251
|
Seeman P. Cannabidiol is a partial agonist at dopamine D2High receptors, predicting its antipsychotic clinical dose. Transl Psychiatry 2016; 6:e920. [PMID: 27754480 PMCID: PMC5315552 DOI: 10.1038/tp.2016.195] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 08/07/2016] [Accepted: 08/18/2016] [Indexed: 12/24/2022] Open
Abstract
Although all current antipsychotics act by interfering with the action of dopamine at dopamine D2 receptors, two recent reports showed that 800 to 1000 mg of cannabidiol per day alleviated the signs and symptoms of schizophrenia, although cannabidiol is not known to act on dopamine receptors. Because these recent clinical findings may indicate an important exception to the general rule that all antipsychotics interfere with dopamine at dopamine D2 receptors, the present study examined whether cannabidiol acted directly on D2 receptors, using tritiated domperidone to label rat brain striatal D2 receptors. It was found that cannabidiol inhibited the binding of radio-domperidone with dissociation constants of 11 nm at dopamine D2High receptors and 2800 nm at dopamine D2Low receptors, in the same biphasic manner as a dopamine partial agonist antipsychotic drug such as aripiprazole. The clinical doses of cannabidiol are sufficient to occupy the functional D2High sites. it is concluded that the dopamine partial agonist action of cannabidiol may account for its clinical antipsychotic effects.
Collapse
Affiliation(s)
- P Seeman
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada,Department of Pharmacology, University of Toronto, Toronto, ON, Canada,Department of Psychiatry, University of Toronto, 260 Heath Street West, Toronto, ON, Canada M5P 3L6. E-mail:
| |
Collapse
|
252
|
Maric NP, Jovicic MJ, Mihaljevic M, Miljevic C. Improving Current Treatments for Schizophrenia. Drug Dev Res 2016; 77:357-367. [DOI: 10.1002/ddr.21337] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Nadja P. Maric
- School of Medicine; University of Belgrade; Belgrade Serbia
- Clinical Centre of Serbia; Clinic for Psychiatry; Belgrade Serbia
| | | | | | - Cedo Miljevic
- School of Medicine; University of Belgrade; Belgrade Serbia
- Institute of Mental Health; Belgrade Serbia
| |
Collapse
|
253
|
Chen XW, Liu Y, Jin XQ, Sun YY, Gu SL, Fu L, Li JQ. Development and Kilogram-Scale Synthesis of a D2/5-HT2A Receptor Dual Antagonist (±)-SIPI 6360. Org Process Res Dev 2016. [DOI: 10.1021/acs.oprd.6b00220] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Xiao-Wen Chen
- School
of Pharmacy, Shanghai JiaoTong University, 800 Dongchuan Road, Shanghai 200240, P. R. China
- Novel Technology
Center of Pharmaceutical Chemistry, Shanghai Institute of Pharmaceutical
Industry, China State Institute of Pharmaceutical Industry, 285 Gebaini Road, Shanghai 201203, P. R. China
| | - Yu Liu
- Novel Technology
Center of Pharmaceutical Chemistry, Shanghai Institute of Pharmaceutical
Industry, China State Institute of Pharmaceutical Industry, 285 Gebaini Road, Shanghai 201203, P. R. China
| | - Xun-Qi Jin
- Novel Technology
Center of Pharmaceutical Chemistry, Shanghai Institute of Pharmaceutical
Industry, China State Institute of Pharmaceutical Industry, 285 Gebaini Road, Shanghai 201203, P. R. China
| | - Yuan-Yuan Sun
- Novel Technology
Center of Pharmaceutical Chemistry, Shanghai Institute of Pharmaceutical
Industry, China State Institute of Pharmaceutical Industry, 285 Gebaini Road, Shanghai 201203, P. R. China
| | - Shun-Lin Gu
- School
of Pharmaceutical Engineering and Life Science, Changzhou University, 1 Gehu Road, Changzhou, Jiangsu Province 213164, PR China
| | - Lei Fu
- School
of Pharmacy, Shanghai JiaoTong University, 800 Dongchuan Road, Shanghai 200240, P. R. China
| | - Jian-Qi Li
- Novel Technology
Center of Pharmaceutical Chemistry, Shanghai Institute of Pharmaceutical
Industry, China State Institute of Pharmaceutical Industry, 285 Gebaini Road, Shanghai 201203, P. R. China
| |
Collapse
|
254
|
De Rossi P, Chiapponi C, Spalletta G. Brain Functional Effects of Psychopharmacological Treatments in Schizophrenia: A Network-based Functional Perspective Beyond Neurotransmitter Systems. Curr Neuropharmacol 2016; 13:435-44. [PMID: 26412063 PMCID: PMC4790396 DOI: 10.2174/1570159x13666150507223542] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Psychopharmacological treatments for schizophrenia have always been a matter of debate and a very important issue in public health given the chronic, relapsing and disabling nature of the disorder. A thorough understanding of the pros and cons of currently available pharmacological treatments for schizophrenia is critical to better capture the features of treatment-refractory clinical pictures and plan the developing of new treatment strategies. This review focuses on brain functional changes induced by antipsychotic drugs as assessed by modern functional neuroimaging techniques (i.e. fMRI, PET, SPECT, MRI spectroscopy). The most important papers on this topic are reviewed in order to draw an ideal map of the main functional changes occurring in the brain during antipsychotic treatment. This supports the hypothesis that a network-based perspective and a functional connectivity approach are needed to fill the currently existing gap of knowledge in the field of psychotropic drugs and their mechanisms of action beyond neurotransmitter systems.
Collapse
Affiliation(s)
| | | | - Gianfranco Spalletta
- Neuropsychiatry Laboratory, Department of Clinical and Behavioural Neurology, IRCCS Santa Lucia Foundation, Via Ardeatina, 306, 00179 Rome, Italy.
| |
Collapse
|
255
|
Oya K, Matsuda Y, Matsunaga S, Kishi T, Iwata N. Efficacy and safety of oxytocin augmentation therapy for schizophrenia: an updated systematic review and meta-analysis of randomized, placebo-controlled trials. Eur Arch Psychiatry Clin Neurosci 2016; 266:439-50. [PMID: 26303414 DOI: 10.1007/s00406-015-0634-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 08/11/2015] [Indexed: 11/26/2022]
Abstract
The aim of this study was to perform a systematic review and an updated and comprehensive meta-analysis of oxytocin augmentation therapy in patients with schizophrenia who received antipsychotic agents. Data published up to 07/11/2015 were obtained from PubMed, PsycINFO, and Cochrane Library databases. We conducted a systematic review and meta-analysis of patients' data from randomized controlled trials (RCTs) comparing oxytocin with placebo. Relative risk (RR), standardized mean difference (SMD), and 95 % confidence intervals (95 % CI) based on the random-effects model were calculated. We included seven RCTs; the total sample size was 206 patients. Oxytocin was superior to placebo for decreasing the Positive and Negative Syndrome Scale (PANSS) general subscale scores (SMD = -0.44, 95 % CI -0.82 to -0.06, p = 0.02, I (2) = 0 %, N = 4, n = 112); however, it was not different from placebo for total symptoms (SMD = -0.46, 95 % CI -1.20 to 0.28, p = 0.22, I (2) = 80 %, N = 6, n = 162), positive symptoms (SMD = -0.18, 95 % CI -0.87 to 0.51, p = 0.60, I (2) = 81 %, N = 6, n = 192), and negative symptoms (SMD = -0.34, 95 % CI -0.76 to 0.08, p = 0.12, I (2) = 55 %, N = 7, n = 214). However, a sensitivity analysis including only oxytocin administration on consecutive days studies was superior to placebo in negative symptoms (SMD = -0.44, 95 % CI -0.87 to -0.01, p = 0.04, I (2) = 51 %, N = 6 n = 192). There were no significant differences for all-cause discontinuation (RR = 1.02) and individual side effects such as headache and dizziness between oxytocin and placebo. Oxytocin may improve PANSS general subscale scores in schizophrenia and seems to be well tolerated. However, because the number of studies in the current analysis was small, further study will be required using larger sample sizes.
Collapse
Affiliation(s)
- Kazuto Oya
- Department of Psychiatry, Fujita Health University School of Medicine, Toyoake, Aichi, 470-1192, Japan
| | - Yuki Matsuda
- Department of Psychiatry, Fujita Health University School of Medicine, Toyoake, Aichi, 470-1192, Japan
| | - Shinji Matsunaga
- Department of Psychiatry, Fujita Health University School of Medicine, Toyoake, Aichi, 470-1192, Japan
| | - Taro Kishi
- Department of Psychiatry, Fujita Health University School of Medicine, Toyoake, Aichi, 470-1192, Japan.
| | - Nakao Iwata
- Department of Psychiatry, Fujita Health University School of Medicine, Toyoake, Aichi, 470-1192, Japan
| |
Collapse
|
256
|
Moe AAK, Scott JG, Burne TH, Eyles DW. Neural changes induced by antipsychotic administration in adolescence: A review of studies in laboratory rodents. J Psychopharmacol 2016; 30:771-94. [PMID: 27413140 DOI: 10.1177/0269881116654776] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Adolescence is characterized by major remodelling processes in the brain. Use of antipsychotic drugs (APDs) in adolescents has increased dramatically in the last 20 years; however, our understanding of the neurobiological consequences of APD treatment on the adolescent brain has not kept the same pace and significant concerns have been raised. In this review, we examined currently available preclinical studies of the effects of APDs on the adolescent brain. In animal models of neuropsychiatric disorders, adolescent APD treatment appears to be protective against selected structural, behavioural and neurochemical phenotypes. In "neurodevelopmentally normal" adolescent animals, a range of short- and long-term alterations in behaviour and neurochemistry have been reported. In particular, the adolescent brain appears to be sensitive to long-term locomotor/reward effects of chronic atypical APDs in contrast with the outcomes in adults. Long-lasting changes in dopaminergic, glutamatergic and gamma-amino butyric acid-ergic systems induced by adolescent APD administration have been observed in the nucleus accumbens. A detailed examination of other potential target regions such as striatum, prefrontal cortex and ventral tegmental area is still required. Through identification of specific neural pathways targeted by adolescent APD treatment, future studies will expand the current knowledge on long-term neural outcomes which are of translational value.
Collapse
Affiliation(s)
- Aung Aung Kywe Moe
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD, Australia
| | - James G Scott
- Queensland Centre for Mental Health Research, Wacol, QLD, Australia Discipline of Psychiatry, School of Medicine, The University of Queensland Centre for Clinical Research, Herston, QLD, Australia Metro North Mental Health Service, Royal Brisbane and Women's Hospital, Herston, QLD, Australia
| | - Thomas Hj Burne
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD, Australia Queensland Centre for Mental Health Research, Wacol, QLD, Australia
| | - Darryl W Eyles
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD, Australia Queensland Centre for Mental Health Research, Wacol, QLD, Australia
| |
Collapse
|
257
|
Abstract
Schizophrenia is a complex, heterogeneous behavioural and cognitive syndrome that seems to originate from disruption of brain development caused by genetic or environmental factors, or both. Dysfunction of dopaminergic neurotransmission contributes to the genesis of psychotic symptoms, but evidence also points to a widespread and variable involvement of other brain areas and circuits. Disturbances of synaptic function might underlie abnormalities of neuronal connectivity that possibly involves interneurons, but the precise nature, location, and timing of these events are uncertain. At present, treatment mainly consists of antipsychotic drugs combined with psychological therapies, social support, and rehabilitation, but a pressing need for more effective treatments and delivery of services exists. Advances in genomics, epidemiology, and neuroscience have led to great progress in understanding the disorder, and the opportunities for further scientific breakthrough are numerous--but so are the challenges.
Collapse
Affiliation(s)
- Michael J Owen
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, UK.
| | - Akira Sawa
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Preben B Mortensen
- Department of Economics, School of Business and Social Science, Aarhus University, Aarhus, Denmark
| |
Collapse
|
258
|
Yang F, Jiang X, Li J, Wang Y, Liu Y, Bi M, Wu C, Zhao Q, Chen W, Yin J, Zhang J, Xie Y, Hu T, Xu M, Guo S, Wang Z, He Y, Shen J. Synthesis, structure–activity relationships, and biological evaluation of a series of benzamides as potential multireceptor antipsychotics. Bioorg Med Chem Lett 2016; 26:3141-3147. [DOI: 10.1016/j.bmcl.2016.04.087] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 04/27/2016] [Accepted: 04/29/2016] [Indexed: 10/21/2022]
|
259
|
Isvoranu AM, Borsboom D, van Os J, Guloksuz S. A Network Approach to Environmental Impact in Psychotic Disorder: Brief Theoretical Framework. Schizophr Bull 2016; 42:870-3. [PMID: 27179124 PMCID: PMC4903069 DOI: 10.1093/schbul/sbw049] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The spectrum of psychotic disorder represents a multifactorial and heterogeneous condition and is thought to result from a complex interplay between genetic and environmental factors. In the current paper, we analyze this interplay using network analysis, which has been recently proposed as a novel psychometric framework for the study of mental disorders. Using general population data, we construct network models for the relation between 3 environmental risk factors (cannabis use, developmental trauma, and urban environment), dimensional measures of psychopathology (anxiety, depression, interpersonal sensitivity, obsessive-compulsive disorder, phobic anxiety, somatizations, and hostility), and a composite measure of psychosis expression. Results indicate the existence of specific paths between environmental factors and symptoms. These paths most often involve cannabis use. In addition, the analyses suggest that symptom networks are more strongly connected for people exposed to environmental risk factors, implying that environmental exposure may lead to less resilient symptom networks.
Collapse
Affiliation(s)
- Adela-Maria Isvoranu
- Department of Psychology, Psychological Methods, University of Amsterdam, Amsterdam, The Netherlands;
| | - Denny Borsboom
- Department of Psychology, Psychological Methods, University of Amsterdam, Amsterdam, The Netherlands
| | - Jim van Os
- Department of Psychiatry and Psychology, School of Mental Health and Neuroscience, Maastricht University Medical Center, Maastricht, The Netherlands;,King’s College London, King’s Health Partners, Department of Psychosis Studies, Institute of Psychiatry, London, UK
| | - Sinan Guloksuz
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT
| |
Collapse
|
260
|
Frangou S. Bulking up the hippocampus in schizophrenia: a role for 5-HT 1A agonists? BJPsych Open 2016; 2:e10-e11. [PMID: 27703789 PMCID: PMC4995174 DOI: 10.1192/bjpo.bp.116.002857] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Revised: 06/27/2016] [Accepted: 06/28/2016] [Indexed: 11/23/2022] Open
Abstract
SUMMARY The volume of the hippocampus is reduced in patients with schizophrenia, and this deficit tends to become more pronounced with chronicity. Newer antipsychotics may protect against the progressive reductions in hippocampal volume while preliminary data offer hope that specific antipsychotics may act to reverse it. DECLARATION OF INTEREST None. COPYRIGHT AND USAGE © The Royal College of Psychiatrists 2016. This is an open access article distributed under the terms of the Creative Commons Non-Commercial, No Derivatives (CC BY-NC-ND) licence.
Collapse
Affiliation(s)
- Sophia Frangou
- , MD, PhD, FRCPsych, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
261
|
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: 67] [Impact Index Per Article: 7.4] [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
|
262
|
Mehta UM, Keshavan MS, Gangadhar BN. Bridging the schism of schizophrenia through yoga-Review of putative mechanisms. Int Rev Psychiatry 2016; 28:254-64. [PMID: 27187680 DOI: 10.1080/09540261.2016.1176905] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Schizophrenia patients experience a 'disconnect' at multiple levels-neuronal networks, mental processes, and interpersonal relationships. The resultant poor quality-of-life and functional disability are related to the persistent cognitive deficits and negative symptoms, which are rather resistant to conventional antipsychotic medications. Yoga has emerged as an important therapeutic intervention to improve quality-of-life in schizophrenia. Recent preliminary evidence suggests that effects of yoga on cognitive and negative symptoms may drive this benefit. This study attempts to integrate evidence from neuroscience-based research, which focuses on the neuroplasticity-harnessing effects of yoga to bridge the schizophrenia connectopathy. In an overarching model to study putative neurobiological mechanisms that drive therapeutic effects of yoga, it is proposed that (a) various styles of meditation may help in strengthening the lateral and medial prefrontal brain networks, thus improving neurocognition and mentalizing abilities, and (b) learning and performing co-ordinated physical postures with a teacher facilitates imitation and the process of being imitated, which can improve social cognition and empathy through reinforcement of the premotor and parietal mirror neuron system. Oxytocin may play a role in mediating these processes, leading to better social connectedness and social outcomes. Clinical and heuristic implications of this model are further discussed.
Collapse
Affiliation(s)
- Urvakhsh Meherwan Mehta
- a Department of Psychiatry , National Institute of Mental Health & Neurosciences (NIMHANS) , Bengaluru , India
| | - Matcheri S Keshavan
- b Department of Psychiatry , Beth Israel Deaconess Medical Center and Harvard Medical School , Boston , MA , USA
| | - Bangalore N Gangadhar
- a Department of Psychiatry , National Institute of Mental Health & Neurosciences (NIMHANS) , Bengaluru , India
| |
Collapse
|
263
|
El-Sayed El-Sisi A, Sokkar SS, El-Sayed El-Sayad M, Sayed Ramadan E, Osman EY. Celecoxib and omega-3 fatty acids alone and in combination with risperidone affect the behavior and brain biochemistry in amphetamine-induced model of schizophrenia. Biomed Pharmacother 2016; 82:425-31. [PMID: 27470381 DOI: 10.1016/j.biopha.2016.05.024] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Revised: 05/16/2016] [Accepted: 05/16/2016] [Indexed: 01/25/2023] Open
Abstract
The implications of oxidative stress and neuro-inflammation in the pathogenesis of schizophrenia have been elucidated. Despite their effectiveness against positive symptoms of schizophrenia, antipsychotics have limited effectiveness against negative and cognitive symptoms and are associated with remarkable adverse effects. The use of celecoxib or omega-3 in schizophrenia may have beneficial effects. This study aimed to evaluate the possible efficacies of celecoxib, omega-3 or the combination of celecoxib+risperidone and omega-3+ risperidone compared to risperidone on the behavior and brain biochemistry in rats. In the present study, an amphetamine-induced model of schizophrenia in adult male rats was used to evaluate the effects of celecoxib, omega-3, celecoxib+risperidone and omega-3+ risperidone on the behavior of animals and on brain lipid peroxidation or tumor necrosis factor-alpha. In the water maze task, celecoxib, omega-3, celecoxib+risperidone, omega-3+ risperidone significantly decreased the latency time compared to amphetamine-treated group. Celecoxib, omega-3, celecoxib+risperidone, omega-3+risperidone also significantly reversed the decreased spontaneous alternation induced by amphetamine in the Y-maze task. In the social interaction task, groups treated with celecoxib, omega-3, celecoxib+risperidone, omega-3+ risperidone spent less time to recognize foreign animals than animals in the amphetamine-treated group. Increased brain MDA and TNF-α levels due to amphetamine were significantly reduced in groups treated with celecoxib+risperidone or omega-3+ risperidone. The present findings showed that celecoxib or omega-3 can attenuate amphetamine- induced behavioral impairment and these effects may be associated with their ability to decrease lipid peroxidation and cytokine release. Celecoxib or omega-3 may be promising candidates as adjuvant therapy for schizophrenia.
Collapse
Affiliation(s)
- Alaa El-Sayed El-Sisi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Egypt
| | - Samia Salem Sokkar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Egypt
| | | | - Ehab Sayed Ramadan
- Department of Neuropsychiatry, Faculty of Medicine, University of Tanta, Egypt
| | - Enass Yossef Osman
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Egypt.
| |
Collapse
|
264
|
Fernandez-Egea E, Vértes PE, Flint SM, Turner L, Mustafa S, Hatton A, Smith KGC, Lyons PA, Bullmore ET. Peripheral Immune Cell Populations Associated with Cognitive Deficits and Negative Symptoms of Treatment-Resistant Schizophrenia. PLoS One 2016; 11:e0155631. [PMID: 27244229 PMCID: PMC4887013 DOI: 10.1371/journal.pone.0155631] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Accepted: 05/01/2016] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Hypothetically, psychotic disorders could be caused or conditioned by immunological mechanisms. If so, one might expect there to be peripheral immune system phenotypes that are measurable in blood cells as biomarkers of psychotic states. METHODS We used multi-parameter flow cytometry of venous blood to quantify and determine the activation state of 73 immune cell subsets for 18 patients with chronic schizophrenia (17 treated with clozapine), and 18 healthy volunteers matched for age, sex, BMI and smoking. We used multivariate methods (partial least squares) to reduce dimensionality and define populations of differentially co-expressed cell counts in the cases compared to controls. RESULTS Schizophrenia cases had increased relative numbers of NK cells, naïve B cells, CXCR5+ memory T cells and classical monocytes; and decreased numbers of dendritic cells (DC), HLA-DR+ regulatory T-cells (Tregs), and CD4+ memory T cells. Likewise, within the patient group, more severe negative and cognitive symptoms were associated with decreased relative numbers of dendritic cells, HLA-DR+ Tregs, and CD4+ memory T cells. Motivated by the importance of central nervous system dopamine signalling for psychosis, we measured dopamine receptor gene expression in separated CD4+ cells. Expression of the dopamine D3 (DRD3) receptor was significantly increased in clozapine-treated schizophrenia and covaried significantly with differentiated T cell classes in the CD4+ lineage. CONCLUSIONS Peripheral immune cell populations and dopaminergic signalling are disrupted in clozapine-treated schizophrenia. Immuno-phenotypes may provide peripherally accessible and mechanistically specific biomarkers of residual cognitive and negative symptoms in this treatment-resistant subgroup of patients.
Collapse
Affiliation(s)
- Emilio Fernandez-Egea
- NIHR Cambridge Biomedical Research Centre, Cambridge University Hospitals NHS Foundation Trust and the University of Cambridge, Cambridge, United Kingdom
- University of Cambridge, Behavioural & Clinical Neuroscience Institute, Department of Psychiatry, Cambridge, United Kingdom
- Centro de Investigación Biomedica en Red de Salud Mental (CIBERSAM), G04, Barcelona, Spain
- Cambridgeshire & Peterborough NHS Foundation Trust, Cambridge, United Kingdom
| | - Petra E. Vértes
- University of Cambridge, Behavioural & Clinical Neuroscience Institute, Department of Psychiatry, Cambridge, United Kingdom
| | - Shaun M. Flint
- Department of Medicine and Cambridge Institute for Medical Research, University of Cambridge, School of Clinical Medicine, Cambridge, United Kingdom
| | - Lorinda Turner
- NIHR Cambridge Biomedical Research Centre, Cambridge University Hospitals NHS Foundation Trust and the University of Cambridge, Cambridge, United Kingdom
- Department of Medicine and Cambridge Institute for Medical Research, University of Cambridge, School of Clinical Medicine, Cambridge, United Kingdom
| | - Syed Mustafa
- University of Cambridge, Behavioural & Clinical Neuroscience Institute, Department of Psychiatry, Cambridge, United Kingdom
| | - Alex Hatton
- Department of Medicine and Cambridge Institute for Medical Research, University of Cambridge, School of Clinical Medicine, Cambridge, United Kingdom
| | - Kenneth G. C. Smith
- NIHR Cambridge Biomedical Research Centre, Cambridge University Hospitals NHS Foundation Trust and the University of Cambridge, Cambridge, United Kingdom
- Department of Medicine and Cambridge Institute for Medical Research, University of Cambridge, School of Clinical Medicine, Cambridge, United Kingdom
| | - Paul A. Lyons
- Department of Medicine and Cambridge Institute for Medical Research, University of Cambridge, School of Clinical Medicine, Cambridge, United Kingdom
| | - Edward T. Bullmore
- NIHR Cambridge Biomedical Research Centre, Cambridge University Hospitals NHS Foundation Trust and the University of Cambridge, Cambridge, United Kingdom
- University of Cambridge, Behavioural & Clinical Neuroscience Institute, Department of Psychiatry, Cambridge, United Kingdom
- Cambridgeshire & Peterborough NHS Foundation Trust, Cambridge, United Kingdom
- GlaxoSmithKline, ImmunoPsychiatry, Alternative Discovery & Development, Pharmaceutical R&D, Cambridge, United Kingdom
| |
Collapse
|
265
|
Butini S, Nikolic K, Kassel S, Brückmann H, Filipic S, Agbaba D, Gemma S, Brogi S, Brindisi M, Campiani G, Stark H. Polypharmacology of dopamine receptor ligands. Prog Neurobiol 2016; 142:68-103. [PMID: 27234980 DOI: 10.1016/j.pneurobio.2016.03.011] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Revised: 01/26/2016] [Accepted: 03/15/2016] [Indexed: 01/11/2023]
Abstract
Most neurological diseases have a multifactorial nature and the number of molecular mechanisms discovered as underpinning these diseases is continuously evolving. The old concept of developing selective agents for a single target does not fit with the medical need of most neurological diseases. The development of designed multiple ligands holds great promises and appears as the next step in drug development for the treatment of these multifactorial diseases. Dopamine and its five receptor subtypes are intimately involved in numerous neurological disorders. Dopamine receptor ligands display a high degree of cross interactions with many other targets including G-protein coupled receptors, transporters, enzymes and ion channels. For brain disorders like Parkinsońs disease, schizophrenia and depression the dopaminergic system, being intertwined with many other signaling systems, plays a key role in pathogenesis and therapy. The concept of designed multiple ligands and polypharmacology, which perfectly meets the therapeutic needs for these brain disorders, is herein discussed as a general ligand-based concept while focusing on dopaminergic agents and receptor subtypes in particular.
Collapse
Affiliation(s)
- S Butini
- Department of Biotechnology, Chemistry and Pharmacy, European Research Centre for Drug Discovery and Development, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - K Nikolic
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11000 Belgrade, Serbia
| | - S Kassel
- Heinrich Heine University Duesseldorf, Institute of Pharmaceutical and Medicinal Chemistry, Universitaetsstr. 1, 40225 Duesseldorf, Germany
| | - H Brückmann
- Heinrich Heine University Duesseldorf, Institute of Pharmaceutical and Medicinal Chemistry, Universitaetsstr. 1, 40225 Duesseldorf, Germany
| | - S Filipic
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11000 Belgrade, Serbia
| | - D Agbaba
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11000 Belgrade, Serbia
| | - S Gemma
- Department of Biotechnology, Chemistry and Pharmacy, European Research Centre for Drug Discovery and Development, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - S Brogi
- Department of Biotechnology, Chemistry and Pharmacy, European Research Centre for Drug Discovery and Development, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - M Brindisi
- Department of Biotechnology, Chemistry and Pharmacy, European Research Centre for Drug Discovery and Development, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - G Campiani
- Department of Biotechnology, Chemistry and Pharmacy, European Research Centre for Drug Discovery and Development, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - H Stark
- Heinrich Heine University Duesseldorf, Institute of Pharmaceutical and Medicinal Chemistry, Universitaetsstr. 1, 40225 Duesseldorf, Germany.
| |
Collapse
|
266
|
Affiliation(s)
- Norah Essali
- Texila American University; 309 Waterford Place NE Atlanta Georgia USA
| | - Ibrahem Hanafi
- Damascus University; Faculty of Medicine; Mazzah Damascus Syrian Arab Republic
| | - Adib Essali
- Waikato District Health Board; Manaaki Centre; crn Rolleston and Mary Streets Thames New Zealand 3575
| |
Collapse
|
267
|
Muguruza C, Meana JJ, Callado LF. Group II Metabotropic Glutamate Receptors as Targets for Novel Antipsychotic Drugs. Front Pharmacol 2016; 7:130. [PMID: 27242534 PMCID: PMC4873505 DOI: 10.3389/fphar.2016.00130] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 05/05/2016] [Indexed: 11/13/2022] Open
Abstract
Schizophrenia is a chronic psychiatric disorder which substantially impairs patients' quality of life. Despite the extensive research in this field, the pathophysiology and etiology of schizophrenia remain unknown. Different neurotransmitter systems and functional networks have been found to be affected in the brain of patients with schizophrenia. In this context, postmortem brain studies as well as genetic assays have suggested alterations in Group II metabotropic glutamate receptors (mGluRs) in schizophrenia. Despite many years of drug research, several needs in the treatment of schizophrenia have not been addressed sufficiently. In fact, only 5-10% of patients with schizophrenia successfully achieve a full recovery after treatment. In recent years mGluRs have turned up as novel targets for the design of new antipsychotic medications for schizophrenia. Concretely, Group II mGluRs are of particular interest due to their regulatory role in neurotransmission modulating glutamatergic activity in brain synapses. Preclinical studies have demonstrated that orthosteric Group II mGluR agonists exhibit antipsychotic-like properties in animal models of schizophrenia. However, when these compounds have been tested in human clinical studies with schizophrenic patients results have been inconclusive. Nevertheless, it has been recently suggested that this apparent lack of efficacy in schizophrenic patients may be related to previous exposure to atypical antipsychotics. Moreover, the role of the functional heterocomplex formed by 5-HT2A and mGlu2 receptors in the clinical response to Group II mGluR agonists is currently under study.
Collapse
Affiliation(s)
- Carolina Muguruza
- Department of Pharmacology, University of the Basque Country, UPV/EHULeioa, Spain
- Centro de Investigación Biomédica en Red de Salud MentalMadrid, Spain
| | - J. Javier Meana
- Department of Pharmacology, University of the Basque Country, UPV/EHULeioa, Spain
- Centro de Investigación Biomédica en Red de Salud MentalMadrid, Spain
| | - Luis F. Callado
- Department of Pharmacology, University of the Basque Country, UPV/EHULeioa, Spain
- Centro de Investigación Biomédica en Red de Salud MentalMadrid, Spain
| |
Collapse
|
268
|
Zeng Z, Wang X, Bhardwaj SK, Zhou X, Little PJ, Quirion R, Srivastava LK, Zheng W. The Atypical Antipsychotic Agent, Clozapine, Protects Against Corticosterone-Induced Death of PC12 Cells by Regulating the Akt/FoxO3a Signaling Pathway. Mol Neurobiol 2016; 54:3395-3406. [DOI: 10.1007/s12035-016-9904-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 05/03/2016] [Indexed: 01/28/2023]
|
269
|
Bolkan SS, Carvalho Poyraz F, Kellendonk C. Using human brain imaging studies as a guide toward animal models of schizophrenia. Neuroscience 2016; 321:77-98. [PMID: 26037801 PMCID: PMC4664583 DOI: 10.1016/j.neuroscience.2015.05.055] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Revised: 05/15/2015] [Accepted: 05/21/2015] [Indexed: 12/17/2022]
Abstract
Schizophrenia is a heterogeneous and poorly understood mental disorder that is presently defined solely by its behavioral symptoms. Advances in genetic, epidemiological and brain imaging techniques in the past half century, however, have significantly advanced our understanding of the underlying biology of the disorder. In spite of these advances clinical research remains limited in its power to establish the causal relationships that link etiology with pathophysiology and symptoms. In this context, animal models provide an important tool for causally testing hypotheses about biological processes postulated to be disrupted in the disorder. While animal models can exploit a variety of entry points toward the study of schizophrenia, here we describe an approach that seeks to closely approximate functional alterations observed with brain imaging techniques in patients. By modeling these intermediate pathophysiological alterations in animals, this approach offers an opportunity to (1) tightly link a single functional brain abnormality with its behavioral consequences, and (2) to determine whether a single pathophysiology can causally produce alterations in other brain areas that have been described in patients. In this review we first summarize a selection of well-replicated biological abnormalities described in the schizophrenia literature. We then provide examples of animal models that were studied in the context of patient imaging findings describing enhanced striatal dopamine D2 receptor function, alterations in thalamo-prefrontal circuit function, and metabolic hyperfunction of the hippocampus. Lastly, we discuss the implications of findings from these animal models for our present understanding of schizophrenia, and consider key unanswered questions for future research in animal models and human patients.
Collapse
Affiliation(s)
- S S Bolkan
- Department of Pharmacology, Columbia University, New York, NY 10032, USA; Department of Psychiatry, Columbia University, New York, NY 10032, USA; Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, USA
| | - F Carvalho Poyraz
- Department of Pharmacology, Columbia University, New York, NY 10032, USA; Department of Psychiatry, Columbia University, New York, NY 10032, USA; Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, USA
| | - C Kellendonk
- Department of Pharmacology, Columbia University, New York, NY 10032, USA; Department of Psychiatry, Columbia University, New York, NY 10032, USA; Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, USA.
| |
Collapse
|
270
|
Keeley JW, Webb C, Peterson D, Roussin L, Flanagan EH. Development of a Response Inconsistency Scale for the Personality Inventory for DSM-5. J Pers Assess 2016; 98:351-9. [PMID: 27049169 DOI: 10.1080/00223891.2016.1158719] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
The advent of a dimensional model of personality disorder included in DSM-5 has necessitated the development of a new measurement scheme, specifically a self-report questionnaire termed the Personality Inventory for DSM-5 (PID-5; Krueger, Derringer, Markon, Watson, & Skodol, 2012 ). However, there are many threats to the validity of a self-report measure, including response inconsistency. This study outlines the development of an inconsistency scale for the PID-5. Across both college student and clinical samples, the inconsistency scale was able to reliably differentiate real from random responding. Random responses led to increased scores on the PID-5 facets, indicating the importance of detecting inconsistent responding prior to test interpretation. Thus, this inconsistency scale could be of use to researchers and clinicians in detecting inconsistent responses to this new personality disorder measure.
Collapse
Affiliation(s)
- Jared W Keeley
- a Department of Psychology , Mississippi State University
| | | | | | | | | |
Collapse
|
271
|
Brühl AB, Sahakian BJ. Drugs, games, and devices for enhancing cognition: implications for work and society. Ann N Y Acad Sci 2016; 1369:195-217. [PMID: 27043232 DOI: 10.1111/nyas.13040] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Revised: 02/15/2016] [Accepted: 02/18/2016] [Indexed: 12/11/2022]
Abstract
As work environments change, the demands on working people change. Cognitive abilities in particular are becoming progressively more important for work performance and successful competition in a global environment. However, work-related stress, performance over long hours, lack of sleep, shift work, and jet lag affect cognitive functions. Therefore, an increasing number of healthy people are reported to use cognitive-enhancing drugs, as well as other interventions, such as noninvasive brain stimulation, to maintain or improve work performance. This review summarizes research on pharmacological and technical methods as well as cognitive training, including game apps for the brain, in healthy people. In neuropsychiatric disorders, impairments in cognitive functions can drastically reduce the chances of returning to work; therefore, this review also summarizes findings from pharmacological and cognitive-training studies in neuropsychiatric disorders.
Collapse
Affiliation(s)
- Annette B Brühl
- Department of Psychiatry, and Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, United Kingdom.,Department of Psychiatry, Psychotherapy and Psychosomatics, University Hospital of Psychiatry Zurich, Zürich, Switzerland
| | - Barbara J Sahakian
- Department of Psychiatry, and Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
272
|
Johnsen E, Fathian F, Kroken RA, Steen VM, Jørgensen HA, Gjestad R, Løberg EM. The serum level of C-reactive protein (CRP) is associated with cognitive performance in acute phase psychosis. BMC Psychiatry 2016; 16:60. [PMID: 26973142 PMCID: PMC4790054 DOI: 10.1186/s12888-016-0769-x] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 03/02/2016] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Inflammatory processes have been implicated in the etiology of schizophrenia and related psychoses, in which cognitive deficits represent core symptoms. The aim of the present study was to investigate possible associations between the level of the inflammation marker C-reactive protein (CRP) and cognitive performance in patients through the acute phase of psychosis. METHODS A total of 124 patients were assessed at admittance to hospital and 62 patients were retested at discharge or after 6 weeks at the latest, with measurements of the CRP levels and alternative forms of the Repeatable Battery for the Assessment of Neuropsychological Status. RESULTS There was an inverse relationship between overall cognitive performance and CRP level at admittance. The association increased in sub-analyses including only patients with schizophrenia. In cognitive subdomain analyses statistically significant inverse associations were found between the CRP level and Delayed memory and Attention, respectively. No associations were found between CRP level and other measures of psychopathology including psychosis symptoms, depression, or functioning. At follow-up the association between CRP level and cognition was no longer present. There was a significant increase in cognitive performance between baseline and follow-up. There was a stronger increase in overall cognition scores in patients with higher baseline CRP levels. CONCLUSIONS The findings indicate that signs of inflammation may serve as a state-dependent marker of cognitive dysfunctions in acute psychosis. TRIAL REGISTRATION ClinicalTrials.gov ID; NCT00932529 , registration date: 02.07.2009.
Collapse
Affiliation(s)
- Erik Johnsen
- Division of Psychiatry, Haukeland University Hospital, Bergen, Norway. .,Department of Clinical Medicine, Section Psychiatry, University of Bergen, Bergen, Norway.
| | | | - Rune A. Kroken
- Division of Psychiatry, Haukeland University Hospital, Bergen, Norway ,Department of Clinical Medicine, Section Psychiatry, University of Bergen, Bergen, Norway
| | - Vidar M. Steen
- NORMENT and KG Jebsen Centre for Psychosis Research, Department of Clinical Science, University of Bergen, Bergen, Norway ,Dr. Einar Martens Research Group for Biological Psychiatry, Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen, Norway
| | - Hugo A. Jørgensen
- Department of Clinical Medicine, Section Psychiatry, University of Bergen, Bergen, Norway
| | - Rolf Gjestad
- Division of Psychiatry, Haukeland University Hospital, Bergen, Norway
| | - Else-Marie Løberg
- Department of Clinical Psychology, University of Bergen, Bergen, Norway ,Department of Addiction Medicine, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
273
|
Sahlholm K, Zeberg H, Nilsson J, Ögren SO, Fuxe K, Århem P. The fast-off hypothesis revisited: A functional kinetic study of antipsychotic antagonism of the dopamine D2 receptor. Eur Neuropsychopharmacol 2016; 26:467-76. [PMID: 26811292 DOI: 10.1016/j.euroneuro.2016.01.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 12/02/2015] [Accepted: 01/11/2016] [Indexed: 11/28/2022]
Abstract
Newer, "atypical" antipsychotics carry a lower risk of motor side-effects than older, "typical" compounds. It has been proposed that a ~100-fold faster dissociation from the dopamine D2 receptor (D2R) distinguishes atypical from typical antipsychotics. Furthermore, differing antipsychotic D2R affinities have been suggested to reflect differences in dissociation rate constants (koff), while association rate constants (kon) were assumed to be similar. However, it was recently demonstrated that lipophilic accumulation of ligand in the cell interior and/or membrane can cause underestimation of koff, and as high-affinity D2R antagonists are frequently lipophilic, this may have been a confounding factor in previous studies. In the present work, a functional electrophysiology assay was used to measure the recovery of dopamine-mediated D2R responsivity from antipsychotic antagonism, using elevated concentrations of dopamine to prevent the potential bias of re-binding of lipophilic ligands. The variability of antipsychotic kon was also reexamined, capitalizing on the temporal resolution of the assay. kon was estimated from the experimental recordings using a simple mathematical model assumed to describe the binding process. The time course of recovery from haloperidol (typical antipsychotic) was only 6.4- to 2.5-fold slower than that of the atypical antipsychotics, amisulpride, clozapine, and quetiapine, while antipsychotic kons were found to vary more widely than previously suggested. Finally, affinities calculated using our kon and koff estimates correlated well with functional potency and with affinities reported from radioligand binding studies. In light of these findings, it appears unlikely that typical and atypical antipsychotics are primarily distinguished by their D2R binding kinetics.
Collapse
Affiliation(s)
- Kristoffer Sahlholm
- Department of Neuroscience, Karolinska Institutet, Retzius väg 8, SE-171 77 Stockholm, Sweden.
| | - Hugo Zeberg
- Department of Neuroscience, Karolinska Institutet, Retzius väg 8, SE-171 77 Stockholm, Sweden
| | - Johanna Nilsson
- Department of Neuroscience, Karolinska Institutet, Retzius väg 8, SE-171 77 Stockholm, Sweden
| | - Sven Ove Ögren
- Department of Neuroscience, Karolinska Institutet, Retzius väg 8, SE-171 77 Stockholm, Sweden
| | - Kjell Fuxe
- Department of Neuroscience, Karolinska Institutet, Retzius väg 8, SE-171 77 Stockholm, Sweden
| | - Peter Århem
- Department of Neuroscience, Karolinska Institutet, Retzius väg 8, SE-171 77 Stockholm, Sweden
| |
Collapse
|
274
|
Abstract
Schizophrenia is a heterogeneous, debilitating disorder characterized by three distinct sets of clinical features: positive symptoms, negative symptoms, and cognitive deficits. Extant antipsychotic drugs have been most successful at treating the positive symptoms of patients with schizophrenia but have minimal therapeutic effects on negative symptoms and cognitive deficits, which are the symptoms that best predict the poor prognosis of these patients. Therefore, there has been a major effort towards identifying compounds that alleviate these symptoms. Oxytocin (OT) is a nonapeptide that regulates peripheral reproductive-relevant functions, and also acts as a neurotransmitter in the brain. Converging evidence from both preclinical and clinical research suggests that OT may have therapeutic efficacy for the positive symptoms, negative symptoms, and cognitive deficits of schizophrenia. In the majority of the small, randomized, placebo-controlled clinical trials conducted to date, OT has shown particular promise in its potential to treat the intractable negative symptoms and social cognitive deficits exhibited by most of the patients with this debilitating disorder. In this leading article, we summarize the clinical evidence relevant to (1) endogenous OT and schizophrenia, and (2) the putative therapeutic effects of OT on each of the three clinical domains.
Collapse
Affiliation(s)
- Paul D Shilling
- Department of Psychiatry, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - David Feifel
- Department of Psychiatry, University of California, San Diego Medical Center, 200 West Arbor Drive, San Diego, CA, 92103-8218, USA.
| |
Collapse
|
275
|
Edwards AC, Bigdeli TB, Docherty AR, Bacanu S, Lee D, de Candia TR, Moscati A, Thiselton DL, Maher BS, Wormley BK, Molecular Genetics of Schizophrenia Collaboration (MGS), Walsh D, O’Neill FA, Kendler KS, Riley BP, Fanous AH. Meta-analysis of Positive and Negative Symptoms Reveals Schizophrenia Modifier Genes. Schizophr Bull 2016; 42:279-87. [PMID: 26316594 PMCID: PMC4753595 DOI: 10.1093/schbul/sbv119] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Evidence suggests that genetic factors may influence both schizophrenia (Scz) and its clinical presentation. In recent years, genome-wide association studies (GWAS) have demonstrated considerable success in identifying risk loci. Detection of "modifier loci" has the potential to further elucidate underlying disease processes. METHODS We performed GWAS of empirically derived positive and negative symptom scales in Irish cases from multiply affected pedigrees and a larger, independent case-control sample, subsequently combining these into a large Irish meta-analysis. In addition to single-SNP associations, we considered gene-based and pathway analyses to better capture convergent genetic effects, and to facilitate biological interpretation of these findings. Replication and testing of aggregate genetic effects was conducted using an independent European-American sample. RESULTS Though no single marker met the genome-wide significance threshold, genes and ontologies/pathways were significantly associated with negative and positive symptoms; notably, NKAIN2 and NRG1, respectively. We observed limited overlap in ontologies/pathways associated with different symptom profiles, with immune-related categories over-represented for negative symptoms, and addiction-related categories for positive symptoms. Replication analyses suggested that genes associated with clinical presentation are generalizable to non-Irish samples. CONCLUSIONS These findings strongly support the hypothesis that modifier loci contribute to the etiology of distinct Scz symptom profiles. The finding that previously implicated "risk loci" actually influence particular symptom dimensions has the potential to better delineate the roles of these genes in Scz etiology. Furthermore, the over-representation of distinct gene ontologies/pathways across symptom profiles suggests that the clinical heterogeneity of Scz is due in part to complex and diverse genetic factors.
Collapse
Affiliation(s)
- Alexis C. Edwards
- Virginia Institute for Psychiatric and Behavioral Genetics, Department of Psychiatry, Virginia Commonwealth University School of Medicine, Richmond, VA;,*To whom correspondence should be addressed; Virginia Institute for Psychiatric and Behavioral Genetics, Department of Psychiatry, Virginia Commonwealth University School of Medicine, PO Box 980126, Richmond, VA 23298-0126, US; tel: 1-804-828-8591, fax: 1-804-828-1471, e-mail:
| | - Tim B. Bigdeli
- Virginia Institute for Psychiatric and Behavioral Genetics, Department of Psychiatry, Virginia Commonwealth University School of Medicine, Richmond, VA
| | - Anna R. Docherty
- Virginia Institute for Psychiatric and Behavioral Genetics, Department of Psychiatry, Virginia Commonwealth University School of Medicine, Richmond, VA
| | - Silviu Bacanu
- Virginia Institute for Psychiatric and Behavioral Genetics, Department of Psychiatry, Virginia Commonwealth University School of Medicine, Richmond, VA
| | - Donghyung Lee
- Virginia Institute for Psychiatric and Behavioral Genetics, Department of Psychiatry, Virginia Commonwealth University School of Medicine, Richmond, VA
| | - Teresa R. de Candia
- Department of Psychology and Neuroscience, University of Colorado, Boulder, CO;,Institute for Behavioral Genetics, University of Colorado, Boulder, CO
| | - Arden Moscati
- Virginia Institute for Psychiatric and Behavioral Genetics, Department of Psychiatry, Virginia Commonwealth University School of Medicine, Richmond, VA
| | - Dawn L. Thiselton
- Virginia Institute for Psychiatric and Behavioral Genetics, Department of Psychiatry, Virginia Commonwealth University School of Medicine, Richmond, VA;,Present address: Health Diagnostic Laboratory, Inc., Richmond, VA
| | - Brion S. Maher
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Brandon K. Wormley
- Virginia Institute for Psychiatric and Behavioral Genetics, Department of Psychiatry, Virginia Commonwealth University School of Medicine, Richmond, VA
| | | | | | - Francis A. O’Neill
- Centre for Public Health, Institute of Clinical Sciences, Queen’s University Belfast, Belfast, UK
| | - Kenneth S. Kendler
- Virginia Institute for Psychiatric and Behavioral Genetics, Department of Psychiatry, Virginia Commonwealth University School of Medicine, Richmond, VA
| | - Brien P. Riley
- Virginia Institute for Psychiatric and Behavioral Genetics, Department of Psychiatry, Virginia Commonwealth University School of Medicine, Richmond, VA
| | - Ayman H. Fanous
- Virginia Institute for Psychiatric and Behavioral Genetics, Department of Psychiatry, Virginia Commonwealth University School of Medicine, Richmond, VA;,Mental Health Service Line, Washington VA Medical Center, Washington, DC;,Department of Psychiatry, Georgetown University School of Medicine, Washington, DC
| |
Collapse
|
276
|
Iranpour N, Zandifar A, Farokhnia M, Goguol A, Yekehtaz H, Khodaie-Ardakani MR, Salehi B, Esalatmanesh S, Zeionoddini A, Mohammadinejad P, Zeinoddini A, Akhondzadeh S. The effects of pioglitazone adjuvant therapy on negative symptoms of patients with chronic schizophrenia: a double-blind and placebo-controlled trial. Hum Psychopharmacol 2016; 31:103-12. [PMID: 26856695 DOI: 10.1002/hup.2517] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2015] [Revised: 11/23/2015] [Accepted: 12/09/2015] [Indexed: 01/03/2023]
Abstract
OBJECTIVE The evident central role of inflammation, oxidative stress, and metabolic derangement in pathophysiology of negative symptoms of schizophrenia has opened new insights into probable pharmacological options for these symptoms. Pioglitazone is an antidiabetic agent with anti-inflammatory and antioxidant properties. In this study, we evaluated the efficacy of pioglitazone as an adjunct to risperidone for reduction of negative symptoms in schizophrenia. METHODS In this randomized, double-blind, placebo-controlled trial, 40 patients with chronic schizophrenia and a minimum score of 20 on the negative subscale of Positive and Negative Syndrome Scale (PANSS) were randomly allocated to receive risperidone plus either pioglitazone (30 mg/day) or placebo for 8 weeks. Patients' symptoms and adverse events were rated at baseline and weeks 2, 4, 6, and 8. The difference between the two groups in decline of PANSS negative subscale scores was considered as the primary outcome of this study. RESULTS At the study endpoint, patients in the pioglitazone group showed significantly more improvement in PANSS negative subscale scores (p < 0.001) as well as PANSS total scores (p = 0.01) compared with the placebo group. CONCLUSION These findings suggest the probable efficacy of pioglitazone as an augmentation therapy in reducing the negative symptoms of schizophrenia.
Collapse
Affiliation(s)
- Negar Iranpour
- Psychiatric Research Center, Roozbeh Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Atefeh Zandifar
- Psychiatric Research Center, Roozbeh Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehdi Farokhnia
- Psychiatric Research Center, Roozbeh Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Amirhossein Goguol
- Psychiatric Research Center, Roozbeh Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Habibeh Yekehtaz
- Psychiatric Research Center, Roozbeh Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Bahman Salehi
- Department of Psychiatry, Arak University of Medical Sciences, Arak, Iran
| | - Sophia Esalatmanesh
- Psychiatric Research Center, Roozbeh Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Atefeh Zeionoddini
- Psychiatric Research Center, Roozbeh Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Payam Mohammadinejad
- Psychiatric Research Center, Roozbeh Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Arefeh Zeinoddini
- Psychiatric Research Center, Roozbeh Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Shahin Akhondzadeh
- Psychiatric Research Center, Roozbeh Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
277
|
Davidson M, Kapara O, Goldberg S, Yoffe R, Noy S, Weiser M. A Nation-Wide Study on the Percentage of Schizophrenia and Bipolar Disorder Patients Who Earn Minimum Wage or Above. Schizophr Bull 2016; 42:443-7. [PMID: 25796051 PMCID: PMC4753584 DOI: 10.1093/schbul/sbv023] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Although it is undisputable that patients with severe mental illness have impaired ability to work, the extent of this is unclear. This is a nation-wide, cross-sectional survey of patients who have been hospitalized with severe mental illness earning minimum wage or above. METHOD Data from the Israeli Psychiatric Hospitalization Case Registry were linked with nation-wide data from the National Insurance Institute (the equivalent of US Social Security) on personal income. Hospitalization data were obtained on all consecutive admissions to any psychiatric hospital in the country between 1990-2008 with a diagnosis of schizophrenia, other nonaffective psychotic disorders, or bipolar disorder (N = 35 673). Earning minimum wage or more was defined as earning at least 1000 USD/month, which was equivalent to minimum wage in Israel in December 2010. RESULTS The percentages of patients with only 1 admission who were earning minimum wage or above in December 2010 were as follows: 10.6% of patients with a diagnosis of schizophrenia; 21.6% of patients with a diagnosis of nonaffective psychotic disorders; and 24.2% of patients with bipolar disorder. The percentages of patients with multiple admissions who were earning minimum wage or above were as follows: 5.8% of patients with schizophrenia; 11.2% of patients with nonaffective psychotic disorders; and 19.9% of patients with bipolar disorder. CONCLUSIONS Despite potential confounders, the results indicate that patients with schizophrenia, nonaffective psychotic disorders, or bipolar disorder have a poor employment outcome, even if they have only been admitted once. These results emphasize the importance of improving interventions to re-integrate these individuals into the work force.
Collapse
Affiliation(s)
- Michael Davidson
- Department of Psychiatry, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel; Sackler School of Medicine, Tel Aviv University, Ramat Aviv, Israel
| | - Ori Kapara
- Department of Psychiatry, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - Shira Goldberg
- Department of Psychiatry, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - Rinat Yoffe
- Department of Mental Health, Ministry of Health, Jerusalem, Israel
| | - Shlomo Noy
- Department of Psychiatry, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel; Sackler School of Medicine, Tel Aviv University, Ramat Aviv, Israel
| | - Mark Weiser
- Department of Psychiatry, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel; Sackler School of Medicine, Tel Aviv University, Ramat Aviv, Israel;
| |
Collapse
|
278
|
Aripiprazole versus chlorpromazine for people with schizophrenia and schizophrenia-like psychoses. Hippokratia 2016. [DOI: 10.1002/14651858.cd012072] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
279
|
Park SM, Chen M, Schmerberg CM, Dulman RS, Rodriguiz RM, Caron MG, Jin J, Wetsel WC. Effects of β-Arrestin-Biased Dopamine D2 Receptor Ligands on Schizophrenia-Like Behavior in Hypoglutamatergic Mice. Neuropsychopharmacology 2016; 41:704-15. [PMID: 26129680 PMCID: PMC4707817 DOI: 10.1038/npp.2015.196] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 06/01/2015] [Accepted: 06/02/2015] [Indexed: 01/20/2023]
Abstract
Current antipsychotic drugs (APDs) show efficacy with positive symptoms, but are limited in treating negative or cognitive features of schizophrenia. Whereas all currently FDA-approved medications target primarily the dopamine D2 receptor (D2R) to inhibit G(i/o)-mediated adenylyl cyclase, a recent study has shown that many APDs affect not only G(i/o)- but they can also influence β-arrestin- (βArr)-mediated signaling. The ability of ligands to differentially affect signaling through these pathways is termed functional selectivity. We have developed ligands that are devoid of D2R-mediated G(i/o) protein signaling, but are simultaneously partial agonists for D2R/βArr interactions. The purpose of this study was to test the effectiveness of UNC9975 or UNC9994 on schizophrenia-like behaviors in phencyclidine-treated or NR1-knockdown hypoglutamatergic mice. We have found the UNC compounds reduce hyperlocomotion in the open field, restore PPI, improve novel object recognition memory, partially normalize social behavior, decrease conditioned avoidance responding, and elicit a much lower level of catalepsy than haloperidol. These preclinical results suggest that exploitation of functional selectivity may provide unique opportunities to develop drugs with fewer side effects, greater therapeutic selectivity, and enhanced efficacy for treating schizophrenia and related conditions than medications that are currently available.
Collapse
Affiliation(s)
- Su M Park
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
| | - Meng Chen
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
| | - Claire M Schmerberg
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
| | - Russell S Dulman
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
| | - Ramona M Rodriguiz
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA,Mouse Behavioral and Neuroendocrine Analysis Core Facility, Duke University Medical Center, Durham, NC, USA
| | - Marc G Caron
- Department of Cell Biology, Duke University Medical Center, Durham, NC, USA
| | - Jian Jin
- Departments of Structural and Chemical Biology, Oncological Sciences, and Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - William C Wetsel
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA,Mouse Behavioral and Neuroendocrine Analysis Core Facility, Duke University Medical Center, Durham, NC, USA,Department of Cell Biology, Duke University Medical Center, Durham, NC, USA,Department of Neurobiology, Duke University Medical Center, Durham, NC, USA,Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, 354 Sands Building, PO Box 103203, Durham, NC 27705, USA, Tel: +1 919 684 4574, E-mail:
| |
Collapse
|
280
|
Feifel D, Shilling PD, MacDonald K. A Review of Oxytocin's Effects on the Positive, Negative, and Cognitive Domains of Schizophrenia. Biol Psychiatry 2016; 79:222-33. [PMID: 26410353 PMCID: PMC5673255 DOI: 10.1016/j.biopsych.2015.07.025] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 07/16/2015] [Accepted: 07/31/2015] [Indexed: 02/06/2023]
Abstract
Schizophrenia is a disabling, heterogeneous disorder with clinical features that can be parsed into three domains: positive symptoms, negative symptoms, and cognitive deficits. Current antipsychotic drugs produce fairly robust clinical benefit against positive symptoms but typically have minimal therapeutic effects on negative symptoms and cognitive deficits. Oxytocin (OT) is a nonapeptide that, in addition to its role as a hormone regulating peripheral reproductive-relevant functions, acts as a neurotransmitter in the brain. Several lines of preclinical and clinical research suggest that the OT system may play a role in regulating the expression of schizophrenia spectrum disorders and that targeting the central OT system may yield novel treatments to address these symptoms. In this review, we summarize the extant preclinical and clinical evidence relevant to the role of OT in schizophrenia with particular emphasis on its putative therapeutic effects on each of the three above-mentioned clinical domains.
Collapse
Affiliation(s)
- David Feifel
- Department of Psychiatry, University of California, San Diego, La Jolla, California.
| | - Paul D Shilling
- Department of Psychiatry, University of California, San Diego, La Jolla, California
| | - Kai MacDonald
- Department of Psychiatry, University of California, San Diego, La Jolla, California
| |
Collapse
|
281
|
Halene TB, Kozlenkov A, Jiang Y, Mitchell A, Javidfar B, Dincer A, Park R, Wiseman J, Croxson P, Giannaris EL, Hof PR, Roussos P, Dracheva S, Hemby SE, Akbarian S. NeuN+ neuronal nuclei in non-human primate prefrontal cortex and subcortical white matter after clozapine exposure. Schizophr Res 2016; 170:235-44. [PMID: 26776227 PMCID: PMC4740223 DOI: 10.1016/j.schres.2015.12.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 12/24/2015] [Accepted: 12/28/2015] [Indexed: 12/01/2022]
Abstract
Increased neuronal densities in subcortical white matter have been reported for some cases with schizophrenia. The underlying cellular and molecular mechanisms remain unresolved. We exposed 26 young adult macaque monkeys for 6 months to either clozapine, haloperidol or placebo and measured by structural MRI frontal gray and white matter volumes before and after treatment, followed by observer-independent, flow-cytometry-based quantification of neuronal and non-neuronal nuclei and molecular fingerprinting of cell-type specific transcripts. After clozapine exposure, the proportion of nuclei expressing the neuronal marker NeuN increased by approximately 50% in subcortical white matter, in conjunction with a more subtle and non-significant increase in overlying gray matter. Numbers and proportions of nuclei expressing the oligodendrocyte lineage marker, OLIG2, and cell-type specific RNA expression patterns, were maintained after antipsychotic drug exposure. Frontal lobe gray and white matter volumes remained indistinguishable between antipsychotic-drug-exposed and control groups. Chronic clozapine exposure increases the proportion of NeuN+ nuclei in frontal subcortical white matter, without alterations in frontal lobe volumes or cell type-specific gene expression. Further exploration of neurochemical plasticity in non-human primate brain exposed to antipsychotic drugs is warranted.
Collapse
Affiliation(s)
- Tobias B. Halene
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA,Corresponding author: Tobias B. Halene, MD PhD, Icahn School of Medicine at Mount Sinai, Department of Psychiatry, 1470 Madison Ave, Hess 9-105, New York, NY 10029, Tel: 646 627 5529, Fax: 646-537-9583,
| | - Alexey Kozlenkov
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yan Jiang
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Amanda Mitchell
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Behnam Javidfar
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Aslihan Dincer
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Royce Park
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jennifer Wiseman
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Paula Croxson
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Eustathia Lela Giannaris
- Department of Cell and Developmental Biology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Patrick R. Hof
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Panos Roussos
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA,Department of Genetics and Genomic Science and Institute for Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Stella Dracheva
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Scott E. Hemby
- Department of Physiology and Pharmacology, Wake Forest University, Winston-Salem, NC, USA
| | - Schahram Akbarian
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
282
|
Watson DJG, King MV, Gyertyán I, Kiss B, Adham N, Fone KCF. The dopamine D₃-preferring D₂/D₃ dopamine receptor partial agonist, cariprazine, reverses behavioural changes in a rat neurodevelopmental model for schizophrenia. Eur Neuropsychopharmacol 2016; 26:208-224. [PMID: 26723167 DOI: 10.1016/j.euroneuro.2015.12.020] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 11/19/2015] [Accepted: 12/04/2015] [Indexed: 02/07/2023]
Abstract
Current antipsychotic medication is largely ineffective against the negative and cognitive symptoms of schizophrenia. One promising therapeutic development is to design new molecules that balance actions on dopamine D2 and D3 receptors to maximise benefits and limit adverse effects. This study used two rodent paradigms to investigate the action of the dopamine D3-preferring D3/D2 receptor partial agonist cariprazine. In adult male rats, cariprazine (0.03-0.3 mg/kg i.p.), and the atypical antipsychotic aripiprazole (1-3 mg/kg i.p.) caused dose-dependent reversal of a delay-induced impairment in novel object recognition (NOR). Treating neonatal rat pups with phencyclidine (PCP) and subsequent social isolation produced a syndrome of behavioural alterations in adulthood including hyperactivity in a novel arena, deficits in NOR and fear motivated learning and memory, and a reduction and change in pattern of social interaction accompanied by increased ultrasonic vocalisations (USVs). Acute administration of cariprazine (0.1 and 0.3 mg/kg) and aripiprazole (3 mg/kg) to resultant adult rats reduced neonatal PCP-social isolation induced locomotor hyperactivity and reversed NOR deficits. Cariprazine (0.3 mg/kg) caused a limited reversal of the social interaction deficit but neither drug affected the change in USVs or the deficit in fear motivated learning and memory. Results suggest that in the behavioural tests investigated cariprazine is at least as effective as aripiprazole and in some paradigms it showed additional beneficial features further supporting the advantage of combined dopamine D3/D2 receptor targeting. These findings support recent clinical studies demonstrating the efficacy of cariprazine in treatment of negative symptoms and functional impairment in schizophrenia patients.
Collapse
Affiliation(s)
- David J G Watson
- School of Life Sciences, Queen׳s Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK
| | - Madeleine V King
- School of Life Sciences, Queen׳s Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK
| | - Istvan Gyertyán
- Pharmacological and Safety Research, Gedeon Richter Plc, Gyömrői út 19-21, Budapest H-1103 Hungary
| | - Béla Kiss
- Pharmacological and Safety Research, Gedeon Richter Plc, Gyömrői út 19-21, Budapest H-1103 Hungary
| | - Nika Adham
- Forest Research Institute, Inc., Harborside Financial Center, Plaza V, Jersey City, NJ 07311, USA
| | - Kevin C F Fone
- School of Life Sciences, Queen׳s Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK.
| |
Collapse
|
283
|
Lemercier CE, Schulz SB, Heidmann KE, Kovács R, Gerevich Z. Dopamine D3 Receptors Inhibit Hippocampal Gamma Oscillations by Disturbing CA3 Pyramidal Cell Firing Synchrony. Front Pharmacol 2016; 6:297. [PMID: 26779018 PMCID: PMC4702013 DOI: 10.3389/fphar.2015.00297] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 12/03/2015] [Indexed: 11/13/2022] Open
Abstract
Cortical gamma oscillations are associated with cognitive processes and are altered in several neuropsychiatric conditions such as schizophrenia and Alzheimer’s disease. Since dopamine D3 receptors are possible targets in treatment of these conditions, it is of great importance to understand their role in modulation of gamma oscillations. The effect of D3 receptors on gamma oscillations and the underlying cellular mechanisms were investigated by extracellular local field potential and simultaneous intracellular sharp micro-electrode recordings in the CA3 region of the hippocampus in vitro. D3 receptors decreased the power and broadened the bandwidth of gamma oscillations induced by acetylcholine or kainate. Blockade of the D3 receptors resulted in faster synchronization of the oscillations, suggesting that endogenous dopamine in the hippocampus slows down the dynamics of gamma oscillations by activation of D3 receptors. Investigating the underlying cellular mechanisms for these effects showed that D3 receptor activation decreased the rate of action potentials (APs) during gamma oscillations and reduced the precision of the AP phase coupling to the gamma cycle in CA3 pyramidal cells. The results may offer an explanation how selective activation of D3 receptors may impair cognition and how, in converse, D3 antagonists may exert pro-cognitive and antipsychotic effects.
Collapse
Affiliation(s)
- Clément E Lemercier
- Institute of Neurophysiology, Charité - Universitätsmedizin Berlin Berlin, Germany
| | - Steffen B Schulz
- Institute of Neurophysiology, Charité - Universitätsmedizin Berlin Berlin, Germany
| | - Karin E Heidmann
- Institute of Neurophysiology, Charité - Universitätsmedizin Berlin Berlin, Germany
| | - Richard Kovács
- Institute of Neurophysiology, Charité - Universitätsmedizin Berlin Berlin, Germany
| | - Zoltan Gerevich
- Institute of Neurophysiology, Charité - Universitätsmedizin Berlin Berlin, Germany
| |
Collapse
|
284
|
Electrochemical detection of nanomolar dopamine in the presence of neurophysiological concentration of ascorbic acid and uric acid using charge-coated carbon nanotubes via facile and green preparation. Talanta 2016; 147:453-9. [DOI: 10.1016/j.talanta.2015.10.020] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 10/06/2015] [Accepted: 10/07/2015] [Indexed: 12/23/2022]
|
285
|
Li P, Snyder GL, Vanover KE. Dopamine Targeting Drugs for the Treatment of Schizophrenia: Past, Present and Future. Curr Top Med Chem 2016; 16:3385-3403. [PMID: 27291902 PMCID: PMC5112764 DOI: 10.2174/1568026616666160608084834] [Citation(s) in RCA: 170] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 05/20/2016] [Accepted: 05/23/2016] [Indexed: 01/18/2023]
Abstract
Schizophrenia is a chronic and debilitating neuropsychiatric disorder affecting approximately 1% of the world's population. This disease is associated with considerable morbidity placing a major financial burden on society. Antipsychotics have been the mainstay of the pharmacological treatment of schizophrenia for decades. The traditional typical and atypical antipsychotics demonstrate clinical efficacy in treating positive symptoms, such as hallucinations and delusions, while are largely ineffective and may worsen negative symptoms, such as blunted affect and social withdrawal, as well as cognitive function. The inability to treat these latter symptoms may contribute to social function impairment associated with schizophrenia. The dysfunction of multiple neurotransmitter systems in schizophrenia suggests that drugs selectively targeting one neurotransmission pathway are unlikely to meet all the therapeutic needs of this heterogeneous disorder. Often, however, the unintentional engagement of multiple pharmacological targets or even the excessive engagement of intended pharmacological targets can lead to undesired consequences and poor tolerability. In this article, we will review marketed typical and atypical antipsychotics and new therapeutic agents targeting dopamine receptors and other neurotransmitters for the treatment of schizophrenia. Representative typical and atypical antipsychotic drugs and new investigational drug candidates will be systematically reviewed and compared by reviewing structure-activity relationships, pharmacokinetic properties, drug metabolism and safety, pharmacological properties, preclinical data in animal models, clinical outcomes and associated side effects.
Collapse
Affiliation(s)
- Peng Li
- Intra-Cellular Therapies Inc, 430 East 29th Street, Suite 900, New York, NY 10016, United States.
| | | | | |
Collapse
|
286
|
Peltola MA, Kuja-Panula J, Liuhanen J, Võikar V, Piepponen P, Hiekkalinna T, Taira T, Lauri SE, Suvisaari J, Kulesskaya N, Paunio T, Rauvala H. AMIGO-Kv2.1 Potassium Channel Complex Is Associated With Schizophrenia-Related Phenotypes. Schizophr Bull 2016; 42:191-201. [PMID: 26240432 PMCID: PMC4681558 DOI: 10.1093/schbul/sbv105] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The enormous variability in electrical properties of neurons is largely affected by a multitude of potassium channel subunits. Kv2.1 is a widely expressed voltage-dependent potassium channel and an important regulator of neuronal excitability. The Kv2.1 auxiliary subunit AMIGO constitutes an integral part of the Kv2.1 channel complex in brain and regulates the activity of the channel. AMIGO and Kv2.1 localize to the distinct somatodendritic clusters at the neuronal plasma membrane. Here we have created and characterized a mouse line lacking the AMIGO gene. Absence of AMIGO clearly reduced the amount of the Kv2.1 channel protein in mouse brain and altered the electrophysiological properties of neurons. These changes were accompanied by behavioral and pharmacological abnormalities reminiscent of those identified in schizophrenia. Concomitantly, we have detected an association of a rare, population-specific polymorphism of KV2.1 (KCNB1) with human schizophrenia in a genetic isolate enriched with schizophrenia. Our study demonstrates the involvement of AMIGO-Kv2.1 channel complex in schizophrenia-related behavioral domains in mice and identifies KV2.1 (KCNB1) as a strong susceptibility gene for schizophrenia spectrum disorders in humans.
Collapse
Affiliation(s)
- Marjaana A. Peltola
- Neuroscience Center, University of Helsinki, Helsinki, Finland;,*To whom correspondence should be addressed; Neuroscience Center, PO Box 56 (Viikinkaari 4), FI-00014 University of Helsinki, Helsinki, Finland; tel: +358-2941-57649, fax: +358-2941-57620, e-mail:
| | | | - Johanna Liuhanen
- Public Health Genomics Unit, National Institute for Health and Welfare, Helsinki, Finland
| | - Vootele Võikar
- Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - Petteri Piepponen
- Division of Pharmacology and Toxicology, University of Helsinki, Helsinki, Finland
| | - Tero Hiekkalinna
- Public Health Genomics Unit, National Institute for Health and Welfare, Helsinki, Finland;,Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Tomi Taira
- Neuroscience Center, University of Helsinki, Helsinki, Finland;,Department of Biosciences, University of Helsinki, Helsinki, Finland;,Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
| | - Sari E. Lauri
- Neuroscience Center, University of Helsinki, Helsinki, Finland;,Department of Biosciences, University of Helsinki, Helsinki, Finland
| | - Jaana Suvisaari
- Department of Mental Health and Substance Abuse Services, National Institute for Health and Welfare, Helsinki, Finland
| | | | - Tiina Paunio
- Public Health Genomics Unit, National Institute for Health and Welfare, Helsinki, Finland;,Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland;,Department of Psychiatry, Institute of Clinical Medicine, University of Helsinki, Helsinki, Finland
| | - Heikki Rauvala
- Neuroscience Center, University of Helsinki, Helsinki, Finland
| |
Collapse
|
287
|
Design, synthesis and evaluation of bitopic arylpiperazinephenyl-1,2,4-oxadiazoles as preferential dopamine D3 receptor ligands. Bioorg Med Chem 2016; 24:191-200. [DOI: 10.1016/j.bmc.2015.12.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 11/30/2015] [Accepted: 12/01/2015] [Indexed: 01/11/2023]
|
288
|
Zink M, Correll CU. Glutamatergic agents for schizophrenia: current evidence and perspectives. Expert Rev Clin Pharmacol 2015; 8:335-52. [PMID: 25916667 DOI: 10.1586/17512433.2015.1040393] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Suboptimal outcomes in schizophrenia are a consequence of lacking insight into the etiology, biomarkers and treatment-relevant subgroups, the therapeutic restriction to dopaminergic-modulating antipsychotics that fail to significantly improve negative and cognitive symptoms, non-adherence, and, in the case of treatment-resistance, the underutilization of clozapine. Evidence suggests additional, extra-dopaminergic abnormalities in amino acid neurotransmission, particularly the glutamatergic system. Antidopaminergic antipsychotics modulate this system on several levels, as do mood stabilizers, including lamotrigine, topiramate and pregabaline. Recently, agonists at metabotropic glutamate receptors and glycine uptake inhibitors failed in large placebo-controlled trials for schizophrenia. Problems to overcome for successfully leveraging glutamatergic agents for schizophrenia are patient selection, focus on positive symptoms and late disease stages, and dose-response relationships. Because glutamate guides processes of brain development and maturation, clinical research should focus on the at-risk mental state or first-episode psychosis, address cognition and negative symptoms and use monotherapy designs in parallel to augmentation strategies.
Collapse
Affiliation(s)
- Mathias Zink
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | | |
Collapse
|
289
|
Brown MA, Bishnoi RJ, Dholakia S, Velligan DI. Methodological issues associated with preclinical drug development and increased placebo effects in schizophrenia clinical trials. Expert Rev Clin Pharmacol 2015; 9:591-604. [PMID: 26696325 DOI: 10.1586/17512433.2016.1135734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Recent failures to detect efficacy in clinical trials investigating pharmacological treatments for schizophrenia raise concerns regarding the potential contribution of methodological shortcomings to this research. This review provides an examination of two key methodological issues currently suspected of playing a role in hampering schizophrenia drug development; 1) limitations on the translational utility of preclinical development models, and 2) methodological challenges posed by increased placebo effects. Recommendations for strategies to address these methodological issues are addressed.
Collapse
Affiliation(s)
- Matt A Brown
- a Department of Psychiatry , University of Texas Health Science Center at San Antonio , San Antonio , TX , USA
| | - Ram J Bishnoi
- a Department of Psychiatry , University of Texas Health Science Center at San Antonio , San Antonio , TX , USA
| | - Sara Dholakia
- a Department of Psychiatry , University of Texas Health Science Center at San Antonio , San Antonio , TX , USA
| | - Dawn I Velligan
- a Department of Psychiatry , University of Texas Health Science Center at San Antonio , San Antonio , TX , USA
| |
Collapse
|
290
|
Kedracka-Krok S, Swiderska B, Jankowska U, Skupien-Rabian B, Solich J, Dziedzicka-Wasylewska M. Stathmin reduction and cytoskeleton rearrangement in rat nucleus accumbens in response to clozapine and risperidone treatment - Comparative proteomic study. Neuroscience 2015; 316:63-81. [PMID: 26708747 DOI: 10.1016/j.neuroscience.2015.12.028] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 12/02/2015] [Accepted: 12/14/2015] [Indexed: 11/17/2022]
Abstract
The complex network of anatomical connections of the nucleus accumbens (NAc) makes it an interface responsible for the selection and integration of cognitive and affective information to modulate appetitive or aversively motivated behaviour. There is evidence for NAc dysfunction in schizophrenia. NAc also seems to be important for antipsychotic drug action, but the biochemical characteristics of drug-induced alterations within NAc remain incompletely characterized. In this study, a comprehensive proteomic analysis was performed to describe the differences in the mechanisms of action of clozapine (CLO) and risperidone (RIS) in the rat NAc. Both antipsychotics influenced the level of microtubule-regulating proteins, i.e., stathmin, and proteins of the collapsin response mediator protein family (CRMPs), and only CLO affected NAD-dependent protein deacetylase sirtuin-2 and septin 6. Both antipsychotics induced changes in levels of other cytoskeleton-related proteins. CLO exclusively up-regulated proteins involved in neuroprotection, such as glutathione synthetase, heat-shock 70-kDa protein 8 and mitochondrial heat-shock protein 75. RIS tuned cell function by changing the pattern of post-translational modifications of some proteins: it down-regulated the phosphorylated forms of stathmin and dopamine and the cyclic AMP-regulated phosphoprotein (DARPP-32) isoform but up-regulated cyclin-dependent kinase 5 (Cdk5). RIS modulated the level and phosphorylation state of synaptic proteins: synapsin-2, synaptotagmin-1 and adaptor-related protein-2 (AP-2) complex.
Collapse
Affiliation(s)
- S Kedracka-Krok
- Department of Physical Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland; Department of Structural Biology, Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland.
| | - B Swiderska
- Department of Physical Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - U Jankowska
- Department of Structural Biology, Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
| | - B Skupien-Rabian
- Department of Physical Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - J Solich
- Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - M Dziedzicka-Wasylewska
- Department of Physical Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland; Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| |
Collapse
|
291
|
Sánchez-González A, Esnal A, Río-Álamos C, Oliveras I, Cañete T, Blázquez G, Tobeña A, Fernández-Teruel A. Association between prepulse inhibition of the startle response and latent inhibition of two-way avoidance acquisition: A study with heterogeneous NIH-HS rats. Physiol Behav 2015; 155:195-201. [PMID: 26700617 DOI: 10.1016/j.physbeh.2015.12.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 12/10/2015] [Accepted: 12/12/2015] [Indexed: 12/20/2022]
Abstract
This study presents the first evaluation of the associations between responses in two paradigms related to schizophrenia in the genetically heterogeneous NIH-HS rat stock. NIH-HS rats are a stock of genetically heterogeneous animals that have been derived from eight different inbred strains. A rotational breeding schedule has been followed for more than eighty generations, leading to a high level of genetic recombination that makes the NIH-HS rats a unique tool for studying the genetic basis of (biological, behavioral, disease-related) complex traits. Previous work has dealt with the characterization of coping styles, cognitive and anxiety/fear-related profiles of NIH-HS rats. In the present study we have completed their characterization in two behavioral models, prepulse inhibition (PPI) and latent inhibition (LI) of the two-way active avoidance response, that appear to be related to schizophrenia or to schizophrenia-relevant symptoms. We have found that these rats display PPI for each of the four prepulse intensities tested, allowing their stratification in high, medium and low PPI subgroups. When testing these three subgroups for LI of two-way active avoidance acquisition it has been observed that the LowPPI and MediumPPI subgroups present impaired LI, which, along with the fact that the HighPPI group presents significant LI, allows us to hypothesize that responses in these two paradigms are somehow related and that selection of NIH-HS rats for Low vs HighPPI could make a promising animal model for the study of clusters of schizophrenia-relevant symptoms and their underlying neurobiological mechanisms.
Collapse
Affiliation(s)
- Ana Sánchez-González
- Medical Psychology Unit, Department of Psychiatry & Forensic Medicine, Institute of Neurosciences, Universitat Autònoma de Barcelona, 08193-Bellaterra, Barcelona, Spain.
| | - Aitor Esnal
- Medical Psychology Unit, Department of Psychiatry & Forensic Medicine, Institute of Neurosciences, Universitat Autònoma de Barcelona, 08193-Bellaterra, Barcelona, Spain
| | - Cristóbal Río-Álamos
- Medical Psychology Unit, Department of Psychiatry & Forensic Medicine, Institute of Neurosciences, Universitat Autònoma de Barcelona, 08193-Bellaterra, Barcelona, Spain
| | - Ignasi Oliveras
- Medical Psychology Unit, Department of Psychiatry & Forensic Medicine, Institute of Neurosciences, Universitat Autònoma de Barcelona, 08193-Bellaterra, Barcelona, Spain
| | - Toni Cañete
- Medical Psychology Unit, Department of Psychiatry & Forensic Medicine, Institute of Neurosciences, Universitat Autònoma de Barcelona, 08193-Bellaterra, Barcelona, Spain
| | - Gloria Blázquez
- Medical Psychology Unit, Department of Psychiatry & Forensic Medicine, Institute of Neurosciences, Universitat Autònoma de Barcelona, 08193-Bellaterra, Barcelona, Spain
| | - Adolf Tobeña
- Medical Psychology Unit, Department of Psychiatry & Forensic Medicine, Institute of Neurosciences, Universitat Autònoma de Barcelona, 08193-Bellaterra, Barcelona, Spain
| | - Alberto Fernández-Teruel
- Medical Psychology Unit, Department of Psychiatry & Forensic Medicine, Institute of Neurosciences, Universitat Autònoma de Barcelona, 08193-Bellaterra, Barcelona, Spain.
| |
Collapse
|
292
|
Carboni L, Domenici E. Proteome effects of antipsychotic drugs: Learning from preclinical models. Proteomics Clin Appl 2015; 10:430-41. [PMID: 26548651 DOI: 10.1002/prca.201500087] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 10/27/2015] [Accepted: 11/03/2015] [Indexed: 02/02/2023]
Abstract
Proteome-wide expression analyses are performed in the brain of schizophrenia patients to understand the biological basis of the disease and discover molecular paths for new clinical interventions. A major issue with postmortem analysis is the lack of tools to discern molecular modulation related to the disease from dysregulation due to medications. We review available proteome-wide analysis of antipsychotic treatment in rodents, highlighting shared dysregulated pathways that may contribute to an extended view of molecular processes underlying their pharmacological activity. Fourteen proteomic studies conducted with typical and atypical antipsychotic treatments were examined; hypothesis-based approaches are also briefly discussed. Treatment with antipsychotics mainly affects proteins belonging to metabolic pathways involved in energy generation, both in glycolytic and oxidative phosphorylation pathways, suggesting antipsychotics-induced impairments in metabolism. Nevertheless, schizophrenic patients show impaired glucose metabolism and mitochondrial dysfunctions independent of therapy. Other antipsychotics-induced changes shared by different studies implicate cytoskeletal and synaptic function proteins. The mechanism can be related to the reorganization of dendritic spines resulting from neural plasticity events induced by treatments affecting neurotransmitter circuitry. However, metabolic and plasticity pathways activated by antipsychotics can also play an authentic role in the etiopathological basis of schizophrenia.
Collapse
Affiliation(s)
- Lucia Carboni
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Enrico Domenici
- Roche Pharmaceutical Research and Early Development, Neuroscience, Ophthalmology and Rare Diseases Discovery & Translational Medicine Area, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| |
Collapse
|
293
|
Habela CW, Song H, Ming GL. Modeling synaptogenesis in schizophrenia and autism using human iPSC derived neurons. Mol Cell Neurosci 2015; 73:52-62. [PMID: 26655799 DOI: 10.1016/j.mcn.2015.12.002] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 11/17/2015] [Accepted: 12/01/2015] [Indexed: 02/08/2023] Open
Abstract
Schizophrenia (SCZ) and autism spectrum disorder (ASD) are genetically and phenotypically complex disorders of neural development. Human genetic studies, as well as studies examining structural changes at the cellular level, have converged on glutamatergic synapse formation, function, and maintenance as common pathophysiologic substrates involved in both disorders. Synapses as basic functional units of the brain are continuously modified by experience throughout life, therefore they are particularly attractive candidates for targeted therapy. Until recently we lacked a system to evaluate dynamic changes that lead to synaptic abnormalities. With the development of techniques to generate induced pluripotent stem cells (iPSCs) from patients, we are now able to study neuronal and synaptic development in cells from individual patients in the context of genetic changes conferring disease susceptibility. In this review, we discuss recent studies focusing on neural cells differentiated from SCZ and ASD patient iPSCs. These studies support a central role for glutamatergic synapse formation and function in both disorders and demonstrate that iPSC derived neurons offer a potential system for further evaluation of processes leading to synaptic dysregulation and for the design and screening of future therapies.
Collapse
Affiliation(s)
- Christa W Habela
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Hongjun Song
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; The Solomon Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Guo-Li Ming
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; The Solomon Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
294
|
Contribution of sex hormones to gender differences in schizophrenia: A review. Asian J Psychiatr 2015; 18:2-14. [PMID: 26321672 DOI: 10.1016/j.ajp.2015.07.016] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 07/14/2015] [Accepted: 07/25/2015] [Indexed: 12/22/2022]
Abstract
Female patients with schizophrenia tend to have a more benign course and better outcomes than males. One proposed explanation is the differential influence of male and female sex hormones, including estrogen, progesterone, testosterone, and dehydroepiandrosterone (DHEA) and its sulfate (DHEAS). Such benefit may be mediated by their effects on neurotransmitters and neuroprotection. Besides altered estrogen and DHEA/DHEAS levels in female patients, data is equivocal on hormonal differences between patients and controls. However, several reports note a mostly negative correlation between estrogen levels and symptom severity in both genders, and a positive correlation between estrogen levels and neurocognition but mainly in females. Adjunctive estrogen appears to improve symptoms in both genders. Progesterone levels have inconsistent links to symptom severity in both genders, and correlate positively with neurocognition but only in males. Estrogen-progesterone combination shows preliminary benefits as augmentation for both symptoms and neurocognition in females. Testosterone levels correlate inversely with negative symptoms in males and have inconsistent associations with neurocognition in both genders. Testosterone augmentation reduced negative symptoms in male patients in a pilot investigation, but has not been evaluated for neurocognition in either gender. DHEA/DHEAS have mixed results for their association with, and clinical utility for, symptoms and neurocognition in both genders. Overall, data on the impact of sex hormones on clinical course or as treatment for schizophrenia is limited, but estrogen has most evidence for positive influence and clinical benefit. The possibly greater tolerability and broader impact of these hormones versus existing medications support further exploration of their use.
Collapse
|
295
|
Zhang JP, Robinson DG, Gallego JA, John M, Yu J, Addington J, Tohen M, Kane JM, Malhotra AK, Lencz T. Association of a Schizophrenia Risk Variant at the DRD2 Locus With Antipsychotic Treatment Response in First-Episode Psychosis. Schizophr Bull 2015; 41:1248-55. [PMID: 26320194 PMCID: PMC4601717 DOI: 10.1093/schbul/sbv116] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Findings from the Psychiatric Genomics Consortium genome-wide association study (GWAS) showed that variation at the DRD2 locus is associated with schizophrenia risk. However, the functional significance of rs2514218, the top DRD2 single nucleotide polymorphism in the GWAS, is unknown. Dopamine D2 receptor binding is a common mechanism of action for all antipsychotic drugs, and DRD2 variants were related to antipsychotic response in previous studies. The present study examined whether rs2514218 genotype could predict antipsychotic response, including efficacy and adverse events, in a cohort of patients with first episode of psychosis treated with either risperidone or aripiprazole for 12 weeks. Subjects were genotyped using the Illumina Infinium HumanOmniExpressExome array platform. After standard quality control, data from 100 subjects (49 randomly assigned to treatment with aripiprazole and 51 assigned to risperidone) was available for analysis. Subjects were assessed for psychotic symptomatology and medication-related adverse events weekly for 4 weeks, then biweekly for 8 weeks. Linear mixed model analysis revealed that the homozygotes for the risk (C) allele at rs2514218 had significantly greater reduction in positive symptoms during 12 weeks of treatment compared to the T allele carriers. In the aripiprazole group, C/C homozygotes also reported more akathisia than the T allele carriers, while in the risperidone group, male T allele carriers demonstrated greater prolactin elevations compared to male C/C homozygotes. These findings suggest that the schizophrenia risk variant at the DRD2 locus (or another variant in close proximity) is associated with observable differences in response to treatments which reduce striatal dopamine signaling.
Collapse
Affiliation(s)
| | | | | | | | - Jin Yu
- Division of Psychiatry Research, Department of Psychiatry, The Zucker Hillside Hospital, North Shore-Long Island Jewish Health System, Glen Oaks, NY
| | - Jean Addington
- Department of Psychiatry, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Mauricio Tohen
- Department of Psychiatry and Behavioral Sciences, University of New Mexico Health Sciences Center, Albuquerque, NM
| | | | | | | |
Collapse
|
296
|
Abstract
Histone modifications and DNA methylation represent central dynamic and reversible processes that regulate gene expression and contribute to cellular phenotypes. These epigenetic marks have been shown to play fundamental roles in a diverse set of signaling and behavioral outcomes. Psychiatric disorders such as schizophrenia and depression are complex and heterogeneous diseases with multiple and independent factors that may contribute to their pathophysiology, making challenging to find a link between specific elements and the underlying mechanisms responsible for the disorder and its treatment. Growing evidences suggest that epigenetic modifications in certain brain regions and neural circuits represent a key mechanism through which environmental factors interact with individual's genetic constitution to affect risk of psychiatric conditions throughout life. This review focuses on recent advances that directly implicate epigenetic modifications in schizophrenia and antipsychotic drug action.
Collapse
Affiliation(s)
- Daisuke Ibi
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Javier González-Maeso
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Physiology and Biophysics, Virginia Commonwealth University Medical School, Richmond, VA 23298, USA.
| |
Collapse
|
297
|
Tang S, Xie M, Cao N, Ding S. Patient-Specific Induced Pluripotent Stem Cells for Disease Modeling and Phenotypic Drug Discovery. J Med Chem 2015; 59:2-15. [PMID: 26322868 DOI: 10.1021/acs.jmedchem.5b00789] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
In vitro cell models are invaluable tools for studying diseases and discovering drugs. Human induced pluripotent stem cells, particularly derived from patients, are an advantageous resource for generating ample supplies of cells to create unique platforms that model disease. This manuscript will review recent developments in modeling a variety of diseases (including their cellular phenotypes) with induced pluripotent stem cells derived from patients. It will also describe how researchers have exploited these models to validate drugs as potential therapeutics for these devastating diseases.
Collapse
Affiliation(s)
- Shibing Tang
- Gladstone Institutes , 1650 Owens Street, San Francisco, California 94158, United States
| | - Min Xie
- Gladstone Institutes , 1650 Owens Street, San Francisco, California 94158, United States
| | - Nan Cao
- Gladstone Institutes , 1650 Owens Street, San Francisco, California 94158, United States
| | - Sheng Ding
- Gladstone Institutes , 1650 Owens Street, San Francisco, California 94158, United States
| |
Collapse
|
298
|
Cao Y, Paudel S, Zhang X, Kim KM, Cheon SH. Synthesis and evaluation of arylpiperazine-reverse amides as biased dopamine D3 receptor ligands. Bioorg Med Chem 2015; 23:5264-72. [DOI: 10.1016/j.bmc.2015.07.072] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 07/30/2015] [Accepted: 07/30/2015] [Indexed: 11/28/2022]
|
299
|
Sinkus ML, Graw S, Freedman R, Ross RG, Lester HA, Leonard S. The human CHRNA7 and CHRFAM7A genes: A review of the genetics, regulation, and function. Neuropharmacology 2015; 96:274-88. [PMID: 25701707 PMCID: PMC4486515 DOI: 10.1016/j.neuropharm.2015.02.006] [Citation(s) in RCA: 134] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Revised: 02/04/2015] [Accepted: 02/06/2015] [Indexed: 01/16/2023]
Abstract
The human α7 neuronal nicotinic acetylcholine receptor gene (CHRNA7) is ubiquitously expressed in both the central nervous system and in the periphery. CHRNA7 is genetically linked to multiple disorders with cognitive deficits, including schizophrenia, bipolar disorder, ADHD, epilepsy, Alzheimer's disease, and Rett syndrome. The regulation of CHRNA7 is complex; more than a dozen mechanisms are known, one of which is a partial duplication of the parent gene. Exons 5-10 of CHRNA7 on chromosome 15 were duplicated and inserted 1.6 Mb upstream of CHRNA7, interrupting an earlier partial duplication of two other genes. The chimeric CHRFAM7A gene product, dupα7, assembles with α7 subunits, resulting in a dominant negative regulation of function. The duplication is human specific, occurring neither in primates nor in rodents. The duplicated α7 sequence in exons 5-10 of CHRFAM7A is almost identical to CHRNA7, and thus is not completely queried in high throughput genetic studies (GWAS). Further, pre-clinical animal models of the α7nAChR utilized in drug development research do not have CHRFAM7A (dupα7) and cannot fully model human drug responses. The wide expression of CHRNA7, its multiple functions and modes of regulation present challenges for study of this gene in disease. This article is part of the Special Issue entitled 'The Nicotinic Acetylcholine Receptor: From Molecular Biology to Cognition'.
Collapse
Affiliation(s)
- Melissa L Sinkus
- Department of Psychiatry, University of Colorado Denver, Aurora, CO 80045, USA.
| | - Sharon Graw
- Department of Psychiatry, University of Colorado Denver, Aurora, CO 80045, USA.
| | - Robert Freedman
- Department of Psychiatry, University of Colorado Denver, Aurora, CO 80045, USA; Veterans Affairs Medical Research Center, Denver, CO 80262, USA.
| | - Randal G Ross
- Department of Psychiatry, University of Colorado Denver, Aurora, CO 80045, USA.
| | - Henry A Lester
- Division of Biology, California Institute of Technology, Pasadena, CA 91125, USA.
| | - Sherry Leonard
- Department of Psychiatry, University of Colorado Denver, Aurora, CO 80045, USA; Veterans Affairs Medical Research Center, Denver, CO 80262, USA.
| |
Collapse
|
300
|
Javitt DC, Sweet RA. Auditory dysfunction in schizophrenia: integrating clinical and basic features. Nat Rev Neurosci 2015; 16:535-50. [PMID: 26289573 PMCID: PMC4692466 DOI: 10.1038/nrn4002] [Citation(s) in RCA: 281] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Schizophrenia is a complex neuropsychiatric disorder that is associated with persistent psychosocial disability in affected individuals. Although studies of schizophrenia have traditionally focused on deficits in higher-order processes such as working memory and executive function, there is an increasing realization that, in this disorder, deficits can be found throughout the cortex and are manifest even at the level of early sensory processing. These deficits are highly amenable to translational investigation and represent potential novel targets for clinical intervention. Deficits, moreover, have been linked to specific structural abnormalities in post-mortem auditory cortex tissue from individuals with schizophrenia, providing unique insights into underlying pathophysiological mechanisms.
Collapse
Affiliation(s)
- Daniel C Javitt
- Division of Experimental Therapeutics, Departments of Psychiatry and Neuroscience, Columbia University College of Physicians and Surgeons, 1051 Riverside Drive, Unit 21, New York, New York 10032, USA
- Program in Cognitive Neuroscience and Schizophrenia, Nathan S. Kline Institute, 140 Old Orangeburg Rd, Orangeburg, New York 10962, USA
| | - Robert A Sweet
- Departments of Psychiatry and Neurology, University of Pittsburgh, 3811 O'Hara Street, Pittsburgh, Pennsylvania 15213, USA
- VISN 4 Mental Illness Research, Education and Clinical Center (MIRECC), VA Pittsburgh Healthcare System, Research Office Building (151R), University Drive C, Pittsburgh, Pennsylvania 15240, USA
| |
Collapse
|