragile X Syndrome/physiopathology
- Gene Expression Regulation, Developmental/genetics
- Gene Expression Regulation, Developmental/physiology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Neocortex/growth & development
- Neocortex/physiology
- Neuronal Plasticity/genetics
- Neuronal Plasticity/physiology
- Receptor, Metabotropic Glutamate 5
- Receptors, Metabotropic Glutamate/genetics
- Receptors, Metabotropic Glutamate/physiology
- Receptors, N-Methyl-D-Aspartate/genetics
- Receptors, N-Methyl-D-Aspartate/physiology
- Synapses/genetics
- Synapses/physiology
- Synaptic Transmission/genetics
- Synaptic Transmission/physiology
Collapse
Affiliation(s)
- Niraj S Desai
- The Neurosciences Fine Institute, San Diego, CA 92121, USA.
| | | | | | | |
Collapse
|
26
|
Wettschureck N, van der Stelt M, Tsubokawa H, Krestel H, Moers A, Petrosino S, Schütz G, Di Marzo V, Offermanns S. Forebrain-specific inactivation of Gq/G11 family G proteins results in age-dependent epilepsy and impaired endocannabinoid formation. Mol Cell Biol 2006; 26:5888-94. [PMID: 16847339 PMCID: PMC1592765 DOI: 10.1128/mcb.00397-06] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Metabotropic receptors coupled to Gq/G11 family G proteins critically contribute to nervous system functions by modulating synaptic transmission, often facilitating excitation. We investigated the role of Gq/G11 family G proteins in the regulation of neuronal excitability in mice that selectively lack the alpha-subunits of Gq and G11, G alpha q and G alpha 11, respectively, in forebrain principal neurons. Surprisingly, mutant mice exhibited increased seizure susceptibility, and the activation of neuroprotective mechanisms was impaired. We found that endocannabinoid levels were reduced under both basal and excitotoxic conditions and that increased susceptibility to kainic acid could be normalized by the enhancement of endocannabinoid levels with an endocannabinoid reuptake inhibitor, while the competitive cannabinoid type 1 receptor antagonist SR141716A did not cause further aggravation. These findings indicate that Gq/G11 family G proteins negatively regulate neuronal excitability in vivo and suggest that impaired endocannabinoid formation in the absence of Gq/G11 contributes to this phenotype.
Collapse
Affiliation(s)
- Nina Wettschureck
- Institute of Pharmacology, University of Heidelberg, Im Neuenheimer Feld 366, 69120 Heidelberg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Abstract
A number of neuronal functions, including synaptic plasticity, depend on proper regulation of synaptic proteins, many of which can be rapidly regulated by phosphorylation. Neuronal activity controls the function of these synaptic proteins by exquisitely regulating the balance of various protein kinase and protein phosphatase activity. Recent understanding of synaptic plasticity mechanisms underscores important roles that these synaptic phosphoproteins play in regulating both pre- and post-synaptic functions. This review will focus on key postsynaptic phosphoproteins that have been implicated to play a role in synaptic plasticity.
Collapse
Affiliation(s)
- Hey-Kyoung Lee
- Department of Biology, Neuroscience and Cognitive Science (NACS) Program, University of Maryland, College Park, MD 20742, USA.
| |
Collapse
|
28
|
Ferraguti F, Shigemoto R. Metabotropic glutamate receptors. Cell Tissue Res 2006; 326:483-504. [PMID: 16847639 DOI: 10.1007/s00441-006-0266-5] [Citation(s) in RCA: 400] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2006] [Accepted: 05/31/2006] [Indexed: 12/22/2022]
Abstract
Metabotropic glutamate receptors (mGlus) are a family of G-protein-coupled receptors activated by the neurotransmitter glutamate. Molecular cloning has revealed eight different subtypes (mGlu1-8) with distinct molecular and pharmacological properties. Multiplicity in this receptor family is further generated through alternative splicing. mGlus activate a multitude of signalling pathways important for modulating neuronal excitability, synaptic plasticity and feedback regulation of neurotransmitter release. In this review, we summarize anatomical findings (from our work and that of other laboratories) describing their distribution in the central nervous system. Recent evidence regarding the localization of these receptors in peripheral tissues will also be examined. The distinct regional, cellular and subcellular distribution of mGlus in the brain will be discussed in view of their relationship to neurotransmitter release sites and of possible functional implications.
Collapse
Affiliation(s)
- Francesco Ferraguti
- Department of Pharmacology, Innsbruck Medical University, Peter Mayr Strasse 1a, A-6020, Innsbruck, Austria
| | | |
Collapse
|
29
|
Alfonso J, Frick LR, Silberman DM, Palumbo ML, Genaro AM, Frasch AC. Regulation of hippocampal gene expression is conserved in two species subjected to different stressors and antidepressant treatments. Biol Psychiatry 2006; 59:244-51. [PMID: 16140276 DOI: 10.1016/j.biopsych.2005.06.036] [Citation(s) in RCA: 145] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2005] [Revised: 06/10/2005] [Accepted: 06/30/2005] [Indexed: 10/25/2022]
Abstract
BACKGROUND Chronic stress has significant effects on hippocampal structure and function. We have previously identified nerve growth factor (NGF), membrane glycoprotein 6a (M6a), the guanine nucleotide binding protein (G protein) alpha q polypeptide (GNAQ), and CDC-like kinase 1 (CLK-1) as genes regulated by psychosocial stress and clomipramine treatment in the hippocampus of tree shrews. These genes encode proteins involved in neurite outgrowth. METHODS To analyze whether regulation of the above-mentioned genes is conserved between different species, stressors, and antidepressant drugs, we subjected mice to repeated restraint stress and tianeptine treatment and measured hippocampal messenger RNA (mRNA) levels by real time reverse transcription polymerase chain reaction (RT-PCR). RESULTS Chronically stressed mice displayed a reduction in transcript levels for NGF, M6a, GNAQ, and CLK-1. In addition, other genes implicated in neuronal plasticity, such as brain-derived neurotrophic factor (BDNF), cyclic adenosine monophosphate (cAMP) response element binding protein (CREB), protein kinase C (PKC), neural cell adhesion molecule (NCAM), and synapsin I were downregulated in stressed mice. Tianeptine treatment reversed the stress effects for the genes analyzed. Alterations in gene expression were dependent on the duration of the stress treatment and, in some cases, were only observed in male mice. CONCLUSIONS These results suggest that genes involved in neurite remodeling are one of the main targets for regulation by chronic stress. The finding that this regulation is conserved in different stress models and antidepressant treatments highlights the biological relevance of the genes analyzed and suggests that they might be involved in stress-related disorders.
Collapse
Affiliation(s)
- Julieta Alfonso
- IIB-INTECH-Universidad Nacional de Gral, San Martin, Argentina.
| | | | | | | | | | | |
Collapse
|
30
|
Kammermeier PJ. Surface clustering of metabotropic glutamate receptor 1 induced by long Homer proteins. BMC Neurosci 2006; 7:1. [PMID: 16393337 PMCID: PMC1361788 DOI: 10.1186/1471-2202-7-1] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2005] [Accepted: 01/04/2006] [Indexed: 12/02/2022] Open
Abstract
Background Metabotropic glutamate receptors (mGluRs) regulate neuronal excitability and synaptic strength. The group I mGluRs, mGluR1 and 5, are widespread in the brain and localize to post-synaptic sites. The Homer protein family regulates group I mGluR function and distribution. Constitutively expressed 'long' Homer proteins (Homer 1b, 1c, 2 and 3) induce dendritic localization of group I mGluRs and receptor clustering, either internally or on the plasma membrane. Short Homer proteins (Homer 1a, Ania-3) exhibit regulated expression and act as dominant negatives, producing effects on mGluR distribution and function that oppose those of the long Homer proteins. There remains some controversy over whether long Homer proteins induce receptor internalization by inducing retention in the endoplasmic reticulum, or induce mGluR clustering on the plasma membrane. Further, an exhaustive study of the effects of each long Homer isoform on mGluR distribution has not been published. Results The distribution of a GFP-tagged group I mGluR, mGluR1-GFP, was examined in the absence of Homer proteins and in the presence of several Homer isoforms expressed in sympathetic neurons from the rat superior cervical ganglion (SCG) using total internal reflection fluorescence (TIRF-M) and confocal microscopy. Quantitative analysis of mGluR1-GFP fluorescence using TIRF-M revealed that expression of each long Homer isoform tested (Homer 1b, 1c, 2b and 3) induced a significant degree of surface clustering. Using confocal imaging, Homer-induced mGluR clusters were observed intra-cellularly as well as on the plasma membrane. Further, in approximately 40% of neurons co-expressing mGluR1-GFP and Homer 1b, intracellular inclusions were observed, but plasma membrane clusters were also documented in some Homer 1b coexpressing cells. Conclusion All long Homer proteins examined (Homer 1b, 1c, 2b and 3) induced a significant degree of mGluR1-GFP clustering on the plasma membrane compared to cells expressing mGluR1-GFP alone. Clusters induced by long Homers appeared on the plasma membrane and intracellularly, suggesting that clusters form prior to plasma membrane insertion and/or persist after internalization. Finally, while Homer 1b induced surface clustering of mGluR1 in some cells, under some conditions intracellular retention may occur.
Collapse
Affiliation(s)
- Paul J Kammermeier
- Department of Physiology and Pharmacology, Northeastern Ohio Universities College of Medicine, Rootstown, OH 44272, USA.
| |
Collapse
|
31
|
Abstract
Heterotrimeric G proteins are key players in transmembrane signaling by coupling a huge variety of receptors to channel proteins, enzymes, and other effector molecules. Multiple subforms of G proteins together with receptors, effectors, and various regulatory proteins represent the components of a highly versatile signal transduction system. G protein-mediated signaling is employed by virtually all cells in the mammalian organism and is centrally involved in diverse physiological functions such as perception of sensory information, modulation of synaptic transmission, hormone release and actions, regulation of cell contraction and migration, or cell growth and differentiation. In this review, some of the functions of heterotrimeric G proteins in defined cells and tissues are described.
Collapse
Affiliation(s)
- Nina Wettschureck
- Institute of Pharmacology, University of Heidelberg, Im Neuenheimer Feld 366, D-69120 Heidelberg, Germany
| | | |
Collapse
|
32
|
Cumbay MG, Watts VJ. Galphaq potentiation of adenylate cyclase type 9 activity through a Ca2+/calmodulin-dependent pathway. Biochem Pharmacol 2005; 69:1247-56. [PMID: 15794946 DOI: 10.1016/j.bcp.2005.02.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2005] [Revised: 02/02/2005] [Accepted: 02/02/2005] [Indexed: 10/25/2022]
Abstract
Adenylate cyclase (EC 4.6.1.1) type 9 (AC9) activity has been shown to be inhibited by PMA activation of novel protein kinase C (nPKC) isoforms. In the current study the effect on AC9 activity of activating PKC in physiological relevant manner was examined. Contrary to the anticipated inhibitory effect of activating PKCs through Gq-coupled receptors, activation of transiently expressed Gq-coupled serotonin 5-HT2A or muscarinic M5 receptors resulted in the potentiation of isoproterenol-stimulated cyclic AMP accumulation in HEK293 cells stably expressing AC9 (HEK-AC9). Consistent with Gq-mediated activation of PKC, the addition of the PKC inhibitor bisindolylmaleimide further potentiated isoproterenol-stimulated cyclic AMP accumulation. Expression of a constitutively active mutant of Galphaq in HEK-AC9 cells also produced an enhancement in basal and isoproterenol-stimulated cyclic AMP accumulation. We also examined the role of Galphaq-mediated release of intracellular Ca2+ on the observed potentiation of AC9 activity, by depleting intracellular Ca2+ stores with thapsigargin. In Ca2+-depleted HEK-AC9 cells, activation of transiently expressed M5 receptors resulted in inhibition of isoproterenol-stimulated cyclic AMP accumulation that was blocked by bisindolylmaleimide, indicating that M5 potentiation of AC9 activity requires Ca2+. This prompted us to examine the effects of the calmodulin antagonist W7 and the Ca2+/calmodulin-dependent kinase II (CaMK II) inhibitor KN-93. Pretreating cells with W7 and KN-93 significantly inhibited M5-mediated potentiation of isoproterenol-stimulated cyclic AMP accumulation in HEK-AC9 cells, suggesting that Galphaq potentiation of AC9 activity involves Ca2+/calmodulin and CaMK II. This data provides evidence for Ca2+-mediated potentiation of AC9 activity.
Collapse
Affiliation(s)
- Medhane G Cumbay
- Purdue University, Department of Medicinal Chemistry and Molecular Pharmacology, 575 Stadium Mall Drive, West Lafayette, IN 47907-2051, USA
| | | |
Collapse
|
33
|
Chen SR, Pan HL. Distinct roles of group III metabotropic glutamate receptors in control of nociception and dorsal horn neurons in normal and nerve-injured Rats. J Pharmacol Exp Ther 2005; 312:120-6. [PMID: 15375175 DOI: 10.1124/jpet.104.073817] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Increased glutamatergic input to spinal dorsal horn neurons constitutes an important mechanism for neuropathic pain. However, the role of group III metabotropic glutamate receptors (mGluRs) in regulation of nociception and dorsal horn neurons in normal and neuropathic pain conditions is not fully known. In this study, we determined the effect of the group III mGluR specific agonist L(+)-2-amino-4-phosphonobutyric acid (L-AP4) on nociception and dorsal horn projection neurons in normal rats and a rat model of neuropathic pain. Tactile allodynia was induced by ligation of L5/L6 left spinal nerves in rats. Allodynia was determined by von Frey filaments in nerve-injured rats. The nociceptive threshold was tested using a radiant heat and a Randall-Selitto pressure device in normal rats. Single-unit activity of ascending dorsal horn neurons was recorded from the lumbar spinal cord in anesthetized rats. An intrathecal (5-30 microg) L-AP4 dose-dependently attenuated allodynia in nerve-injured rats but had no antinociceptive effect in normal rats. Topical spinal application of 5 to 50 microM L-AP4 also significantly inhibited the evoked responses of ascending dorsal horn neurons in nerve-ligated but not normal rats. Furthermore, blockade of spinal group III mGluRs significantly decreased the withdrawal threshold and increased the evoked responses of dorsal horn neurons in normal but not nerve-injured rats. These data suggest that group III mGluRs play distinct roles in regulation of nociception and dorsal horn neurons in normal and neuropathic pain states. Activation of spinal group III mGluRs suppresses allodynia and inhibits the hypersensitivity of dorsal horn projection neurons associated with neuropathic pain.
Collapse
Affiliation(s)
- Shao-Rui Chen
- Department of Anesthesiology, Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA 17033, USA
| | | |
Collapse
|
34
|
Chevaleyre V, Castillo PE. Endocannabinoid-Mediated Metaplasticity in the Hippocampus. Neuron 2004; 43:871-81. [PMID: 15363397 DOI: 10.1016/j.neuron.2004.08.036] [Citation(s) in RCA: 232] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2004] [Revised: 07/29/2004] [Accepted: 08/26/2004] [Indexed: 10/26/2022]
Abstract
Repetitive activation of glutamatergic fibers that normally induces long-term potentiation (LTP) at excitatory synapses in the hippocampus also triggers long-term depression at inhibitory synapses (I-LTD) via retrograde endocannabinoid signaling. Little is known, however, about the physiological significance of I-LTD. Here, we show that synaptic-driven release of endocannabinoids is a highly localized and efficient process that strongly depresses cannabinoid-sensitive inhibitory inputs within the dendritic compartment of CA1 pyramidal cells. By removing synaptic inhibition in a restricted area of the dendritic tree, endocannabinoids selectively "primed" nearby excitatory synapses, thereby facilitating subsequent induction of LTP. This induction of local metaplasticity is a novel mechanism by which endocannabinoids can contribute to the storage of information in the brain.
Collapse
Affiliation(s)
- Vivien Chevaleyre
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | |
Collapse
|
35
|
Abstract
The G-protein-mediated signaling system has evolved as one of the most widely used transmembrane signaling mechanisms in eukaryotic organisms. Mammalian cells express many G-protein-coupled receptors as well as several types of heterotrimeric G-proteins and effectors. This review focuses on recent data from studies in mutant mice, which have elucidated some of the roles of G-protein-mediated signaling in physiology and pathophysiology.
Collapse
Affiliation(s)
- Nina Wettschureck
- Institute of Pharmacology, University of Heidelberg, Im Neuenheimer Feld 366, 69120, Heidelberg, Germany.
| | | | | |
Collapse
|
36
|
Kobayashi K, Poo MM. Spike Train Timing-Dependent Associative Modification of Hippocampal CA3 Recurrent Synapses by Mossy Fibers. Neuron 2004; 41:445-54. [PMID: 14766182 DOI: 10.1016/s0896-6273(03)00873-0] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2003] [Revised: 09/26/2003] [Accepted: 12/05/2003] [Indexed: 11/26/2022]
Abstract
In the CA3 region of the hippocampus, extensive recurrent associational/commissural (A/C) connections made by pyramidal cells may function as a network for associative memory storage and recall. We here report that long-term potentiation (LTP) at the A/C synapses can be induced by association of brief spike trains in mossy fibers (MFs) from the dentate gyrus and A/C fibers. This LTP not only required substantial overlap between spike trains in MFs and A/C fibers, but also depended on the temporal order of these spike trains in a manner not predicted by the well-known rule of spike timing-dependent plasticity and requiring activation of type 1 metabotropic glutamate receptors. Importantly, spike trains in a putative single MF input provided effective postsynaptic activity for the induction of LTP at A/C synapses. Thus, the timing of spike trains in individual MFs may code information that is crucial for the associative modification of CA3 recurrent synapses.
Collapse
Affiliation(s)
- Katsunori Kobayashi
- Division of Neurobiology, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | | |
Collapse
|
37
|
Offermanns S. G-proteins as transducers in transmembrane signalling. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2003; 83:101-30. [PMID: 12865075 DOI: 10.1016/s0079-6107(03)00052-x] [Citation(s) in RCA: 201] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The G-protein-mediated signalling system has evolved as one of the most widely used transmembrane signalling mechanisms in mammalian organisms. All mammalian cells express G-protein-coupled receptors as well as several types of heterotrimeric G-proteins and effectors. G-protein-mediated signalling is involved in many physiological and pathological processes. This review summarizes some general aspects of G-protein-mediated signalling and focusses on recent data especially from studies in mutant mice which have elucidated some of the cellular and biological functions of heterotrimeric G-prtoteins.
Collapse
Affiliation(s)
- Stefan Offermanns
- Institute of Pharmacology, University of Heidelberg, Im Neuenheimer Feld 366, 69120 Heidelberg, Germany.
| |
Collapse
|
38
|
Higashida H, Zhang JS, Mochida S, Chen XL, Shin Y, Noda M, Hossain KZ, Hoshi N, Hashii M, Shigemoto R, Nakanishi S, Fukuda Y, Yokoyama S. Subtype-specific coupling with ADP-ribosyl cyclase of metabotropic glutamate receptors in retina, cervical superior ganglion and NG108-15 cells. J Neurochem 2003; 85:1148-58. [PMID: 12753074 DOI: 10.1046/j.1471-4159.2003.01751.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Cyclic ADP-ribose (cADP-ribose) is a putative second messenger or modulator. However, the role of cADP-ribose in the downstream signals of the metabotropic glutamate receptors (mGluRs) is unclear. Here, we show that glutamate stimulates ADP-ribosyl cyclase activity in rat or mouse crude membranes of retina via group III mGluRs or in superior cervical ganglion via group I mGluRs. The retina of mGluR6-deficient mice showed no increase in the ADP-ribosyl cyclase level in response to glutamate. GTP enhanced the initial rate of basal and glutamate-stimulated cyclase activity. GTP-gamma-S also stimulated basal activity. To determine whether the coupling mode of mGluRs to ADP-ribosyl cyclase is a feature common to individual cloned mGluRs, we expressed each mGluR subtype in NG108-15 neuroblastoma x glioma hybrid cells. The glutamate-induced stimulation of the cyclase occurs preferentially in NG108-15 cells over-expressing mGluRs1, 3, 5, and 6. Cells expressing mGluR2 or mGluRs4 and 7 exhibit inhibition or no coupling, respectively. Glutamate-induced activation or inhibition of the cyclase activity was eliminated after pre-treatment with cholera or pertussis toxin, respectively. Thus, the subtype-specific coupling of mGluRs to ADP-ribosyl cyclase via G proteins suggests that some glutamate-evoked neuronal functions are mediated by cADP-ribose.
Collapse
Affiliation(s)
- Haruhiro Higashida
- Department of Biophysical Genetics, Kanazawa University Graduate School of Medicine, Kanazawa, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Vigh J, Lasater EM. Intracellular calcium release resulting from mGluR1 receptor activation modulates GABAA currents in wide-field retinal amacrine cells: a study with caffeine. Eur J Neurosci 2003; 17:2237-48. [PMID: 12814357 DOI: 10.1046/j.1460-9568.2003.02652.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The modulatory action of calcium (Ca2+) released from intracellular stores on GABAA receptor-mediated current was investigated in wide-field amacrine cells isolated from the teleost, Morone chrysops, retina. Caffeine, ryanodine or inositol 1,4,5-triphosphate (IP3) markedly inhibited the GABAA current by elevating [Ca2+]i. The inhibition resulted from the activation of a Ca2+--> Ca2+/calmodulin --> calcineurin cascade. Long (>12 s) exposure to glutamate mimicked the caffeine effect, i.e. it inhibited the GABAA current by elevating [Ca2+]i through mGluR1 receptor activation and consequent IP3 generation. This pathway provides a 'timed' disinhibitory mechanism to potentiate excitatory postsynaptic potentials in wide-field amacrine cells. It occurs as a result of the suppression of GABA-mediated conductances as a function of the duration of presynaptic excitatory input activity. This is much like some forms of long-term potentiation in the central nervous system. In a local retinal circuit this will selectively accentuate particular excitatory inputs to the wide-field amacrine cell. Similar to other neural systems, we suggest that activity-dependent postsynaptic disinhibition is an important feature of the signal processing in the inner retina.
Collapse
MESH Headings
- Amacrine Cells/drug effects
- Amacrine Cells/physiology
- Animals
- Anticoagulants/pharmacology
- Bicuculline/pharmacology
- Caffeine/pharmacology
- Calcium/metabolism
- Calcium Channels/metabolism
- Carps
- Cells, Cultured
- Central Nervous System Stimulants/pharmacology
- Chelating Agents/pharmacology
- Dose-Response Relationship, Drug
- Drug Interactions
- Egtazic Acid/analogs & derivatives
- Egtazic Acid/pharmacology
- Electric Conductivity
- Enzyme Inhibitors/pharmacology
- Excitatory Amino Acid Agonists/pharmacology
- Extracellular Space/metabolism
- GABA Antagonists/pharmacology
- Glutamic Acid/physiology
- Heparin/pharmacology
- Immunohistochemistry
- Inositol 1,4,5-Trisphosphate/pharmacology
- Inositol 1,4,5-Trisphosphate Receptors
- Kainic Acid/pharmacology
- Membrane Potentials/drug effects
- Methoxyhydroxyphenylglycol/analogs & derivatives
- Methoxyhydroxyphenylglycol/pharmacology
- Organophosphorus Compounds/pharmacology
- Patch-Clamp Techniques/methods
- Receptors, Cytoplasmic and Nuclear/metabolism
- Receptors, GABA-A/physiology
- Receptors, Metabotropic Glutamate/antagonists & inhibitors
- Receptors, Metabotropic Glutamate/metabolism
- Retina/cytology
- Ryanodine/pharmacology
- alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/pharmacology
- gamma-Aminobutyric Acid/pharmacology
Collapse
Affiliation(s)
- Jozsef Vigh
- Dept. of Ophthalmology and Visual Sciences, John Moran Eye Center, University of Utah, Health Sciences Center, Salt Lake City, UT 84132, USA
| | | |
Collapse
|