151
|
Lucchese G. From Toxoplasmosis to Schizophrenia via NMDA Dysfunction: Peptide Overlap between Toxoplasma gondii and N-Methyl-d-Aspartate Receptors As a Potential Mechanistic Link. Front Psychiatry 2017; 8:37. [PMID: 28360866 PMCID: PMC5350139 DOI: 10.3389/fpsyt.2017.00037] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 02/27/2017] [Indexed: 12/24/2022] Open
Abstract
The present work aims at investigating how Toxoplasma gondii (T. gondii) infection may be linked to N-methyl-d-aspartate receptor (NMDAR) dysfunction in schizophrenia and related disorders and puts forward the hypothesis that immune responses against T. gondii may involve NMDARs. Indeed, the analysis of the protozoan proteome and NMDAR subunits for peptide commonalities shows a massive peptide overlap and supports the possibility that anti-T. gondii immune responses raised during active protozoan infection may cross-react with host NMDARs, determining disruption of neural circuits and cognitive deficits. In particular, the NMDA 2D subunit, which is mainly expressed in parvalbumin-positive interneurons, appears to be a hotspot for potential T. gondii-induced cross-reactive immune attacks.
Collapse
|
152
|
Hansen KB, Yi F, Perszyk RE, Menniti FS, Traynelis SF. NMDA Receptors in the Central Nervous System. Methods Mol Biol 2017; 1677:1-80. [PMID: 28986865 DOI: 10.1007/978-1-4939-7321-7_1] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
NMDA-type glutamate receptors are ligand-gated ion channels that mediate a major component of excitatory neurotransmission in the central nervous system (CNS). They are widely distributed at all stages of development and are critically involved in normal brain functions, including neuronal development and synaptic plasticity. NMDA receptors are also implicated in the pathophysiology of numerous neurological and psychiatric disorders, such as ischemic stroke, traumatic brain injury, Alzheimer's disease, epilepsy, mood disorders, and schizophrenia. For these reasons, NMDA receptors have been intensively studied in the past several decades to elucidate their physiological roles and to advance them as therapeutic targets. Seven NMDA receptor subunits exist that assemble into a diverse array of tetrameric receptor complexes, which are differently regulated, have distinct regional and developmental expression, and possess a wide range of functional and pharmacological properties. The diversity in subunit composition creates NMDA receptor subtypes with distinct physiological roles across neuronal cell types and brain regions, and enables precise tuning of synaptic transmission. Here, we will review the relationship between NMDA receptor structure and function, the diversity and significance of NMDA receptor subtypes in the CNS, as well as principles and rules by which NMDA receptors operate in the CNS under normal and pathological conditions.
Collapse
Affiliation(s)
- Kasper B Hansen
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT, USA. .,Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, MT, USA.
| | - Feng Yi
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT, USA
| | - Riley E Perszyk
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA, USA
| | - Frank S Menniti
- MindImmune Therapeutics, Inc., George & Anne Ryan Institute for Neuroscience, Kingston, RI, USA
| | - Stephen F Traynelis
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
153
|
Tonin FS, Wiens A, Fernandez-Llimos F, Pontarolo R. Iloperidone in the treatment of schizophrenia: an evidence-based review of its place in therapy. CORE EVIDENCE 2016; 11:49-61. [PMID: 28008301 PMCID: PMC5167526 DOI: 10.2147/ce.s114094] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Introduction Schizophrenia is a chronic and debilitating mental disorder that affects the patient’s and their family’s quality of life, as well as financial costs and health care settings. Despite the variety of available antipsychotics, optimal treatment outcomes are not always achieved. Novel drugs, such as iloperidone, can provide more effective, tolerable and safer strategies. Aim To review the evidence for the clinical impact of iloperidone on the treatment of patients with schizophrenia. Evidence review Clinical trials, observational studies and meta-analyses reached a common consensus that iloperidone is as effective as haloperidol, risperidone and ziprasidone in reducing schizophrenia symptoms. Similar amounts of adverse events and discontinuations were observed with iloperidone compared to placebo and active treatments. Common adverse events are mild and include dizziness, hypotension, dry mouth and weight gain. Iloperidone can induce extension of QTc interval, and clinicians should be aware of its contraindications. In long-term trials, iloperidone also showed promising safety and tolerability profiles. The low propensity to cause akathisia, extrapyramidal symptoms (EPS), increased prolactin levels or changes to metabolic laboratory parameters support its use in practice. Results showed that iloperidone prevents relapse in stabilized patients, with a time to relapse superior to placebo and similar to haloperidol. Patients using a prior antipsychotic (eg, risperidone and aripiprazole) can easily switch to iloperidone with no serious impact on safety or efficacy. However, the acquisition costs of iloperidone may hamper its use. Further evidence comparing iloperidone with other antipsychotics, and pharmacoeconomic studies would be welcome. Place in therapy Considering just the clinical profile of iloperidone, it represents a promising drug for treating schizophrenia, particularly in patients who are intolerant to previous antipsychotics, as well as being suitable as first-line therapy. Cost-effectiveness comparisons are needed to justify its use in clinical practice.
Collapse
Affiliation(s)
| | - Astrid Wiens
- Department of Pharmacy, Federal University of Parana, Curitiba, Brazil
| | - Fernando Fernandez-Llimos
- Department of Social Pharmacy, Faculty of Pharmacy, Research Institute for Medicines (iMed.ULisboa), Universidade de Lisboa, Lisbon, Portugal
| | - Roberto Pontarolo
- Department of Pharmacy, Federal University of Parana, Curitiba, Brazil
| |
Collapse
|
154
|
Yoo CH, Song KH, Lim SI, Lee DW, Woo DC, Choe BY. Effects of repeated dizocilpine treatment on glutamatergic activity in the prefrontal cortex in an animal model of schizophrenia: An in vivo proton magnetic resonance spectroscopy study at 9.4T. Neurosci Lett 2016; 637:57-63. [PMID: 27894920 DOI: 10.1016/j.neulet.2016.11.053] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 11/24/2016] [Accepted: 11/24/2016] [Indexed: 12/30/2022]
Abstract
Repeated exposure to dizocilpine (MK-801) can be used as a model of schizophrenia that incorporates disease progression. Proton magnetic resonance spectroscopy (1H MRS) has been widely used to investigate schizophrenia-related alterations in glutamate (Glu). The purpose of this study was to investigate metabolic alterations in the prefrontal cortex (PFC) in an animal model of schizophrenia by using in vivo 1H MRS. Because of the spectral overlap of Glu and glutamine (Gln), high-field 1H MRS with short echo time (TE) was used. A point-resolved spectroscopy sequence was used to measure the levels of Glu and Gln, and the brain metabolites in a volume of interest (22.5μL) located in the PFC region of rats (n=13) before and after 6days of MK-801 (0.5mg/kg) treatment. Analysis of the spectra showed that the cross-contamination of Glu and Gln can be considered to comparably low. No metabolic parameters were altered (p>0.05). However, differences in Glu and N-acetylaspartate (NAA) levels between two times were significantly correlated (p<0.01). The results showed both decreased (in 6 of the 13 rats) and increased (7 of the 13 rats) levels of Glu and NAA, which suggested that these opposite metabolic alterations reflect two stage of disease progression. The results suggest that high-field and short TE in vivo 1H MRS can quantify Glu and Gln with reliably low level of cross-contamination and that repeated exposure to MK-801 induces the progressive development of schizophrenia.
Collapse
Affiliation(s)
- Chi-Hyeon Yoo
- Department of Biomedical Engineering, Research Institute of Biomedical Engineering, College of Medicine, The Catholic University of Korea, Seoul, 137-701, Republic of Korea; Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Kyu-Ho Song
- Department of Biomedical Engineering, Research Institute of Biomedical Engineering, College of Medicine, The Catholic University of Korea, Seoul, 137-701, Republic of Korea
| | - Song-I Lim
- Department of Biomedical Engineering, Research Institute of Biomedical Engineering, College of Medicine, The Catholic University of Korea, Seoul, 137-701, Republic of Korea; Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Do-Wan Lee
- Ehwa Brain Institute, Ehwa Womans University, Seoul, Republic of Korea
| | - Dong-Cheol Woo
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Bo-Young Choe
- Department of Biomedical Engineering, Research Institute of Biomedical Engineering, College of Medicine, The Catholic University of Korea, Seoul, 137-701, Republic of Korea.
| |
Collapse
|
155
|
Van Snellenberg JX, Girgis RR, Horga G, van de Giessen E, Slifstein M, Ojeil N, Weinstein JJ, Moore H, Lieberman JA, Shohamy D, Smith EE, Abi-Dargham A. Mechanisms of Working Memory Impairment in Schizophrenia. Biol Psychiatry 2016; 80:617-26. [PMID: 27056754 PMCID: PMC4995154 DOI: 10.1016/j.biopsych.2016.02.017] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 02/15/2016] [Accepted: 02/16/2016] [Indexed: 11/19/2022]
Abstract
BACKGROUND The neural correlates of working memory (WM) impairment in schizophrenia remain a key puzzle in understanding the cognitive deficits and dysfunction of dorsolateral prefrontal cortex observed in this disorder. We sought to determine whether patients with schizophrenia exhibit an alteration in the inverted-U relationship between WM load and activation that we recently observed in healthy individuals and whether this could account for WM deficits in this population. METHODS Medicated (n = 30) and unmedicated (n = 21) patients with schizophrenia and healthy control subjects (n = 45) performed the self-ordered WM task during functional magnetic resonance imaging. We identified regions exhibiting an altered fit to an inverted-U relationship between WM load and activation that were also predictive of WM performance. RESULTS A blunted inverted-U response was observed in left dorsolateral prefrontal cortex in patients and was associated with behavioral deficits in WM capacity. In addition, suppression of medial prefrontal cortex during WM was reduced in patients and was associated with poorer WM capacity in patients. Finally, activation of visual cortex in the cuneus was elevated in patients and associated with improved WM capacity. Together, these findings explained 55% of the interindividual variance in WM capacity when combined with diagnostic and medication status, which alone accounted for only 22% of the variance in WM capacity. CONCLUSIONS These findings identify a novel biomarker and putative mechanism of WM deficits in patients with schizophrenia, a reduction or flattening of the inverted-U relationship between activation and WM load observed in healthy individuals in left dorsolateral prefrontal cortex.
Collapse
Affiliation(s)
- Jared X Van Snellenberg
- Department of Psychiatry, Columbia University College of Physicians and Surgeons, New York, New York; Divisions of Translational Imaging, New York, New York; Cognitive Neuroscience, New York, New York.
| | - Ragy R Girgis
- Department of Psychiatry, Columbia University College of Physicians and Surgeons, New York, New York; Divisions of Translational Imaging, New York, New York
| | - Guillermo Horga
- Department of Psychiatry, Columbia University College of Physicians and Surgeons, New York, New York; Divisions of Translational Imaging, New York, New York
| | - Elsmarieke van de Giessen
- Department of Psychiatry, Columbia University College of Physicians and Surgeons, New York, New York; Divisions of Translational Imaging, New York, New York; Department of Nuclear Medicine, University of Amsterdam, Amsterdam, The Netherlands
| | - Mark Slifstein
- Department of Psychiatry, Columbia University College of Physicians and Surgeons, New York, New York; Divisions of Translational Imaging, New York, New York
| | - Najate Ojeil
- Department of Psychiatry, Columbia University College of Physicians and Surgeons, New York, New York; Divisions of Translational Imaging, New York, New York
| | - Jodi J Weinstein
- Department of Psychiatry, Columbia University College of Physicians and Surgeons, New York, New York; Divisions of Translational Imaging, New York, New York
| | - Holly Moore
- Department of Psychiatry, Columbia University College of Physicians and Surgeons, New York, New York; Integrative Neuroscience, New York, New York
| | - Jeffrey A Lieberman
- Department of Psychiatry, Columbia University College of Physicians and Surgeons, New York, New York; New York State Psychiatric Institute, New York, New York
| | - Daphna Shohamy
- Department of Psychology, Columbia University, New York, New York
| | - Edward E Smith
- Cognitive Neuroscience, New York, New York; Department of Psychology, Columbia University, New York, New York
| | - Anissa Abi-Dargham
- Department of Psychiatry, Columbia University College of Physicians and Surgeons, New York, New York; Divisions of Translational Imaging, New York, New York
| |
Collapse
|
156
|
Locklear MN, Michaelos M, Collins WF, Kritzer MF. Gonadectomy but not biological sex affects burst-firing in dopamine neurons of the ventral tegmental area and in prefrontal cortical neurons projecting to the ventral tegmentum in adult rats. Eur J Neurosci 2016; 45:106-120. [PMID: 27564091 DOI: 10.1111/ejn.13380] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 08/03/2016] [Accepted: 08/22/2016] [Indexed: 12/25/2022]
Abstract
The mesocortical and mesolimbic dopamine systems regulate cognitive and motivational processes and are strongly implicated in neuropsychiatric disorders in which these processes are disturbed. Sex differences and sex hormone modulation are also known for these dopamine-sensitive behaviours in health and disease. One relevant mechanism of hormone impact appears to be regulation of cortical and subcortical dopamine levels. This study asked whether this regulation of dopamine tone is a consequence of sex or sex hormone impact on the firing modes of ventral midbrain dopamine neurons. To address this, single unit extracellular recordings made in the ventral tegmental area and substantia nigra were compared among urethane-anaesthetized adult male, female, gonadectomized male rats. These comparisons showed that gonadectomy had no effect on nigral cells and no effects on pacemaker, bursty, single-spiking or random modes of dopamine activity in the ventral tegmental area. However, it did significantly and selectively increase burst firing in these cells in a testosterone-sensitive, estradiol-insensitive manner. Given the roles of prefrontal cortex (PFC) in modulating midbrain dopamine cell firing, we next asked whether gonadectomy's effects on dopamine cell bursting had correlated effects on the activity of ventral tegmentally projecting prefrontal cortical neurons. We found that gonadectomy indeed significantly and selectively increased burst firing in ventral tegmentally projecting but not neighbouring prefrontal cells. These effects were also androgen-sensitive. Together, these findings suggest a working model wherein androgen influence over the activity of PFC neurons regulates its top-down modulation of mesocortical and mesolimbic dopamine systems and related dopamine-sensitive behaviours.
Collapse
Affiliation(s)
- Mallory N Locklear
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, 11794-5230, USA
| | - Michalis Michaelos
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, 11794-5230, USA
| | - William F Collins
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, 11794-5230, USA
| | - Mary F Kritzer
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, 11794-5230, USA
| |
Collapse
|
157
|
Choy KHC, Shackleford DM, Malone DT, Mistry SN, Patil RT, Scammells PJ, Langmead CJ, Pantelis C, Sexton PM, Lane JR, Christopoulos A. Positive Allosteric Modulation of the Muscarinic M1 Receptor Improves Efficacy of Antipsychotics in Mouse Glutamatergic Deficit Models of Behavior. J Pharmacol Exp Ther 2016; 359:354-365. [PMID: 27630144 DOI: 10.1124/jpet.116.235788] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 09/13/2016] [Indexed: 12/14/2022] Open
Abstract
Current antipsychotics are effective in treating the positive symptoms associated with schizophrenia, but they remain suboptimal in targeting cognitive dysfunction. Recent studies have suggested that positive allosteric modulation of the M1 muscarinic acetylcholine receptor (mAChR) may provide a novel means of improving cognition. However, very little is known about the potential of combination therapies in extending coverage across schizophrenic symptom domains. This study investigated the effect of the M1 mAChR positive allosteric modulator BQCA [1-(4-methoxybenzyl)-4-oxo-1,4-dihydroquinoline-3-carboxylic acid], alone or in combination with haloperidol (a first-generation antipsychotic), clozapine (a second-generation atypical antipsychotic), or aripiprazole (a third-generation atypical antipsychotic), in reversing deficits in sensorimotor gating and spatial memory induced by the N-methyl-d-aspartate receptor antagonist, MK-801 [(5R,10S)-(+)-5-methyl-10,11-dihydro-5H-dibenzo[a,d]cyclohepten-5,10-imine]. Sensorimotor gating and spatial memory induction are two models that represent aspects of schizophrenia modeled in rodents. In prepulse inhibition (an operational measure of sensorimotor gating), BQCA alone had minimal effects but exhibited different levels of efficacy in reversing MK-801-induced prepulse inhibition disruptions when combined with a subeffective dose of each of the three (currently prescribed) antipsychotics. Furthermore, the combined effect of BQCA and clozapine was absent in M1-/- mice. Interestingly, although BQCA alone had no effect in reversing MK-801-induced memory impairments in a Y-maze spatial test, we observed a reversal upon the combination of BQCA with atypical antipsychotics, but not with haloperidol. These findings provide proof of concept that a judicious combination of existing antipsychotics with a selective M1 mAChR positive allosteric modulator can extend antipsychotic efficacy in glutamatergic deficit models of behavior.
Collapse
Affiliation(s)
- Kwok H C Choy
- Drug Discovery Biology (K.H.C.C., D.T.M, C.J.L, P.M.S, J.R.L, A.C.), Centre for Drug Candidate Optimization (D.M.S., R.T.P.), and Medicinal Chemistry (S.N.M, P.J.S.), Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia; and Melbourne Neuropsychiatry Centre, Department of Psychiatry and Centre for Neural Engineering, University of Melbourne, Melbourne, Australia (C.P.)
| | - David M Shackleford
- Drug Discovery Biology (K.H.C.C., D.T.M, C.J.L, P.M.S, J.R.L, A.C.), Centre for Drug Candidate Optimization (D.M.S., R.T.P.), and Medicinal Chemistry (S.N.M, P.J.S.), Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia; and Melbourne Neuropsychiatry Centre, Department of Psychiatry and Centre for Neural Engineering, University of Melbourne, Melbourne, Australia (C.P.)
| | - Daniel T Malone
- Drug Discovery Biology (K.H.C.C., D.T.M, C.J.L, P.M.S, J.R.L, A.C.), Centre for Drug Candidate Optimization (D.M.S., R.T.P.), and Medicinal Chemistry (S.N.M, P.J.S.), Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia; and Melbourne Neuropsychiatry Centre, Department of Psychiatry and Centre for Neural Engineering, University of Melbourne, Melbourne, Australia (C.P.)
| | - Shailesh N Mistry
- Drug Discovery Biology (K.H.C.C., D.T.M, C.J.L, P.M.S, J.R.L, A.C.), Centre for Drug Candidate Optimization (D.M.S., R.T.P.), and Medicinal Chemistry (S.N.M, P.J.S.), Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia; and Melbourne Neuropsychiatry Centre, Department of Psychiatry and Centre for Neural Engineering, University of Melbourne, Melbourne, Australia (C.P.)
| | - Rahul T Patil
- Drug Discovery Biology (K.H.C.C., D.T.M, C.J.L, P.M.S, J.R.L, A.C.), Centre for Drug Candidate Optimization (D.M.S., R.T.P.), and Medicinal Chemistry (S.N.M, P.J.S.), Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia; and Melbourne Neuropsychiatry Centre, Department of Psychiatry and Centre for Neural Engineering, University of Melbourne, Melbourne, Australia (C.P.)
| | - Peter J Scammells
- Drug Discovery Biology (K.H.C.C., D.T.M, C.J.L, P.M.S, J.R.L, A.C.), Centre for Drug Candidate Optimization (D.M.S., R.T.P.), and Medicinal Chemistry (S.N.M, P.J.S.), Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia; and Melbourne Neuropsychiatry Centre, Department of Psychiatry and Centre for Neural Engineering, University of Melbourne, Melbourne, Australia (C.P.)
| | - Christopher J Langmead
- Drug Discovery Biology (K.H.C.C., D.T.M, C.J.L, P.M.S, J.R.L, A.C.), Centre for Drug Candidate Optimization (D.M.S., R.T.P.), and Medicinal Chemistry (S.N.M, P.J.S.), Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia; and Melbourne Neuropsychiatry Centre, Department of Psychiatry and Centre for Neural Engineering, University of Melbourne, Melbourne, Australia (C.P.)
| | - Christos Pantelis
- Drug Discovery Biology (K.H.C.C., D.T.M, C.J.L, P.M.S, J.R.L, A.C.), Centre for Drug Candidate Optimization (D.M.S., R.T.P.), and Medicinal Chemistry (S.N.M, P.J.S.), Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia; and Melbourne Neuropsychiatry Centre, Department of Psychiatry and Centre for Neural Engineering, University of Melbourne, Melbourne, Australia (C.P.)
| | - Patrick M Sexton
- Drug Discovery Biology (K.H.C.C., D.T.M, C.J.L, P.M.S, J.R.L, A.C.), Centre for Drug Candidate Optimization (D.M.S., R.T.P.), and Medicinal Chemistry (S.N.M, P.J.S.), Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia; and Melbourne Neuropsychiatry Centre, Department of Psychiatry and Centre for Neural Engineering, University of Melbourne, Melbourne, Australia (C.P.)
| | - Johnathan R Lane
- Drug Discovery Biology (K.H.C.C., D.T.M, C.J.L, P.M.S, J.R.L, A.C.), Centre for Drug Candidate Optimization (D.M.S., R.T.P.), and Medicinal Chemistry (S.N.M, P.J.S.), Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia; and Melbourne Neuropsychiatry Centre, Department of Psychiatry and Centre for Neural Engineering, University of Melbourne, Melbourne, Australia (C.P.)
| | - Arthur Christopoulos
- Drug Discovery Biology (K.H.C.C., D.T.M, C.J.L, P.M.S, J.R.L, A.C.), Centre for Drug Candidate Optimization (D.M.S., R.T.P.), and Medicinal Chemistry (S.N.M, P.J.S.), Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia; and Melbourne Neuropsychiatry Centre, Department of Psychiatry and Centre for Neural Engineering, University of Melbourne, Melbourne, Australia (C.P.)
| |
Collapse
|
158
|
Koga M, Serritella AV, Sawa A, Sedlak TW. Implications for reactive oxygen species in schizophrenia pathogenesis. Schizophr Res 2016; 176:52-71. [PMID: 26589391 DOI: 10.1016/j.schres.2015.06.022] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Revised: 06/20/2015] [Accepted: 06/23/2015] [Indexed: 12/18/2022]
Abstract
Oxidative stress is a well-recognized participant in the pathophysiology of multiple brain disorders, particularly neurodegenerative conditions such as Alzheimer's and Parkinson's diseases. While not a dementia, a wide body of evidence has also been accumulating for aberrant reactive oxygen species and inflammation in schizophrenia. Here we highlight roles for oxidative stress as a common mechanism by which varied genetic and epidemiologic risk factors impact upon neurodevelopmental processes that underlie the schizophrenia syndrome. While there is longstanding evidence that schizophrenia may not have a single causative lesion, a common pathway involving oxidative stress opens the possibility for intervention at susceptible phases.
Collapse
Affiliation(s)
- Minori Koga
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, 600 North Wolfe Street, Meyer 3-166, Baltimore, MD 21287, USA
| | - Anthony V Serritella
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, 600 North Wolfe Street, Meyer 3-166, Baltimore, MD 21287, USA
| | - Akira Sawa
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, 600 North Wolfe Street, Meyer 3-166, Baltimore, MD 21287, USA
| | - Thomas W Sedlak
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, 600 North Wolfe Street, Meyer 3-166, Baltimore, MD 21287, USA.
| |
Collapse
|
159
|
The Influence of the CB1 Receptor Ligands on the Schizophrenia-Like Effects in Mice Induced by MK-801. Neurotox Res 2016; 30:658-676. [PMID: 27577742 PMCID: PMC5047950 DOI: 10.1007/s12640-016-9662-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Revised: 08/12/2016] [Accepted: 08/16/2016] [Indexed: 11/20/2022]
Abstract
A growing body of psychiatric research has emerged, focusing on the role of endocannabinoid system in psychiatric disorders. For example, the endocannabinoid system, via cannabinoid CB (CB1 and CB2) receptors, is able to control the function of many receptors, such as N-methyl-d-aspartate (NMDA) receptors connected strictly with psychosis or other schizophrenia-associated symptoms. The aim of the present research was to investigate the impact of the CB1 receptor ligands on the symptoms typical for schizophrenia. We provoked psychosis-like effects in mice by an acute administration of NMDA receptor antagonist, MK-801 (0.1–0.6 mg/kg). An acute administration of MK-801 induced psychotic symptoms, manifested in the increase in locomotor activity (hyperactivity), measured in actimeters, as well as the memory impairment, assessed in the passive avoidance task. We revealed that an acute injection of CB1 receptor agonist, oleamide (5–20 mg/kg), had no influence on the short- and long-term memory-related disturbances, as well as on the hyperlocomotion in mice, provoking by an acute MK-801. In turn, an amnestic effects or hyperactivity induced by an acute MK-801 was attenuated by an acute administration of AM 251 (0.25–3 mg/kg), a CB1 receptor antagonist. The present findings confirm that endocannabinoid system is able to modify a variety of schizophrenia-like responses, including the cognitive disturbances and hyperlocomotion in mice. Antipsychotic-like effects induced by CB1 receptor antagonist, obtained in our research, confirm the potential effect of CB1 receptor blockade and could have important therapeutic implications on clinical settings, in the future.
Collapse
|
160
|
Thomson PA, Duff B, Blackwood DHR, Romaniuk L, Watson A, Whalley HC, Li X, Dauvermann MR, Moorhead TWJ, Bois C, Ryan NM, Redpath H, Hall L, Morris SW, van Beek EJR, Roberts N, Porteous DJ, St Clair D, Whitcher B, Dunlop J, Brandon NJ, Hughes ZA, Hall J, McIntosh A, Lawrie SM. Balanced translocation linked to psychiatric disorder, glutamate, and cortical structure/function. NPJ SCHIZOPHRENIA 2016; 2:16024. [PMID: 27602385 PMCID: PMC4994153 DOI: 10.1038/npjschz.2016.24] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 06/30/2016] [Accepted: 07/01/2016] [Indexed: 01/01/2023]
Abstract
Rare genetic variants of large effect can help elucidate the pathophysiology of brain disorders. Here we expand the clinical and genetic analyses of a family with a (1;11)(q42;q14.3) translocation multiply affected by major psychiatric illness and test the effect of the translocation on the structure and function of prefrontal, and temporal brain regions. The translocation showed significant linkage (LOD score 6.1) with a clinical phenotype that included schizophrenia, schizoaffective disorder, bipolar disorder, and recurrent major depressive disorder. Translocation carriers showed reduced cortical thickness in the left temporal lobe, which correlated with general psychopathology and positive psychotic symptom severity. They showed reduced gyrification in prefrontal cortex, which correlated with general psychopathology severity. Translocation carriers also showed significantly increased activation in the caudate nucleus on increasing verbal working memory load, as well as statistically significant reductions in the right dorsolateral prefrontal cortex glutamate concentrations. These findings confirm that the t(1;11) translocation is associated with a significantly increased risk of major psychiatric disorder and suggest a general vulnerability to psychopathology through altered cortical structure and function, and decreased glutamate levels.
Collapse
Affiliation(s)
- Pippa A Thomson
- Medical Genetics Section, Centre for Genomic and Experimental Medicine, University of Edinburgh, MRC Institute of Genetics and Molecular Medicine at the University of Edinburgh, Western General Hospital , Edinburgh, UK
| | - Barbara Duff
- Division of Psychiatry, Deanery of Clinical Sciences, University of Edinburgh, Royal Edinburgh Hospital, Morningside Park , Edinburgh, UK
| | - Douglas H R Blackwood
- Division of Psychiatry, Deanery of Clinical Sciences, University of Edinburgh, Royal Edinburgh Hospital, Morningside Park , Edinburgh, UK
| | - Liana Romaniuk
- Division of Psychiatry, Deanery of Clinical Sciences, University of Edinburgh, Royal Edinburgh Hospital, Morningside Park , Edinburgh, UK
| | - Andrew Watson
- Division of Psychiatry, Deanery of Clinical Sciences, University of Edinburgh, Royal Edinburgh Hospital, Morningside Park , Edinburgh, UK
| | - Heather C Whalley
- Division of Psychiatry, Deanery of Clinical Sciences, University of Edinburgh, Royal Edinburgh Hospital, Morningside Park , Edinburgh, UK
| | - Xiang Li
- Clinical Research Imaging Centre (CRIC), The Queen's Medical Research Institute, University of Edinburgh , UK
| | - Maria R Dauvermann
- McGovern Institute for Brain Research, Massachusetts Institute of Technology , Cambridge, MA, USA
| | - T William J Moorhead
- Division of Psychiatry, Deanery of Clinical Sciences, University of Edinburgh, Royal Edinburgh Hospital, Morningside Park , Edinburgh, UK
| | - Catherine Bois
- Division of Psychiatry, Deanery of Clinical Sciences, University of Edinburgh, Royal Edinburgh Hospital, Morningside Park , Edinburgh, UK
| | - Niamh M Ryan
- Medical Genetics Section, Centre for Genomic and Experimental Medicine, University of Edinburgh, MRC Institute of Genetics and Molecular Medicine at the University of Edinburgh, Western General Hospital , Edinburgh, UK
| | - Holly Redpath
- Division of Psychiatry, Deanery of Clinical Sciences, University of Edinburgh, Royal Edinburgh Hospital, Morningside Park , Edinburgh, UK
| | - Lynsey Hall
- Division of Psychiatry, Deanery of Clinical Sciences, University of Edinburgh, Royal Edinburgh Hospital, Morningside Park , Edinburgh, UK
| | - Stewart W Morris
- Medical Genetics Section, Centre for Genomic and Experimental Medicine, University of Edinburgh, MRC Institute of Genetics and Molecular Medicine at the University of Edinburgh, Western General Hospital , Edinburgh, UK
| | - Edwin J R van Beek
- Clinical Research Imaging Centre (CRIC), The Queen's Medical Research Institute, University of Edinburgh , UK
| | - Neil Roberts
- Clinical Research Imaging Centre (CRIC), The Queen's Medical Research Institute, University of Edinburgh , UK
| | - David J Porteous
- Medical Genetics Section, Centre for Genomic and Experimental Medicine, University of Edinburgh, MRC Institute of Genetics and Molecular Medicine at the University of Edinburgh, Western General Hospital , Edinburgh, UK
| | - David St Clair
- Institute of Medical Sciences, University of Aberdeen , Aberdeen, UK
| | - Brandon Whitcher
- Clinical & Translational Imaging Group, Pfizer Global Research , Cambridge, MA, USA
| | - John Dunlop
- Neuroscience Research Unit, Pfizer Global Research, Cambridge, MA, USA; AstraZeneca Neuroscience, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Cambridge, MA, USA
| | - Nicholas J Brandon
- Neuroscience Research Unit, Pfizer Global Research, Cambridge, MA, USA; AstraZeneca Neuroscience, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Cambridge, MA, USA
| | - Zoë A Hughes
- Neuroscience Research Unit, Pfizer Global Research , Cambridge, MA, USA
| | - Jeremy Hall
- Neuroscience and Mental Health Research Institute, Cardiff University, Hadyn Ellis Building , Cardiff, UK
| | - Andrew McIntosh
- Division of Psychiatry, Deanery of Clinical Sciences, University of Edinburgh, Royal Edinburgh Hospital, Morningside Park , Edinburgh, UK
| | - Stephen M Lawrie
- Division of Psychiatry, Deanery of Clinical Sciences, University of Edinburgh, Royal Edinburgh Hospital, Morningside Park , Edinburgh, UK
| |
Collapse
|
161
|
Demir S, Bulut M, Atli A, Kaplan İ, Kaya MC, Bez Y, Özdemir PG, Sır A. Decreased Prolidase Activity in Patients with Posttraumatic Stress Disorder. Psychiatry Investig 2016; 13:420-6. [PMID: 27482243 PMCID: PMC4965652 DOI: 10.4306/pi.2016.13.4.420] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 09/06/2015] [Accepted: 09/17/2015] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE Many neurochemical systems have been implicated in the development of Posttraumatic Stress Disorder (PTSD). The prolidase enzyme is a cytosolic exopeptidase that detaches proline or hydroxyproline from the carboxyl terminal position of dipeptides. Prolidase has important biological effects, and to date, its role in the etiology of PTSD has not been studied. In the present study, we aimed to evaluate prolidase activity in patients with PTSD. METHODS The study group consisted of patients who were diagnosed with PTSD after the earthquake that occurred in the province of Van in Turkey in 2011 (n=25); the first control group consisted of patients who experienced the earthquake but did not show PTSD symptoms (n=26) and the second control group consisted of patients who have never been exposed to a traumatic event (n=25). Prolidase activities in the patients and the control groups were determined by the ELISA method using commercial kits. RESULTS Prolidase activity in the patient group was significantly lower when compared to the control groups. Prolidase activity was also significantly lower in the traumatized healthy subjects compared to the other healthy group (p<0.01). CONCLUSION The findings of the present study suggest that the decrease in prolidase activity may have neuroprotective effects in patients with PTSD.
Collapse
Affiliation(s)
- Süleyman Demir
- Department of Psychiatry, Dicle University, Diyarbakir, Turkey
| | - Mahmut Bulut
- Department of Psychiatry, Dicle University, Diyarbakir, Turkey
| | - Abdullah Atli
- Department of Psychiatry, Dicle University, Diyarbakir, Turkey
| | - İbrahim Kaplan
- Department of Biochemistry, Dicle University, Diyarbakir, Turkey
| | | | - Yasin Bez
- Department of Psychiatry, Marmara University, Istanbul, Turkey
| | | | - Aytekin Sır
- Department of Psychiatry, Dicle University, Diyarbakir, Turkey
| |
Collapse
|
162
|
Lieberman JA, Davis RE, Correll CU, Goff DC, Kane JM, Tamminga CA, Mates S, Vanover KE. ITI-007 for the Treatment of Schizophrenia: A 4-Week Randomized, Double-Blind, Controlled Trial. Biol Psychiatry 2016; 79:952-61. [PMID: 26444072 DOI: 10.1016/j.biopsych.2015.08.026] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Revised: 08/14/2015] [Accepted: 08/17/2015] [Indexed: 01/03/2023]
Abstract
BACKGROUND An urgent need exists for new treatments of schizophrenia that are effective against a broad range of symptoms and free of limiting safety issues. ITI-007 is a new molecular entity with a pharmacologic profile that combines dose-related monoamine modulation with phosphorylation of intracellular signaling proteins. METHODS A phase II randomized, double-blind, placebo-controlled, and active-controlled trial was conducted at eight sites in the United States with randomization of 335 acutely psychotic adults with schizophrenia. ITI-007 (60 mg and 120 mg), placebo, and risperidone, included for assay sensitivity, were evaluated as monotherapy for 4 weeks. The primary outcome measure was the Positive and Negative Syndrome Scale total score, with secondary analyses conducted on symptom subscales. RESULTS ITI-007 60 mg (p = .017, effect size = .4) and risperidone (p = .013, effect size = .4) demonstrated antipsychotic efficacy superiority over placebo on the primary end point. The results of secondary analyses reflected improvements in negative and depressive symptoms by ITI-007 60 mg. ITI-007 120 mg did not separate from placebo. However, both doses of ITI-007 were well tolerated in this patient population, as evidenced by low discontinuation and adverse event rates, and were associated with a benign metabolic profile as evidenced by significantly lower levels of prolactin, fasting glucose, total cholesterol, and triglycerides than risperidone. CONCLUSIONS The mechanistically novel investigational drug ITI-007 was effective for the treatment of schizophrenia and comparable with placebo on safety measures in this trial. Secondary analyses indicated that ITI-007 improved negative and depression symptoms and might have expanded therapeutic efficacy in comparison with current antipsychotic drugs.
Collapse
Affiliation(s)
- Jeffrey A Lieberman
- Department of Psychiatry, Columbia University College of Physicians and Surgeons, New York Presbyterian Hospital, New York, New York
| | - Robert E Davis
- Intra-Cellular Therapies, Inc., New York, New York; 3-D Pharmaceutical Consultants, San Diego, California
| | - Christoph U Correll
- Department of Psychiatry, Hofstra North Shore Long Island Jewish School of Medicine, Hempstead; Zucker Hillside Hospital Glen Oaks, New York
| | - Donald C Goff
- Department of Psychiatry, New York University Langone Medical Center, New York; Nathan Kline Institute for Psychiatric Research, Orangeburg, New York
| | - John M Kane
- Department of Psychiatry, Hofstra North Shore Long Island Jewish School of Medicine, Hempstead; Zucker Hillside Hospital Glen Oaks, New York
| | - Carol A Tamminga
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Sharon Mates
- Intra-Cellular Therapies, Inc., New York, New York
| | | |
Collapse
|
163
|
Stanciu CN, Penders TM, Rouse EM. Recreational use of dextromethorphan, “Robotripping”-A brief review. Am J Addict 2016; 25:374-7. [DOI: 10.1111/ajad.12389] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Revised: 05/04/2016] [Accepted: 05/14/2016] [Indexed: 11/27/2022] Open
Affiliation(s)
- Cornel N. Stanciu
- Department of Psychiatric Medicine, Brody School of Medicine; East Carolina University; Greenville North Carolina
| | - Thomas M. Penders
- Department of Psychiatric Medicine, Brody School of Medicine; East Carolina University; Greenville North Carolina
| | - Eden M. Rouse
- Department of Psychiatric Medicine, Brody School of Medicine; East Carolina University; Greenville North Carolina
| |
Collapse
|
164
|
Bhakta SG, Chou HH, Rana B, Talledo JA, Balvaneda B, Gaddis L, Light GA, Swerdlow NR. Effects of acute memantine administration on MATRICS Consensus Cognitive Battery performance in psychosis: Testing an experimental medicine strategy. Psychopharmacology (Berl) 2016; 233:2399-410. [PMID: 27076209 PMCID: PMC5214934 DOI: 10.1007/s00213-016-4291-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 04/01/2016] [Indexed: 02/06/2023]
Abstract
RATIONALE Pro-cognitive agents for chronic psychotic disorders (CPDs) might be detected via experimental medicine models, in which neural targets engaged by the drug predict sensitivity to the drug's pro-cognitive effects. OBJECTIVE This study aims to use an experimental medicine model to test the hypothesis that "target engagement" predicts pro-cognitive effects of the NMDA antagonist, memantine (MEM), in CPDs. METHODS MATRICS Consensus Cognitive Battery (MCCB) performance was assessed in CPD (n = 41) and healthy subjects (HS; n = 41) in a double-blind, randomized cross-over design of acute (single dose) MEM (placebo vs. 10 or 20 mg p.o.). Measures of prepulse inhibition (PPI) and mismatch negativity previously reported from this cohort substantiated target engagement. Biomarkers predicting MEM neurocognitive sensitivity were assessed. RESULTS Testing confirmed MCCB deficits associated with CPD diagnosis, age, and anticholinergic exposure. MEM (20 mg p.o.) reduced MCCB performance in HS. To control for significant test order effects, an "order-corrected MEM effect" (OCME) was calculated. In CPD subjects, greater age, positive MEM effects on PPI, and SNP rs1337697 (within the ionotropic NMDA receptor gene, GRIN3A) predicted greater positive OCME with 20 mg MEM. CONCLUSIONS An experimental medicine model to assess acute pro-cognitive drug effects in CPD subjects is feasible but not without challenges. A single MEM 20 mg dose had a negative impact on neurocognition among HS. In CPD patients, age, MEM effects on PPI, and rs1337697 predicted sensitivity to the neurocognitive effects of MEM. Any potential clinical utility of these predictive markers for pro-cognitive effects of MEM in subgroups of CPD patients cannot be inferred without a validating clinical trial.
Collapse
Affiliation(s)
- Savita G Bhakta
- Department of Psychiatry, UC San Diego School of Medicine, 9500 Gilman Dr., MC 0804, La Jolla, CA, 92093-0804, USA
| | - Hsun-Hua Chou
- Department of Psychiatry, UC San Diego School of Medicine, 9500 Gilman Dr., MC 0804, La Jolla, CA, 92093-0804, USA
| | - Brinda Rana
- Department of Psychiatry, UC San Diego School of Medicine, 9500 Gilman Dr., MC 0804, La Jolla, CA, 92093-0804, USA
| | - Jo A Talledo
- Department of Psychiatry, UC San Diego School of Medicine, 9500 Gilman Dr., MC 0804, La Jolla, CA, 92093-0804, USA
| | - Bryan Balvaneda
- Department of Psychiatry, UC San Diego School of Medicine, 9500 Gilman Dr., MC 0804, La Jolla, CA, 92093-0804, USA
| | - Laura Gaddis
- Department of Psychiatry, UC San Diego School of Medicine, 9500 Gilman Dr., MC 0804, La Jolla, CA, 92093-0804, USA
| | - Gregory A Light
- Department of Psychiatry, UC San Diego School of Medicine, 9500 Gilman Dr., MC 0804, La Jolla, CA, 92093-0804, USA
| | - Neal R Swerdlow
- Department of Psychiatry, UC San Diego School of Medicine, 9500 Gilman Dr., MC 0804, La Jolla, CA, 92093-0804, USA.
| |
Collapse
|
165
|
Mouchlianitis E, Bloomfield MAP, Law V, Beck K, Selvaraj S, Rasquinha N, Waldman A, Turkheimer FE, Egerton A, Stone J, Howes OD. Treatment-Resistant Schizophrenia Patients Show Elevated Anterior Cingulate Cortex Glutamate Compared to Treatment-Responsive. Schizophr Bull 2016; 42:744-52. [PMID: 26683625 PMCID: PMC4838083 DOI: 10.1093/schbul/sbv151] [Citation(s) in RCA: 162] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
INTRODUCTION Resistance to antipsychotic treatment is a significant clinical problem in patients with schizophrenia with approximately 1 in 3 showing limited or no response to repeated treatments with antipsychotic medication. The neurobiological basis for treatment resistance is unknown but recent evidence implicates glutamatergic function in the anterior cingulate cortex. We examined glutamate levels of chronically ill treatment-resistant patients directly compared to treatment-responsive patients. METHODS We acquired proton magnetic resonance spectroscopy (1H-MRS) at 3 Tesla from 21 treatment-resistant and 20 treatment-responsive patients. All participants had a DSM-IV diagnosis of schizophrenia. Treatment-resistant patients were classified using the modified Kane criteria. The groups were matched for age, sex, smoking status, and illness duration. RESULTS Glutamate to creatine ratio levels were higher in treatment-resistant patients (Mean [SD] = 1.57 [0.24]) than in treatment-responsive patients (Mean[SD] = 1.38 [0.23]), (T[35] = 2.34, P = .025, 2-tailed), with a large effect size of d = 0.76. A model assuming 2 populations showed a 25% improvement in the fit of the Akaike weights (0.55) over a model assuming 1 population (0.44), producing group values almost identical to actual group means. DISCUSSION Increased anterior cingulate glutamate level is associated with treatment-resistant schizophrenia. This appears to be a stable neurobiological trait of treatment-resistant patients. We discuss possible explanations for glutamatergic dysfunction playing a significant role in resistance to conventional antipsychotic treatments, which are all dopamine-2 receptor blockers. Our findings suggest that glutamatergic treatments may be particularly effective in resistant patients and that 1H-MRS glutamate indices can potentially have clinical use.
Collapse
Affiliation(s)
- Elias Mouchlianitis
- Medical Research Council Clinical Sciences Centre, Psychiatric Imaging Group, Hammersmith Hospital, London, UK; Institute of Psychiatry Psychology and Neuroscience, Department of Psychosis Studies, King's College London, UK;
| | - Michael A. P. Bloomfield
- Medical Research Council Clinical Sciences Centre, Psychiatric Imaging Group, Hammersmith Hospital, London, UK;,University College London, Division of Psychiatry, London, UK
| | - Vincent Law
- Medical Research Council Clinical Sciences Centre, Psychiatric Imaging Group, Hammersmith Hospital, London, UK
| | - Katherine Beck
- Institute of Psychiatry Psychology and Neuroscience, Department of Psychosis Studies, King’s College London, UK
| | - Sudhakar Selvaraj
- Department of Psychiatry and Behavioral Sciences, University of Texas, Houston, TX
| | | | - Adam Waldman
- Division of Brain Sciences, Imperial College London, Hammersmith Hospital, London, UK
| | - Federico E. Turkheimer
- Institute of Psychiatry Psychology and Neuroscience, Department of Psychosis Studies, King’s College London, UK
| | - Alice Egerton
- Institute of Psychiatry Psychology and Neuroscience, Department of Psychosis Studies, King’s College London, UK
| | - James Stone
- Medical Research Council Clinical Sciences Centre, Psychiatric Imaging Group, Hammersmith Hospital, London, UK;,Institute of Psychiatry Psychology and Neuroscience, Department of Psychosis Studies, King’s College London, UK
| | - Oliver D. Howes
- Medical Research Council Clinical Sciences Centre, Psychiatric Imaging Group, Hammersmith Hospital, London, UK;,Institute of Psychiatry Psychology and Neuroscience, Department of Psychosis Studies, King’s College London, UK
| |
Collapse
|
166
|
Gragnoli C, Reeves GM, Reazer J, Postolache TT. Dopamine-prolactin pathway potentially contributes to the schizophrenia and type 2 diabetes comorbidity. Transl Psychiatry 2016; 6:e785. [PMID: 27093067 PMCID: PMC4872408 DOI: 10.1038/tp.2016.50] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 02/15/2016] [Accepted: 02/19/2016] [Indexed: 01/10/2023] Open
Abstract
Schizophrenia (SCZ) and type 2 diabetes (T2D) are clinically associated, and common knowledge attributes this association to side effects of antipsychotic treatment. However, even drug-naive patients with SCZ are at increased risk for T2D. Dopamine dysfunction has a central role in SCZ. It is well-known that dopamine constitutively inhibits prolactin (PRL) secretion via the dopamine receptor 2 (DR2D). If dopamine is increased or if dopamine receptors hyperfunction, PRL may be reduced. During the first SCZ episode, low PRL levels are associated with worse symptoms. PRL is essential in human and social bonding, as well as it is implicated in glucose homeostasis. Dopamine dysfunction, beyond contributing to SCZ symptoms, may lead to altered appetite and T2D. To our knowledge, there are no studies of the genetics of the SCZ-T2D comorbidity focusing jointly on the dopamine and PRL pathway in the attempt to capture molecular heterogeneity correlated to possible disease manifestation heterogeneity. In this dopamine-PRL pathway-focused-hypothesis-driven review on the association of SCZ with T2D, we report a specific revision of what it is known about PRL and dopamine in relation to what we theorize is one of the missing links between the two disorders. We suggest that new studies are necessary to establish the genetic role of PRL and dopamine pathway in SCZ-T2D comorbidity.
Collapse
Affiliation(s)
- C Gragnoli
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Florida College of Medicine, Jacksonville, FL, USA,Department of Public Health Sciences, Penn State College of Medicine, Hershey, PA, USA,Molecular Biology Laboratory, Bios Biotech Multi-Diagnostic Health Center, Rome, Italy,Division of Endocrinology, Diabetes, and Metabolism, University of Florida College of Medicine, 653-1 West 8th Street, Learning Resource Center, L14, Jacksonville, FL 32209, USA. E-mail:
| | - G M Reeves
- Division of Child and Adolescent Psychiatry, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - J Reazer
- Borland Health Sciences Library, University of Florida, Jacksonville, FL, USA
| | - T T Postolache
- Rocky Mountain Mental Illness Research Education and Clinical Center, Denver, CO, USA,Veterans Integrated Service Network 5 MIRECC, Baltimore, MD, USA,Department of Psychiatry, University of Maryland, Baltimore, MD, USA
| |
Collapse
|
167
|
Physical activity and exercise attenuate neuroinflammation in neurological diseases. Brain Res Bull 2016; 125:19-29. [PMID: 27021169 DOI: 10.1016/j.brainresbull.2016.03.012] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 03/17/2016] [Accepted: 03/22/2016] [Indexed: 12/15/2022]
Abstract
Major depressive disorder (MDD), schizophrenia (SCH), Alzheimer's disease (AD), and Parkinson's disease (PD) are devastating neurological disorders, which increasingly contribute to global morbidity and mortality. Although the pathogenic mechanisms of these conditions are quite diverse, chronic neuroinflammation is one underlying feature shared by all these diseases. Even though the specific root causes of these diseases remain to be identified, evidence indicates that the observed neuroinflammation is initiated by unique pathological features associated with each specific disease. If the initial acute inflammation is not resolved, a chronic neuroinflammatory state develops and ultimately contributes to disease progression. Chronic neuroinflammation is characterized by adverse and non-specific activation of glial cells, which can lead to collateral damage of nearby neurons and other glia. This misdirected neuroinflammatory response is hypothesized to contribute to neuropathology in MDD, SCH, AD, and PD. Physical activity (PA), which is critical for maintenance of whole body and brain health, may also beneficially modify neuroimmune responses. Since PA has neuroimmune-modifying properties, and the common underlying feature of MDD, SCH, AD, and PD is chronic neuroinflammation, we hypothesize that PA could minimize brain diseases by modifying glia-mediated neuroinflammation. This review highlights current evidence supporting the disease-altering potential of PA and exercise through modifications of neuroimmune responses, specifically in MDD, SCH, AD and PD.
Collapse
|
168
|
Leung C, Jia Z. Mouse Genetic Models of Human Brain Disorders. Front Genet 2016; 7:40. [PMID: 27047540 PMCID: PMC4803727 DOI: 10.3389/fgene.2016.00040] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 03/08/2016] [Indexed: 01/29/2023] Open
Abstract
Over the past three decades, genetic manipulations in mice have been used in neuroscience as a major approach to investigate the in vivo function of genes and their alterations. In particular, gene targeting techniques using embryonic stem cells have revolutionized the field of mammalian genetics and have been at the forefront in the generation of numerous mouse models of human brain disorders. In this review, we will first examine childhood developmental disorders such as autism, intellectual disability, Fragile X syndrome, and Williams-Beuren syndrome. We will then explore psychiatric disorders such as schizophrenia and lastly, neurodegenerative disorders including Alzheimer’s disease and Parkinson’s disease. We will outline the creation of these mouse models that range from single gene deletions, subtle point mutations to multi-gene manipulations, and discuss the key behavioral phenotypes of these mice. Ultimately, the analysis of the models outlined in this review will enhance our understanding of the in vivo role and underlying mechanisms of disease-related genes in both normal brain function and brain disorders, and provide potential therapeutic targets and strategies to prevent and treat these diseases.
Collapse
Affiliation(s)
- Celeste Leung
- The Hospital for Sick Children, Program in Neurosciences and Mental Health, Peter Gilgan Centre for Research and Learning, TorontoON, Canada; Program in Physiology, University of Toronto, TorontoON, Canada
| | - Zhengping Jia
- The Hospital for Sick Children, Program in Neurosciences and Mental Health, Peter Gilgan Centre for Research and Learning, TorontoON, Canada; Program in Physiology, University of Toronto, TorontoON, Canada
| |
Collapse
|
169
|
Pitsikas N. Constituents of Saffron (Crocus sativus L.) as Potential Candidates for the Treatment of Anxiety Disorders and Schizophrenia. Molecules 2016; 21:303. [PMID: 26950102 PMCID: PMC6273654 DOI: 10.3390/molecules21030303] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 02/10/2016] [Accepted: 02/29/2016] [Indexed: 02/07/2023] Open
Abstract
Anxiety disorders and schizophrenia are common public health issues. The dried stigma of the plant Crocus sativus L., (C. sativus) commonly known as saffron are used in folk medicine for various purposes. Several lines of evidence suggest that C. sativus, crocins and safranal are implicated in anxiety and schizophrenia. Here, I intend to critically review advances in research of these emerging molecules for the treatment of anxiety and schizophrenia, discuss their advantages over currently used anxiolytics and neuroleptics, as well remaining challenges. Current analysis shows that C. sativus and its components might be a promising class of compounds for the treatment of the above mentioned psychiatric diseases.
Collapse
Affiliation(s)
- Nikolaos Pitsikas
- Department of Pharmacology, School of Medicine, Faculty of Health Sciences, University of Thessaly, Panepistimiou 3 (Biopolis), Larissa 41500, Greece.
| |
Collapse
|
170
|
Hayrynen LK, Hamm JP, Sponheim SR, Clementz BA. Frequency-specific disruptions of neuronal oscillations reveal aberrant auditory processing in schizophrenia. Psychophysiology 2016; 53:786-95. [PMID: 26933842 DOI: 10.1111/psyp.12635] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 02/03/2016] [Indexed: 01/26/2023]
Abstract
Individuals with schizophrenia exhibit abnormalities in evoked brain responses in oddball paradigms. These could result from (a) insufficient salience-related cortical signaling (P300), (b) insufficient suppression of irrelevant aspects of the auditory environment, or (c) excessive neural noise. We tested whether disruption of ongoing auditory steady-state responses at predetermined frequencies informed which of these issues contribute to auditory stimulus relevance processing abnormalities in schizophrenia. Magnetoencephalography data were collected for 15 schizophrenia and 15 healthy subjects during an auditory oddball paradigm (25% targets; 1-s interstimulus interval). Auditory stimuli (pure tones: 1 kHz standards, 2 kHz targets) were administered during four continuous background (auditory steady-state) stimulation conditions: (1) no stimulation, (2) 24 Hz, (3) 40 Hz, and (4) 88 Hz. The modulation of the auditory steady-state response (aSSR) and the evoked responses to the transient stimuli were quantified and compared across groups. In comparison to healthy participants, the schizophrenia group showed greater disruption of the ongoing aSSR by targets regardless of steady-state frequency, and reduced amplitude of both M100 and M300 event-related field components. During the no-stimulation condition, schizophrenia patients showed accentuation of left hemisphere 40 Hz response to both standard and target stimuli, indicating an effort to enhance local stimulus processing. Together, these findings suggest abnormalities in auditory stimulus relevance processing in schizophrenia patients stem from insufficient amplification of salient stimuli.
Collapse
Affiliation(s)
- Lauren K Hayrynen
- Departments of Psychology and Neuroscience, University of Georgia, Athens, Georgia, USA
| | - Jordan P Hamm
- Departments of Psychology and Neuroscience, University of Georgia, Athens, Georgia, USA.,Department of Biological Sciences, Columbia University, New York, New York, USA
| | - Scott R Sponheim
- Minneapolis VA Health Care System, Minneapolis, Minnesota, USA.,Department of Psychiatry, University of Minnesota, Minneapolis, Minnesota, USA
| | - Brett A Clementz
- Departments of Psychology and Neuroscience, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
171
|
Granger KT, Moran PM, Buckley MG, Haselgrove M. Enhanced latent inhibition in high schizotypy individuals. PERSONALITY AND INDIVIDUAL DIFFERENCES 2016. [DOI: 10.1016/j.paid.2015.11.040] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
172
|
Lindberg D, Shan D, Ayers-Ringler J, Oliveros A, Benitez J, Prieto M, McCullumsmith R, Choi DS. Purinergic signaling and energy homeostasis in psychiatric disorders. Curr Mol Med 2016; 15:275-95. [PMID: 25950756 DOI: 10.2174/1566524015666150330163724] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Revised: 02/22/2015] [Accepted: 03/24/2015] [Indexed: 12/11/2022]
Abstract
Purinergic signaling regulates numerous vital biological processes in the central nervous system (CNS). The two principle purines, ATP and adenosine act as excitatory and inhibitory neurotransmitters, respectively. Compared to other classical neurotransmitters, the role of purinergic signaling in psychiatric disorders is not well understood or appreciated. Because ATP exerts its main effect on energy homeostasis, neuronal function of ATP has been underestimated. Similarly, adenosine is primarily appreciated as a precursor of nucleotide synthesis during active cell growth and division. However, recent findings suggest that purinergic signaling may explain how neuronal activity is associated neuronal energy charge and energy homeostasis, especially in mental disorders. In this review, we provide an overview of the synaptic function of mitochondria and purines in neuromodulation, synaptic plasticity, and neuron-glia interactions. We summarize how mitochondrial and purinergic dysfunction contribute to mental illnesses such as schizophrenia, bipolar disorder, autism spectrum disorder (ASD), depression, and addiction. Finally, we discuss future implications regarding the pharmacological targeting of mitochondrial and purinergic function for the treatment of psychiatric disorders.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - D-S Choi
- Neurobiology of Disease Program, Mayo Clinic College of Medicine, Rochester, MN 55905, USA.
| |
Collapse
|
173
|
Vitucci D, Di Giorgio A, Napolitano F, Pelosi B, Blasi G, Errico F, Attrotto MT, Gelao B, Fazio L, Taurisano P, Di Maio A, Marsili V, Pasqualetti M, Bertolino A, Usiello A. Rasd2 Modulates Prefronto-Striatal Phenotypes in Humans and 'Schizophrenia-Like Behaviors' in Mice. Neuropsychopharmacology 2016; 41:916-27. [PMID: 26228524 PMCID: PMC4707838 DOI: 10.1038/npp.2015.228] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 07/03/2015] [Accepted: 07/25/2015] [Indexed: 12/18/2022]
Abstract
Rasd2 is a thyroid hormone target gene, which encodes for a GTP-binding protein enriched in the striatum where, among other functions, it modulates dopaminergic neurotransmission. Here we report that human RASD2 mRNA is abundant in putamen, but it also occurs in the cerebral cortex, with a distinctive expression pattern that differs from that present in rodents. Consistent with its localization, we found that a genetic variation in RASD2 (rs6518956) affects postmortem prefrontal mRNA expression in healthy humans and is associated with phenotypes of relevance to schizophrenia, including prefrontal and striatal grey matter volume and physiology during working memory, as measured with magnetic resonance imaging. Interestingly, quantitative real-time PCR analysis indicated that RASD2 mRNA is slightly reduced in postmortem prefrontal cortex of patients with schizophrenia. In the attempt to uncover the neurobiological substrates associated with Rasd2 activity, we used knockout mice to analyze the in vivo influence of this G-protein on the prepulse inhibition of the startle response and psychotomimetic drug-related behavioral response. Data showed that Rasd2 mutants display deficits in basal prepulse inhibition that, in turn, exacerbate gating disruption under psychotomimetic drug challenge. Furthermore, we documented that lack of Rasd2 strikingly enhances the behavioral sensitivity to motor stimulation elicited by amphetamine and phencyclidine. Based on animal model data, along with the finding that RASD2 influences prefronto-striatal phenotypes in healthy humans, we suggest that genetic mutation or reduced levels of this G-protein might have a role in cerebral circuitry dysfunction underpinning exaggerated psychotomimetic drugs responses and development of specific biological phenotypes linked to schizophrenia.
Collapse
Affiliation(s)
- Daniela Vitucci
- Ceinge Biotecnologie Avanzate, Naples, Italy,Dipartimento di Scienze Motorie e del Benessere DiSMeB, Università degli Studi di Napoli Parthenope, Naples, Italy
| | - Annabella Di Giorgio
- Istituto di Ricovero e Cura a Carattere Scientifico ‘Casa Sollievo della Sofferenza', Foggia, Italy
| | - Francesco Napolitano
- Ceinge Biotecnologie Avanzate, Naples, Italy,Department of Molecular Medicine and Medical Biotechnology, University of Naples ‘Federico II', Naples, Italy
| | - Barbara Pelosi
- Department of Biology, Unit of Cell and Developmental Biology, University of Pisa, Pisa, Italy
| | - Giuseppe Blasi
- Group of Psychiatric Neuroscience, Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari ‘Aldo Moro', Bari, Italy
| | - Francesco Errico
- Ceinge Biotecnologie Avanzate, Naples, Italy,Department of Molecular Medicine and Medical Biotechnology, University of Naples ‘Federico II', Naples, Italy
| | - Maria Teresa Attrotto
- Group of Psychiatric Neuroscience, Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari ‘Aldo Moro', Bari, Italy
| | - Barbara Gelao
- Group of Psychiatric Neuroscience, Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari ‘Aldo Moro', Bari, Italy
| | - Leonardo Fazio
- Group of Psychiatric Neuroscience, Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari ‘Aldo Moro', Bari, Italy
| | - Paolo Taurisano
- Group of Psychiatric Neuroscience, Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari ‘Aldo Moro', Bari, Italy
| | | | | | - Massimo Pasqualetti
- Department of Biology, Unit of Cell and Developmental Biology, University of Pisa, Pisa, Italy,Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems, Rovereto (Trento), Italy
| | - Alessandro Bertolino
- Group of Psychiatric Neuroscience, Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari ‘Aldo Moro', Bari, Italy,pRED, Neuroscience DTA, Hoffmann-La Roche, Basel, Switzerland,Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari ‘Aldo Moro', Piazza G. Cesare 11, Bari 70124, Italy, Tel: +39 0805478572, Fax: +39 0805593172,
| | - Alessandro Usiello
- Ceinge Biotecnologie Avanzate, Naples, Italy,Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, Second University of Naples (SUN), Caserta, Italy,Ceinge Biotecnologie Avanzate, Via G. Salvatore 486, Naples 80145, Italy, Tel: +39 0813737899, Fax: +39 0813737808. E-mail:
| |
Collapse
|
174
|
Güneş M, Bulut M, Demir S, İbiloğlu AO, Kaya MC, Atlı A, Kaplan İ, Camkurt MA, Sir A. Diagnostic performance of increased prolidase activity in schizophrenia. Neurosci Lett 2016; 613:36-40. [DOI: 10.1016/j.neulet.2015.12.036] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Revised: 11/22/2015] [Accepted: 12/17/2015] [Indexed: 12/22/2022]
|
175
|
Locklear MN, Cohen AB, Jone A, Kritzer MF. Sex Differences Distinguish Intracortical Glutamate Receptor-Mediated Regulation of Extracellular Dopamine Levels in the Prefrontal Cortex of Adult Rats. Cereb Cortex 2016; 26:599-610. [PMID: 25260707 PMCID: PMC4712796 DOI: 10.1093/cercor/bhu222] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Executive functions of the prefrontal cortex (PFC) are sensitive to local dopamine (DA) levels. Although sex differences distinguish these functions and their dysfunction in disease, the basis for this is unknown. We asked whether sex differences might result from dimorphisms in the glutamatergic mechanisms that regulate PFC DA levels. Using antagonists selective for α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) and N-methyl-d-aspartate (NMDA) receptors, we compared drug effects on in vivo microdialysis DA measurements in the PFC of adult male and female rats. We found that baseline DA levels were similar across sex, AMPA antagonism decreased PFC DA in both sexes, and NMDA antagonism increased DA in males but decreased DA in females. We also found that, at subseizure-producing drug levels, γ-aminobutyric acid (GABA)-A antagonism did not affect DA in either sex but that GABA-B antagonism transiently increased PFC DA in both sexes, albeit more so in females. Finally, when NMDA antagonism was coincident with GABA-B antagonism, PFC DA levels in males responded as if to GABA-B antagonism alone, whereas in females, DA effects mirrored those induced by NMDA antagonism. Taken together, these data suggest commonalities and fundamental differences in the intracortical amino acid transmitter mechanisms that regulate DA homeostasis in the male and female rat PFCs.
Collapse
Affiliation(s)
- M N Locklear
- Graduate Program in Neuroscience Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY 11794-5230, USA
| | - A B Cohen
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY 11794-5230, USA
| | - A Jone
- Graduate Program in Neuroscience Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY 11794-5230, USA
| | - M F Kritzer
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY 11794-5230, USA
| |
Collapse
|
176
|
Ahnaou A, Biermans R, Drinkenburg WH. Modulation of mGlu2 Receptors, but Not PDE10A Inhibition Normalizes Pharmacologically-Induced Deviance in Auditory Evoked Potentials and Oscillations in Conscious Rats. PLoS One 2016; 11:e0147365. [PMID: 26808689 PMCID: PMC4726622 DOI: 10.1371/journal.pone.0147365] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 01/04/2016] [Indexed: 12/20/2022] Open
Abstract
Improvement of cognitive impairments represents a high medical need in the development of new antipsychotics. Aberrant EEG gamma oscillations and reductions in the P1/N1 complex peak amplitude of the auditory evoked potential (AEP) are neurophysiological biomarkers for schizophrenia that indicate disruption in sensory information processing. Inhibition of phosphodiesterase (i.e. PDE10A) and activation of metabotropic glutamate receptor (mGluR2) signaling are believed to provide antipsychotic efficacy in schizophrenia, but it is unclear whether this occurs with cognition-enhancing potential. The present study used the auditory paired click paradigm in passive awake Sprague Dawley rats to 1) model disruption of AEP waveforms and oscillations as observed in schizophrenia by peripheral administration of amphetamine and the N-methyl-D-aspartate (NMDA) antagonist phencyclidine (PCP); 2) confirm the potential of the antipsychotics risperidone and olanzapine to attenuate these disruptions; 3) evaluate the potential of mGluR2 agonist LY404039 and PDE10 inhibitor PQ-10 to improve AEP deficits in both the amphetamine and PCP models. PCP and amphetamine disrupted auditory information processing to the first click, associated with suppression of the P1/N1 complex peak amplitude, and increased cortical gamma oscillations. Risperidone and olanzapine normalized PCP and amphetamine-induced abnormalities in AEP waveforms and aberrant gamma/alpha oscillations, respectively. LY404039 increased P1/N1 complex peak amplitudes and potently attenuated the disruptive effects of both PCP and amphetamine on AEPs amplitudes and oscillations. However, PQ-10 failed to show such effect in either models. These outcomes indicate that modulation of the mGluR2 results in effective restoration of abnormalities in AEP components in two widely used animal models of psychosis, whereas PDE10A inhibition does not.
Collapse
Affiliation(s)
- Abdallah Ahnaou
- Department of Neuroscience, Janssen Research & Development, A Division of Janssen Pharmaceutica N.V., Turnhoutseweg 30, B-2340 Beerse, Belgium
- * E-mail:
| | - Ria Biermans
- Department of Neuroscience, Janssen Research & Development, A Division of Janssen Pharmaceutica N.V., Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Wilhelmus H. Drinkenburg
- Department of Neuroscience, Janssen Research & Development, A Division of Janssen Pharmaceutica N.V., Turnhoutseweg 30, B-2340 Beerse, Belgium
| |
Collapse
|
177
|
Zhang Q, Yu Y, Huang XF. Olanzapine Prevents the PCP-induced Reduction in the Neurite Outgrowth of Prefrontal Cortical Neurons via NRG1. Sci Rep 2016; 6:19581. [PMID: 26781398 PMCID: PMC4726088 DOI: 10.1038/srep19581] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 12/15/2015] [Indexed: 11/09/2022] Open
Abstract
Accumulating evidence suggests that reducing neurite outgrowth and synaptic plasticity plays a critical role in the pathology of cognitive deficits in schizophrenia. The N-methyl-D-aspartate receptor antagonist phencyclidine (PCP) can induce symptoms of schizophrenia as well as reduce dendritic spine density and neurite growth. The antipsychotic drug olanzapine may improve these deficits. This study aimed to investigate: (1) if olanzapine prevents PCP-induced suppression of neurite outgrowth and synaptic protein expression; (2) if olanzapine affects the Akt-GSK3 signaling pathway; and (3) the role of neuregulin 1 (NRG1) in this process. Immunofluorescence revealed that PCP treatment for 24 hours reduces both neurite length (28.5%) and the number of neurite branches (35.6%) in primary prefrontal cortical neuron cultures. PCP reduced protein and mRNA expressions of synaptophysin (24.9% and 23.2%, respectively) and PSD95 (31.5% and 21.4%, respectively), and the protein expression of p-Akt (26.7%) and p-GSK3β (35.2%). Olanzapine co-treatment prevented these PCP-induced effects in normal neurons but not in neurons from NRG1-knockout mice. These results indicate that NRG1 mediates the preventive effects of olanzapine on the PCP-induced impairment of neurite outgrowth and synaptic protein expression. This study provides potential targets for interventions on improving the efficacy of olanzapine on preventing cognitive deficits in schizophrenia.
Collapse
Affiliation(s)
- Qingsheng Zhang
- Centre for Translational Neuroscience, School of Medicine, University of Wollongong, Wollongong, 2522, NSW, Australia.,Illawarra Health and Medical Research Institute, Wollongong, 2522, NSW, Australia
| | - Yinghua Yu
- Centre for Translational Neuroscience, School of Medicine, University of Wollongong, Wollongong, 2522, NSW, Australia.,Illawarra Health and Medical Research Institute, Wollongong, 2522, NSW, Australia.,Schizophrenia Research Institute, 384 Victoria Street, Darlinghurst, 2010, NSW, Australia
| | - Xu-Feng Huang
- Centre for Translational Neuroscience, School of Medicine, University of Wollongong, Wollongong, 2522, NSW, Australia.,Illawarra Health and Medical Research Institute, Wollongong, 2522, NSW, Australia.,Schizophrenia Research Institute, 384 Victoria Street, Darlinghurst, 2010, NSW, Australia
| |
Collapse
|
178
|
Zhou Y, Wang J, Gu Z, Wang S, Zhu W, Aceña JL, Soloshonok VA, Izawa K, Liu H. Next Generation of Fluorine-Containing Pharmaceuticals, Compounds Currently in Phase II-III Clinical Trials of Major Pharmaceutical Companies: New Structural Trends and Therapeutic Areas. Chem Rev 2016; 116:422-518. [PMID: 26756377 DOI: 10.1021/acs.chemrev.5b00392] [Citation(s) in RCA: 1823] [Impact Index Per Article: 227.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Yu Zhou
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 555 Zu Chong Zhi Road, Shanghai 201203, China
| | - Jiang Wang
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 555 Zu Chong Zhi Road, Shanghai 201203, China
| | - Zhanni Gu
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 555 Zu Chong Zhi Road, Shanghai 201203, China
| | - Shuni Wang
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 555 Zu Chong Zhi Road, Shanghai 201203, China
| | - Wei Zhu
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 555 Zu Chong Zhi Road, Shanghai 201203, China
| | - José Luis Aceña
- Department of Organic Chemistry I, Faculty of Chemistry, University of the Basque Country UPV/EHU , Paseo Manuel Lardizábal 3, 20018 San Sebastián, Spain.,Department of Organic Chemistry, Autónoma University of Madrid , Cantoblanco, 28049 Madrid, Spain
| | - Vadim A Soloshonok
- Department of Organic Chemistry I, Faculty of Chemistry, University of the Basque Country UPV/EHU , Paseo Manuel Lardizábal 3, 20018 San Sebastián, Spain.,IKERBASQUE, Basque Foundation for Science, María Díaz de Haro 3, 48013 Bilbao, Spain
| | - Kunisuke Izawa
- Hamari Chemicals Ltd., 1-4-29 Kunijima, Higashi-Yodogawa-ku, Osaka, Japan 533-0024
| | - Hong Liu
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 555 Zu Chong Zhi Road, Shanghai 201203, China
| |
Collapse
|
179
|
Engen K, Agartz I. Anti-NMDA-reseptorencefalitt. TIDSSKRIFT FOR DEN NORSKE LEGEFORENING 2016; 136:1006-9. [DOI: 10.4045/tidsskr.15.0795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
|
180
|
Pollak TA, Beck K, Irani SR, Howes OD, David AS, McGuire PK. Autoantibodies to central nervous system neuronal surface antigens: psychiatric symptoms and psychopharmacological implications. Psychopharmacology (Berl) 2016; 233:1605-21. [PMID: 26667479 PMCID: PMC4828500 DOI: 10.1007/s00213-015-4156-y] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 11/05/2015] [Indexed: 12/30/2022]
Abstract
RATIONALE Autoantibodies to central nervous system (CNS) neuronal surface antigens have been described in association with autoimmune encephalopathies which prominently feature psychiatric symptoms in addition to neurological symptoms. The potential role of these autoantibodies in primary psychiatric diseases such as schizophrenia or bipolar affective disorder is of increasing interest. OBJECTIVES We aimed to review the nature of psychiatric symptoms associated with neuronal surface autoantibodies, in the context of autoimmune encephalopathies as well as primary psychiatric disorders, and to review the mechanisms of action of these autoantibodies from a psychopharmacological perspective. RESULTS The functional effects of the autoantibodies on their target antigens are described; their clinical expression is at least in part mediated by their effects on neuronal receptor function, primarily at the synapse, usually resulting in receptor hypofunction. The psychiatric effects of the antibodies are related to known functions of the receptor target or its complexed proteins, with reference to supportive genetic and pharmacological evidence where relevant. Evidence for a causal role of these autoantibodies in primary psychiatric disease is increasing but remains controversial; relevant methodological controversies are outlined. Non-receptor-based mechanisms of autoantibody action, including neuroinflammatory mechanisms, and therapeutic implications are discussed. CONCLUSIONS An analysis of the autoantibodies from a psychopharmacological perspective, as endogenous, bioactive, highly specific, receptor-targeting molecules, provides a valuable opportunity to understand the neurobiological basis of associated psychiatric symptoms. Potentially, new treatment strategies will emerge from the improving understanding of antibody-antigen interaction within the CNS.
Collapse
Affiliation(s)
- T A Pollak
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's Health Partners, King's College London, De Crespigny Park, Denmark Hill, London, SE5 8AF, UK.
| | - K Beck
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's Health Partners, King's College London, De Crespigny Park, Denmark Hill, London, SE5 8AF, UK
| | - S R Irani
- Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, Oxford, UK
| | - O D Howes
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's Health Partners, King's College London, De Crespigny Park, Denmark Hill, London, SE5 8AF, UK
| | - A S David
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's Health Partners, King's College London, De Crespigny Park, Denmark Hill, London, SE5 8AF, UK
| | - P K McGuire
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's Health Partners, King's College London, De Crespigny Park, Denmark Hill, London, SE5 8AF, UK
| |
Collapse
|
181
|
Influence of Antipsychotic and Anticholinergic Loads on Cognitive Functions in Patients with Schizophrenia. SCHIZOPHRENIA RESEARCH AND TREATMENT 2016; 2016:8213165. [PMID: 27144021 PMCID: PMC4842070 DOI: 10.1155/2016/8213165] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 02/19/2016] [Accepted: 03/07/2016] [Indexed: 11/18/2022]
Abstract
Many patients with schizophrenia show cognitive impairment. There is evidence that, beyond a certain dose of antipsychotic medication, the antipsychotic daily dose (ADD) may impair cognitive performance. Parallel to their D2 receptor antagonism, many antipsychotics show a significant binding affinity to cholinergic muscarinic receptors. Pharmacological treatment with a high anticholinergic daily dose (CDD) significantly impairs attention and memory performance. To examine the relationships between individual cognitive performance and ADD and/or CDD, we conducted a retrospective record-based analysis of a sample of n = 104 in patients with a diagnosis of schizophrenia, all of whom had completed a comprehensive neuropsychological test battery. To calculate the individual ADD and CDD, the medication at the time of testing was converted according to equivalence models. After extracting five principal cognitive components, we examined the impact of ADD and CDD on cognitive performance in the medicated sample and subgroups using multiple regression analysis. Finally, locally weighted scatterplot smoothing (Loess) was applied to further explore the course of cognitive performance under increasing dosage. Results showed significant negative effects of ADD on performance in tests of information processing speed and verbal memory. No effects were found for CDD. The potential neuropsychopharmacological and clinical implications are discussed.
Collapse
|
182
|
Riebe I, Seth H, Culley G, Dósa Z, Radi S, Strand K, Fröjd V, Hanse E. Tonically active NMDA receptors--a signalling mechanism critical for interneuronal excitability in the CA1 stratum radiatum. Eur J Neurosci 2015; 43:169-78. [PMID: 26547631 DOI: 10.1111/ejn.13128] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 11/02/2015] [Accepted: 11/03/2015] [Indexed: 11/30/2022]
Abstract
In contrast to tonic extrasynaptic γ-aminobutyric acid (GABA)A receptor-mediated signalling, the physiological significance of tonic extrasynaptic N-methyl-D-aspartate (NMDA) receptor (NMDAR)-mediated signalling remains uncertain. In this study, reversible open-channel blockers of NMDARs, memantine and phencyclidine (PCP) were used as tools to examine tonic NMDAR-mediated signalling in rat hippocampal slices. Memantine in concentrations up to 10 μM had no effect on synaptically evoked NMDAR-mediated responses in pyramidal neurons or GABAergic interneurons. On the other hand, 10 μM memantine reduced tonic NMDAR-mediated currents in GABAergic interneurons by approximately 50%. These tonic NMDAR-mediated currents in interneurons contributed significantly to the excitability of the interneurons as 10 μM memantine reduced the disynaptic inhibitory postsynaptic current in pyramidal cells by about 50%. Moreover, 10 μM memantine, but also PCP in concentrations ≤ 1 μM, increased the magnitude of the population spike, likely because of disinhibition. The relatively higher impact of tonic NMDAR-mediated signalling in interneurons was at least partly explained by the expression of GluN2D-containing NMDARs, which was not observed in mature pyramidal cells. The current results are consistent with the idea that low doses of readily reversible NMDAR open-channel blockers preferentially inhibit tonically active extrasynaptic NMDARs, and they suggest that tonically active NMDARs contribute more prominently to the intrinsic excitation in GABAergic interneurons than in pyramidal cells. It is proposed that this specific difference between interneurons and pyramidal cells can explain the disinhibition caused by the Alzheimer's disease medication memantine.
Collapse
Affiliation(s)
- Ilse Riebe
- Department of Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, Gothenburg University, Medicinaregatan 11, 405 30, Gothenburg, Sweden
| | - Henrik Seth
- Department of Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, Gothenburg University, Medicinaregatan 11, 405 30, Gothenburg, Sweden
| | - Georgia Culley
- Department of Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, Gothenburg University, Medicinaregatan 11, 405 30, Gothenburg, Sweden
| | - Zita Dósa
- Synaptic Physiology Laboratory, Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Shayma Radi
- Department of Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, Gothenburg University, Medicinaregatan 11, 405 30, Gothenburg, Sweden
| | - Karin Strand
- Department of Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, Gothenburg University, Medicinaregatan 11, 405 30, Gothenburg, Sweden
| | - Victoria Fröjd
- Department of Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, Gothenburg University, Medicinaregatan 11, 405 30, Gothenburg, Sweden
| | - Eric Hanse
- Department of Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, Gothenburg University, Medicinaregatan 11, 405 30, Gothenburg, Sweden
| |
Collapse
|
183
|
Li Y, Wang X, Hou Y, Zhou X, Chen Q, Guo C, Xia Q, Zhang Y, Zhao P. Integrative Proteomics and Metabolomics Analysis of Insect Larva Brain: Novel Insights into the Molecular Mechanism of Insect Wandering Behavior. J Proteome Res 2015; 15:193-204. [PMID: 26644297 DOI: 10.1021/acs.jproteome.5b00736] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Before metamorphosis, most holometabolous insects, such as the silkworm studied here, undergo a special phase called the wandering stage. Insects in this stage often display enhanced locomotor activity (ELA). ELA is vital because it ensures that the insect finds a safe and suitable place to live through the pupal stage. The physiological mechanisms of wandering behavior are still unclear. Here, we integrated proteomics and metabolomics approaches to analyze the brain of the lepidopteran insect, silkworm, at the feeding and wandering stages. Using LC-MS/MS and GC-MS, in all we identified 3004 proteins and 37 metabolites at these two stages. Among them, 465 proteins and 22 metabolites were changed. Neural signal transduction proteins and metabolites, such as neurofilament, dopaminergic synapse related proteins, and glutamic acid, were significantly altered, which suggested that active neural conduction occurred in the brain at the wandering stage. We also found decreased dopamine degradation at the wandering stage. The proposed changes in active neural conduction and increased dopamine concentration might induce ELA. In addition, proteins involved in the ubiquitin proteasome system and lysosome pathway were upregulated, revealing that the brain experiences morphological remodeling during metamorphosis. These findings yielded novel insights into the molecular mechanism underlying insect wandering behavior.
Collapse
Affiliation(s)
- Yi Li
- State Key Laboratory of Silkworm Genome Biology, Southwest University , Chongqing 400716, China
| | - Xin Wang
- State Key Laboratory of Silkworm Genome Biology, Southwest University , Chongqing 400716, China
| | - Yong Hou
- State Key Laboratory of Silkworm Genome Biology, Southwest University , Chongqing 400716, China
| | - Xiaoying Zhou
- State Key Laboratory of Silkworm Genome Biology, Southwest University , Chongqing 400716, China
| | - Quanmei Chen
- Department of Biochemistry & Molecular Biology, Chongqing Medical University , Chongqing 400016, China
| | - Chao Guo
- State Key Laboratory of Silkworm Genome Biology, Southwest University , Chongqing 400716, China
| | - Qingyou Xia
- State Key Laboratory of Silkworm Genome Biology, Southwest University , Chongqing 400716, China
| | - Yan Zhang
- State Key Laboratory of Silkworm Genome Biology, Southwest University , Chongqing 400716, China
| | - Ping Zhao
- State Key Laboratory of Silkworm Genome Biology, Southwest University , Chongqing 400716, China
| |
Collapse
|
184
|
Sapkota K, Mao Z, Synowicki P, Lieber D, Liu M, Ikezu T, Gautam V, Monaghan DT. GluN2D N-Methyl-d-Aspartate Receptor Subunit Contribution to the Stimulation of Brain Activity and Gamma Oscillations by Ketamine: Implications for Schizophrenia. J Pharmacol Exp Ther 2015; 356:702-11. [PMID: 26675679 DOI: 10.1124/jpet.115.230391] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 12/15/2015] [Indexed: 01/01/2023] Open
Abstract
The dissociative anesthetic ketamine elicits symptoms of schizophrenia at subanesthetic doses by blocking N-methyl-d-aspartate receptors (NMDARs). This property led to a variety of studies resulting in the now well-supported theory that hypofunction of NMDARs is responsible for many of the symptoms of schizophrenia. However, the roles played by specific NMDAR subunits in different symptom components are unknown. To evaluate the potential contribution of GluN2D NMDAR subunits to antagonist-induced cortical activation and schizophrenia symptoms, we determined the ability of ketamine to alter regional brain activity and gamma frequency band neuronal oscillations in wild-type (WT) and GluN2D-knockout (GluN2D-KO) mice. In WT mice, ketamine (30 mg/kg, i.p.) significantly increased [(14)C]-2-deoxyglucose ([(14)C]-2DG) uptake in the medial prefrontal cortex (mPFC), entorhinal cortex and other brain regions, and decreased activity in the somatosensory cortex and inferior colliculus. In GluN2D-KO mice, however, ketamine did not significantly increase [(14)C]-2DG uptake in any brain region examined, yet still decreased [(14)C]-2DG uptake in the somatosensory cortex and inferior colliculus. Ketamine also increased locomotor activity in WT mice but not in GluN2D-KO mice. In electrocorticographic analysis, ketamine induced a 111% ± 16% increase in cortical gamma-band oscillatory power in WT mice, but only a 15% ± 12% increase in GluN2D-KO mice. Consistent with GluN2D involvement in schizophrenia-related neurologic changes, GluN2D-KO mice displayed impaired spatial memory acquisition and reduced parvalbumin (PV)-immunopositive staining compared with control mice. These results suggest a critical role of GluN2D-containing NMDARs in neuronal oscillations and ketamine's psychotomimetic, dissociative effects and hence suggests a critical role for GluN2D subunits in cognition and perception.
Collapse
Affiliation(s)
- Kiran Sapkota
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska (K.S., Z.M., P.S., D.L., M.L., D.T.M.); Departments of Pharmacology & Experimental Therapeutics and Neurology, School of Medicine, Boston University, Boston, Massachusetts (T.I.); Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts (V.G.)
| | - Zhihao Mao
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska (K.S., Z.M., P.S., D.L., M.L., D.T.M.); Departments of Pharmacology & Experimental Therapeutics and Neurology, School of Medicine, Boston University, Boston, Massachusetts (T.I.); Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts (V.G.)
| | - Paul Synowicki
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska (K.S., Z.M., P.S., D.L., M.L., D.T.M.); Departments of Pharmacology & Experimental Therapeutics and Neurology, School of Medicine, Boston University, Boston, Massachusetts (T.I.); Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts (V.G.)
| | - Dillon Lieber
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska (K.S., Z.M., P.S., D.L., M.L., D.T.M.); Departments of Pharmacology & Experimental Therapeutics and Neurology, School of Medicine, Boston University, Boston, Massachusetts (T.I.); Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts (V.G.)
| | - Meng Liu
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska (K.S., Z.M., P.S., D.L., M.L., D.T.M.); Departments of Pharmacology & Experimental Therapeutics and Neurology, School of Medicine, Boston University, Boston, Massachusetts (T.I.); Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts (V.G.)
| | - Tsuneya Ikezu
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska (K.S., Z.M., P.S., D.L., M.L., D.T.M.); Departments of Pharmacology & Experimental Therapeutics and Neurology, School of Medicine, Boston University, Boston, Massachusetts (T.I.); Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts (V.G.)
| | - Vivek Gautam
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska (K.S., Z.M., P.S., D.L., M.L., D.T.M.); Departments of Pharmacology & Experimental Therapeutics and Neurology, School of Medicine, Boston University, Boston, Massachusetts (T.I.); Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts (V.G.)
| | - Daniel T Monaghan
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska (K.S., Z.M., P.S., D.L., M.L., D.T.M.); Departments of Pharmacology & Experimental Therapeutics and Neurology, School of Medicine, Boston University, Boston, Massachusetts (T.I.); Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts (V.G.)
| |
Collapse
|
185
|
Kobayashi Y, Kulikova SP, Shibato J, Rakwal R, Satoh H, Pinault D, Masuo Y. DNA microarray unravels rapid changes in transcriptome of MK-801 treated rat brain. World J Biol Chem 2015; 6:389-408. [PMID: 26629322 PMCID: PMC4657125 DOI: 10.4331/wjbc.v6.i4.389] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 01/20/2015] [Accepted: 08/31/2015] [Indexed: 02/05/2023] Open
Abstract
AIM: To investigate the impact of MK-801 on gene expression patterns genome wide in rat brain regions.
METHODS: Rats were treated with an intraperitoneal injection of MK-801 [0.08 (low-dose) and 0.16 (high-dose) mg/kg] or NaCl (vehicle control). In a first series of experiment, the frontoparietal electrocorticogram was recorded 15 min before and 60 min after injection. In a second series of experiments, the whole brain of each animal was rapidly removed at 40 min post-injection, and different regions were separated: amygdala, cerebral cortex, hippocampus, hypothalamus, midbrain and ventral striatum on ice followed by DNA microarray (4 × 44 K whole rat genome chip) analysis.
RESULTS: Spectral analysis revealed that a single systemic injection of MK-801 significantly and selectively augmented the power of baseline gamma frequency (30-80 Hz) oscillations in the frontoparietal electroencephalogram. DNA microarray analysis showed the largest number (up- and down- regulations) of gene expressions in the cerebral cortex (378), midbrain (376), hippocampus (375), ventral striatum (353), amygdala (301), and hypothalamus (201) under low-dose (0.08 mg/kg) of MK-801. Under high-dose (0.16 mg/kg), ventral striatum (811) showed the largest number of gene expression changes. Gene expression changes were functionally categorized to reveal expression of genes and function varies with each brain region.
CONCLUSION: Acute MK-801 treatment increases synchrony of baseline gamma oscillations, and causes very early changes in gene expressions in six individual rat brain regions, a first report.
Collapse
|
186
|
Rodríguez-Muñoz M, Cortés-Montero E, Pozo-Rodrigálvarez A, Sánchez-Blázquez P, Garzón-Niño J. The ON:OFF switch, σ1R-HINT1 protein, controls GPCR-NMDA receptor cross-regulation: implications in neurological disorders. Oncotarget 2015; 6:35458-77. [PMID: 26461475 PMCID: PMC4742118 DOI: 10.18632/oncotarget.6064] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 09/23/2015] [Indexed: 12/11/2022] Open
Abstract
In the brain, the histidine triad nucleotide-binding protein 1 (HINT1) and sigma 1 receptors (σ1Rs) coordinate the activity of certain G-protein coupled receptors (GPCRs) with that of glutamate N-methyl-D-aspartate receptors (NMDARs). To determine the role of HINT1-σ1R in the plasticity of GPCR-NMDAR interactions, substances acting at MOR, cannabinoid CB1 receptor, NMDAR and σ1R were injected into mice, and their effects were evaluated through in vivo, ex vivo, and in vitro assays. It was observed that HINT1 protein binds to GPCRs and NMDAR NR1 subunits in a calcium-independent manner, whereas σ1R binding to these proteins increases in the presence of calcium. In this scenario, σ1R agonists keep HINT1 at the GPCR and stimulate GPCR-NMDAR interaction, whereas σ1R antagonists transfer HINT1 to NR1 subunits and disengage both receptors. This regulation is lost in σ1R-/- mice, where HINT1 proteins mostly associate with NMDARs, and GPCRs are physically and functionally disconnected from NMDARs. In HINT1-/- mice, ischemia produces low NMDAR-mediated brain damage, suggesting that several different GPCRs enhance glutamate excitotoxicity via HINT1-σ1R. Thus, several GPCRs associate with NMDARs by a dynamic process under the physiological control of HINT1 proteins and σ1Rs. The NMDAR-HINT1-σ1R complex deserves attention because it offers new therapeutic opportunities.
Collapse
Affiliation(s)
- María Rodríguez-Muñoz
- Department of Molecular, Cellular and Developmental Neurobiology, Laboratory of Neuropharmacology. Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC). Madrid, Spain
| | - Elsa Cortés-Montero
- Department of Molecular, Cellular and Developmental Neurobiology, Laboratory of Neuropharmacology. Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC). Madrid, Spain
| | - Andrea Pozo-Rodrigálvarez
- Department of Molecular, Cellular and Developmental Neurobiology, Laboratory of Neuropharmacology. Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC). Madrid, Spain
| | - Pilar Sánchez-Blázquez
- Department of Molecular, Cellular and Developmental Neurobiology, Laboratory of Neuropharmacology. Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC). Madrid, Spain
| | - Javier Garzón-Niño
- Department of Molecular, Cellular and Developmental Neurobiology, Laboratory of Neuropharmacology. Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC). Madrid, Spain
| |
Collapse
|
187
|
The role of nitric oxide donors in schizophrenia: Basic studies and clinical applications. Eur J Pharmacol 2015; 766:106-13. [DOI: 10.1016/j.ejphar.2015.09.045] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 09/14/2015] [Accepted: 09/28/2015] [Indexed: 01/04/2023]
|
188
|
Cannon TD. How Schizophrenia Develops: Cognitive and Brain Mechanisms Underlying Onset of Psychosis. Trends Cogn Sci 2015; 19:744-756. [PMID: 26493362 DOI: 10.1016/j.tics.2015.09.009] [Citation(s) in RCA: 144] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2015] [Revised: 09/10/2015] [Accepted: 09/11/2015] [Indexed: 12/12/2022]
Abstract
Identifying cognitive and neural mechanisms involved in the development of schizophrenia requires longitudinal observation of individuals prior to onset. Here recent studies of prodromal individuals who progress to full psychosis are briefly reviewed in relation to models of schizophrenia pathophysiology. Together, this body of work suggests that disruption in brain connectivity, driven primarily by a progressive reduction in dendritic spines on cortical pyramidal neurons, may represent a key triggering mechanism. The earliest disruptions appear to be in circuits involved in referencing experiences according to time, place, and agency, which may result in a failure to recognize particular cognitions as self-generated or to constrain interpretations of the meaning of events based on prior experiences, providing the scaffolding for faulty reality testing.
Collapse
Affiliation(s)
- Tyrone D Cannon
- Department of Psychology, Yale University, 2 Hillhouse Avenue, P.O. Box 208205, New Haven, CT 06520, USA.
| |
Collapse
|
189
|
Galderisi S, Merlotti E, Mucci A. Neurobiological background of negative symptoms. Eur Arch Psychiatry Clin Neurosci 2015; 265:543-58. [PMID: 25797499 DOI: 10.1007/s00406-015-0590-4] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2015] [Accepted: 03/15/2015] [Indexed: 01/29/2023]
Abstract
Studies investigating neurobiological bases of negative symptoms of schizophrenia failed to provide consistent findings, possibly due to the heterogeneity of this psychopathological construct. We tried to review the findings published to date investigating neurobiological abnormalities after reducing the heterogeneity of the negative symptoms construct. The literature in electronic databases as well as citations and major articles are reviewed with respect to the phenomenology, pathology, genetics and neurobiology of schizophrenia. We searched PubMed with the keywords "negative symptoms," "deficit schizophrenia," "persistent negative symptoms," "neurotransmissions," "neuroimaging" and "genetic." Additional articles were identified by manually checking the reference lists of the relevant publications. Publications in English were considered, and unpublished studies, conference abstracts and poster presentations were not included. Structural and functional imaging studies addressed the issue of neurobiological background of negative symptoms from several perspectives (considering them as a unitary construct, focusing on primary and/or persistent negative symptoms and, more recently, clustering them into factors), but produced discrepant findings. The examined studies provided evidence suggesting that even primary and persistent negative symptoms include different psychopathological constructs, probably reflecting the dysfunction of different neurobiological substrates. Furthermore, they suggest that complex alterations in multiple neurotransmitter systems and genetic variants might influence the expression of negative symptoms in schizophrenia. On the whole, the reviewed findings, representing the distillation of a large body of disparate data, suggest that further deconstruction of negative symptomatology into more elementary components is needed to gain insight into underlying neurobiological mechanisms.
Collapse
Affiliation(s)
- Silvana Galderisi
- Department of Psychiatry, Second University of Naples (SUN), L.go Madonna delle Grazie, 1, 80138, Naples, Italy.
| | - Eleonora Merlotti
- Department of Psychiatry, Second University of Naples (SUN), L.go Madonna delle Grazie, 1, 80138, Naples, Italy
| | - Armida Mucci
- Department of Psychiatry, Second University of Naples (SUN), L.go Madonna delle Grazie, 1, 80138, Naples, Italy
| |
Collapse
|
190
|
Chang EH, Kirtley A, Chandon TSS, Borger P, Husain-Krautter S, Vingtdeux V, Malhotra AK. Postnatal neurodevelopmental expression and glutamate-dependent regulation of the ZNF804A rodent homologue. Schizophr Res 2015; 168:402-410. [PMID: 26164821 PMCID: PMC4591171 DOI: 10.1016/j.schres.2015.06.023] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Revised: 06/19/2015] [Accepted: 06/23/2015] [Indexed: 12/28/2022]
Abstract
The zinc finger protein ZNF804A rs1344706 variant is a replicated genome-wide significant risk variant for schizophrenia and bipolar disorder. While its association with altered brain structure and cognition in patients and healthy risk allele carriers is well documented, the characteristics and function of the gene in the brain remains poorly understood. Here, we used in situ hybridization to determine mRNA expression levels of the ZNF804A rodent homologue, Zfp804a, across multiple postnatal neurodevelopmental time points in the rat brain. We found changes in Zfp804a expression in the rat hippocampus, frontal cortex, and thalamus across postnatal neurodevelopment. Zfp804a mRNA peaked at postnatal day (P) 21 in hippocampal CA1 and DG regions and was highest in the lower cortical layers of frontal cortex at P1, possibly highlighting a role in developmental migration. Using immunofluorescence, we found that Zfp804a mRNA and ZFP804A co-localized with neurons and not astrocytes. In primary cultured cortical neurons, we found that Zfp804a expression was significantly increased when neurons were exposed to glutamate [20μM], but this increase was blocked by the N-methyl-d-aspartate receptor (NMDAR) antagonist MK-801. Expression of Comt, Pde4b, and Drd2, genes previously shown to be regulated by ZNF804A overexpression, was also significantly changed in an NMDA-dependent manner. Our results describe, for the first time, the unique postnatal neurodevelopmental expression of Zfp804a in the rodent brain and demonstrate that glutamate potentially plays an important role in the regulation of this psychiatric susceptibility gene. These are critical steps toward understanding the biological function of ZNF804A in the mammalian brain.
Collapse
Affiliation(s)
- Eric H. Chang
- Center for Psychiatric Neuroscience, The Feinstein Institute for Medical Research, North Shore-LIJ Health System, 350 Community Drive, Manhasset, NY 11030, USA, Department of Psychiatry Research, Zucker Hillside Hospital, North Shore-LIJ Health System, 75-59 263rd Street, Glen Oaks, NY 11004, USA
| | - Anne Kirtley
- Center for Psychiatric Neuroscience, The Feinstein Institute for Medical Research, North Shore-LIJ Health System, 350 Community Drive, Manhasset, NY 11030, USA, Department of Psychiatry Research, Zucker Hillside Hospital, North Shore-LIJ Health System, 75-59 263rd Street, Glen Oaks, NY 11004, USA
| | - Toni-Shay S. Chandon
- Center for Psychiatric Neuroscience, The Feinstein Institute for Medical Research, North Shore-LIJ Health System, 350 Community Drive, Manhasset, NY 11030, USA, Department of Psychiatry Research, Zucker Hillside Hospital, North Shore-LIJ Health System, 75-59 263rd Street, Glen Oaks, NY 11004, USA
| | - Philip Borger
- Center for Psychiatric Neuroscience, The Feinstein Institute for Medical Research, North Shore-LIJ Health System, 350 Community Drive, Manhasset, NY 11030, USA, Department of Psychiatry Research, Zucker Hillside Hospital, North Shore-LIJ Health System, 75-59 263rd Street, Glen Oaks, NY 11004, USA
| | - Sehba Husain-Krautter
- Center for Psychiatric Neuroscience, The Feinstein Institute for Medical Research, North Shore-LIJ Health System, 350 Community Drive, Manhasset, NY 11030, USA, Department of Psychiatry Research, Zucker Hillside Hospital, North Shore-LIJ Health System, 75-59 263rd Street, Glen Oaks, NY 11004, USA, Center for Oncology and Cell Biology, The Feinstein Institute for Medical Research, North Shore-LIJ Health System, 350 Community Drive, Manhasset, NY 11030, USA
| | - Valerie Vingtdeux
- Litwin Center for the Study of Alzheimer's Disease, The Feinstein Institute for Medical Research, North Shore-LIJ Health System, 350 Community Drive, Manhasset, NY 11030, USA
| | - Anil K. Malhotra
- Center for Psychiatric Neuroscience, The Feinstein Institute for Medical Research, North Shore-LIJ Health System, 350 Community Drive, Manhasset, NY 11030, USA, Department of Psychiatry Research, Zucker Hillside Hospital, North Shore-LIJ Health System, 75-59 263rd Street, Glen Oaks, NY 11004, USA, Hofstra North Shore-LIJ School of Medicine, Departments of Psychiatry and Molecular Medicine, Hofstra University, Hempstead, NY, USA
| |
Collapse
|
191
|
Pinacho R, Saia G, Meana JJ, Gill G, Ramos B. Transcription factor SP4 phosphorylation is altered in the postmortem cerebellum of bipolar disorder and schizophrenia subjects. Eur Neuropsychopharmacol 2015; 25:1650-1660. [PMID: 26049820 PMCID: PMC4600646 DOI: 10.1016/j.euroneuro.2015.05.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Revised: 12/12/2014] [Accepted: 05/13/2015] [Indexed: 11/27/2022]
Abstract
Transcription factors play important roles in the control of neuronal function in physiological and pathological conditions. We previously reported reduced levels of transcription factor SP4 protein, but not transcript, in the cerebellum in bipolar disorder and associated with more severe negative symptoms in schizophrenia. We have recently reported phosphorylation of Sp4 at S770, which is regulated by membrane depolarization and NMDA receptor activity. The aim of this study was to investigate SP4 S770 phosphorylation in bipolar disorder and its association with negative symptoms in schizophrenia, and to explore the potential relationship between phosphorylation and protein abundance. Here we report a significant increase in SP4 phosphorylation in the cerebellum, but not the prefrontal cortex, of bipolar disorder subjects (n=10) (80% suicide) compared to matched controls (n=10). We found that SP4 phosphorylation inversely correlated with SP4 levels independently of disease status in both areas of the human brain. Moreover, SP4 phosphorylation in the cerebellum positively correlated with negative symptoms in schizophrenia subjects (n=15). Further, we observed that a phospho-mimetic mutation in truncated Sp4 was sufficient to significantly decrease Sp4 steady-state levels, while a non-phosphorylatable mutant showed increased stability in cultured rat cerebellar granule neurons. Our results indicate that SP4 S770 phosphorylation is increased in the cerebellum in bipolar disorder subjects that committed suicide and in severe schizophrenia subjects, and may be part of a degradation signal that controls Sp4 abundance in cerebellar granule neurons. This opens the possibility that modulation of SP4 phosphorylation may contribute to the molecular pathophysiology of psychotic disorders.
Collapse
Affiliation(s)
- Raquel Pinacho
- Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM. Dr. Antoni Pujadas, 42, 08830- Sant Boi de Llobregat, Barcelona, Spain
| | - Gregory Saia
- Department of Developmental, Molecular, and Chemical Biology, Sackler School of Biomedical Sciences, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111.,Cell, Molecular and Developmental Biology Program, Sackler School of Biomedical Sciences, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111
| | - J Javier Meana
- Departamento de Farmacología, Universidad del País Vasco / Euskal Herriko Unibertsitatea UPV/EHU, Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, B Sarriena s/n 48940-Leioa, Bizkaia, Spain
| | - Grace Gill
- Department of Developmental, Molecular, and Chemical Biology, Sackler School of Biomedical Sciences, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111
| | - Belén Ramos
- Unitat de recerca, Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Universitat de Barcelona, Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM. Dr. Antoni Pujadas, 42, 08830- Sant Boi de Llobregat, Barcelona, Spain
| |
Collapse
|
192
|
Wallace TL, Bertrand D. Neuronal α7 Nicotinic Receptors as a Target for the Treatment of Schizophrenia. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2015; 124:79-111. [PMID: 26472526 DOI: 10.1016/bs.irn.2015.08.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Schizophrenia is a lifelong disease, the burden of which is often underestimated. Characterized by positive (e.g., hallucinations) and negative (e.g., avolition, amotivation) symptoms, schizophrenia is also accompanied with profound impairments in cognitive function that progress throughout the development of the disease. Although treatment with antipsychotic medications can effectively dampen some of the positive symptoms, these medications largely fail to reverse cognitive deficits or to mitigate negative symptoms. With a worldwide prevalence of approximately 1%, schizophrenia remains a large unmet medical need that stands to benefit greatly from (1) continued research to better understand the biological underpinnings of the disease and (2) the targeted development of novel therapeutics to improve the lives of those affected individuals. Improvements in our understanding of the neuronal networks associated with schizophrenia as well as progress in identifying genetic risk factors and environmental conditions that may predispose individuals to developing the disease are advancing new strategies to study and treat it. Herein, we review the evidence that supports the role of α7 nicotinic acetylcholine receptors in the central nervous system and why these receptors constitute a promising target to treat some of the prominent symptoms of schizophrenia.
Collapse
|
193
|
Abstract
We describe the case of a 17-year-old male who presented with acute onset of seizures and malignant catatonia with psychosis, agitation, and hypermetabolism, who responded to electroconvulsive therapy (ECT). Soon after he began to respond, he was diagnosed with anti-N-methyl-D-aspartate (NMDA) receptor encephalitis and then given immunosuppressive therapy. Anti-NMDA receptor encephalitis is an increasingly recognized autoimmune disorder that often presents with neuropsychiatric symptoms. The mainstays for treatment have been early diagnosis, tumor work-up and removal if found, and initiation of immunosuppressive therapy. Treatment response is often slow and residual symptoms common. In this case, ECT produced clinical stabilization before the underlying diagnosis of anti-NMDA receptor encephalitis was made and standard treatment initiated. We suggest that ECT may be highly beneficial for stabilizing life-threatening neuropsychiatric symptoms in this syndrome and should be considered as a potentially additive treatment to immunotherapy when rapid relief is sought.
Collapse
|
194
|
Tsapakis EM, Dimopoulou T, Tarazi FI. Clinical management of negative symptoms of schizophrenia: An update. Pharmacol Ther 2015; 153:135-47. [DOI: 10.1016/j.pharmthera.2015.06.008] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 06/15/2015] [Indexed: 02/07/2023]
|
195
|
Raiteri L, Raiteri M. Multiple functions of neuronal plasma membrane neurotransmitter transporters. Prog Neurobiol 2015; 134:1-16. [PMID: 26300320 DOI: 10.1016/j.pneurobio.2015.08.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 07/09/2015] [Accepted: 08/18/2015] [Indexed: 12/11/2022]
Abstract
Removal from receptors of neurotransmitters just released into synapses is one of the major steps in neurotransmission. Transporters situated on the plasma membrane of nerve endings and glial cells perform the process of neurotransmitter (re)uptake. Because the density of transporters in the membranes can fluctuate, transporters can determine the transmitter concentrations at receptors, thus modulating indirectly the excitability of neighboring neurons. Evidence is accumulating that neurotransmitter transporters can exhibit multiple functions. Being bidirectional, neurotransmitter transporters can mediate transmitter release by working in reverse, most often under pathological conditions that cause ionic gradient dysregulations. Some transporters reverse to release transmitters, like dopamine or serotonin, when activated by 'indirectly acting' substrates, like the amphetamines. Some transporters exhibit as one major function the ability to capture transmitters into nerve terminals that perform insufficient synthesis. Transporter activation can generate conductances that regulate directly neuronal excitability. Synaptic and non-synaptic transporters play different roles. Cytosolic Na(+) elevations accompanying transport can interact with plasmalemmal or/and mitochondrial Na(+)/Ca(2+) exchangers thus generating calcium signals. Finally, neurotransmitter transporters can behave as receptors mediating releasing stimuli able to cause transmitter efflux through multiple mechanisms. Neurotransmitter transporters are therefore likely to play hitherto unknown roles in multiple therapeutic treatments.
Collapse
Affiliation(s)
- Luca Raiteri
- Department of Pharmacy, Pharmacology and Toxicology Section, University of Genoa, Genoa, Italy; Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy; National Institute of Neuroscience, Genoa, Italy
| | - Maurizio Raiteri
- Department of Pharmacy, Pharmacology and Toxicology Section, University of Genoa, Genoa, Italy; Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy; National Institute of Neuroscience, Genoa, Italy.
| |
Collapse
|
196
|
Flores FJ, Ching S, Hartnack K, Fath AB, Purdon PL, Wilson MA, Brown EN. A PK-PD model of ketamine-induced high-frequency oscillations. J Neural Eng 2015; 12:056006. [PMID: 26268223 DOI: 10.1088/1741-2560/12/5/056006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
OBJECTIVE Ketamine is a widely used drug with clinical and research applications, and also known to be used as a recreational drug. Ketamine produces conspicuous changes in the electrocorticographic (ECoG) signals observed both in humans and rodents. In rodents, the intracranial ECoG displays a high-frequency oscillation (HFO) which power is modulated nonlinearly by ketamine dose. Despite the widespread use of ketamine there is no model description of the relationship between the pharmacokinetic-pharmacodynamics (PK-PDs) of ketamine and the observed HFO power. APPROACH In the present study, we developed a PK-PD model based on estimated ketamine concentration, its known pharmacological actions, and observed ECoG effects. The main pharmacological action of ketamine is antagonism of the NMDA receptor (NMDAR), which in rodents is accompanied by an HFO observed in the ECoG. At high doses, however, ketamine also acts at non-NMDAR sites, produces loss of consciousness, and the transient disappearance of the HFO. We propose a two-compartment PK model that represents the concentration of ketamine, and a PD model based in opposing effects of the NMDAR and non-NMDAR actions on the HFO power. MAIN RESULTS We recorded ECoG from the cortex of rats after two doses of ketamine, and extracted the HFO power from the ECoG spectrograms. We fit the PK-PD model to the time course of the HFO power, and showed that the model reproduces the dose-dependent profile of the HFO power. The model provides good fits even in the presence of high variability in HFO power across animals. As expected, the model does not provide good fits to the HFO power after dosing the pure NMDAR antagonist MK-801. SIGNIFICANCE Our study provides a simple model to relate the observed electrophysiological effects of ketamine to its actions at the molecular level at different concentrations. This will improve the study of ketamine and rodent models of schizophrenia to better understand the wide and divergent range of effects that ketamine has.
Collapse
Affiliation(s)
- Francisco J Flores
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA. Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | | | | | | | | | | |
Collapse
|
197
|
Lodge D, Mercier MS. Ketamine and phencyclidine: the good, the bad and the unexpected. Br J Pharmacol 2015; 172:4254-76. [PMID: 26075331 DOI: 10.1111/bph.13222] [Citation(s) in RCA: 128] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Revised: 05/29/2015] [Accepted: 06/03/2015] [Indexed: 12/21/2022] Open
Abstract
The history of ketamine and phencyclidine from their development as potential clinical anaesthetics through drugs of abuse and animal models of schizophrenia to potential rapidly acting antidepressants is reviewed. The discovery in 1983 of the NMDA receptor antagonist property of ketamine and phencyclidine was a key step to understanding their pharmacology, including their psychotomimetic effects in man. This review describes the historical context and the course of that discovery and its expansion into other hallucinatory drugs. The relevance of these findings to modern hypotheses of schizophrenia and the implications for drug discovery are reviewed. The findings of the rapidly acting antidepressant effects of ketamine in man are discussed in relation to other glutamatergic mechanisms.
Collapse
Affiliation(s)
- D Lodge
- Centre for Synaptic Plasticity, School of Physiology and Pharmacology, University of Bristol, Bristol, UK
| | - M S Mercier
- Centre for Synaptic Plasticity, School of Physiology and Pharmacology, University of Bristol, Bristol, UK
| |
Collapse
|
198
|
Vreeker A, van Bergen AH, Kahn RS. Cognitive enhancing agents in schizophrenia and bipolar disorder. Eur Neuropsychopharmacol 2015; 25:969-1002. [PMID: 25957798 DOI: 10.1016/j.euroneuro.2015.04.014] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2014] [Accepted: 04/10/2015] [Indexed: 12/20/2022]
Abstract
Cognitive dysfunction is a core feature of schizophrenia and is also present in bipolar disorder (BD). Whereas decreased intelligence precedes the onset of psychosis in schizophrenia and remains relatively stable thereafter; high intelligence is a risk factor for bipolar illness but cognitive function decreases after onset of symptoms. While in schizophrenia, many studies have been conducted on the development of cognitive enhancing agents; in BD such studies are almost non-existent. This review focuses on the pharmacological agents with putative effects on cognition in both schizophrenia and bipolar illness; specifically agents targeting the dopaminergic, cholinergic and glutamatergic neurotransmitter pathways in schizophrenia and the cognitive effects of lithium, anticonvulsants and antipsychotics in BD. In the final analysis we conclude that cognitive enhancing agents have not yet been produced convincingly for schizophrenia and have hardly been studied in BD. Importantly, studies should focus on other phases of the illness. To be able to treat cognitive deficits effectively in schizophrenia, patients in the very early stages of the illness, or even before - in the ultra-high risk stages - should be targeted. In contrast, cognitive deficits occur later in BD, and therefore drugs should be tested in BD after the onset of illness. Hopefully, we will then find effective drugs for the incapacitating effects of cognitive deficits in these patients.
Collapse
Affiliation(s)
- Annabel Vreeker
- University Medical Center Utrecht, Department of Psychiatry, Brain Center Rudolf Magnus, The Netherlands
| | - Annet H van Bergen
- University Medical Center Utrecht, Department of Psychiatry, Brain Center Rudolf Magnus, The Netherlands
| | - René S Kahn
- University Medical Center Utrecht, Department of Psychiatry, Brain Center Rudolf Magnus, The Netherlands.
| |
Collapse
|
199
|
Martin MV, Mirnics K, Nisenbaum LK, Vawter MP. Olanzapine Reversed Brain Gene Expression Changes Induced by Phencyclidine Treatment in Non-Human Primates. MOLECULAR NEUROPSYCHIATRY 2015; 1:82-93. [PMID: 26405684 DOI: 10.1159/000430786] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The NMDA receptor antagonist phencyclidine (PCP) creates schizophrenia-like symptoms in normal controls. The effect of PCP on non-human primate brain gene expression was examined and compared to changes induced by olanzapine treatment. Experimental studies of PCP and antipsychotic drugs have direct relevance to understanding the patho-physiology and treatment of schizophrenia. Genome-wide changes in prefrontal cortex gene expression revealed alterations of 146 transcripts in the PCP treatment group compared to vehicle controls. Dysregulated genes were enriched in identified classes implicated in neurological and genetic disorders, including schizophrenia genes from the Psychiatric Genomics Consortium 108 loci as well as cell death in PCP-treated primates. Canonical pathway analysis revealed a significant overrepresentation of several groups including synaptic long-term potentiation and calcium signaling. Olanzapine coadministered with PCP normalized 34% of the 146 PCP-induced probe set expression changes, and a network of 17 olanzapine-normalized genes was identified enriched in schizophrenia candidate genes containing RGS4, SYN1 and AKT as nodes. The results of this study support the use of PCP administration in non-human primates as a glutamatergic model of schizophrenia and suggest that a large number of PCP-induced expression differences can be reversed by olanzapine. The results of this study may be informative for identification of potential candidates for pharmacogenetics and biomarker research related to the treatment of schizophrenia.
Collapse
Affiliation(s)
- Maureen V Martin
- Functional Genomics Laboratory, Department of Psychiatry and Human Behavior, University of California, Irvine, Irvine, Calif
| | - Karoly Mirnics
- Department of Psychiatry, Vanderbilt University, Nashville, Tenn
| | - Laura K Nisenbaum
- Neuroscience Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Ind., USA
| | - Marquis P Vawter
- Functional Genomics Laboratory, Department of Psychiatry and Human Behavior, University of California, Irvine, Irvine, Calif
| |
Collapse
|
200
|
Abstract
Although visual processing impairments are common in schizophrenia, it is not clear to what extent these originate in the eye vs. the brain. This review highlights potential contributions, from the retina and other structures of the eye, to visual processing impairments in schizophrenia and high-risk states. A second goal is to evaluate the status of retinal abnormalities as biomarkers for schizophrenia. The review was motivated by known retinal changes in other disorders (e.g., Parkinson’s disease, multiple sclerosis), and their relationships to perceptual and cognitive impairments, and disease progression therein. The evidence reviewed suggests two major conclusions. One is that there are multiple structural and functional disturbances of the eye in schizophrenia, all of which could be factors in the visual disturbances of patients. These include retinal venule widening, retinal nerve fiber layer thinning, dopaminergic abnormalities, abnormal ouput of retinal cells as measured by electroretinography (ERG), maculopathies and retinopathies, cataracts, poor acuity, and strabismus. Some of these are likely to be illness-related, whereas others may be due to medication or comorbid conditions. The second conclusion is that certain retinal findings can serve as biomarkers of neural pathology, and disease progression, in schizophrenia. The strongest evidence for this to date involves findings of widened retinal venules, thinning of the retinal nerve fiber layer, and abnormal ERG amplitudes. These data suggest that a greater understanding of the contribution of retinal and other ocular pathology to the visual and cognitive disturbances of schizophrenia is warranted, and that retinal changes have untapped clinical utility.
Collapse
|