1
|
Meur S, Karati D. Fyn Kinase in Alzheimer's Disease: Unraveling Molecular Mechanisms and Therapeutic Implications. Mol Neurobiol 2025; 62:643-660. [PMID: 38890236 DOI: 10.1007/s12035-024-04286-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 06/05/2024] [Indexed: 06/20/2024]
Abstract
Alzheimer's disease, characterized by the accumulation of abnormal protein aggregates and neuronal damage in the brain, leads to a gradual decline in cognitive function and memory. As a complex neurodegenerative disorder, it involves disruptions in various biochemical pathways and neurotransmitter systems, contributing to the progressive loss of neurons and synaptic connections. The complexity of Alzheimer's signaling pathways complicates treatment, presenting a formidable challenge in the quest for effective therapeutic interventions. A member of the Src family of kinases (SFKs), Fyn, is a type of non-receptor tyrosine kinase that has been linked to multiple essential CNS processes, such as myelination and synaptic transmission. Fyn is an appealing target for AD treatments because it is uniquely linked to the two major pathologies in AD by its interaction with tau, in addition to being activated by amyloid-beta (Aβ) through PrPC. Fyn mediates neurotoxicity and synaptic impairments caused by Aβ and is involved in regulating the process of Aβ synthesis.Additionally, the tau protein's tyrosine phosphorylation is induced by Fyn. Fyn is also a challenging target because of its widespread body expression and strong homology with other kinases of the Src family, which could cause unintentional off-target effects. This review emphasizes signaling pathways mediated by Fyn that govern neuronal development and plasticity while also summarizing the most noteworthy recent research relevant to Fyn kinase's function in the brain. Additionally, the therapeutic inhibition of Fyn kinase has been discussed, with a focus on the Fyn kinase inhibitors that are in clinical trials, which presents a fascinating opportunity for targeting Fyn kinase in the creation of possible therapeutic approaches for the management of Alzheimer's disease.
Collapse
Affiliation(s)
- Shreyasi Meur
- Department of Pharmaceutical Technology, School of Pharmacy, Techno India University, Kolkata, 700091, West Bengal, India
| | - Dipanjan Karati
- Department of Pharmaceutical Technology, School of Pharmacy, Techno India University, Kolkata, 700091, West Bengal, India.
| |
Collapse
|
2
|
Deep SN, Seelig S, Paul S, Poddar R. Homocysteine-induced sustained GluN2A NMDA receptor stimulation leads to mitochondrial ROS generation and neurotoxicity. J Biol Chem 2024; 300:107253. [PMID: 38569938 PMCID: PMC11081806 DOI: 10.1016/j.jbc.2024.107253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 03/11/2024] [Accepted: 03/24/2024] [Indexed: 04/05/2024] Open
Abstract
Homocysteine, a sulfur-containing amino acid derived from methionine metabolism, is a known agonist of N-methyl-D-aspartate receptor (NMDAR) and is involved in neurotoxicity. Our previous findings showed that neuronal exposure to elevated homocysteine levels leads to sustained low-level increase in intracellular Ca2+, which is dependent on GluN2A subunit-containing NMDAR (GluN2A-NMDAR) stimulation. These studies further showed a role of ERK MAPK in homocysteine-GluN2A-NMDAR-mediated neuronal death. However, the intracellular mechanisms associated with such sustained GluN2A-NMDAR stimulation and subsequent Ca2+ influx have remained unexplored. Using live-cell imaging with Fluo3-AM and biochemical approaches, we show that homocysteine-GluN2A NMDAR-induced initial Ca2+ influx triggers sequential phosphorylation and subsequent activation of the proline rich tyrosine kinase 2 (Pyk2) and Src family kinases, which in turn phosphorylates GluN2A-Tyr1325 residue of GluN2A-NMDARs to maintain channel activity. The continuity of this cycle of events leads to sustained Ca2+ influx through GluN2A-NMDAR. Our findings also show that lack of activation of the regulatory tyrosine phosphatase STEP, which can limit Pyk2 and Src family kinase activity further contributes to the maintenance of this cycle. Additional studies using live-cell imaging of neurons expressing a redox-sensitive GFP targeted to the mitochondrial matrix show that treatment with homocysteine leads to a progressive increase in mitochondrial reactive oxygen species generation, which is dependent on GluN2A-NMDAR-mediated sustained ERK MAPK activation. This later finding demonstrates a novel role of GluN2A-NMDAR in homocysteine-induced mitochondrial ROS generation and highlights the role of ERK MAPK as the intermediary signaling pathway between GluN2A-NMDAR stimulation and mitochondrial reactive oxygen species generation.
Collapse
Affiliation(s)
- Satya Narayan Deep
- Department of Neurology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA
| | - Sarah Seelig
- Department of Neurology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA
| | - Surojit Paul
- Department of Neurology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA
| | - Ranjana Poddar
- Department of Neurology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA.
| |
Collapse
|
3
|
Rodríguez-Durán LF, López-Ibarra DL, Herrera-Xithe G, Bermúdez-Rattoni F, Osorio-Gómez D, Escobar ML. Synergistic photoactivation of VTA-catecholaminergic and BLA-glutamatergic projections induces long-term potentiation in the insular cortex. Neurobiol Learn Mem 2023; 205:107845. [PMID: 37865264 DOI: 10.1016/j.nlm.2023.107845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 10/13/2023] [Accepted: 10/18/2023] [Indexed: 10/23/2023]
Abstract
The presentation of novel stimuli induces a reliable dopamine release in the insular cortex (IC) from the ventral tegmental area (VTA). The novel stimuli could be associated with motivational and emotional signals induced by cortical glutamate release from the basolateral amygdala (BLA). Dopamine and glutamate are essential for acquiring and maintaining behavioral tasks, including visual and taste recognition memories. In this study, we hypothesize that the simultaneous activation of dopaminergic and glutamatergic projections to the neocortex can underlie synaptic plasticity. High-frequency stimulation of the BLA-IC circuit has demonstrated a reliable long-term potentiation (LTP), a widely acknowledged synaptic plasticity that underlies memory consolidation. Therefore, the concurrent optogenetic stimulation of the insula's glutamatergic and dopaminergic terminal fibers would induce reliable LTP. Our results confirmed that combined photostimulation of the VTA and BLA projections to the IC induces a slow-onset LTP. We also found that optogenetically-induced LTP in the IC relies on both glutamatergic NMDA receptors and dopaminergic D1/D5 receptors, suggesting that the combined effects of these neurotransmitters can trigger synaptic plasticity in the neocortex. Overall, our findings provide compelling evidence supporting the essential role of both dopaminergic and glutamatergic projections in modulating synaptic plasticity within the IC. Furthermore, our results suggest that the synergistic actions of these projections have a pivotal influence on the formation of motivational memories.
Collapse
Affiliation(s)
- Luis F Rodríguez-Durán
- Instituto de Fisiología Celular, UNAM, División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Circuito Exterior, Ciudad Universitaria, 04510 Mexico City, Mexico
| | - Diana L López-Ibarra
- Instituto de Fisiología Celular, UNAM, División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Circuito Exterior, Ciudad Universitaria, 04510 Mexico City, Mexico
| | - Gabriela Herrera-Xithe
- Instituto de Fisiología Celular, UNAM, División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Circuito Exterior, Ciudad Universitaria, 04510 Mexico City, Mexico
| | - Federico Bermúdez-Rattoni
- Instituto de Fisiología Celular, UNAM, División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Circuito Exterior, Ciudad Universitaria, 04510 Mexico City, Mexico
| | - Daniel Osorio-Gómez
- Instituto de Fisiología Celular, UNAM, División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Circuito Exterior, Ciudad Universitaria, 04510 Mexico City, Mexico.
| | - Martha L Escobar
- Facultad de Psicología, UNAM, División de Investigación y Estudios de Posgrado, Universidad Nacional Autónoma de México, Ciudad Universitaria, 04510 Mexico City, Mexico.
| |
Collapse
|
4
|
Efficacy of Serotonin and Dopamine Activity Modulators in the Treatment of Negative Symptoms in Schizophrenia: A Rapid Review. Biomedicines 2023; 11:biomedicines11030921. [PMID: 36979900 PMCID: PMC10046337 DOI: 10.3390/biomedicines11030921] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 03/10/2023] [Accepted: 03/13/2023] [Indexed: 03/19/2023] Open
Abstract
Schizophrenia is among the fifteen most disabling diseases worldwide. Negative symptoms (NS) are highly prevalent in schizophrenia, negatively affect the functional outcome of the disorder, and their treatment is difficult and rarely specifically investigated. Serotonin-dopamine activity modulators (SDAMs), of which aripiprazole, cariprazine, brexpiprazole, and lumateperone were approved for schizophrenia treatment, represent a possible therapy to reduce NS. The aim of this rapid review is to summarize the evidence on this topic to make it readily available for psychiatrists treating NS and for further research. We searched the PubMed database for original studies using SDAM, aripiprazole, cariprazine, brexpiprazole, lumateperone, schizophrenia, and NS as keywords. We included four mega-analyses, eight meta-analyses, two post hoc analyses, and 20 clinical trials. Aripiprazole, cariprazine, and brexpiprazole were more effective than placebo in reducing NS. Only six studies compared SDAMs with other classes of antipsychotics, demonstrating a superiority in the treatment of NS mainly for cariprazine. The lack of specific research and various methodological issues, related to the study population and the assessment of NS, may have led to these partial results. Here, we highlight the need to conduct new methodologically robust investigations with head-to-head treatment comparisons and long-term observational studies on homogeneous groups of patients evaluating persistent NS with first- and second-generation scales, namely the Brief Negative Symptom Scale and the Clinical Assessment Interview for Negative Symptoms. This rapid review can expand research on NS therapeutic strategies in schizophrenia, which is fundamental for the long-term improvement of patients’ quality of life.
Collapse
|
5
|
The paradigm of amyloid precursor protein in amyotrophic lateral sclerosis: The potential role of the 682YENPTY 687 motif. Comput Struct Biotechnol J 2023; 21:923-930. [PMID: 36698966 PMCID: PMC9860402 DOI: 10.1016/j.csbj.2023.01.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 01/07/2023] [Accepted: 01/07/2023] [Indexed: 01/12/2023] Open
Abstract
Neurodegenerative diseases are characterized by the progressive decline of neuronal function in several brain areas, and are always associated with cognitive, psychiatric, or motor deficits due to the atrophy of certain neuronal populations. Most neurodegenerative diseases share common pathological mechanisms, such as neurotoxic protein misfolding, oxidative stress, and impairment of autophagy machinery. Amyotrophic lateral sclerosis (ALS) is one of the most common adult-onset motor neuron disorders worldwide. It is clinically characterized by the selective and progressive loss of motor neurons in the motor cortex, brain stem, and spinal cord, ultimately leading to muscle atrophy and rapidly progressive paralysis. Multiple recent studies have indicated that the amyloid precursor protein (APP) and its proteolytic fragments are not only drivers of Alzheimer's disease (AD) but also one of the earliest signatures in ALS, preceding or anticipating neuromuscular junction instability and denervation. Indeed, altered levels of APP peptides have been found in the brain, muscles, skin, and cerebrospinal fluid of ALS patients. In this short review, we discuss the nature and extent of research evidence on the role of APP peptides in ALS, focusing on the intracellular C-terminal peptide and its regulatory motif 682YENPTY687, with the overall aim of providing new frameworks and perspectives for intervention and identifying key questions for future investigations.
Collapse
|
6
|
Titulaer J, Radhe O, Danielsson K, Dutheil S, Marcus MM, Jardemark K, Svensson TH, Snyder GL, Ericson M, Davis RE, Konradsson-Geuken Å. Lumateperone-mediated effects on prefrontal glutamatergic receptor-mediated neurotransmission: A dopamine D 1 receptor dependent mechanism. Eur Neuropsychopharmacol 2022; 62:22-35. [PMID: 35878581 DOI: 10.1016/j.euroneuro.2022.06.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 06/27/2022] [Accepted: 06/29/2022] [Indexed: 11/25/2022]
Abstract
Lumateperone is a novel drug approved for the treatment of schizophrenia in adults and depressive episodes associated with bipolar depression in adults, as monotherapy and as adjunctive therapy with lithium or valproate treatment in the United States. Lumateperone simultaneously modulates key neurotransmitters, such as serotonin, dopamine, and glutamate, implicated in serious mental illness. In patients with schizophrenia, lumateperone was shown to improve positive symptoms along with negative and depressive symptoms, while also enhancing prosocial behavior. Moreover, in patients with bipolar I or II disorder, lumateperone improved depressive symptoms as well. To further understand the mechanisms related to lumateperone's clinical response, the aim of this study was to investigate the effect of lumateperone on dopaminergic- and glutamatergic signaling in the rat medial prefrontal cortex (mPFC). We used the conditioned avoidance response (CAR) test to determine the antipsychotic-like effect of lumateperone, electrophysiology in vitro to study lumateperone's effects on NMDA- and AMPA-induced currents in the mPFC, and the neurochemical techniques microdialysis and amperometry to measure dopamine- and glutamate release in the rat mPFC. Our results demonstrate that lumateperone; i) significantly suppressed CAR in rats, indicating an antipsychotic-like effect, ii) facilitated NMDA and AMPA receptor-mediated currents in the mPFC, in a dopamine D1-dependent manner, and iii) significantly increased dopamine and glutamate release in the rat mPFC. To the extent that these findings can be translated to humans, the ability of lumateperone to activate these pathways may contribute to its demonstrated effectiveness in safely improving symptoms related to neuropsychiatric disorder including mood alterations.
Collapse
Affiliation(s)
- J Titulaer
- Section of Neuropharmacology and Addiction Research, Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden; Section of Neuropsychopharmacology, Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden.
| | - O Radhe
- Section of Neuropharmacology and Addiction Research, Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - K Danielsson
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden
| | - S Dutheil
- Intra-Cellular Therapies, Inc., New York, NY, United States
| | - M M Marcus
- Section of Neuropsychopharmacology, Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - K Jardemark
- Section of Neuropsychopharmacology, Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - T H Svensson
- Section of Neuropsychopharmacology, Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - G L Snyder
- Intra-Cellular Therapies, Inc., New York, NY, United States
| | - M Ericson
- Addiction Biology Unit, Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden
| | - R E Davis
- Intra-Cellular Therapies, Inc., New York, NY, United States
| | - Å Konradsson-Geuken
- Section of Neuropharmacology and Addiction Research, Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden; Section of Neuropsychopharmacology, Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
7
|
Differential Transcriptome Profiling Unveils Novel Deregulated Gene Signatures Involved in Pathogenesis of Alzheimer's Disease. Biomedicines 2022; 10:biomedicines10030611. [PMID: 35327413 PMCID: PMC8945049 DOI: 10.3390/biomedicines10030611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 02/13/2022] [Accepted: 02/28/2022] [Indexed: 02/01/2023] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disorder that is characterized by a progressive loss of cognitive functions at a higher level than normal aging. Although the apolipoprotein (APOE) gene is a major risk factor in developing AD, other genes have also been reported to be linked with complex phenotypes. Therefore, this genome-wide expression study explored differentially expressed genes as possible novel biomarkers involved in AD. The mRNA expression dataset, GSE28146, containing 15 sample data composed of 7 AD cases from the hippocampus region with age-matched control (n = 8, >80 years), was analyzed. Using “affy” R-package, mRNA expression was calculated, while pathway enrichment analysis was performed to determine related biological processes. Of 58 differentially expressed genes, 44 downregulated and 14 upregulated genes were found to be significantly (p < 0.001) altered. The pathway enrichment analysis revealed two altered genes, i.e., dynein light chain 1 (DYNLL1) and kalirin (KLRN), associated with AD in the elderly population. The majority of genes were associated with retrograde endocannabinoid as well as vascular endothelial growth factors affecting the complex phenotypes. The DYNLL1 and KLRN genes may be involved with AD and Huntington’s disease (HD) phenotypes and represent a common genetic basis of these diseases. However, the hallmark of AD is dementia, while the classic motor sign of HD includes chorea. Our data warrant further investigation to identify the role of these genes in disease pathogenesis.
Collapse
|
8
|
Jones-Tabah J, Mohammad H, Paulus EG, Clarke PBS, Hébert TE. The Signaling and Pharmacology of the Dopamine D1 Receptor. Front Cell Neurosci 2022; 15:806618. [PMID: 35110997 PMCID: PMC8801442 DOI: 10.3389/fncel.2021.806618] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 12/23/2021] [Indexed: 12/30/2022] Open
Abstract
The dopamine D1 receptor (D1R) is a Gαs/olf-coupled GPCR that is expressed in the midbrain and forebrain, regulating motor behavior, reward, motivational states, and cognitive processes. Although the D1R was initially identified as a promising drug target almost 40 years ago, the development of clinically useful ligands has until recently been hampered by a lack of suitable candidate molecules. The emergence of new non-catechol D1R agonists, biased agonists, and allosteric modulators has renewed clinical interest in drugs targeting this receptor, specifically for the treatment of motor impairment in Parkinson's Disease, and cognitive impairment in neuropsychiatric disorders. To develop better therapeutics, advances in ligand chemistry must be matched by an expanded understanding of D1R signaling across cell populations in the brain, and in disease states. Depending on the brain region, the D1R couples primarily to either Gαs or Gαolf through which it activates a cAMP/PKA-dependent signaling cascade that can regulate neuronal excitability, stimulate gene expression, and facilitate synaptic plasticity. However, like many GPCRs, the D1R can signal through multiple downstream pathways, and specific signaling signatures may differ between cell types or be altered in disease. To guide development of improved D1R ligands, it is important to understand how signaling unfolds in specific target cells, and how this signaling affects circuit function and behavior. In this review, we provide a summary of D1R-directed signaling in various neuronal populations and describe how specific pathways have been linked to physiological and behavioral outcomes. In addition, we address the current state of D1R drug development, including the pharmacology of newly developed non-catecholamine ligands, and discuss the potential utility of D1R-agonists in Parkinson's Disease and cognitive impairment.
Collapse
|
9
|
Oleson EB, Hamilton LR, Gomez DM. Cannabinoid Modulation of Dopamine Release During Motivation, Periodic Reinforcement, Exploratory Behavior, Habit Formation, and Attention. Front Synaptic Neurosci 2021; 13:660218. [PMID: 34177546 PMCID: PMC8222827 DOI: 10.3389/fnsyn.2021.660218] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 05/05/2021] [Indexed: 12/12/2022] Open
Abstract
Motivational and attentional processes energize action sequences to facilitate evolutionary competition and promote behavioral fitness. Decades of neuropharmacology, electrophysiology and electrochemistry research indicate that the mesocorticolimbic DA pathway modulates both motivation and attention. More recently, it was realized that mesocorticolimbic DA function is tightly regulated by the brain's endocannabinoid system and greatly influenced by exogenous cannabinoids-which have been harnessed by humanity for medicinal, ritualistic, and recreational uses for 12,000 years. Exogenous cannabinoids, like the primary psychoactive component of cannabis, delta-9-tetrahydrocannabinol, produce their effects by acting at binding sites for naturally occurring endocannabinoids. The brain's endocannabinoid system consists of two G-protein coupled receptors, endogenous lipid ligands for these receptor targets, and several synthetic and metabolic enzymes involved in their production and degradation. Emerging evidence indicates that the endocannabinoid 2-arachidonoylglycerol is necessary to observe concurrent increases in DA release and motivated behavior. And the historical pharmacology literature indicates a role for cannabinoid signaling in both motivational and attentional processes. While both types of behaviors have been scrutinized under manipulation by either DA or cannabinoid agents, there is considerably less insight into prospective interactions between these two important signaling systems. This review attempts to summate the relevance of cannabinoid modulation of DA release during operant tasks designed to investigate either motivational or attentional control of behavior. We first describe how cannabinoids influence DA release and goal-directed action under a variety of reinforcement contingencies. Then we consider the role that endocannabinoids might play in switching an animal's motivation from a goal-directed action to the search for an alternative outcome, in addition to the formation of long-term habits. Finally, dissociable features of attentional behavior using both the 5-choice serial reaction time task and the attentional set-shifting task are discussed along with their distinct influences by DA and cannabinoids. We end with discussing potential targets for further research regarding DA-cannabinoid interactions within key substrates involved in motivation and attention.
Collapse
Affiliation(s)
- Erik B. Oleson
- Department of Psychology, University of Colorado Denver, Denver, CO, United States
| | - Lindsey R. Hamilton
- Department of Psychology, University of Colorado Denver, Denver, CO, United States
| | - Devan M. Gomez
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI, United States
| |
Collapse
|
10
|
Li Y, Bao Y, Zheng H, Qin Y, Hua B. Can Src protein tyrosine kinase inhibitors be combined with opioid analgesics? Src and opioid-induced tolerance, hyperalgesia and addiction. Biomed Pharmacother 2021; 139:111653. [PMID: 34243625 DOI: 10.1016/j.biopha.2021.111653] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/17/2021] [Accepted: 04/21/2021] [Indexed: 10/21/2022] Open
Abstract
The clinical application of opioids may be accompanied by a series of adverse consequences, such as opioid tolerance, opioid-induced hyperalgesia, opioid dependence or addiction. In view of this issue, clinicians are faced with the dilemma of treating various types of pain with or without opioids. In this review, we discuss that Src protein tyrosine kinase plays an important role in these adverse consequences, and Src inhibitors can solve these problems well. Therefore, Src inhibitors have the potential to be used in combination with opioids to achieve synergy. How to combine them together to maximize the analgesic effect while avoiding unnecessary trouble provides a topic for follow-up research.
Collapse
Affiliation(s)
- Yaoyuan Li
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yanju Bao
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Honggang Zheng
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yinggang Qin
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Baojin Hua
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
11
|
Martel JC, Gatti McArthur S. Dopamine Receptor Subtypes, Physiology and Pharmacology: New Ligands and Concepts in Schizophrenia. Front Pharmacol 2020; 11:1003. [PMID: 32765257 PMCID: PMC7379027 DOI: 10.3389/fphar.2020.01003] [Citation(s) in RCA: 143] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 06/22/2020] [Indexed: 12/14/2022] Open
Abstract
Dopamine receptors are widely distributed within the brain where they play critical modulator roles on motor functions, motivation and drive, as well as cognition. The identification of five genes coding for different dopamine receptor subtypes, pharmacologically grouped as D1- (D1 and D5) or D2-like (D2S, D2L, D3, and D4) has allowed the demonstration of differential receptor function in specific neurocircuits. Recent observation on dopamine receptor signaling point at dopamine-glutamate-NMDA neurobiology as the most relevant in schizophrenia and for the development of new therapies. Progress in the chemistry of D1- and D2-like receptor ligands (agonists, antagonists, and partial agonists) has provided more selective compounds possibly able to target the dopamine receptors homo and heterodimers and address different schizophrenia symptoms. Moreover, an extensive evaluation of the functional effect of these agents on dopamine receptor coupling and intracellular signaling highlights important differences that could also result in highly differentiated clinical pharmacology. The review summarizes the recent advances in the field, addressing the relevance of emerging new targets in schizophrenia in particular in relation to the dopamine - glutamate NMDA systems interactions.
Collapse
|
12
|
Carceles‐Cordon M, Mannara F, Aguilar E, Castellanos A, Planagumà J, Dalmau J. NMDAR
Antibodies Alter Dopamine Receptors and Cause Psychotic Behavior in Mice. Ann Neurol 2020; 88:603-613. [DOI: 10.1002/ana.25829] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 06/17/2020] [Accepted: 06/21/2020] [Indexed: 01/11/2023]
Affiliation(s)
- Marc Carceles‐Cordon
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Universitat de Barcelona Barcelona Spain
| | - Francesco Mannara
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Universitat de Barcelona Barcelona Spain
| | - Esther Aguilar
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Universitat de Barcelona Barcelona Spain
| | - Aida Castellanos
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Universitat de Barcelona Barcelona Spain
| | - Jesús Planagumà
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Universitat de Barcelona Barcelona Spain
| | - Josep Dalmau
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Universitat de Barcelona Barcelona Spain
- Department of NeurologyUniversity of Pennsylvania Philadelphia PA USA
- Institució Catalana de Recerca i Estudis Avançats (ICREA) Barcelona Spain
| |
Collapse
|
13
|
Tevzadze G, Zhuravliova E, Barbakadze T, Shanshiashvili L, Dzneladze D, Nanobashvili Z, Lordkipanidze T, Mikeladze D. Gut neurotoxin p-cresol induces differential expression of GLUN2B and GLUN2A subunits of the NMDA receptor in the hippocampus and nucleus accumbens in healthy and audiogenic seizure-prone rats. AIMS Neurosci 2020; 7:30-42. [PMID: 32455164 PMCID: PMC7242059 DOI: 10.3934/neuroscience.2020003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 02/17/2020] [Indexed: 01/18/2023] Open
Abstract
Mislocalization and abnormal expression of N-methyl-D-aspartate glutamate receptor (NMDAR) subunits is observed in several brain disorders and pathological conditions. Recently, we have shown that intraperitoneal injection of the gut neurotoxin p-cresol induces autism-like behavior and accelerates seizure reactions in healthy and epilepsy-prone rats, respectively. In this study, we evaluated the expression of GLUN2B and GLUN2A NMDAR subunits, and assessed the activity of cAMP-response element binding protein (CREB) and Rac1 in the hippocampi and nucleus accumbens of healthy and epilepsy-prone rats following p-cresol administration. We have found that subchronic intraperitoneal injection of p-cresol induced differential expression of GLUN2B and GLUN2A between the two brain regions, and altered the GLUN2B/GLUN2A ratio, in rats in both groups. Moreover, p-cresol impaired CREB phosphorylation in both brain structures and stimulated Rac activity in the hippocampus. These data indicate that p-cresol differently modulates the expression of NMDAR subunits in the nucleus accumbens and hippocampi of healthy and epilepsy-prone rats. We propose that these differences are due to the specificity of interactions between dopaminergic and glutamatergic pathways in these structures.
Collapse
Affiliation(s)
- Gigi Tevzadze
- 4-D Research Institute, Ilia State University, 3/5 Cholokashvili av, Tbilisi, 0162, Georgia
| | - Elene Zhuravliova
- Institute of Chemical Biology, Ilia State University, 3/5 Cholokashvili av, Tbilisi, 0162, Georgia.,I. Beritashvili Center of Experimental Biomedicine 14, Gotua Str., Tbilisi 0160, Georgia
| | - Tamar Barbakadze
- Institute of Chemical Biology, Ilia State University, 3/5 Cholokashvili av, Tbilisi, 0162, Georgia.,I. Beritashvili Center of Experimental Biomedicine 14, Gotua Str., Tbilisi 0160, Georgia
| | - Lali Shanshiashvili
- Institute of Chemical Biology, Ilia State University, 3/5 Cholokashvili av, Tbilisi, 0162, Georgia.,I. Beritashvili Center of Experimental Biomedicine 14, Gotua Str., Tbilisi 0160, Georgia
| | - Davit Dzneladze
- I. Beritashvili Center of Experimental Biomedicine 14, Gotua Str., Tbilisi 0160, Georgia
| | - Zaqaria Nanobashvili
- I. Beritashvili Center of Experimental Biomedicine 14, Gotua Str., Tbilisi 0160, Georgia
| | - Tamar Lordkipanidze
- Institute of Chemical Biology, Ilia State University, 3/5 Cholokashvili av, Tbilisi, 0162, Georgia.,I. Beritashvili Center of Experimental Biomedicine 14, Gotua Str., Tbilisi 0160, Georgia
| | - David Mikeladze
- Institute of Chemical Biology, Ilia State University, 3/5 Cholokashvili av, Tbilisi, 0162, Georgia.,I. Beritashvili Center of Experimental Biomedicine 14, Gotua Str., Tbilisi 0160, Georgia
| |
Collapse
|
14
|
Mao Z, Wang W, Gong H, Wu Y, Zhang Y, Wang X. Upregulation of miR-496 Rescues Propofol-induced Neurotoxicity by Targeting Rho Associated Coiled-coil Containing Protein Kinase 2 (ROCK2) in Prefrontal Cortical Neurons. Curr Neurovasc Res 2020; 17:188-195. [PMID: 32370715 DOI: 10.2174/1567202617666200506101926] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 01/14/2020] [Accepted: 01/17/2020] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Early exposure to general anesthesia in children might be a potentially highrisk factor for learning and behavioral disorders. The mechanism of neurotoxicity induced by general anesthesia was not defined. miR-496 could regulate cerebral injury, while the roles of miR- 496 in neurotoxicity were not elucidated. Therefore, we aimed to investigate the effects of miR- 496 in neurotoxicity induced by propofol. METHODS Primary Prefrontal Cortical (PFC) neurons were isolated from neonatal rats and treated with propofol to induce neurotoxicity. Cell viability was detected by (3-(4,5-Dimethylthiazol- 2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, and cell apoptosis was assessed by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay. The target relationship of miR-496 and Rho Associated Coiled-Coil Containing Protein Kinase 2 (ROCK2) was explored using luciferase assays. RESULTS Propofol decreased cell viability, promoted cell apoptosis, and decreased the expression of miR-496 in PFC neurons in a dose-dependent manner. Overexpression of miR-496 attenuated neurotoxicity induced by propofol in PFC neurons. ROCK2 was a target of miR-496, and miR-496 oppositely modulated the expression of ROCK2. Besides, propofol increased the expression of ROCK2 through inhibiting miR-496 in PFC neurons. Overexpression of miR-496 attenuated propofol- induced neurotoxicity by targeting ROCK2 in PFC neurons. CONCLUSION miR-496 was decreased in PFC neurons treated with propofol, and overexpression of miR-496 attenuated propofol-induced neurotoxicity by targeting ROCK2. miR-496 and ROCK2 may be promising targets for protecting propofol-induced neurotoxicity.
Collapse
Affiliation(s)
- Zemei Mao
- Department of Anesthesiology, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science & Technology, Wuhan City, Hubei Province, 430016, China
| | - Wanju Wang
- Department of General Surgery, Hubei Provincial Hospital of Integrated Chinese & Western Medicine, Wuhan City, Hubei Province, 430015, China
| | - Haixia Gong
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang City, Jiangxi Province, 330006, China
| | - Yinghui Wu
- Department of Anesthesiology, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science & Technology, Wuhan City, Hubei Province, 430016, China
| | - Yang Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang City, Jiangxi Province, 330006, China
| | - Xinlei Wang
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang City, Jiangxi Province, 330006, China
| |
Collapse
|
15
|
Mitlöhner J, Kaushik R, Niekisch H, Blondiaux A, Gee CE, Happel MFK, Gundelfinger E, Dityatev A, Frischknecht R, Seidenbecher C. Dopamine Receptor Activation Modulates the Integrity of the Perisynaptic Extracellular Matrix at Excitatory Synapses. Cells 2020; 9:cells9020260. [PMID: 31972963 PMCID: PMC7073179 DOI: 10.3390/cells9020260] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 01/14/2020] [Accepted: 01/17/2020] [Indexed: 01/08/2023] Open
Abstract
In the brain, Hebbian-type and homeostatic forms of plasticity are affected by neuromodulators like dopamine (DA). Modifications of the perisynaptic extracellular matrix (ECM), which control the functions and mobility of synaptic receptors as well as the diffusion of transmitters and neuromodulators in the extracellular space, are crucial for the manifestation of plasticity. Mechanistic links between synaptic activation and ECM modifications are largely unknown. Here, we report that neuromodulation via D1-type DA receptors can induce targeted ECM proteolysis specifically at excitatory synapses of rat cortical neurons via proteases ADAMTS-4 and -5. We showed that receptor activation induces increased proteolysis of brevican (BC) and aggrecan, two major constituents of the adult ECM both in vivo and in vitro. ADAMTS immunoreactivity was detected near synapses, and shRNA-mediated knockdown reduced BC cleavage. We have outlined a molecular scenario of how synaptic activity and neuromodulation are linked to ECM rearrangements via increased cAMP levels, NMDA receptor activation, and intracellular calcium signaling.
Collapse
Affiliation(s)
- Jessica Mitlöhner
- Leibniz Institute for Neurobiology (LIN), Department of Neurochemistry and Molecular Biology, 39118 Magdeburg, Germany; (J.M.); (A.B.); (E.G.)
| | - Rahul Kaushik
- German Center for Neurodegenerative Diseases (DZNE), Molecular Neuroplasticity Group, 39120 Magdeburg, Germany;
- Center for Behavioral Brain Sciences (CBBS), 39120 Magdeburg, Germany
| | - Hartmut Niekisch
- Leibniz Institute for Neurobiology (LIN), Department of Systems Physiology of Learning, 39118 Magdeburg, Germany; (H.N.); (M.F.K.H.)
| | - Armand Blondiaux
- Leibniz Institute for Neurobiology (LIN), Department of Neurochemistry and Molecular Biology, 39118 Magdeburg, Germany; (J.M.); (A.B.); (E.G.)
| | - Christine E. Gee
- Center for Molecular Neurobiology Hamburg (ZMNH), Institute for Synaptic Physiology, 20251 Hamburg, Germany;
| | - Max F. K. Happel
- Leibniz Institute for Neurobiology (LIN), Department of Systems Physiology of Learning, 39118 Magdeburg, Germany; (H.N.); (M.F.K.H.)
| | - Eckart Gundelfinger
- Leibniz Institute for Neurobiology (LIN), Department of Neurochemistry and Molecular Biology, 39118 Magdeburg, Germany; (J.M.); (A.B.); (E.G.)
- Center for Behavioral Brain Sciences (CBBS), 39120 Magdeburg, Germany
- Otto-von-Guericke University, Medical Faculty, 39120 Magdeburg, Germany
| | - Alexander Dityatev
- German Center for Neurodegenerative Diseases (DZNE), Molecular Neuroplasticity Group, 39120 Magdeburg, Germany;
- Center for Behavioral Brain Sciences (CBBS), 39120 Magdeburg, Germany
- Otto-von-Guericke University, Medical Faculty, 39120 Magdeburg, Germany
- Correspondence: (A.D.); (R.F.); (C.S.); Tel.: +49-391 67-24526 (A.D.); +49-9131 85-28051 (R.F.); +49-391-6263-92401 (C.S.)
| | - Renato Frischknecht
- Leibniz Institute for Neurobiology (LIN), Department of Neurochemistry and Molecular Biology, 39118 Magdeburg, Germany; (J.M.); (A.B.); (E.G.)
- Center for Behavioral Brain Sciences (CBBS), 39120 Magdeburg, Germany
- Correspondence: (A.D.); (R.F.); (C.S.); Tel.: +49-391 67-24526 (A.D.); +49-9131 85-28051 (R.F.); +49-391-6263-92401 (C.S.)
| | - Constanze Seidenbecher
- Leibniz Institute for Neurobiology (LIN), Department of Neurochemistry and Molecular Biology, 39118 Magdeburg, Germany; (J.M.); (A.B.); (E.G.)
- Center for Behavioral Brain Sciences (CBBS), 39120 Magdeburg, Germany
- Otto-von-Guericke University, Medical Faculty, 39120 Magdeburg, Germany
- Correspondence: (A.D.); (R.F.); (C.S.); Tel.: +49-391 67-24526 (A.D.); +49-9131 85-28051 (R.F.); +49-391-6263-92401 (C.S.)
| |
Collapse
|
16
|
Nedunchezhian D, Langeswaran K, Santhoshkumar S. Identification of novel inhibitor targeting Fyn kinase using molecular docking analysis. Bioinformation 2019; 15:419-424. [PMID: 31312079 PMCID: PMC6614121 DOI: 10.6026/97320630015419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 06/03/2019] [Indexed: 12/02/2022] Open
Abstract
Identification of tyrosine Fyn kinase inhibitor is recognized as an effective and feasible therapeutic measure in reducing consequences of memory loss disorder Alzheimer's. The present investigation has been attempted with an objective to find out a novel potent inhibitor with similar homological structure to Fyn kinase using structure based in silico screening measure. Such derived structure was compared with natural data base pool and were systematically analyzed. Ligand based interaction was also tested and evaluated. We applied a molecular dynamic simulation technique to validate the stability of the identified complexes and to understand the ligand binding mechanism. Results provide information on the characteristics of novel and potent inhibitor for tyrosinase Fyn kinase protein so as to develop an innovative strategy to treat AD.
Collapse
Affiliation(s)
- Dharani Nedunchezhian
- Department of Biotechnology and Bioinformatics,Bishop Heber College,Bharathidasan University,Tiruchirapalli,Tamil Nadu,India
| | - Kulanthaivel Langeswaran
- Cancer Genetics and Molecular Biology Laboratory, Department of Bioinformatics, Science Campus, Alagappa University, Karaikudi, Tamil Nadu, India
| | - Sundar Santhoshkumar
- Department of Computer Science, Science Campus, Alagappa University, Karaikudi, Tamil Nadu, India
| |
Collapse
|
17
|
Abstract
Cerebral organoids are an emerging cutting-edge technology to model human brain
development and neurodevelopmental disorders, for which mouse models exhibit significant
limitations. In the human brain, synaptic connections define neural circuits, and synaptic
deficits account for various neurodevelopmental disorders. Thus, harnessing the full power
of cerebral organoids for human brain modeling requires the ability to visualize and
analyze synapses in cerebral organoids. Previously, we devised an optimized method to
generate human cerebral organoids, and showed that optimal organoids express mature-neuron
markers, including synaptic proteins and neurotransmitter receptors and transporters.
Here, we give evidence for synaptogenesis in cerebral organoids, via microscopical
visualization of synapses. We also describe multiple approaches to quantitatively analyze
synapses in cerebral organoids. Collectively, our work provides sufficient evidence for
the possibility of modeling synaptogenesis and synaptic disorders in cerebral organoids,
and may help advance the use of cerebral organoids in molecular neuroscience and studies
of neurodevelopmental disorders such as autism.
Collapse
Affiliation(s)
- Abraam M Yakoub
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Mark Sadek
- Department of Pharmaceutical Biotechnology, University of Illinois College of Pharmacy, Chicago, IL, USA.,Department of Research and Development, Akorn Pharmaceuticals, Vernon Hills, IL, USA
| |
Collapse
|
18
|
Jin DZ, Mao LM, Wang JQ. Amphetamine activates non-receptor tyrosine kinase Fyn and stimulates ERK phosphorylation in the rat striatum in vivo. Eur J Pharmacol 2018; 843:45-54. [PMID: 30419241 DOI: 10.1016/j.ejphar.2018.11.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 11/02/2018] [Accepted: 11/06/2018] [Indexed: 12/16/2022]
Abstract
The psychostimulant amphetamine (AMPH) has an impact on a variety of cellular activities in striatal neurons, although underlying signaling mechanisms are incompletely understood. The Src family kinase (SFK) is among key signaling molecules enriched in striatal neurons and is involved in the regulation of a set of discrete downstream targets. Given the likelihood that AMPH may regulate SFKs, we investigated and characterized the effect of AMPH on SFK phosphorylation and enzymatic activity in rat striatal neurons in vivo. We found that AMPH elevated SFK Y416 phosphorylation in striatal slices and the adult rat striatum. This elevation was concentration- and time-dependent and occurred in all subdivisions of the striatum, including the caudate putamen and nucleus accumbens (core and shell). The dopamine D1 receptor antagonist SCH23390 blocked the effect of AMPH. Between Fyn and Src, AMPH elevated phosphorylation of immunoprecipitated Fyn but not Src and increased Fyn kinase activity in the striatum. In parallel with SFKs, striatal ERK phosphorylation was increased by AMPH. This increase in ERK phosphorylation was reduced by the SFK inhibitor PP2. These results demonstrate that AMPH is able to activate SFKs (mainly Fyn) in striatal neurons via a D1 receptor-dependent mechanism. Activated SFKs participate in processing the concomitant ERK response to AMPH.
Collapse
Affiliation(s)
- Dao-Zhong Jin
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City, Kansas City, MO 64108, USA.
| | - Li-Min Mao
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City, Kansas City, MO 64108, USA
| | - John Q Wang
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City, Kansas City, MO 64108, USA; Department of Anesthesiology, School of Medicine, University of Missouri-Kansas City, Kansas City, MO 64108, USA.
| |
Collapse
|
19
|
Sun Y, Zhan L, Cheng X, Zhang L, Hu J, Gao Z. The Regulation of GluN2A by Endogenous and Exogenous Regulators in the Central Nervous System. Cell Mol Neurobiol 2017; 37:389-403. [PMID: 27255970 PMCID: PMC11482088 DOI: 10.1007/s10571-016-0388-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Accepted: 05/25/2016] [Indexed: 12/25/2022]
Abstract
The NMDA receptor is the most widely studied ionotropic glutamate receptor, and it is central to many physiological and pathophysiological processes in the central nervous system. GluN2A is one of the two main types of GluN2 NMDA receptor subunits in the forebrain. The proper activity of GluN2A is important to brain function, as the abnormal regulation of GluN2A may induce some neuropsychiatric disorders. This review will examine the regulation of GluN2A by endogenous and exogenous regulators in the central nervous system.
Collapse
Affiliation(s)
- Yongjun Sun
- Department of Pharmacy, Hebei University of Science and Technology, Yuhua East Road 70, Shijiazhuang, 050018, People's Republic of China
- Hebei Research Center of Pharmaceutical and Chemical Engineering, Hebei University of Science and Technology, Shijiazhuang, 050018, People's Republic of China
| | - Liying Zhan
- Department of Pharmacy, Hebei University of Science and Technology, Yuhua East Road 70, Shijiazhuang, 050018, People's Republic of China
| | - Xiaokun Cheng
- North China Pharmaceutical Group New Drug Research and Development Co., Ltd, Shijiazhuang, 050015, People's Republic of China
| | - Linan Zhang
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, 050017, People's Republic of China
| | - Jie Hu
- School of Nursing, Hebei Medical University, Shijiazhuang, 050017, People's Republic of China
| | - Zibin Gao
- Department of Pharmacy, Hebei University of Science and Technology, Yuhua East Road 70, Shijiazhuang, 050018, People's Republic of China.
- Hebei Research Center of Pharmaceutical and Chemical Engineering, Hebei University of Science and Technology, Shijiazhuang, 050018, People's Republic of China.
- State Key Laboratory Breeding Base-Hebei Province Key Laboratory of Molecular Chemistry for Drug, Shijiazhuang, 050018, People's Republic of China.
| |
Collapse
|
20
|
Mao LM, Wang JQ. Antagonism of Dopamine D2 Receptors Alters Phosphorylation of Fyn in the Rat Medial Prefrontal Cortex. J Mol Neurosci 2017; 61:524-530. [PMID: 28176147 DOI: 10.1007/s12031-017-0894-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 01/27/2017] [Indexed: 02/06/2023]
Abstract
Several Src family kinase (SFK) members are expressed in the mammalian brain and serve as key kinases in the regulation of a variety of cellular and synaptic events. These SFKs may be subject to the modulation by dopamine, although this topic has been investigated incompletely. In this study, we explored whether dopamine D2 receptors (D2Rs) regulate SFKs in adult rat brains in vivo. We investigated the role of D2Rs in two forebrain areas, the medial prefrontal cortex (mPFC) and hippocampus, since dopamine plays a pivotal role in regulating activity of mPFC and hippocampal neurons and D2Rs are expressed in these regions. We found that a systemic injection of a D2R selective antagonist eticlopride elevated phosphorylation of SFKs at a conserved autophosphorylation site, an event correlated with activation of SFKs, in the mPFC. Similarly, antagonism of D2Rs by haloperidol increased SFK phosphorylation. In contrast, eticlopride and haloperidol did not alter SFK phosphorylation in the hippocampus. The effect of eticlopride was time-dependent and relatively delayed. Among two common SFK members enriched at synaptic sites, eticlopride selectively altered phosphorylation of Fyn but not Src. Our data suggest that D2Rs exert an inhibitory effect on the activity-related phosphorylation of Fyn in the mPFC under normal conditions.
Collapse
Affiliation(s)
- Li-Min Mao
- Department of Basic Medical Science, School of Medicine, University of Missouri-Kansas City, 2411 Holmes Street, Kansas City, MO, 64108, USA
| | - John Q Wang
- Department of Basic Medical Science, School of Medicine, University of Missouri-Kansas City, 2411 Holmes Street, Kansas City, MO, 64108, USA. .,Department of Anesthesiology, School of Medicine, University of Missouri-Kansas City, Kansas City, MO, 64108, USA.
| |
Collapse
|
21
|
de Bartolomeis A, Marmo F, Buonaguro EF, Latte G, Tomasetti C, Iasevoli F. Switching antipsychotics: Imaging the differential effect on the topography of postsynaptic density transcripts in antipsychotic-naïve vs. antipsychotic-exposed rats. Prog Neuropsychopharmacol Biol Psychiatry 2016; 70:24-38. [PMID: 27177972 DOI: 10.1016/j.pnpbp.2016.04.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 04/17/2016] [Accepted: 04/27/2016] [Indexed: 10/24/2022]
Abstract
The postsynaptic density (PSD) has been regarded as a functional switchboard at the crossroads of a dopamine-glutamate interaction, and it is putatively involved in the pathophysiology of psychosis. Indeed, it has been demonstrated that antipsychotics may modulate several PSD transcripts, such as PSD-95, Shank, and Homer. Despite switching antipsychotics is a frequent strategy to counteract lack of efficacy and/or side effect onset in clinical practice, no information is available on the effects of sequential treatments with different antipsychotics on PSD molecules. The aim of this study was to evaluate whether a previous exposure to a typical antipsychotic and a switch to an atypical one may affect the expression of PSD transcripts, in order to evaluate potential neurobiological correlates of this common clinical practice, with specific regards to putative synaptic plasticity processes. We treated male Sprague-Dawley rats intraperitoneally for 15days with haloperidol or vehicle, then from the sixteenth day we switched the animals to amisulpride or continued to treat them with vehicle or haloperidol for 15 additional days. In this way we got six first treatment/second treatment groups: vehicle/vehicle, vehicle/haloperidol, vehicle/amisulpride, haloperidol/vehicle, haloperidol/haloperidol, haloperidol/amisulpride. In this paradigm, we evaluated the expression of brain transcripts belonging to relevant and interacting PSD proteins, both of the Immediate-Early Gene (Homer1a, Arc) and the constitutive classes (Homer1b/c and PSD-95). The major finding was the differential effect of amisulpride on gene transcripts when administered in naïve vs. antipsychotic-pretreated rats, with modifications of the ratio between Homer1a/Homer1b transcripts and differential effects in cortex and striatum. These results suggest that the neurobiological effects on PSD transcripts of amisulpride, and possibly of other antipsychotics, may be greatly affected by prior antipsychotic treatments and may impact significantly on the switching procedure.
Collapse
Affiliation(s)
- Andrea de Bartolomeis
- Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, School of Medicine, University "Federico II", Naples, Italy.
| | - Federica Marmo
- Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, School of Medicine, University "Federico II", Naples, Italy
| | - Elisabetta F Buonaguro
- Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, School of Medicine, University "Federico II", Naples, Italy
| | - Gianmarco Latte
- Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, School of Medicine, University "Federico II", Naples, Italy
| | - Carmine Tomasetti
- Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, School of Medicine, University "Federico II", Naples, Italy
| | - Felice Iasevoli
- Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, School of Medicine, University "Federico II", Naples, Italy
| |
Collapse
|
22
|
Jacobs S, Tsien JZ. Adult forebrain NMDA receptors gate social motivation and social memory. Neurobiol Learn Mem 2016; 138:164-172. [PMID: 27575297 DOI: 10.1016/j.nlm.2016.08.019] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 08/12/2016] [Accepted: 08/25/2016] [Indexed: 01/13/2023]
Abstract
Motivation to engage in social interaction is critical to ensure normal social behaviors, whereas dysregulation in social motivation can contribute to psychiatric diseases such as schizophrenia, autism, social anxiety disorders and post-traumatic stress disorder (PTSD). While dopamine is well known to regulate motivation, its downstream targets are poorly understood. Given the fact that the dopamine 1 (D1) receptors are often physically coupled with the NMDA receptors, we hypothesize that the NMDA receptor activity in the adult forebrain principal neurons are crucial not only for learning and memory, but also for the proper gating of social motivation. Here, we tested this hypothesis by examining sociability and social memory in inducible forebrain-specific NR1 knockout mice. These mice are ideal for exploring the role of the NR1 subunit in social behavior because the NR1 subunit can be selectively knocked out after the critical developmental period, in which NR1 is required for normal development. We found that the inducible deletion of the NMDA receptors prior to behavioral assays impaired, not only object and social recognition memory tests, but also resulted in profound deficits in social motivation. Mice with ablated NR1 subunits in the forebrain demonstrated significant decreases in sociability compared to their wild type counterparts. These results suggest that in addition to its crucial role in learning and memory, the NMDA receptors in the adult forebrain principal neurons gate social motivation, independent of neuronal development.
Collapse
Affiliation(s)
- Stephanie Jacobs
- Brain and Behavior Discovery Institute and Department of Neurology, Medical College of Georgia at Augusta University, Augusta, GA 30907, USA
| | - Joe Z Tsien
- Brain and Behavior Discovery Institute and Department of Neurology, Medical College of Georgia at Augusta University, Augusta, GA 30907, USA.
| |
Collapse
|
23
|
Zamzow DR, Elias V, Acosta VA, Escobedo E, Magnusson KR. Higher levels of phosphorylated Y1472 on GluN2B subunits in the frontal cortex of aged mice are associated with good spatial reference memory, but not cognitive flexibility. AGE (DORDRECHT, NETHERLANDS) 2016; 38:50. [PMID: 27094400 PMCID: PMC5005925 DOI: 10.1007/s11357-016-9913-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2015] [Accepted: 04/06/2016] [Indexed: 06/05/2023]
Abstract
The N-methyl-D-aspartate receptor (NMDAr) is particularly vulnerable to aging. The GluN2B subunit of the NMDAr, compared to other NMDAr subunits, suffers the greatest losses of expression in the aging brain, especially in the frontal cortex. While expression levels of GluN2B mRNA and protein in the aged brain are well documented, there has been little investigation into age-related posttranslational modifications of the subunit. In this study, we explored some of the mechanisms that may promote differences in the NMDAr complex in the frontal cortex of aged animals. Two ages of mice, 3 and 24 months, were behaviorally tested in the Morris water maze. The frontal cortex and hippocampus from each mouse were subjected to differential centrifugation followed by solubilization in Triton X-100. Proteins from Triton-insoluble membranes, Triton-soluble membranes, and intracellular membranes/cytosol were examined by Western blot. Higher levels of GluN2B tyrosine 1472 phosphorylation in frontal cortex synaptic fractions of old mice were associated with better reference learning but poorer cognitive flexibility. Levels of GluN2B phosphotyrosine 1336 remained steady, but there were greater levels of the calpain-induced 115 kDa GluN2B cleavage product on extrasynaptic membranes in these old good learners. There was an age-related increase in calpain activity, but it was not associated with better learning. These data highlight a unique aging change for aged mice with good spatial learning that might be detrimental to cognitive flexibility. This study also suggests that higher levels of truncated GluN2B on extrasynaptic membranes are not deleterious to spatial memory in aged mice.
Collapse
Affiliation(s)
| | - Val Elias
- Oregon State University, Corvallis, OR, USA
| | | | | | | |
Collapse
|
24
|
Moraga-Amaro R, González H, Ugalde V, Donoso-Ramos JP, Quintana-Donoso D, Lara M, Morales B, Rojas P, Pacheco R, Stehberg J. Dopamine receptor D5 deficiency results in a selective reduction of hippocampal NMDA receptor subunit NR2B expression and impaired memory. Neuropharmacology 2016; 103:222-35. [DOI: 10.1016/j.neuropharm.2015.12.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 11/30/2015] [Accepted: 12/17/2015] [Indexed: 11/16/2022]
|
25
|
Yuan Xiang P, Janc O, Grochowska KM, Kreutz MR, Reymann KG. Dopamine agonists rescue Aβ-induced LTP impairment by Src-family tyrosine kinases. Neurobiol Aging 2016; 40:98-102. [PMID: 26973108 DOI: 10.1016/j.neurobiolaging.2016.01.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2015] [Revised: 01/13/2016] [Accepted: 01/14/2016] [Indexed: 11/17/2022]
Abstract
Soluble forms of oligomeric amyloid beta (AβO) are involved in the loss of synaptic plasticity and memory, especially in early phases of Alzheimer's disease. Stimulation of dopamine D1/D5 receptors (D1R/D5R) is known to increase surface expression of synaptic α-amino-3-hydroxyl-5-methyl-4-isoxazole-propionate subtype glutamate and N-methyl-D-aspartate subtype glutamate receptors and facilitates the induction of the late phase of long-term potentiation (LTP), probably via a related mechanism. In this study, we show that the D1/D5R agonist SKF38393 protects LTP of hippocampal CA1 synapses from the deleterious action of oligomeric amyloid beta. Unexpectedly, the D1R/D5R-mediated recovery of LTP is independent of protein kinase A or phospholipase C pathways. Instead, we found that the inhibition of Src-family tyrosine kinases completely abolished the protective effects of D1R/D5R stimulation in a cellular model of learning and memory.
Collapse
Affiliation(s)
- PingAn Yuan Xiang
- Research Group Neuroplasticity, Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Oliwia Janc
- RG Neuropharmacology, Leibniz Institute for Neurobiology, Magdeburg, Germany
| | | | - Michael R Kreutz
- Research Group Neuroplasticity, Leibniz Institute for Neurobiology, Magdeburg, Germany; Leibniz Group 'Dendritic Organelles and Synaptic Function', Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; German Center for Neurodegenerative Diseases, Magdeburg, Germany.
| | - Klaus G Reymann
- RG Neuropharmacology, Leibniz Institute for Neurobiology, Magdeburg, Germany; German Center for Neurodegenerative Diseases, Magdeburg, Germany.
| |
Collapse
|
26
|
Norepinephrine versus dopamine and their interaction in modulating synaptic function in the prefrontal cortex. Brain Res 2016; 1641:217-33. [PMID: 26790349 DOI: 10.1016/j.brainres.2016.01.005] [Citation(s) in RCA: 118] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 12/16/2015] [Accepted: 01/05/2016] [Indexed: 01/11/2023]
Abstract
Among the neuromodulators that regulate prefrontal cortical circuit function, the catecholamine transmitters norepinephrine (NE) and dopamine (DA) stand out as powerful players in working memory and attention. Perturbation of either NE or DA signaling is implicated in the pathogenesis of several neuropsychiatric disorders, including attention deficit hyperactivity disorder (ADHD), post-traumatic stress disorder (PTSD), schizophrenia, and drug addiction. Although the precise mechanisms employed by NE and DA to cooperatively control prefrontal functions are not fully understood, emerging research indicates that both transmitters regulate electrical and biochemical aspects of neuronal function by modulating convergent ionic and synaptic signaling in the prefrontal cortex (PFC). This review summarizes previous studies that investigated the effects of both NE and DA on excitatory and inhibitory transmissions in the prefrontal cortical circuitry. Specifically, we focus on the functional interaction between NE and DA in prefrontal cortical local circuitry, synaptic integration, signaling pathways, and receptor properties. Although it is clear that both NE and DA innervate the PFC extensively and modulate synaptic function by activating distinctly different receptor subtypes and signaling pathways, it remains unclear how these two systems coordinate their actions to optimize PFC function for appropriate behavior. Throughout this review, we provide perspectives and highlight several critical topics for future studies. This article is part of a Special Issue entitled SI: Noradrenergic System.
Collapse
|
27
|
de Bartolomeis A, Errico F, Aceto G, Tomasetti C, Usiello A, Iasevoli F. D-aspartate dysregulation in Ddo(-/-) mice modulates phencyclidine-induced gene expression changes of postsynaptic density molecules in cortex and striatum. Prog Neuropsychopharmacol Biol Psychiatry 2015; 62:35-43. [PMID: 25979765 DOI: 10.1016/j.pnpbp.2015.05.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 05/06/2015] [Accepted: 05/06/2015] [Indexed: 10/23/2022]
Abstract
N-methyl-D-aspartate receptor (NMDAR) hypofunction has been considered a key alteration in schizophrenia pathophysiology. Thus, several strategies aimed at enhancing glutamatergic transmission, included the introduction in therapy of D-amino acids, such as D-serine and D-cycloserine augmentation, have been proposed to counteract difficult-to-treat symptoms or treatment-resistant forms of schizophrenia. Another D-amino acid, D-aspartate, has recently gained increasing interest for its role in NMDAR activation and has been found reduced in post-mortem cortex of schizophrenia patients. NMDAR is the core of the postsynaptic density (PSD), a postsynaptic site involved in glutamate signaling and responsive to antipsychotic treatment. In this study, we investigated striatal and cortical gene expression of key PSD transcripts (i.e. Homer1a, Homer1b/c, and PSD-95) in mice with persistently elevated brain D-aspartate-levels, i.e. the D-aspartate-oxidase knockout mice (Ddo(-/-)). These animal models were analyzed both in naive condition and after phencyclidine (PCP) treatment. Naive Ddo(-/-) mice showed decreased Homer1a expression in the prefrontal cortex, increased Homer1b/c expression in the striatum, and decreased PSD-95 expression in the striatum and in the cortex. Acute PCP treatment restored, and even potentiated, Homer1a expression in the prefrontal cortex of mutant mice, while it had limited effects on the other genes. These results suggest that persistently elevated D-aspartate, by enhancing NMDA transmission, may cause complex adaptive mechanisms affecting Homer1a, which in turn may explain the recently demonstrated protective effects of this D-amino acid against PCP-induced behavioral alterations, such as ataxic behavior.
Collapse
Affiliation(s)
- Andrea de Bartolomeis
- Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, Reproductive and Odontostomatological Sciences, University School of Medicine Federico II, Naples, Italy.
| | - Francesco Errico
- CEINGE Biotecnologie Avanzate, Naples, Italy; Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | | | - Carmine Tomasetti
- Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, Reproductive and Odontostomatological Sciences, University School of Medicine Federico II, Naples, Italy
| | - Alessandro Usiello
- CEINGE Biotecnologie Avanzate, Naples, Italy; Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, Second University of Naples (SUN), Caserta, Italy
| | - Felice Iasevoli
- Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, Reproductive and Odontostomatological Sciences, University School of Medicine Federico II, Naples, Italy
| |
Collapse
|
28
|
Falsafi SK, Dierssen M, Ghafari M, Pollak A, Lubec G. Reduced cortical neurotransmitter receptor complex levels in fetal Down syndrome brain. Amino Acids 2015; 48:103-16. [PMID: 26269195 DOI: 10.1007/s00726-015-2062-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 07/24/2015] [Indexed: 12/27/2022]
Abstract
In this study, cortical receptor complex levels were determined in fetal Down syndrome (DS, trisomy 21) brain. Frontal cortices were obtained from individuals with DS (19th-22nd week of gestation) and controls. Membrane proteins were extracted, assayed on blue native gels and immunoblotted with brain receptor antibodies. Levels of a D1R-containing complex were markedly decreased in male and female cortices of DS individuals. Females with DS had significant reductions of nicotinic acetylcholine receptors α4 and α7, NMDA receptor GluN1 and AMPA receptor GluA1- and GluA3-containing receptor complexes. Levels of other brain receptor complexes (5-hydroxytryptamine 1A, GluA2 and GluR4 receptor-containing complexes) were comparable between the groups of females. Levels of GluA2- and GluA3-containing complexes were significantly increased in males. Decreased levels of D1R complexes in both sexes, along with the significant reduction of α4, α7-containing receptor complexes observed in females, may explain the brain deficits and impaired cognition observed in DS.
Collapse
Affiliation(s)
- Soheil Keihan Falsafi
- Department of Pediatrics, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria.
| | - Mara Dierssen
- Genes and Disease Program, Center for Genomic Regulation and CIBERER, Barcelona, Spain
| | - Maryam Ghafari
- Department of Pediatrics, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Arnold Pollak
- Department of Pediatrics, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Gert Lubec
- Department of Pediatrics, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria.
| |
Collapse
|
29
|
Di Miceli M, Gronier B. Psychostimulants and atomoxetine alter the electrophysiological activity of prefrontal cortex neurons, interaction with catecholamine and glutamate NMDA receptors. Psychopharmacology (Berl) 2015; 232:2191-205. [PMID: 25572531 DOI: 10.1007/s00213-014-3849-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 12/12/2014] [Indexed: 12/29/2022]
Abstract
RATIONALE Attention-deficit hyperactivity disorder (ADHD) is the most frequently diagnosed neuropsychiatric disorder in childhood. Currently available ADHD drugs include the psychostimulants methylphenidate (MPH) and D-amphetamine (D-AMP), acting on norepinephrine and dopamine transporters/release, and atomoxetine (ATX), a selective norepinephrine uptake inhibitor. Recent evidence suggests an involvement of glutamate neurotransmission in the pathology and treatment of ADHD, via mechanisms to be clarified. OBJECTIVE We have investigated how ADHD drugs could modulate, through interaction with catecholamine receptors, basal and glutamate-induced excitability of pyramidal neurons in the prefrontal cortex (PFC), a region which plays a major role in control of attention and impulsivity. METHODS We have used the technique of extracellular single-unit recording in anaesthetised rats coupled with microiontophoresis. RESULTS Both MPH (1-3 mg/kg) and D-AMP (1-9 mg/kg) increased the firing activity of PFC neurons in a dopamine D1 receptor-dependent manner. ATX administration (1-6 mg/kg) also increased the firing of neurons, but this effect is not significantly reversed by D1 (SCH 23390) or alpha1 (prazosin) receptor antagonists but potentiated by alpha2 antagonist (yohimbine). All drugs induced a clear potentiation of the excitatory response of PFC neurons to the microiontophoretic application of the glutamate agonist N-methyl-D-aspartate (NMDA), but not to the glutamate agonist α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA). The potentiating effect of D-AMP on NMDA-induced activation of PFC neurons was partially reversed or prevented by dopamine D1 receptor blockade. CONCLUSION Our data shows that increase in excitability of PFC neurons in basal conditions and via NMDA receptor activation may be involved in the therapeutic response to ADHD drugs.
Collapse
Affiliation(s)
- Mathieu Di Miceli
- Leicester School of Pharmacy, De Montfort University, The Gateway, Leicester, LE1 9BH, UK
| | | |
Collapse
|
30
|
Buchta WC, Riegel AC. Chronic cocaine disrupts mesocortical learning mechanisms. Brain Res 2015; 1628:88-103. [PMID: 25704202 DOI: 10.1016/j.brainres.2015.02.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Revised: 01/28/2015] [Accepted: 02/01/2015] [Indexed: 01/06/2023]
Abstract
The addictive power of drugs of abuse such as cocaine comes from their ability to hijack natural reward and plasticity mechanisms mediated by dopamine signaling in the brain. Reward learning involves burst firing of midbrain dopamine neurons in response to rewards and cues predictive of reward. The resulting release of dopamine in terminal regions is thought to act as a teaching signaling to areas such as the prefrontal cortex and striatum. In this review, we posit that a pool of extrasynaptic dopaminergic D1-like receptors activated in response to dopamine neuron burst firing serve to enable synaptic plasticity in the prefrontal cortex in response to rewards and their cues. We propose that disruptions in these mechanisms following chronic cocaine use contribute to addiction pathology, in part due to the unique architecture of the mesocortical pathway. By blocking dopamine reuptake in the cortex, cocaine elevates dopamine signaling at these extrasynaptic receptors, prolonging D1-receptor activation and the subsequent activation of intracellular signaling cascades, and thus inducing long-lasting maladaptive plasticity. These cellular adaptations may account for many of the changes in cortical function observed in drug addicts, including an enduring vulnerability to relapse. Therefore, understanding and targeting these neuroadaptations may provide cognitive benefits and help prevent relapse in human drug addicts.
Collapse
Affiliation(s)
- William C Buchta
- Neurobiology of Addiction Research Center (NARC), Medical University of South Carolina, Charleston, SC 29425, USA
| | - Arthur C Riegel
- Neurobiology of Addiction Research Center (NARC), Medical University of South Carolina, Charleston, SC 29425, USA.
| |
Collapse
|
31
|
Recent progress in understanding subtype specific regulation of NMDA receptors by G Protein Coupled Receptors (GPCRs). Int J Mol Sci 2014; 15:3003-24. [PMID: 24562329 PMCID: PMC3958896 DOI: 10.3390/ijms15023003] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Revised: 12/30/2013] [Accepted: 02/12/2014] [Indexed: 12/24/2022] Open
Abstract
G Protein Coupled Receptors (GPCRs) are the largest family of receptors whose ligands constitute nearly a third of prescription drugs in the market. They are widely involved in diverse physiological functions including learning and memory. NMDA receptors (NMDARs), which belong to the ionotropic glutamate receptor family, are likewise ubiquitously expressed in the central nervous system (CNS) and play a pivotal role in learning and memory. Despite its critical contribution to physiological and pathophysiological processes, few pharmacological interventions aimed directly at regulating NMDAR function have been developed to date. However, it is well established that NMDAR function is precisely regulated by cellular signalling cascades recruited downstream of G protein coupled receptor (GPCR) stimulation. Accordingly, the downstream regulation of NMDARs likely represents an important determinant of outcome following treatment with neuropsychiatric agents that target selected GPCRs. Importantly, the functional consequence of such regulation on NMDAR function varies, based not only on the identity of the GPCR, but also on the cell type in which relevant receptors are expressed. Indeed, the mechanisms responsible for regulating NMDARs by GPCRs involve numerous intracellular signalling molecules and regulatory proteins that vary from one cell type to another. In the present article, we highlight recent findings from studies that have uncovered novel mechanisms by which selected GPCRs regulate NMDAR function and consequently NMDAR-dependent plasticity.
Collapse
|
32
|
Liu Y, Yang X, Suo Z, Xu Y, Hu X. Fyn kinase-regulated NMDA receptor- and AMPA receptor-dependent pain sensitization in spinal dorsal horn of mice. Eur J Pain 2014; 18:1120-8. [DOI: 10.1002/j.1532-2149.2014.00455.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/10/2013] [Indexed: 11/05/2022]
Affiliation(s)
- Y.N. Liu
- Department of Molecular Pharmacology; School of Pharmacy; Lanzhou University; Gansu China
- College of Chemistry and Chemical Engineering; Lanzhou University; Gansu China
| | - X. Yang
- Department of Molecular Pharmacology; School of Pharmacy; Lanzhou University; Gansu China
| | - Z.W. Suo
- Department of Molecular Pharmacology; School of Pharmacy; Lanzhou University; Gansu China
| | - Y.M. Xu
- Department of Molecular Pharmacology; School of Pharmacy; Lanzhou University; Gansu China
| | - X.D. Hu
- Department of Molecular Pharmacology; School of Pharmacy; Lanzhou University; Gansu China
| |
Collapse
|
33
|
Cahill E, Salery M, Vanhoutte P, Caboche J. Convergence of dopamine and glutamate signaling onto striatal ERK activation in response to drugs of abuse. Front Pharmacol 2014; 4:172. [PMID: 24409148 PMCID: PMC3884214 DOI: 10.3389/fphar.2013.00172] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Accepted: 12/19/2013] [Indexed: 12/31/2022] Open
Abstract
Despite their distinct targets, all addictive drugs commonly abused by humans evoke increases in dopamine (DA) concentration within the striatum. The main DA Guanine nucleotide binding protein couple receptors (GPCRs) expressed by medium-sized spiny neurons of the striatum are the D1R and D2R, which are positively and negatively coupled to cyclic adenosine monophosphate (cAMP)/protein kinase A (PKA) signaling, respectively. These two DA GPCRs are largely segregated into distinct neuronal populations, where they are co-expressed with glutamate receptors in dendritic spines. Direct and indirect interactions between DA GPCRs and glutamate receptors are the molecular basis by which DA modulates glutamate transmission and controls striatal plasticity and behavior induced by drugs of abuse. A major downstream target of striatal D1R is the extracellular signal-regulated kinase (ERK) kinase pathway. ERK activation by drugs of abuse behaves as a key integrator of D1R and glutamate NMDAR signaling. Once activated, ERK can trigger chromatin remodeling and induce gene expression that permits long-term cellular alterations and drug-induced morphological and behavioral changes. Besides the classical cAMP/PKA pathway, downstream of D1R, recent evidence implicates a cAMP-independent crosstalk mechanism by which the D1R potentiates NMDAR-mediated calcium influx and ERK activation. The mounting evidence of reciprocal modulation of DA and glutamate receptors adds further intricacy to striatal synaptic signaling and is liable to prove relevant for addictive drug-induced signaling, plasticity, and behavior. Herein, we review the evidence that built our understanding of the consequences of this synergistic signaling for the actions of drugs of abuse.
Collapse
Affiliation(s)
- Emma Cahill
- UMRS 952, INSERM, Physiopathologie des Maladies du Système Nerveux Central Paris, France ; UMR7224, CNRS, Physiopathologie des Maladies du Système Nerveux Central Paris, France ; University Pierre and Marie Curie-Paris 6 Paris, France
| | - Marine Salery
- UMRS 952, INSERM, Physiopathologie des Maladies du Système Nerveux Central Paris, France ; UMR7224, CNRS, Physiopathologie des Maladies du Système Nerveux Central Paris, France ; University Pierre and Marie Curie-Paris 6 Paris, France
| | - Peter Vanhoutte
- UMRS 952, INSERM, Physiopathologie des Maladies du Système Nerveux Central Paris, France ; UMR7224, CNRS, Physiopathologie des Maladies du Système Nerveux Central Paris, France ; University Pierre and Marie Curie-Paris 6 Paris, France
| | - Jocelyne Caboche
- UMRS 952, INSERM, Physiopathologie des Maladies du Système Nerveux Central Paris, France ; UMR7224, CNRS, Physiopathologie des Maladies du Système Nerveux Central Paris, France ; University Pierre and Marie Curie-Paris 6 Paris, France
| |
Collapse
|
34
|
Dopamine and extinction: a convergence of theory with fear and reward circuitry. Neurobiol Learn Mem 2013; 108:65-77. [PMID: 24269353 DOI: 10.1016/j.nlm.2013.11.007] [Citation(s) in RCA: 153] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Revised: 11/01/2013] [Accepted: 11/08/2013] [Indexed: 01/11/2023]
Abstract
Research on dopamine lies at the intersection of sophisticated theoretical and neurobiological approaches to learning and memory. Dopamine has been shown to be critical for many processes that drive learning and memory, including motivation, prediction error, incentive salience, memory consolidation, and response output. Theories of dopamine's function in these processes have, for the most part, been developed from behavioral approaches that examine learning mechanisms in reward-related tasks. A parallel and growing literature indicates that dopamine is involved in fear conditioning and extinction. These studies are consistent with long-standing ideas about appetitive-aversive interactions in learning theory and they speak to the general nature of cellular and molecular processes that underlie behavior. We review the behavioral and neurobiological literature showing a role for dopamine in fear conditioning and extinction. At a cellular level, we review dopamine signaling and receptor pharmacology, cellular and molecular events that follow dopamine receptor activation, and brain systems in which dopamine functions. At a behavioral level, we describe theories of learning and dopamine function that could describe the fundamental rules underlying how dopamine modulates different aspects of learning and memory processes.
Collapse
|
35
|
Ladépêche L, Dupuis JP, Groc L. Surface trafficking of NMDA receptors: gathering from a partner to another. Semin Cell Dev Biol 2013; 27:3-13. [PMID: 24177014 DOI: 10.1016/j.semcdb.2013.10.005] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Revised: 10/11/2013] [Accepted: 10/22/2013] [Indexed: 10/26/2022]
Abstract
Understanding the molecular and cellular pathways by which neurons integrate signals from different neurotransmitter systems has been among the major challenges of modern neuroscience. The ionotropic glutamate NMDA receptor plays a key role in the maturation and plasticity of glutamate synapses, both in physiology and pathology. It recently appeared that the surface distribution of NMDA receptors is dynamically regulated through lateral diffusion, providing for instance a powerful way to rapidly affect the content and composition of synaptic receptors. The ability of various neuromodulators to regulate NMDA receptor signaling revealed that this receptor can also serve as a molecular integrator of the ambient neuronal environment. Although still in its infancy, we here review our current understanding of the cellular regulation of NMDA receptor surface dynamics. We specifically discuss the roles of well-known modulators, such as dopamine, and membrane interactors in these regulatory processes, exemplifying the recent evidence that the direct interaction between NMDAR and dopamine receptors regulates their surface diffusion and distribution. In addition to the well-established modulation of NMDA receptor signaling by intracellular pathways, the surface dynamics of the receptor is now emerging as the first level of regulation, opening new pathophysiological perspectives for innovative therapeutical strategies.
Collapse
Affiliation(s)
- Laurent Ladépêche
- Université de Bordeaux, Interdisciplinary Institute for Neuroscience, UMR 5297, F-33000 Bordeaux, France; CNRS, Interdisciplinary Institute for Neuroscience, UMR 5297, F-33000 Bordeaux, France
| | - Julien Pierre Dupuis
- Université de Bordeaux, Interdisciplinary Institute for Neuroscience, UMR 5297, F-33000 Bordeaux, France; CNRS, Interdisciplinary Institute for Neuroscience, UMR 5297, F-33000 Bordeaux, France
| | - Laurent Groc
- Université de Bordeaux, Interdisciplinary Institute for Neuroscience, UMR 5297, F-33000 Bordeaux, France; CNRS, Interdisciplinary Institute for Neuroscience, UMR 5297, F-33000 Bordeaux, France.
| |
Collapse
|
36
|
Single-molecule imaging of the functional crosstalk between surface NMDA and dopamine D1 receptors. Proc Natl Acad Sci U S A 2013; 110:18005-10. [PMID: 24127604 DOI: 10.1073/pnas.1310145110] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Dopamine is a powerful modulator of glutamatergic neurotransmission and NMDA receptor-dependent synaptic plasticity. Although several intracellular cascades participating in this functional dialogue have been identified over the last few decades, the molecular crosstalk between surface dopamine and glutamate NMDA receptor (NMDAR) signaling still remains poorly understood. Using a combination of single-molecule detection imaging and electrophysiology in live hippocampal neurons, we demonstrate here that dopamine D1 receptors (D1Rs) and NMDARs form dynamic surface clusters in the vicinity of glutamate synapses. Strikingly, D1R activation or D1R/NMDAR direct interaction disruption decreases the size of these clusters, increases NMDAR synaptic content through a fast lateral redistribution of the receptors, and favors long-term synaptic potentiation. Together, these data demonstrate the presence of dynamic D1R/NMDAR perisynaptic reservoirs favoring a rapid and bidirectional surface crosstalk between receptors and set the plasma membrane as the primary stage of the dopamine-glutamate interplay.
Collapse
|
37
|
Ladepeche L, Yang L, Bouchet D, Groc L. Regulation of dopamine D1 receptor dynamics within the postsynaptic density of hippocampal glutamate synapses. PLoS One 2013; 8:e74512. [PMID: 24040266 PMCID: PMC3765443 DOI: 10.1371/journal.pone.0074512] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Accepted: 08/02/2013] [Indexed: 01/12/2023] Open
Abstract
Dopamine receptor potently modulates glutamate signalling, synaptic plasticity and neuronal network adaptations in various pathophysiological processes. Although key intracellular signalling cascades have been identified, the cellular mechanism by which dopamine and glutamate receptor-mediated signalling interplay at glutamate synapse remain poorly understood. Among the cellular mechanisms proposed to aggregate D1R in glutamate synapses, the direct interaction between D1R and the scaffold protein PSD95 or the direct interaction with the glutamate NMDA receptor (NMDAR) have been proposed. To tackle this question we here used high-resolution single nanoparticle imaging since it provides a powerful way to investigate at the sub-micron resolution the dynamic interaction between these partners in live synapses. We demonstrate in hippocampal neuronal networks that dopamine D1 receptors (D1R) laterally diffuse within glutamate synapses, in which their diffusion is reduced. Disrupting the interaction between D1R and PSD95, through genetical manipulation and competing peptide, did not affect D1R dynamics in glutamatergic synapses. However, preventing the physical interaction between D1R and the GluN1 subunit of NMDAR abolished the synaptic stabilization of diffusing D1R. Together, these data provide direct evidence that the interaction between D1R and NMDAR in synapses participate in the building of the dopamine-receptor-mediated signalling, and most likely to the glutamate-dopamine cross-talk.
Collapse
Affiliation(s)
- Laurent Ladepeche
- Univ. de Bordeaux, Interdisciplinary Institute for Neuroscience, Unité Mixte de recherche UMR 5297, Bordeaux, France
- Centre National de la Recherche Scientifique CNRS, IINS UMR 5297, Bordeaux, France
| | - Luting Yang
- Univ. de Bordeaux, Interdisciplinary Institute for Neuroscience, Unité Mixte de recherche UMR 5297, Bordeaux, France
- Centre National de la Recherche Scientifique CNRS, IINS UMR 5297, Bordeaux, France
| | - Delphine Bouchet
- Univ. de Bordeaux, Interdisciplinary Institute for Neuroscience, Unité Mixte de recherche UMR 5297, Bordeaux, France
- Centre National de la Recherche Scientifique CNRS, IINS UMR 5297, Bordeaux, France
| | - Laurent Groc
- Univ. de Bordeaux, Interdisciplinary Institute for Neuroscience, Unité Mixte de recherche UMR 5297, Bordeaux, France
- Centre National de la Recherche Scientifique CNRS, IINS UMR 5297, Bordeaux, France
- * E-mail:
| |
Collapse
|
38
|
Extrasynaptic targeting of NMDA receptors following D1 dopamine receptor activation and cocaine self-administration. J Neurosci 2013; 33:9451-61. [PMID: 23719812 DOI: 10.1523/jneurosci.5730-12.2013] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
We previously showed that after repeated exposure to cocaine, D1-like dopamine receptor (D1DR) stimulation reverses plastic changes of AMPA receptor-mediated signaling in the nucleus accumbens shell. However, there is little information on the impact of cocaine self-administration on D1-NMDA receptor interactions in this brain region. Here, using whole-cell patch-clamp recordings, we assessed whether cocaine self-administration alters the effects of D1DR stimulation on synaptic and extrasynaptic NMDA receptors (NMDARs). In slices from cocaine-naive rats, pretreatment with a D1DR agonist decreased synaptic NMDAR-mediated currents and increased the contribution of extrasynaptic NMDARs. In contrast, neither cocaine self-administration alone nor cocaine experience followed by D1DR stimulation had an effect on synaptic or extrasynaptic NMDAR signaling. Activation of extrasynaptic NMDARs relies on the availability of extracellular glutamate, which is regulated primarily by glutamate transporters. In cocaine-experienced animals, relative to cocaine-naive rats, administration of a glutamate reuptake blocker, DL-threo-β-benzyloxyaspartic acid, revealed increased extrasynaptic NMDAR activity and stronger baseline activity of glutamate uptake transporters. In cocaine-naive rats, the D1DR-mediated increase in extrasynaptic NMDAR signaling was independent of the activity of glutamate reuptake transporters. Together, these results indicate that cocaine experience blunts the influence of D1DRs on synaptic and extrasynaptic NMDAR signaling. Additionally, prior cocaine self-administration limits activation of the extrasynaptic NMDAR pool by increasing glutamate reuptake. These findings outline a pattern of adaptive interactions between D1DRs and NMDARs in the nucleus accumbens shell and demonstrate upregulation of extrasynaptic NMDAR signaling as a novel consequence of cocaine self-administration.
Collapse
|
39
|
Bazyan AS, van Luijtelaar G. Neurochemical and behavioral features in genetic absence epilepsy and in acutely induced absence seizures. ISRN NEUROLOGY 2013; 2013:875834. [PMID: 23738145 PMCID: PMC3664506 DOI: 10.1155/2013/875834] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Accepted: 02/06/2013] [Indexed: 02/08/2023]
Abstract
The absence epilepsy typical electroencephalographic pattern of sharp spikes and slow waves (SWDs) is considered to be due to an interaction of an initiation site in the cortex and a resonant circuit in the thalamus. The hyperpolarization-activated cyclic nucleotide-gated cationic I h pacemaker channels (HCN) play an important role in the enhanced cortical excitability. The role of thalamic HCN in SWD occurrence is less clear. Absence epilepsy in the WAG/Rij strain is accompanied by deficiency of the activity of dopaminergic system, which weakens the formation of an emotional positive state, causes depression-like symptoms, and counteracts learning and memory processes. It also enhances GABAA receptor activity in the striatum, globus pallidus, and reticular thalamic nucleus, causing a rise of SWD activity in the cortico-thalamo-cortical networks. One of the reasons for the occurrence of absences is that several genes coding of GABAA receptors are mutated. The question arises: what the role of DA receptors is. Two mechanisms that cause an infringement of the function of DA receptors in this genetic absence epilepsy model are proposed.
Collapse
Affiliation(s)
- A. S. Bazyan
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Science, Russian Federation, 5A Butlerov Street, Moscow 117485, Russia
| | - G. van Luijtelaar
- Biological Psychology, Donders Centre for Cognition, Donders Institute for Brain, Cognition and Behavior, Radboud University Nijmegen, P.O. Box 9104, 6500 HE Nijmegen, The Netherlands
| |
Collapse
|
40
|
Wang X, Pinto-Duarte A, Sejnowski TJ, Behrens MM. How Nox2-containing NADPH oxidase affects cortical circuits in the NMDA receptor antagonist model of schizophrenia. Antioxid Redox Signal 2013; 18:1444-62. [PMID: 22938164 PMCID: PMC3603498 DOI: 10.1089/ars.2012.4907] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2012] [Accepted: 09/02/2012] [Indexed: 12/13/2022]
Abstract
SIGNIFICANCE Schizophrenia is a complex neuropsychiatric disorder affecting around 1% of the population worldwide. Its mode of inheritance suggests a multigenic neurodevelopmental disorder with symptoms appearing during late adolescence/early adulthood, with its onset strongly influenced by environmental stimuli. Many neurotransmitter systems, including dopamine, glutamate, and gamma-aminobutyric acid, show alterations in affected individuals, and the behavioral and physiological characteristics of the disease can be mimicked by drugs that produce blockade of N-methyl-d-aspartate glutamate receptors (NMDARs). RECENT ADVANCES Mounting evidence suggests that drugs that block NMDARs specifically impair the inhibitory capacity of parvalbumin-expressing (PV+) fast-spiking neurons in adult and developing rodents, and alterations in these inhibitory neurons is one of the most consistent findings in the schizophrenic postmortem brain. Disruption of the inhibitory capacity of PV+ inhibitory neurons will alter the functional balance between excitation and inhibition in prefrontal cortical circuits producing impairment of working memory processes such as those observed in schizophrenia. CRITICAL ISSUES Mechanistically, the effect of NMDAR antagonists can be attributed to the activation of the Nox2-dependent reduced form of nicotinamide adenine dinucleotide phosphate oxidase pathway in cortical neurons, which is consistent with the emerging role of oxidative stress in the pathogenesis of mental disorders, specifically schizophrenia. Here we review the mechanisms by which NMDAR antagonists produce lasting impairment of the cortical PV+ neuronal system and the roles played by Nox2-dependent oxidative stress mechanisms. FUTURE DIRECTIONS The discovery of the pathways by which oxidative stress leads to unbalanced excitation and inhibition in cortical neural circuits opens a new perspective toward understanding the biological underpinnings of schizophrenia.
Collapse
Affiliation(s)
- Xin Wang
- The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | | | | | | |
Collapse
|
41
|
Wang MJ, Li YC, Snyder MA, Wang H, Li F, Gao WJ. Group II metabotropic glutamate receptor agonist LY379268 regulates AMPA receptor trafficking in prefrontal cortical neurons. PLoS One 2013; 8:e61787. [PMID: 23593498 PMCID: PMC3625159 DOI: 10.1371/journal.pone.0061787] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Accepted: 03/13/2013] [Indexed: 12/26/2022] Open
Abstract
Group II metabotropic glutamate receptor (mGluR) agonists have emerged as potential treatment drugs for schizophrenia and other neurological disorders, whereas the mechanisms involved remain elusive. Here we examined the effects of LY379268 (LY37) on the expression and trafficking of the α-amino-3-hydroxy-5-methyl-4-isoxazole propionate (AMPA) receptor subunits GluA1 and GluA2 in prefrontal neurons. We show that LY37 significantly increased the surface and total expression of both GluA1 and GluA2 subunits in cultured prefrontal neurons and in vivo. This effect was mimicked by the selective mGluR2 agonist LY395756 and was blocked by mGluR2/3 antagonist LY341495. Moreover, we found that both GluA1 and GluA2 subunits were colocalized with PSD95 but not synapsin I, suggesting a postsynaptic localization. Consistently, treatment with LY37 significantly increased the amplitude, but not frequency, of miniature excitatory postsynaptic currents. Further, actinomycin-D blocked LY37's effects, suggesting a transcriptional regulation. In addition, application of glycogen synthase kinase-3beta (GSK-3β) inhibitor completely blocked LY37's effect on GluA2 surface expression, whereas GSK-3β inhibitor itself induced decreases in the surface and total protein levels of GluA1, but not GluA2 subunits. This suggests that GSK-3β differentially mediates GluA1 and GluA2 trafficking. Further, LY37 significantly increased the phosphorylation, but not total protein, of extracellular signal-regulated kinase 1/2 (ERK1/2). Neither ERK1/2 inhibitor PD98059 alone nor PD98059 combined with LY37 treatment induced changes in GluA1 or GluA2 surface expression or total protein levels. Our data thus suggest that mGluR2/3 agonist regulates postsynaptic AMPA receptors by affecting the synaptic trafficking of both GluA1 and GluA2 subunits and that the regulation is likely through ERK1/2 signaling in GluA1 and/or both ERK1/2 and GSK-3β signaling pathways in the GluA2 subunit.
Collapse
Affiliation(s)
- Min-Juan Wang
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
- Department of Neurobiology and Anatomy, Zhongshan College of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Yan-Chun Li
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Melissa A. Snyder
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Huaixing Wang
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Feng Li
- Department of Neurobiology and Anatomy, Zhongshan College of Medicine, Sun Yat-Sen University, Guangzhou, China
- * E-mail: (WJG); (FL)
| | - Wen-Jun Gao
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
- * E-mail: (WJG); (FL)
| |
Collapse
|
42
|
Yang B, Ni J, Zeng Z, Shi B, You W, Ke C. Cloning and characterization of the dopamine like receptor in the oyster Crassostrea angulata: expression during the ovarian cycle. Comp Biochem Physiol B Biochem Mol Biol 2012; 164:168-75. [PMID: 23274282 DOI: 10.1016/j.cbpb.2012.12.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Revised: 12/20/2012] [Accepted: 12/20/2012] [Indexed: 01/11/2023]
Abstract
We cloned and characterized a complete cDNA encoding a dopamine receptor (DAR) named Ca-DA1R from Fujian oyster, Crassostrea angulata. The 2843 bp long cDNA sequence includes a 916-bp 5'-UTR, the 1197 bp ORF which encodes a putative protein of 399 amino acids, and a 729 bp 3'-UTR. The Ca-DA1R sequence possesses typical characteristics of a D1 receptor: two main features being a short third intracellular loop and a long inner COOH-terminal tail domain. Using a real-time PCR approach, expression profiles of Ca-DA1R were analyzed in adult tissues and during the four stages of ovarian development. Ca-DA1R was expressed ubiquitously, although transcript levels varied between tissues, with higher mRNA levels detected in the ovary, labial palps and mantle. During the four stages of ovarian development, Ca-DA1R mRNA expression level was higher in the proliferation stage than in the other three stages during the ovary cycle. In situ hybridization results reveal that the Ca-DA1R mRNA is mainly expressed in the epithelium of the gonoducts. These observations suggest that Ca-DA1R binding of DA probably plays an important role in early ovarian development and via regulating oocyte locomotion cooperates with the 5-HT receptor system during the ovarian cycle in C. angulata.
Collapse
Affiliation(s)
- Bingye Yang
- State Key Laboratory of Marine Environmental Science, Xiamen University, Xiamen 361005, PR China
| | | | | | | | | | | |
Collapse
|
43
|
Wang M, Wong AH, Liu F. Interactions between NMDA and dopamine receptors: A potential therapeutic target. Brain Res 2012; 1476:154-63. [DOI: 10.1016/j.brainres.2012.03.029] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2012] [Revised: 03/08/2012] [Accepted: 03/10/2012] [Indexed: 11/26/2022]
|
44
|
Directional gating of synaptic plasticity by GPCRs and their distinct downstream signalling pathways. EMBO J 2012; 31:783-5. [PMID: 22334046 DOI: 10.1038/emboj.2012.3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|
45
|
Remington G, Agid O, Foussias G. Schizophrenia as a disorder of too little dopamine: implications for symptoms and treatment. Expert Rev Neurother 2011; 11:589-607. [PMID: 21469931 DOI: 10.1586/ern.10.191] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Antipsychotics represent the first effective therapy for schizophrenia, with their benefits linked to dopamine D2 blockade. Schizophrenia was soon identified as a hyperdopaminergic disorder, and antipsychotics proved to be reasonably effective in controlling positive symptoms. However, over the years, schizophrenia has been reconceptualized more broadly, now defined as a heterogeneous disorder with multiple symptom domains. Negative and cognitive features, not particularly responsive to antipsychotic therapy, have taken on increased importance--current thinking suggests that these domains predate the onset of positive symptoms and are more closely tied to functional outcome. That they are better understood in the context of decreased dopamine activity suggests that schizophrenia may fundamentally represent a hypodopaminergic disorder. This shift in thinking has important theoretical implications from the standpoint of etiology and pathophysiology, but also clinically in terms of treatment and drug development.
Collapse
|
46
|
Groveman BR, Feng S, Fang XQ, Pflueger M, Lin SX, Bienkiewicz EA, Yu X. The regulation of N-methyl-D-aspartate receptors by Src kinase. FEBS J 2011; 279:20-8. [PMID: 22060915 DOI: 10.1111/j.1742-4658.2011.08413.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Src family kinases (SFKs) play critical roles in the regulation of many cellular functions by growth factors, G-protein-coupled receptors and ligand-gated ion channels. Recent data have shown that SFKs serve as a convergent point of multiple signaling pathways regulating N-methyl-d-aspartate (NMDA) receptors in the central nervous system. Multiple SFK molecules, such as Src and Fyn, closely associate with their substrate, NMDA receptors, via indirect and direct binding mechanisms. The NMDA receptor is associated with an SFK signaling complex consisting of SFKs; the SFK-activating phosphatase, protein tyrosine phosphatase α; and the SFK-inactivating kinase, C-terminal Src kinase. Early studies have demonstrated that intramolecular interactions with the SH2 or SH3 domain lock SFKs in a closed conformation. Disruption of the interdomain interactions can induce the activation of SFKs with multiple signaling pathways involved in regulation of this process. The enzyme activity of SFKs appears 'graded', exhibiting different levels coinciding with activation states. It has also been proposed that the SH2 and SH3 domains may stimulate catalytic activity of protein tyrosine kinases, such as Abl. Recently, it has been found that the enzyme activity of neuronal Src protein is associated with its stability, and that the SH2 and SH3 domain interactions may act not only to constrain the activation of neuronal Src, but also to regulate the enzyme activity of active neuronal Src. Collectively, these findings demonstrate novel mechanisms underlying the regulation of SFKs.
Collapse
Affiliation(s)
- Bradley R Groveman
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL 32306, USA.
| | | | | | | | | | | | | |
Collapse
|
47
|
|
48
|
Ehrnhoefer DE, Wong BKY, Hayden MR. Convergent pathogenic pathways in Alzheimer's and Huntington's diseases: shared targets for drug development. Nat Rev Drug Discov 2011; 10:853-67. [PMID: 22015920 DOI: 10.1038/nrd3556] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Neurodegenerative diseases, exemplified by Alzheimer's disease and Huntington's disease, are characterized by progressive neuropsychiatric dysfunction and loss of specific neuronal subtypes. Although there are differences in the exact sites of pathology, and the clinical profiles of these two conditions only partially overlap, considerable similarities in disease mechanisms and pathogenic pathways can be observed. These shared mechanisms raise the possibility of exploiting common therapeutic targets for drug development. As Huntington's disease has a monogenic cause, it is possible to accurately identify individuals who carry the Huntington's disease mutation but do not yet manifest symptoms. These individuals could act as a model for Alzheimer's disease to test therapeutic interventions that target shared pathogenic pathways.
Collapse
Affiliation(s)
- Dagmar E Ehrnhoefer
- Centre for Molecular Medicine and Therapeutics, Department of Medical Genetics, Child & Family Research Institute, University of British Columbia, 950 West 28th Avenue, Vancouver, British Columbia V5Z 4H4, Canada
| | | | | |
Collapse
|
49
|
MPTP-meditated hippocampal dopamine deprivation modulates synaptic transmission and activity-dependent synaptic plasticity. Toxicol Appl Pharmacol 2011; 254:332-41. [DOI: 10.1016/j.taap.2011.05.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2011] [Revised: 05/06/2011] [Accepted: 05/08/2011] [Indexed: 11/20/2022]
|
50
|
Abstract
INTRODUCTION Since the development of clozapine, scores of antipsychotics have been introduced. These are, with one exception (aripiprazole), based on the pharmacological principle of 5-HT(2)/dopamine antagonism. Research on other treatment targets, which, in part, influence dopaminergic pathways directly or indirectly, is mounting. Managing psychotic symptoms is only one facet of successful treatment of schizophrenia. Effective remedies against negative symptoms and cognitive deficits are still an unmet clinical need. AREAS COVERED With the focus on the topics mentioned above, the authors briefly review the latest clinical research organized on the basis of receptor systems and other drug targets, which are discussed to be involved in the pathophysiology of schizophrenia. EXPERT OPINION In conclusion, although clinicians will have to have considerable patience before truly novel anti-schizophrenia treatments become obtainable, a number of interesting leads with considerable theoretical potential are being explored. As yet, it is difficult to predict which of these mechanisms will effectively augment the currently available treatments.
Collapse
Affiliation(s)
- Falko Biedermann
- Medical University Innsbruck, Department for Psychiatry and Psychotherapy, Clinic for Biological Psychiatry, Anichstrasse 35, A-6020 Innsbruck Austria.
| | | |
Collapse
|