1
|
Rahiminezhad Seta R, Eftekhari Mahabadi S, Delphi L, Alijanpour S, Rezayof A. Hippocampal nicotinic acetylcholine receptor signaling mediates the anti-allodynic effect of ketamine and morphine on neuropathic pain. Neuroscience 2025; 565:138-147. [PMID: 39615650 DOI: 10.1016/j.neuroscience.2024.11.067] [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: 09/09/2024] [Revised: 11/21/2024] [Accepted: 11/25/2024] [Indexed: 12/08/2024]
Abstract
The present study investigated the involvement of hippocampal nicotinic acetylcholine receptors (nAChRs) in the anti-allodynic effect of ketamine/morphine on neuropathic pain in adult male Wistar rats. Morphine or ketamine administration decreased the percentage of maximum possible effect (MPE%), indicating an analgesic effect. The most significant decrease occurred with a 5 mg/kg dose of morphine (average MPE% = 98), while a 0.5 mg/kg dose of ketamine resulted in a high response (average MPE% = 91), using decision trees as a machine learning tool. Combining morphine and ketamine improved neuropathic pain (average MPE% = 91). Intra-CA1 microinjection of mecamylamine (2 μg/rat) with morphine (3 mg/kg) reduced neuropathic pain (average MPE% = 94). Co-administration of lower doses of ketamine (0.1 mg/kg, i.p.) and mecamylamine (0.5 or 1 μg/rat) with morphine (3 mg/kg) led to a considerable reduction in pain (average MPE% = 91). Utilizing the generalized least squares (GLS) model enabled the establishment of a continuous relation between drug dose and MPE% as the outcome of interest. There was a 19.60 higher average MPE% for each mg/kg increase in morphine dose. In contrast, there was a 17.05 higher average MPE% for every 0.1 mg/kg increase in ketamine dose. Each 0.1 mg/kg increase in ketamine dose, when combined with morphine (3 mg/kg), led to a 30.85 higher average MPE%. A tenfold impact of increasing mecamylamine dosage on MPE% was observed when paired with morphine. Thus, hippocampal nAChRs play a significant role in mediating the anti-allodynic effect of ketamine and morphine in neuropathic pain.
Collapse
Affiliation(s)
- Romina Rahiminezhad Seta
- Department of Animal Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Samaneh Eftekhari Mahabadi
- School of Mathematics, Statistics and Computer Science, College of Science, University of Tehran, Tehran, Iran
| | - Ladan Delphi
- Department of Animal Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Sakineh Alijanpour
- Department of Animal Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran; Department of Biology, Faculty of Science, Gonbad Kavous University, Gonbad Kavous, Iran
| | - Ameneh Rezayof
- Department of Animal Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran.
| |
Collapse
|
2
|
Sansone L, Milani F, Fabrizi R, Belli M, Cristina M, Zagà V, de Iure A, Cicconi L, Bonassi S, Russo P. Nicotine: From Discovery to Biological Effects. Int J Mol Sci 2023; 24:14570. [PMID: 37834017 PMCID: PMC10572882 DOI: 10.3390/ijms241914570] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 09/21/2023] [Accepted: 09/23/2023] [Indexed: 10/15/2023] Open
Abstract
Nicotine, the primary psychoactive agent in tobacco leaves, has led to the widespread use of tobacco, with over one billion smokers globally. This article provides a historical overview of tobacco and discusses tobacco dependence, as well as the biological effects induced by nicotine on mammalian cells. Nicotine induces various biological effects, such as neoangiogenesis, cell division, and proliferation, and it affects neural and non-neural cells through specific pathways downstream of nicotinic receptors (nAChRs). Specific effects mediated by α7 nAChRs are highlighted. Nicotine is highly addictive and hazardous. Public health initiatives should prioritize combating smoking and its associated risks. Understanding nicotine's complex biological effects is essential for comprehensive research and informed health policies. While potential links between nicotine and COVID-19 severity warrant further investigation, smoking remains a significant cause of morbidity and mortality globally. Effective public health strategies are vital to promote healthier lifestyles.
Collapse
Affiliation(s)
- Luigi Sansone
- Department of Human Sciences and Quality, Life Promotion San Raffaele University, Via di Val Cannuta 247, 00166 Rome, Italy; (L.S.); (M.B.); (M.C.); (A.d.I.); (S.B.)
- MEBIC Consortium, San Raffaele University, 00166 Rome, Italy
| | - Francesca Milani
- Clinical and Molecular Epidemiology, IRCCS San Raffaele Roma, Via di Val Cannuta 247, 00166 Rome, Italy; (F.M.); (R.F.); (L.C.)
| | - Riccardo Fabrizi
- Clinical and Molecular Epidemiology, IRCCS San Raffaele Roma, Via di Val Cannuta 247, 00166 Rome, Italy; (F.M.); (R.F.); (L.C.)
| | - Manuel Belli
- Department of Human Sciences and Quality, Life Promotion San Raffaele University, Via di Val Cannuta 247, 00166 Rome, Italy; (L.S.); (M.B.); (M.C.); (A.d.I.); (S.B.)
- MEBIC Consortium, San Raffaele University, 00166 Rome, Italy
| | - Mario Cristina
- Department of Human Sciences and Quality, Life Promotion San Raffaele University, Via di Val Cannuta 247, 00166 Rome, Italy; (L.S.); (M.B.); (M.C.); (A.d.I.); (S.B.)
- Department of Molecular Medicine, University La Sapienza, Viale del Policlinico 155, 00161 Rome, Italy
| | - Vincenzo Zagà
- Italian Society of Tabaccology (SITAB), 00136 Bologna, Italy;
| | - Antonio de Iure
- Department of Human Sciences and Quality, Life Promotion San Raffaele University, Via di Val Cannuta 247, 00166 Rome, Italy; (L.S.); (M.B.); (M.C.); (A.d.I.); (S.B.)
- Experimental Neurophysiology IRCCS San Raffaele Roma, Via di Val Cannuta 247, 00166 Rome, Italy
| | - Luca Cicconi
- Clinical and Molecular Epidemiology, IRCCS San Raffaele Roma, Via di Val Cannuta 247, 00166 Rome, Italy; (F.M.); (R.F.); (L.C.)
| | - Stefano Bonassi
- Department of Human Sciences and Quality, Life Promotion San Raffaele University, Via di Val Cannuta 247, 00166 Rome, Italy; (L.S.); (M.B.); (M.C.); (A.d.I.); (S.B.)
- Clinical and Molecular Epidemiology, IRCCS San Raffaele Roma, Via di Val Cannuta 247, 00166 Rome, Italy; (F.M.); (R.F.); (L.C.)
| | - Patrizia Russo
- Department of Human Sciences and Quality, Life Promotion San Raffaele University, Via di Val Cannuta 247, 00166 Rome, Italy; (L.S.); (M.B.); (M.C.); (A.d.I.); (S.B.)
- MEBIC Consortium, San Raffaele University, 00166 Rome, Italy
| |
Collapse
|
3
|
NMDA Receptor and Its Emerging Role in Cancer. Int J Mol Sci 2023; 24:ijms24032540. [PMID: 36768862 PMCID: PMC9917092 DOI: 10.3390/ijms24032540] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/23/2023] [Accepted: 01/25/2023] [Indexed: 01/31/2023] Open
Abstract
Glutamate is a key player in excitatory neurotransmission in the central nervous system (CNS). The N-methyl-D-aspartate receptor (NMDAR) is a glutamate-gated ion channel which presents several unique features and is involved in various physiological and pathological neuronal processes. Thanks to great efforts in neuroscience, its structure and the molecular mechanisms controlling its localization and functional regulation in neuronal cells are well known. The signaling mediated by NMDAR in neurons is very complex as it depends on its localization, composition, Ca2+ influx, and ion flow-independent conformational changes. Moreover, NMDA receptors are highly diffusive in the plasma membrane of neurons, where they form heterocomplexes with other membrane receptors and scaffold proteins which determine the receptor function and activation of downstream signaling. Interestingly, a recent paper demonstrates that NMDAR signaling is involved in epithelial cell competition, an evolutionary conserved cell fitness process influencing cancer initiation and progress. The idea that NMDAR signaling is limited to CNS has been challenged in the past two decades. A large body of evidence suggests that NMDAR is expressed in cancer cells outside the CNS and can respond to the autocrine/paracrine release of glutamate. In this review, we survey research on NMDAR signaling and regulation in neurons that can help illuminate its role in tumor biology. Finally, we will discuss existing data on the role of the glutamine/glutamate metabolism, the anticancer action of NMDAR antagonists in experimental models, NMDAR synaptic signaling in tumors, and clinical evidence in human cancer.
Collapse
|
4
|
Findley CA, McFadden SA, Cox MF, Sime LN, Peck MR, Quinn K, Bartke A, Hascup KN, Hascup ER. Prodromal Glutamatergic Modulation with Riluzole Impacts Glucose Homeostasis and Spatial Cognition in Alzheimer's Disease Mice. J Alzheimers Dis 2023; 94:371-392. [PMID: 37248899 PMCID: PMC10357216 DOI: 10.3233/jad-221245] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/26/2023] [Indexed: 05/31/2023]
Abstract
BACKGROUND Prior research supports a strong link between Alzheimer's disease (AD) and metabolic dysfunction that involves a multi-directional interaction between glucose, glutamatergic homeostasis, and amyloid pathology. Elevated soluble amyloid-β (Aβ) is an early biomarker for AD-associated cognitive decline that contributes to concurrent glutamatergic and metabolic dyshomeostasis in humans and male transgenic AD mice. Yet, it remains unclear how primary time-sensitive targeting of hippocampal glutamatergic activity may impact glucose regulation in an amyloidogenic mouse model. Previous studies have illustrated increased glucose uptake and metabolism using a neuroprotective glutamate modulator (riluzole), supporting the link between glucose and glutamatergic homeostasis. OBJECTIVE We hypothesized that targeting early glutamatergic hyperexcitation through riluzole treatment could aid in attenuating co-occurring metabolic and amyloidogenic pathologies with the intent of ameliorating cognitive decline. METHODS We conducted an early intervention study in male and female transgenic (AβPP/PS1) and knock-in (APPNL - F/NL - F) AD mice to assess the on- and off-treatment effects of prodromal glutamatergic modulation (2-6 months of age) on glucose homeostasis and spatial cognition through riluzole treatment. RESULTS Results indicated a sex- and genotype-specific effect on glucose homeostasis and spatial cognition with riluzole intervention that evolved with disease progression and time since treatment. CONCLUSION These findings support the interconnected nature of glucose and glutamatergic homeostasis with amyloid pathology and petition for further investigation into the targeting of this relationship to improve cognitive performance.
Collapse
Affiliation(s)
- Caleigh A. Findley
- Neuroscience Institute, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Department of Neurology, Southern Illinois University School of Medicine, Springfield, IL, USA
- Departments of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Samuel A. McFadden
- Neuroscience Institute, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Department of Neurology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - MaKayla F. Cox
- Neuroscience Institute, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Department of Neurology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Lindsey N. Sime
- Neuroscience Institute, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Department of Neurology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Mackenzie R. Peck
- Neuroscience Institute, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Department of Neurology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Kathleen Quinn
- Neuroscience Institute, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Department of Neurology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Andrzej Bartke
- Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, USA
- Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Kevin N. Hascup
- Neuroscience Institute, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Department of Neurology, Southern Illinois University School of Medicine, Springfield, IL, USA
- Departments of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, USA
- Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Erin R. Hascup
- Neuroscience Institute, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Department of Neurology, Southern Illinois University School of Medicine, Springfield, IL, USA
- Departments of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, USA
| |
Collapse
|
5
|
Alpha7 nicotinic acetylcholine receptor agonist PHA-543613 improves memory deficits in presenilin 1 and presenilin 2 conditional double knockout mice. Exp Neurol 2023; 359:114271. [PMID: 36370840 DOI: 10.1016/j.expneurol.2022.114271] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 10/18/2022] [Accepted: 11/06/2022] [Indexed: 11/11/2022]
Abstract
Cholinergic system dysfunction has been considered as a critical feature of neurodegenerative progression in Alzheimer's disease (AD). The α7 nicotinic acetylcholine receptors (α7-nAChRs) are widely expressed in the hippocampus cortex and play an important role in memory formation, considered as potential therapeutic agents targets. However, underlying mechanisms have not been fully elucidated. Here, we combine behavioral, molecular biological methods with in vitro slice and in vivo multichannel electrophysiological recording techniques to investigate the molecular, cellular synaptic and neuronal mechanisms of activating α7-nAChR by PHA-543613 (a selective α7-nAChR agonist), which influences the impaired cognitive function using presenilin 1 (PS1) and presenilin 2 (PS2) conditional double knockout (cDKO) mice. Our results demonstrated that PHA-543613 treatment significantly improved the impaired hippocampus-related memory via recovering the reduced the hippocampal synaptic protein levels of α7-nAChR, NMADAR and AMPAR, thereby restoring the impaired post-tetanic potentiation (PTP), long-term potentiation (LTP), activation of molecular signaling pathway for neuronal protection, theta power and strength of theta-gamma phase-amplitude coupling (PAC) at hippocampus in 6-month-old cDKO mice. For the first time, we systematically reveal the mechanisms by which PHA-543613 improves memory deficits at different levels. Therefore, our findings may be significant for the development of therapeutic strategies for AD.
Collapse
|
6
|
Ventral hippocampal NMDA receptors mediate the effects of nicotine on stress-induced anxiety/exploratory behaviors in rats. Neurosci Lett 2022; 780:136649. [DOI: 10.1016/j.neulet.2022.136649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 03/19/2022] [Accepted: 04/19/2022] [Indexed: 11/22/2022]
|
7
|
Duan Y, Lv J, Zhang Z, Chen Z, Wu H, Chen J, Chen Z, Yang J, Wang D, Liu Y, Chen F, Tian Y, Cao X. Exogenous Aβ 1-42 monomers improve synaptic and cognitive function in Alzheimer's disease model mice. Neuropharmacology 2022; 209:109002. [PMID: 35196539 DOI: 10.1016/j.neuropharm.2022.109002] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/28/2022] [Accepted: 02/15/2022] [Indexed: 01/16/2023]
Abstract
Growing evidence has suggested the poor correlation between brain amyloid plaque and Alzheimer's disease (AD). Presenilin1 (PS1) and presenilin2 (PS2) conditional double knockout (cDKO) mice exhibited the reduced 42-amino acid amyloid-β peptide (Aβ1-42) level and AD-like symptoms, indicating a different pathological mechanism from the amyloid cascade hypothesis for AD. Here we found that exogenous synthetic Aβ1-42 monomers could improve the impaired memory not only in cDKO mice without Aβ1-42 deposition but also in the APP/PS1/Tau triple transgenic 3 × Tg-AD mice with Aβ1-42 deposition, which were mediated by α7-nAChR. Our findings demonstrate for the first time that reduced soluble Aβ1-42 level is the main cause of cognitive dysfunction in cDKO mice, and support the opinions that low soluble Aβ1-42 level due to Aβ1-42 deposition may also cause cognitive deficits in 3 × Tg-AD mice. Therefore, "loss-of-function" of Aβ1-42 should be avoided when designing therapies aimed at reducing Aβ1-42 burden in AD.
Collapse
Affiliation(s)
- Yanhong Duan
- Key Laboratory of Brain Functional Genomics, Ministry of Education, School of Life Sciences, East China Normal University, 3663 N. Zhongshan Rd., Shanghai, 200062, China
| | - Junyan Lv
- Key Laboratory of Brain Functional Genomics, Ministry of Education, School of Life Sciences, East China Normal University, 3663 N. Zhongshan Rd., Shanghai, 200062, China
| | - Zhonghui Zhang
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, East China Normal University, 3663 N. Zhongshan Rd., Shanghai, 200062, China
| | - Zhenzhen Chen
- Key Laboratory of Brain Functional Genomics, Ministry of Education, School of Life Sciences, East China Normal University, 3663 N. Zhongshan Rd., Shanghai, 200062, China
| | - Hao Wu
- Key Laboratory of Brain Functional Genomics, Ministry of Education, School of Life Sciences, East China Normal University, 3663 N. Zhongshan Rd., Shanghai, 200062, China
| | - Jinnan Chen
- Key Laboratory of Brain Functional Genomics, Ministry of Education, School of Life Sciences, East China Normal University, 3663 N. Zhongshan Rd., Shanghai, 200062, China
| | - Zhidong Chen
- Key Laboratory of Brain Functional Genomics, Ministry of Education, School of Life Sciences, East China Normal University, 3663 N. Zhongshan Rd., Shanghai, 200062, China
| | - Jiarun Yang
- Key Laboratory of Brain Functional Genomics, Ministry of Education, School of Life Sciences, East China Normal University, 3663 N. Zhongshan Rd., Shanghai, 200062, China
| | - Dasheng Wang
- Key Laboratory of Brain Functional Genomics, Ministry of Education, School of Life Sciences, East China Normal University, 3663 N. Zhongshan Rd., Shanghai, 200062, China
| | - Yamei Liu
- School of Life Sciences, Shanghai University, No. 99 Shangda Rd., Shanghai, 200444, PR China
| | - Fuxue Chen
- School of Life Sciences, Shanghai University, No. 99 Shangda Rd., Shanghai, 200444, PR China
| | - Yang Tian
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, East China Normal University, 3663 N. Zhongshan Rd., Shanghai, 200062, China
| | - Xiaohua Cao
- Key Laboratory of Brain Functional Genomics, Ministry of Education, School of Life Sciences, East China Normal University, 3663 N. Zhongshan Rd., Shanghai, 200062, China.
| |
Collapse
|
8
|
Calpain Inhibitors as Potential Therapeutic Modulators in Neurodegenerative Diseases. Neurochem Res 2022; 47:1125-1149. [PMID: 34982393 DOI: 10.1007/s11064-021-03521-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 12/27/2021] [Accepted: 12/28/2021] [Indexed: 02/07/2023]
Abstract
It is considered a significant challenge to understand the neuronal cell death mechanisms with a suitable cure for neurodegenerative disorders in the coming years. Calpains are one of the best-considered "cysteine proteases activated" in brain disorders. Calpain is an important marker and mediator in the pathophysiology of neurodegeneration. Calpain activation being the essential neurodegenerative factor causing apoptotic machinery activation, it is crucial to develop reliable and effective approaches to prevent calpain-mediated apoptosis in degenerating neurons. It has been recently seen that the "inhibition of calpain activation" has appeared as a possible therapeutic target for managing neurodegenerative diseases. A systematic literature review of PubMed, Medline, Bentham, Scopus, and EMBASE (Elsevier) databases was conducted. The present article reviews the basic pathobiology and role of selective calpain inhibitors used in various neurodegenerative diseases as a therapeutic target.
Collapse
|
9
|
Jiang A, Su P, Li S, Wong AHC, Liu F. Disrupting the α7nAChR-NR2A protein complex exerts antidepressant-like effects. Mol Brain 2021; 14:107. [PMID: 34225758 PMCID: PMC8256601 DOI: 10.1186/s13041-021-00817-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 06/24/2021] [Indexed: 11/10/2022] Open
Abstract
Major depressive disorder (MDD) is associated with significant morbidity and mortality. Most antidepressant medications target the serotonin and norepinephrine transporters, but a significant minority of patients do not respond to treatment and novel therapeutic targets are needed. We previously identified a protein complex composed of the α7 nicotinic acetylcholine receptor (nAChR) and NMDA glutamate receptors (NMDARs), through which α7nAChR upregulates NMDAR function. Disruption of the α7nAChR-NMDAR complex with an interfering peptide blocked α7nAChR-mediated upregulation of NMDAR function and cue-induced reinstatement of nicotine seeking in rat models of relapse. Here we report that disrupting the α7nAChR-NMDAR complex with the interfering peptide also has antidepressant-like effects in the forced swim test (FST), a common rat behaviour screening test for antidepressant effects. Furthermore, the interfering peptide significantly increases extracellular signal-regulated kinase (ERK) activity in the animals subjected to the FST. Our results provide a novel potential therapeutic target for the development of new antidepressant medications.
Collapse
Affiliation(s)
- Anlong Jiang
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, 250 College Street, Toronto, ON, M5T 1R8, Canada
| | - Ping Su
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, 250 College Street, Toronto, ON, M5T 1R8, Canada
| | - Shupeng Li
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, 250 College Street, Toronto, ON, M5T 1R8, Canada
| | - Albert H C Wong
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, 250 College Street, Toronto, ON, M5T 1R8, Canada
- Departments of Pharmacology, University of Toronto, Toronto, ON, M5T 1R8, Canada
- Institutes of Medical Science, University of Toronto, Toronto, ON, M5T 1R8, Canada
| | - Fang Liu
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, 250 College Street, Toronto, ON, M5T 1R8, Canada.
- Institutes of Medical Science, University of Toronto, Toronto, ON, M5T 1R8, Canada.
- University of Toronto, Toronto, ON, M5T 1R8, Canada.
- University of Toronto, Toronto, ON, M5T 1R8, Canada.
| |
Collapse
|
10
|
Stone TW. Relationships and Interactions between Ionotropic Glutamate Receptors and Nicotinic Receptors in the CNS. Neuroscience 2021; 468:321-365. [PMID: 34111447 DOI: 10.1016/j.neuroscience.2021.06.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 06/03/2021] [Accepted: 06/04/2021] [Indexed: 02/07/2023]
Abstract
Although ionotropic glutamate receptors and nicotinic receptors for acetylcholine (ACh) have usually been studied separately, they are often co-localized and functionally inter-dependent. The objective of this review is to survey the evidence for interactions between the two receptor families and the mechanisms underlying them. These include the mutual regulation of subunit expression, which change the NMDA:AMPA response balance, and the existence of multi-functional receptor complexes which make it difficult to distinguish between individual receptor sites, especially in vivo. This is followed by analysis of the functional relationships between the receptors from work on transmitter release, cellular electrophysiology and aspects of behavior where these can contribute to understanding receptor interactions. It is clear that nicotinic receptors (nAChRs) on axonal terminals directly regulate the release of glutamate and other neurotransmitters, α7-nAChRs generally promoting release. Hence, α7-nAChR responses will be prevented not only by a nicotinic antagonist, but also by compounds blocking the indirectly activated glutamate receptors. This accounts for the apparent anticholinergic activity of some glutamate antagonists, including the endogenous antagonist kynurenic acid. The activation of presynaptic nAChRs is by the ambient levels of ACh released from pre-terminal synapses, varicosities and glial cells, acting as a 'volume neurotransmitter' on synaptic and extrasynaptic sites. In addition, ACh and glutamate are released as CNS co-transmitters, including 'cholinergic' synapses onto spinal Renshaw cells. It is concluded that ACh should be viewed primarily as a modulator of glutamatergic neurotransmission by regulating the release of glutamate presynaptically, and the location, subunit composition, subtype balance and sensitivity of glutamate receptors, and not primarily as a classical fast neurotransmitter. These conclusions and caveats should aid clarification of the sites of action of glutamate and nicotinic receptor ligands in the search for new centrally-acting drugs.
Collapse
Affiliation(s)
- Trevor W Stone
- The Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7FY, UK; Institute of Neuroscience, University of Glasgow, G12 8QQ, UK.
| |
Collapse
|
11
|
Petit-Pedrol M, Groc L. Regulation of membrane NMDA receptors by dynamics and protein interactions. J Cell Biol 2021; 220:211609. [PMID: 33337489 PMCID: PMC7754687 DOI: 10.1083/jcb.202006101] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 11/19/2020] [Accepted: 11/20/2020] [Indexed: 12/16/2022] Open
Abstract
Understanding neurotransmitter system crosstalk in the brain is a major challenge in neurobiology. Several intracellular and genomic cascades have been identified in this crosstalk. However, the discovery that neurotransmitter receptors are highly diffusive in the plasma membrane of neurons, where they form heterocomplexes with other proteins, has profoundly changed our view of neurotransmitter signaling. Here, we review new insights into neurotransmitter crosstalk at the plasma membrane. We focus on the membrane organization and interactome of the ionotropic glutamate N-methyl-D-aspartate receptor (NMDAR) that plays a central role in excitatory synaptic and network physiology and is involved in the etiology of several major neuropsychiatric disorders. The nanoscale organization and dynamics of NMDAR is a key regulatory process for glutamate synapse transmission, plasticity, and crosstalk with other neurotransmitter systems, such as the monoaminergic ones. The plasma membrane appears to be a prime regulatory compartment for spatial and temporal crosstalk between neurotransmitter systems in the healthy and diseased brain. Understanding the molecular mechanisms regulating membrane neurotransmitter receptor crosstalk will likely open research avenues for innovative therapeutical strategies.
Collapse
Affiliation(s)
- Mar Petit-Pedrol
- Université de Bordeaux, Centre National de la Recherche Scientifique, Interdisciplinary Institute for Neuroscience, Unité Mixte de Recherche 5297, Bordeaux, France
| | - Laurent Groc
- Université de Bordeaux, Centre National de la Recherche Scientifique, Interdisciplinary Institute for Neuroscience, Unité Mixte de Recherche 5297, Bordeaux, France
| |
Collapse
|
12
|
Angrand L, Takillah S, Malissin I, Berriche A, Cervera C, Bel R, Gerard Q, Knoertzer J, Baati R, Kononchik JP, Megarbane B, Thibault K, Dal Bo G. Persistent brainwave disruption and cognitive impairment induced by acute sarin surrogate sub-lethal dose exposure. Toxicology 2021; 456:152787. [PMID: 33887375 DOI: 10.1016/j.tox.2021.152787] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 04/11/2021] [Accepted: 04/15/2021] [Indexed: 11/15/2022]
Abstract
Warfare neurotoxicants such as sarin, soman or VX, are organophosphorus compounds which irreversibly inhibit cholinesterase. High-dose exposure with nerve agents (NA) is known to produce seizure activity and related brain damage, while less is known about the effects of acute sub-lethal dose exposure. The aim of this study was to characterize behavioral, brain activity and neuroinflammatory modifications at different time points after exposure to 4-nitrophenyl isopropyl methylphosphonate (NIMP), a sarin surrogate. In order to decipher the impacts of sub-lethal exposure, we chose 4 different doses of NIMP each corresponding to a fraction of the median lethal dose (LD50). First, we conducted a behavioral analysis of symptoms during the first hour following NIMP challenge and established a specific scoring scale for the intoxication severity. The intensity of intoxication signs was dose-dependent and proportional to the cholinesterase activity inhibition evaluated in mice brain. The lowest dose (0.3 LD50) did not induce significant behavioral, electrocorticographic (ECoG) nor cholinesterase activity changes. Animals exposed to one of the other doses (0.5, 0.7 and 0.9 LD50) exhibited substantial changes in behavior, significant cholinesterase activity inhibition, and a disruption of brainwave distribution that persisted in a dose-dependent manner. To evaluate long lasting changes, we conducted ECoG recording for 30 days on mice exposed to 0.5 or 0.9 LD50 of NIMP. Mice in both groups showed long-lasting impairment of theta rhythms, and a lack of restoration in hippocampal ChE activity after 1-month post-exposure. In addition, an increase in neuroinflammatory markers (IBA-1, TNF-α, NF-κB) and edema were transiently observed in mice hippocampus. Furthermore, a novel object recognition test showed an alteration of short-term memory in both groups, 1-month post-NIMP intoxication. Our findings identified both transient and long-term ECoG alterations and some long term cognitive impairments following exposure to sub-lethal doses of NIMP. These may further impact morphopathological alterations in the brain.
Collapse
Affiliation(s)
- Loïc Angrand
- Departement of Toxicology and Chemical Risks, French Armed Forces Biomedical Research Institute, Bretigny sur Orge, France; EnvA, IMRB, Maisons-Alfort, France; Université Paris-Est Créteil, INSERM, Team Relaix, Créteil, France
| | - Samir Takillah
- Departement of Neuroscience, Unit of Fatigue and Vigilance, French Armed Forces Biomedical Research Institute, Bretigny sur Orge, France; VIFASOM Team (EA 7330), Paris Descartes University, Sorbonne Paris Cité, Hôtel Dieu, Paris, France
| | - Isabelle Malissin
- Department of Medical and Toxicological Critical Care, Lariboisière Hospital, Federation of Toxicology APHP, Paris-Diderot University, INSERM UMRS-1144, Paris, France
| | - Asma Berriche
- Departement of Toxicology and Chemical Risks, French Armed Forces Biomedical Research Institute, Bretigny sur Orge, France; CEA, Fontenay aux roses, France
| | - Chloe Cervera
- Departement of Toxicology and Chemical Risks, French Armed Forces Biomedical Research Institute, Bretigny sur Orge, France
| | - Rosalie Bel
- Departement of Toxicology and Chemical Risks, French Armed Forces Biomedical Research Institute, Bretigny sur Orge, France
| | - Quentin Gerard
- Departement of Toxicology and Chemical Risks, French Armed Forces Biomedical Research Institute, Bretigny sur Orge, France; Normandie University, UNICAEN, INSERM, GIP Cyceron, Institut Blood and Brain @Caen-Normandie (BB@C), UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), Caen, France
| | - Julie Knoertzer
- Departement of Toxicology and Chemical Risks, French Armed Forces Biomedical Research Institute, Bretigny sur Orge, France
| | - Rachid Baati
- ICPEES UMR CNRS 7515, Institut de Chimie des Procédés, pour l'Energie, l'Environnement, et la Santé, Strasbourg, France
| | - Joseph P Kononchik
- Departement of Toxicology and Chemical Risks, French Armed Forces Biomedical Research Institute, Bretigny sur Orge, France
| | - Bruno Megarbane
- VIFASOM Team (EA 7330), Paris Descartes University, Sorbonne Paris Cité, Hôtel Dieu, Paris, France; Department of Medical and Toxicological Critical Care, Lariboisière Hospital, Federation of Toxicology APHP, Paris-Diderot University, INSERM UMRS-1144, Paris, France
| | - Karine Thibault
- Departement of Toxicology and Chemical Risks, French Armed Forces Biomedical Research Institute, Bretigny sur Orge, France.
| | - Gregory Dal Bo
- Departement of Toxicology and Chemical Risks, French Armed Forces Biomedical Research Institute, Bretigny sur Orge, France.
| |
Collapse
|
13
|
Koola MM. Alpha7 nicotinic-N-methyl-D-aspartate hypothesis in the treatment of schizophrenia and beyond. Hum Psychopharmacol 2021; 36:1-16. [PMID: 32965756 DOI: 10.1002/hup.2758] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 09/08/2020] [Accepted: 09/09/2020] [Indexed: 12/12/2022]
Abstract
Development of novel treatments for positive, cognitive, and negative symptoms continue to be a high-priority area of schizophrenia research and a major unmet clinical need. Given that all randomized controlled trials (RCTs) conducted to date failed with one add-on medication/mechanism of action, future RCTs with the same approach are not warranted. Even if the field develops a medication for cognition, others are still needed to treat negative and positive symptoms. Therefore, fixing one domain does not completely solve the problem. Also, targeting the cholinergic system, glutamatergic system, and cholinergic plus alpha7 nicotinic and N-methyl-D-aspartate (NMDA) receptors failed independently. Hence, targeting other less important pathophysiological mechanisms/targets is unlikely to be successful. Meta-analyses of RCTs targeting major pathophysiological mechanisms have found some efficacy signal in schizophrenia; thus, combination treatments with different mechanisms of action may enhance the efficacy signal. The objective of this article is to highlight the importance of conducting RCTs with novel combination treatments in schizophrenia to develop antischizophrenia treatments. Positive RCTs with novel combination treatments that target the alpha7 nicotinic and NMDA receptors simultaneously may lead to a disease-modifying therapeutic armamentarium in schizophrenia. Novel combination treatments that concurrently improve the three domains of psychopathology and several prognostic and theranostic biomarkers may facilitate therapeutic discovery in schizophrenia.
Collapse
Affiliation(s)
- Maju Mathew Koola
- Department of Psychiatry and Behavioral Health, Stony Brook University Renaissance School of Medicine, Stony Brook, New York, USA
| |
Collapse
|
14
|
Galantamine-Memantine combination in the treatment of Alzheimer's disease and beyond. Psychiatry Res 2020; 293:113409. [PMID: 32829072 DOI: 10.1016/j.psychres.2020.113409] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/17/2020] [Accepted: 08/17/2020] [Indexed: 02/06/2023]
Abstract
Alzheimer's disease (AD) is the most prevalent form of dementia in the elderly population worldwide. Despite the major unmet clinical need, no new medications for the treatment of AD have been approved since 2003. Galantamine is an acetylcholinesterase inhibitor that is also a positive allosteric modulator at the α4β2 and α7nACh receptors. Memantine is an N-methyl-d-aspartate receptor modulator/agonist. Both galantamine and memantine are FDA-approved medications for the treatment of AD. The objective of this review is to highlight the potential of the galantamine-memantine combination to conduct randomized controlled trials (RCTs) in AD. Several studies have shown the combination to be effective. Neurodegenerative diseases involve multiple pathologies; therefore, combination treatment appears to be a rational approach. Although underutilized, the galantamine-memantine combination is the standard of care in the treatment of AD. Positive RCTs with the combination with concurrent improvement in symptoms and biomarkers may lead to FDA approval, which may lead to greater utilization of this combination in clinical practice.
Collapse
|
15
|
Mamiya T, Tanase S, Takeuchi S, Kato S, Ito A, Hiramatsu M, Nabeshima T. Galantamine improves enhanced impulsivity, impairments of attention and long-term potentiation induced by prenatal nicotine exposure to mice. Biochem Pharmacol 2020; 180:114139. [PMID: 32652142 DOI: 10.1016/j.bcp.2020.114139] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 07/02/2020] [Accepted: 07/06/2020] [Indexed: 12/21/2022]
Abstract
Prenatal nicotine exposure (PNE) causes behavioral abnormalities in offspring, such as an enhancement of impulsivity and decrease in attention at adolescence. Here we examined the effects of galantamine (GAL) on the behavioral and electrophysiological changes induced by PNE in mice. Pregnant C57BL/6J mice were exposed to nicotine (0.2 mg/mL) dissolved in sweetened (2% saccharin) drinking water during gestational day 14 and perinatal day 0 (P0). At the ages of postnatal days 42-49 (P42-P49), female offspring displayed impulsivity in the cliff avoidance test and impairment of visual attention in the object-based attention test. Decrease of long-term potentiation (LTP) and extracellular glutamate levels were observed in the prefrontal cortex of PNE mice. Systemic treatment with GAL (1 mg/kg, s.c.), an allosteric potentiating ligand for the nicotinic acetylcholine receptor (nAChR) and a weak cholinesterase inhibitor, attenuated the enhancement of impulsivity and impairment of attention induced by PNE in mice. Further, GAL reversed the impairment of LTP induced by PNE in the prefrontal cortex of mice, although it failed to attenuate the decrease of extracellular glutamate levels. The effects of GAL were blocked by an α 7 nAChR antagonist, methyllycaconitine (1 mg/kg, i.p.). These results suggest that PNE during cortex development affects nicotinic cholinergic-dependent plasticity and formation of impulsivity and attention. Furthermore, GAL could be a useful drug for cognitive impairments-related to attention deficit hyperactivity disorder.
Collapse
Affiliation(s)
- Takayoshi Mamiya
- Department of Chemical Pharmacology, Faculty of Pharmacy, Meijo University, Nagoya, Japan; Japanese Drug Organization of Appropriate Use and Research, Nagoya, Japan.
| | - Shota Tanase
- Department of Chemical Pharmacology, Faculty of Pharmacy, Meijo University, Nagoya, Japan
| | - Shino Takeuchi
- Department of Chemical Pharmacology, Faculty of Pharmacy, Meijo University, Nagoya, Japan
| | - Shunsuke Kato
- Department of Chemical Pharmacology, Faculty of Pharmacy, Meijo University, Nagoya, Japan
| | - Ai Ito
- Department of Chemical Pharmacology, Faculty of Pharmacy, Meijo University, Nagoya, Japan
| | - Masayuki Hiramatsu
- Department of Chemical Pharmacology, Faculty of Pharmacy, Meijo University, Nagoya, Japan; Japanese Drug Organization of Appropriate Use and Research, Nagoya, Japan
| | - Toshitaka Nabeshima
- Advanced Diagnostic System Research Laboratory, Graduate School of Health Sciences, Fujita Health University, Toyoake, Japan; Japanese Drug Organization of Appropriate Use and Research, Nagoya, Japan
| |
Collapse
|
16
|
Activation of alpha7 nicotinic and NMDA receptors is necessary for performance in a working memory task. Psychopharmacology (Berl) 2020; 237:1723-1735. [PMID: 32162104 PMCID: PMC7313359 DOI: 10.1007/s00213-020-05495-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 02/19/2020] [Indexed: 10/24/2022]
Abstract
RATIONALE Working memory deficits are present in schizophrenia (SZ) but remain insufficiently resolved by medications. Similar cognitive dysfunctions can be produced acutely in animals by elevating brain levels of kynurenic acid (KYNA). KYNA's effects may reflect interference with the function of both the α7 nicotinic acetylcholine receptor (α7nAChR) and the glycineB site of the NMDA receptor. OBJECTIVES The aim of the present study was to examine, using pharmacological tools, the respective roles of these two receptor sites on performance in a delayed non-match-to-position working memory (WM) task (DNMTP). METHODS DNMTP consisted of 120 trials/session (5, 10, and 15 s delays). Rats received two doses (25 or 100 mg/kg, i.p.) of L-kynurenine (KYN; bioprecursor of KYNA) or L-4-chlorokynurenine (4-Cl-KYN; bioprecursor of the selective glycineB site antagonist 7-Cl-kynurenic acid). Attenuation of KYN- or 4-Cl-KYN-induced deficits was assessed by co-administration of galantamine (GAL, 3 mg/kg) or PAM-2 (1 mg/kg), two positive modulators of α7nAChR function. Reversal of 4-Cl-KYN-induced deficits was examined using D-cycloserine (DCS; 30 mg/kg), a partial agonist at the glycineB site. RESULTS Both KYN and 4-Cl-KYN administration produced dose-related deficits in DNMTP accuracy that were more severe at the longer delays. In KYN-treated rats, these deficits were reversed to control levels by GAL or PAM-2 but not by DCS. In contrast, DCS eliminated performance deficits in 4-Cl-KYN-treated animals. CONCLUSIONS These experiments reveal that both α7nAChR and NMDAR activity are necessary for normal WM accuracy. They provide substantive new support for the therapeutic potential of positive modulators at these two receptor sites in SZ and other major brain diseases.
Collapse
|
17
|
The medial prefrontal cortex - hippocampus circuit that integrates information of object, place and time to construct episodic memory in rodents: Behavioral, anatomical and neurochemical properties. Neurosci Biobehav Rev 2020; 113:373-407. [PMID: 32298711 DOI: 10.1016/j.neubiorev.2020.04.007] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 02/25/2020] [Accepted: 04/06/2020] [Indexed: 12/31/2022]
Abstract
Rats and mice have been demonstrated to show episodic-like memory, a prototype of episodic memory, as defined by an integrated memory of the experience of an object or event, in a particular place and time. Such memory can be assessed via the use of spontaneous object exploration paradigms, variably designed to measure memory for object, place, temporal order and object-location inter-relationships. We review the methodological properties of these tests, the neurobiology about time and discuss the evidence for the involvement of the medial prefrontal cortex (mPFC), entorhinal cortex (EC) and hippocampus, with respect to their anatomy, neurotransmitter systems and functional circuits. The systematic analysis suggests that a specific circuit between the mPFC, lateral EC and hippocampus encodes the information for event, place and time of occurrence into the complex episodic-like memory, as a top-down regulation from the mPFC onto the hippocampus. This circuit can be distinguished from the neuronal component memory systems for processing the individual information of object, time and place.
Collapse
|
18
|
Santoro A, Tomino C, Prinzi G, Lamonaca P, Cardaci V, Fini M, Russo P. Tobacco Smoking: Risk to Develop Addiction, Chronic Obstructive Pulmonary Disease, and Lung Cancer. Recent Pat Anticancer Drug Discov 2019; 14:39-52. [PMID: 30605063 DOI: 10.2174/1574892814666190102122848] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 12/11/2018] [Accepted: 12/27/2018] [Indexed: 12/24/2022]
Abstract
BACKGROUND The morbidity and mortality associated with tobacco smoking is well established. Nicotine is the addictive component of tobacco. Nicotine, through the non-neuronal α7nicotinic receptor, induces cell proliferation, neo-angiogenesis, epithelial to mesenchymal transition, and inhibits drug-induced apoptosis. OBJECTIVE To understand the genetic, molecular and cellular biology of addiction, chronic obstructive pulmonary disease and lung cancer. METHODS The search for papers to be included in the review was performed during the months of July- September 2018 in the following databases: PubMed (http://www.ncbi.nlm.nih.gov), Scopus (http://www.scopus.com), EMBASE (http://www.elsevier.com/online-tools/embase), and ISI Web of Knowledge (http://apps.webofknowledge.com/). The following searching terms: "nicotine", "nicotinic receptor", and "addiction" or "COPD" or "lung cancer" were used. Patents were retrieved in clinicaltrials.gov (https://clinicaltrials.gov/). All papers written in English were evaluated. The reference list of retrieved articles was also reviewed to identify other eligible studies that were not indexed by the above-mentioned databases. New experimental data on the ability of nicotine to promote transformation of human bronchial epithelial cells, exposed for one hour to Benzo[a]pyrene-7,8-diol-9-10-epoxide, are reported. RESULTS Nicotinic receptors variants and nicotinic receptors upregulation are involved in addiction, chronic obstructive pulmonary disease and/or lung cancer. Nicotine through α7nicotinic receptor upregulation induces complete bronchial epithelial cells transformation. CONCLUSION Genetic studies highlight the involvement of nicotinic receptors variants in addiction, chronic obstructive pulmonary disease and/or lung cancer. A future important step will be to translate these genetic findings to clinical practice. Interventions able to help smoking cessation in nicotine dependence subjects, under patent, are reported.
Collapse
Affiliation(s)
- Alessia Santoro
- Clinical and Molecular Epidemiology, IRCSS San Raffaele Pisana, Via di Valcannuta 247, I-00166 Rome, Italy
| | - Carlo Tomino
- Scientific Direction, IRCSS San Raffaele Pisana, Via di Valcannuta 247, I-00166 Rome, Italy
| | - Giulia Prinzi
- Clinical and Molecular Epidemiology, IRCSS San Raffaele Pisana, Via di Valcannuta 247, I-00166 Rome, Italy
| | - Palma Lamonaca
- Clinical and Molecular Epidemiology, IRCSS San Raffaele Pisana, Via di Valcannuta 247, I-00166 Rome, Italy
| | - Vittorio Cardaci
- Pulmonary Rehabilitation, IRCCS San Raffaele Pisana, Via della Pisana, 235, I-00163 Rome, Italy
| | - Massimo Fini
- Scientific Direction, IRCSS San Raffaele Pisana, Via di Valcannuta 247, I-00166 Rome, Italy
| | - Patrizia Russo
- Clinical and Molecular Epidemiology, IRCSS San Raffaele Pisana, Via di Valcannuta 247, I-00166 Rome, Italy
| |
Collapse
|
19
|
Livingstone RW, Elder MK, Barrett MC, Westlake CM, Peppercorn K, Tate WP, Abraham WC, Williams JM. Secreted Amyloid Precursor Protein-Alpha Promotes Arc Protein Synthesis in Hippocampal Neurons. Front Mol Neurosci 2019; 12:198. [PMID: 31474829 PMCID: PMC6702288 DOI: 10.3389/fnmol.2019.00198] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 07/30/2019] [Indexed: 12/22/2022] Open
Abstract
Secreted amyloid precursor protein-α (sAPPα) is a neuroprotective and memory-enhancing molecule, however, the mechanisms through which sAPPα promotes these effects are not well understood. Recently, we have shown that sAPPα enhances cell-surface expression of glutamate receptors. Activity-related cytoskeletal-associated protein Arc (Arg3.1) is an immediate early gene capable of modulating long-term potentiation, long-term depression and homeostatic plasticity through regulation of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor localization. Accordingly, we hypothesized that sAPPα may enhance synaptic plasticity, in part, by the de novo synthesis of Arc. Using primary cortical and hippocampal neuronal cultures we found that sAPPα (1 nM, 2 h) enhances levels of Arc mRNA and protein. Arc protein levels were increased in both the neuronal somata and dendrites in a Ca2+/calmodulin-dependent protein kinase II-dependent manner. Additionally, dendritic Arc expression was dependent upon activation of mitogen-activated protein kinase and protein kinase G. The enhancement of dendritic Arc protein was significantly reduced by antagonism of N-methyl-D-aspartate (NMDA) and nicotinic acetylcholine (α7nACh) receptors, and fully eliminated by dual application of these antagonists. This effect was further corroborated in area CA1 of acute hippocampal slices. These data suggest sAPPα-regulated plasticity within hippocampal neurons is mediated by cooperation of NMDA and α7nACh receptors to engage a cascade of signal transduction molecules to enhance the transcription and translation of Arc.
Collapse
Affiliation(s)
- Rhys W Livingstone
- Department of Anatomy, Brain Health Research Centre, Brain Research New Zealand, Rangahau Roro Aotearoa, University of Otago, Dunedin, New Zealand
| | - Megan K Elder
- Department of Anatomy, Brain Health Research Centre, Brain Research New Zealand, Rangahau Roro Aotearoa, University of Otago, Dunedin, New Zealand
| | - Maya C Barrett
- Department of Anatomy, Brain Health Research Centre, Brain Research New Zealand, Rangahau Roro Aotearoa, University of Otago, Dunedin, New Zealand
| | - Courteney M Westlake
- Department of Anatomy, Brain Health Research Centre, Brain Research New Zealand, Rangahau Roro Aotearoa, University of Otago, Dunedin, New Zealand
| | - Katie Peppercorn
- Department of Biochemistry, Brain Health Research Centre, Brain Research New Zealand, Rangahau Roro Aotearoa, University of Otago, Dunedin, New Zealand
| | - Warren P Tate
- Department of Biochemistry, Brain Health Research Centre, Brain Research New Zealand, Rangahau Roro Aotearoa, University of Otago, Dunedin, New Zealand
| | - Wickliffe C Abraham
- Department of Psychology, Brain Health Research Centre, Brain Research New Zealand, Rangahau Roro Aotearoa, University of Otago, Dunedin, New Zealand
| | - Joanna M Williams
- Department of Anatomy, Brain Health Research Centre, Brain Research New Zealand, Rangahau Roro Aotearoa, University of Otago, Dunedin, New Zealand
| |
Collapse
|
20
|
Palacios-Filardo J, Mellor JR. Neuromodulation of hippocampal long-term synaptic plasticity. Curr Opin Neurobiol 2018; 54:37-43. [PMID: 30212713 PMCID: PMC6367596 DOI: 10.1016/j.conb.2018.08.009] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 07/26/2018] [Accepted: 08/15/2018] [Indexed: 12/31/2022]
Abstract
Acetylcholine, noradrenaline, dopamine and serotonin all facilitate long-term synaptic plasticity. Neuromodulators facilitate long-term synaptic plasticity by common and divergent mechanisms. Common mechanisms include NMDA receptor facilitation by potassium channel inhibition, gliotransmission and disinhibition. Divergent mechanisms include diversity of disinhibition and temporal and spatial neuromodulator release.
Multiple neuromodulators including acetylcholine, noradrenaline, dopamine and serotonin are released in response to uncertainty to focus attention on events where the predicted outcome does not match observed reality. In these situations, internal representations need to be updated, a process that requires long-term synaptic plasticity. Through a variety of common and divergent mechanisms, it is recently shown that all these neuromodulators facilitate the induction and/or expression of long-term synaptic plasticity within the hippocampus. Under physiological conditions, this may be critical for suprathreshold induction of plasticity endowing neuromodulators with a gating function and providing a mechanism by which neuromodulators enable the targeted updating of memory with relevant information to improve the accuracy of future predictions.
Collapse
Affiliation(s)
- Jon Palacios-Filardo
- Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol BS8 1TD, UK
| | - Jack R Mellor
- Centre for Synaptic Plasticity, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol BS8 1TD, UK.
| |
Collapse
|
21
|
Blockade of glutamate receptor ameliorates lipopolysaccharide-induced sepsis through regulation of neuropeptides. Biosci Rep 2018; 38:BSR20171629. [PMID: 29440461 PMCID: PMC5938426 DOI: 10.1042/bsr20171629] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Revised: 01/25/2018] [Accepted: 02/12/2018] [Indexed: 11/23/2022] Open
Abstract
Glutamate receptors (N-methyl-d-aspartate receptor (NMDAR)) are expressed mainly in the central nervous system (CNS), but several potentially important exceptions are worth mentioning. Recently, NMDAR, a glutamate receptor, has been reported to be found in the lungs. NMDAR is activated in acute lung injury (ALI). Here, the present experiment was designed to examine whether NMDAR blockade (MK-801) ameliorates ALI through affecting neuropeptides in LPS-induced sepsis animal models. Male Kunming mice were divided into control group, LPS group, control + MK-801 group, and LPS + MK-801 group. Bronchoalveolar lavage fluid (BALF) was collected and evaluated. The lung histological pathology was assayed by immunocytochemistry staining. Western blot was used to measure PGP9.5, substance P (SP), and vasoactive intestinal polypeptide (VIP). Results showed that LPS-induced mice animal models were ameliorated by co-treatment with the MK-801, an uncompetitive NMDAR antagonist. Moreover, the protective effects of MK-801 attributed to the increased secretion of VIP and decreased secretion of SP. The results of the present study indicated that the blockade of NMDAR may represent a promising therapeutic strategy for the treatment of sepsis-associated ALI through regulation of neuropeptides.
Collapse
|
22
|
Elnagar MR, Walls AB, Helal GK, Hamada FM, Thomsen MS, Jensen AA. Functional characterization of α7 nicotinic acetylcholine and NMDA receptor signaling in SH-SY5Y neuroblastoma cells in an ERK phosphorylation assay. Eur J Pharmacol 2018; 826:106-113. [DOI: 10.1016/j.ejphar.2018.02.047] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 02/20/2018] [Accepted: 02/28/2018] [Indexed: 12/23/2022]
|
23
|
Role of α7nAChR-NMDAR in sevoflurane-induced memory deficits in the developing rat hippocampus. PLoS One 2018; 13:e0192498. [PMID: 29401517 PMCID: PMC5798850 DOI: 10.1371/journal.pone.0192498] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 01/24/2018] [Indexed: 11/28/2022] Open
Abstract
Detrimental effects of volatile anaesthetics, including sevoflurane, on the structure and function of the developing brain have been reported. The internalization of N-methyl-D-aspartate receptors (NMDARs) contributes to anaesthetic neurotoxicity. Both nicotinic acetylcholine receptors (nAChRs) and NMDAR play a critical role in the development of the nervous system. Moreover, nAChR can interact with NMDAR, and previous studies have demonstrated modulation of NMDAR by nAChR. In our study, we used an α7 nicotinic acetylcholine receptor (α7nAChR) agonist and α7nAChR antagonist to explore the role of α7nAChR and NMDAR in sevoflurane-induced long-term effects on memory and dendritic spine both in vivo and in vitro. The results revealed that the activation of α7nAChR attenuated the development of sevoflurane-induced memory deficit and dendritic spine changes, which might be by regulating NR2B-containing NMDAR trafficking from the intracellular pool to the cell surface pool in the hippocampus. Moreover, we demonstrated that α7nAChR could regulate NR2B-containing NMDAR via Src-family tyrosine kinase (SFK). Thus, our current study indicates that the trafficking of NR2B-containing NMDAR is regulated by α7nAChR via SFK in neonatal rat hippocampus, which may be secondary to sevoflurane-induced cognitive deficits in the developing hippocampus.
Collapse
|
24
|
Happ DF, Tasker RA. Effects of α7 Nicotinic Receptor Activation on Cell Survival in Rat Organotypic Hippocampal Slice Cultures. Neurotox Res 2017; 33:887-895. [DOI: 10.1007/s12640-017-9854-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 12/04/2017] [Accepted: 12/13/2017] [Indexed: 11/28/2022]
|
25
|
Elnagar MR, Walls AB, Helal GK, Hamada FM, Thomsen MS, Jensen AA. Probing the putative α7 nAChR/NMDAR complex in human and murine cortex and hippocampus: Different degrees of complex formation in healthy and Alzheimer brain tissue. PLoS One 2017; 12:e0189513. [PMID: 29261717 PMCID: PMC5738045 DOI: 10.1371/journal.pone.0189513] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 11/28/2017] [Indexed: 01/20/2023] Open
Abstract
α7 nicotinic acetylcholine receptors (nAChRs) and N-methyl-D-aspartate receptors (NMDARs) are key mediators of central cholinergic and glutamatergic neurotransmission, respectively. In addition to numerous well-established functional interactions between α7 nAChRs and NMDARs, the two receptors have been proposed to form a multimeric complex, and in the present study we have investigated this putative α7 nAChR/NMDAR assembly in human and murine brain tissues. By α-bungarotoxin (BGT) affinity purification, α7 and NMDAR subunits were co-purified from human and murine cortical and hippocampal homogenates, substantiating the notion that the receptors are parts of a multimeric complex in the human and rodent brain. Interestingly, the ratios between GluN1 and α7 levels in BGT pull-downs from cortical homogenates from Alzheimer's disease (AD) brains were significantly lower than those in pull-downs from non-AD controls, indicating a reduced degree of α7 nAChR/NMDAR complex formation in the diseased tissue. A similar difference in GluN1/α7 ratios was observed between pull-downs from cortical homogenates from adult 3xTg-AD and age-matched wild type (WT) mice, whereas the GluN1/α7 ratios determined in pull-downs from young 3xTg-AD and age-matched WT mice did not differ significantly. The observation that pretreatment with oligomeric amyloid-β1-42 reduced GluN1/α7 ratios in BGT pull-downs from human cortical homogenate in a concentration-dependent manner provided a plausible molecular mechanism for this observed reduction. In conclusion, while it will be important to further challenge the existence of the putative α7 nAChR/NMDAR complex in future studies applying other methodologies than biochemical assays and to investigate the functional implications of this complex for cholinergic and glutamatergic neurotransmission, this work supports the formation of the complex and presents new insights into its regulation in healthy and diseased brain tissue.
Collapse
Affiliation(s)
- Mohamed R. Elnagar
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken, Copenhagen Ø, Denmark
- Faculty of Pharmacy, Al-Azhar University, Al-Mokhaym Al-Daem, Nasr City, Cairo, Egypt
| | - Anne Byriel Walls
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken, Copenhagen Ø, Denmark
| | - Gouda K. Helal
- Faculty of Pharmacy, Al-Azhar University, Al-Mokhaym Al-Daem, Nasr City, Cairo, Egypt
| | - Farid M. Hamada
- Faculty of Pharmacy, Al-Azhar University, Al-Mokhaym Al-Daem, Nasr City, Cairo, Egypt
| | - Morten Skøtt Thomsen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken, Copenhagen Ø, Denmark
| | - Anders A. Jensen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken, Copenhagen Ø, Denmark
| |
Collapse
|
26
|
Corsi-Zuelli FMDG, Brognara F, Quirino GFDS, Hiroki CH, Fais RS, Del-Ben CM, Ulloa L, Salgado HC, Kanashiro A, Loureiro CM. Neuroimmune Interactions in Schizophrenia: Focus on Vagus Nerve Stimulation and Activation of the Alpha-7 Nicotinic Acetylcholine Receptor. Front Immunol 2017; 8:618. [PMID: 28620379 PMCID: PMC5449450 DOI: 10.3389/fimmu.2017.00618] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 05/10/2017] [Indexed: 12/28/2022] Open
Abstract
Schizophrenia is one of the most debilitating mental disorders and is aggravated by the lack of efficacious treatment. Although its etiology is unclear, epidemiological studies indicate that infection and inflammation during development induces behavioral, morphological, neurochemical, and cognitive impairments, increasing the risk of developing schizophrenia. The inflammatory hypothesis of schizophrenia is also supported by clinical studies demonstrating systemic inflammation and microglia activation in schizophrenic patients. Although elucidating the mechanism that induces this inflammatory profile remains a challenge, mounting evidence suggests that neuroimmune interactions may provide therapeutic advantages to control inflammation and hence schizophrenia. Recent studies have indicated that vagus nerve stimulation controls both peripheral and central inflammation via alpha-7 nicotinic acetylcholine receptor (α7nAChR). Other findings have indicated that vagal stimulation and α7nAChR-agonists can provide therapeutic advantages for neuropsychiatric disorders, such as depression and epilepsy. This review analyzes the latest results regarding: (I) the immune-to-brain pathogenesis of schizophrenia; (II) the regulation of inflammation by the autonomic nervous system in psychiatric disorders; and (III) the role of the vagus nerve and α7nAChR in schizophrenia.
Collapse
Affiliation(s)
| | - Fernanda Brognara
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | | | - Carlos Hiroji Hiroki
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Rafael Sobrano Fais
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Cristina Marta Del-Ben
- Department of Neuroscience and Behavior, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Luis Ulloa
- Department of Surgery, Center of Immunology and Inflammation, Rutgers University-New Jersey Medical School, Newark, NJ, United States
| | - Helio Cesar Salgado
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Alexandre Kanashiro
- Department of Physiological Sciences, Federal University of São Carlos, São Carlos, Brazil
| | - Camila Marcelino Loureiro
- Department of Internal Medicine, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
27
|
McGregor AL, D'Souza G, Kim D, Tingle MD. Varenicline improves motor and cognitive deficits and decreases depressive-like behaviour in late-stage YAC128 mice. Neuropharmacology 2017; 116:233-246. [DOI: 10.1016/j.neuropharm.2016.12.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 12/21/2016] [Accepted: 12/21/2016] [Indexed: 02/03/2023]
|
28
|
Wang J, He X, Guo F, Cheng X, Wang Y, Wang X, Feng Z, Vreugdenhil M, Lu C. Multiple Kinases Involved in the Nicotinic Modulation of Gamma Oscillations in the Rat Hippocampal CA3 Area. Front Cell Neurosci 2017; 11:57. [PMID: 28321180 PMCID: PMC5337687 DOI: 10.3389/fncel.2017.00057] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 02/15/2017] [Indexed: 11/29/2022] Open
Abstract
Neuronal synchronization at gamma band frequency (20–80 Hz, γ oscillations) is closely associated with higher brain function, such as learning, memory and attention. Nicotinic acetylcholine receptors (nAChRs) are highly expressed in the hippocampus, and modulate hippocampal γ oscillations, but the intracellular mechanism underlying such modulation remains elusive. We explored multiple kinases by which nicotine can modulate γ oscillations induced by kainate in rat hippocampal area CA3 in vitro. We found that inhibitors of cyclic AMP dependent kinase (protein kinase A, PKA), protein kinase C (PKC), N-methyl-D-aspartate receptor (NMDA) receptors, Phosphoinositide 3-kinase (PI3K) and extracellular signal-related kinases (ERK), each individually could prevent the γ oscillation-enhancing effect of 1 μM nicotine, whereas none of them affected baseline γ oscillation strength. Inhibition of the serine/threonine kinase Akt increased baseline γ oscillations and partially blocked its nicotinic enhancement. We propose that the PKA-NMDAR-PI3K-ERK pathway modifies cellular properties required for the nicotinic enhancement of γ oscillations, dependent on a PKC-ERK mediated pathway. These signaling pathways provide clues for restoring γ oscillations in pathological conditions affecting cognition. The suppression of γ oscillations at 100 μM nicotine was only dependent on PKA-NMDAR activation and may be due to very high intracellular calcium levels.
Collapse
Affiliation(s)
- JianGang Wang
- Key Laboratory for the Brain Research of Henan Province, Xinxiang Medical UniversityXinxinang, China; Department of Pathophysiology, Xinxiang Medical UniversityXinxinang, China
| | - XiaoLong He
- Key Laboratory of Neuronal Oscillation and Disease, Yantze University Medical School JingZhou, China
| | - Fangli Guo
- Key Laboratory for the Brain Research of Henan Province, Xinxiang Medical UniversityXinxinang, China; Department of Neurobiology and Physiology, Xinxiang Medical UniversityXinxinang, China
| | - XiangLin Cheng
- Department of Laboratory Medicine, Yantze University Affiliated Hospital JingZhou, China
| | - Yali Wang
- Key Laboratory for the Brain Research of Henan Province, Xinxiang Medical UniversityXinxinang, China; Department of Neurobiology and Physiology, Xinxiang Medical UniversityXinxinang, China
| | - XiaoFang Wang
- Key Laboratory for the Brain Research of Henan Province, Xinxiang Medical University Xinxinang, China
| | - ZhiWei Feng
- Key Laboratory for the Brain Research of Henan Province, Xinxiang Medical University Xinxinang, China
| | - Martin Vreugdenhil
- Department of Psychology, Xinxiang Medical UniversityXinxinang, China; School of Life Sciences, Birmingham City UniversityBirmingham, UK
| | - ChengBiao Lu
- Key Laboratory for the Brain Research of Henan Province, Xinxiang Medical UniversityXinxinang, China; Key Laboratory of Neuronal Oscillation and Disease, Yantze University Medical SchoolJingZhou, China; Department of Neurobiology and Physiology, Xinxiang Medical UniversityXinxinang, China
| |
Collapse
|
29
|
Hopf FW. Do specific NMDA receptor subunits act as gateways for addictive behaviors? GENES BRAIN AND BEHAVIOR 2016; 16:118-138. [PMID: 27706932 DOI: 10.1111/gbb.12348] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 09/27/2016] [Accepted: 10/03/2016] [Indexed: 12/19/2022]
Abstract
Addiction to alcohol and drugs is a major social and economic problem, and there is considerable interest in understanding the molecular mechanisms that promote addictive drives. A number of proteins have been identified that contribute to expression of addictive behaviors. NMDA receptors (NMDARs), a subclass of ionotropic glutamate receptors, have been of particular interest because their physiological properties make them an attractive candidate for gating induction of synaptic plasticity, a molecular change thought to mediate learning and memory. NMDARs are generally inactive at the hyperpolarized resting potentials of many neurons. However, given sufficient depolarization, NMDARs are activated and exhibit long-lasting currents with significant calcium permeability. Also, in addition to stimulating neurons by direct depolarization, NMDARs and their calcium signaling can allow strong and/or synchronized inputs to produce long-term changes in other molecules (such as AMPA-type glutamate receptors) which can last from days to years, binding internal and external stimuli in a long-term memory trace. Such memories could allow salient drug-related stimuli to exert strong control over future behaviors and thus promote addictive drives. Finally, NMDARs may themselves undergo plasticity, which can alter subsequent neuronal stimulation and/or the ability to induce plasticity. This review will address recent and past findings suggesting that NMDAR activity promotes drug- and alcohol-related behaviors, with a particular focus on GluN2B subunits as possible central regulators of many addictive behaviors, as well as newer studies examining the importance of non-canonical NMDAR subunits and endogenous NMDAR cofactors.
Collapse
Affiliation(s)
- F W Hopf
- Alcohol and Addiction Research Group, Department of Neurology, University of California at San Francisco, San Francisco, CA, USA
| |
Collapse
|
30
|
Marcus MM, Björkholm C, Malmerfelt A, Möller A, Påhlsson N, Konradsson-Geuken Å, Feltmann K, Jardemark K, Schilström B, Svensson TH. Alpha7 nicotinic acetylcholine receptor agonists and PAMs as adjunctive treatment in schizophrenia. An experimental study. Eur Neuropsychopharmacol 2016; 26:1401-1411. [PMID: 27474687 DOI: 10.1016/j.euroneuro.2016.07.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 06/14/2016] [Accepted: 07/02/2016] [Indexed: 10/21/2022]
Abstract
Nicotine has been found to improve cognition and reduce negative symptoms in schizophrenia and a genetic and pathophysiological link between the α7 nicotinic acetylcholine receptors (nAChRs) and schizophrenia has been demonstrated. Therefore, there has been a large interest in developing drugs affecting the α7 nAChRs for schizophrenia. In the present study we investigated, in rats, the effects of a selective α7 agonist (PNU282987) and a α7 positive allosteric modulator (PAM; NS1738) alone and in combination with the atypical antipsychotic drug risperidone for their utility as adjunct treatment in schizophrenia. Moreover we also investigated their utility as adjunct treatment in depression in combination with the SSRI citalopram. We found that NS1738 and to some extent also PNU282987, potentiated a subeffective dose of risperidone in the conditioned avoidance response test. Both drugs also potentiated the effect of a sub-effective concentration of risperidone on NMDA-induced currents in pyramidal cells of the medial prefrontal cortex. Moreover, NS1738 and PNU282987 enhanced recognition memory in the novel object recognition test, when given separately. Both drugs also potentiated accumbal but not prefrontal risperidone-induced dopamine release. Finally, PNU282987 reduced immobility in the forced swim test, indicating an antidepressant-like effect. Taken together, our data support the utility of drugs targeting the α7 nAChRs, perhaps especially α7 PAMs, to potentiate the effect of atypical antipsychotic drugs. Moreover, our data suggest that α7 agonists and PAMs can be used to ameliorate cognitive symptoms in schizophrenia and depression.
Collapse
Affiliation(s)
- Monica M Marcus
- Department of Physiology and Pharmacology, Section of Neuropsychopharmacology, Karolinska Institutet, S-171 77 Stockholm, Sweden
| | - Carl Björkholm
- Department of Physiology and Pharmacology, Section of Neuropsychopharmacology, Karolinska Institutet, S-171 77 Stockholm, Sweden
| | - Anna Malmerfelt
- Department of Physiology and Pharmacology, Section of Neuropsychopharmacology, Karolinska Institutet, S-171 77 Stockholm, Sweden
| | - Annie Möller
- Department of Physiology and Pharmacology, Section of Neuropsychopharmacology, Karolinska Institutet, S-171 77 Stockholm, Sweden
| | - Ninni Påhlsson
- Department of Physiology and Pharmacology, Section of Neuropsychopharmacology, Karolinska Institutet, S-171 77 Stockholm, Sweden
| | - Åsa Konradsson-Geuken
- Department of Physiology and Pharmacology, Section of Neuropsychopharmacology, Karolinska Institutet, S-171 77 Stockholm, Sweden
| | - Kristin Feltmann
- Department of Physiology and Pharmacology, Section of Neuropsychopharmacology, Karolinska Institutet, S-171 77 Stockholm, Sweden
| | - Kent Jardemark
- Department of Physiology and Pharmacology, Section of Neuropsychopharmacology, Karolinska Institutet, S-171 77 Stockholm, Sweden
| | - Björn Schilström
- Department of Physiology and Pharmacology, Section of Neuropsychopharmacology, Karolinska Institutet, S-171 77 Stockholm, Sweden
| | - Torgny H Svensson
- Department of Physiology and Pharmacology, Section of Neuropsychopharmacology, Karolinska Institutet, S-171 77 Stockholm, Sweden.
| |
Collapse
|
31
|
Chen T, Zhang B, Li G, Chen L, Chen L. Simvastatin enhances NMDA receptor GluN2B expression and phosphorylation of GluN2B and GluN2A through increased histone acetylation and Src signaling in hippocampal CA1 neurons. Neuropharmacology 2016; 107:411-421. [PMID: 27016018 DOI: 10.1016/j.neuropharm.2016.03.028] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 03/18/2016] [Accepted: 03/21/2016] [Indexed: 11/25/2022]
Abstract
Simvastatin (SV) can improve cognitive deficits in Alzheimer's disease patients and mice. Herein, we report that the administration of SV (20 mg/kg) for 5 days in mice (SV-mice) or the treatment of slices with SV (10 μM) for 4 h (SV-slices) could increase the density of NMDA-evoked inward currents (INMDA) in hippocampal CA1 pyramidal cells, which were blocked by farnesol (FOH) that converts farnesyl pyrophosphate (FPP), but not geranylgeraniol (GGOH) that increases geranylgeranylpyrophosphate (GGPP). Sensitivity of INMDA to ifenprodil in SV-mice or SV-slices was significantly increased. The levels of hippocampal GluN2B and GluN2A or Src phosphorylation in SV-mice or SV-slices were higher than controls, which were sensitive to FOH. The Src inhibitor PP2 could inhibit the SV-enhanced phosphorylation of GluN2B and GluN2A and SV-augmented INMDA, but PI3K inhibitor LY294002 did not. The levels of GluN2B mRNA and protein were elevated in SV-mice, which was abolished by FOH, but not by GGOH or PP2. Furthermore, the histone H3K9 and H3K27 acetylation of GluN2B promoter was increased in SV-mice, which was suppressed by FOH rather than GGOH or PP2. In control mice and slices, the reduction of FPP by farnesyl transferase inhibitor could increase the levels of GluN2B expression, the histone H3K9 and H3K27 acetylation and enhance the phosphorylation of GluN2B, GluN2A and Src. The findings indicate that the administration of SV can enhance GluN2B expression and GluN2B and GluN2A phosphorylation leading to augmentation of NMDAR activity through reducing FPP to increase histone acetylation of GluN2B and Src signaling.
Collapse
Affiliation(s)
- Tingting Chen
- State Key Laboratory of Reproductive Medicine, China; Department of Physiology, Nanjing Medical University, Nanjing 210029, China
| | - Baofeng Zhang
- State Key Laboratory of Reproductive Medicine, China; Department of Physiology, Nanjing Medical University, Nanjing 210029, China
| | - Guoxi Li
- Department of Physiology, Nanjing Medical University, Nanjing 210029, China
| | - Lei Chen
- Department of Physiology, Nanjing Medical University, Nanjing 210029, China
| | - Ling Chen
- State Key Laboratory of Reproductive Medicine, China; Department of Physiology, Nanjing Medical University, Nanjing 210029, China.
| |
Collapse
|
32
|
Zhang H, Li T, Li S, Liu F. Cross-talk between α7 nAchR and NMDAR revealed by protein profiling. J Proteomics 2015; 131:113-121. [PMID: 26498070 DOI: 10.1016/j.jprot.2015.10.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 09/15/2015] [Accepted: 10/14/2015] [Indexed: 10/22/2022]
Abstract
Functional regulation of NMDA receptor (NMDAR) by the activation of α7 nicotinic acetylcholine receptor (α7nAChR) has been reported, although the molecular signaling pathway underlying this process remains largely unknown. We employed a label-free quantitative proteomics approach to identify potential intracellular molecules and pathways that might be involved in the functional cross-talk between NMDAR and α7nAChR. 43 proteins showed significantly expression changes after choline treatment in which 35 out of 43 proteins was significantly altered by co-treatment with NMDA. Western blot analysis verified proteins expression determined by LC-MS. Furthermore, protein expression in vivo in neurons from fetal rats were visualized and quantified by Confocal microscopy,which showed consistency of relative changes of AHA-1 expressionmeasured by LC-MS and Western blot. Biological network analysis of differently expressed proteins found 21 kind of biological pathways involved. Of those pathways, 6 pathways were directly involved in regulation of neurotransmitters. Lastly, the levels of neurotransmitters (dopamine, glutamate, GABA and 5-HT) were measured by HPLC-ECD. Co-treatment choline/NMDA significantly enhances the release of dopamine, glutamate and GABA and dramatically decrease 5-HT to only 65% of control level, which is consist with this protein interaction network analysis, providing an additional evidence for the cross-talk between NMDAR and α7nAChR.
Collapse
Affiliation(s)
- Hailong Zhang
- Campbell Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario M5T 1R8, Canada
| | - Tao Li
- Campbell Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario M5T 1R8, Canada; Medical and Forensic College, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, PR China
| | - Shupeng Li
- Campbell Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario M5T 1R8, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario M5T 1R8, Canada
| | - Fang Liu
- Campbell Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario M5T 1R8, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario M5T 1R8, Canada.
| |
Collapse
|
33
|
Bertrand D, Lee CHL, Flood D, Marger F, Donnelly-Roberts D. Therapeutic Potential of α7 Nicotinic Acetylcholine Receptors. Pharmacol Rev 2015; 67:1025-73. [PMID: 26419447 DOI: 10.1124/pr.113.008581] [Citation(s) in RCA: 118] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2025] Open
Abstract
Progress in the fields of neuroscience and molecular biology has identified the forebrain cholinergic system as being important in many higher order brain functions. Further analysis of the genes encoding the nicotinic acetylcholine receptors (nAChRs) has highlighted, in particular, the role of α7 nAChRs in these higher order brain functions as evidenced by their peculiar physiologic and pharmacological properties. As this receptor has gained the attention of scientists from academia and industry, our knowledge of its roles in various brain and bodily functions has increased immensely. We have also seen the development of small molecules that have further refined our understanding of the roles of α7 nAChRs, and these molecules have begun to be tested in clinical trials for several indications. Although a large body of data has confirmed a role of α7 nAChRs in cognition, the translation of small molecules affecting α7 nAChRs into therapeutics has to date only progressed to the stage of testing in clinical trials. Notably, however, most recent human genetic and biochemical studies are further underscoring the crucial role of α7 nAChRs and associated genes in multiple organ systems and disease states. The aim of this review is to discuss our current knowledge of α7 nAChRs and their relevance as a target in specific functional systems and disease states.
Collapse
Affiliation(s)
- Daniel Bertrand
- HiQScreen Sàrl, Geneva, Switzerland (D.B., F.M.); AbbVie Inc., North Chicago, Illinois (C-H.L.L., D.D-R.); and FORUM Pharmaceuticals Inc., Waltham, Massachusetts (D.F.)
| | - Chih-Hung L Lee
- HiQScreen Sàrl, Geneva, Switzerland (D.B., F.M.); AbbVie Inc., North Chicago, Illinois (C-H.L.L., D.D-R.); and FORUM Pharmaceuticals Inc., Waltham, Massachusetts (D.F.)
| | - Dorothy Flood
- HiQScreen Sàrl, Geneva, Switzerland (D.B., F.M.); AbbVie Inc., North Chicago, Illinois (C-H.L.L., D.D-R.); and FORUM Pharmaceuticals Inc., Waltham, Massachusetts (D.F.)
| | - Fabrice Marger
- HiQScreen Sàrl, Geneva, Switzerland (D.B., F.M.); AbbVie Inc., North Chicago, Illinois (C-H.L.L., D.D-R.); and FORUM Pharmaceuticals Inc., Waltham, Massachusetts (D.F.)
| | - Diana Donnelly-Roberts
- HiQScreen Sàrl, Geneva, Switzerland (D.B., F.M.); AbbVie Inc., North Chicago, Illinois (C-H.L.L., D.D-R.); and FORUM Pharmaceuticals Inc., Waltham, Massachusetts (D.F.)
| |
Collapse
|
34
|
Cheng Q, Yakel JL. The effect of α7 nicotinic receptor activation on glutamatergic transmission in the hippocampus. Biochem Pharmacol 2015. [PMID: 26212541 DOI: 10.1016/j.bcp.2015.07.015] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Nicotinic acetylcholine receptors (nAChRs) are expressed widely in the CNS, and mediate both synaptic and perisynaptic activities of endogenous cholinergic inputs and pharmacological actions of exogenous compounds (e.g., nicotine and choline). Behavioral studies indicate that nicotine improves such cognitive functions as learning and memory, however the cellular mechanism of these actions remains elusive. With help from newly developed biosensors and optogenetic tools, recent studies provide new insights on signaling mechanisms involved in the activation of nAChRs. Here we will review α7 nAChR's action in the tri-synaptic pathway in the hippocampus. The effects of α7 nAChR activation via either exogenous compounds or endogenous cholinergic innervation are detailed for spontaneous and evoked glutamatergic synaptic transmission and synaptic plasticity, as well as the underlying signaling mechanisms. In summary, α7 nAChRs trigger intracellular calcium rise and calcium-dependent signaling pathways to enhance glutamate release and induce glutamatergic synaptic plasticity.
Collapse
Affiliation(s)
- Qing Cheng
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC 27709, USA
| | - Jerrel L Yakel
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC 27709, USA
| |
Collapse
|
35
|
Varga D, Herédi J, Kánvási Z, Ruszka M, Kis Z, Ono E, Iwamori N, Iwamori T, Takakuwa H, Vécsei L, Toldi J, Gellért L. Systemic L-Kynurenine sulfate administration disrupts object recognition memory, alters open field behavior and decreases c-Fos immunopositivity in C57Bl/6 mice. Front Behav Neurosci 2015; 9:157. [PMID: 26136670 PMCID: PMC4468612 DOI: 10.3389/fnbeh.2015.00157] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 06/01/2015] [Indexed: 01/31/2023] Open
Abstract
L-Kynurenine (L-KYN) is a central metabolite of tryptophan degradation through the kynurenine pathway (KP). The systemic administration of L-KYN sulfate (L-KYNs) leads to a rapid elevation of the neuroactive KP metabolite kynurenic acid (KYNA). An elevated level of KYNA may have multiple effects on the synaptic transmission, resulting in complex behavioral changes, such as hypoactivity or spatial working memory deficits. These results emerged from studies that focused on rats, after low-dose L-KYNs treatment. However, in several studies neuroprotection was achieved through the administration of high-dose L-KYNs. In the present study, our aim was to investigate whether the systemic administration of a high dose of L-KYNs (300 mg/bwkg; i.p.) would produce alterations in behavioral tasks (open field or object recognition) in C57Bl/6j mice. To evaluate the changes in neuronal activity after L-KYNs treatment, in a separate group of animals we estimated c-Fos expression levels in the corresponding subcortical brain areas. The L-KYNs treatment did not affect the general ambulatory activity of C57Bl/6j mice, whereas it altered their moving patterns, elevating the movement velocity and resting time. Additionally, it seemed to increase anxiety-like behavior, as peripheral zone preference of the open field arena emerged and the rearing activity was attenuated. The treatment also completely abolished the formation of object recognition memory and resulted in decreases in the number of c-Fos-immunopositive-cells in the dorsal part of the striatum and in the CA1 pyramidal cell layer of the hippocampus. We conclude that a single exposure to L-KYNs leads to behavioral disturbances, which might be related to the altered basal c-Fos protein expression in C57Bl/6j mice.
Collapse
Affiliation(s)
- Dániel Varga
- Department of Physiology, Anatomy and Neuroscience, University of Szeged Szeged, Hungary
| | - Judit Herédi
- Department of Physiology, Anatomy and Neuroscience, University of Szeged Szeged, Hungary
| | - Zita Kánvási
- Department of Physiology, Anatomy and Neuroscience, University of Szeged Szeged, Hungary
| | - Marian Ruszka
- Department of Physiology, Anatomy and Neuroscience, University of Szeged Szeged, Hungary ; Department of Neurology, Faculty of Medicine, MTA-SZTE Neuroscience Research Group, University of Szeged Szeged, Hungary
| | - Zsolt Kis
- Department of Physiology, Anatomy and Neuroscience, University of Szeged Szeged, Hungary
| | - Etsuro Ono
- Department of Biomedicine, Graduate School of Medical Sciences, Kyushu University Fukuoka, Japan ; Center of Biomedical Research, Research Center for Human Disease Modeling, Department of Physiological Sciences, Graduate School of Medical Sciences, Kyushu University Fukuoka, Japan
| | - Naoki Iwamori
- Department of Biomedicine, Graduate School of Medical Sciences, Kyushu University Fukuoka, Japan ; Center of Biomedical Research, Research Center for Human Disease Modeling, Department of Physiological Sciences, Graduate School of Medical Sciences, Kyushu University Fukuoka, Japan
| | - Tokuko Iwamori
- Department of Biomedicine, Graduate School of Medical Sciences, Kyushu University Fukuoka, Japan ; Center of Biomedical Research, Research Center for Human Disease Modeling, Department of Physiological Sciences, Graduate School of Medical Sciences, Kyushu University Fukuoka, Japan
| | - Hiroki Takakuwa
- Faculty of Life Sciences, Kyoto Sangyo University, Kamigamo-Motoyama Kita, Kyoto, Japan
| | - László Vécsei
- Department of Neurology, Faculty of Medicine, MTA-SZTE Neuroscience Research Group, University of Szeged Szeged, Hungary ; Department of Neurology, University of Szeged, Hungary Szeged, Hungary
| | - József Toldi
- Department of Physiology, Anatomy and Neuroscience, University of Szeged Szeged, Hungary ; Department of Neurology, Faculty of Medicine, MTA-SZTE Neuroscience Research Group, University of Szeged Szeged, Hungary
| | - Levente Gellért
- Department of Physiology, Anatomy and Neuroscience, University of Szeged Szeged, Hungary ; Department of Neurology, Faculty of Medicine, MTA-SZTE Neuroscience Research Group, University of Szeged Szeged, Hungary
| |
Collapse
|
36
|
Yu JY, Zhang B, Peng L, Wu CH, Cao H, Zhong JF, Hoffman J, Huang SH. Repositioning of Memantine as a Potential Novel Therapeutic Agent against Meningitic E. coli-Induced Pathogenicities through Disease-Associated Alpha7 Cholinergic Pathway and RNA Sequencing-Based Transcriptome Analysis of Host Inflammatory Responses. PLoS One 2015; 10:e0121911. [PMID: 25993608 PMCID: PMC4437645 DOI: 10.1371/journal.pone.0121911] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 02/07/2015] [Indexed: 01/09/2023] Open
Abstract
Neonatal sepsis and meningitis (NSM) remains a leading cause worldwide of mortality and morbidity in newborn infants despite the availability of antibiotics over the last several decades. E. coli is the most common gram-negative pathogen causing NSM. Our previous studies show that α7 nicotinic receptor (α7 nAChR), an essential regulator of inflammation, plays a detrimental role in the host defense against NSM. Despite notable successes, there still exists an unmet need for new effective therapeutic approaches to treat this disease. Using the in vitro/in vivo models of the blood-brain barrier (BBB) and RNA-seq, we undertook a drug repositioning study to identify unknown antimicrobial activities for known drugs. We have demonstrated for the first time that memantine (MEM), a FDA-approved drug for treatment of Alzheimer’s disease, could very efficiently block E. coli-caused bacteremia and meningitis in a mouse model of NSM in a manner dependent on α7 nAChR. MEM was able to synergistically enhance the antibacterial activity of ampicillin in HBMEC infected with E. coli K1 (E44) and in neonatal mice with E44-caused bacteremia and meningitis. Differential gene expression analysis of RNA-Seq data from mouse BMEC infected with E. coli K1 showed that several E44-increased inflammatory factors, including IL33, IL18rap, MMP10 and Irs1, were significantly reduced by MEM compared to the infected cells without drug treatment. MEM could also significantly up-regulate anti-inflammatory factors, including Tnfaip3, CISH, Ptgds and Zfp36. Most interestingly, these factors may positively and negatively contribute to regulation of NF-κB, which is a hallmark feature of bacterial meningitis. Furthermore, we have demonstrated that circulating BMEC (cBMEC) are the potential novel biomarkers for NSM. MEM could significantly reduce E44-increased blood level of cBMEC in mice. Taken together, our data suggest that memantine can efficiently block host inflammatory responses to bacterial infection through modulation of both inflammatory and anti-inflammatory pathways.
Collapse
Affiliation(s)
- Jing-Yi Yu
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health and Tropical Medicine, Southern Medical University, Guangzhou 510515, China; Saban Research Institute of Children's Hospital Los Angeles, Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90027, United States of America
| | - Bao Zhang
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health and Tropical Medicine, Southern Medical University, Guangzhou 510515, China; Saban Research Institute of Children's Hospital Los Angeles, Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90027, United States of America
| | - Liang Peng
- Saban Research Institute of Children's Hospital Los Angeles, Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90027, United States of America; Department of Clinic Laboratory, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Chun-Hua Wu
- Saban Research Institute of Children's Hospital Los Angeles, Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90027, United States of America
| | - Hong Cao
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health and Tropical Medicine, Southern Medical University, Guangzhou 510515, China
| | - John F Zhong
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, United States of America; Department of Perio, Diagnostic Sciences & Biomedical Sciences, School of Dentistry, University of Southern California, Los Angeles, CA, 93003, United States of America; Department of Pediatrics, School of Medicine, University of Southern California, Los Angeles, CA, 93003, United States of America
| | - Jill Hoffman
- Saban Research Institute of Children's Hospital Los Angeles, Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90027, United States of America
| | - Sheng-He Huang
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health and Tropical Medicine, Southern Medical University, Guangzhou 510515, China; Saban Research Institute of Children's Hospital Los Angeles, Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90027, United States of America
| |
Collapse
|
37
|
Marchi M, Grilli M, Pittaluga AM. Nicotinic modulation of glutamate receptor function at nerve terminal level: a fine-tuning of synaptic signals. Front Pharmacol 2015; 6:89. [PMID: 25972809 PMCID: PMC4413670 DOI: 10.3389/fphar.2015.00089] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 04/10/2015] [Indexed: 11/13/2022] Open
Abstract
This review focuses on a specific interaction occurring between the nicotinic cholinergic receptors (nAChRs) and the glutamatergic receptors (GluRs) at the nerve endings level. We have employed synaptosomes in superfusion and supplemented and integrated our findings with data obtained using techniques from molecular biology and immuno-cytochemistry, and the assessment of receptor trafficking. In particular, we characterize the following: (1) the direct and unequivocal localization of native α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) and N-methyl-D-aspartate (NMDA) glutamatergic receptors on specific nerve terminals, (2) their pharmacological characterization and functional co-localization with nAChRs on the same nerve endings, and (3) the existence of synergistic or antagonistic interactions among them. Indeed, in the rat nucleus accumbens (NAc), the function of some AMPA and NMDA receptors present on the dopaminergic and glutamatergic nerve terminals can be regulated negatively or positively in response to a brief activation of nAChRs. This effect occurs rapidly and involves the trafficking of AMPA and NMDA receptors. The event takes place also at very low concentrations of nicotine and involves the activation of several nAChRs subtypes. This dynamic control by cholinergic nicotinic system of glutamatergic NMDA and AMPA receptors might therefore represent an important neuronal presynaptic adaptation associated with nicotine administration. The understanding of the role of these nicotine-induced functional changes might open new and interesting perspectives both in terms of explaining the mechanisms that underlie some of the effects of nicotine addiction and in the development of new drugs for smoking cessation.
Collapse
Affiliation(s)
- Mario Marchi
- Department of Pharmacy, Pharmacology and Toxicology Section, University of Genoa , Genoa, Italy ; Center of Excellence for Biomedical Research, University of Genoa , Genoa, Italy
| | - Massimo Grilli
- Department of Pharmacy, Pharmacology and Toxicology Section, University of Genoa , Genoa, Italy
| | - Anna M Pittaluga
- Department of Pharmacy, Pharmacology and Toxicology Section, University of Genoa , Genoa, Italy ; Center of Excellence for Biomedical Research, University of Genoa , Genoa, Italy
| |
Collapse
|
38
|
Enhanced NMDA receptor-mediated modulation of excitatory neurotransmission in the dorsal vagal complex of streptozotocin-treated, chronically hyperglycemic mice. PLoS One 2015; 10:e0121022. [PMID: 25799386 PMCID: PMC4370733 DOI: 10.1371/journal.pone.0121022] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 02/09/2015] [Indexed: 11/19/2022] Open
Abstract
A variety of metabolic disorders, including complications experienced by diabetic patients, have been linked to altered neural activity in the dorsal vagal complex. This study tested the hypothesis that augmentation of N-Methyl-D-Aspartate (NMDA) receptor-mediated responses in the vagal complex contributes to increased glutamate release in the dorsal motor nucleus of the vagus nerve (DMV) in mice with streptozotocin-induced chronic hyperglycemia (i.e., hyperglycemic mice), a model of type 1 diabetes. Antagonism of NMDA receptors with AP-5 (100 μM) suppressed sEPSC frequency in vagal motor neurons recorded in vitro, confirming that constitutively active NMDA receptors regulate glutamate release in the DMV. There was a greater relative effect of NMDA receptor antagonism in hyperglycemic mice, suggesting that augmented NMDA effects occur in neurons presynaptic to the DMV. Effects of NMDA receptor blockade on mEPSC frequency were equivalent in control and diabetic mice, suggesting that differential effects on glutamate release were due to altered NMDA function in the soma-dendritic membrane of intact afferent neurons. Application of NMDA (300 μM) resulted in greater inward current and current density in NTS neurons recorded from hyperglycemic than control mice, particularly in glutamatergic NTS neurons identified by single-cell RT-PCR for VGLUT2. Overall expression of NR1 protein and message in the dorsal vagal complex were not different between the two groups. Enhanced postsynaptic NMDA responsiveness of glutamatergic NTS neurons is consistent with tonically-increased glutamate release in the DMV in mice with chronic hyperglycemia. Functional augmentation of NMDA-mediated responses may serve as a physiological counter-regulatory mechanism to control pathological disturbances of homeostatic autonomic function in type 1 diabetes.
Collapse
|
39
|
Tian S, Pan S, You Y. Nicotine enhances the reconsolidation of novel object recognition memory in rats. Pharmacol Biochem Behav 2015; 129:14-8. [DOI: 10.1016/j.pbb.2014.11.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 11/24/2014] [Accepted: 11/29/2014] [Indexed: 12/23/2022]
|
40
|
Zappettini S, Grilli M, Olivero G, Chen J, Padolecchia C, Pittaluga A, Tomé AR, Cunha RA, Marchi M. Nicotinic α7 receptor activation selectively potentiates the function of NMDA receptors in glutamatergic terminals of the nucleus accumbens. Front Cell Neurosci 2014; 8:332. [PMID: 25360085 PMCID: PMC4199379 DOI: 10.3389/fncel.2014.00332] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 09/30/2014] [Indexed: 11/13/2022] Open
Abstract
We here provide functional and immunocytochemical evidence supporting the co-localization and functional interaction between nicotinic acetylcholine receptors (nAChRs) and N-methyl-D-aspartic acid receptors (NMDARs) in glutamatergic terminals of the nucleus accumbens (NAc). Immunocytochemical studies showed that a significant percentage of NAc terminals were glutamatergic and possessed GluN1 and α7-containing nAChR. A short-term pre-exposure of synaptosomes to nicotine (30 µM) or choline (1 mM) caused a significant potentiation of the 100 µM NMDA-evoked [3H]D-aspartate ([3H]D-Asp) outflow, which was prevented by α-bungarotoxin (100 nM). The pre-exposure to nicotine (100 µM) or choline (1 mM) also enhanced the NMDA-induced cytosolic free calcium levels, as measured by FURA-2 fluorescence imaging in individual NAc terminals, an effect also prevented by α-bungarotoxin. Pre-exposure to the α4-nAChR agonists 5IA85380 (10 nM) or RJR2429 (1 µM) did not modify NMDA-evoked ([3H]D-Asp) outflow and calcium transients. The NMDA-evoked ([3H]D-Asp) overflow was partially antagonized by the NMDAR antagonists MK801, D-AP5, 5,7-DCKA and R(-)CPP and unaffected by the GluN2B-NMDAR antagonists Ro256981 and ifenprodil. Notably, pre-treatment with choline increased GluN2A biotin-tagged proteins. In conclusion, our results show that the GluN2A-NMDA receptor function can be positively regulated in NAc terminals in response to a brief incubation with α7 but not α4 nAChRs agonists. This might be a general feature in different brain areas since a similar nAChR-mediated bolstering of NMDA-induced ([3H]D-Asp) overflow was also observed in hippocampal synaptosomes.
Collapse
Affiliation(s)
- Stefania Zappettini
- Faculté de Médecine, Institut de Neurosciences des Systèmes Inserm UMR1106, Aix Marseille Université La Timone Marseille, France
| | - Massimo Grilli
- Department of Pharmacy, University of Genoa, Viale Cembrano Genoa, Italy
| | - Guendalina Olivero
- Department of Pharmacy, University of Genoa, Viale Cembrano Genoa, Italy
| | - Jiayang Chen
- Department of Pharmacy, University of Genoa, Viale Cembrano Genoa, Italy
| | | | - Anna Pittaluga
- Department of Pharmacy, University of Genoa, Viale Cembrano Genoa, Italy ; Center of Excellence for Biomedical Research, University of Genoa Genoa, Italy
| | - Angelo R Tomé
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra Coimbra, Portugal ; Department of Life Sciences, Faculty of Sciences and Technology, University of Coimbra Coimbra, Portugal
| | - Rodrigo A Cunha
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra Coimbra, Portugal ; Faculty of Medicine, University of Coimbra Coimbra, Portugal
| | - Mario Marchi
- Department of Pharmacy, University of Genoa, Viale Cembrano Genoa, Italy ; Center of Excellence for Biomedical Research, University of Genoa Genoa, Italy
| |
Collapse
|
41
|
Li S, Wong AHC, Liu F. Ligand-gated ion channel interacting proteins and their role in neuroprotection. Front Cell Neurosci 2014; 8:125. [PMID: 24847210 PMCID: PMC4023026 DOI: 10.3389/fncel.2014.00125] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Accepted: 04/21/2014] [Indexed: 11/29/2022] Open
Abstract
Ion channel receptors are a vital component of nervous system signaling, allowing rapid and direct conversion of a chemical neurotransmitter message to an electrical current. In recent decades, it has become apparent that ionotropic receptors are regulated by protein-protein interactions with other ion channels, G-protein coupled receptors and intracellular proteins. These other proteins can also be modulated by these interactions with ion channel receptors. This bidirectional functional cross-talk is important for critical cellular functions such as excitotoxicity in pathological and disease states like stroke, and for the basic dynamics of activity-dependent synaptic plasticity. Protein interactions with ion channel receptors can therefore increase the computational capacity of neuronal signaling cascades and also represent a novel target for therapeutic intervention in neuropsychiatric disease. This review will highlight some examples of ion channel receptor interactions and their potential clinical utility for neuroprotection.
Collapse
Affiliation(s)
- Shupeng Li
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, University of Toronto Toronto, ON, Canada
| | - Albert H C Wong
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, University of Toronto Toronto, ON, Canada
| | - Fang Liu
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, University of Toronto Toronto, ON, Canada
| |
Collapse
|