1
|
Lyu Z, Gong Z, Huang M, Xin S, Zou M, Ding Y. Benefits of exercise on cognitive impairment in alcohol use disorder following alcohol withdrawal. FEBS Open Bio 2024; 14:1540-1558. [PMID: 39054261 PMCID: PMC11492329 DOI: 10.1002/2211-5463.13865] [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: 10/10/2023] [Revised: 06/21/2024] [Accepted: 07/11/2024] [Indexed: 07/27/2024] Open
Abstract
Although most cognitive impairments induced by prolonged alcohol consumption tend to improve within the initial months of abstinence, there is evidence suggesting certain cognitive deficits may persist. This study aimed to investigate the impact of aerobic exercise on learning and memory in alcohol use disorder (AUD) mice following a period of abstinence from alcohol. We also sought to assess the levels of monoamine neurotransmitters in the hippocampus. To this end, we established an AUD mouse model through a two-bottle choice (sucrose fading mode and normal mode) and chronic intermittent alcohol vapor (combined with intraperitoneal injection) and randomly allocated mice into exercise groups to undergo treadmill training. Learning and memory abilities were assessed through the Morris water maze test and spontaneous activity was evaluated using the open field test. The levels of dopamine, norepinephrine, serotonin, and brain-derived neurotrophic factor in the hippocampus were quantified using enzyme-linked immunoassay (ELISA) kits. The findings reveal that after cessation of alcohol consumption, learning and memory abilities in AUD mice did not completely return to normal levels. The observed enhancement of cognitive functions in AUD mice through aerobic exercise may be attributed to restoring levels of monoamine neurotransmitters in the hippocampus, boosting brain-derived neurotrophic factor (BDNF) concentrations, and facilitating an increase in hippocampal mass. These results offer empirical evidence to support aerobic exercise as a viable therapeutic strategy to alleviate cognitive deficits associated with AUD.
Collapse
Affiliation(s)
- Zhen Lyu
- Key Lab of Aquatic Sports Training Monitoring and Intervention of General Administration of Sport of China, Faculty of Physical EducationJiangxi Normal UniversityNanchangChina
- School of PsychologyShanghai University of SportChina
| | - Zhi‐Gang Gong
- Key Lab of Aquatic Sports Training Monitoring and Intervention of General Administration of Sport of China, Faculty of Physical EducationJiangxi Normal UniversityNanchangChina
| | - Min‐Xia Huang
- Science and Technology College of Nanchang Hangkong UniversityJiujiangChina
| | - Si‐Ping Xin
- Key Lab of Aquatic Sports Training Monitoring and Intervention of General Administration of Sport of China, Faculty of Physical EducationJiangxi Normal UniversityNanchangChina
| | - Mao‐Zhong Zou
- Key Lab of Aquatic Sports Training Monitoring and Intervention of General Administration of Sport of China, Faculty of Physical EducationJiangxi Normal UniversityNanchangChina
| | - Yu‐Quan Ding
- Key Lab of Aquatic Sports Training Monitoring and Intervention of General Administration of Sport of China, Faculty of Physical EducationJiangxi Normal UniversityNanchangChina
| |
Collapse
|
2
|
Wang C, Hei Y, Liu Y, Bajpai AK, Li Y, Guan Y, Xu F, Yao C. Systems genetics identifies methionine as a high risk factor for Alzheimer's disease. Front Neurosci 2024; 18:1381889. [PMID: 39081851 PMCID: PMC11286400 DOI: 10.3389/fnins.2024.1381889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Accepted: 06/25/2024] [Indexed: 08/02/2024] Open
Abstract
As a dietary strategy, methionine restriction has been reported to promote longevity and regulate metabolic disorders. However, the role and possible regulatory mechanisms underlying methionine in neurodegenerative diseases such as Alzheimer's disease (AD), remain unexplored. This study utilized the data from BXD recombinant inbred (RI) mice to establish a correlation between the AD phenotype in mice and methionine level. Gene enrichment analysis indicated that the genes associated with the concentration of methionine in the midbrain are involved in the dopaminergic synaptic signaling pathway. Protein interaction network analysis revealed that glycogen synthase kinase 3 beta (GSK-3β) was a key regulator of the dopaminergic synaptic pathway and its expression level was significantly correlated with the AD phenotype. Finally, in vitro experiments demonstrated that methionine deprivation could reduce the expression of Aβ and phosphorylated Tau, suggesting that lowering methionine levels in humans may be a preventive or therapeutic strategy for AD. In conclusion, our findings support that methionine is a high risk factor for AD. These findings predict potential regulatory network, theoretically supporting methionine restriction to prevent AD.
Collapse
Affiliation(s)
- Congmin Wang
- School of Pharmacy, Binzhou Medical University, Yantai, China
| | - Yu Hei
- School of Pharmacy, Binzhou Medical University, Yantai, China
| | - Yu Liu
- School of Pharmacy, Binzhou Medical University, Yantai, China
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, China
| | - Akhilesh Kumar Bajpai
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Yuhe Li
- School of Pharmacy, Binzhou Medical University, Yantai, China
| | - Yawen Guan
- School of Pharmacy, Binzhou Medical University, Yantai, China
| | - Fuyi Xu
- School of Pharmacy, Binzhou Medical University, Yantai, China
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, China
| | - Cuifang Yao
- School of Pharmacy, Binzhou Medical University, Yantai, China
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, China
| |
Collapse
|
3
|
Oka M, Yoshino R, Kitanaka N, Hall FS, Uhl GR, Kitanaka J. Role of glycogen synthase kinase-3β in dependence and abuse liability of alcohol. Alcohol Alcohol 2024; 59:agad086. [PMID: 38145944 DOI: 10.1093/alcalc/agad086] [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: 07/30/2023] [Revised: 11/25/2023] [Accepted: 11/28/2023] [Indexed: 12/27/2023] Open
Abstract
BACKGROUND Alcohol is a major abused drug worldwide that contributes substantially to health and social problems. These problems result from acute alcohol overuse as well as chronic use, leading to alcohol use disorder (AUD). A major goal of this field is to establish a treatment for alcohol abuse and dependence in patients with AUD. The central molecular mechanisms of acute alcohol actions have been extensively investigated in rodent models. AIMS One of the central mechanisms that may be involved is glycogen synthase kinase-3β (GSK-3β) activity, a key enzyme involved in glycogen metabolism but which has crucial roles in numerous cellular processes. Although the exact mechanisms leading from acute alcohol actions to these chronic changes in GSK-3β function are not yet clear, GSK-3β nonetheless constitutes a potential therapeutic target for AUD by reducing its function using GSK-3β inhibitors. This review is focused on the correlation between GSK-3β activity and the degree of alcohol consumption. METHODS Research articles regarding investigation of effect of GSK-3β on alcohol consumption in rodents were searched on PubMed, Embase, and Scopus databases using keywords "glycogen synthase kinase," "alcohol (or ethanol)," "intake (or consumption)," and evaluated by changes in ratios of pGSK-3βSer9/pGSK-3β. RESULTS In animal experiments, GSK-3β activity decreases in the brain under forced and voluntary alcohol consumption while GSK-3β activity increases under alcohol-seeking behavior. CONCLUSIONS Several pieces of evidence suggest that alterations in GSK-3β function are important mediators of chronic ethanol actions, including those related to alcohol dependence and the adverse effects of chronic ethanol exposure.
Collapse
Affiliation(s)
- Masahiro Oka
- Laboratory of Drug Addiction and Experimental Therapeutics, Department of Pharmacy, School of Pharmacy, Hyogo Medical University, 1-3-6 Minatojima, Chuo-ku, Kobe, Hyogo 650-8530, Japan
| | - Rui Yoshino
- Laboratory of Drug Addiction and Experimental Therapeutics, Department of Pharmacy, School of Pharmacy, Hyogo Medical University, 1-3-6 Minatojima, Chuo-ku, Kobe, Hyogo 650-8530, Japan
| | - Nobue Kitanaka
- Department of Pharmacology, School of Medicine, Hyogo Medical University, Nishinomiya, Hyogo 663-8501, Japan
| | - F Scott Hall
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Frederic and Mary Wolfe Center HEB 282D, Mail Stop 1015, 3000 Arlington Avenue,Toledo, OH 43614, United States
| | - George R Uhl
- Neurology Service, VA Maryland Healthcare System, 10 North Greene Street, Baltimore, MD 21201, United States
- Departments of Neurology and Pharmacology, University of Maryland School of Medicine, 655 W. Baltimore Street, Baltimore, MD 21201, United States
| | - Junichi Kitanaka
- Laboratory of Drug Addiction and Experimental Therapeutics, Department of Pharmacy, School of Pharmacy, Hyogo Medical University, 1-3-6 Minatojima, Chuo-ku, Kobe, Hyogo 650-8530, Japan
| |
Collapse
|
4
|
Kim B, Kim JS, Youn B, Moon C. Dopamine depletion alters neuroplasticity-related signaling in the rat hippocampus. Anim Cells Syst (Seoul) 2023; 27:436-446. [PMID: 38125760 PMCID: PMC10732217 DOI: 10.1080/19768354.2023.2294308] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 12/08/2023] [Indexed: 12/23/2023] Open
Abstract
Dopamine (DA) plays a significant role in regulating hippocampal function, particularly in modulating synaptic plasticity. Despite this, a comprehensive understanding of the molecular mechanisms involved in neuroplasticity-related signaling influenced by DA remains incomplete. This study aimed to elucidate the changes in the expression of key molecules related to hippocampal neuroplasticity following DA depletion in rats. To induce DA depletion, unilateral striatal infusions of 6-hydroxydopamine (6-OHDA) were administered to adult Sprague-Dawley rats. The subsequent loss of nigrostriatal DAergic signaling in these 6-OHDA-lesioned rats was confirmed using an apomorphine-induced rotation test at 4 weeks post-infusion and by assessing the expression levels of tyrosine hydroxylase (TH) through immunohistochemistry and western blotting at 7 weeks post-infusion. A decrease in DAergic signaling, evidenced by reduced TH-positive immunoreactivity, was also noted in the ipsilateral hippocampus of the lesioned rats. Interestingly, 6-OHDA infusion led to increased phosphorylation of pivotal hippocampal plasticity-related proteins, including extracellular signal-regulated kinase (ERK), protein kinase B (Akt), glycogen synthase kinase 3β (GSK3β), and cAMP response element-binding protein (CREB), in the ipsilateral hippocampus 7 weeks following the infusion. To extend these findings, in vitro experiments were conducted on primary hippocampal neurons exposed to DA and/or the active D1/D2 DA receptor antagonist, flupentixol (Flux). DA inhibited the constitutive phosphorylation of ERK, Akt, GSK3, and CREB, while Flux restored these phosphorylation levels. Taken together, these findings indicate that DA depletion triggers an increase in plasticity-related signaling in the hippocampus, suggesting a possible compensatory mechanism that promotes activity-independent neuroplasticity following DA depletion.
Collapse
Affiliation(s)
- Bohye Kim
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, Republic of Korea
| | - Joong-Sun Kim
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, Republic of Korea
| | - BuHyun Youn
- Department of Biological Science, Pusan National University, Busan, Republic of Korea
| | - Changjong Moon
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, Republic of Korea
| |
Collapse
|
5
|
Wang X, Yang J, Wang W, Li Y, Yang Y. Decreasing REDD1 expression protects against high glucose-induced apoptosis, oxidative stress and inflammatory injury in podocytes through regulation of the AKT/GSK-3β/Nrf2 pathway. Immunopharmacol Immunotoxicol 2023; 45:527-538. [PMID: 36883011 DOI: 10.1080/08923973.2023.2183351] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 02/10/2023] [Indexed: 02/24/2023]
Abstract
OBJECTIVE Our goal in this work was to investigate the possible role and mechanism of regulated in development and DNA damage response 1 (REDD1) in mediating high glucose (HG)-induced podocyte injury in vitro. MATERIALS AND METHODS Mouse podocytes were stimulated with HG to establish HG injury model. Protein expression was examined by Western blotting. Cell viability was measured by cell counting kit-8 assay. Cell apoptosis was assessed by annexin V-FITC/propidium iodide and TUNEL apoptotic assays. Levels of reactive oxygen species (ROS), malondialdehyde (MDA), superoxide dismutase (SOD), and glutathione peroxidase (GPx) were quantified by commercial kits. Concentrations of tumor necrosis factor (TNF)-α, interleukin (IL)-6, and IL-1β were measured by ELISA. RESULTS A marked increase in REDD1 expression was observed in podocytes stimulated with HG. Reduced REDD1 expression strikingly restrained HG-induced increases in apoptosis, oxidative stress, and inflammation response in cultured podocytes. Decreasing REDD1 expression enhanced nuclear factor erythroid 2-related factor 2 (Nrf2) activation in HG-exposed podocytes via regulation of the AKT/glycogen synthase kinase-3 beta (GSK-3β) pathway. Inhibition of AKT or reactivation of GSK-3β prominently abolished Nrf2 activation induced by decreasing REDD1 expression. Pharmacological repression of Nrf2 markedly reversed the protective effects of decreasing REDD1 expression in HG-injured podocytes. CONCLUSION Our data demonstrate that decreasing REDD1 expression protects cultured podocytes from HG-induced injuries by potentiating Nrf2 signaling through regulation of the AKT/GSK-3β pathway. Our work underscores the potential role of REDD1-mediated podocyte injury during the development of diabetic kidney disease.
Collapse
Affiliation(s)
- Xiaojing Wang
- The First Clinical Medical College of Shanxi Medical University, Taiyuan, China
- Department of Endocrinology, Shanxi Yuncheng Central Hospital, Yuncheng, China
| | - Jing Yang
- The First Clinical Medical College of Shanxi Medical University, Taiyuan, China
| | - Wenxing Wang
- Department of Endocrinology, Shanxi Yuncheng Central Hospital, Yuncheng, China
| | - Yun Li
- Department of Endocrinology, Shanxi Yuncheng Central Hospital, Yuncheng, China
| | - Yue Yang
- Department of Endocrinology, Shanxi Yuncheng Central Hospital, Yuncheng, China
| |
Collapse
|
6
|
Murata M, Bilim V, Shirono Y, Kazama A, Hiruma K, Tasaki M, Tomita Y. MicroRNAs as Potential Regulators of GSK-3β in Renal Cell Carcinoma. Curr Issues Mol Biol 2023; 45:7432-7448. [PMID: 37754254 PMCID: PMC10529713 DOI: 10.3390/cimb45090470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 09/07/2023] [Accepted: 09/09/2023] [Indexed: 09/28/2023] Open
Abstract
The prognosis of patients with advanced renal cell carcinoma (RCC) has improved with newer therapies, including molecular-targeted therapies and immuno-oncology agents. Despite these therapeutic advances, many patients with metastatic disease remain uncured. Inhibition of glycogen synthase kinase-3β (GSK-3β) is a promising new therapeutic strategy for RCC; however, the precise regulatory mechanism has not yet been fully elucidated. MicroRNAs (miRNAs) act as post-translational regulators of target genes, and we investigated the potential regulation of miRNAs on GSK-3β in RCC. We selected nine candidate miRNAs from three databases that could potentially regulate GSK-3β. Among these, hsa-miR-4465 (miR-4465) was downregulated in RCC cell lines and renal cancer tissues. Furthermore, luciferase assays revealed that miR-4465 directly interacted with the 3' untranslated region of GSK-3β, and Western blot analysis showed that overexpression of miR-4465 significantly decreased GSK-3β protein expression. Functional assays showed that miR-4465 overexpression significantly suppressed cell invasion of A498 and Caki-1 cells; however, cell proliferation and migration were suppressed only in Caki-1 and A498 cells, respectively, with no effect on cell cycle and apoptosis. In conclusion, miR-4465 regulates GSK-3β expression but does not consistently affect RCC cell function as a single molecule. Further comprehensive investigation of regulatory networks is required in this field.
Collapse
Affiliation(s)
- Masaki Murata
- Department of Urology, Division of Molecular Oncology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8510, Japan; (V.B.); (Y.S.); (A.K.); (K.H.); (M.T.); (Y.T.)
| | - Vladimir Bilim
- Department of Urology, Division of Molecular Oncology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8510, Japan; (V.B.); (Y.S.); (A.K.); (K.H.); (M.T.); (Y.T.)
- Department of Urology, Kameda Daiichi Hospital, Niigata 950-0165, Japan
| | - Yuko Shirono
- Department of Urology, Division of Molecular Oncology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8510, Japan; (V.B.); (Y.S.); (A.K.); (K.H.); (M.T.); (Y.T.)
| | - Akira Kazama
- Department of Urology, Division of Molecular Oncology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8510, Japan; (V.B.); (Y.S.); (A.K.); (K.H.); (M.T.); (Y.T.)
- Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Kaede Hiruma
- Department of Urology, Division of Molecular Oncology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8510, Japan; (V.B.); (Y.S.); (A.K.); (K.H.); (M.T.); (Y.T.)
| | - Masayuki Tasaki
- Department of Urology, Division of Molecular Oncology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8510, Japan; (V.B.); (Y.S.); (A.K.); (K.H.); (M.T.); (Y.T.)
| | - Yoshihiko Tomita
- Department of Urology, Division of Molecular Oncology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8510, Japan; (V.B.); (Y.S.); (A.K.); (K.H.); (M.T.); (Y.T.)
| |
Collapse
|
7
|
Zalouli V, Rajavand H, Bayat M, Khaleghnia J, Sharifianjazi F, Jafarinazhad F, Beheshtizadeh N. Adult hippocampal neurogenesis (AHN) controls central nervous system and promotes peripheral nervous system regeneration via physical exercise. Biomed Pharmacother 2023; 165:115078. [PMID: 37390707 DOI: 10.1016/j.biopha.2023.115078] [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: 05/08/2023] [Revised: 06/21/2023] [Accepted: 06/23/2023] [Indexed: 07/02/2023] Open
Abstract
Physical exercise has beneficial effects on adult hippocampal neurogenesis (AHN) and cognitive processes, including learning. Although it is not known if anaerobic resistance training and high-intensity interval training, which involve alternating brief bouts of highly intense anaerobic activity with rest periods, have comparable effects on AHN. Also, while less thoroughly investigated, individual genetic diversity in the overall response to physical activity is likely to play a key role in the effects of exercise on AHN. Physical exercise has been shown to improve health on average, although the benefits may vary from person to person, perhaps due to genetic differences. Maximal aerobic capacity and metabolic health may improve significantly with aerobic exercise for some people, while the same amount of training may have little effect on others. This review discusses the AHN's capability for peripheral nervous system (PNS) regeneration and central nervous system (CNS) control via physical exercise. Exercise neurogenicity, effective genes, growth factors, and the neurotrophic factors involved in PNS regeneration and CNS control were discussed. Also, some disorders that could be affected by AHN and physical exercise are summarized.
Collapse
Affiliation(s)
- Vahideh Zalouli
- Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Hosnieh Rajavand
- Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mahdi Bayat
- Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran; Department of Medicine and Surgery, Physical Activity and Health Promotion, University of Tor Vergata, Rome, Italy
| | - Jalil Khaleghnia
- Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran; Department of Sport Sciences, Khavaran Institute of Higher Education, Mashhad, Iran
| | - Fariborz Sharifianjazi
- Department of Natural Sciences, School of Science and Technology, University of Georgia, Tbilisi 0171, Georgia
| | - Farzad Jafarinazhad
- Yeditepe University, Faculty of Health Sciences, Department of Physiotherapy and Rehabilitation, Istanbul, Turkey.
| | - Nima Beheshtizadeh
- Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran; Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
8
|
Hu X, Liu L, Wang Z, Sun Y, Li Z, Zhou G, Yue K, Wang L, Lian B, Lu G, Li C, Sun L. The potential role of GSK-3β signaling pathway for amelioration actions of ketamine on the PTSD rodent model. Brain Res Bull 2023; 200:110697. [PMID: 37392896 DOI: 10.1016/j.brainresbull.2023.110697] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 06/26/2023] [Accepted: 06/28/2023] [Indexed: 07/03/2023]
Abstract
RATIONALE Post-traumatic stress disorder (PTSD) is a complex, chronic psychiatric disorder typically triggered by life-threatening events and, as yet, lacks a specialized pharmacological treatment. The potential therapeutic role of ketamine, an N-methyl-D-aspartate receptor antagonist, in mitigating PTSD has been the subject of investigation. OBJECTIVE The aim of this study was to elucidate alterations in the glycogen synthase kinase-3β (GSK-3β) signaling pathway in response to ketamine intervention, using the single prolonged stress (SPS) model of PTSD at a molecular level. METHODS PTSD-like symptoms were simulated using the SPS model. Ketamine (10mg/kg) and GSK-3β antagonist SB216763 (5mg/kg) were then administered intraperitoneally. Stress-related behavior was evaluated through the open field test (OFT) and the elevated plus maze test (EMPT). Additionally, brain activity was analyzed using quantitative electroencephalography (qEEG). Changes in protein and mRNA expressions of glucocorticoid receptor (GR), brain-derived neurotrophic factor (BDNF), GSK-3β, phosphorylated ser-9 GSK-3β (p-GSK-3β), FK506 binding protein 5 (FKBP5), and corticotropin-releasing hormone (CRH) were assessed in the hypothalamus via western blot and qPCR. RESULTS SPS-exposed rats exhibited reduced distance and time spent in the center of the open arms, a pattern divergent from control rats. qEEG readings revealed SPS-induced increases in alpha power, low gamma and high gamma power. Furthermore, SPS triggered an upregulation in the protein and gene expression of GSK-3β, GR, BDNF, p-GSK-3β, and FKBP5, and downregulated CRH expression in the hypothalamus. Ketamine administration following the SPS procedure counteracted these changes by increasing the time spent in the center of the OFT, the distance traversed in the open arms of the EMPT, and mitigating SPS-induced alterations in cerebral cortex oscillations. Moreover, ketamine reduced the protein levels of GSK-3β, GR, p-GSK-3β, and altered the ratio of p-GSK-3β to GSK-3β. Gene expression of GSK-3β, GR, BDNF, and FKBP5 decreased in the SPS-Ket group compared to the SPS-Sal group. CONCLUSIONS Ketamine appeared to remediate the abnormal GSK-3β signaling pathway induced by SPS. These findings collectively suggest that ketamine could be a promising therapeutic agent for PTSD symptoms, working through the modulation of the GSK-3β signaling pathway.
Collapse
Affiliation(s)
- Xinyu Hu
- School of Psychology, Weifang Medical University, 7166# Baotong West Street, Weifang 261053, China.
| | - Lifen Liu
- School of Psychology, Weifang Medical University, 7166# Baotong West Street, Weifang 261053, China.
| | - Zixun Wang
- School of Clinical Medicine, Weifang Medical University, 7166# Baotong West Street, Weifang, Shandong, 261053, PR China.
| | - Yongjing Sun
- School of Clinical Medicine, Weifang Medical University, 7166# Baotong West Street, Weifang, Shandong, 261053, PR China.
| | - Zhi Li
- School of Clinical Medicine, Weifang Medical University, 7166# Baotong West Street, Weifang, Shandong, 261053, PR China.
| | - Guorun Zhou
- School of Psychology, Weifang Medical University, 7166# Baotong West Street, Weifang 261053, China.
| | - Kuitao Yue
- Medical Imaging Center, Affiliated Hospital of Weifang Medical University, Weifang, 261035, PR China.
| | - Lin Wang
- Clinical Competency Training Center, Medical experiment and training center, Weifang Medical University, 7166# Baotong West Street, Weifang Shandong, 261053, P. R. China.
| | - Bo Lian
- Department of Bioscience and Technology, Weifang Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, China.
| | - Guohua Lu
- School of Psychology, Weifang Medical University, 7166# Baotong West Street, Weifang 261053, China.
| | - Changjiang Li
- School of Psychology, Weifang Medical University, 7166# Baotong West Street, Weifang 261053, China.
| | - Lin Sun
- School of Psychology, Weifang Medical University, 7166# Baotong West Street, Weifang 261053, China.
| |
Collapse
|
9
|
Zhang X, An H, Chen Y, Shu N. Neurobiological Mechanisms of Cognitive Decline Correlated with Brain Aging. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1419:127-146. [PMID: 37418211 DOI: 10.1007/978-981-99-1627-6_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 07/08/2023]
Abstract
Cognitive decline has emerged as one of the greatest health threats of old age. Meanwhile, aging is the primary risk factor for Alzheimer's disease (AD) and other prevalent neurodegenerative disorders. Developing therapeutic interventions for such conditions demands a greater understanding of the processes underlying normal and pathological brain aging. Despite playing an important role in the pathogenesis and incidence of disease, brain aging has not been well understood at a molecular level. Recent advances in the biology of aging in model organisms, together with molecular- and systems-level studies of the brain, are beginning to shed light on these mechanisms and their potential roles in cognitive decline. This chapter seeks to integrate the knowledge about the neurological mechanisms of age-related cognitive changes that underlie aging.
Collapse
Affiliation(s)
- Xiaxia Zhang
- State Key Laboratory of Cognitive Neuroscience and Learning, Faculty of Psychology, Beijing Normal University, Beijing, China
- Beijing Aging Brain Rejuvenation Initiative (BABRI) Centre, Beijing Normal University, Beijing, China
| | - Haiting An
- Beijing Aging Brain Rejuvenation Initiative (BABRI) Centre, Beijing Normal University, Beijing, China
- Beijing Neurosurgical Institute, Beijing Tian Tan Hospital, Capital Medical University, Beijing, China
| | - Yuan Chen
- State Key Laboratory of Cognitive Neuroscience and Learning, Faculty of Psychology, Beijing Normal University, Beijing, China
- Beijing Aging Brain Rejuvenation Initiative (BABRI) Centre, Beijing Normal University, Beijing, China
| | - Ni Shu
- State Key Laboratory of Cognitive Neuroscience and Learning, Faculty of Psychology, Beijing Normal University, Beijing, China.
| |
Collapse
|
10
|
Mahmoodkhani M, Ghasemi M, Derafshpour L, Amini M, Mehranfard N. Developmental effects of early-life stress on dopamine D2 receptor and proteins involved in noncanonical D2 dopamine receptor signaling pathway in the prefrontal cortex of male rats. JOURNAL OF COMPLEMENTARY & INTEGRATIVE MEDICINE 2022; 19:697-703. [PMID: 33962496 DOI: 10.1515/jcim-2020-0539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 03/02/2021] [Indexed: 06/12/2023]
Abstract
OBJECTIVES Dopamine neurotransmission is implicated in multiple neuropsychiatric disorders, most strikingly in Parkinson's disease, bipolar disorder, attention-deficit hyperactivity disorder and schizophrenia. In addition to canonical pathway, D2-receptor (D2R) exerts some of its biological actions through regulating the activity of Akt and GSK3, which in turn were found to be altered in several psychiatric illnesses. The present study examined the impacts of maternal separation, an early-life stress model which has been associated with disturbed neurodevelopment and appearance of many psychiatric disorders, on developmental changes in dopamine concentration and the expression of D2Rs, Akt and GSK-3β in the medial prefrontal cortex (PFC; a key target of stress) in adolescent and young adult male rats. METHODS Maternal separation was performed 3 h per day from postnatal days 2 to 11. The PFC protein and dopamine contents were determined using western blotting analysis and Eliza, respectively. RESULTS Results indicated long-term increases in the prefrontal dopamine levels in stressed adolescent and young adult male rats, accompanied by significant downregulation of D2R as well as upregulation of p-Akt and GSK-3β contents in stressed adolescence compared to controls, with all protein levels that returned to control values in stressed adult rats. CONCLUSIONS Our findings suggest that early-life stress differentially modulates prefrontal D2R/Akt/GSK-3β levels during development. Since adolescence period is susceptible to the onset of specific mental illnesses, disruption of noncanonical components of D2R signaling during this critical period may have an important role in programming neurobehavioral phenotypes in adulthood and manipulations influencing Akt/GSK-3β pathway may improve the expression of specific dopamine-related behaviors and the effects of dopaminergic drugs.
Collapse
Affiliation(s)
- Maryam Mahmoodkhani
- Neurophysiology Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Maedeh Ghasemi
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Leila Derafshpour
- Neurophysiology Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Mohammad Amini
- School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Nasrin Mehranfard
- Neurophysiology Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
11
|
de Bartolomeis A, Vellucci L, Austin MC, De Simone G, Barone A. Rational and Translational Implications of D-Amino Acids for Treatment-Resistant Schizophrenia: From Neurobiology to the Clinics. Biomolecules 2022; 12:biom12070909. [PMID: 35883465 PMCID: PMC9312470 DOI: 10.3390/biom12070909] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 06/25/2022] [Accepted: 06/26/2022] [Indexed: 12/13/2022] Open
Abstract
Schizophrenia has been conceptualized as a neurodevelopmental disorder with synaptic alterations and aberrant cortical–subcortical connections. Antipsychotics are the mainstay of schizophrenia treatment and nearly all share the common feature of dopamine D2 receptor occupancy, whereas glutamatergic abnormalities are not targeted by the presently available therapies. D-amino acids, acting as N-methyl-D-aspartate receptor (NMDAR) modulators, have emerged in the last few years as a potential augmentation strategy in those cases of schizophrenia that do not respond well to antipsychotics, a condition defined as treatment-resistant schizophrenia (TRS), affecting almost 30–40% of patients, and characterized by serious cognitive deficits and functional impairment. In the present systematic review, we address with a direct and reverse translational perspective the efficacy of D-amino acids, including D-serine, D-aspartate, and D-alanine, in poor responders. The impact of these molecules on the synaptic architecture is also considered in the light of dendritic spine changes reported in schizophrenia and antipsychotics’ effect on postsynaptic density proteins. Moreover, we describe compounds targeting D-amino acid oxidase and D-aspartate oxidase enzymes. Finally, other drugs acting at NMDAR and proxy of D-amino acids function, such as D-cycloserine, sarcosine, and glycine, are considered in the light of the clinical burden of TRS, together with other emerging molecules.
Collapse
Affiliation(s)
- Andrea de Bartolomeis
- Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Sciences and Dentistry, University of Naples Federico II, 80131 Naples, Italy; (L.V.); (G.D.S.); (A.B.)
- Correspondence: ; Tel.: +39-081-7463673 or +39-081-7463884 or +39-3662745592; Fax: +39-081-7462644
| | - Licia Vellucci
- Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Sciences and Dentistry, University of Naples Federico II, 80131 Naples, Italy; (L.V.); (G.D.S.); (A.B.)
| | - Mark C. Austin
- Clinical Psychopharmacology Program, College of Pharmacy, Idaho State University, Pocatello, ID 83209, USA;
| | - Giuseppe De Simone
- Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Sciences and Dentistry, University of Naples Federico II, 80131 Naples, Italy; (L.V.); (G.D.S.); (A.B.)
| | - Annarita Barone
- Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Sciences and Dentistry, University of Naples Federico II, 80131 Naples, Italy; (L.V.); (G.D.S.); (A.B.)
| |
Collapse
|
12
|
Brown RW, Varnum CG, Wills LJ, Peeters LD, Gass JT. Modulation of mGlu5 improves sensorimotor gating deficits in rats neonatally treated with quinpirole through changes in dopamine D2 signaling. Pharmacol Biochem Behav 2021; 211:173292. [PMID: 34710401 PMCID: PMC9176413 DOI: 10.1016/j.pbb.2021.173292] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/19/2021] [Accepted: 10/20/2021] [Indexed: 12/19/2022]
Abstract
This study analyzed whether the positive allosteric modulator of metabotropic glutamate receptor type 5 (mGlu5) 3-Cyano-N-(1,3-diphenyl-1H-pyrazol-5-yl) benzamide (CDPPB) would alleviate deficits in prepulse inhibition (PPI) and affect dopamine (DA) D2 signaling in the dorsal striatum and prefrontal cortex (PFC) in the neonatal quinpirole (NQ) model of schizophrenia (SZ). Male and female Sprague-Dawley rats were neonatally treated with either saline (NS) or quinpirole HCL (1 mg/kg; NQ), a DAD2 receptor agonist, from postnatal days (P) 1-21. Rats were raised to P44 and behaviorally tested on PPI from P44-P48. Before each trial, rats were subcutaneous (sc) administered saline or CDPPB (10 mg/kg or 30 mg/kg). On P50, rats were given a spontaneous locomotor activity test after CDPPB or saline administration. On P51, the dorsal striatum and PFC were evaluated for both arrestin-2 (βA-2) and phospho-AKT protein levels. NQ-treated rats demonstrated a significant deficit in PPI, which was alleviated to control levels by the 30 mg/kg dose of CDPPB. There were no significant effects of CDPPB on locomotor activity. NQ treatment increased βA-2 and decreased phospho-AKT in both the dorsal striatum and PFC, consistent with an increase DAD2 signaling. The 30 mg/kg dose of CDPPB significantly reversed changes in βA-2 in the dorsal striatum and PFC and phospho-AKT in the PFC equivalent to controls. Both doses of CDPPB produced a decrease of phospho-AKT in the PFC compared to controls. This study revealed that a mGlu5 positive allosteric modulator was effective to alleviate PPI deficits and striatal DAD2 signaling in the NQ model of SZ.
Collapse
Affiliation(s)
- Russell W Brown
- Department of Biomedical Sciences James H. Quillen College of Medicine, East Tennessee State University Johnson City, TN 37614, United States of America.
| | - Christopher G Varnum
- Department of Biomedical Sciences James H. Quillen College of Medicine, East Tennessee State University Johnson City, TN 37614, United States of America
| | - Liza J Wills
- Department of Biomedical Sciences James H. Quillen College of Medicine, East Tennessee State University Johnson City, TN 37614, United States of America
| | - Loren D Peeters
- Department of Biomedical Sciences James H. Quillen College of Medicine, East Tennessee State University Johnson City, TN 37614, United States of America
| | - Justin T Gass
- Department of Biomedical Sciences James H. Quillen College of Medicine, East Tennessee State University Johnson City, TN 37614, United States of America
| |
Collapse
|
13
|
Keegan BM, Dreitzler AL, Sexton T, Beveridge TJR, Smith HR, Miller MD, Blough BE, Porrino LJ, Childers SR, Howlett AC. Chronic phenmetrazine treatment promotes D 2 dopaminergic and α2-adrenergic receptor desensitization and alters phosphorylation of signaling proteins and local cerebral glucose metabolism in the rat brain. Brain Res 2021; 1761:147387. [PMID: 33631209 PMCID: PMC8552242 DOI: 10.1016/j.brainres.2021.147387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 02/15/2021] [Accepted: 02/16/2021] [Indexed: 11/21/2022]
Abstract
Phenmetrazine (PHEN) is a putative treatment for cocaine and psychostimulant recidivism; however, neurochemical changes underlying its activity have not been fully elucidated. We sought to characterize brain homeostatic adaptations to chronic PHEN, specifically on functional brain activity (local cerebral glucose utilization), G-Protein Coupled Receptor-stimulated G-protein activation, and phosphorylation of ERK1/2Thr202/Tyr204, GSK3βTyr216, and DARPP-32Thr34. Male Sprague-Dawley rats were implanted with sub-cutaneous minipumps delivering either saline (vehicle), acute (2-day) or chronic (14-day) low dose (25 mg/kg/day) or high dose (50 mg/kg/day) PHEN. Acute administration of high dose PHEN increased local cerebral glucose utilization measured by 2-[14C]-deoxyglucose uptake in basal ganglia and motor-related regions of the rat brain. However, chronically treated animals developed tolerance to these effects. To identify the neurochemical changes associated with PHEN's activity, we performed [35S]GTPγS binding assays on unfixed and immunohistochemistry on fixed coronal brain sections. Chronic PHEN treatment dose-dependently attenuated D2 dopamine and α2-adrenergic, but not 5-HT1A, receptor-mediated G-protein activation. Two distinct patterns of effects on pERK1/2 and pDARPP-32 were observed: 1) chronic low dose PHEN decreased pERK1/2, and also significantly increased pDARPP-32 levels in some regions; 2) acute and chronic PHEN increased pERK1/2, but chronic high dose PHEN treatment tended to decrease pDARPP-32. Chronic low dose, but not high dose, PHEN significantly reduced pGSK3β levels in several regions. Our study provides definitive evidence that extended length PHEN dosage schedules elicit distinct modes of neuronal acclimatization in cellular signaling. These pharmacodynamic modifications should be considered in drug development for chronic use.
Collapse
Affiliation(s)
- Bradley M Keegan
- Center for the Neurobiology of Addiction Treatment, Department of Physiology and Pharmacology, Wake Forest School of Medicine, Medical Center Blvd., Winston-Salem, NC 27157, USA
| | - Annie L Dreitzler
- Center for the Neurobiology of Addiction Treatment, Department of Physiology and Pharmacology, Wake Forest School of Medicine, Medical Center Blvd., Winston-Salem, NC 27157, USA
| | - Tammy Sexton
- Center for the Neurobiology of Addiction Treatment, Department of Physiology and Pharmacology, Wake Forest School of Medicine, Medical Center Blvd., Winston-Salem, NC 27157, USA
| | - Thomas J R Beveridge
- Center for the Neurobiology of Addiction Treatment, Department of Physiology and Pharmacology, Wake Forest School of Medicine, Medical Center Blvd., Winston-Salem, NC 27157, USA
| | - Hilary R Smith
- Center for the Neurobiology of Addiction Treatment, Department of Physiology and Pharmacology, Wake Forest School of Medicine, Medical Center Blvd., Winston-Salem, NC 27157, USA
| | - Mack D Miller
- Center for the Neurobiology of Addiction Treatment, Department of Physiology and Pharmacology, Wake Forest School of Medicine, Medical Center Blvd., Winston-Salem, NC 27157, USA
| | - Bruce E Blough
- Center for Drug Discovery, RTI International, Research Triangle Park, NC 27709, USA
| | - Linda J Porrino
- Center for the Neurobiology of Addiction Treatment, Department of Physiology and Pharmacology, Wake Forest School of Medicine, Medical Center Blvd., Winston-Salem, NC 27157, USA
| | - Steven R Childers
- Center for the Neurobiology of Addiction Treatment, Department of Physiology and Pharmacology, Wake Forest School of Medicine, Medical Center Blvd., Winston-Salem, NC 27157, USA
| | - Allyn C Howlett
- Center for the Neurobiology of Addiction Treatment, Department of Physiology and Pharmacology, Wake Forest School of Medicine, Medical Center Blvd., Winston-Salem, NC 27157, USA.
| |
Collapse
|
14
|
Chaves Filho AJM, Cunha NL, Rodrigues PDA, de Souza AG, Soares MVR, Jucá PM, de Queiroz T, Clemente DCDS, Mottin M, Andrade CH, Peixoto CA, Macedo DS. Doxycycline reverses cognitive impairment, neuroinflammation and oxidative imbalance induced by D-amphetamine mania model in mice: A promising drug repurposing for bipolar disorder treatment? Eur Neuropsychopharmacol 2021; 42:57-74. [PMID: 33191076 DOI: 10.1016/j.euroneuro.2020.11.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 10/22/2020] [Accepted: 11/02/2020] [Indexed: 11/15/2022]
Abstract
Immune-inflammatory mechanisms are involved in the pathophysiology of bipolar disorder. Tetracyclines present neuroprotective actions based on their anti-inflammatory and microglia suppressant effects. Doxycycline (DOXY) is a tetracycline that demonstrates a better usage profile with protective actions against inflammation and CNS injury. Here, we investigated the effects of DOXY against behavioral, neuroinflammatory, and pro-oxidative changes induced by the d-amphetamine mania model. Adult mice were given d-amphetamine 2.0 mg/kg or saline for 14 days. Between days 8 and 14, received lithium, DOXY (25 or 50 mg/kg), or their combination (lithium+DOXY) on both doses. We collected the brain areas prefrontal cortex (PFC), hippocampus, and amygdala to evaluate inflammatory and oxidative alterations. D-amphetamine induced hyperlocomotion and impairment in recognition and working memory. Lithium reversed hyperlocomotion but could not restore cognitive alterations. DOXY alone (at both doses) or combined with lithium reversed d-amphetamine-induced cognitive changes. DOXY, better than lithium, reversed the d-amphetamine-induced rise in TNFα, MPO, and lipid peroxidation. DOXY reduced the hippocampal expression of Iba1 (a marker of microglial activation), inducible nitric oxide synthase (iNOS), and nitrite. Combined with lithium, DOXY increased the phosphorylated (inactivated) form of GSK3β (Ser9). Therefore, DOXY alone or combined with lithium reversed cognitive impairment and neuroinflammation induced by the mice's d-amphetamine model. This study points to DOXY as a promising adjunctive tool for bipolar disorder treatment focused on cognition and neuroimmune changes. Our data provide the first rationale for clinical trials investigating DOXY therapeutic actions in bipolar disorder mania.
Collapse
Affiliation(s)
- Adriano José Maia Chaves Filho
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil; LabMol - Laboratory for Molecular Modeling and Drug Design, Faculty of Pharmacy, Federal University of Goiás, Goiânia, Brazil.
| | - Natássia Lopes Cunha
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil.
| | - Patrícia de Araújo Rodrigues
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil.
| | - Alana Gomes de Souza
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil.
| | - Michele Verde-Ramo Soares
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil.
| | - Paloma Marinho Jucá
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil.
| | - Tatiana de Queiroz
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil.
| | - Dino César da Silva Clemente
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil.
| | - Melina Mottin
- LabMol - Laboratory for Molecular Modeling and Drug Design, Faculty of Pharmacy, Federal University of Goiás, Goiânia, Brazil.
| | - Carolina Horta Andrade
- LabMol - Laboratory for Molecular Modeling and Drug Design, Faculty of Pharmacy, Federal University of Goiás, Goiânia, Brazil.
| | - Christina Alves Peixoto
- Laboratório de Ultraestrutura, Instituto Aggeu Magalhães - FIOCRUZ, Recife, Brazil; Instituto Nacional de Ciência e Tecnologia de Neuroimunomodulação (NIM), Rio de Janeiro, Brazil..
| | - Danielle S Macedo
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil; National Institute for Translational Medicine (INCT-TM, CNPq), Ribeirão Preto, SP, Brazil..
| |
Collapse
|
15
|
Arrestin recruitment to dopamine D2 receptor mediates locomotion but not incentive motivation. Mol Psychiatry 2020; 25:2086-2100. [PMID: 30120413 PMCID: PMC6378141 DOI: 10.1038/s41380-018-0212-4] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 06/20/2018] [Accepted: 06/25/2018] [Indexed: 01/11/2023]
Abstract
The dopamine (DA) D2 receptor (D2R) is an important target for the treatment of neuropsychiatric disorders such as schizophrenia and Parkinson's disease. However, the development of improved therapeutic strategies has been hampered by our incomplete understanding of this receptor's downstream signaling processes in vivo and how these relate to the desired and undesired effects of drugs. D2R is a G protein-coupled receptor (GPCR) that activates G protein-dependent as well as non-canonical arrestin-dependent signaling pathways. Whether these effector pathways act alone or in concert to facilitate specific D2R-dependent behaviors is unclear. Here, we report on the development of a D2R mutant that recruits arrestin but is devoid of G protein activity. When expressed virally in "indirect pathway" medium spiny neurons (iMSNs) in the ventral striatum of D2R knockout mice, this mutant restored basal locomotor activity and cocaine-induced locomotor activity in a manner indistinguishable from wild-type D2R, indicating that arrestin recruitment can drive locomotion in the absence of D2R-mediated G protein signaling. In contrast, incentive motivation was enhanced only by wild-type D2R, signifying a dissociation in the mechanisms that underlie distinct D2R-dependent behaviors, and opening the door to more targeted therapeutics.
Collapse
|
16
|
Barr JL, Unterwald EM. Glycogen synthase kinase-3 signaling in cellular and behavioral responses to psychostimulant drugs. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118746. [PMID: 32454064 DOI: 10.1016/j.bbamcr.2020.118746] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 05/13/2020] [Accepted: 05/14/2020] [Indexed: 12/15/2022]
Abstract
Glycogen synthase kinase-3 (GSK-3) is a serine/threonine kinase implicated in numerous physiological processes and cellular functions through its ability to regulate the function of many proteins, including transcription factors and structural proteins. GSK-3β has been demonstrated to function as a regulator of multiple behavioral processes induced by drugs of abuse, particularly psychostimulant drugs. In this review, we provide an overview of the regulation of GSK-3β activity produced by psychostimulants, and the role of GSK-3β signaling in psychostimulant-induced behaviors including drug reward, associative learning and memory which play a role in the maintenance of drug-seeking. Evidence supports the conclusion that GSK-3β is an important component of the actions of psychostimulant drugs and that GSK-3β is a valid target for developing novel therapeutics. Additional studies are required to examine the role of GSK-3β in distinct cell types within the mesolimbic and memory circuits to further elucidate the mechanisms related to the acquisition, consolidation, and recall of drug-related memories, and potentially countering neuroadaptations that reinforce drug-seeking behaviors that maintain drug dependence.
Collapse
Affiliation(s)
- Jeffrey L Barr
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Ellen M Unterwald
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA.
| |
Collapse
|
17
|
Li YC, Panikker P, Xing B, Yang SS, Alexandropoulos C, McEachern EP, Akumuo R, Zhao E, Gulchina Y, Pletnikov MV, Urs NM, Caron MG, Elefant F, Gao WJ. Deletion of Glycogen Synthase Kinase-3β in D 2 Receptor-Positive Neurons Ameliorates Cognitive Impairment via NMDA Receptor-Dependent Synaptic Plasticity. Biol Psychiatry 2020; 87:745-755. [PMID: 31892408 PMCID: PMC7103512 DOI: 10.1016/j.biopsych.2019.10.025] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 10/28/2019] [Accepted: 10/29/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Cortical dopaminergic systems are critically involved in prefrontal cortex (PFC) functions, especially in working memory and neurodevelopmental disorders such as schizophrenia. GSK-3β (glycogen synthase kinase-3β) is highly associated with cAMP (cyclic adenosine monophosphate)-independent dopamine D2 receptor (D2R)-mediated signaling to affect dopamine-dependent behaviors. However, the mechanisms underlying the GSK-3β modulation of cognitive function via D2Rs remains unclear. METHODS This study explored how conditional cell-type-specific ablation of GSK-3β in D2R+ neurons (D2R-GSK-3β-/-) in the brain affects synaptic function in the medial PFC (mPFC). Both male and female (postnatal days 60-90) mice, including 140 D2R, 24 D1R, and 38 DISC1 mice, were used. RESULTS This study found that NMDA receptor (NMDAR) function was significantly increased in layer V pyramidal neurons in mPFC of D2R-GSK-3β-/- mice, along with increased dopamine modulation of NMDAR-mediated current. Consistently, NR2A and NR2B protein levels were elevated in mPFC of D2R-GSK-3β-/- mice. This change was accompanied by a significant increase in enrichment of activator histone mark H3K27ac at the promoters of both Grin2a and Grin2b genes. In addition, altered short- and long-term synaptic plasticity, along with an increased spine density in layer V pyramidal neurons, were detected in D2R-GSK-3β-/- mice. Indeed, D2R-GSK-3β-/- mice also exhibited a resistance of working memory impairment induced by injection of NMDAR antagonist MK-801. Notably, either inhibiting GSK-3β or disrupting the D2R-DISC1 complex was able to reverse the mutant DISC1-induced decrease of NMDAR-mediated currents in the mPFC. CONCLUSIONS This study demonstrates that GSK-3β modulates cognition via D2R-DISC1 interaction and epigenetic regulation of NMDAR expression and function.
Collapse
Affiliation(s)
- Yan-Chun Li
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania.
| | - Priyalakshmi Panikker
- Department of Biology, Drexel University College of Medicine, Philadelphia, PA 19104, USA
| | - Bo Xing
- Department of Neurobiology & Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Sha-Sha Yang
- Department of Neurobiology & Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Cassandra Alexandropoulos
- Department of Neurobiology & Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Erin P McEachern
- Department of Neurobiology & Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Rita Akumuo
- Department of Neurobiology & Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Elise Zhao
- Department of Neurobiology & Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Yelena Gulchina
- Department of Neurobiology & Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Mikhail V. Pletnikov
- Departments of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Nikhil M. Urs
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL 32610, USA
| | - Marc G. Caron
- Departments of Cell Biology, Neurobiology, and Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Felice Elefant
- Department of Biology, Drexel University College of Medicine, Philadelphia, PA 19104, USA
| | - Wen-Jun Gao
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania.
| |
Collapse
|
18
|
Lithium for schizophrenia: supporting evidence from a 12-year, nationwide health insurance database and from Akt1-deficient mouse and cellular models. Sci Rep 2020; 10:647. [PMID: 31959776 PMCID: PMC6971245 DOI: 10.1038/s41598-019-57340-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 12/19/2019] [Indexed: 12/11/2022] Open
Abstract
Accumulating evidence suggests AKT1 and DRD2-AKT-GSK3 signaling involvement in schizophrenia. AKT1 activity is also required for lithium, a GSK3 inhibitor, to modulate mood-related behaviors. Notably, GSK3 inhibitor significantly alleviates behavioral deficits in Akt1−/− female mice, whereas typical/atypical antipsychotics have no effect. In agreement with adjunctive therapy with lithium in treating schizophrenia, our data mining indicated that the average utilization rates of lithium in the Taiwan National Health Insurance Research Database from 2002 to 2013 are 10.9% and 6.63% in inpatients and outpatients with schizophrenia, respectively. Given that lithium is commonly used in clinical practice, it is of great interest to evaluate the effect of lithium on alleviating Akt1-related deficits. Taking advantage of Akt1+/− mice to mimic genetic deficiency in patients, behavioral impairments were replicated in female Akt1+/− mice but were alleviated by subchronic lithium treatment for 13 days. Lithium also effectively alleviated the observed reduction in phosphorylated GSK3α/β expression in the brains of Akt1+/− mice. Furthermore, inhibition of Akt expression using an Akt1/2 inhibitor significantly reduced neurite length in P19 cells and primary hippocampal cell cultures, which was also ameliorated by lithium. Collectively, our findings implied the therapeutic potential of lithium and the importance of the AKT1-GSK3 signaling pathway.
Collapse
|
19
|
Xia QP, Cheng ZY, He L. The modulatory role of dopamine receptors in brain neuroinflammation. Int Immunopharmacol 2019; 76:105908. [PMID: 31622861 DOI: 10.1016/j.intimp.2019.105908] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 09/03/2019] [Accepted: 09/08/2019] [Indexed: 01/11/2023]
Abstract
Neuroinflammation is a general pathological feature of central nervous system (CNS) diseases, primarily caused by activation of astrocytes and microglia, as well as the infiltration of peripheral immune cells. Inhibition of neuroinflammation is an important strategy in the treatment of brain disorders. Dopamine (DA) receptor, a significant G protein-coupled receptor (GPCR), is classified into two families: D1-like (D1 and D5) and D2-like (D2, D3 and D4) receptor families, according to their downstream signaling pathways. Traditionally, DA receptor forms a wide variety of psychological activities and motor functions, such as voluntary movement, working memory and learning. Recently, the role of DA receptor in neuroinflammation has been investigated widely, mainly focusing on nucleotide-binding oligomerization domain-like receptor pyrin domain-containing 3 (NLRP3) inflammasome, renin-angiotensin system, αB-crystallin, as well as invading peripheral immune cells, including T cells, dendritic cells, macrophages and monocytes. This review briefly outlined the functions and signaling pathways of DA receptor subtypes as well as its role in inflammation-related glial cells, and subsequently summarized the mechanisms of DA receptors affecting neuroinflammation. Meaningfully, this article provided a theoretical basis for drug development targeting DA receptors in inflammation-related brain diseases.
Collapse
Affiliation(s)
- Qing-Peng Xia
- Department of Pharmacology, China Pharmaceutical University, Nanjing 210009, China
| | - Zhao-Yan Cheng
- Department of Pharmacology, China Pharmaceutical University, Nanjing 210009, China
| | - Ling He
- Department of Pharmacology, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
20
|
Zhou Y, Zhang L, Fu X, Jiang Z, Tong R, Shi J, Li J, Zhong L. Design, Synthesis and in Vitro Tumor Cytotoxicity Evaluation of 3,5-Diamino-N-substituted Benzamide Derivatives as Novel GSK-3β Small Molecule Inhibitors. Chem Biodivers 2019; 16:e1900304. [PMID: 31338947 DOI: 10.1002/cbdv.201900304] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Accepted: 07/23/2019] [Indexed: 02/05/2023]
Abstract
Glycogen synthase kinase-3 (GSK-3) plays an important regulatory role in various signaling pathways; such as PI3 K/AKT, which is closely related to the occurrence and development of tumors. At present, the most reported active GSK-3 inhibitors have the same structure: lactam ring or amide structure. To find out the GSK-3β small molecule inhibitor with novel, safe, efficient and more uncomplicated synthesis method, we analyzed in-depth reported crystal-binding patterns of GSK-3β small molecule inhibitor with GSK-3β protein, and designed and synthesized 17 non-reported 3,5-diamino-N-substituted benzamide compounds. Their structures were confirmed by 1 H-NMR, 13 C-NMR, and HR-MS. The preliminary screening of tumor cytotoxicity of compounds in vitro was detected by MTT, and their structure-activity relationships were illustrated. The results have shown that 3,5-diamino-N-[3-(trifluoromethyl)phenyl]benzamide (4d) exhibited significant tumor cytotoxicity against human colon cancer cells (HCT-116) with IC50 of 8.3 μm and showed commendable selectivity to GSK-3β. In addition, Compound 4d induced apoptosis to some extent and possessed modest PK properties.
Collapse
Affiliation(s)
- Yanping Zhou
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, No. 32 West Second Section First Ring Road, Chengdu, 610072, P. R. China
| | - Lijuan Zhang
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, No. 32 West Second Section First Ring Road, Chengdu, 610072, P. R. China
| | - Xiujuan Fu
- School of Pharmacy, Southwest Medicinal University, No. 319 Section 3, Zhongshan Road, Luzhou, 646000, P. R. China
| | - Zhongliang Jiang
- Department of Hematology, Miller School of Medicine, University of Miami, Miami, USA
| | - Rongsheng Tong
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, No. 32 West Second Section First Ring Road, Chengdu, 610072, P. R. China
| | - Jianyou Shi
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, No. 32 West Second Section First Ring Road, Chengdu, 610072, P. R. China
| | - Jian Li
- Department of Pharmacy, West China Hospital Sichuan University, Chengdu, 610041, P. R. China
| | - Lei Zhong
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, No. 32 West Second Section First Ring Road, Chengdu, 610072, P. R. China
| |
Collapse
|
21
|
Tremblay M, Barrus MM, Cocker PJ, Baunez C, Winstanley CA. Increased motor impulsivity in a rat gambling task during chronic ropinirole treatment: potentiation by win-paired audiovisual cues. Psychopharmacology (Berl) 2019; 236:1901-1915. [PMID: 30706098 DOI: 10.1007/s00213-019-5173-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 01/16/2019] [Indexed: 02/04/2023]
Abstract
RATIONALE Chronic administration of D2/3 receptor agonists ropinirole or pramipexole can increase the choice of uncertain rewards in rats, theoretically approximating iatrogenic gambling disorder (iGD). OBJECTIVES We aimed to assess the effect of chronic ropinirole in animal models that attempt to capture critical aspects of commercial gambling, including the risk of losing rather than failing to gain, and the use of win-paired sensory stimuli heavily featured in electronic gambling machines (EGMs). METHODS Male Long-Evans rats learned the rat gambling task (rGT; n = 24), in which animals sample between four options that differ in the magnitude and probability of rewards and time-out punishments. In the cued rGT (n = 40), reward-concurrent audiovisual cues were added that scaled in complexity with win size. Rats were then implanted with an osmotic pump delivering ropinirole (5 mg/kg/day) or saline for 28 days. RESULTS Chronic ropinirole did not unequivocally increase preference for more uncertain outcomes in either the cued or uncued rGT. Ropinirole transiently increased premature responses, a measure of motor impulsivity, and this change was larger and more long-lasting in the cued task. CONCLUSIONS These data suggest that explicitly signaling loss prevents the increase in preference for uncertain rewards caused by D2/3 receptor agonists observed previously. The ability of win-paired cues to amplify ropinirole-induced increases in motor impulsivity may explain why compulsive use of EGMs is particularly common in iGD. These data offer valuable insight into the cognitive-behavioral mechanisms through which chronic dopamine agonist treatments may induce iGD and related impulse control disorders.
Collapse
Affiliation(s)
- Melanie Tremblay
- Department of Psychology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Westbrook Mall, Vancouver, BC, V6T 1Z3, Canada. .,Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada.
| | - Michael M Barrus
- Department of Psychology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Westbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Paul J Cocker
- Department of Psychology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Westbrook Mall, Vancouver, BC, V6T 1Z3, Canada.,Department of Experimental Psychology, University of Cambridge, Cambridge, UK
| | - Christelle Baunez
- Institut de Neurosciences de la Timone (INT), UMR7289, Centre National de la Recherche Scientifique (CNRS) ∓ Aix-Marseille Université (AMU), Marseille, France
| | - Catharine A Winstanley
- Department of Psychology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Westbrook Mall, Vancouver, BC, V6T 1Z3, Canada.
| |
Collapse
|
22
|
Wang X, Luo C, Mao XY, Li X, Yin JY, Zhang W, Zhou HH, Liu ZQ. Metformin reverses the schizophrenia-like behaviors induced by MK-801 in rats. Brain Res 2019; 1719:30-39. [PMID: 31121159 DOI: 10.1016/j.brainres.2019.05.023] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 05/16/2019] [Accepted: 05/19/2019] [Indexed: 12/11/2022]
Abstract
Schizophrenia is known to be a complex and disabling psychiatric disorder. Dopamine receptor antagonists have a significant therapeutic effect in improving the positive symptoms that are associated with the illness. Therefore, dopamine receptor antagonists are commonly used in the treatment of schizophrenia; however, they do not achieve satisfactory results in improving negative symptoms and cognitive impairment. Metformin, widely known as an antidiabetic drug, has been found to enhance spatial memory formation and improve anxiety-like behaviors in rodents. Metformin's neuroprotective effect has been well documented in several neurological disorders including Alzheimer's disease, Parkinson's disease, strokes, Huntington's disease, and seizures. In the present study, we used a rat model to explore the effect of metformin on schizophrenia-like behaviors induced by MK-801 (dizocilpine), an N-methyl-D-aspartate (NMDA) receptor antagonist. We found that the pre-pulse inhibition (PPI) deficit caused by MK-801 could be alleviated by metformin. The hyperlocomotion in the open field test induced by chronic treatment of MK-801 was reversed by administration of metformin. Metformin has no effect on the baseline level of anxiety in normal naive rats, while metformin could relieve the anxiety-like behaviors in MK-801-treatment rats, though this effect is not reaching a significant level. Additionally, metformin could significantly ameliorate working memory impairments induced by MK-801. Moreover, the increased level of phosphorylation of Akt and GSK3β in the frontal cortex induced by MK-801 was normalized by metformin. In conclusion, our results demonstrate that metformin improved schizophrenia-like symptoms in rats, and is therefore a potential agent for the treatment of schizophrenia.
Collapse
Affiliation(s)
- Xu Wang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, PR China
| | - Chao Luo
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, PR China; School of Life Sciences, Central South University, Changsha, Hunan 410078, PR China
| | - Xiao-Yuan Mao
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, PR China
| | - Xi Li
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, PR China
| | - Ji-Ye Yin
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, PR China
| | - Wei Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, PR China
| | - Hong-Hao Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, PR China
| | - Zhao-Qian Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, PR China.
| |
Collapse
|
23
|
Cocker PJ, Lin MY, Tremblay M, Kaur S, Winstanley CA. The β-adrenoceptor blocker propranolol ameliorates compulsive-like gambling behaviour in a rodent slot machine task: implications for iatrogenic gambling disorder. Eur J Neurosci 2018; 50:2401-2414. [PMID: 30019362 DOI: 10.1111/ejn.14070] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 06/25/2018] [Accepted: 07/06/2018] [Indexed: 01/22/2023]
Abstract
Previous work has shown that chronic administration of the dopamine D2/3 receptor agonist ropinirole invigorates performance on a rodent slot machine task (rSMT). This behavioural change appears superficially similar to the iatrogenic gambling disorder (GD) observed in a sub-set of patients with Parkinson's disease (PD), and has been associated with increased activation of the intra-cellular signalling proteins GSK3β and CREB in the striatum. Here, we wanted to determine whether this response to ropinirole could be attenuated by targeting these signalling proteins, and if the loss of dopaminergic innervation characteristic of PD would alter ropinirole's effects on the rSMT. Male Long Evans rats were trained on the rSMT. Dopaminergic terminals innervating the dorsolateral striatum were then lesioned bilaterally using the neurotoxin 6-hydroxydopamine hydrochloride (6-OHDA). Subsequently animals were implanted with osmotic mini-pumps delivering ropinirole. Lastly, animals were given dietary lithium (Li+ ), to inhibit the activation of GSK3β, or injections of the ß-adrenoceptor antagonist propranolol, which potently inhibits CREB as a secondary mechanism of action, and any changes in ropinirole-induced increases in compulsive-like engagement in the rSMT evaluated. Chronic ropinirole increased the number of trials animals completed, reproducing our original finding. This increase in task engagement was not altered in animals with 6-OHDA lesions, a putative model of early PD. In addition, the effects of ropinirole were not attenuated by administration of Li+ , but were ameliorated by propranolol. These data suggest that propranolol may represent a potential pharmacotherapy for the treatment of iatrogenic gambling.
Collapse
Affiliation(s)
- P J Cocker
- Department of Psychology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - M Y Lin
- Department of Psychology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - M Tremblay
- Department of Psychology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - S Kaur
- Department of Psychology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - C A Winstanley
- Department of Psychology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
24
|
Scala F, Nenov MN, Crofton EJ, Singh AK, Folorunso O, Zhang Y, Chesson BC, Wildburger NC, James TF, Alshammari MA, Alshammari TK, Elfrink H, Grassi C, Kasper JM, Smith AE, Hommel JD, Lichti CF, Rudra JS, D'Ascenzo M, Green TA, Laezza F. Environmental Enrichment and Social Isolation Mediate Neuroplasticity of Medium Spiny Neurons through the GSK3 Pathway. Cell Rep 2018; 23:555-567. [PMID: 29642012 PMCID: PMC6150488 DOI: 10.1016/j.celrep.2018.03.062] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 02/05/2018] [Accepted: 03/14/2018] [Indexed: 11/29/2022] Open
Abstract
Resilience and vulnerability to neuropsychiatric disorders are linked to molecular changes underlying excitability that are still poorly understood. Here, we identify glycogen-synthase kinase 3β (GSK3β) and voltage-gated Na+ channel Nav1.6 as regulators of neuroplasticity induced by environmentally enriched (EC) or isolated (IC) conditions-models for resilience and vulnerability. Transcriptomic studies in the nucleus accumbens from EC and IC rats predicted low levels of GSK3β and SCN8A mRNA as a protective phenotype associated with reduced excitability in medium spiny neurons (MSNs). In vivo genetic manipulations demonstrate that GSK3β and Nav1.6 are molecular determinants of MSN excitability and that silencing of GSK3β prevents maladaptive plasticity of IC MSNs. In vitro studies reveal direct interaction of GSK3β with Nav1.6 and phosphorylation at Nav1.6T1936 by GSK3β. A GSK3β-Nav1.6T1936 competing peptide reduces MSNs excitability in IC, but not EC rats. These results identify GSK3β regulation of Nav1.6 as a biosignature of MSNs maladaptive plasticity.
Collapse
Affiliation(s)
- Federico Scala
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX 77550, USA; Biophysics Graduate Program, Institute of Human Physiology, Università Cattolica, Rome, Italy
| | - Miroslav N Nenov
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Elizabeth J Crofton
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX 77550, USA; Neuroscience Graduate Program, The University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Aditya K Singh
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Oluwarotimi Folorunso
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Yafang Zhang
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX 77550, USA; Pharmacology and Toxicology Graduate Program, The University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Brent C Chesson
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX 77550, USA; Pharmacology and Toxicology Graduate Program, The University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Norelle C Wildburger
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Thomas F James
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX 77550, USA; Neuroscience Graduate Program, The University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Musaad A Alshammari
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX 77550, USA; Pharmacology and Toxicology Graduate Program, The University of Texas Medical Branch, Galveston, TX 77550, USA; Studies Abroad Program, King Saud University, Riyadh, Saudi Arabia
| | - Tahani K Alshammari
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX 77550, USA; Pharmacology and Toxicology Graduate Program, The University of Texas Medical Branch, Galveston, TX 77550, USA; Studies Abroad Program, King Saud University, Riyadh, Saudi Arabia
| | - Hannah Elfrink
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX 77550, USA; Bench Tutorials Program: Scientific Research and Design, The University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Claudio Grassi
- Institute of Human Physiology, Università Cattolica, Rome, Italy; Fondazione Policlinico Universitario A. Gemelli, Rome, Italy
| | - James M Kasper
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX 77550, USA; Center for Addiction Research, The University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Ashley E Smith
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX 77550, USA; Center for Addiction Research, The University of Texas Medical Branch, Galveston, TX 77550, USA; Cell Biology Graduate Program, The University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Jonathan D Hommel
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX 77550, USA; Center for Addiction Research, The University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Cheryl F Lichti
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX 77550, USA; Mitchell Center for Neurodegenerative Diseases, The University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Jai S Rudra
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX 77550, USA
| | | | - Thomas A Green
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX 77550, USA; Center for Addiction Research, The University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Fernanda Laezza
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch, Galveston, TX 77550, USA; Mitchell Center for Neurodegenerative Diseases, The University of Texas Medical Branch, Galveston, TX 77550, USA; Center for Addiction Research, The University of Texas Medical Branch, Galveston, TX 77550, USA.
| |
Collapse
|
25
|
Monaco SA, Ferguson BR, Gao WJ. Lithium Inhibits GSK3β and Augments GluN2A Receptor Expression in the Prefrontal Cortex. Front Cell Neurosci 2018; 12:16. [PMID: 29449801 PMCID: PMC5799274 DOI: 10.3389/fncel.2018.00016] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2017] [Accepted: 01/11/2018] [Indexed: 12/12/2022] Open
Abstract
Glycogen synthase kinase 3β (GSK3β) is a highly conserved serine/threonine kinase that has been implicated in both psychiatric and neurodegenerative diseases including schizophrenia, bipolar disorder, and Alzheimer's disease; therefore regulating its activity has become an important strategy for treatment of cognitive impairments in these disorders. This study examines the effects of lithium on GSK3β and its interaction with β-catenin and NMDA receptors within the prefrontal cortex. Lithium, a clinically relevant drug commonly prescribed as a mood stabilizer for psychiatric disorders, significantly increased levels of phosphorylated GSK3β serine 9, an inhibitory phosphorylation site, and decreased β-catenin ser33/37/thr41 phosphorylation in vitro, indicating GSK3β inhibition and reduced β-catenin degradation. GluN2A subunit levels were concurrently increased following lithium treatment. Similar alterations were also demonstrated in vivo; lithium administration increased GSK3β serine 9 phosphorylation and GluN2A levels, suggesting a reduced GSK3β activity and augmented GluN2A expression. Correspondingly, we observed that the amplitudes of evoked GluN2A-mediated excitatory postsynaptic currents in mPFC pyramidal neurons were significantly increased following lithium administration. Our data suggest that GSK3β activity negatively regulates GluN2A expression, likely by mediating upstream β-catenin phosphorylation, in prefrontal cortical neurons. Furthermore, our biochemical and electrophysiological experiments demonstrate that lithium mediates a specific increase in GluN2A subunit expression, ultimately augmenting GluN2A-mediated currents in the prefrontal cortex.
Collapse
Affiliation(s)
| | | | - Wen-Jun Gao
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, United States
| |
Collapse
|
26
|
Tuplin EW, Holahan MR. Aripiprazole, A Drug that Displays Partial Agonism and Functional Selectivity. Curr Neuropharmacol 2017; 15:1192-1207. [PMID: 28412910 PMCID: PMC5725548 DOI: 10.2174/1570159x15666170413115754] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 03/06/2017] [Accepted: 04/07/2017] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The treatment of schizophrenia is challenging due to the wide range of symptoms (positive, negative, cognitive) associated with the disease. Typical antipsychotics that antagonize D2 receptors are effective in treating positive symptoms, but extrapyramidal side-effects (EPS) are a common occurrence. Atypical antipsychotics targeting 5-HT2A and D2 receptors are more effective at treating cognitive and negative symptoms compared to typical antipsychotics, but these drugs also result in side-effects such as metabolic syndromes. OBJECTIVE To identify evidence in the literature that elucidates the pharmacological profile of aripiprazole.s. METHODS We searched PubMed for peer reviewed articles on aripiprazole and its clinical efficacy, side-effects, pharmacology, and effects in animal models of schizophrenia symptoms. RESULTS Aripiprazole is a newer atypical antipsychotic that displays a unique pharmacological profile, including partial D2 agonism and functionally selective properties. Aripiprazole is effective at treating the positive symptoms of schizophrenia and has the potential to treat negative and cognitive symptoms at least as well as other atypical antipsychotics. The drug has a favorable side-effect profile and has a low propensity to result in EPS or metabolic syndromes. Animal models of schizophrenia have been used to determine the efficacy of aripiprazole in symptom management. In these instances, aripiprazole resulted in the reversal of deficits in extinction, pre-pulse inhibition, and social withdrawal. Because aripiprazole requires a greater than 90% occupancy rate at D2 receptors to be clinically active and does not produce EPS, this suggests a functionally selective effect on intracellular signaling pathways. CONCLUSION A combination of factors such as dopamine system stabilization via partial agonism, functional selectivity at D2 receptors, and serotonin-dopamine system interaction may contribute to the ability of aripiprazole to successfully manage schizophrenia symptoms. This review examines these mechanisms of action to further clarify the pharmacological actions of aripiprazole.
Collapse
Affiliation(s)
- Erin W. Tuplin
- Department of Neuroscience, Faculty of Science, Carleton University, 3414 Herzberg, 1125 Colonel By Drive, Ottawa, K1S 5B6, ON Canada
| | - Matthew R. Holahan
- Department of Neuroscience, Faculty of Science, Carleton University, 3414 Herzberg, 1125 Colonel By Drive, Ottawa, K1S 5B6, ON Canada
| |
Collapse
|
27
|
Inhibition of glycogen synthase kinase-3 by SB 216763 affects acquisition at lower doses than expression of amphetamine-conditioned place preference in rats. Behav Pharmacol 2017; 28:262-271. [DOI: 10.1097/fbp.0000000000000283] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
28
|
Cocker PJ, Tremblay M, Kaur S, Winstanley CA. Chronic administration of the dopamine D 2/3 agonist ropinirole invigorates performance of a rodent slot machine task, potentially indicative of less distractible or compulsive-like gambling behaviour. Psychopharmacology (Berl) 2017; 234:137-153. [PMID: 27714426 DOI: 10.1007/s00213-016-4447-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 09/19/2016] [Indexed: 01/11/2023]
Abstract
RATIONALE Whilst dopamine agonist therapies can successfully manage the symptoms of diseases such as Parkinson's disease (PD), fibromyalgia and restless leg syndrome, they can also cause impulse control and addiction disorders such as gambling disorder (GD). These compulsive behaviours seriously undermine the utility of such treatments. OBJECTIVES The objective of the study was to model this phenomenon using a rodent slot machine task (rSMT) in order to investigate the neurobiological basis underlying such behavioural changes. METHODS Male Long Evans rats were trained to perform the rSMT. The D2-like agonist ropinirole, or saline, was then delivered continuously for 28 days via osmotic mini-pump. The effects of ropinirole on baseline rSMT performance, as well as extinction and reinstatement sessions, were determined during this time. Brain samples from key frontostriatal regions implicated in GD and PD were then harvested immediately or after a 4-week washout period during which behaviour returned to pre-drug baseline. RESULTS Ropinirole invigorated task performance, in that drug treatment resulted in a robust and sustained increase in the number of trials completed. Ex vivo analyses revealed that chronic ropinirole treatment led to a pattern of changes indicative of upregulation within the β-arrestin-AKT-GSK3β intracellular cascade, recently theorised to dominate D2-mediated signalling under hyperdopaminergic conditions, in the dorsal striatum, rather than the canonical PKA-dependent signalling pathway associated with D2 receptor activation. CONCLUSIONS Such findings provide novel insight into the role of dopamine signalling in mediating compulsive-like gambling behaviour and may inform more directed pharmacotherapies for the treatment of both idiopathic and iatrogenic GD.
Collapse
Affiliation(s)
- Paul J Cocker
- Department of Psychology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada.
| | - M Tremblay
- Department of Psychology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| | - S Kaur
- Department of Psychology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Catharine A Winstanley
- Department of Psychology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada.
| |
Collapse
|
29
|
Wang JR, Sun PH, Ren ZX, Meltzer HY, Zhen XC. GSK-3β Interacts with Dopamine D1 Receptor to Regulate Receptor Function: Implication for Prefrontal Cortical D1 Receptor Dysfunction in Schizophrenia. CNS Neurosci Ther 2016; 23:174-187. [PMID: 27996211 DOI: 10.1111/cns.12664] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 11/09/2016] [Accepted: 11/12/2016] [Indexed: 01/11/2023] Open
Abstract
INTRODUCTION Impaired dopamine D1 receptor (D1R) function in prefrontal cortex (PFC) is believed to contribute to the PFC hypofunction that has been hypothesized to be associated with negative symptoms and cognitive deficits in schizophrenia. It is therefore critical to understand the mechanisms for modulation of D1R function. AIMS To investigate the physical interaction and functional modulation between D1R and GSK-3β. RESULTS D1R and GSK-3β physically interact in cultured cells and native brain tissues. This direct interaction was found to occur at the S(417)PALS(421) motif in the C-terminus of D1R. Inhibition of GSK-3β impaired D1R activation along with a decrease in D1R-GSK-3β interaction. GSK-3β inhibition reduced agonist-stimulated D1R desensitization and endocytosis, the latter associated with the reduction of membrane translocation of β-arrestin-2. Similarly, inhibition of GSK-3β in rat PFC also resulted in impaired D1R activation and association with GSK-3β. Moreover, in a NMDA antagonist animal model of schizophrenia, we detected a decrease in prefrontal GSK-3β activity and D1R-GSK-3β association and decreased D1R activation in the PFC. CONCLUSIONS The present work identified GSK-3β as a new interacting protein for D1R functional regulation and revealed a novel mechanism for GSK-3β-regulated D1R function which may underlie D1R dysfunction in schizophrenia.
Collapse
Affiliation(s)
- Jing-Ru Wang
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Soochow University, Suzhou, Jiangsu, China
| | - Pei-Hua Sun
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Soochow University, Suzhou, Jiangsu, China
| | - Zhao-Xiang Ren
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Soochow University, Suzhou, Jiangsu, China
| | - Herbert Y Meltzer
- Department of Psychiatry and Behavioral Sciences, Northwestern Feinberg School of Medicine, Chicago, IL, USA
| | - Xue-Chu Zhen
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Soochow University, Suzhou, Jiangsu, China.,College of Pharmaceutical Sciences and the Collaborative Innovation Center for Brain Science, Soochow University, Suzhou, China
| |
Collapse
|
30
|
McCubrey JA, Rakus D, Gizak A, Steelman LS, Abrams SL, Lertpiriyapong K, Fitzgerald TL, Yang LV, Montalto G, Cervello M, Libra M, Nicoletti F, Scalisi A, Torino F, Fenga C, Neri LM, Marmiroli S, Cocco L, Martelli AM. Effects of mutations in Wnt/β-catenin, hedgehog, Notch and PI3K pathways on GSK-3 activity-Diverse effects on cell growth, metabolism and cancer. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:2942-2976. [PMID: 27612668 DOI: 10.1016/j.bbamcr.2016.09.004] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 08/14/2016] [Accepted: 09/02/2016] [Indexed: 02/07/2023]
Abstract
Glycogen synthase kinase-3 (GSK-3) is a serine/threonine kinase that participates in an array of critical cellular processes. GSK-3 was first characterized as an enzyme that phosphorylated and inactivated glycogen synthase. However, subsequent studies have revealed that this moon-lighting protein is involved in numerous signaling pathways that regulate not only metabolism but also have roles in: apoptosis, cell cycle progression, cell renewal, differentiation, embryogenesis, migration, regulation of gene transcription, stem cell biology and survival. In this review, we will discuss the roles that GSK-3 plays in various diseases as well as how this pivotal kinase interacts with multiple signaling pathways such as: PI3K/PTEN/Akt/mTOR, Ras/Raf/MEK/ERK, Wnt/beta-catenin, hedgehog, Notch and TP53. Mutations that occur in these and other pathways can alter the effects that natural GSK-3 activity has on regulating these signaling circuits that can lead to cancer as well as other diseases. The novel roles that microRNAs play in regulation of the effects of GSK-3 will also be evaluated. Targeting GSK-3 and these other pathways may improve therapy and overcome therapeutic resistance.
Collapse
Affiliation(s)
- James A McCubrey
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University Greenville, NC 27858, USA.
| | - Dariusz Rakus
- Department of Animal Molecular Physiology, Institute of Experimental Biology, Wroclaw University, Wroclaw, Poland
| | - Agnieszka Gizak
- Department of Animal Molecular Physiology, Institute of Experimental Biology, Wroclaw University, Wroclaw, Poland
| | - Linda S Steelman
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University Greenville, NC 27858, USA
| | - Steve L Abrams
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University Greenville, NC 27858, USA
| | - Kvin Lertpiriyapong
- Department of Comparative Medicine, Brody School of Medicine at East Carolina University, USA
| | - Timothy L Fitzgerald
- Department of Surgery, Brody School of Medicine at East Carolina University, USA
| | - Li V Yang
- Department of Internal Medicine, Hematology/Oncology Section, Brody School of Medicine at East Carolina University, USA
| | - Giuseppe Montalto
- Biomedical Department of Internal Medicine and Specialties, University of Palermo, Palermo, Italy; Consiglio Nazionale delle Ricerche, Istituto di Biomedicina e Immunologia Molecolare "Alberto Monroy", Palermo, Italy
| | - Melchiorre Cervello
- Consiglio Nazionale delle Ricerche, Istituto di Biomedicina e Immunologia Molecolare "Alberto Monroy", Palermo, Italy
| | - Massimo Libra
- Department of Bio-medical Sciences, University of Catania, Catania, Italy
| | | | - Aurora Scalisi
- Unit of Oncologic Diseases, ASP-Catania, Catania 95100, Italy
| | - Francesco Torino
- Department of Systems Medicine, Chair of Medical Oncology, Tor Vergata University of Rome, Rome, Italy
| | - Concettina Fenga
- Department of Biomedical, Odontoiatric, Morphological and Functional Images, Occupational Medicine Section - Policlinico "G. Martino" - University of Messina, Messina 98125, Italy
| | - Luca M Neri
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Sandra Marmiroli
- Department of Surgery, Medicine, Dentistry and Morphology, University of Modena and Reggio Emilia, Modena, Italy
| | - Lucio Cocco
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, Bologna, Italy
| | - Alberto M Martelli
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, Bologna, Italy
| |
Collapse
|
31
|
Tremblay M, Silveira MM, Kaur S, Hosking JG, Adams WK, Baunez C, Winstanley CA. Chronic D 2/3 agonist ropinirole treatment increases preference for uncertainty in rats regardless of baseline choice patterns. Eur J Neurosci 2016; 45:159-166. [PMID: 27422144 DOI: 10.1111/ejn.13332] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Revised: 06/08/2016] [Accepted: 07/03/2016] [Indexed: 12/17/2022]
Abstract
D2/3 receptor agonists are effective treatments for Parkinson's disease (PD), but can precipitate impulse control disorders (ICDs) including gambling disorder (GD). The neurobiological mechanisms underlying this devastating side-effect of dopamine agonist replacement therapy (DRT), and any dependence on the dopamine depletion caused by PD, are unclear. It is also unclear whether previous biases towards risk or uncertainty are a risk factor for developing these ICDs. We investigated whether chronic D2/3 agonist administration (5 mg/kg/day ropinirole for 28 days) altered performance of a rat model of gambling-like behaviour, the rodent betting task (rBT), and examined if baseline behaviour predicted this behavioural change. The rBT captures individual differences in subjective preference for uncertain outcomes: animals choose between guaranteed or probabilistic reinforcement of equal expected value. Chronic ropinirole dramatically increased selection of the uncertain option in two-thirds of animals, regardless of baseline preferences. The effect on choice in the rBT was replicated in a dorsolateral striatal 6-hydroxydopamine (6-OHDA) rat model of early PD. These studies are the first to look at individual differences in response to chronic, rather than pulsatile, dosing of DRT in a rodent model of gambling behaviour. These findings suggest that DRT-induced PG may stem from increases in subjective valuation of uncertainty. Such symptoms likely arise because of changes in dopaminergic striatal signalling caused by DRT rather than from an interaction between pre-morbid behaviours or PD itself.
Collapse
Affiliation(s)
- Melanie Tremblay
- Department of Psychology, University of British Columbia, 2136 West Mall, Vancouver, BC, V6T 1Z4, Canada
| | - Mason M Silveira
- Department of Psychology, University of British Columbia, 2136 West Mall, Vancouver, BC, V6T 1Z4, Canada
| | - Sukhbir Kaur
- Department of Psychology, University of British Columbia, 2136 West Mall, Vancouver, BC, V6T 1Z4, Canada
| | - Jay G Hosking
- Department of Psychology, University of British Columbia, 2136 West Mall, Vancouver, BC, V6T 1Z4, Canada
| | - Wendy K Adams
- Department of Psychology, University of British Columbia, 2136 West Mall, Vancouver, BC, V6T 1Z4, Canada
| | - Christelle Baunez
- Institut de Neurosciences de la Timone (INT), UMR7289, Centre National de la Recherche Scientifique (CNRS) & Aix-Marseille Université (AMU), Marseille, France
| | - Catharine A Winstanley
- Department of Psychology, University of British Columbia, 2136 West Mall, Vancouver, BC, V6T 1Z4, Canada
| |
Collapse
|
32
|
Zuo D, Lin L, Liu Y, Wang C, Xu J, Sun F, Li L, Li Z, Wu Y. Baicalin Attenuates Ketamine-Induced Neurotoxicity in the Developing Rats: Involvement of PI3K/Akt and CREB/BDNF/Bcl-2 Pathways. Neurotox Res 2016; 30:159-72. [PMID: 26932180 DOI: 10.1007/s12640-016-9611-y] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 12/29/2015] [Accepted: 02/19/2016] [Indexed: 12/22/2022]
Abstract
Ketamine is widely used as an anesthetic in pediatric clinical practice. However, numerous studies have reported that exposure to ketamine during the developmental period induces neurotoxicity. Here we investigate the neuroprotective effects of baicalin, a natural flavonoid compound, against ketamine-induced apoptotic neurotoxicity in the cortex and hippocampus of the Sprague-Dawley postnatal day 7 (PND7) rat pups. Our results revealed that five continuous injections of ketamine (20 mg/kg) at 90-min intervals over 6 h induced obvious morphological damages of neuron by Nissl staining and apoptosis by TUNEL assays in the prefrontal cortex and hippocampus of PND7 rat pups. Baicalin (100 mg/kg) pretreatment alleviated ketamine-induced morphological change and apoptosis. Caspase-3 activity and caspase-3 mRNA expression increase induced by ketamine were also inhibited by baicalin treatment. LY294002, an inhibitor of PI3K, abrogated the effect of baicalin against ketamine-induced caspase-3 activity and caspase-3 mRNA expression increase. In addition, Western blot studies indicated that baicalin not only inhibited ketamine-induced p-Akt and p-GSK-3β decrease, but also relieved ketamine-induced p-CREB and BDNF expression decrease. Baicalin also attenuated ketamine-induced Bcl-2/Bax decrease and caspase-3 expression increase. Further in vitro experiments proved that baicalin mitigated ketamine-induced cell viability decrease in the MTT assay, morphological change by Rosenfeld's staining, and caspase-3 expression increase by Western blot in the primary neuron-glia mixed cultures. LY294002 abrogated the protective effect of baicalin. These data demonstrate that baicalin exerts neuroprotective effect against ketamine-induced neuronal apoptosis by activating the PI3K/Akt and its downstream CREB/BDNF/Bcl-2 signaling pathways. Therefore, baicalin appears to be a promising agent in preventing or reversing ketamine's apoptotic neurotoxicity at an early developmental stage.
Collapse
Affiliation(s)
- Daiying Zuo
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, People's Republic of China
| | - Li Lin
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, People's Republic of China
| | - Yumiao Liu
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, People's Republic of China
| | - Chengna Wang
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, People's Republic of China
| | - Jingwen Xu
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, People's Republic of China
| | - Feng Sun
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, People's Republic of China
| | - Lin Li
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, People's Republic of China
| | - Zengqiang Li
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, People's Republic of China
| | - Yingliang Wu
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, People's Republic of China.
| |
Collapse
|
33
|
Kalkman HO, Feuerbach D. Modulatory effects of α7 nAChRs on the immune system and its relevance for CNS disorders. Cell Mol Life Sci 2016; 73:2511-30. [PMID: 26979166 PMCID: PMC4894934 DOI: 10.1007/s00018-016-2175-4] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 02/25/2016] [Accepted: 03/01/2016] [Indexed: 02/08/2023]
Abstract
The clinical development of selective alpha-7 nicotinic acetylcholine receptor (α7 nAChR) agonists has hitherto been focused on disorders characterized by cognitive deficits (e.g., Alzheimer's disease, schizophrenia). However, α7 nAChRs are also widely expressed by cells of the immune system and by cells with a secondary role in pathogen defense. Activation of α7 nAChRs leads to an anti-inflammatory effect. Since sterile inflammation is a frequently observed phenomenon in both psychiatric disorders (e.g., schizophrenia, melancholic and bipolar depression) and neurological disorders (e.g., Alzheimer's disease, Parkinson's disease, and multiple sclerosis), α7 nAChR agonists might show beneficial effects in these central nervous system disorders. In the current review, we summarize information on receptor expression, the intracellular signaling pathways they modulate and reasons for receptor dysfunction. Information from tobacco smoking, vagus nerve stimulation, and cholinesterase inhibition is used to evaluate the therapeutic potential of selective α7 nAChR agonists in these inflammation-related disorders.
Collapse
Affiliation(s)
- Hans O Kalkman
- Neuroscience Research, NIBR, Fabrikstrasse 22-3.001.02, 4002, Basel, Switzerland.
- , Gänsbühlgartenweg 7, 4132, Muttenz, Switzerland.
| | - Dominik Feuerbach
- Neuroscience Research, NIBR, Fabrikstrasse 22-3.001.02, 4002, Basel, Switzerland
| |
Collapse
|
34
|
Pei Y, Asif-Malik A, Canales JJ. Trace Amines and the Trace Amine-Associated Receptor 1: Pharmacology, Neurochemistry, and Clinical Implications. Front Neurosci 2016; 10:148. [PMID: 27092049 PMCID: PMC4820462 DOI: 10.3389/fnins.2016.00148] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Accepted: 03/21/2016] [Indexed: 01/30/2023] Open
Abstract
Biogenic amines are a collection of endogenous molecules that play pivotal roles as neurotransmitters and hormones. In addition to the "classical" biogenic amines resulting from decarboxylation of aromatic acids, including dopamine (DA), norepinephrine, epinephrine, serotonin (5-HT), and histamine, other biogenic amines, present at much lower concentrations in the central nervous system (CNS), and hence referred to as "trace" amines (TAs), are now recognized to play significant neurophysiological and behavioral functions. At the turn of the century, the discovery of the trace amine-associated receptor 1 (TAAR1), a phylogenetically conserved G protein-coupled receptor that is responsive to both TAs, such as β-phenylethylamine, octopamine, and tyramine, and structurally-related amphetamines, unveiled mechanisms of action for TAs other than interference with aminergic pathways, laying the foundations for deciphering the functional significance of TAs and its mammalian CNS receptor, TAAR1. Although, its molecular interactions and downstream targets have not been fully elucidated, TAAR1 activation triggers accumulation of intracellular cAMP, modulates PKA and PKC signaling and interferes with the β-arrestin2-dependent pathway via G protein-independent mechanisms. TAAR1 is uniquely positioned to exert direct control over DA and 5-HT neuronal firing and release, which has profound implications for understanding the pathophysiology of, and therefore designing more efficacious therapeutic interventions for, a range of neuropsychiatric disorders that involve aminergic dysregulation, including Parkinson's disease, schizophrenia, mood disorders, and addiction. Indeed, the recent development of novel pharmacological tools targeting TAAR1 has uncovered the remarkable potential of TAAR1-based medications as new generation pharmacotherapies in neuropsychiatry. This review summarizes recent developments in the study of TAs and TAAR1, their intricate neurochemistry and pharmacology, and their relevance for neurodegenerative and neuropsychiatric disease.
Collapse
Affiliation(s)
| | | | - Juan J. Canales
- Department of Neuroscience, Psychology and Behaviour, University of LeicesterLeicester, UK
| |
Collapse
|
35
|
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: 109] [Impact Index Per Article: 13.6] [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
|
36
|
Oda Y, Kanahara N, Iyo M. Alterations of Dopamine D2 Receptors and Related Receptor-Interacting Proteins in Schizophrenia: The Pivotal Position of Dopamine Supersensitivity Psychosis in Treatment-Resistant Schizophrenia. Int J Mol Sci 2015; 16:30144-63. [PMID: 26694375 PMCID: PMC4691170 DOI: 10.3390/ijms161226228] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 12/01/2015] [Accepted: 12/08/2015] [Indexed: 12/22/2022] Open
Abstract
Although the dopamine D2 receptor (DRD2) has been a main target of antipsychotic pharmacotherapy for the treatment of schizophrenia, the standard treatment does not offer sufficient relief of symptoms to 20%-30% of patients suffering from this disorder. Moreover, over 80% of patients experience relapsed psychotic episodes within five years following treatment initiation. These data strongly suggest that the continuous blockade of DRD2 by antipsychotic(s) could eventually fail to control the psychosis in some point during long-term treatment, even if such treatment has successfully provided symptomatic improvement for the first-episode psychosis, or stability for the subsequent chronic stage. Dopamine supersensitivity psychosis (DSP) is historically known as a by-product of antipsychotic treatment in the manner of tardive dyskinesia or transient rebound psychosis. Numerous data in psychopharmacological studies suggest that the up-regulation of DRD2, caused by antipsychotic(s), is likely the mechanism underlying the development of the dopamine supersensitivity state. However, regardless of evolving notions of dopamine signaling, particularly dopamine release, signal transduction, and receptor recycling, most of this research has been conducted and discussed from the standpoint of disease etiology or action mechanism of the antipsychotic, not of DSP. Hence, the mechanism of the DRD2 up-regulation or mechanism evoking clinical DSP, both of which are caused by pharmacotherapy, remains unknown. Once patients experience a DSP episode, they become increasingly difficult to treat. Light was recently shed on a new aspect of DSP as a treatment-resistant factor. Clarification of the detailed mechanism of DSP is therefore crucial, and a preventive treatment strategy for DSP or treatment-resistant schizophrenia is urgently needed.
Collapse
Affiliation(s)
- Yasunori Oda
- Department of Psychiatry, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuou-ku, Chiba 260-8670, Japan.
| | - Nobuhisa Kanahara
- Division of Medical Treatment and Rehabilitation, Chiba University Center for Forensic Mental Health, 1-8-1 Inohana, Chuou-ku, Chiba 260-8670, Japan.
| | - Masaomi Iyo
- Department of Psychiatry, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuou-ku, Chiba 260-8670, Japan.
| |
Collapse
|
37
|
Gancarz A, Jouroukhin Y, Saito A, Shevelkin A, Mueller LE, Kamiya A, Dietz DM, Pletnikov MV. DISC1 signaling in cocaine addiction: Towards molecular mechanisms of co-morbidity. Neurosci Res 2015; 105:70-4. [PMID: 26385055 DOI: 10.1016/j.neures.2015.09.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Revised: 09/03/2015] [Accepted: 09/07/2015] [Indexed: 11/30/2022]
Abstract
Substance abuse and other psychiatric diseases may share molecular pathology. In order to test this hypothesis, we examined the role of Disrupted In Schizophrenia 1 (DISC1), a psychiatric risk factor, in cocaine self-administration (SA). Cocaine SA significantly increased expression of DISC1 in the nucleus accumbens (NAc); while knockdown of DISC1 in NAc significantly increased cocaine SA and decreased phosphorylation of GSK-3β at Ser9 compared to scrambled shRNA. Our study provides the first mechanistic evidence of a critical role of DISC1 in drug-induced behavioral neuroadaptations and sheds more light at the shared molecular pathology of drug abuse and other major psychiatric disorders.
Collapse
Affiliation(s)
- Amy Gancarz
- The State University of New York at Buffalo, Buffalo, NY, USA
| | - Yan Jouroukhin
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Atsushi Saito
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Alexey Shevelkin
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, USA
| | | | - Atsushi Kamiya
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, USA
| | - David M Dietz
- The State University of New York at Buffalo, Buffalo, NY, USA.
| | - Mikhail V Pletnikov
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, USA; Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, USA; Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, USA; Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, USA.
| |
Collapse
|
38
|
Abstract
The variety of physiological functions controlled by dopamine in the brain and periphery is mediated by the D1, D2, D3, D4 and D5 dopamine GPCRs. Drugs acting on dopamine receptors are significant tools for the management of several neuropsychiatric disorders including schizophrenia, bipolar disorder, depression and Parkinson's disease. Recent investigations of dopamine receptor signalling have shown that dopamine receptors, apart from their canonical action on cAMP-mediated signalling, can regulate a myriad of cellular responses to fine-tune the expression of dopamine-associated behaviours and functions. Such signalling mechanisms may involve alternate G protein coupling or non-G protein mechanisms involving ion channels, receptor tyrosine kinases or proteins such as β-arrestins that are classically involved in GPCR desensitization. Another level of complexity is the growing appreciation of the physiological roles played by dopamine receptor heteromers. Applications of new in vivo techniques have significantly furthered the understanding of the physiological functions played by dopamine receptors. Here we provide an update of the current knowledge regarding the complex biology, signalling, physiology and pharmacology of dopamine receptors.
Collapse
|
39
|
LY395756, an mGluR2 agonist and mGluR3 antagonist, enhances NMDA receptor expression and function in the normal adult rat prefrontal cortex, but fails to improve working memory and reverse MK801-induced working memory impairment. Exp Neurol 2015; 273:190-201. [PMID: 26341392 DOI: 10.1016/j.expneurol.2015.08.019] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Revised: 08/06/2015] [Accepted: 08/25/2015] [Indexed: 11/22/2022]
Abstract
Targeting group II metabotropic glutamate receptors (mGluR2/3) has been proposed to correct the dysfunctional glutamatergic system, particularly NMDA receptor (NMDAR) hypofunction, for treatment of schizophrenia. However, how activation of mGluR2/3 affects NMDAR function in adult animals remains elusive. Here we show the effects of LY395756 (LY39), a compound acting as both an mGluR2 agonist and mGluR3 antagonist, on the NMDAR expression and function of normal adult rat prefrontal cortex (PFC) as well as working memory function in the MK801 model of schizophrenia. We found that in vivo administration of LY39 significantly increased the total protein levels of NMDAR subunits and NR2B phosphorylationin the PFC, along with the amplitude of NMDAR-mediated miniature excitatory postsynaptic currents (mEPSC) in the prefrontal cortical neurons. Moreover, LY39 also significantly increased mTOR and pmTOR expression, but not ERK1/2, Akt, and GSK3β, suggesting an activation of mTOR signaling. Indeed, the mTOR inhibitor rapamycin, and actinomycin-D, a transcription inhibitor, blocked the enhanced effects of LY39 on NMDAR-mEPSCs. These results indicate that LY39 regulates NMDAR expression and function through unidentified mTOR-mediated protein synthesis in the normal adult rat PFC. However, this change is insufficient to affect working memory function in normal animals, nor to reverse the MK801-induced working memory deficit. Our data provide the first evidence of an in vivo effect of a novel compound that acts as both an mGluR2 agonist and mGluR3 antagonist on synaptic NMDAR expression and function in the adult rat PFC, although its effect -on PFC-dependent cognitive function remains to be explored.
Collapse
|
40
|
Keegan BMT, Beveridge TJR, Pezor JJ, Xiao R, Sexton T, Childers SR, Howlett AC. Chronic baclofen desensitizes GABA(B)-mediated G-protein activation and stimulates phosphorylation of kinases in mesocorticolimbic rat brain. Neuropharmacology 2015; 95:492-502. [PMID: 25724082 PMCID: PMC4537290 DOI: 10.1016/j.neuropharm.2015.02.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Revised: 12/30/2014] [Accepted: 02/11/2015] [Indexed: 01/19/2023]
Abstract
The GABAB receptor is a therapeutic target for CNS and neuropathic disorders; however, few preclinical studies have explored effects of chronic stimulation. This study evaluated acute and chronic baclofen treatments on GABAB-activated G-proteins and signaling protein phosphorylation as indicators of GABAB signaling capacity. Brain sections from rats acutely administered baclofen (5 mg/kg, i.p.) showed no significant differences from controls in GABAB-stimulated GTPγS binding in any brain region, but displayed significantly greater phosphorylation/activation of focal adhesion kinase (pFAK(Tyr397)) in mesocorticolimbic regions (caudate putamen, cortex, hippocampus, thalamus) and elevated phosphorylated/activated glycogen synthase kinase 3-β (pGSK3β(Tyr216)) in the prefrontal cortex, cerebral cortex, caudate putamen, nucleus accumbens, thalamus, septum, and globus pallidus. In rats administered chronic baclofen (5 mg/kg, t.i.d. for five days), GABAB-stimulated GTPγS binding was significantly diminished in the prefrontal cortex, septum, amygdala, and parabrachial nucleus compared to controls. This effect was specific to GABAB receptors: there was no effect of chronic baclofen treatment on adenosine A1-stimulated GTPγS binding in any region. Chronically-treated rats also exhibited increases in pFAK(Tyr397) and pGSK3β(Tyr216) compared to controls, and displayed wide-spread elevations in phosphorylated dopamine- and cAMP-regulated phosphoprotein-32 (pDARPP-32(Thr34)) compared to acutely-treated or control rats. We postulate that those neuroadaptive effects of GABAB stimulation mediated by G-proteins and their sequelae correlate with tolerance to several of baclofen's effects, whereas sustained signaling via kinase cascades points to cross-talk between GABAB receptors and alternative mechanisms that are resistant to desensitization. Both desensitized and sustained signaling pathways should be considered in the development of pharmacotherapies targeting the GABA system.
Collapse
Affiliation(s)
- Bradley M T Keegan
- Center for the Neurobiology of Addiction Treatment, Winston-Salem, NC 27157, USA; Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Thomas J R Beveridge
- Center for the Neurobiology of Addiction Treatment, Winston-Salem, NC 27157, USA; Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Jeffrey J Pezor
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA; Department of Chemistry, Wake Forest University, Winston-Salem, NC 27157, USA
| | - Ruoyu Xiao
- Center for the Neurobiology of Addiction Treatment, Winston-Salem, NC 27157, USA; Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Tammy Sexton
- Center for the Neurobiology of Addiction Treatment, Winston-Salem, NC 27157, USA; Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Steven R Childers
- Center for the Neurobiology of Addiction Treatment, Winston-Salem, NC 27157, USA; Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Allyn C Howlett
- Center for the Neurobiology of Addiction Treatment, Winston-Salem, NC 27157, USA; Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA.
| |
Collapse
|
41
|
Li ML, Hu XQ, Li F, Gao WJ. Perspectives on the mGluR2/3 agonists as a therapeutic target for schizophrenia: Still promising or a dead end? Prog Neuropsychopharmacol Biol Psychiatry 2015; 60:66-76. [PMID: 25724760 PMCID: PMC4426221 DOI: 10.1016/j.pnpbp.2015.02.012] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2014] [Revised: 02/16/2015] [Accepted: 02/17/2015] [Indexed: 01/08/2023]
Abstract
Group II metabotropic glutamate receptor (mGluR2/3) agonists once showed promise as non-dopaminergic antipsychotic drugs because of their efficacy in alleviating symptoms of schizophrenia (SZ) in both animal models and human patients. However, the recent failure of Phase III clinical trials dealt a huge blow to the scientific community and the aftershock of the setback in mGluR2/3 research can be felt everywhere from grant support and laboratory studies to paper publication. An immediate question raised is whether mGluR2/3 is still a promising therapeutic target for schizophrenia. Answering this question is not easy, but apparently a new strategy is needed. This article provides a focused review of literature on the study of mGluR2/3 agonists, especially on mGluR2/3 agonists' mechanism of action and efficacy in both normal conditions and animal models of SZ, as well as clinical studies in human patients with the disease. We argue that the cellular and molecular actions of mGluR2/3 agonists, the distinct roles between mGluR2 and mGluR3, as well as their effects on different stages of the disease and different subpopulations of patients, remain incompletely studied. Until the mechanisms associated with mGluR2/3 are clearly elucidated and all treatment options are tested, it would be a great mistake to terminate the study of mGluR2/3 as a therapeutic target for schizophrenia. This review will thus shed light on the comprehensive features of the translational potential mGluR2/3 agonists as well as the need for further research into the more selective activation of mGluR2.
Collapse
Affiliation(s)
- Meng-Lin Li
- Drexel University College of Medicine, Philadelphia, PA, USA,Department of Rehabilitation, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xi-Quan Hu
- Department of Rehabilitation, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Feng Li
- Department of Neurobiology and Anatomy, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Wen-Jun Gao
- Drexel University College of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
42
|
Xing B, Liang XP, Liu P, Zhao Y, Chu Z, Dang YH. Valproate Inhibits Methamphetamine Induced Hyperactivity via Glycogen Synthase Kinase 3β Signaling in the Nucleus Accumbens Core. PLoS One 2015; 10:e0128068. [PMID: 26030405 PMCID: PMC4452337 DOI: 10.1371/journal.pone.0128068] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 04/23/2015] [Indexed: 11/20/2022] Open
Abstract
Valproate (VPA) has recently been shown to influence the behavioral effects of psycho-stimulants. Although glycogen synthase kinase 3β (GSK3β) signaling in the nucleus accumbens (NAc) plays a key role in mediating dopamine (DA)-dependent behaviors, there is less direct evidence that how VPA acts on the GSK3β signaling in the functionally distinct sub-regions of the NAc, the NAc core (NAcC) and the NAc shell (NAcSh), during psycho-stimulant-induced hyperactivity. In the present study, we applied locomotion test after acute methamphetamine (MA) (2 mg/kg) injection to identify the locomotor activity of rats received repeated VPA (300 mg/kg) pretreatment. We next measured phosphor-GSK3β at serine 9 and total GSK3β levels in NAcC and NAcSh respectively to determine the relationship between the effect of VPA on MA-induced hyperlocomotor and changes in GSK3β activity. We further investigated whether microinjection of VPA (300 μg/0.5 μl/side, once daily for 7 consecutive days) into NAcC or NAcSh could affect hyperactivity induced by MA. Our data indicated that repeated VPA treatment attenuated MA-induced hyperlocomotor, and the effect was associated with decreased levels of phosphorylated GSK3β at Ser 9 in the NAcC. Moreover, repeated bilateral intra-NAcC, but not intra-NAcSh VPA treatment, significantly attenuated MA-induced hyperactivity. Our results suggested that GSK3β activity in NAcC contributes to the inhibitory effects of VPA on MA-induced hyperactivity.
Collapse
Affiliation(s)
- Bo Xing
- Department of Forensic Medicine, Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education, Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi, PR China
- Xi’an Mental Health Center, Xi’an, Shaanxi, PR China
| | | | - Peng Liu
- Department of Forensic Medicine, Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education, Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi, PR China
| | - Yan Zhao
- Department of Forensic Medicine, Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education, Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi, PR China
- Department of Pathophysiology, School of basic Medicine Sciences, Xi'an Medical University, Xi'an, Shaanxi, PR China
| | - Zheng Chu
- Department of Forensic Medicine, Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education, Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi, PR China
| | - Yong-hui Dang
- Department of Forensic Medicine, Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education, Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi, PR China
- * E-mail:
| |
Collapse
|
43
|
Golpich M, Amini E, Hemmati F, Ibrahim NM, Rahmani B, Mohamed Z, Raymond AA, Dargahi L, Ghasemi R, Ahmadiani A. Glycogen synthase kinase-3 beta (GSK-3β) signaling: Implications for Parkinson's disease. Pharmacol Res 2015; 97:16-26. [PMID: 25829335 DOI: 10.1016/j.phrs.2015.03.010] [Citation(s) in RCA: 194] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 03/05/2015] [Accepted: 03/16/2015] [Indexed: 01/02/2023]
Abstract
Glycogen synthase kinase 3 (GSK-3) dysregulation plays an important role in the pathogenesis of numerous disorders, affecting the central nervous system (CNS) encompassing both neuroinflammation and neurodegenerative diseases. Several lines of evidence have illustrated a key role of the GSK-3 and its cellular and molecular signaling cascades in the control of neuroinflammation. Glycogen synthase kinase 3 beta (GSK-3β), one of the GSK-3 isomers, plays a major role in neuronal apoptosis and its inhibition decreases expression of alpha-Synuclein (α-Synuclein), which make this kinase an attractive therapeutic target for neurodegenerative disorders. Parkinson's disease (PD) is a chronic neurodegenerative movement disorder characterized by the progressive and massive loss of dopaminergic neurons by neuronal apoptosis in the substantia nigra pars compacta and depletion of dopamine in the striatum, which lead to pathological and clinical abnormalities. Thus, understanding the role of GSK-3β in PD will enhance our knowledge of the basic mechanisms underlying the pathogenesis of this disorder and facilitate the identification of new therapeutic avenues. In recent years, GSK-3β has been shown to play essential roles in modulating a variety of cellular functions, which have prompted efforts to develop GSK-3β inhibitors as therapeutics. In this review, we summarize GSK-3 signaling pathways and its association with neuroinflammation. Moreover, we highlight the interaction between GSK-3β and several cellular processes involved in the pathogenesis of PD, including the accumulation of α-Synuclein aggregates, oxidative stress and mitochondrial dysfunction. Finally, we discuss about GSK-3β inhibitors as a potential therapeutic strategy in PD.
Collapse
Affiliation(s)
- Mojtaba Golpich
- Department of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Cheras, Kuala Lumpur, Malaysia
| | - Elham Amini
- Department of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Cheras, Kuala Lumpur, Malaysia
| | - Fatemeh Hemmati
- Department of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Cheras, Kuala Lumpur, Malaysia
| | - Norlinah Mohamed Ibrahim
- Department of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Cheras, Kuala Lumpur, Malaysia
| | - Behrouz Rahmani
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahurin Mohamed
- Department of Pharmacology, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Azman Ali Raymond
- Department of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Cheras, Kuala Lumpur, Malaysia
| | - Leila Dargahi
- NeuroBiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Rasoul Ghasemi
- Neurophysiology Research Center and Department of Physiology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Abolhassan Ahmadiani
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Pharmacology, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia.
| |
Collapse
|
44
|
Huang X, Huang K, Zheng W, Beveridge TJR, Yang S, Li X, Li P, Zhou W, Liu Y. The effects of GSK-3β blockade on ketamine self-administration and relapse to drug-seeking behavior in rats. Drug Alcohol Depend 2015; 147:257-65. [PMID: 25497591 DOI: 10.1016/j.drugalcdep.2014.10.028] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Revised: 10/23/2014] [Accepted: 10/25/2014] [Indexed: 11/26/2022]
Abstract
RATIONALE The role of glycogen synthase kinase-3 (GSK-3) has recently been implicated in the neurochemical mechanism underlying ketamine-induced neuronal toxicity and behavioral disturbance. OBJECTIVES The primary goal of the present study was to determine the role of GSK-3β in ketamine self-administration (SA) and relapse to drug-seeking behavior after abstinence. METHODS In Experiment 1, the level of phosphorylated GSK-3β (p-GSK-3β) and total GSK-3β (t-GSK-3β) was determined in various brain areas following 14 days of ketamine SA. In Experiments 2 and 3, the effects of a GSK-3β inhibitor, SB216763 (2 and 4 mg/kg) and a GSK-3 inhibitor, lithium (LiCl, 100mg/kg) on the responding maintained by 0.5mg/kg/infusion ketamine SA were evaluated. In Experiments 4 and 5, rats underwent ketamine SA for 14 days followed by a 10-day abstinence period. The animals were treated with 2 or 4 mg/kg GSK-3β inhibitor, or 100mg/kg LiCl during the cue-induced relapse test. Seven days later, animals received the same drug treatment and underwent the drug-induced relapse test. Finally, the effect of saline and DMSO on locomotor activity was evaluated in Experiment 6. RESULTS Ketamine SA significantly decreased the ratio p-GSK-3β and t-GSK-3β (p-GSK-3β:t-GSK-3β) in the caudate putamen, nucleus accumbens, and ventral tegmental area. Both SB216763 and LiCl decreased responding on a progressive ratio schedule, but not on a fixed ratio schedule. Cue-induced relapse was suppressed only by 4mg/kg SB216763, whereas drug-induced relapse was inhibited by 2, 4 mg/kg SB216763 and LiCl. However, inactive responses were also suppressed by LiCl during progressive ratio and drug-induced relapse testing. CONCLUSIONS SB216763 was effective at decreasing ketamine SA under the PR schedule and reducing drug-seeking behavior after abstinence.
Collapse
Affiliation(s)
- Xianni Huang
- Ningbo University School of Medicine, 818 Fenghua Road, Ningbo 315211, Zhejiang, PR China
| | - Kunyu Huang
- Ningbo University School of Medicine, 818 Fenghua Road, Ningbo 315211, Zhejiang, PR China
| | - Wenhui Zheng
- Ningbo University School of Medicine, 818 Fenghua Road, Ningbo 315211, Zhejiang, PR China
| | - Thomas J R Beveridge
- Center for the Neurobiology of Addiction Treatment, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA; Ferring Pharmaceuticals Inc., 100 Interpace Pkwy, Parsippany, NJ 07054 USA
| | - Shujun Yang
- Ningbo University School of Medicine, 818 Fenghua Road, Ningbo 315211, Zhejiang, PR China
| | - Xingxing Li
- Ningbo University School of Medicine, 818 Fenghua Road, Ningbo 315211, Zhejiang, PR China
| | - Pengping Li
- Ningbo University School of Medicine, 818 Fenghua Road, Ningbo 315211, Zhejiang, PR China
| | - Wenhua Zhou
- Ningbo University School of Medicine, 818 Fenghua Road, Ningbo 315211, Zhejiang, PR China; Drug Addiction Research and Treatment Center of Ningbo, 42 Xibei St., Ningbo 315010, Zhejiang, PR China.
| | - Yu Liu
- Ningbo University School of Medicine, 818 Fenghua Road, Ningbo 315211, Zhejiang, PR China.
| |
Collapse
|
45
|
McCubrey JA, Steelman LS, Bertrand FE, Davis NM, Sokolosky M, Abrams SL, Montalto G, D'Assoro AB, Libra M, Nicoletti F, Maestro R, Basecke J, Rakus D, Gizak A, Demidenko ZN, Cocco L, Martelli AM, Cervello M. GSK-3 as potential target for therapeutic intervention in cancer. Oncotarget 2015; 5:2881-911. [PMID: 24931005 PMCID: PMC4102778 DOI: 10.18632/oncotarget.2037] [Citation(s) in RCA: 377] [Impact Index Per Article: 41.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The serine/threonine kinase glycogen synthase kinase-3 (GSK-3) was initially identified and studied in the regulation of glycogen synthesis. GSK-3 functions in a wide range of cellular processes. Aberrant activity of GSK-3 has been implicated in many human pathologies including: bipolar depression, Alzheimer's disease, Parkinson's disease, cancer, non-insulin-dependent diabetes mellitus (NIDDM) and others. In some cases, suppression of GSK-3 activity by phosphorylation by Akt and other kinases has been associated with cancer progression. In these cases, GSK-3 has tumor suppressor functions. In other cases, GSK-3 has been associated with tumor progression by stabilizing components of the beta-catenin complex. In these situations, GSK-3 has oncogenic properties. While many inhibitors to GSK-3 have been developed, their use remains controversial because of the ambiguous role of GSK-3 in cancer development. In this review, we will focus on the diverse roles that GSK-3 plays in various human cancers, in particular in solid tumors. Recently, GSK-3 has also been implicated in the generation of cancer stem cells in various cell types. We will also discuss how this pivotal kinase interacts with multiple signaling pathways such as: PI3K/PTEN/Akt/mTORC1, Ras/Raf/MEK/ERK, Wnt/beta-catenin, Hedgehog, Notch and others.
Collapse
Affiliation(s)
- James A McCubrey
- Department of Microbiology and Immunology,Brody School of Medicine at East Carolina University Greenville, NC 27858 USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Gomez-Sintes R, Bortolozzi A, Artigas F, Lucas JJ. Reduced striatal dopamine DA D2 receptor function in dominant-negative GSK-3 transgenic mice. Eur Neuropsychopharmacol 2014; 24:1524-33. [PMID: 25088904 DOI: 10.1016/j.euroneuro.2014.07.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2013] [Revised: 06/07/2014] [Accepted: 07/11/2014] [Indexed: 11/17/2022]
Abstract
Glycogen synthase kinase-3 (GSK-3) is a serine/threonine kinase with constitutive activity involved in cellular architecture, gene expression, cell proliferation, fate decision and apoptosis, among others. GSK-3 expression is particularly high in brain where it may be involved in neurological and psychiatric disorders such as Alzheimer׳s disease, bipolar disorder and major depression. A link with schizophrenia is suggested by the antipsychotic drug-induced GSK-3 regulation and by the involvement of the Akt/GSK-3 pathway in dopaminergic neurotransmission. Taking advantage of the previous development of dominant negative GSK-3 transgenic mice (Tg) showing a selective reduction of GSK-3 activity in forebrain neurons but not in dopaminergic neurons, we explored the relationship between GSK-3 and dopaminergic neurotransmission in vivo. In microdialysis experiments, local quinpirole (DA D2-R agonist) in dorsal striatum reduced dopamine (DA) release significantly less in Tg mice than in wild-type (WT) mice. However, local SKF-81297 (selective DA D1-R agonist) in dorsal striatum reduced DA release equally in both control and Tg mice indicating a comparable function of DA D1-R in the direct striato-nigral pathway. Likewise, systemic quinpirole administration - acting preferentially on presynaptic DA D2- autoreceptors to modulate DA release-reduced striatal DA release similarly in both control and Tg mice. Quinpirole reduced locomotor activity and induced c-fos expression in globus pallidus (both striatal DA D2-R-mediated effects) significantly more in WT than in Tg mice. Taking together, the present results show that dominant negative GSK-3 transgenic mice show reduced DA D2-R-mediated function in striatum and further support a link between dopaminergic neurotransmission and GSK-3 activity.
Collapse
Affiliation(s)
- Raquel Gomez-Sintes
- Centro de Biología Molecular "Severo Ochoa" (CBM"SO"), CSIC/UAM, 28049 Madrid, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Analia Bortolozzi
- Department of Neurochemistry and Neuropharmacology, IIBB - Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), CSIC, Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
| | - Francesc Artigas
- Department of Neurochemistry and Neuropharmacology, IIBB - Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), CSIC, Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain.
| | - José J Lucas
- Centro de Biología Molecular "Severo Ochoa" (CBM"SO"), CSIC/UAM, 28049 Madrid, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
47
|
Edut S, Rubovitch V, Rehavi M, Schreiber S, Pick CG. A study on the mechanism by which MDMA protects against dopaminergic dysfunction after minimal traumatic brain injury (mTBI) in mice. J Mol Neurosci 2014; 54:684-97. [PMID: 25124230 DOI: 10.1007/s12031-014-0399-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Accepted: 08/05/2014] [Indexed: 12/11/2022]
Abstract
Driving under methylenedioxymethamphetamine (MDMA) influence increases the risk of being involved in a car accident, which in turn can lead to traumatic brain injury. The behavioral deficits after traumatic brain injury (TBI) are closely connected to dopamine pathway dysregulation. We have previously demonstrated in mice that low MDMA doses prior to mTBI can lead to better performances in cognitive tests. The purpose of this study was to assess in mice the changes in the dopamine system that occurs after both MDMA and minimal traumatic brain injury (mTBI). Experimental mTBI was induced using a concussive head trauma device. One hour before injury, animals were subjected to MDMA. Administration of MDMA before injury normalized the alterations in tyrosine hydroxylase (TH) levels that were observed in mTBI mice. This normalization was also able to lower the elevated dopamine receptor type 2 (D2) levels observed after mTBI. Brain-derived neurotrophic factor (BDNF) levels did not change following injury alone, but in mice subjected to MDMA and mTBI, significant elevations were observed. In the behavioral tests, haloperidol reversed the neuroprotection seen when MDMA was administered prior to injury. Altered catecholamine synthesis and high D2 receptor levels contribute to cognitive dysfunction, and strategies to normalize TH signaling and D2 levels may provide relief for the deficits observed after injury. Pretreatment with MDMA kept TH and D2 receptor at normal levels, allowing regular dopamine system activity. While the beneficial effect we observe was due to a dangerous recreational drug, understanding the alterations in dopamine and the mechanism of dysfunction at a cellular level can lead to legal therapies and potential candidates for clinical use.
Collapse
Affiliation(s)
- S Edut
- Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel Aviv University, 69978, Tel Aviv, Israel,
| | | | | | | | | |
Collapse
|
48
|
Li Y, Jia L, Ren D, Liu C, Gong Y, Wang N, Zhang X, Zhao Y. Axl mediates tumor invasion and chemosensitivity through PI3K/Akt signaling pathway and is transcriptionally regulated by slug in breast carcinoma. IUBMB Life 2014; 66:507-18. [PMID: 24984960 DOI: 10.1002/iub.1285] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Accepted: 06/16/2014] [Indexed: 11/06/2022]
Abstract
The invasion and chemoresistance are crucial causes of morbidity and relapse for cancer patients. Axl is implicated in the modulation of cell invasion, cancer metastasis, and chemosensitivity in human breast carcinoma cell lines. Both breast cancer cell lines and tissues displayed increased expression of Axl, and it over expressed in highly metastatic breast cancer. The altered expression level of Axl was corresponding to the changed invasive phenotype and chemosensitivity of MDA-MB-231 cells both in vitro and in vivo. Further data indicated that experimental inhibition of Axl by RNAi assay inhibited phosphatidylinositol 3-kinase (PI3K)/Akt/GSK3β signaling pathway, resulted in the decrease of Slug expression, and further suppressed cell invasion properties and chemosensitivity. What is more, after the detection and statistics in human breast cancer specimens, we found the Axl expression was closely correlated with histological grade, lymph node metastasis, and clinical stage (P < 0.01). Taken together, these findings indicate that Axl exerts the role of tumor metastasis and chemosensitivity through activation of the PI3K/Akt/GSK3β signaling pathway, which is transcriptionally regulated by Slug. Our findings support the possibility that Axl is a novel regulator. It means by targeting Axl or its related signaling pathways, we can reduce the invasion and chemosensitivity of breast tumor.
Collapse
Affiliation(s)
- Yanyan Li
- Department of General Surgery, Dalian Medical University, Dalian, Liaoning Province, China
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Zhou J, Ping FF, Lv WT, Feng JY, Shang J. Interleukin-18 directly protects cortical neurons by activating PI3K/AKT/NF-κB/CREB pathways. Cytokine 2014; 69:29-38. [PMID: 25022959 DOI: 10.1016/j.cyto.2014.05.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Revised: 04/12/2014] [Accepted: 05/01/2014] [Indexed: 12/11/2022]
Abstract
Interleukin-18 (IL-18), a member of the IL-1 family of cytokines, was initially identified as an interferon (IFN)-γ-inducing factor. IL-18 is expressed in both immune and non-immune cells and participates in the adjustment of multitude cellular functions. Nonetheless, the effects of IL-18 on cortical neurons have not been explored. The present study was conducted to investigate the influence of IL-18 on rat primary cortical neurons and elucidate the underlying mechanisms. We proved that rrIL-18 increased the brain-derived neurotrophic factor (BDNF) expression in a time-dependent manner. Treatment with rrIL-18 (50 ng/ml) deactivated phosphatase and tensin homolog deleted on chromosome 10 (PTEN) by facilitating its phosphorylation, enhanced the expression of Phosphoinositide 3-OH kinase (PI3K) and p-Akt, standing for the activation of the PI3K/Akt pathway. As its pivotal downstream pathways, nuclear factor-kappa B (NF-κB), cAMP-responsive element binding protein (CREB)/Bcl-2 and glycogen synthase kinase-3β (GSK-3β) were examined in further steps. Our data revealed that rrIL-18 stimulated NF-κB activation, improved p-CREB and anti-apoptotic Bcl-2 expression levels. But rrIL-18 had little or no effect on GSK-3β pathway. Besides, rrIL-18 increased levels of BDNF and Bcl-2/Bax ratio and decreased cleaved caspase-3 expression to protect cortical neurons from damage induced by oxygen-glucose deprivation (OGD). These results in vitro showed the protection of IL-18 on cortical neurons. And this direct neuroprotective effect of IL-18 is crippled by PI3K inhibitor wortmannin.
Collapse
Affiliation(s)
- Jia Zhou
- Center for Drug Screening, China Pharmaceutical University, Nanjing 210009, China; State Key Laboratory of Natural Medicines, China Pharmaceutical University, China
| | - Feng-feng Ping
- Center for Drug Screening, China Pharmaceutical University, Nanjing 210009, China; Department of Clinical Laboratory Science, Wuxi People's Hospital affiliated to Nanjing Medical University, China
| | - Wen-ting Lv
- Center for Drug Screening, China Pharmaceutical University, Nanjing 210009, China; State Key Laboratory of Natural Medicines, China Pharmaceutical University, China
| | - Jun-yi Feng
- Center for Drug Screening, China Pharmaceutical University, Nanjing 210009, China; State Key Laboratory of Natural Medicines, China Pharmaceutical University, China
| | - Jing Shang
- Center for Drug Screening, China Pharmaceutical University, Nanjing 210009, China; State Key Laboratory of Natural Medicines, China Pharmaceutical University, China.
| |
Collapse
|
50
|
González-López A, López-Alonso I, Aguirre A, Amado-Rodríguez L, Batalla-Solís E, Astudillo A, Tomás-Zapico C, Fueyo A, dos Santos CC, Talbot K, Albaiceta GM. Mechanical ventilation triggers hippocampal apoptosis by vagal and dopaminergic pathways. Am J Respir Crit Care Med 2013; 188:693-702. [PMID: 23962032 DOI: 10.1164/rccm.201304-0691oc] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Critically ill patients frequently develop neuropsychological disturbances including acute delirium or memory impairment. The need for mechanical ventilation is a risk factor for these adverse events, but a mechanism that links lung stretch and brain injury has not been identified. OBJECTIVES To identify the mechanisms that lead to brain dysfunction during mechanical ventilation. METHODS Brains from mechanically ventilated mice were harvested, and signals of apoptosis and alterations in the Akt survival pathway were studied. These measurements were repeated in vagotomized or haloperidol-treated mice, and in animals intracerebroventricularly injected with selective dopamine-receptor blockers. Hippocampal slices were cultured and treated with micromolar concentrations of dopamine, with or without dopamine receptor blockers. Last, levels of dysbindin, a regulator of the membrane availability of dopamine receptors, were assessed in the experimental model and in brain samples from ventilated patients. MEASUREMENTS AND MAIN RESULTS Mechanical ventilation triggers hippocampal apoptosis as a result of type 2 dopamine receptor activation in response to vagal signaling. Activation of these receptors blocks the Akt/GSK3β prosurvival pathway and activates the apoptotic cascade, as demonstrated in vivo and in vitro. Vagotomy, systemic haloperidol, or intracerebroventricular raclopride (a type 2 dopamine receptor blocker) ameliorated this effect. Moreover, ventilation induced a concomitant change in the expression of dysbindin-1C. These results were confirmed in brain samples from ventilated patients. CONCLUSIONS These results prove the existence of a pathogenic mechanism of lung stretch-induced hippocampal apoptosis that could explain the neurological changes in ventilated patients and may help to identify novel therapeutic approaches.
Collapse
Affiliation(s)
- Adrián González-López
- 1 Departamento de Biología Funcional, Área de Fisiología, Instituto Universitario de Oncología del Principado de Asturias, and
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|