1
|
Lopes JJ, Carruthers SP, Meyer D, Dean B, Rossell SL. Glutamatergic neurotransmission in schizophrenia: A systematic review and quantitative synthesis of proton magnetic resonance spectroscopy studies across schizophrenia spectrum disorders. Aust N Z J Psychiatry 2024; 58:930-951. [PMID: 38812258 DOI: 10.1177/00048674241254216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
Abstract
OBJECTIVE Studies using proton magnetic resonance spectroscopy reveal substantial inconsistencies in the levels of brain glutamate, glutamine and glutamate + glutamine across schizophrenia spectrum disorders. This systematic review employs qualitative and quantitative methods to analyse the patterns and relationships between glutamatergic metabolites, schizophrenia spectrum disorders and brain regions. METHODS A literature search was conducted using various databases with keywords including glutamate, glutamine, schizophrenia, psychosis and proton magnetic resonance spectroscopy. Inclusion criteria were limited to case-control studies that reported glutamatergic metabolite levels in adult patients with a schizophrenia spectrum disorder diagnosis - i.e. first-episode psychosis, schizophrenia, treatment-resistant schizophrenia and/or ultra-treatment-resistant schizophrenia - using proton magnetic resonance spectroscopy at 3 T or above. Pooled study data were synthesized and analysed. RESULTS A total of 92 studies met the inclusion criteria, including 2721 healthy controls and 2822 schizophrenia spectrum disorder participants. Glu levels were higher in the basal ganglia, frontal cortex and medial prefrontal of first-episode psychosis participants, contrasting overall lower levels in schizophrenia participants. For Gln, strong differences in metabolite levels were evident in the basal ganglia, dorsolateral prefrontal cortex and frontal cortex, with first-episode psychosis showing significantly higher levels in the basal ganglia. In glutamate + glutamine, higher metabolite levels were found across schizophrenia spectrum disorder groups, particularly in the basal ganglia and dorsolateral prefrontal cortex of treatment-resistant schizophrenia participants. Significant relationships were found between metabolite levels and medication status, clinical measures and methodological variables. CONCLUSION The review highlights abnormal glutamatergic metabolite levels throughout schizophrenia spectrum disorders and in specific brain regions. The review underscores the importance of standardized future research assessing glutamatergic metabolites using proton magnetic resonance spectroscopy due to considerable literature heterogeneity.
Collapse
Affiliation(s)
- Jamie J Lopes
- Centre for Mental Health, Swinburne University of Technology, Melbourne, VIC, Australia
| | - Sean P Carruthers
- Centre for Mental Health, Swinburne University of Technology, Melbourne, VIC, Australia
| | - Denny Meyer
- Centre for Mental Health, Swinburne University of Technology, Melbourne, VIC, Australia
| | - Brian Dean
- Centre for Mental Health, Swinburne University of Technology, Melbourne, VIC, Australia
- Molecular Psychiatry Laboratory, The Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia
| | - Susan L Rossell
- Centre for Mental Health, Swinburne University of Technology, Melbourne, VIC, Australia
- Department of Psychiatry, St Vincent's Hospital, Melbourne, VIC, Australia
| |
Collapse
|
2
|
Fan L, Liang L, Wang Y, Ma X, Yuan L, Ouyang L, He Y, Li Z, Li C, Chen X, Palaniyappan L. Glutamatergic basis of antipsychotic response in first-episode psychosis: a dual voxel study of the anterior cingulate cortex. Neuropsychopharmacology 2024; 49:845-853. [PMID: 37752221 PMCID: PMC10948866 DOI: 10.1038/s41386-023-01741-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/31/2023] [Accepted: 09/11/2023] [Indexed: 09/28/2023]
Abstract
A subgroup of patients with schizophrenia is believed to have aberrant excess of glutamate in the frontal cortex; this subgroup is thought to show poor response to first-line antipsychotic treatments that focus on dopamine blockade. If we can identify this subgroup early in the course of illness, we can reduce the repeated use of first-line antipsychotics and potentially stratify first-episode patients to intervene early with second-line treatments such as clozapine. The use of proton magnetic resonance spectroscopy (1H-MRS) to measure glutamate and Glx (glutamate plus glutamine) may provide a means for such a stratification. We must first establish if there is robust evidence linking elevations in anterior cingulate cortex (ACC) glutamate metabolites to poor response, and determine if the use of antipsychotics worsens the glutamatergic excess in eventual nonresponders. In this study, we estimated glutamate levels at baseline in 42 drug-naive patients with schizophrenia. We then treated them all with risperidone at a standard dose range of 2-6 mg/day and followed them up for 3 months to categorize their response status. We expected to see baseline "hyperglutamatergia" in nonresponders, and expected this to worsen over time at the follow-up. In line with our predictions, nonresponders had higher glutamate than responders, but patients as a group did not differ in glutamate and Glx from the healthy control (HC) group before treatment-onset (F1,79 = 3.20, p = 0.046, partial η2 = 0.075). Glutamatergic metabolites did not change significantly over time in both nonresponders and responders over the 3 months of antipsychotic exposure (F1,31 = 1.26, p = 0.270, partial η2 = 0.039). We conclude that the use of antipsychotics without prior knowledge of later response delays symptom relief in a subgroup of first-episode patients, but does not worsen the glutamatergic excess seen at the baseline. Given the current practice of nonstratified use of antipsychotics, longer-time follow-up MRS studies are required to see if improvement in symptoms accompanies a dynamic shift in glutamate profile.
Collapse
Affiliation(s)
- Lejia Fan
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, China
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Liangbing Liang
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Yujue Wang
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Xiaoqian Ma
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Liu Yuan
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Lijun Ouyang
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Ying He
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Zongchang Li
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Chunwang Li
- Department of Radiology, Hunan Children's Hospital, Changsha, China
| | - Xiaogang Chen
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, China.
| | - Lena Palaniyappan
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montreal, QC, Canada.
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada.
| |
Collapse
|
3
|
Singh U, Das B, Khanra S, Roy C. Resting state and activated brain glutamate-glutamine, brain lactate, cognition, and psychopathology among males with schizophrenia: A 3 Tesla proton magnetic resonance spectroscopic (1H-MRS) study. Indian J Psychiatry 2024; 66:82-89. [PMID: 38419937 PMCID: PMC10898519 DOI: 10.4103/indianjpsychiatry.indianjpsychiatry_621_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 11/09/2023] [Accepted: 12/25/2023] [Indexed: 03/02/2024] Open
Abstract
Background Research on glutamate (Glu) in schizophrenia has so far been inconclusive. Based on preclinical studies on Glu lactate interaction, researchers have now focused on brain lactate level as a sign of major pathology, including cognitive dysfunctions in the brain. Our study aimed to examine changes at resting and activated states in brain lactate and Glu-glutamine (Glx) at the anterior cingulate cortex (ACC) in schizophrenia. Methods A hospital-based prospective study was conducted with twenty-two male cases of schizophrenia and matched healthy controls (HCs). Positive and Negative Syndrome Scale (PANSS), Montreal Cognitive Assessment (MoCA), and Stroop tasks were administered among patients. Brain lactate and Glx at ACC were measured at resting state and during the Stroop test with proton magnetic resonance spectroscopy (1H-MRS) both at baseline and at remission and once among HC. Result Though MoCA scores improved significantly (P < 0.001) at remission from baseline among cases, repeated-measures analysis of variance (RM-ANOVA) did not find a significant time effect for Glx (P = 0.82) and lactate (P = 0.30) among cases from baseline to remission. Glx and lactate changed differently from baseline to remission. Conclusion Our study did not find significant differences in Glx and lactate between schizophrenia patients and HC. No significant time effect on Glx and lactate was observed from baseline to remission among schizophrenia cases. Different changes observed in Glx and lactate from baseline to remission require replication in future studies with larger sample size, longer follow-up period, and multivoxel MR assessment.
Collapse
Affiliation(s)
- Ujjwal Singh
- Department of Psychiatry, Central Institute of Psychiatry, Ranchi, Jharkhand, India
| | - Basudeb Das
- Department of Psychiatry, Central Institute of Psychiatry, Ranchi, Jharkhand, India
| | - Sourav Khanra
- Department of Psychiatry, Central Institute of Psychiatry, Ranchi, Jharkhand, India
| | - Chandramouli Roy
- Department of Psychiatry, Central Institute of Psychiatry, Ranchi, Jharkhand, India
| |
Collapse
|
4
|
Stein A, Zhu C, Du F, Öngür D. Magnetic Resonance Spectroscopy Studies of Brain Energy Metabolism in Schizophrenia: Progression from Prodrome to Chronic Psychosis. Curr Psychiatry Rep 2023; 25:659-669. [PMID: 37812338 DOI: 10.1007/s11920-023-01457-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/05/2023] [Indexed: 10/10/2023]
Abstract
PURPOSE OF REVIEW Schizophrenia (SZ) is a debilitating mental illness; existing treatments are partially effective and associated with significant side effect burden, largely due to our limited understanding of disease mechanisms and the trajectory of disease progression. Accumulating evidence suggests that metabolic changes associated with glucose metabolism, mitochondrial dysfunction, and redox imbalance play an important role in the pathophysiology of schizophrenia. However, the molecular mechanisms associated with these abnormalities in the brains of schizophrenia patients and the ways in which they change over time remain unclear. This paper aims to review the current literature on molecular mechanisms and in vivo magnetic resonance spectroscopy (MRS) studies of impaired energy metabolism in patients at clinical high risk for psychosis, with first-episode SZ, and with chronic SZ. Our review covers research related to high-energy phosphate metabolism, lactate, intracellular pH, redox ratio, and the antioxidant glutathione. RECENT FINDINGS Both first-episode and chronic SZ patients display a significant reduction in creatine kinase reaction activity and redox (NAD + /NADH) ratio in the prefrontal cortex. Chronic, but not first-episode, SZ patients also show a trend toward increased lactate levels and decreased pH value. These findings suggest a progressive shift from oxidative phosphorylation to glycolysis for energy production over the course of SZ, which is associated with redox imbalance and mitochondrial dysfunction. Accumulating evidence indicates that aberrant brain energy metabolism associated with mitochondrial dysfunction and redox imbalance plays a critical role in SZ and will be a promising target for future treatments.
Collapse
Affiliation(s)
- Abigail Stein
- Psychotic Disorders Division, McLean Hospital, Belmont, 02478, USA
- McLean Imaging Center, McLean Hospital, Belmont, 02478, USA
| | - Chenyanwen Zhu
- Psychotic Disorders Division, McLean Hospital, Belmont, 02478, USA
- McLean Imaging Center, McLean Hospital, Belmont, 02478, USA
| | - Fei Du
- Psychotic Disorders Division, McLean Hospital, Belmont, 02478, USA.
- McLean Imaging Center, McLean Hospital, Belmont, 02478, USA.
- Harvard Medical School, Boston, MA, 02115, USA.
| | - Dost Öngür
- Psychotic Disorders Division, McLean Hospital, Belmont, 02478, USA.
- Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
5
|
van Boxel R, Gangadin SS, Janssen H, van der Steur S, van der Vinne LJC, Dortants L, Pelgrim TAD, Draisma LWR, Tuura R, van der Meer P, Batalla A, Bossong MG. The impact of cannabidiol treatment on resting state functional connectivity, prefrontal metabolite levels and reward processing in recent-onset patients with a psychotic disorder. J Psychiatr Res 2023; 163:93-101. [PMID: 37207437 DOI: 10.1016/j.jpsychires.2023.05.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 04/14/2023] [Accepted: 05/01/2023] [Indexed: 05/21/2023]
Abstract
The first clinical trials with cannabidiol (CBD) as treatment for psychotic disorders have shown its potential as an effective and well-tolerated antipsychotic agent. However, the neurobiological mechanisms underlying the antipsychotic profile of CBD are currently unclear. Here we investigated the impact of 28-day adjunctive CBD or placebo treatment (600 mg daily) on brain function and metabolism in 31 stable recent-onset psychosis patients (<5 years after diagnosis). Before and after treatment, patients underwent a Magnetic Resonance Imaging (MRI) session including resting state functional MRI, proton Magnetic Resonance Spectroscopy (1H-MRS) and functional MRI during reward processing. Symptomatology and cognitive functioning were also assessed. CBD treatment significantly changed functional connectivity in the default mode network (DMN; time × treatment interaction p = 0.037), with increased connectivity in the CBD (from 0.59 ± 0.39 to 0.80 ± 0.32) and reduced connectivity in the placebo group (from 0.77 ± 0.37 to 0.62 ± 0.33). Although there were no significant treatment effects on prefrontal metabolite concentrations, we showed that decreased positive symptom severity over time was associated with both diminishing glutamate (p = 0.029) and N-acetyl-aspartate (NAA; neuronal integrity marker) levels (p = 0.019) in the CBD, but not the placebo group. CBD treatment did not have an impact on brain activity patterns during reward anticipation and receipt or functional connectivity in executive and salience networks. Our results show that adjunctive CBD treatment of recent-onset psychosis patients induced changes in DMN functional connectivity, but not prefrontal metabolite concentrations or brain activity during reward processing. These findings suggest that DMN connectivity alteration may be involved in the therapeutic effects of CBD.
Collapse
Affiliation(s)
- Ruben van Boxel
- Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, the Netherlands
| | - Shiral S Gangadin
- Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, the Netherlands; Section of Neuropsychiatry, Department of Biomedical Sciences of Cells and Systems, University Medical Center Groningen, Groningen, the Netherlands
| | - Hella Janssen
- Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, the Netherlands
| | - Sanne van der Steur
- Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, the Netherlands
| | - Lucia J C van der Vinne
- Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, the Netherlands
| | - Lon Dortants
- Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, the Netherlands
| | - Teuntje A D Pelgrim
- Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, the Netherlands; Department of Psychiatry, Parnassia Psychiatric Institute, Amsterdam, the Netherlands
| | - Luc W R Draisma
- Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, the Netherlands
| | - Ruth Tuura
- Center of MR Research, University Children's Hospital Zurich, Zurich, Switzerland
| | - Pim van der Meer
- Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, the Netherlands
| | - Albert Batalla
- Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, the Netherlands
| | - Matthijs G Bossong
- Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, the Netherlands.
| |
Collapse
|
6
|
Jauhar S, McCutcheon RA, Veronese M, Borgan F, Nour M, Rogdaki M, Pepper F, Stone JM, Egerton A, Vamvakas G, Turkheimer F, McGuire PK, Howes OD. The relationship between striatal dopamine and anterior cingulate glutamate in first episode psychosis changes with antipsychotic treatment. Transl Psychiatry 2023; 13:184. [PMID: 37253720 PMCID: PMC10229638 DOI: 10.1038/s41398-023-02479-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 05/15/2023] [Indexed: 06/01/2023] Open
Abstract
The neuromodulator dopamine and excitatory neurotransmitter glutamate have both been implicated in the pathogenesis of psychosis, and dopamine antagonists remain the predominant treatment for psychotic disorders. To date no study has measured the effect of antipsychotics on both of these indices together, in the same population of people with psychosis. Striatal dopamine synthesis capacity (Kicer) and anterior cingulate glutamate were measured using 18F-DOPA positron emission tomography and proton magnetic resonance spectroscopy respectively, before and after at least 5 weeks' naturalistic antipsychotic treatment in people with first episode psychosis (n = 18) and matched healthy controls (n = 20). The relationship between both measures at baseline and follow-up, and the change in this relationship was analyzed using a mixed linear model. Neither anterior cingulate glutamate concentrations (p = 0.75) nor striatal Kicer (p = 0.79) showed significant change following antipsychotic treatment. The change in relationship between whole striatal Kicer and anterior cingulate glutamate, however, was statistically significant (p = 0.017). This was reflected in a significant difference in relationship between both measures for patients and controls at baseline (t = 2.1, p = 0.04), that was not present at follow-up (t = 0.06, p = 0.96). Although we did not find any effect of antipsychotic treatment on absolute measures of dopamine synthesis capacity and anterior cingulate glutamate, the relationship between anterior cingluate glutamate and striatal dopamine synthesis capacity did change, suggesting that antipsychotic treatment affects the relationship between glutamate and dopamine.
Collapse
Affiliation(s)
- Sameer Jauhar
- Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College, London, UK.
| | - Robert A McCutcheon
- Department of Psychiatry, University of Oxford, Oxford, UK
- Oxford Health NHS Foundation Trust, Oxford, UK
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College, London, UK
| | - Mattia Veronese
- Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College, London, UK
| | - Faith Borgan
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College, London, UK
| | - Matthew Nour
- Max Planck UCL Centre for Computational Psychiatry and Ageing Research, Berlin, Germany
| | - Maria Rogdaki
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College, London, UK
| | - Fiona Pepper
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College, London, UK
| | - James M Stone
- Department of Neuroscience and Imaging, University of Sussex, Brighton and Hove, UK
| | - Alice Egerton
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College, London, UK
| | - George Vamvakas
- Department of Biostatistics, Institute of Psychiatry, Psychology and Neuroscience, King's College, London, UK
| | - Federico Turkheimer
- Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College, London, UK
| | | | - Oliver D Howes
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College, London, UK
- MRC London Institute of Medical Sciences, Imperial College, London, UK
- H Lundbeck A/s, St Albans, UK
| |
Collapse
|
7
|
Merritt K, McCutcheon RA, Aleman A, Ashley S, Beck K, Block W, Bloemen OJN, Borgan F, Boules C, Bustillo JR, Capizzano AA, Coughlin JM, David A, de la Fuente-Sandoval C, Demjaha A, Dempster K, Do KQ, Du F, Falkai P, Galińska-Skok B, Gallinat J, Gasparovic C, Ginestet CE, Goto N, Graff-Guerrero A, Ho BC, Howes O, Jauhar S, Jeon P, Kato T, Kaufmann CA, Kegeles LS, Keshavan MS, Kim SY, King B, Kunugi H, Lauriello J, León-Ortiz P, Liemburg E, Mcilwain ME, Modinos G, Mouchlianitis E, Nakamura J, Nenadic I, Öngür D, Ota M, Palaniyappan L, Pantelis C, Patel T, Plitman E, Posporelis S, Purdon SE, Reichenbach JR, Renshaw PF, Reyes-Madrigal F, Russell BR, Sawa A, Schaefer M, Shungu DC, Smesny S, Stanley JA, Stone J, Szulc A, Taylor R, Thakkar KN, Théberge J, Tibbo PG, van Amelsvoort T, Walecki J, Williamson PC, Wood SJ, Xin L, Yamasue H, McGuire P, Egerton A. Variability and magnitude of brain glutamate levels in schizophrenia: a meta and mega-analysis. Mol Psychiatry 2023; 28:2039-2048. [PMID: 36806762 PMCID: PMC10575771 DOI: 10.1038/s41380-023-01991-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 01/18/2023] [Accepted: 01/31/2023] [Indexed: 02/19/2023]
Abstract
Glutamatergic dysfunction is implicated in schizophrenia pathoaetiology, but this may vary in extent between patients. It is unclear whether inter-individual variability in glutamate is greater in schizophrenia than the general population. We conducted meta-analyses to assess (1) variability of glutamate measures in patients relative to controls (log coefficient of variation ratio: CVR); (2) standardised mean differences (SMD) using Hedges g; (3) modal distribution of individual-level glutamate data (Hartigan's unimodality dip test). MEDLINE and EMBASE databases were searched from inception to September 2022 for proton magnetic resonance spectroscopy (1H-MRS) studies reporting glutamate, glutamine or Glx in schizophrenia. 123 studies reporting on 8256 patients and 7532 controls were included. Compared with controls, patients demonstrated greater variability in glutamatergic metabolites in the medial frontal cortex (MFC, glutamate: CVR = 0.15, p < 0.001; glutamine: CVR = 0.15, p = 0.003; Glx: CVR = 0.11, p = 0.002), dorsolateral prefrontal cortex (glutamine: CVR = 0.14, p = 0.05; Glx: CVR = 0.25, p < 0.001) and thalamus (glutamate: CVR = 0.16, p = 0.008; Glx: CVR = 0.19, p = 0.008). Studies in younger, more symptomatic patients were associated with greater variability in the basal ganglia (BG glutamate with age: z = -0.03, p = 0.003, symptoms: z = 0.007, p = 0.02) and temporal lobe (glutamate with age: z = -0.03, p = 0.02), while studies with older, more symptomatic patients associated with greater variability in MFC (glutamate with age: z = 0.01, p = 0.02, glutamine with symptoms: z = 0.01, p = 0.02). For individual patient data, most studies showed a unimodal distribution of glutamatergic metabolites. Meta-analysis of mean differences found lower MFC glutamate (g = -0.15, p = 0.03), higher thalamic glutamine (g = 0.53, p < 0.001) and higher BG Glx in patients relative to controls (g = 0.28, p < 0.001). Proportion of males was negatively associated with MFC glutamate (z = -0.02, p < 0.001) and frontal white matter Glx (z = -0.03, p = 0.02) in patients relative to controls. Patient PANSS total score was positively associated with glutamate SMD in BG (z = 0.01, p = 0.01) and temporal lobe (z = 0.05, p = 0.008). Further research into the mechanisms underlying greater glutamatergic metabolite variability in schizophrenia and their clinical consequences may inform the identification of patient subgroups for future treatment strategies.
Collapse
Affiliation(s)
- Kate Merritt
- Division of Psychiatry, UCL, Institute of Mental Health, London, UK.
| | | | - André Aleman
- Center for Brain Disorder and Cognitive Science, Shenzhen University, Shenzhen, China
- University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Sarah Ashley
- Division of Psychiatry, UCL, Institute of Mental Health, London, UK
| | - Katherine Beck
- Psychosis Studies Department, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Wolfgang Block
- Department of Diagnostic and Interventional Radiology, University Hospital Bonn, Bonn, Germany
| | - Oswald J N Bloemen
- Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, the Netherlands
| | - Faith Borgan
- Psychosis Studies Department, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Christiana Boules
- Psychosis Studies Department, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Juan R Bustillo
- Department of Psychiatry and Behavioral Sciences, Center for Psychiatric Research, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Aristides A Capizzano
- Department of Radiology, Division of Neuroradiology, University of Michigan, 1500 E Medical Center Dr, Ann Arbor, MI, 48109, USA
| | - Jennifer M Coughlin
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Anthony David
- Division of Psychiatry, UCL, Institute of Mental Health, London, UK
| | - Camilo de la Fuente-Sandoval
- Laboratory of Experimental Psychiatry, Instituto Nacional de Neurología y Neurocirugía, Mexico City, Mexico
- Neuropsychiatry Department, Instituto Nacional de Neurología y Neurocirugía, Mexico City, Mexico
| | - Arsime Demjaha
- Psychosis Studies Department, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Kara Dempster
- Department of Psychiatry, Dalhousie University, Halifax, NS, Canada
| | - Kim Q Do
- Center for Psychiatric Neuroscience (CNP), Department of Psychiatry, Lausanne University Hospital-CHUV, Prilly-Lausanne, Switzerland
| | - Fei Du
- Psychotic Disorders Division, McLean Hospital, Harvard Medical School, Belmont, MA, USA
| | - Peter Falkai
- Department of Psychiatry, University Hospital, LMU Munich, Nussbaumstrasse 7, 80336, Munich, Germany
| | - Beata Galińska-Skok
- Department of Psychiatry, Medical University of Bialystok, Bialystok, Poland
| | - Jürgen Gallinat
- Department of Psychiatry and Psychotherapy, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | | | - Cedric E Ginestet
- Department of Biostatistics and Health Informatics (S2.06), Institute of Psychiatry, Psychology and Neuroscience King's College London, London, UK
| | - Naoki Goto
- Department of Psychiatry, Kokura Gamo Hospital, Kitakyushu, Fukuoka, 8020978, Japan
| | - Ariel Graff-Guerrero
- Multimodal Neuroimaging Schizophrenia Group, Research Imaging Centre, Geriatric Mental Health Program at Centre for Addiction and Mental Health, and Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Beng-Choon Ho
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Oliver Howes
- Psychosis Studies Department, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Sameer Jauhar
- Psychosis Studies Department, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Peter Jeon
- Department of Medical Biophysics, University of Western Ontario, London, ON, Canada
| | - Tadafumi Kato
- Department of Psychiatry and Behavioral Science, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Charles A Kaufmann
- Department of Psychiatry, Columbia University, New York State Psychiatric Institute (NYSPI), New York, NY, USA
| | - Lawrence S Kegeles
- Columbia University, Department of Psychiatry, New York State Psychiatric Institute (NYSPI), New York, NY, USA
| | | | | | - Bridget King
- Psychosis Studies Department, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Hiroshi Kunugi
- National Center of Neurology and Psychiatry, Kodaira, Tokyo, 187-0031, Japan
| | - J Lauriello
- Jefferson Health-Sidney Kimmel Medical College, Philadelphia, PA, USA
| | - Pablo León-Ortiz
- Laboratory of Experimental Psychiatry, Instituto Nacional de Neurología y Neurocirugía, Mexico City, Mexico
- Neuropsychiatry Department, Instituto Nacional de Neurología y Neurocirugía, Mexico City, Mexico
| | - Edith Liemburg
- Rob Giel Research Center, Department of Psychiatry, University Medical Center Groningen, Groningen, the Netherlands
| | - Meghan E Mcilwain
- School of Pharmacy, University of Auckland, 85 Park Road, Grafton, Auckland, 1023, New Zealand
| | - Gemma Modinos
- Psychosis Studies Department, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Department of Neuroimaging, Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology & Neuroscience, De Crespigny Park, London, SE5 8AF, UK
| | - Elias Mouchlianitis
- Psychosis Studies Department, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Jun Nakamura
- Department of Psychiatry, University of Occupational and Environmental Health, Kitakyushu, Fukuoka, Japan
| | - Igor Nenadic
- Department of Psychiatry and Psychotherapy, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Dost Öngür
- Psychotic Disorders Division, McLean Hospital, Harvard Medical School, Belmont, MA, USA
| | - Miho Ota
- National Center of Neurology and Psychiatry, Kodaira, Tokyo, 187-0031, Japan
| | - Lena Palaniyappan
- Department of Medical Biophysics, University of Western Ontario, London, ON, Canada
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Christos Pantelis
- Melbourne Neuropsychiatry Centre, The University of Melbourne and Melbourne Health, Carlton, VIC, Australia
- The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
| | - Tulsi Patel
- Division of Psychiatry, UCL, Institute of Mental Health, London, UK
| | - Eric Plitman
- Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Sotirios Posporelis
- Psychosis Studies Department, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- South London and Maudsley, Bethlem Royal Hospital, Monks Orchard Road, Beckenham, BR3 3BX, UK
| | - Scot E Purdon
- Neuropsychology Department, Alberta Hospital Edmonton, Edmonton, AB, Canada
- Department of Psychiatry, University of Alberta, Edmonton, AB, Canada
| | - Jürgen R Reichenbach
- Medical Physics Group, Institute for Diagnostic and Interventional Radiology (IDIR), Jena University Hospital, Jena, Germany
| | - Perry F Renshaw
- Department of Psychiatry, University of Utah, Salt Lake City, UT, USA
| | - Francisco Reyes-Madrigal
- Laboratory of Experimental Psychiatry, Instituto Nacional de Neurología y Neurocirugía, Mexico City, Mexico
| | - Bruce R Russell
- School of Pharmacy, University of Otago, Dunedin, New Zealand
| | - Akira Sawa
- Departments of Psychiatry, Neuroscience, Mental Health, Biomedical Engineering, and Genetic Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Martin Schaefer
- Department of Psychiatry, Psychotherapy, Psychosomatics and Addiction Medicine, Kliniken Essen-Mitte, Essen, Germany
- Department of Psychiatry and Psychotherapy, Charité-Universitätsmedizin Berlin, Campus Charité Mitte, Berlin, Germany
| | - Dikoma C Shungu
- Department of Radiology, Weill Cornell Medical College, New York City, NY, USA
| | - Stefan Smesny
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Jena, Germany
| | - Jeffrey A Stanley
- Brain Imaging Research Division, Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA
| | - James Stone
- Department of Neuroimaging, Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology & Neuroscience, De Crespigny Park, London, SE5 8AF, UK
- Brighton and Sussex Medical School, University of Sussex, Brighton, UK
| | - Agata Szulc
- Department of Psychiatry, Medical University of Warsaw, Warsaw, Poland
| | - Reggie Taylor
- Department of Medical Biophysics, University of Western Ontario, London, ON, Canada
- Lawson Health Research Institute, London, ON, Canada
| | - Katharine N Thakkar
- Department of Psychology, Michigan State University, East Lansing, MI, USA
- Division of Psychiatry and Behavioral Medicine, Michigan State University, East Lansing, MI, USA
| | - Jean Théberge
- Department of Medical Biophysics, University of Western Ontario, London, ON, Canada
- Lawson Health Research Institute, London, ON, Canada
- Department of Psychiatry, Western University, London, ON, Canada
| | - Philip G Tibbo
- Department of Psychiatry, Dalhousie University, Halifax, NS, Canada
| | - Thérèse van Amelsvoort
- Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, the Netherlands
| | | | - Peter C Williamson
- Lawson Health Research Institute, London, ON, Canada
- Department of Psychiatry, Western University, London, ON, Canada
| | - Stephen J Wood
- Orygen, Melbourne, VIC, Australia
- Institute for Mental Health, University of Birmingham, Edgbaston, UK
- Centre for Youth Mental Health, University of Melbourne, Melbourne, VIC, Australia
| | - Lijing Xin
- Animal Imaging and Technology Core (AIT), Center for Biomedical Imaging (CIBM), Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Hidenori Yamasue
- Department of Psychiatry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Philip McGuire
- Psychosis Studies Department, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Alice Egerton
- Psychosis Studies Department, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| |
Collapse
|
8
|
Wang M, Barker PB, Cascella NG, Coughlin JM, Nestadt G, Nucifora FC, Sedlak TW, Kelly A, Younes L, Geman D, Palaniyappan L, Sawa A, Yang K. Longitudinal changes in brain metabolites in healthy controls and patients with first episode psychosis: a 7-Tesla MRS study. Mol Psychiatry 2023; 28:2018-2029. [PMID: 36732587 PMCID: PMC10394114 DOI: 10.1038/s41380-023-01969-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 01/13/2023] [Accepted: 01/17/2023] [Indexed: 02/04/2023]
Abstract
Seven Tesla magnetic resonance spectroscopy (7T MRS) offers a precise measurement of metabolic levels in the human brain via a non-invasive approach. Studying longitudinal changes in brain metabolites could help evaluate the characteristics of disease over time. This approach may also shed light on how the age of study participants and duration of illness may influence these metabolites. This study used 7T MRS to investigate longitudinal patterns of brain metabolites in young adulthood in both healthy controls and patients. A four-year longitudinal cohort with 38 patients with first episode psychosis (onset within 2 years) and 48 healthy controls was used to examine 10 brain metabolites in 5 brain regions associated with the pathophysiology of psychosis in a comprehensive manner. Both patients and controls were found to have significant longitudinal reductions in glutamate in the anterior cingulate cortex (ACC). Only patients were found to have a significant decrease over time in γ-aminobutyric acid, N-acetyl aspartate, myo-inositol, total choline, and total creatine in the ACC. Together we highlight the ACC with dynamic changes in several metabolites in early-stage psychosis, in contrast to the other 4 brain regions that also are known to play roles in psychosis. Meanwhile, glutathione was uniquely found to have a near zero annual percentage change in both patients and controls in all 5 brain regions during a four-year follow-up in young adulthood. Given that a reduction of the glutathione in the ACC has been reported as a feature of treatment-refractory psychosis, this observation further supports the potential of glutathione as a biomarker for this subset of patients with psychosis.
Collapse
Affiliation(s)
- Min Wang
- Russell H Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Peter B Barker
- Russell H Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA.
| | - Nicola G Cascella
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jennifer M Coughlin
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Gerald Nestadt
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Frederick C Nucifora
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Thomas W Sedlak
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alexandra Kelly
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Laurent Younes
- Department of Applied Mathematics and Statistics, Johns Hopkins University, Baltimore, MD, USA
| | - Donald Geman
- Department of Applied Mathematics and Statistics, Johns Hopkins University, Baltimore, MD, USA
| | - Lena Palaniyappan
- Robarts Research Institution, University of Western Ontario, London, ON, Canada
- Department of Psychiatry, University of Western Ontario, London, ON, Canada
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Akira Sawa
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA.
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Mental Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA.
| | - Kun Yang
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
9
|
Sreeraj VS, Shivakumar V, Bhalerao GV, Kalmady SV, Narayanaswamy JC, Venkatasubramanian G. Resting-state functional connectivity correlates of antipsychotic treatment in unmedicated schizophrenia. Asian J Psychiatr 2023; 82:103459. [PMID: 36682158 DOI: 10.1016/j.ajp.2023.103459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 01/03/2023] [Accepted: 01/07/2023] [Indexed: 01/09/2023]
Abstract
BACKGROUND Antipsychotics may modulate the resting state functional connectivity(rsFC) to improve clinical symptoms in schizophrenia(Sz). Existing literature has potential confounders like past medication effects and evaluating preselected regions/networks. We aimed to evaluate connectivity pattern changes with antipsychotics in unmedicated Sz using Multivariate pattern analysis(MVPA), a data-driven technique for whole-brain connectome analysis. METHODS Forty-seven unmedicated patients with Sz(DSM-IV-TR) underwent clinical evaluation and neuroimaging at baseline and after 3-months of antipsychotic treatment. Resting-state functional MRI was analysed using group-MVPA to derive 5-components. The brain region with significant connectivity pattern changes with antipsychotics was identified, and post-hoc seed-to-voxel analysis was performed to identify connectivity changes and their association with symptom changes. RESULTS Connectome-MVPA analysis revealed the connectivity pattern of a cluster localised to left anterior cingulate and paracingulate gyri (ACC/PCG) (peak coordinates:x = -04,y = +30,z = +26;k = 12;cluster-pFWE=0.002) to differ significantly after antipsychotics. Specifically, its connections with clusters of precuneus/posterior cingulate cortex(PCC) and left inferior temporal gyrus(ITG) correlated with improvement in positive and negative symptoms scores, respectively. CONCLUSION ACC/PCG, a hub of the default mode network, seems to mediate the antipsychotic effects in unmedicated Sz. Evaluating causality models with data from randomised controlled design using the MVPA approach would further enhance our understanding of therapeutic connectomics in Sz.
Collapse
Affiliation(s)
- Vanteemar S Sreeraj
- InSTAR Clinic and Translational Psychiatry Lab, Department of Psychiatry, National Institute of Mental Health and Neurosciences, Bengaluru, India.
| | - Venkataram Shivakumar
- InSTAR Clinic and Translational Psychiatry Lab, Department of Psychiatry, National Institute of Mental Health and Neurosciences, Bengaluru, India; Department of Integrative Medicine, National Institute of Mental Health and Neurosciences, Bengaluru, India
| | | | - Sunil V Kalmady
- Alberta Machine Intelligence Institute, Department of Computing Science, University of Alberta, Edmonton, Alberta, Canada; Canadian VIGOUR Centre, University of Alberta, Edmonton, Alberta, Canada
| | | | - Ganesan Venkatasubramanian
- InSTAR Clinic and Translational Psychiatry Lab, Department of Psychiatry, National Institute of Mental Health and Neurosciences, Bengaluru, India
| |
Collapse
|
10
|
Abstract
People with psychotic disorders can show marked interindividual variations in the onset of illness, responses to treatment and relapse, but they receive broadly similar clinical care. Precision psychiatry is an approach that aims to stratify people with a given disorder according to different clinical outcomes and tailor treatment to their individual needs. At present, interindividual differences in outcomes of psychotic disorders are difficult to predict on the basis of clinical assessment alone. Therefore, current research in psychosis seeks to build models that predict outcomes by integrating clinical information with a range of biological measures. Here, we review recent progress in the application of precision psychiatry to psychotic disorders and consider the challenges associated with implementing this approach in clinical practice.
Collapse
|
11
|
Egerton A, Griffiths K, Casetta C, Deakin B, Drake R, Howes OD, Kassoumeri L, Khan S, Lankshear S, Lees J, Lewis S, Mikulskaya E, Millgate E, Oloyede E, Pollard R, Rich N, Segev A, Sendt KV, MacCabe JH. Anterior cingulate glutamate metabolites as a predictor of antipsychotic response in first episode psychosis: data from the STRATA collaboration. Neuropsychopharmacology 2023; 48:567-575. [PMID: 36456813 PMCID: PMC9852590 DOI: 10.1038/s41386-022-01508-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 11/07/2022] [Accepted: 11/09/2022] [Indexed: 12/03/2022]
Abstract
Elevated brain glutamate has been implicated in non-response to antipsychotic medication in schizophrenia. Biomarkers that can accurately predict antipsychotic non-response from the first episode of psychosis (FEP) could allow stratification of patients; for example, patients predicted not to respond to standard antipsychotics could be fast-tracked to clozapine. Using proton magnetic resonance spectroscopy (1H-MRS), we examined the ability of glutamate and Glx (glutamate plus glutamine) in the anterior cingulate cortex (ACC) and caudate to predict response to antipsychotic treatment. A total of 89 minimally medicated patients with FEP not meeting symptomatic criteria for remission were recruited across two study sites. 1H-MRS and clinical data were acquired at baseline, 2 and 6 weeks. Response was defined as >20% reduction in Positive and Negative Syndrome Scale (PANSS) Total score from baseline to 6 weeks. In the ACC, baseline glutamate and Glx were higher in Non-Responders and significantly predicted response (P < 0.02; n = 42). Overall accuracy was greatest for ACC Glx (69%) and increased to 75% when symptom severity at baseline was included in the model. Glutamate metabolites in the caudate were not associated with response, and there was no significant change in glutamate metabolites over time in either region. These results add to the evidence linking elevations in ACC glutamate metabolites to a poor antipsychotic response. They indicate that glutamate may have utility in predicting response during early treatment of first episode psychosis. Improvements in accuracy may be made by combining glutamate measures with other response biomarkers.
Collapse
Affiliation(s)
- Alice Egerton
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK.
- NIHR Biomedical Research Centre at South London and Maudsley NHS Foundation Trust, London, UK.
| | - Kira Griffiths
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Cecila Casetta
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Bill Deakin
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Greater Manchester Mental Health NHS Foundation Trust Bury New Road, Prestwich, Manchester, M25 3BL, UK
| | - Richard Drake
- Greater Manchester Mental Health NHS Foundation Trust Bury New Road, Prestwich, Manchester, M25 3BL, UK
- Division of Psychology and Mental Health, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Oliver D Howes
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
- NIHR Biomedical Research Centre at South London and Maudsley NHS Foundation Trust, London, UK
| | - Laura Kassoumeri
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Sobia Khan
- Greater Manchester Mental Health NHS Foundation Trust Bury New Road, Prestwich, Manchester, M25 3BL, UK
- Division of Psychology and Mental Health, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Steve Lankshear
- Greater Manchester Mental Health NHS Foundation Trust Bury New Road, Prestwich, Manchester, M25 3BL, UK
- Division of Psychology and Mental Health, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Jane Lees
- Division of Psychology and Mental Health, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Shon Lewis
- Greater Manchester Mental Health NHS Foundation Trust Bury New Road, Prestwich, Manchester, M25 3BL, UK
- Division of Psychology and Mental Health, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Elena Mikulskaya
- Greater Manchester Mental Health NHS Foundation Trust Bury New Road, Prestwich, Manchester, M25 3BL, UK
- Division of Psychology and Mental Health, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Edward Millgate
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Ebenezer Oloyede
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Rebecca Pollard
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Nathalie Rich
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Aviv Segev
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Kyra-Verena Sendt
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - James H MacCabe
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
- NIHR Biomedical Research Centre at South London and Maudsley NHS Foundation Trust, London, UK
| |
Collapse
|
12
|
Zahid U, Onwordi EC, Hedges EP, Wall MB, Modinos G, Murray RM, Egerton A. Neurofunctional correlates of glutamate and GABA imbalance in psychosis: A systematic review. Neurosci Biobehav Rev 2023; 144:105010. [PMID: 36549375 DOI: 10.1016/j.neubiorev.2022.105010] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 12/01/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022]
Abstract
Glutamatergic and GABAergic dysfunction are implicated in the pathophysiology of schizophrenia. Previous work has shown relationships between glutamate, GABA, and brain activity in healthy volunteers. We conducted a systematic review to evaluate whether these relationships are disrupted in psychosis. Primary outcomes were the relationship between metabolite levels and fMRI BOLD response in psychosis relative to healthy volunteers. 17 case-control studies met inclusion criteria (594 patients and 538 healthy volunteers). Replicated findings included that in psychosis, positive associations between ACC glutamate levels and brain activity are reduced during resting state conditions and increased during cognitive control tasks, and negative relationships between GABA and local activation in the ACC are reduced. There was evidence that antipsychotic medication may alter the relationship between glutamate levels and brain activity. Emerging literature is providing insights into disrupted relationships between neurometabolites and brain activity in psychosis. Future studies determining a link to clinical variables may develop this approach for biomarker applications, including development or targeting novel therapeutics.
Collapse
Affiliation(s)
- Uzma Zahid
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK; Department of Psychiatry, University of Oxford, UK.
| | - Ellis C Onwordi
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK; MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital Campus, London, UK; Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, UK; South London and Maudsley NHS Foundation Trust, Camberwell, London, UK
| | - Emily P Hedges
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK
| | - Matthew B Wall
- Invicro London, Hammersmith Hospital, UK; Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, UK; Clinical Psychopharmacology Unit, University College London, UK
| | - Gemma Modinos
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK
| | - Robin M Murray
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK
| | - Alice Egerton
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, UK
| |
Collapse
|
13
|
Zahid U, McCutcheon RA, Borgan F, Jauhar S, Pepper F, Nour MM, Rogdaki M, Osugo M, Murray GK, Hathway P, Murray RM, Egerton A, Howes OD. The effect of antipsychotics on glutamate levels in the anterior cingulate cortex and clinical response: A 1H-MRS study in first-episode psychosis patients. Front Psychiatry 2022; 13:967941. [PMID: 36032237 PMCID: PMC9403834 DOI: 10.3389/fpsyt.2022.967941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 07/19/2022] [Indexed: 11/14/2022] Open
Abstract
Introduction Glutamatergic dysfunction is implicated in the pathophysiology of schizophrenia. It is unclear whether glutamatergic dysfunction predicts response to treatment or if antipsychotic treatment influences glutamate levels. We investigated the effect of antipsychotic treatment on glutamatergic levels in the anterior cingulate cortex (ACC), and whether there is a relationship between baseline glutamatergic levels and clinical response after antipsychotic treatment in people with first episode psychosis (FEP). Materials and methods The sample comprised 25 FEP patients; 22 completed magnetic resonance spectroscopy scans at both timepoints. Symptoms were assessed using the Positive and Negative Syndrome Scale (PANSS). Results There was no significant change in glutamate [baseline 13.23 ± 2.33; follow-up 13.89 ± 1.74; t(21) = -1.158, p = 0.260], or Glx levels [baseline 19.64 ± 3.26; follow-up 19.66 ± 2.65; t(21) = -0.034, p = 0.973]. There was no significant association between glutamate or Glx levels at baseline and the change in PANSS positive (Glu r = 0.061, p = 0.777, Glx r = -0.152, p = 0.477), negative (Glu r = 0.144, p = 0.502, Glx r = 0.052, p = 0.811), general (Glu r = 0.110, p = 0.607, Glx r = -0.212, p = 0.320), or total scores (Glu r = 0.078, p = 0.719 Glx r = -0.155, p = 0.470). Conclusion These findings indicate that treatment response is unlikely to be associated with baseline glutamatergic metabolites prior to antipsychotic treatment, and there is no major effect of antipsychotic treatment on glutamatergic metabolites in the ACC.
Collapse
Affiliation(s)
- Uzma Zahid
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Robert A. McCutcheon
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Faith Borgan
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Sameer Jauhar
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Fiona Pepper
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
- Department of Clinical and Movement Neurosciences, Queen Square Institute of Neurology, University College London Centre, London, United Kingdom
| | - Matthew M. Nour
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
- Max Planck University College London Centre for Computational Psychiatry and Ageing Research, London, United Kingdom
- Wellcome Trust Centre for Neuroimaging, University College London, London, United Kingdom
| | - Maria Rogdaki
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
- Department of Child and Adolescent Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Martin Osugo
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Graham K. Murray
- Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
| | - Pamela Hathway
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Robin M. Murray
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Alice Egerton
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Oliver D. Howes
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom
- H. Lundbeck UK, Valby, Denmark
| |
Collapse
|
14
|
di Hou M, Santoro V, Biondi A, Shergill SS, Premoli I. A systematic review of TMS and neurophysiological biometrics in patients with schizophrenia. J Psychiatry Neurosci 2021; 46:E675-E701. [PMID: 34933940 PMCID: PMC8695525 DOI: 10.1503/jpn.210006] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 08/06/2021] [Accepted: 09/06/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Transcranial magnetic stimulation can be combined with electromyography (TMS-EMG) and electroencephalography (TMS-EEG) to evaluate the excitatory and inhibitory functions of the cerebral cortex in a standardized manner. It has been postulated that schizophrenia is a disorder of functional neural connectivity underpinned by a relative imbalance of excitation and inhibition. The aim of this review was to provide a comprehensive overview of TMS-EMG and TMS-EEG research in schizophrenia, focused on excitation or inhibition, connectivity, motor cortical plasticity and the effect of antipsychotic medications, symptom severity and illness duration on TMS-EMG and TMS-EEG indices. METHODS We searched PsycINFO, Embase and Medline, from database inception to April 2020, for studies that included TMS outcomes in patients with schizophrenia. We used the following combination of search terms: transcranial magnetic stimulation OR tms AND interneurons OR glutamic acid OR gamma aminobutyric acid OR neural inhibition OR pyramidal neurons OR excita* OR inhibit* OR GABA* OR glutam* OR E-I balance OR excitation-inhibition balance AND schizoaffective disorder* OR Schizophrenia OR schizophreni*. RESULTS TMS-EMG and TMS-EEG measurements revealed deficits in excitation or inhibition, functional connectivity and motor cortical plasticity in patients with schizophrenia. Increased duration of the cortical silent period (a TMS-EMG marker of γ-aminobutyric acid B receptor activity) with clozapine was a relatively consistent finding. LIMITATIONS Most of the studies used patients with chronic schizophrenia and medicated patients, employed cross-sectional group comparisons and had small sample sizes. CONCLUSION TMS-EMG and TMS-EEG offer an opportunity to develop a novel and improved understanding of the physiologic processes that underlie schizophrenia and to assess the therapeutic effect of antipsychotic medications. In the future, these techniques may also help predict disease progression and further our understanding of the excitatory/inhibitory balance and its implications for mechanisms that underlie treatment-resistant schizophrenia.
Collapse
Affiliation(s)
- Meng di Hou
- From the Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK (Hou, Shergill); the Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK (Santoro, Biondi, Premoli); and the Kent and Medway Medical School, Canterbury, UK (Shergill)
| | | | | | | | | |
Collapse
|
15
|
Frontal neural metabolite changes in schizophrenia and their association with cognitive control: A systematic review. Neurosci Biobehav Rev 2021; 132:224-247. [PMID: 34864431 PMCID: PMC8830497 DOI: 10.1016/j.neubiorev.2021.11.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 11/05/2021] [Accepted: 11/09/2021] [Indexed: 01/01/2023]
Abstract
GABA levels are decreased in medial frontal brain areas of schizophrenia patients. Glutamate levels are lower in medial and lateral frontal areas in chronic patients. Working memory performance is associated with frontal GABA and Glu. Prediction errors are associated Glu and medial frontal GABA. Processing speed correlates with medial frontal GABA levels.
A large proportion of patients with schizophrenia exhibit deficits in cognitive control functions including working memory, processing speed and inhibitory control, which have been associated with frontal brain areas. In this systematic review, we investigated differences between chronic schizophrenia patients, first-episode (FEP) patients and healthy control groups in the neurometabolite levels of GABA, glutamate, glutamine and Glx in frontal brain areas. Additionally, we reviewed correlations between cognitive control functions or negative symptoms and these neurometabolite levels. Several studies reported decreased GABA or glutamate concentrations in frontal lobe areas, particularly in chronic schizophrenia patients, while the results were mixed for FEP patients. Working memory performance and prediction errors have been associated with frontal GABA and glutamate levels, and processing speed with frontomedial GABA levels in chronic patients. The relationship between metabolites and negative symptom severity was somewhat inconsistent. Future studies should take the participants' age, medication status or responsivity, disease stage and precise anatomical location of the voxel into account when comparing neurometabolite levels between schizophrenia patients and healthy controls.
Collapse
|
16
|
Jeon P, Limongi R, Ford SD, Mackinley M, Dempster K, Théberge J, Palaniyappan L. Progressive Changes in Glutamate Concentration in Early Stages of Schizophrenia: A Longitudinal 7-Tesla MRS Study. ACTA ACUST UNITED AC 2021; 2:sgaa072. [PMID: 34746793 PMCID: PMC8561748 DOI: 10.1093/schizbullopen/sgaa072] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Progressive reduction in glutamatergic transmission has been proposed as an important component of the illness trajectory of schizophrenia. Despite its popularity, to date, this notion has not been convincingly tested in patients in early stages of schizophrenia. In a longitudinal 7T magnetic resonance spectroscopy (1H-MRS), we quantified glutamate at the dorsal anterior cingulate cortex in 21 participants with a median lifetime antipsychotic exposure of less than 3 days and followed them up after 6 months of treatment. Ten healthy controls were also scanned at 2 time points. While patients had significantly lower overall glutamate levels than healthy controls (F(1,27) = 5.23, P = .03), we did not observe a progressive change of glutamate concentration in patients (F(1,18) = 0.47, P = .50), and the group by time interaction was not significant (F(1,27) = 0.86, P = .36). On average, patients with early psychosis receiving treatment showed a 0.02 mM/y increase, while healthy controls showed a 0.06 mM/y reduction of MRS glutamate levels. Bayesian analysis of our observations does not support early, post-onset glutamate loss in schizophrenia. Interestingly, it provides evidence in favor of a lack of progressive glutamate change in our schizophrenia sample—indicating that the glutamate level at the onset of illness was the best predictor of the levels 6 months after treatment. A more nuanced view of glutamatergic physiology, linked to early cortical maturation, may be required to understand glutamate-mediated dynamics in schizophrenia.
Collapse
Affiliation(s)
- Peter Jeon
- Department of Medical Biophysics, Western University, London, Ontario, Canada.,Lawson Health Research Institute, Imaging Division, London, Ontario, Canada
| | - Roberto Limongi
- Robarts Research Institute, Western University, London, Ontario, Canada
| | - Sabrina D Ford
- Department of Psychiatry, Western University, London, Ontario, Canada
| | - Michael Mackinley
- Department of Neuroscience, Western University, London, Ontario, Canada
| | - Kara Dempster
- Department of Psychiatry, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Jean Théberge
- Department of Medical Biophysics, Western University, London, Ontario, Canada.,Lawson Health Research Institute, Imaging Division, London, Ontario, Canada.,St. Joseph's Health Care, Diagnostic Imaging, London, Ontario, Canada.,Department of Medical Imaging, Western University, London, Ontario, Canada
| | - Lena Palaniyappan
- Department of Medical Biophysics, Western University, London, Ontario, Canada.,Robarts Research Institute, Western University, London, Ontario, Canada.,Department of Psychiatry, Western University, London, Ontario, Canada
| |
Collapse
|
17
|
Kraguljac NV, Anthony T, Morgan CJ, Jindal RD, Burger MS, Lahti AC. White matter integrity, duration of untreated psychosis, and antipsychotic treatment response in medication-naïve first-episode psychosis patients. Mol Psychiatry 2021; 26:5347-5356. [PMID: 32398721 PMCID: PMC7658031 DOI: 10.1038/s41380-020-0765-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 04/24/2020] [Accepted: 04/27/2020] [Indexed: 01/10/2023]
Abstract
It is becoming increasingly clear that longer duration of untreated psychosis (DUP) is associated with adverse clinical outcomes in patients with psychosis spectrum disorders. Because this association is often cited when justifying early intervention efforts, it is imperative to better understand underlying biological mechanisms. We enrolled 66 antipsychotic-naïve first-episode psychosis (FEP) patients and 45 matched healthy controls in this trial. At baseline, we used a human connectome style diffusion-weighted imaging (DWI) sequence to quantify white matter integrity in both groups. Patients then received 16 weeks of treatment with risperidone, 51 FEP completed the trial. We compared whole-brain fractional anisotropy (FA), mean diffusivity, axial diffusivity (AD), and radial diffusivity between groups. To test if structural white matter integrity mediates the relationship between longer DUP and poorer treatment response, we fit a mediator model and estimated indirect effects. We found decreased whole-brain FA and AD in medication-naive FEP compared with controls. In patients, lower FA was correlated with longer DUP (r = -0.32; p = 0.03) and poorer subsequent response to antipsychotic treatment (r = 0.40; p = 0.01). Importantly, we found a significant mediation effect for FA (indirect effect: -2.70; p = 0.03), indicating that DUP exerts its effects on treatment response through affecting white matter integrity. Our data provide empirical support to the idea the DUP may have fundamental pathogenic effects on the natural history of psychosis, suggest a biological mechanism underlying this phenomenon, and underscore the importance of early intervention efforts in this disabling neuropsychiatric syndrome.
Collapse
Affiliation(s)
- Nina Vanessa Kraguljac
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Thomas Anthony
- Department of Electrical and Computer Engineering/ IT Research Computing, University of Alabama at Birmingham
| | | | - Ripu Daman Jindal
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham,Department of Neurology, Birmingham VA Medical Center
| | - Mark Steven Burger
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham
| | - Adrienne Carol Lahti
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham
| |
Collapse
|
18
|
Onwordi EC, Whitehurst T, Mansur A, Statton B, Berry A, Quinlan M, O'Regan DP, Rogdaki M, Marques TR, Rabiner EA, Gunn RN, Vernon AC, Natesan S, Howes OD. The relationship between synaptic density marker SV2A, glutamate and N-acetyl aspartate levels in healthy volunteers and schizophrenia: a multimodal PET and magnetic resonance spectroscopy brain imaging study. Transl Psychiatry 2021; 11:393. [PMID: 34282130 PMCID: PMC8290006 DOI: 10.1038/s41398-021-01515-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Accepted: 06/22/2021] [Indexed: 02/07/2023] Open
Abstract
Glutamatergic excitotoxicity is hypothesised to underlie synaptic loss in schizophrenia pathogenesis, but it is unknown whether synaptic markers are related to glutamatergic function in vivo. Additionally, it has been proposed that N-acetyl aspartate (NAA) levels reflect neuronal integrity. Here, we investigated whether synaptic vesicle glycoprotein 2 A (SV2A) levels are related to glutamatergic markers and NAA in healthy volunteers (HV) and schizophrenia patients (SCZ). Forty volunteers (SCZ n = 18, HV n = 22) underwent [11C]UCB-J positron emission tomography and proton magnetic resonance spectroscopy (1H-MRS) imaging in the left hippocampus and anterior cingulate cortex (ACC) to index [11C]UCB-J distribution volume ratio (DVR), and creatine-scaled glutamate (Glu/Cr), glutamate and glutamine (Glx/Cr) and NAA (NAA/Cr). In healthy volunteers, but not patients, [11C]UCB-J DVR was significantly positively correlated with Glu/Cr, in both the hippocampus and ACC. Furthermore, in healthy volunteers, but not patients, [11C]UCB-J DVR was significantly positively correlated with Glx/Cr, in both the hippocampus and ACC. There were no significant relationships between [11C]UCB-J DVR and NAA/Cr in the hippocampus or ACC in healthy volunteers or patients. Therefore, an appreciable proportion of the brain 1H-MRS glutamatergic signal is related to synaptic density in healthy volunteers. This relationship is not seen in schizophrenia, which, taken with lower synaptic marker levels, is consistent with lower levels of glutamatergic terminals and/or a lower proportion of glutamatergic relative to GABAergic terminals in the ACC in schizophrenia.
Collapse
Affiliation(s)
- Ellis Chika Onwordi
- MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital Campus, London, W12 0NN, UK.
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, W12 0NN, UK.
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, De Crespigny Park, London, SE5 8AF, UK.
- South London and Maudsley NHS Foundation Trust, Camberwell, London, SE5 8AF, UK.
| | - Thomas Whitehurst
- MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital Campus, London, W12 0NN, UK
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, W12 0NN, UK
| | - Ayla Mansur
- Department of Brain Sciences, Imperial College London, The Commonwealth Building, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
- Invicro, Burlington Danes Building, Du Cane Road, London, W12 0NN, UK
| | - Ben Statton
- MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital Campus, London, W12 0NN, UK
| | - Alaine Berry
- MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital Campus, London, W12 0NN, UK
| | - Marina Quinlan
- MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital Campus, London, W12 0NN, UK
| | - Declan P O'Regan
- MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital Campus, London, W12 0NN, UK
| | - Maria Rogdaki
- MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital Campus, London, W12 0NN, UK
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, W12 0NN, UK
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, De Crespigny Park, London, SE5 8AF, UK
- South London and Maudsley NHS Foundation Trust, Camberwell, London, SE5 8AF, UK
| | - Tiago Reis Marques
- MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital Campus, London, W12 0NN, UK
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, De Crespigny Park, London, SE5 8AF, UK
- South London and Maudsley NHS Foundation Trust, Camberwell, London, SE5 8AF, UK
| | - Eugenii A Rabiner
- Invicro, Burlington Danes Building, Du Cane Road, London, W12 0NN, UK
- Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, De Crespigny Park, London, SE5 8AF, UK
| | - Roger N Gunn
- Department of Brain Sciences, Imperial College London, The Commonwealth Building, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
- Invicro, Burlington Danes Building, Du Cane Road, London, W12 0NN, UK
| | - Anthony C Vernon
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King's College London, 5 Cutcombe Road, London, SE5 9RT, UK
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, SE1 1UL, UK
| | - Sridhar Natesan
- MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital Campus, London, W12 0NN, UK
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, De Crespigny Park, London, SE5 8AF, UK
| | - Oliver D Howes
- MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital Campus, London, W12 0NN, UK.
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, W12 0NN, UK.
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, De Crespigny Park, London, SE5 8AF, UK.
- South London and Maudsley NHS Foundation Trust, Camberwell, London, SE5 8AF, UK.
| |
Collapse
|
19
|
Shared Biological Pathways between Antipsychotics and Omega-3 Fatty Acids: A Key Feature for Schizophrenia Preventive Treatment? Int J Mol Sci 2021; 22:ijms22136881. [PMID: 34206945 PMCID: PMC8269187 DOI: 10.3390/ijms22136881] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/18/2021] [Accepted: 06/23/2021] [Indexed: 12/25/2022] Open
Abstract
Schizophrenia typically emerges during adolescence, with progression from an ultra-high risk state (UHR) to the first episode of psychosis (FEP) followed by a chronic phase. The detailed pathophysiology of schizophrenia and the factors leading to progression across these stages remain relatively unknown. The current treatment relies on antipsychotics, which are effective for FEP and chronic schizophrenia but ineffective for UHR patients. Antipsychotics modulate dopaminergic and glutamatergic neurotransmission, inflammation, oxidative stress, and membrane lipids pathways. Many of these biological pathways intercommunicate and play a role in schizophrenia pathophysiology. In this context, research of preventive treatment in early stages has explored the antipsychotic effects of omega-3 supplementation in UHR and FEP patients. This review summarizes the action of omega-3 in various biological systems involved in schizophrenia. Similar to antipsychotics, omega-3 supplementation reduces inflammation and oxidative stress, improves myelination, modifies the properties of cell membranes, and influences dopamine and glutamate pathways. Omega-3 supplementation also modulates one-carbon metabolism, the endocannabinoid system, and appears to present neuroprotective properties. Omega-3 has little side effects compared to antipsychotics and may be safely prescribed for UHR patients and as an add-on for FEP patients. This could to lead to more efficacious individualised treatments, thus contributing to precision medicine in psychiatry.
Collapse
|
20
|
Merritt K, McGuire PK, Egerton A, Aleman A, Block W, Bloemen OJN, Borgan F, Bustillo JR, Capizzano AA, Coughlin JM, De la Fuente-Sandoval C, Demjaha A, Dempster K, Do KQ, Du F, Falkai P, Galinska-Skok B, Gallinat J, Gasparovic C, Ginestet CE, Goto N, Graff-Guerrero A, Ho BC, Howes OD, Jauhar S, Jeon P, Kato T, Kaufmann CA, Kegeles LS, Keshavan M, Kim SY, Kunugi H, Lauriello J, Liemburg EJ, Mcilwain ME, Modinos G, Mouchlianitis ED, Nakamura J, Nenadic I, Öngür D, Ota M, Palaniyappan L, Pantelis C, Plitman E, Posporelis S, Purdon SE, Reichenbach JR, Renshaw PF, Russell BR, Sawa A, Schaefer M, Shungu DC, Smesny S, Stanley JA, Stone JM, Szulc A, Taylor R, Thakkar K, Théberge J, Tibbo PG, van Amelsvoort T, Walecki J, Williamson PC, Wood SJ, Xin L, Yamasue H. Association of Age, Antipsychotic Medication, and Symptom Severity in Schizophrenia With Proton Magnetic Resonance Spectroscopy Brain Glutamate Level: A Mega-analysis of Individual Participant-Level Data. JAMA Psychiatry 2021; 78:667-681. [PMID: 33881460 PMCID: PMC8060889 DOI: 10.1001/jamapsychiatry.2021.0380] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Importance Proton magnetic resonance spectroscopy (1H-MRS) studies indicate that altered brain glutamatergic function may be associated with the pathophysiology of schizophrenia and the response to antipsychotic treatment. However, the association of altered glutamatergic function with clinical and demographic factors is unclear. Objective To assess the associations of age, symptom severity, level of functioning, and antipsychotic treatment with brain glutamatergic metabolites. Data Sources The MEDLINE database was searched to identify journal articles published between January 1, 1980, and June 3, 2020, using the following search terms: MRS or magnetic resonance spectroscopy and (1) schizophrenia or (2) psychosis or (3) UHR or (4) ARMS or (5) ultra-high risk or (6) clinical high risk or (7) genetic high risk or (8) prodrome* or (9) schizoaffective. Authors of 114 1H-MRS studies measuring glutamate (Glu) levels in patients with schizophrenia were contacted between January 2014 and June 2020 and asked to provide individual participant data. Study Selection In total, 45 1H-MRS studies contributed data. Data Extraction and Synthesis Associations of Glu, Glu plus glutamine (Glx), or total creatine plus phosphocreatine levels with age, antipsychotic medication dose, symptom severity, and functioning were assessed using linear mixed models, with study as a random factor. Main Outcomes and Measures Glu, Glx, and Cr values in the medial frontal cortex (MFC) and medial temporal lobe (MTL). Results In total, 42 studies were included, with data for 1251 patients with schizophrenia (mean [SD] age, 30.3 [10.4] years) and 1197 healthy volunteers (mean [SD] age, 27.5 [8.8] years). The MFC Glu (F1,1211.9 = 4.311, P = .04) and Glx (F1,1079.2 = 5.287, P = .02) levels were lower in patients than in healthy volunteers, and although creatine levels appeared lower in patients, the difference was not significant (F1,1395.9 = 3.622, P = .06). In both patients and volunteers, the MFC Glu level was negatively associated with age (Glu to Cr ratio, F1,1522.4 = 47.533, P < .001; cerebrospinal fluid-corrected Glu, F1,1216.7 = 5.610, P = .02), showing a 0.2-unit reduction per decade. In patients, antipsychotic dose (in chlorpromazine equivalents) was negatively associated with MFC Glu (estimate, 0.10 reduction per 100 mg; SE, 0.03) and MFC Glx (estimate, -0.11; SE, 0.04) levels. The MFC Glu to Cr ratio was positively associated with total symptom severity (estimate, 0.01 per 10 points; SE, 0.005) and positive symptom severity (estimate, 0.04; SE, 0.02) and was negatively associated with level of global functioning (estimate, 0.04; SE, 0.01). In the MTL, the Glx to Cr ratio was positively associated with total symptom severity (estimate, 0.06; SE, 0.03), negative symptoms (estimate, 0.2; SE, 0.07), and worse Clinical Global Impression score (estimate, 0.2 per point; SE, 0.06). The MFC creatine level increased with age (estimate, 0.2; SE, 0.05) but was not associated with either symptom severity or antipsychotic medication dose. Conclusions and Relevance Findings from this mega-analysis suggest that lower brain Glu levels in patients with schizophrenia may be associated with antipsychotic medication exposure rather than with greater age-related decline. Higher brain Glu levels may act as a biomarker of illness severity in schizophrenia.
Collapse
Affiliation(s)
- Kate Merritt
- Division of Psychiatry, Institute of Mental Health, UCL, London, United Kingdom
- Psychosis Studies Department, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, United Kingdom
| | - Philip K McGuire
- Psychosis Studies Department, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, United Kingdom
| | - Alice Egerton
- Psychosis Studies Department, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, United Kingdom
| | - André Aleman
- Center for Brain Disorder and Cognitive Science, Shenzhen University, Shenzhen, China
- University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Wolfgang Block
- Department of Diagnostic and Interventional Radiology, University Hospital Bonn, Bonn, Germany
| | - Oswald J N Bloemen
- Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, The Netherlands
| | - Faith Borgan
- Psychosis Studies Department, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, United Kingdom
| | - Juan R Bustillo
- Department of Psychiatry and Behavioral Sciences, Center for Psychiatric Research, University of New Mexico School of Medicine, Albuquerque
| | - Aristides A Capizzano
- Department of Radiology, Division of Neuroradiology, University of Michigan, Ann Arbor
| | - Jennifer Marie Coughlin
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Camilo De la Fuente-Sandoval
- Laboratory of Experimental Psychiatry, Instituto Nacional de Neurología y Neurocirugía, Mexico City, Mexico
- Neuropsychiatry Department, Instituto Nacional de Neurología y Neurocirugía, Mexico City, Mexico
| | - Arsime Demjaha
- Psychosis Studies Department, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, United Kingdom
| | - Kara Dempster
- Department of Psychiatry, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Kim Q Do
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital-CHUV, Prilly-Lausanne, Switzerland
| | - Fei Du
- Psychotic Disorders Division, McLean Hospital, Harvard Medical School, Belmont, Massachusetts
| | - Peter Falkai
- Department of Psychiatry, University Hospital, LMU Munich, Munich, Germany
| | - Beata Galinska-Skok
- Department of Psychiatry, Medical University of Bialystok, Bialystok, Poland
| | - Jurgen Gallinat
- Department of Psychiatry and Psychotherapy, University Medical Center Hamburg-Eppendorf (UKE), Germany
| | | | - Cedric E Ginestet
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology and Neuroscience King's College London, London, United Kingdom
| | - Naoki Goto
- Department of Psychiatry, Kokura Gamo Hospital, Kitakyushu, Fukuoka, Japan
| | - Ariel Graff-Guerrero
- Multimodal Neuroimaging Schizophrenia Group, Research Imaging Centre, Geriatric Mental Health Program at Centre for Addiction and Mental Health, Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Beng Choon Ho
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City
| | - Oliver D Howes
- Psychosis Studies Department, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, United Kingdom
| | - Sameer Jauhar
- Psychosis Studies Department, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, United Kingdom
| | - Peter Jeon
- Department of Medical Biophysics, University of Western Ontario, London, Ontario, Canada
| | - Tadafumi Kato
- Department of Psychiatry and Behavioral Science, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Charles A Kaufmann
- Department of Psychiatry, Columbia University, New York State Psychiatric Institute, New York
| | - Lawrence S Kegeles
- Department of Psychiatry, Columbia University, New York State Psychiatric Institute, New York
| | | | | | - Hiroshi Kunugi
- National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| | - John Lauriello
- Jefferson Health-Sidney Kimmel Medical College, Philadelphia, Pennsylvania
| | - Edith Jantine Liemburg
- Rob Giel Research Center, Department of Psychiatry, University Medical Center Groningen, Groningen, The Netherlands
| | - Meghan E Mcilwain
- School of Pharmacy, University of Auckland, Grafton, Auckland, New Zealand
| | - Gemma Modinos
- Psychosis Studies Department, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, United Kingdom
- Department of Neuroimaging, Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology & Neuroscience, De Crespigny Park, London, United Kingdom
| | - Elias D Mouchlianitis
- Psychosis Studies Department, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, United Kingdom
| | - Jun Nakamura
- Department of Psychiatry, University of Occupational and Environmental Health, Kitakyushu, Fukuoka, Japan
| | - Igor Nenadic
- Department of Psychiatry and Psychotherapy, University Medical Center Hamburg-Eppendorf (UKE), Germany
| | - Dost Öngür
- Psychotic Disorders Division, McLean Hospital, Harvard Medical School, Belmont, Massachusetts
- Editor, JAMA Psychiatry
| | - Miho Ota
- National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| | - Lena Palaniyappan
- Department of Medical Biophysics, University of Western Ontario, London, Ontario, Canada
- Department of Psychiatry, Western University, London, Ontario, Canada
| | - Christos Pantelis
- Melbourne Neuropsychiatry Centre, The University of Melbourne and Melbourne Health, Carlton, Victoria, Australia
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
| | - Eric Plitman
- Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec, Canada
- Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| | - Sotirios Posporelis
- Psychosis Studies Department, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, United Kingdom
- South London and Maudsley, Bethlem Royal Hospital, Beckenham, United Kingdom
| | - Scot E Purdon
- Neuropsychology Department, Alberta Hospital Edmonton, Edmonton, Alberta, Canada
- Edmonton Early Intervention in Psychosis Clinic, Edmonton, Alberta, Canada
| | - Jürgen R Reichenbach
- Medical Physics Group, Institute for Diagnostic and Interventional Radiology, Jena University Hospital, Jena, Germany
| | - Perry F Renshaw
- Department of Psychiatry, University of Utah, Salt Lake City
| | - Bruce R Russell
- School of Pharmacy, University of Otago, Dunedin, New Zealand
| | - Akira Sawa
- Department of Psychiatry, Johns Hopkins University, Baltimore, Maryland
- Department of Neuroscience, Johns Hopkins University, Baltimore, Maryland
- Department of Mental Health, Johns Hopkins University, Baltimore, Maryland
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland
- Department of Genetic Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Martin Schaefer
- Department of Psychiatry, Psychotherapy, Psychosomatics and Addiction Medicine, Kliniken Essen-Mitte, Essen, Germany
- Department of Psychiatry and Psychotherapy, Charité-Universitätsmedizin Berlin, Campus Charité Mitte, Berlin, Germany
| | - Dikoma C Shungu
- Department of Radiology, Weill Cornell Medical College, New York, New York
| | - Stefan Smesny
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Jena, Germany
| | - Jeffrey A Stanley
- Brain Imaging Research Division, Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, Michigan
| | - James M Stone
- Department of Neuroimaging, Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology & Neuroscience, De Crespigny Park, London, United Kingdom
- Brighton and Sussex Medical School, University of Sussex, Brighton, United Kingdom
| | - Agata Szulc
- Department of Psychiatry, Medical University of Warsaw, Poland
| | - Reggie Taylor
- Brighton and Sussex Medical School, University of Sussex, Brighton, United Kingdom
- Lawson Health Research Institute, London, Ontario, Canada
| | - Katy Thakkar
- Department of Psychology, Michigan State University, East Lansing
- Division of Psychiatry and Behavioral Medicine, Michigan State University, East Lansing
| | - Jean Théberge
- Department of Medical Biophysics, University of Western Ontario, London, Ontario, Canada
- Department of Psychiatry, Western University, London, Ontario, Canada
- Lawson Health Research Institute, London, Ontario, Canada
| | - Philip G Tibbo
- Department of Psychiatry, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Therese van Amelsvoort
- Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, The Netherlands
| | | | - Peter C Williamson
- Department of Psychiatry, Western University, London, Ontario, Canada
- Lawson Health Research Institute, London, Ontario, Canada
| | - Stephen James Wood
- Orygen, Melbourne, Australia
- Institute for Mental Health, University of Birmingham, Edgbaston, United Kingdom
- Centre for Youth Mental Health, University of Melbourne, Australia
| | - Lijing Xin
- Animal Imaging and Technology Core, Center for Biomedical Imaging, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Hidenori Yamasue
- Department of Psychiatry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| |
Collapse
|
21
|
Kraguljac NV, McDonald WM, Widge AS, Rodriguez CI, Tohen M, Nemeroff CB. Neuroimaging Biomarkers in Schizophrenia. Am J Psychiatry 2021; 178:509-521. [PMID: 33397140 PMCID: PMC8222104 DOI: 10.1176/appi.ajp.2020.20030340] [Citation(s) in RCA: 119] [Impact Index Per Article: 39.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Schizophrenia is a complex neuropsychiatric syndrome with a heterogeneous genetic, neurobiological, and phenotypic profile. Currently, no objective biological measures-that is, biomarkers-are available to inform diagnostic or treatment decisions. Neuroimaging is well positioned for biomarker development in schizophrenia, as it may capture phenotypic variations in molecular and cellular disease targets, or in brain circuits. These mechanistically based biomarkers may represent a direct measure of the pathophysiological underpinnings of the disease process and thus could serve as true intermediate or surrogate endpoints. Effective biomarkers could validate new treatment targets or pathways, predict response, aid in selection of patients for therapy, determine treatment regimens, and provide a rationale for personalized treatments. In this review, the authors discuss a range of mechanistically plausible neuroimaging biomarker candidates, including dopamine hyperactivity, N-methyl-d-aspartate receptor hypofunction, hippocampal hyperactivity, immune dysregulation, dysconnectivity, and cortical gray matter volume loss. They then focus on the putative neuroimaging biomarkers for disease risk, diagnosis, target engagement, and treatment response in schizophrenia. Finally, they highlight areas of unmet need and discuss strategies to advance biomarker development.
Collapse
Affiliation(s)
- Nina V. Kraguljac
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL,Corresponding Author: Nina Vanessa Kraguljac, MD, Department of Psychiatry and Behavioral Neurobiology, The University of Alabama at Birmingham, SC 501, 1720 7th Ave S, Birmingham, AL 35294-0017, 205-996-7171,
| | - William M. McDonald
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine
| | - Alik S. Widge
- Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis, MN
| | - Carolyn I. Rodriguez
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA,Veterans Affairs Palo Alto Health Care System, Palo Alto, CA
| | - Mauricio Tohen
- Department of Psychiatry and Behavioral Sciences, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Charles B. Nemeroff
- Department of Psychiatry, University of Texas Dell Medical School, Austin, TX
| |
Collapse
|
22
|
McQueen G, Sendt KV, Gillespie A, Avila A, Lally J, Vallianatou K, Chang N, Ferreira D, Borgan F, Howes OD, Barker GJ, Lythgoe DJ, Stone JM, McGuire P, MacCabe JH, Egerton A. Changes in Brain Glutamate on Switching to Clozapine in Treatment-Resistant Schizophrenia. Schizophr Bull 2021; 47:662-671. [PMID: 33398325 PMCID: PMC8084451 DOI: 10.1093/schbul/sbaa156] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
It has been suggested that the antipsychotic clozapine may modulate brain glutamate, and that this effect could contribute to its efficacy in treatment-resistant schizophrenia (TRS). The aim of this study was to examine the effects of clozapine on brain glutamate in TRS longitudinally. This study examined individuals with TRS before and 12 weeks after switching from a non-clozapine antipsychotic to treatment with clozapine as part of their normal clinical care. Proton magnetic resonance spectroscopy (1H-MRS) measured concentrations, corrected for voxel tissue content, of glutamate (Glucorr), and glutamate plus glutamine (Glxcorr) in the anterior cingulate cortex (ACC) and right caudate nucleus. Symptoms were monitored using the Positive and Negative Syndrome Scale (PANSS). Of 37 recruited patients (27 men, 39.30 years old, 84% clozapine naïve), 25 completed 1H-MRS at both timepoints. 12 weeks of clozapine was associated with a longitudinal reduction in Glucorr in the caudate (n = 23, F = 7.61 P = .01) but not in the ACC (n = 24, F = 0.02, P = .59). Percentage reduction in caudate Glucorr was positively correlated with percentage improvement in symptoms (total PANSS score, n = 23, r = .42, P = .04). These findings indicate that reductions in glutamate in the caudate nucleus may contribute to symptomatic improvement during the first months of clozapine treatment.
Collapse
Affiliation(s)
- Grant McQueen
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London, UK
| | - Kyra-Verena Sendt
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London, UK
| | - Amy Gillespie
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London, UK
| | - Alessia Avila
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London, UK
| | - John Lally
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London, UK.,Department of Psychiatry, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Kalliopi Vallianatou
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London, UK
| | - Nynn Chang
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London, UK
| | - Diogo Ferreira
- Hospital de Santa Maria, Centro Hospitalar Universitário de Lisboa Norte, Lisbon, Portugal
| | - Faith Borgan
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London, UK
| | - Oliver D Howes
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London, UK
| | - Gareth J Barker
- Department of Neuroimaging, Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology & Neuroimaging, King's College London, De Crespigny Park, London, UK
| | - David J Lythgoe
- Department of Neuroimaging, Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology & Neuroimaging, King's College London, De Crespigny Park, London, UK
| | - James M Stone
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London, UK.,South London and Maudsley NHS Trust, London, UK
| | - Philip McGuire
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London, UK
| | - James H MacCabe
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London, UK
| | - Alice Egerton
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London, UK
| |
Collapse
|
23
|
Egerton A, Murphy A, Donocik J, Anton A, Barker GJ, Collier T, Deakin B, Drake R, Eliasson E, Emsley R, Gregory CJ, Griffiths K, Kapur S, Kassoumeri L, Knight L, Lambe EJB, Lawrie SM, Lees J, Lewis S, Lythgoe DJ, Matthews J, McGuire P, McNamee L, Semple S, Shaw AD, Singh KD, Stockton-Powdrell C, Talbot PS, Veronese M, Wagner E, Walters JTR, Williams SR, MacCabe JH, Howes OD. Dopamine and Glutamate in Antipsychotic-Responsive Compared With Antipsychotic-Nonresponsive Psychosis: A Multicenter Positron Emission Tomography and Magnetic Resonance Spectroscopy Study (STRATA). Schizophr Bull 2021; 47:505-516. [PMID: 32910150 PMCID: PMC7965076 DOI: 10.1093/schbul/sbaa128] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The variability in the response to antipsychotic medication in schizophrenia may reflect between-patient differences in neurobiology. Recent cross-sectional neuroimaging studies suggest that a poorer therapeutic response is associated with relatively normal striatal dopamine synthesis capacity but elevated anterior cingulate cortex (ACC) glutamate levels. We sought to test whether these measures can differentiate patients with psychosis who are antipsychotic responsive from those who are antipsychotic nonresponsive in a multicenter cross-sectional study. 1H-magnetic resonance spectroscopy (1H-MRS) was used to measure glutamate levels (Glucorr) in the ACC and in the right striatum in 92 patients across 4 sites (48 responders [R] and 44 nonresponders [NR]). In 54 patients at 2 sites (25 R and 29 NR), we additionally acquired 3,4-dihydroxy-6-[18F]fluoro-l-phenylalanine (18F-DOPA) positron emission tomography (PET) to index striatal dopamine function (Kicer, min-1). The mean ACC Glucorr was higher in the NR than the R group after adjustment for age and sex (F1,80 = 4.27; P = .04). This was associated with an area under the curve for the group discrimination of 0.59. There were no group differences in striatal dopamine function or striatal Glucorr. The results provide partial further support for a role of ACC glutamate, but not striatal dopamine synthesis, in determining the nature of the response to antipsychotic medication. The low discriminative accuracy might be improved in groups with greater clinical separation or increased in future studies that focus on the antipsychotic response at an earlier stage of the disorder and integrate other candidate predictive biomarkers. Greater harmonization of multicenter PET and 1H-MRS may also improve sensitivity.
Collapse
Affiliation(s)
- Alice Egerton
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
- NIHR Biomedical Research Centre at South London and Maudsley NHS Foundation Trust, London, UK
| | - Anna Murphy
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Jacek Donocik
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
| | - Adriana Anton
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Academic Unit of Radiology, Medical School, Faculty of Medicine, Dentistry & Health, University of Sheffield, Sheffield, UK
| | - Gareth J Barker
- NIHR Biomedical Research Centre at South London and Maudsley NHS Foundation Trust, London, UK
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
| | - Tracy Collier
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
- NIHR Biomedical Research Centre at South London and Maudsley NHS Foundation Trust, London, UK
| | - Bill Deakin
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Richard Drake
- Division of Psychology and Mental Health, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Emma Eliasson
- Division of Psychiatry, University of Edinburgh, Edinburgh, UK
| | - Richard Emsley
- NIHR Biomedical Research Centre at South London and Maudsley NHS Foundation Trust, London, UK
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
| | - Catherine J Gregory
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Kira Griffiths
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
| | - Shitij Kapur
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
- NIHR Biomedical Research Centre at South London and Maudsley NHS Foundation Trust, London, UK
- Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria, Australia
| | - Laura Kassoumeri
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
- NIHR Biomedical Research Centre at South London and Maudsley NHS Foundation Trust, London, UK
| | - Laura Knight
- CUBRIC, School of Psychology, College of Biomedical and Life Sciences, Cardiff University, Cardiff, UK
| | - Emily J B Lambe
- CUBRIC, School of Psychology, College of Biomedical and Life Sciences, Cardiff University, Cardiff, UK
| | | | - Jane Lees
- Division of Psychology and Mental Health, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Shôn Lewis
- Division of Psychology and Mental Health, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - David J Lythgoe
- NIHR Biomedical Research Centre at South London and Maudsley NHS Foundation Trust, London, UK
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
| | - Julian Matthews
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Philip McGuire
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
- NIHR Biomedical Research Centre at South London and Maudsley NHS Foundation Trust, London, UK
| | - Lily McNamee
- Division of Psychiatry, University of Edinburgh, Edinburgh, UK
| | - Scott Semple
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Alexander D Shaw
- CUBRIC, School of Psychology, College of Biomedical and Life Sciences, Cardiff University, Cardiff, UK
| | - Krish D Singh
- CUBRIC, School of Psychology, College of Biomedical and Life Sciences, Cardiff University, Cardiff, UK
| | - Charlotte Stockton-Powdrell
- Division of Psychology and Mental Health, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Peter S Talbot
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Mattia Veronese
- NIHR Biomedical Research Centre at South London and Maudsley NHS Foundation Trust, London, UK
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
| | - Ernest Wagner
- Division of Psychiatry, University of Edinburgh, Edinburgh, UK
| | - James T R Walters
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
| | - Stephen R Williams
- Division of Informatics, Imaging and Data Sciences, University of Manchester, Manchester, UK
| | - James H MacCabe
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
- NIHR Biomedical Research Centre at South London and Maudsley NHS Foundation Trust, London, UK
| | - Oliver D Howes
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
- NIHR Biomedical Research Centre at South London and Maudsley NHS Foundation Trust, London, UK
- Psychiatric Imaging Group, MRC London Institute of Medical Sciences, Hammersmith Hospital, London, UK
| |
Collapse
|
24
|
Smesny S, Berberich D, Gussew A, Schönfeld N, Langbein K, Walther M, Reichenbach JR. Alterations of neurometabolism in the dorsolateral prefrontal cortex and thalamus in transition to psychosis patients change under treatment as usual - A two years follow-up 1H/ 31P-MR-spectroscopy study. Schizophr Res 2021; 228:7-18. [PMID: 33429152 DOI: 10.1016/j.schres.2020.11.063] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 11/22/2020] [Accepted: 11/27/2020] [Indexed: 11/25/2022]
Abstract
BACKGROUND The ultra-high risk (UHR) paradigm allows early contact with patients developing acute psychosis and the study of treatment effects on the underlying pathology. METHODS 29 patients with first acute psychosis according to CAARMS criteria (transition patients, TP) (T0) and thereof 22 patients after two-year follow-up (mean 788 d) (T1) underwent 1H-/31P-MR spectroscopy of the prefrontal (DLPFC) and anterior midcingulate (aMCC) cortices and the thalamus. N-acetylaspartate (NAA), glutamate (Glu, Glx), energy (PCr, ATP) and phospholipid metabolites (PME, PDE) were compared to 27 healthy controls by ANCOVA and correlated with patients' symptom ratings (BPRS-E, SCL-90R). For longitudinal analysis, linear mixed model (LMM) and ANCOVA for repeated measures were used. RESULTS DLPFC: In patients, NAA and PME were decreased bilaterally and Glu on the left side at T0. Left-sided Glu and NAA (trend) and bilateral Glx increased during follow-up. Thalamus: In TP, bilateral NAA, left-sided Glu and right-sided Glx were decreased at T0; bilateral NAA and left-sided Glx increased during follow-up. aMCC: In TP, bilateral NAA, right-sided Glu, and bilateral PME and PDE were decreased, while left-sided PCr was increased at T0. No changes were observed during follow-up. CONCLUSION Regardless of the long-term diagnosis, the psychotic state of illness includes disturbed neuronal function in the DLPFC, thalamus and aMCC. Treatment-as-usual (TAU), including antipsychotic/antidepressant medication and supportive psychotherapy, had an effect on the thalamo-frontal area but not or less pronounced on the neurometabolic deficits of the aMCC.
Collapse
Affiliation(s)
- Stefan Smesny
- Department of Psychiatry, Jena University Hospital, Philosophenweg 3, D-07743 Jena, Germany.
| | - Diana Berberich
- Department of Psychiatry, Jena University Hospital, Philosophenweg 3, D-07743 Jena, Germany
| | - Alexander Gussew
- Department of Radiology, University Hospital Halle (Saale), Ernst-Grube-Str. 40, D-06120 Halle (Saale), Germany
| | - Nils Schönfeld
- Department of Psychiatry, Jena University Hospital, Philosophenweg 3, D-07743 Jena, Germany
| | - Kerstin Langbein
- Department of Psychiatry, Jena University Hospital, Philosophenweg 3, D-07743 Jena, Germany
| | - Mario Walther
- Jena University of Applied Sciences, Department of Fundamental Sciences, Carl-Zeiss-Promenade 2, D-07745 Jena, Germany
| | - Jürgen R Reichenbach
- Medical Physics Group, Department of Diagnostic and Interventional Radiology, Jena University Hospital, Philosophenweg 3, D-07740 Jena, Germany
| |
Collapse
|
25
|
Kaminski J, Mascarell-Maricic L, Fukuda Y, Katthagen T, Heinz A, Schlagenhauf F. Glutamate in the Dorsolateral Prefrontal Cortex in Patients With Schizophrenia: A Meta-analysis of 1H-Magnetic Resonance Spectroscopy Studies. Biol Psychiatry 2021; 89:270-277. [PMID: 33129486 DOI: 10.1016/j.biopsych.2020.09.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 08/28/2020] [Accepted: 09/01/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND To date, there is no systematic overview of glutamate in the dorsolateral prefrontal cortex (DLPFC) of patients with schizophrenia. Here, we meta-analyzed case-control studies of high-field proton magnetic resonance spectroscopy (1H-MRS) investigating glutamate in DLPFC. Additionally, we estimated variance ratios to investigate homo/heterogeneity. METHODS Preregistration of the study was performed on September 20, 2019. The predefined literature search on PubMed comprised articles with search terms (magnetic resonance spectroscopy OR MRS) AND (glutamate OR glut∗ OR GLX) AND (schizophrenia OR psychosis OR schizophren∗). Meta-analyses with a fixed- and random-effects model with inverse variance method, DerSimonian-Laird estimator for τ2, and Cohen's d were calculated. For differences in variability, we calculated a random-effects model for measures of variance ratios. The primary study outcome was the difference in glutamate in the DLPFC in cases versus controls. Secondary outcomes were differences in variability. RESULTS The quantitative analysis comprised 429 cases and 365 controls. Overall, we found no group difference (d = 0.03 [95% confidence interval (CI), -0.20 to 0.26], z = 0.28, p = .78). Sensitivity analysis revealed an effect for medication status (Q = 8.35, p = .039), i.e., increased glutamate in antipsychotic-naïve patients (d = 0.46 [95% CI, 0.08 to 0.84], z = 2.37, p = .018). Concerning variance ratios, we found an effect of medication status (Q = 16.95, p < .001) due to lower coefficient of variation ratio (CVR) in medication-naïve patients (logCVR = -0.49 [95% CI, -0.78 to -0.20], z = -3.33, p < .001). In studies with medicated patients, we found higher CVR (logCVR = 0.22 [95% CI, 0.06 to 0.39], z = 2.67; p = .008). CONCLUSIONS We carefully interpret the higher levels and lower variability in cortical glutamate in antipsychotic-naïve patients as a possible key factor resulting from a putative allostatic mechanism. We conclude that care has to be taken when evaluating metabolite levels in clinical samples in which medication might confound findings.
Collapse
Affiliation(s)
- Jakob Kaminski
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Psychiatry and Psychotherapy CCM, Berlin, Germany; Berlin Institute of Health, Berlin, Germany.
| | - Lea Mascarell-Maricic
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Psychiatry and Psychotherapy CCM, Berlin, Germany
| | - Yu Fukuda
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Psychiatry and Psychotherapy CCM, Berlin, Germany
| | - Teresa Katthagen
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Psychiatry and Psychotherapy CCM, Berlin, Germany
| | - Andreas Heinz
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Psychiatry and Psychotherapy CCM, Berlin, Germany; Bernstein Center for Computational Neuroscience, Berlin, Germany
| | - Florian Schlagenhauf
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Psychiatry and Psychotherapy CCM, Berlin, Germany; Berlin Institute of Health, Berlin, Germany; Bernstein Center for Computational Neuroscience, Berlin, Germany; Department of Neurology, Max Planck Institute for Human Cognitive and Brain Science, Leipzig, Germany
| |
Collapse
|
26
|
Granger KT, Talwar A, Barnett JH. Latent inhibition and its potential as a biomarker for schizophrenia. Biomark Neuropsychiatry 2020. [DOI: 10.1016/j.bionps.2020.100025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
|
27
|
Egerton A, Grace AA, Stone J, Bossong MG, Sand M, McGuire P. Glutamate in schizophrenia: Neurodevelopmental perspectives and drug development. Schizophr Res 2020; 223:59-70. [PMID: 33071070 DOI: 10.1016/j.schres.2020.09.013] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 08/12/2020] [Accepted: 09/20/2020] [Indexed: 12/14/2022]
Abstract
Research into the neurobiological processes that may lead to the onset of schizophrenia places growing emphasis on the glutamatergic system and brain development. Preclinical studies have shown that neurodevelopmental, genetic, and environmental factors contribute to glutamatergic dysfunction and schizophrenia-related phenotypes. Clinical research has suggested that altered brain glutamate levels may be present before the onset of psychosis and relate to outcome in those at clinical high risk. After psychosis onset, glutamate dysfunction may also relate to the degree of antipsychotic response and clinical outcome. These findings support ongoing efforts to develop pharmacological interventions that target the glutamate system and could suggest that glutamatergic compounds may be more effective in specific patient subgroups or illness stages. In this review, we consider the updated glutamate hypothesis of schizophrenia, from a neurodevelopmental perspective, by reviewing recent preclinical and clinical evidence, and discuss the potential implications for novel therapeutics.
Collapse
Affiliation(s)
- Alice Egerton
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
| | - Anthony A Grace
- Departments of Neuroscience, Psychiatry and Psychology, University of Pittsburgh, Pittsburgh, PA, USA
| | - James Stone
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Matthijs G Bossong
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Michael Sand
- Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, CT, USA
| | - Philip McGuire
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| |
Collapse
|
28
|
Kubota M, Moriguchi S, Takahata K, Nakajima S, Horita N. Treatment effects on neurometabolite levels in schizophrenia: A systematic review and meta-analysis of proton magnetic resonance spectroscopy studies. Schizophr Res 2020; 222:122-132. [PMID: 32505446 DOI: 10.1016/j.schres.2020.03.069] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 01/29/2020] [Accepted: 03/29/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND Although there is growing evidence of alterations in the neurometabolite status associated with the pathophysiology of schizophrenia, how treatments influence these metabolite levels in patients with schizophrenia remains poorly studied. METHODS We conducted a literature search using Embase, Medline, and PsycINFO to identify proton magnetic resonance spectroscopy studies that compared neurometabolite levels before and after treatment in patients with schizophrenia. Six neurometabolites (glutamate, glutamine, glutamate + glutamine, gamma-aminobutyric acid, N-acetylaspartate, myo-inositol) and six regions of interest (frontal cortex, temporal cortex, parieto-occipital cortex, thalamus, basal ganglia, hippocampus) were investigated. RESULTS Thirty-two studies (n = 773 at follow-up) were included in our meta-analysis. Our results demonstrated that the frontal glutamate + glutamine level was significantly decreased (14 groups; n = 292 at follow-up; effect size = -0.35, P = 0.0003; I2 = 22%) and the thalamic N-acetylaspartate level was significantly increased (7 groups; n = 184 at follow-up; effect size = 0.47, P < 0.00001; I2 = 0%) after treatment in schizophrenia patients. No significant associations were found between neurometabolite changes and age, gender, duration of illness, duration of treatment, or baseline symptom severity. CONCLUSIONS The current results suggest that glutamatergic neurometabolite levels in the frontal cortex and neuronal integrity in the thalamus in schizophrenia might be modified following treatment.
Collapse
Affiliation(s)
- Manabu Kubota
- Department of Functional Brain Imaging, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan; Department of Psychiatry, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan.
| | - Sho Moriguchi
- Department of Functional Brain Imaging, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan; Research Imaging Centre, Centre for Addiction and Mental Health, 250 College Street, Toronto, Ontario M5T1R8, Canada
| | - Keisuke Takahata
- Department of Functional Brain Imaging, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan; Department of Neuropsychiatry, Keio University Graduate School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Shinichiro Nakajima
- Research Imaging Centre, Centre for Addiction and Mental Health, 250 College Street, Toronto, Ontario M5T1R8, Canada; Department of Neuropsychiatry, Keio University Graduate School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Nobuyuki Horita
- Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan
| |
Collapse
|
29
|
Wenneberg C, Nordentoft M, Rostrup E, Glenthøj LB, Bojesen KB, Fagerlund B, Hjorthøj C, Krakauer K, Kristensen TD, Schwartz C, Edden RAE, Broberg BV, Glenthøj BY. Cerebral Glutamate and Gamma-Aminobutyric Acid Levels in Individuals at Ultra-high Risk for Psychosis and the Association With Clinical Symptoms and Cognition. BIOLOGICAL PSYCHIATRY: COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2020; 5:569-579. [PMID: 32008981 DOI: 10.1016/j.bpsc.2019.12.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 12/09/2019] [Accepted: 12/10/2019] [Indexed: 12/31/2022]
Abstract
BACKGROUND Studies examining glutamate or gamma-aminobutyric acid (GABA) in ultra-high risk for psychosis (UHR) and the association with pathophysiology and cognition have shown conflicting results. We aimed to determine whether perturbed glutamate and GABA levels in the anterior cingulate cortex and glutamate levels in the left thalamus were present in UHR individuals and to investigate associations between metabolite levels and clinical symptoms and cognition. METHODS We included 122 UHR individuals and 60 healthy control subjects. Participants underwent proton magnetic resonance spectroscopy to estimate glutamate and GABA levels and undertook clinical and cognitive assessments. RESULTS We found no differences in metabolite levels between UHR individuals and healthy control subjects. In UHR individuals, we found negative correlations in the anterior cingulate cortex between the composite of glutamate and glutamine (Glx) and the Comprehensive Assessment of At-Risk Mental States composite score (p = .04) and between GABA and alogia (p = .01); positive associations in the anterior cingulate cortex between glutamate (p = .01) and Glx (p = .01) and spatial working memory and between glutamate (p = .04), Glx (p = .04), and GABA (p = .02) and set-shifting; and a positive association in the thalamus between glutamate and attention (p = .04). No associations between metabolites and clinical or cognitive scores were found in the healthy control subjects. CONCLUSIONS An association between glutamate and GABA levels and clinical symptoms and cognition found only in UHR individuals suggests a loss of the normal relationship between metabolite levels and cognitive function. Longitudinal studies with investigation of clinical and cognitive outcome and the association with baseline levels of glutamate and GABA could illuminate whether glutamatergic and GABAergic dysfunction predicts clinical outcome.
Collapse
Affiliation(s)
- Christina Wenneberg
- Copenhagen Research Center for Mental Health, Mental Health Center Copenhagen, Copenhagen University Hospital, Hellerup, Denmark; Center for Neuropsychiatric Schizophrenia Research and Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, Mental Health Center, University of Copenhagen, Glostrup, Denmark; Functional Imaging Unit, Department of Clinical Physiology, Nuclear Medicine and PET, Copenhagen University Hospital Rigshospitalet, Glostrup, Denmark.
| | - Merete Nordentoft
- Copenhagen Research Center for Mental Health, Mental Health Center Copenhagen, Copenhagen University Hospital, Hellerup, Denmark
| | - Egill Rostrup
- Center for Neuropsychiatric Schizophrenia Research and Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, Mental Health Center, University of Copenhagen, Glostrup, Denmark; Functional Imaging Unit, Department of Clinical Physiology, Nuclear Medicine and PET, Copenhagen University Hospital Rigshospitalet, Glostrup, Denmark
| | - Louise Birkedal Glenthøj
- Copenhagen Research Center for Mental Health, Mental Health Center Copenhagen, Copenhagen University Hospital, Hellerup, Denmark; Center for Neuropsychiatric Schizophrenia Research and Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, Mental Health Center, University of Copenhagen, Glostrup, Denmark
| | - Kirsten Borup Bojesen
- Center for Neuropsychiatric Schizophrenia Research and Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, Mental Health Center, University of Copenhagen, Glostrup, Denmark
| | - Birgitte Fagerlund
- Center for Neuropsychiatric Schizophrenia Research and Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, Mental Health Center, University of Copenhagen, Glostrup, Denmark
| | - Carsten Hjorthøj
- Copenhagen Research Center for Mental Health, Mental Health Center Copenhagen, Copenhagen University Hospital, Hellerup, Denmark; Section of Epidemiology, Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Kristine Krakauer
- Copenhagen Research Center for Mental Health, Mental Health Center Copenhagen, Copenhagen University Hospital, Hellerup, Denmark; Center for Neuropsychiatric Schizophrenia Research and Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, Mental Health Center, University of Copenhagen, Glostrup, Denmark; Functional Imaging Unit, Department of Clinical Physiology, Nuclear Medicine and PET, Copenhagen University Hospital Rigshospitalet, Glostrup, Denmark
| | - Tina Dam Kristensen
- Copenhagen Research Center for Mental Health, Mental Health Center Copenhagen, Copenhagen University Hospital, Hellerup, Denmark; Center for Neuropsychiatric Schizophrenia Research and Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, Mental Health Center, University of Copenhagen, Glostrup, Denmark
| | - Camilla Schwartz
- Functional Imaging Unit, Department of Clinical Physiology, Nuclear Medicine and PET, Copenhagen University Hospital Rigshospitalet, Glostrup, Denmark
| | - Richard A E Edden
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland; F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland
| | - Brian Villumsen Broberg
- Center for Neuropsychiatric Schizophrenia Research and Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, Mental Health Center, University of Copenhagen, Glostrup, Denmark
| | - Birte Yding Glenthøj
- Center for Neuropsychiatric Schizophrenia Research and Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, Mental Health Center, University of Copenhagen, Glostrup, Denmark
| |
Collapse
|
30
|
McCutcheon RA, Krystal JH, Howes OD. Dopamine and glutamate in schizophrenia: biology, symptoms and treatment. World Psychiatry 2020; 19:15-33. [PMID: 31922684 PMCID: PMC6953551 DOI: 10.1002/wps.20693] [Citation(s) in RCA: 301] [Impact Index Per Article: 75.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Glutamate and dopamine systems play distinct roles in terms of neuronal signalling, yet both have been proposed to contribute significantly to the pathophysiology of schizophrenia. In this paper we assess research that has implicated both systems in the aetiology of this disorder. We examine evidence from post-mortem, preclinical, pharmacological and in vivo neuroimaging studies. Pharmacological and preclinical studies implicate both systems, and in vivo imaging of the dopamine system has consistently identified elevated striatal dopamine synthesis and release capacity in schizophrenia. Imaging of the glutamate system and other aspects of research on the dopamine system have produced less consistent findings, potentially due to methodological limitations and the heterogeneity of the disorder. Converging evidence indicates that genetic and environmental risk factors for schizophrenia underlie disruption of glutamatergic and dopaminergic function. However, while genetic influences may directly underlie glutamatergic dysfunction, few genetic risk variants directly implicate the dopamine system, indicating that aberrant dopamine signalling is likely to be predominantly due to other factors. We discuss the neural circuits through which the two systems interact, and how their disruption may cause psychotic symptoms. We also discuss mechanisms through which existing treatments operate, and how recent research has highlighted opportunities for the development of novel pharmacological therapies. Finally, we consider outstanding questions for the field, including what remains unknown regarding the nature of glutamate and dopamine function in schizophrenia, and what needs to be achieved to make progress in developing new treatments.
Collapse
Affiliation(s)
- Robert A McCutcheon
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital, London, UK
- South London and Maudsley Foundation NHS Trust, Maudsley Hospital, London, UK
| | - John H Krystal
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- VA National Center for PTSD, VA Connecticut Healthcare System, West Haven, CT, USA
| | - Oliver D Howes
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital, London, UK
- South London and Maudsley Foundation NHS Trust, Maudsley Hospital, London, UK
| |
Collapse
|
31
|
Kaminski J, Gleich T, Fukuda Y, Katthagen T, Gallinat J, Heinz A, Schlagenhauf F. Association of Cortical Glutamate and Working Memory Activation in Patients With Schizophrenia: A Multimodal Proton Magnetic Resonance Spectroscopy and Functional Magnetic Resonance Imaging Study. Biol Psychiatry 2020; 87:225-233. [PMID: 31521336 DOI: 10.1016/j.biopsych.2019.07.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 07/01/2019] [Accepted: 07/04/2019] [Indexed: 01/06/2023]
Abstract
BACKGROUND Cognitive deficits such as working memory (WM) impairment are core features of schizophrenia. One candidate marker for the integrity of synaptic neurotransmission necessary for cognitive processes is glutamate. It is frequently postulated that antipsychotic medication possibly alters functional mechanisms in the living brain. We tested in vivo for group differences in activation of the dorsolateral prefrontal cortex (DLPFC) during WM performance and the association with glutamate concentration in DLPFC depending on medication status. METHODS A total of 90 subjects (35 control subjects, 36 medicated patients, and 19 unmedicated patients) contributed magnetic resonance spectroscopy data. We estimated glutamate in left DLPFC. Subjects performed an n-back WM task (2-back vs. 0-back) during functional magnetic resonance imaging, and local activation in left DLPFC was measured. For analysis of association with medication status, we calculated linear regression models including an interaction effect with group. RESULTS Medicated and unmedicated patients with schizophrenia showed impaired performance. We found significantly reduced WM activation in left DLPFC in medicated patients and a trendwise reduction in unmedicated patients as compared with control subjects. We found no group difference in local glutamate concentration. However, we found differential effects of medication status on the association between local glutamate concentration and WM activation in left DLPFC, with a positive association in unmedicated patients but not in medicated patients. CONCLUSIONS We provide evidence that WM-dependent activation is associated with glutamate concentration in unmedicated patients with schizophrenia. Our finding points to putative allostatic changes that affect the functioning of the brain and might be altered through medication.
Collapse
Affiliation(s)
- Jakob Kaminski
- Department of Psychiatry and Psychotherapy Charité Campus Mitte, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany; Berlin Institute of Health, Berlin, Germany.
| | - Tobias Gleich
- Department of Psychiatry and Psychotherapy Charité Campus Mitte, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Yu Fukuda
- Department of Psychiatry and Psychotherapy Charité Campus Mitte, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Teresa Katthagen
- Department of Psychiatry and Psychotherapy Charité Campus Mitte, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Jürgen Gallinat
- University Clinic Hamburg-Eppendorf, Clinic and Policlinic for Psychiatry and Psychotherapy, Hamburg, Germany
| | - Andreas Heinz
- Department of Psychiatry and Psychotherapy Charité Campus Mitte, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Florian Schlagenhauf
- Department of Psychiatry and Psychotherapy Charité Campus Mitte, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany; Berlin Institute of Health, Berlin, Germany; Bernstein Center for Computational Neuroscience, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
32
|
Neurometabolic correlates of 6 and 16 weeks of treatment with risperidone in medication-naive first-episode psychosis patients. Transl Psychiatry 2020; 10:15. [PMID: 32066680 PMCID: PMC7026447 DOI: 10.1038/s41398-020-0700-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 11/25/2019] [Accepted: 12/08/2019] [Indexed: 02/06/2023] Open
Abstract
Antipsychotic medications are the cornerstone of treatment in schizophrenia spectrum disorders. In first-episode psychosis, the recommended time for an antipsychotic medication trial is up to 16 weeks, but the biological correlates of shorter and longer antipsychotic treatment trials in these cohorts remain largely unknown. We enrolled 29 medication-naive first-episode patients (FEP) and 22 matched healthy controls (HC) in this magnetic resonance spectroscopy (MRS) study, examining the levels of combined glutamate and glutamine (commonly referred to as Glx) in the bilateral medial prefrontal cortex (MPFC) with a PRESS sequence (TR/TE = 2000/80 ms) before initiation of antipsychotic treatment, after 6 and 16 weeks of treatment with risperidone. Data were quantified in 18 HC and 20 FEP at baseline, for 19 HC and 15 FEP at week 6, and for 14 HC and 16 FEP at week 16. At baseline, none of the metabolites differed between groups. Metabolite levels did not change after 6 or 16 weeks of treatment in patients. Our data suggest that metabolite levels do not change after 6 or 16 weeks of treatment with risperidone in FEP. It is possible that our choice of sequence parameters and the limited sample size contributed to negative findings reported here. On the other hand, longer follow-up may be needed to detect treatment-related metabolic changes with MRS. In summary, our study adds to the efforts in better understanding glutamatergic neurometabolism in schizophrenia, especially as it relates to antipsychotic exposure.
Collapse
|
33
|
Wenneberg C, Glenthøj BY, Hjorthøj C, Buchardt Zingenberg FJ, Glenthøj LB, Rostrup E, Broberg BV, Nordentoft M. Cerebral glutamate and GABA levels in high-risk of psychosis states: A focused review and meta-analysis of 1H-MRS studies. Schizophr Res 2020; 215:38-48. [PMID: 31784336 DOI: 10.1016/j.schres.2019.10.050] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 10/21/2019] [Accepted: 10/24/2019] [Indexed: 12/24/2022]
Abstract
Disturbances in the brain glutamate and GABA (γ-aminobutyric acid) homeostasis may be markers of transition to psychosis in individuals at high-risk (HR). Knowledge of GABA and glutamate levels in HR stages could give an insight into changes in the neurochemistry underlying psychosis. Studies on glutamate in HR have provided conflicting data, and GABA studies have only recently been initialized. In this meta-analysis, we compared cerebral levels of glutamate and GABA in HR individuals with healthy controls (HC). We searched Medline and Embase for articles published on 1H-MRS studies on glutamate and GABA in HR states until April 9th, 2019. We identified a total of 28 eligible studies, of which eight reported GABA (243 HR, 356 HC) and 26 reported glutamate (299 HR, 279 HC) or Glx (glutamate + glutamine) (584 HR, 632 HC) levels. Sample sizes varied from 6 to 75 for HR and 10 to 184 for HC. Our meta-analysis of 1H-MRS studies on glutamate and GABA in HR states displayed significantly lower (P = 0.0003) levels of thalamic glutamate in HR individuals than in HC and significantly higher (P = 0.001) Glx in the frontal lobe of genetic HR individuals (1st-degree relatives) than in HC. No other significant differences in glutamate and GABA levels were found. Subject numbers in the studies on glutamate as well as GABA levels were generally small and the data conflicting. Our meta-analytical findings highlight the need for larger and more homogeneous studies of glutamate and GABA in high-risk states.
Collapse
Affiliation(s)
- Christina Wenneberg
- Copenhagen Research Center for Mental Health, CORE, Mental Health Center Copenhagen, Copenhagen University Hospital, Gentofte Hospitalsvej 15.4, 2900, Hellerup, Denmark; Center for Neuropsychiatric Schizophrenia Research, CNSR, Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, CINS, Mental Health Centre Glostrup, University of Copenhagen, Ndr. Ringvej 29-67, 2600, Glostrup, Denmark.
| | - Birte Yding Glenthøj
- Center for Neuropsychiatric Schizophrenia Research, CNSR, Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, CINS, Mental Health Centre Glostrup, University of Copenhagen, Ndr. Ringvej 29-67, 2600, Glostrup, Denmark.
| | - Carsten Hjorthøj
- Copenhagen Research Center for Mental Health, CORE, Mental Health Center Copenhagen, Copenhagen University Hospital, Gentofte Hospitalsvej 15.4, 2900, Hellerup, Denmark; University of Copenhagen, Department of Public Health, Section of Epidemiology, Øster Farimagsgade 5, Postboks 2099, 1014, Copenhagen K, Denmark.
| | - Frederik Johan Buchardt Zingenberg
- Copenhagen Research Center for Mental Health, CORE, Mental Health Center Copenhagen, Copenhagen University Hospital, Gentofte Hospitalsvej 15.4, 2900, Hellerup, Denmark.
| | - Louise Birkedal Glenthøj
- Copenhagen Research Center for Mental Health, CORE, Mental Health Center Copenhagen, Copenhagen University Hospital, Gentofte Hospitalsvej 15.4, 2900, Hellerup, Denmark; Center for Neuropsychiatric Schizophrenia Research, CNSR, Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, CINS, Mental Health Centre Glostrup, University of Copenhagen, Ndr. Ringvej 29-67, 2600, Glostrup, Denmark.
| | - Egill Rostrup
- Center for Neuropsychiatric Schizophrenia Research, CNSR, Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, CINS, Mental Health Centre Glostrup, University of Copenhagen, Ndr. Ringvej 29-67, 2600, Glostrup, Denmark.
| | - Brian Villumsen Broberg
- Center for Neuropsychiatric Schizophrenia Research, CNSR, Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, CINS, Mental Health Centre Glostrup, University of Copenhagen, Ndr. Ringvej 29-67, 2600, Glostrup, Denmark.
| | - Merete Nordentoft
- Copenhagen Research Center for Mental Health, CORE, Mental Health Center Copenhagen, Copenhagen University Hospital, Gentofte Hospitalsvej 15.4, 2900, Hellerup, Denmark.
| |
Collapse
|
34
|
Glutamatergic function in a genetic high-risk group for psychosis: A proton magnetic resonance spectroscopy study in individuals with 22q11.2 deletion. Eur Neuropsychopharmacol 2019; 29:1333-1342. [PMID: 31648854 DOI: 10.1016/j.euroneuro.2019.09.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 09/10/2019] [Accepted: 09/13/2019] [Indexed: 11/21/2022]
Abstract
Glutamatergic dysregulation is one of the leading theories regarding the pathoaetiolopy of schizophrenia. Meta-analysis of magnetic resonance spectroscopy studies in schizophrenia shows increased levels of glutamate and glutamine (Glx) in the medial frontal cortex and basal ganglia in clinical high-risk groups for psychosis and increased glutamine levels in the thalamus, but it is unclear if this is also the case in people at genetic high risk for psychosis. The aim of this study was to investigate glutamatergic function in the anterior cingulate cortex, striatum and thalamus in carriers of a genetic variant (22q11.2 deletion) associated with a high risk for psychosis. 53 volunteers (23 22q11.2 deletion carriers and 30 controls) underwent proton magnetic resonance spectroscopy imaging and neuropsychological assessments for prodromal psychotic symptoms, schizotypy, anxiety, depression and FSIQ. We did not find any difference between groups in Glx in the anterior cingulate cortex, striatum or thalamus (Glx: t(50)=-1.26, p = 0.21; U = 251, z = -0.7, p = 0.49; U = 316, z= -0.26, p = 0.79, respectively). No correlation was detected between Glx levels in any region and symptomatology or FSIQ. Our findings indicate that glutamatergic function is not altered in people at genetic high risk of psychosis due to the 22q11.2 deletion, which could suggest that this is not the mechanism underlying psychosis risk in 22q11.2 deletion carriers.
Collapse
|
35
|
Using proton magnetic resonance spectroscopic imaging to study glutamatergic alterations in patients with schizophrenia: A systematic review. Schizophr Res 2019; 210:13-20. [PMID: 31272905 DOI: 10.1016/j.schres.2019.06.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 06/05/2019] [Accepted: 06/16/2019] [Indexed: 12/21/2022]
Abstract
The glutamate hypothesis of schizophrenia posits aberrant glutamatergic activity in patients with schizophrenia. Levels of glutamate and glutamine can be detected and quantified in vivo by proton magnetic resonance spectroscopy. A related technique, proton magnetic resonance spectroscopic imaging (1H-MRSI), is particularly useful as it simultaneously collects multiple spectra, across multiple voxels, from a single acquisition. The primary aim of this study was to review and discuss the use of 1H-MRSI to measure levels of glutamate and glutamine in patients with schizophrenia. Additionally, the advantages and disadvantages of using 1H-MRSI to examine schizophrenia pathophysiology are discussed. A literature search was conducted through Ovid. English language studies utilizing 1H-MRSI to measure glutamate and glutamine in patients with schizophrenia were identified. Six studies met the inclusion criteria. The included studies provide inconclusive support for glutamatergic elevations within frontal brain regions in patients with schizophrenia. The key benefit of employing 1H-MRSI to examine schizophrenia pathophysiology appears to be its broader spatial coverage. Future 1H-MRSI studies utilizing large sample sizes and longitudinal study designs are necessitated to further our understanding of glutamatergic alterations in patients with schizophrenia.
Collapse
|
36
|
Merritt K, Perez-Iglesias R, Sendt KV, Goozee R, Jauhar S, Pepper F, Barker GJ, Glenthøj B, Arango C, Lewis S, Kahn R, Stone J, Howes O, Dazzan P, McGuire P, Egerton A. Remission from antipsychotic treatment in first episode psychosis related to longitudinal changes in brain glutamate. NPJ SCHIZOPHRENIA 2019; 5:12. [PMID: 31371817 PMCID: PMC6672005 DOI: 10.1038/s41537-019-0080-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Accepted: 06/19/2019] [Indexed: 02/04/2023]
Abstract
Neuroimaging studies in schizophrenia have linked elevated glutamate metabolite levels to non-remission following antipsychotic treatment, and also indicate that antipsychotics can reduce glutamate metabolite levels. However, the relationship between symptomatic reduction and change in glutamate during initial antipsychotic treatment is unclear. Here we report proton magnetic resonance spectroscopy (1H-MRS) measurements of Glx and glutamate in the anterior cingulate cortex (ACC) and thalamus in patients with first episode psychosis (n = 23) at clinical presentation, and after 6 weeks and 9 months of treatment with antipsychotic medication. At 9 months, patients were classified into Remission (n = 12) and Non-Remission (n = 11) subgroups. Healthy volunteers (n = 15) were scanned at the same three time-points. In the thalamus, Glx varied over time according to remission status (P = 0.020). This reflected an increase in Glx between 6 weeks and 9 months in the Non-Remission subgroup that was not evident in the Remission subgroup (P = 0.031). In addition, the change in Glx in the thalamus over the 9 months of treatment was positively correlated with the change in the severity of Positive and Negative Syndrome Scale (PANSS) positive, total and general symptoms (P<0.05). There were no significant effects of group or time on glutamate metabolites in the ACC, and no differences between either patient subgroup and healthy volunteers. These data suggest that the nature of the response to antipsychotic medication may be related to the pattern of changes in glutamatergic metabolite levels over the course of treatment. Specifically, longitudinal reductions in thalamic Glx levels following antipsychotic treatment are associated with symptomatic improvement.
Collapse
Affiliation(s)
- Kate Merritt
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, De Crespigny Park, London, SE5 8AF, UK.
| | - Rocio Perez-Iglesias
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, De Crespigny Park, London, SE5 8AF, UK.,CIBERSAM: Centro Investigación Biomédica en Red Salud Mental, Santander, Spain
| | - Kyra-Verena Sendt
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, De Crespigny Park, London, SE5 8AF, UK
| | - Rhianna Goozee
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, De Crespigny Park, London, SE5 8AF, UK
| | - Sameer Jauhar
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, De Crespigny Park, London, SE5 8AF, UK
| | - Fiona Pepper
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, De Crespigny Park, London, SE5 8AF, UK
| | - Gareth J Barker
- Department of Neuroimaging, Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology & Neuroscience, De Crespigny Park, London, SE5 8AF, UK
| | - Birte Glenthøj
- Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, CINS, & Center for Neuropsychiatric Schizophrenia Research, CNSR, Mental Health Center Glostrup, University of Copenhagen, København, Denmark
| | - Celso Arango
- CIBERSAM: Centro Investigación Biomédica en Red Salud Mental, Santander, Spain
| | - Shôn Lewis
- Institute of Brain, Behaviour and Mental Health, Manchester Academic Health Sciences Centre and Manchester Mental Health and Social Care Trust, Manchester, M13 9PL, UK
| | - René Kahn
- Department of Psychiatry, Icahn School of Medicine, New York, USA
| | - James Stone
- Department of Neuroimaging, Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology & Neuroscience, De Crespigny Park, London, SE5 8AF, UK
| | - Oliver Howes
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, De Crespigny Park, London, SE5 8AF, UK
| | - Paola Dazzan
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, De Crespigny Park, London, SE5 8AF, UK
| | - Philip McGuire
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, De Crespigny Park, London, SE5 8AF, UK
| | - Alice Egerton
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, De Crespigny Park, London, SE5 8AF, UK
| |
Collapse
|
37
|
Kraguljac NV, Morgan CJ, Reid MA, White DM, Jindal RD, Sivaraman S, Martinak BK, Lahti AC. A longitudinal magnetic resonance spectroscopy study investigating effects of risperidone in the anterior cingulate cortex and hippocampus in schizophrenia. Schizophr Res 2019; 210:239-244. [PMID: 30630705 PMCID: PMC7881837 DOI: 10.1016/j.schres.2018.12.028] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 12/18/2018] [Accepted: 12/19/2018] [Indexed: 12/30/2022]
Abstract
Magnetic Resonance Spectroscopy is a popular approach to probe brain chemistry in schizophrenia (SZ), but no consensus exists as to the extent of alterations. This may be attributable to differential effects of populations studied, brain regions examined, or antipsychotic medication effects. Here, we measured neurometabolites in the anterior cingulate cortex (ACC) and hippocampus, two structurally dissimilar brain regions implicated in the SZ pathophysiology. We enrolled 61 SZ with the goal to scan them before and after six weeks of treatment with risperidone. We also scanned 31 matched healthy controls twice, six weeks apart. Using mixed effect repeated measures linear models to examine the effect of group and time on metabolite levels in each voxel, we report an increase in hippocampal glutamate + glutamine (Glx) in SZ compared to controls (p = 0.043), but no effect of antipsychotic medication (p = 0.330). In the ACC, we did not find metabolite alterations or antipsychotic medication related changes after six weeks of treatment with risperidone. The coefficients for the discriminant function (differentiating SZ from HC) in the ACC were greatest for NAA (-0.83), and in the hippocampus for Glx (0.76), the same metabolites were associated with greater treatment response in patients at trend level. Taken together, our data extends the existing literature by demonstrating regionally distinct metabolite alterations in the same patient group and suggests that antipsychotic medications may have limited effects on metabolite levels in these regions.
Collapse
Affiliation(s)
- Nina V. Kraguljac
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham
| | | | - Meredith A. Reid
- MRI Research Center, Department of Electrical and Computer Engineering, Auburn University
| | - David M. White
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham
| | - Ripu D. Jindal
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham,Department of Neurology, Birmingham VA Medical Center
| | - Soumya Sivaraman
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham
| | - Bridgette K. Martinak
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham
| | - Adrienne C. Lahti
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham
| |
Collapse
|
38
|
Borgan FR, Jauhar S, McCutcheon RA, Pepper FS, Rogdaki M, Lythgoe DJ, Howes OD. Glutamate levels in the anterior cingulate cortex in un-medicated first episode psychosis: a proton magnetic resonance spectroscopy study. Sci Rep 2019; 9:8685. [PMID: 31266965 PMCID: PMC6606579 DOI: 10.1038/s41598-019-45018-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 05/22/2019] [Indexed: 01/09/2023] Open
Abstract
Converging lines of evidence suggest that glutamatergic dysfunction may contribute to the pathophysiology of first episode psychosis. We investigated whether first episode psychosis patients free from all pharmacological treatments and illicit substances show cortical glutamatergic alterations. One-hundred and eleven volunteers including 65 healthy volunteers and 46 first episode psychosis patients free from all pharmacological treatments (28 drug naïve) underwent a proton magnetic resonance spectroscopy scan measuring glutamate levels in the bilateral anterior cingulate cortex. Symptom severity was measured using the Positive and Negative Syndrome Scale (PANSS) and cognition was measured using the Wechsler Adult Intelligence Scale (WAIS) digit symbol test. There were no differences in glutamate levels between patients and controls. These findings remained unchanged when adjusting for the effects of age, sex and ethnicity or when restricting the analyses to patients who were both medication naïve to all pharmacological treatments and illicit substances. Whilst these findings do not preclude glutamatergic alterations in psychosis, methodological advances are needed for us to investigate whether patients show alterations in other aspects of glutamate function, such as pre-synaptic glutamate or release.
Collapse
Affiliation(s)
- Faith R Borgan
- Psychosis Studies Department, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, England. .,Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, W12 0NN, UK.
| | - Sameer Jauhar
- Psychosis Studies Department, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, England
| | - Robert A McCutcheon
- Psychosis Studies Department, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, England.,Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, W12 0NN, UK
| | - Fiona S Pepper
- Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, England
| | - Maria Rogdaki
- Psychosis Studies Department, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, England.,Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, W12 0NN, UK
| | - David J Lythgoe
- Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, England
| | - Oliver D Howes
- Psychosis Studies Department, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, England. .,Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, W12 0NN, UK.
| |
Collapse
|
39
|
Pillinger T, Rogdaki M, McCutcheon RA, Hathway P, Egerton A, Howes OD. Altered glutamatergic response and functional connectivity in treatment resistant schizophrenia: the effect of riluzole and therapeutic implications. Psychopharmacology (Berl) 2019; 236:1985-1997. [PMID: 30820633 PMCID: PMC6642056 DOI: 10.1007/s00213-019-5188-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 01/30/2019] [Indexed: 01/09/2023]
Abstract
RATIONALE Anterior cingulate cortex (ACC) glutamatergic abnormalities are reported in treatment-resistant schizophrenia (TRS) and implicated in functional dysconnectivity and psychopathology. Preclinical evidence indicates riluzole reduces synaptic glutamate. However, it is unknown whether riluzole can modulate glutamate metabolite levels and associated functional connectivity in TRS. OBJECTIVES To examine the relationship between glutamatergic function and cortical connectivity and determine if riluzole can modulate glutamate metabolite levels and cortical functional connectivity in TRS. METHODS Nineteen TRS patients and 18 healthy volunteers (HV) underwent magnetic resonance imaging consisting of MR spectroscopy measuring ACC glutamate plus glutamine (Glx), fMRI measuring resting ACC-functional connectivity, and arterial spin labelling measuring regional cerebral blood flow (rCBF), and clinical measures. They then received 50 mg riluzole twice daily for 2 days when imaging was repeated. RESULTS Baseline (pre-riluzole) Glx levels were correlated directly with negative symptom severity (r = 0.49; p = 0.03) and inversely with verbal learning in TRS (r = - 0.63; p = 0.002), but not HV (r = - 0.24; p = 0.41). Connectivity between the ACC and anterior prefrontal cortex (aPFC) was correlated with verbal learning in TRS (r = 0.49; p = 0.04), but not HV (r = 0.28; p = 0.33). There was a significant group × time interaction effect on Glx levels (p < 0.05) and on ACC connectivity to the aPFC (p < 0.05, FWE-corrected). Riluzole decreased Glx and increased ACC-aPFC connectivity in TRS relative to HV. Change in Glx correlated inversely with change in ACC-aPFC connectivity in TRS (r = - 0.52; p = 0.02) but not HV (r = 0.01; p = 0.98). Riluzole did not alter rCBF (p > 0.05), indicating absence of a non-specific blood flow effect. CONCLUSION Results indicate glutamatergic function and cortical connectivity are linked to symptoms and cognitive measures and that it is possible to pharmacologically modulate them in TRS.
Collapse
Affiliation(s)
- Toby Pillinger
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, England
- Medical Research Council London Institute of Medical Sciences, London, England
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, England
| | - Maria Rogdaki
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, England
- Medical Research Council London Institute of Medical Sciences, London, England
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, England
| | - Robert A. McCutcheon
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, England
- Medical Research Council London Institute of Medical Sciences, London, England
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, England
| | - Pamela Hathway
- Department of Electrical and Electronic Engineering, Imperial College London, London, England
| | - Alice Egerton
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, England
| | - Oliver D. Howes
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, England
- Medical Research Council London Institute of Medical Sciences, London, England
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, England
| |
Collapse
|
40
|
Fernandez G, Kuruvilla S, Hines CDG, Poignant F, Marr J, Forest T, Briscoe R. Brain findings associated with risperidone in rhesus monkeys: magnetic resonance imaging and pathology perspectives. J Toxicol Pathol 2019; 32:233-243. [PMID: 31719750 PMCID: PMC6831502 DOI: 10.1293/tox.2019-0004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Accepted: 05/13/2019] [Indexed: 12/19/2022] Open
Abstract
Brain changes associated with risperidone, a dopamine-2/serotonin-2 receptor antagonist, have been documented in rats and humans, but not in nonhuman primates. This study characterized brain changes associated with risperidone in nonhuman primates. Rhesus monkeys were orally administered risperidone in a dose-escalation paradigm up to a maximum tolerated dose of 0.5 mg/kg/day for 3 weeks, or 3 months followed by a 3-month recovery period. Transient and fully reversible neurological signs consistent with risperidone pharmacology were observed. The results of a magnetic resonance imaging evaluation after 3 months of treatment and at the end of the 3-month recovery period showed no meaningful changes in the brain. There were no risperidone-related brain weight changes or gross findings. Histomorphological evaluation of brain sections stained with hematoxylin and eosin, ionized calcium binding adaptor molecule 1 (Iba1), and luxol fast blue/cresyl violet double staining showed no notable differences between control and risperidone groups. However, evaluation of the brain after glial fibrillary acidic protein (GFAP) immunohistochemical staining revealed increased staining in the cell bodies and processes of astrocytes in the putamen without apparent alterations in numbers or distribution. The increase in GFAP staining was present after 3 weeks and 3 months of treatment, but no increase in staining was observed after the 3-month recovery period, demonstrating the reversibility of this finding. The reversible increase in GFAP expression was likely an adaptive, non-adverse response of astrocytes, associated with the pharmacology of risperidone. These observations are valuable considerations in the nonclinical risk assessment of new drug candidates for psychiatric disorders.
Collapse
Affiliation(s)
- Guillermo Fernandez
- Safety Assessment and Laboratory Animal Resources, Merck & Co., Inc., 770 Sumneytown Pike, West Point, PA 19486, USA
| | - Sabu Kuruvilla
- Safety Assessment and Laboratory Animal Resources, Merck & Co., Inc., 770 Sumneytown Pike, West Point, PA 19486, USA
| | - Catherine D G Hines
- Translational Imaging Biomarkers, Merck & Co., Inc., 770 Sumneytown Pike, West Point, PA 19486, USA
| | - Frédéric Poignant
- Safety Assessment and Laboratory Animal Resources, Laboratoires Merck Sharp & Dohme-Chibret, Route de Marsat - Riom, 63963 Clermont-Ferrand cedex 9, France
| | - James Marr
- Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Inc., 770 Sumneytown Pike, West Point, PA 19486, USA
| | - Thomas Forest
- Safety Assessment and Laboratory Animal Resources, Merck & Co., Inc., 770 Sumneytown Pike, West Point, PA 19486, USA
| | - Richard Briscoe
- Safety Assessment and Laboratory Animal Resources, Merck & Co., Inc., 770 Sumneytown Pike, West Point, PA 19486, USA
| |
Collapse
|
41
|
Vita A, Minelli A, Barlati S, Deste G, Giacopuzzi E, Valsecchi P, Turrina C, Gennarelli M. Treatment-Resistant Schizophrenia: Genetic and Neuroimaging Correlates. Front Pharmacol 2019; 10:402. [PMID: 31040787 PMCID: PMC6476957 DOI: 10.3389/fphar.2019.00402] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Accepted: 04/01/2019] [Indexed: 12/11/2022] Open
Abstract
Schizophrenia is a severe neuropsychiatric disorder that affects approximately 0.5–1% of the population. Response to antipsychotic therapy is highly variable, and it is not currently possible to predict those patients who will or will not respond to antipsychotic medication. Furthermore, a high percentage of patients, approximately 30%, are classified as treatment-resistant (treatment-resistant schizophrenia; TRS). TRS is defined as a non-response to at least two trials of antipsychotic medication of adequate dose and duration. These patients are usually treated with clozapine, the only evidence-based pharmacotherapy for TRS. However, clozapine is associated with severe adverse events. For these reasons, there is an increasing interest to identify better targets for drug development of new compounds and to establish better biomarkers for existing medications. The ability of antipsychotics to improve psychotic symptoms is dependent on their antagonist and reverse agonist activities at different neuroreceptors, and some genetic association studies of TRS have focused on different pharmacodynamic factors. Some genetic studies have shown an association between antipsychotic response or TRS and neurodevelopment candidate genes, antipsychotic mechanisms of action (such as dopaminergic, serotonergic, GABAergic, and glutamatergic) or pharmacokinetic factors (i.e., differences in the cytochrome families). Moreover, there is a growing body of literature on the structural and functional neuroimaging research into TRS. Neuroimaging studies can help to uncover the underlying neurobiological reasons for such resistance and identify resistant patients earlier. Studies examining the neuropharmacological mechanisms of antipsychotics, including clozapine, can help to improve our knowledge of their action on the central nervous system, with further implications for the discovery of biomarkers and the development of new treatments. The identification of the underlying mechanisms of TRS is a major challenge for developing personalized medicine in the psychiatric field for schizophrenia treatment. The main goal of precision medicine is to use genetic and brain-imaging information to improve the safety, effectiveness, and health outcomes of patients via more efficiently targeted risk stratification, prevention, and tailored medication and treatment management approaches. The aim of this review is to summarize the state of art of pharmacogenetic, pharmacogenomic and neuroimaging studies in TRS.
Collapse
Affiliation(s)
- Antonio Vita
- Department of Mental Health and Addiction Services, ASST Spedali Civili, Brescia, Italy.,Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Alessandra Minelli
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Stefano Barlati
- Department of Mental Health and Addiction Services, ASST Spedali Civili, Brescia, Italy.,Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Giacomo Deste
- Department of Mental Health and Addiction Services, ASST Spedali Civili, Brescia, Italy
| | - Edoardo Giacopuzzi
- Genetic Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Paolo Valsecchi
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Cesare Turrina
- Department of Mental Health and Addiction Services, ASST Spedali Civili, Brescia, Italy.,Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Massimo Gennarelli
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.,Genetic Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| |
Collapse
|
42
|
Iwata Y, Nakajima S, Plitman E, Caravaggio F, Kim J, Shah P, Mar W, Chavez S, De Luca V, Mimura M, Remington G, Gerretsen P, Graff-Guerrero A. Glutamatergic Neurometabolite Levels in Patients With Ultra-Treatment-Resistant Schizophrenia: A Cross-Sectional 3T Proton Magnetic Resonance Spectroscopy Study. Biol Psychiatry 2019; 85:596-605. [PMID: 30389132 DOI: 10.1016/j.biopsych.2018.09.009] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 09/13/2018] [Accepted: 09/13/2018] [Indexed: 12/21/2022]
Abstract
BACKGROUND In terms of antipsychotic treatment response, patients with schizophrenia can be classified into three groups: 1) treatment resistant to both non-clozapine (non-CLZ) antipsychotics and CLZ (ultra-treatment-resistant schizophrenia [URS]), 2) treatment resistant to non-CLZ antipsychotics but CLZ-responsive schizophrenia [non-URS]), and 3) responsive to first-line antipsychotics (non-treatment-resistant schizophrenia). This study aimed to compare glutamatergic neurometabolite levels among these three patient groups and healthy control subjects using proton magnetic resonance spectroscopy. METHODS Glutamate and glutamate+glutamine levels were assessed in the caudate, the dorsal anterior cingulate cortex (dACC), and the dorsolateral prefrontal cortex using 3T proton magnetic resonance spectroscopy (point-resolved spectroscopy, echo time = 35 ms). Glutamatergic neurometabolite levels were compared between the groups. RESULTS A total of 100 participants were included, consisting of 26 patients with URS, 27 patients with non-URS, 21 patients with non-treatment-resistant schizophrenia, and 26 healthy control subjects. Group differences were detected in ACC glutamate+glutamine levels (F3,96 = 2.93, p = .038); patients with URS showed higher dACC glutamate+glutamine levels than healthy control subjects (p = .038). There were no group differences in the caudate or dorsolateral prefrontal cortex. CONCLUSIONS Taken together with previous studies that demonstrated higher ACC glutamate levels in patients with treatment-resistant schizophrenia, this study suggests that higher levels of ACC glutamatergic metabolites may be among the shared biological characteristics of treatment resistance to antipsychotics, including CLZ.
Collapse
Affiliation(s)
- Yusuke Iwata
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Geriatric Mental Health Division, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada; Department of Neuropsychiatry, School of Medicine, Keio University, Tokyo, Japan
| | - Shinichiro Nakajima
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Neuropsychiatry, School of Medicine, Keio University, Tokyo, Japan
| | - Eric Plitman
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Fernando Caravaggio
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Julia Kim
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Parita Shah
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Wanna Mar
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Sofia Chavez
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Vincenzo De Luca
- Geriatric Mental Health Division, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Campbell Institute Research Program, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Masaru Mimura
- Department of Neuropsychiatry, School of Medicine, Keio University, Tokyo, Japan
| | - Gary Remington
- Schizophrenia Division, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Campbell Institute Research Program, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Philip Gerretsen
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Geriatric Mental Health Division, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Campbell Institute Research Program, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Ariel Graff-Guerrero
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Geriatric Mental Health Division, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Campbell Institute Research Program, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
43
|
Chouinard VA. Glutamatergic System and Neuroimaging Studies of Treatment-Resistant Schizophrenia. Biol Psychiatry 2019; 85:e31-e32. [PMID: 30871691 DOI: 10.1016/j.biopsych.2019.02.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 02/01/2019] [Accepted: 02/04/2019] [Indexed: 12/31/2022]
Affiliation(s)
- Virginie-Anne Chouinard
- Psychotic Disorders Division, McLean Hospital, Belmont, Massachusetts; Department of Psychiatry, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
44
|
Legind CS, Broberg BV, Mandl RCW, Brouwer R, Anhøj SJ, Hilker R, Jensen MH, McGuire P, Pol HH, Fagerlund B, Rostrup E, Glenthøj BY. Heritability of cerebral glutamate levels and their association with schizophrenia spectrum disorders: a 1[H]-spectroscopy twin study. Neuropsychopharmacology 2019; 44:581-589. [PMID: 30301944 PMCID: PMC6333786 DOI: 10.1038/s41386-018-0236-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 09/03/2018] [Accepted: 10/03/2018] [Indexed: 01/10/2023]
Abstract
Research findings implicate cerebral glutamate in the pathophysiology of schizophrenia, including genetic studies reporting associations with glutamatergic neurotransmission. The extent to which aberrant glutamate levels can be explained by genetic factors is unknown, and if glutamate can serve as a marker of genetic susceptibility for schizophrenia remains to be established. We investigated the heritability of cerebral glutamate levels and whether a potential association with schizophrenia spectrum disorders could be explained by genetic factors. Twenty-three monozygotic (MZ) and 20 dizygotic (DZ) proband pairs con- or discordant for schizophrenia spectrum disorders, along with healthy control pairs (MZ = 28, DZ = 18) were recruited via the National Danish Twin Register and the Psychiatric Central Register (17 additional twins were scanned without their siblings). Glutamate levels in the left thalamus and the anterior cingulate cortex (ACC) were measured using 1[H]-magnetic resonance spectroscopy at 3 Tesla and analyzed by structural equation modeling. Glutamate levels in the left thalamus were heritable and positively correlated with liability for schizophrenia spectrum disorders (phenotypic correlation, 0.16, [0.02-0.29]; p = 0.010). The correlation was explained by common genes influencing both the levels of glutamate and liability for schizophrenia spectrum disorders. In the ACC, glutamate and glx levels were heritable, but not correlated to disease liability. Increases in thalamic glutamate levels found in schizophrenia spectrum disorders are explained by genetic influences related to the disease, and as such the measure could be a potential marker of genetic susceptibility, useful in early detection and stratification of patients with psychosis.
Collapse
Affiliation(s)
- Christian Stefan Legind
- Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, CINS, & Center for Neuropsychiatric Schizophrenia Research, CNSR, Mental Health Center Glostrup, University of Copenhagen, Copenhagen, Denmark.
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Brian Villumsen Broberg
- Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, CINS, & Center for Neuropsychiatric Schizophrenia Research, CNSR, Mental Health Center Glostrup, University of Copenhagen, Copenhagen, Denmark
| | - René Christiaan William Mandl
- Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, CINS, & Center for Neuropsychiatric Schizophrenia Research, CNSR, Mental Health Center Glostrup, University of Copenhagen, Copenhagen, Denmark
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Rachel Brouwer
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Simon Jesper Anhøj
- Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, CINS, & Center for Neuropsychiatric Schizophrenia Research, CNSR, Mental Health Center Glostrup, University of Copenhagen, Copenhagen, Denmark
| | - Rikke Hilker
- Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, CINS, & Center for Neuropsychiatric Schizophrenia Research, CNSR, Mental Health Center Glostrup, University of Copenhagen, Copenhagen, Denmark
| | - Maria Høj Jensen
- Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, CINS, & Center for Neuropsychiatric Schizophrenia Research, CNSR, Mental Health Center Glostrup, University of Copenhagen, Copenhagen, Denmark
| | - Philip McGuire
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience (IoPPN), King's College London, De Crespigny Park, London, UK
| | - Hilleke Hulshoff Pol
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Birgitte Fagerlund
- Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, CINS, & Center for Neuropsychiatric Schizophrenia Research, CNSR, Mental Health Center Glostrup, University of Copenhagen, Copenhagen, Denmark
- Department of Psychology, University of Copenhagen, Copenhagen, Denmark
| | - Egill Rostrup
- Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, CINS, & Center for Neuropsychiatric Schizophrenia Research, CNSR, Mental Health Center Glostrup, University of Copenhagen, Copenhagen, Denmark
- Functional Imaging Unit, Department of Clinical Physiology, Nuclear Medicine and PET, Rigshospitalet - Glostrup, Copenhagen, Denmark
| | - Birte Yding Glenthøj
- Center for Clinical Intervention and Neuropsychiatric Schizophrenia Research, CINS, & Center for Neuropsychiatric Schizophrenia Research, CNSR, Mental Health Center Glostrup, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
45
|
Cadena EJ, White DM, Kraguljac NV, Reid MA, Maximo JO, Nelson EA, Gawronski BA, Lahti AC. A Longitudinal Multimodal Neuroimaging Study to Examine Relationships Between Resting State Glutamate and Task Related BOLD Response in Schizophrenia. Front Psychiatry 2018; 9:632. [PMID: 30555359 PMCID: PMC6281980 DOI: 10.3389/fpsyt.2018.00632] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 11/07/2018] [Indexed: 01/10/2023] Open
Abstract
Previous studies have observed impairments in both brain function and neurometabolite levels in schizophrenia. In this study, we investigated the relationship between brain activity and neurochemistry in off-medication patients with schizophrenia and if this relationship is altered following antipsychotic medication by combining proton magnetic resonance spectroscopy (1H-MRS) with functional magnetic resonance imaging (fMRI). We used single voxel MRS acquired in the bilateral dorsal anterior cingulate cortex (ACC) and fMRI during performance of a Stroop color-naming task in 22 patients with schizophrenia (SZ), initially off-medication and after a 6-week course of risperidone, and 20 matched healthy controls (HC) twice, 6 weeks apart. We observed a significant decrease in ACC glutamate + glutamine (Glx)/Creatine (Cr) levels in medicated SZ patients compared to HC but not compared to their off-medication baseline. In off-medication SZ, the relationship between ACC Glx/Cr levels and the blood oxygen level-dependent (BOLD) response in regions of the salience network (SN) and posterior default mode network (DMN) was opposite than of HC. After 6 weeks, the relationship between Glx and the BOLD response was still opposite between the groups; however for both groups the direction of the relationship changed from baseline to week 6. These results suggest a mechanism whereby alterations in the relationship between cortical glutamate and BOLD response is disrupting the modulation of major neural networks subserving cognitive processes, potentially affecting cognition. While these relationships appear to normalize with treatment in patients, the interpretations of the results are confounded by significant group differences in Glx levels, as well as the variability of the relationship between Glx and BOLD response in HC over time, which may be driven by factors including habituation to task or scanner environment.
Collapse
Affiliation(s)
- Elyse J. Cadena
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - David M. White
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Nina V. Kraguljac
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Meredith A. Reid
- Magnetic Resonance Imaging Research Center, Auburn University at Birmingham, Birmingham, AL, United States
| | - Jose O. Maximo
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Eric A. Nelson
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Brian A. Gawronski
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Adrienne C. Lahti
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
46
|
Response to initial antipsychotic treatment in first episode psychosis is related to anterior cingulate glutamate levels: a multicentre 1H-MRS study (OPTiMiSE). Mol Psychiatry 2018; 23:2145-2155. [PMID: 29880882 DOI: 10.1038/s41380-018-0082-9] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 03/15/2018] [Accepted: 04/04/2018] [Indexed: 12/19/2022]
Abstract
Conventional antipsychotic medication is ineffective in around a third of patients with schizophrenia, and the nature of the therapeutic response is unpredictable. We investigated whether response to antipsychotics is related to brain glutamate levels prior to treatment. Proton magnetic resonance spectroscopy was used to measure glutamate levels (Glu/Cr) in the anterior cingulate cortex (ACC) and in the thalamus in antipsychotic-naive or minimally medicated patients with first episode psychosis (FEP, n = 71) and healthy volunteers (n = 60), at three sites. Following scanning, patients were treated with amisulpride for 4 weeks (n = 65), then 1H-MRS was repeated (n = 46). Remission status was defined in terms of Positive and Negative Syndrome Scale for Schizophrenia (PANSS) scores. Higher levels of Glu/Cr in the ACC were associated with more severe symptoms at presentation and a lower likelihood of being in remission at 4 weeks (P < 0.05). There were longitudinal reductions in Glu/Cr in both the ACC and thalamus over the treatment period (P < 0.05), but these changes were not associated with the therapeutic response. There were no differences in baseline Glu/Cr between patients and controls. These results extend previous evidence linking higher levels of ACC glutamate with a poor antipsychotic response by showing that the association is evident before the initiation of treatment.
Collapse
|
47
|
Overbeek G, Gawne TJ, Reid MA, Salibi N, Kraguljac NV, White DM, Lahti AC. Relationship Between Cortical Excitation and Inhibition and Task-Induced Activation and Deactivation: A Combined Magnetic Resonance Spectroscopy and Functional Magnetic Resonance Imaging Study at 7T in First-Episode Psychosis. BIOLOGICAL PSYCHIATRY: COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2018; 4:121-130. [PMID: 30470582 DOI: 10.1016/j.bpsc.2018.10.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 09/18/2018] [Accepted: 10/07/2018] [Indexed: 12/29/2022]
Abstract
BACKGROUND Schizophrenia is thought to be a disorder of brain dysconnectivity. An imbalance between cortical excitation/inhibition is also implicated, but the link between these abnormalities remains unclear. The present study used magnetic resonance spectroscopy and functional magnetic resonance imaging at 7T to investigate how measurements of glutamate and gamma-aminobutyric acid (GABA) relate to the blood oxygen level-dependent (BOLD) response during a cognitive task, and how these relationships are altered in schizophrenia. METHODS Usable functional magnetic resonance imaging data from 17 first-episode psychosis (FEP) patients (4 women, 13 men) and 21 matched healthy control subjects (HCs) (5 women, 16 men) were acquired during a Stroop task. Within- and between-group comparisons of the BOLD response were performed. Neurometabolite levels were measured in the dorsal anterior cingulate cortex. Two multiple regressions investigated how glutamate, glutamine, and GABA related to the BOLD response in HCs and FEP patients separately. A third investigated between-group differences in the relationships between the BOLD response and each of these neurometabolites. RESULTS Compared with HCs, FEP patients showed an increased BOLD response within regions of the executive and default mode networks. In FEP patients, the relationship between anterior cingulate cortex glutamate levels and the BOLD response in regions of the posterior default mode network was opposite to that of HCs. In FEP patients but not HCs, anterior cingulate cortex GABA levels correlated with the local BOLD response and with the Stroop reaction time. CONCLUSION These results suggest a mechanism whereby alterations in the relationship between cortical glutamate/GABA and BOLD response is disrupting the dynamic of major neural networks, possibly affecting cognition.
Collapse
Affiliation(s)
- Gregory Overbeek
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Timothy J Gawne
- Department of Vision Sciences, University of Alabama at Birmingham, Birmingham, Alabama
| | - Meredith A Reid
- Magnetic Resonance Imaging Research Center, Auburn University, Auburn, Alabama
| | - Nouha Salibi
- Siemens Healthineers MR R&D, Malvern, Pennsylvania
| | - Nina V Kraguljac
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, Alabama
| | - David M White
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Adrienne C Lahti
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, Alabama.
| |
Collapse
|
48
|
Jauhar S, McCutcheon R, Borgan F, Veronese M, Nour M, Pepper F, Rogdaki M, Stone J, Egerton A, Turkheimer F, McGuire P, Howes OD. The relationship between cortical glutamate and striatal dopamine in first-episode psychosis: a cross-sectional multimodal PET and magnetic resonance spectroscopy imaging study. Lancet Psychiatry 2018; 5:816-823. [PMID: 30236864 PMCID: PMC6162342 DOI: 10.1016/s2215-0366(18)30268-2] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Revised: 07/02/2018] [Accepted: 07/05/2018] [Indexed: 12/25/2022]
Abstract
BACKGROUND The pathophysiology of psychosis is incompletely understood. Disruption in cortical glutamatergic signalling causing aberrant striatal dopamine synthesis capacity is a proposed model for psychosis, but has not been tested in vivo. We therefore aimed to test the relationship between cortical glutamate concentrations and striatal dopamine synthesis capacity, and psychotic symptoms. METHODS In this cross-sectional multimodal imaging study, 28 individuals with first-episode psychosis and 28 healthy controls underwent 18F-DOPA PET (measuring striatal dopamine synthesis capacity), and proton magnetic resonance spectroscopy (measuring anterior cingulate cortex glutamate concentrations). Participants were recruited from first-episode psychosis services in London, UK and were required to be in the first episode of a psychotic illness, with no previous illness or treatment episodes. Exclusion criteria for all participants were: history of substantial head trauma, dependence on illicit substances, medical comorbidity (other than minor illnesses), and contraindications to scanning (such as pregnancy). Symptoms were measured using the Positive and Negative Syndrome Scale. The primary endpoint was the relationship between anterior cingulate cortex glutamate concentrations and striatal dopamine synthesis capacity in individuals with their first episode of psychosis as shown by imaging, examined by linear regression. Linear regression was used to examine relationships between measures. FINDINGS Glutamate concentrations showed a significant inverse relationship with striatal dopamine synthesis capacity in patients with psychosis (R2=0·16, p=0·03, β -1·71 × 10-4, SE 0·76 × 10-4). This relationship remained significant after the addition of age, gender, ethnicity, and medication status to the model (p=0·015). In healthy controls, there was no significant relationship between dopamine and glutamate measures (R2=0·04, p=0·39). Positive and Negative Syndrome Scale positive psychotic symptoms were positively associated with striatal dopamine synthesis capacity (R2=0·14, p=0·046, β 2546, SE 1217) and showed an inverse relationship with anterior cingulate glutamate concentrations (R2=0·16, p=0·03, β -1·71 × 10-4, SE 7·63 × 10-5). No relationships were seen with negative symptoms (positive symptoms, mean [SD] -18·4 (6·6) negative symptoms, mean [SD] -15·4 [6·1]). INTERPRETATION These observations are consistent with the hypothesis that cortical glutamate dysfunction is related to subcortical dopamine synthesis capacity and psychosis. Although the precise mechanistic relationship between cortical glutamate and dopamine in vivo remains unclear, our findings support further studies to test the effect of modulating cortical glutamate in the treatment of psychosis. FUNDING Medical Research Council, Wellcome Trust, Biomedical Research Council, South London and Maudsley NHS Foundation Trust, JMAS Sim Fellowship, Royal College of Physicians (Edinburgh) (SJ).
Collapse
Affiliation(s)
- Sameer Jauhar
- Department of Psychological Medicine, King's College, London, UK; Early Intervention Psychosis Clinical Academic Group, South London and Maudsley NHS Trust, London, UK
| | | | - Faith Borgan
- Department of Psychosis Studies, King's College, London, UK
| | - Mattia Veronese
- Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College, London, UK
| | - Matthew Nour
- Department of Psychosis Studies, King's College, London, UK
| | - Fiona Pepper
- Department of Psychosis Studies, King's College, London, UK
| | - M Rogdaki
- Department of Psychosis Studies, King's College, London, UK
| | - James Stone
- Department of Psychological Medicine, King's College, London, UK; Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College, London, UK
| | - Alice Egerton
- Department of Psychosis Studies, King's College, London, UK
| | - Frederico Turkheimer
- Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College, London, UK
| | - Philip McGuire
- Department of Psychosis Studies, King's College, London, UK; Early Intervention Psychosis Clinical Academic Group, South London and Maudsley NHS Trust, London, UK
| | - Oliver D Howes
- Department of Psychosis Studies, King's College, London, UK; Psychiatric Imaging Group MRC London Institute of Medical Sciences, Hammersmith Hospital, London, UK; Institute of Clinical Sciences, Faculty of Medicine, Imperial College, Hammersmith Hospital, London, UK.
| |
Collapse
|
49
|
McQueen G, Lally J, Collier T, Zelaya F, Lythgoe DJ, Barker GJ, Stone JM, McGuire P, MacCabe JH, Egerton A. Effects of N-acetylcysteine on brain glutamate levels and resting perfusion in schizophrenia. Psychopharmacology (Berl) 2018; 235:3045-3054. [PMID: 30141055 PMCID: PMC6182588 DOI: 10.1007/s00213-018-4997-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 08/06/2018] [Indexed: 01/16/2023]
Abstract
RATIONALE N-Acetylcysteine (NAC) is currently under investigation as an adjunctive treatment for schizophrenia. The therapeutic potential of NAC may involve modulation of brain glutamate function, but its effects on brain glutamate levels in schizophrenia have not been evaluated. OBJECTIVES The aim of this study was to examine whether a single dose of NAC can alter brain glutamate levels. A secondary aim was to characterise its effects on regional brain perfusion. METHODS In a double-blind placebo-controlled crossover study, 19 patients with a diagnosis of schizophrenia underwent two MRI scans, following oral administration of 2400 mg NAC or matching placebo. Proton magnetic resonance spectroscopy was used to investigate the effect of NAC on glutamate and Glx (glutamate plus glutamine) levels scaled to creatine (Cr) in the anterior cingulate cortex (ACC) and in the right caudate nucleus. Pulsed continuous arterial spin labelling was used to assess the effects of NAC on resting cerebral blood flow (rCBF) in the same regions. RESULTS Relative to the placebo condition, the NAC condition was associated with lower levels of Glx/Cr, in the ACC (P < 0.05), but not in the caudate nucleus. There were no significant differences in CBF in the NAC compared to placebo condition. CONCLUSIONS These data provide preliminary evidence that NAC can modulate ACC glutamate in patients with schizophrenia. In contrast, physiological effects of NAC on the brain were not detectable as between session changes in rCBF. Future studies assessing the effects of a course of treatment with NAC on glutamate metabolites in schizophrenia are indicated.
Collapse
Affiliation(s)
- Grant McQueen
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London, UK.
| | - John Lally
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London, UK
- Department of Psychiatry, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Tracy Collier
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London, UK
| | - Fernando Zelaya
- Department of Neuroimaging, Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology & Neuroimaging, King's College London, De Crespigny Park, London, UK
| | - David J Lythgoe
- Department of Neuroimaging, Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology & Neuroimaging, King's College London, De Crespigny Park, London, UK
| | - Gareth J Barker
- Department of Neuroimaging, Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology & Neuroimaging, King's College London, De Crespigny Park, London, UK
| | - James M Stone
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London, UK
- Experimental Medicine, Hammersmith Hospital, Imperial College London, London, UK
| | - Philip McGuire
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London, UK
| | - James H MacCabe
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London, UK
| | - Alice Egerton
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London, UK
| |
Collapse
|
50
|
Modinos G, Egerton A, McLaughlin A, McMullen K, Kumari V, Lythgoe DJ, Barker GJ, Aleman A, Williams SCR. Neuroanatomical changes in people with high schizotypy: relationship to glutamate levels. Psychol Med 2018; 48:1880-1889. [PMID: 29198207 PMCID: PMC5884418 DOI: 10.1017/s0033291717003403] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
BACKGROUND Cortical glutamatergic dysfunction is thought to be fundamental for psychosis development, and may lead to structural degeneration through excitotoxicity. Glutamate levels have been related to gray matter volume (GMV) alterations in people at ultra-high risk of psychosis, and we previously reported GMV changes in individuals with high schizotypy (HS), which refers to the expression of schizophrenia-like characteristics in healthy people. This study sought to examine whether GMV changes in HS subjects are related to glutamate levels. METHODS We selected 22 healthy subjects with HS and 23 healthy subjects with low schizotypy (LS) based on their rating on a self-report questionnaire for psychotic-like experiences. Glutamate levels were measured in the bilateral anterior cingulate cortex (ACC) using proton magnetic resonance spectroscopy, and GMV was assessed using voxel-based morphometry. RESULTS Subjects with HS showed GMV decreases in the rolandic operculum/superior temporal gyrus (pFWE = 0.045). Significant increases in GMV were also detected in HS, in the precuneus (pFWE = 0.043), thereby replicating our previous finding in a separate cohort, as well as in the ACC (pFWE = 0.041). While the HS and LS groups did not differ in ACC glutamate levels, in HS subjects ACC glutamate was negatively correlated with ACC GMV (pFWE = 0.026). Such association was absent in LS. CONCLUSIONS Our study shows that GMV findings in schizotypy are related to glutamate levels, supporting the hypothesis that glutamatergic function may lead to structural changes associated with the expression of psychotic-like experiences.
Collapse
Affiliation(s)
- Gemma Modinos
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, UK
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, UK
| | - Alice Egerton
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, UK
| | - Anna McLaughlin
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, UK
| | - Katrina McMullen
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, UK
| | - Veena Kumari
- Department of Psychology, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, UK
| | - David J Lythgoe
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, UK
| | - Gareth J Barker
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, UK
| | - André Aleman
- University of Groningen, Department of Neuroscience, University Medical Centre Groningen, The Netherlands
| | - Steve CR Williams
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, UK
| |
Collapse
|