1
|
Magagnoli J, Cummings TH, Wyatt MD, Shtutman M, Sutton SS. Schizophrenia, Off-Label Antipsychotics, and Dementia Risk in People With HIV. J Acquir Immune Defic Syndr 2025; 98:133-142. [PMID: 39418469 DOI: 10.1097/qai.0000000000003545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 08/27/2024] [Indexed: 10/19/2024]
Abstract
BACKGROUND Comorbidities such as schizophrenia and medication such as antipsychotics may influence the risk of dementia among people living with HIV (PLWH). The objective of this article is to assess the associations among HIV patients with schizophrenia, off-label antipsychotics, and dementia risk. SETTING US Department of Veterans Affairs health care facilities from 2000 to September 2023. METHODS Retrospective cohort study of PLWH treated by the US Department of Veterans Affairs with history of schizophrenia, off-label antipsychotic use, and neither schizophrenia nor antipsychotic use. Propensity score-matched non-HIV controls were included for the respective HIV groups. The hazard of dementia is estimated using Cox proportional hazards models. RESULTS PLWH and schizophrenia, were found to have a 2.49 higher hazard of dementia than HIV patients with no history of schizophrenia or antipsychotic medication use (hazard ratio [HR] = 2.49, 95% confidence interval [CI]: 1.85 to 3.35). PLWH and off-label antipsychotic use were found to have a 1.77-fold higher hazard of dementia than HIV patients with no history of schizophrenia or antipsychotic medication use (HR = 1.77, 95% CI: 1.37 to 2.28). Propensity score-matched analysis reveals that, among patients with schizophrenia, those with HIV had a 1.65-fold higher hazard of dementia (HR = 1.65, 95% CI: 1.12 to 2.44). Among patients with no schizophrenia or antipsychotic medication, those with HIV had a 1.47-fold higher hazard of dementia (HR = 1.47, 95% CI: 1.33 to 1.63). CONCLUSIONS This study demonstrates that among PLWH, history of schizophrenia or off-label antipsychotic medication use are associated with substantial increases in dementia incidence. Furthermore, propensity score-matched control analysis reveals that HIV infection itself is independently and significantly associated with elevated dementia risk.
Collapse
Affiliation(s)
- Joseph Magagnoli
- Dorn Research Institute, Columbia VA Health Care System, Columbia, SC
- Department of Clinical Pharmacy and Outcomes Sciences, College of Pharmacy, University of South Carolina, Columbia, SC; and
| | - Tammy H Cummings
- Dorn Research Institute, Columbia VA Health Care System, Columbia, SC
- Department of Clinical Pharmacy and Outcomes Sciences, College of Pharmacy, University of South Carolina, Columbia, SC; and
| | - Michael D Wyatt
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC
| | - Michael Shtutman
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC
| | - S Scott Sutton
- Dorn Research Institute, Columbia VA Health Care System, Columbia, SC
- Department of Clinical Pharmacy and Outcomes Sciences, College of Pharmacy, University of South Carolina, Columbia, SC; and
| |
Collapse
|
2
|
Chopra S, Levi PT, Holmes A, Orchard ER, Segal A, Francey SM, O'Donoghue B, Cropley VL, Nelson B, Graham J, Baldwin L, Yuen HP, Allott K, Alvarez-Jimenez M, Harrigan S, Pantelis C, Wood SJ, McGorry P, Fornito A. Brainwide Anatomical Connectivity and Prediction of Longitudinal Outcomes in Antipsychotic-Naïve First-Episode Psychosis. Biol Psychiatry 2025; 97:157-166. [PMID: 39069164 DOI: 10.1016/j.biopsych.2024.07.016] [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] [Received: 02/17/2024] [Revised: 06/05/2024] [Accepted: 07/16/2024] [Indexed: 07/30/2024]
Abstract
BACKGROUND Disruptions of axonal connectivity are thought to be a core pathophysiological feature of psychotic illness, but whether they are present early in the illness, prior to antipsychotic exposure, and whether they can predict clinical outcome remain unknown. METHODS We acquired diffusion-weighted magnetic resonance images to map structural connectivity between each pair of 319 parcellated brain regions in 61 antipsychotic-naïve individuals with first-episode psychosis (15-25 years, 46% female) and a demographically matched sample of 27 control participants. Clinical follow-up data were also acquired in patients 3 and 12 months after the scan. We used connectome-wide analyses to map disruptions of inter-regional pairwise connectivity and connectome-based predictive modeling to predict longitudinal change in symptoms and functioning. RESULTS Individuals with first-episode psychosis showed disrupted connectivity in a brainwide network linking all brain regions compared with controls (familywise error-corrected p = .03). Baseline structural connectivity significantly predicted change in functioning over 12 months (r = 0.44, familywise error-corrected p = .041), such that lower connectivity within fronto-striato-thalamic systems predicted worse functional outcomes. CONCLUSIONS Brainwide reductions of structural connectivity exist during the early stages of psychotic illness and cannot be attributed to antipsychotic medication. Moreover, baseline measures of structural connectivity can predict change in patient functional outcomes up to 1 year after engagement with treatment services.
Collapse
Affiliation(s)
- Sidhant Chopra
- Turner Institute for Brain and Mental Health, School of Psychological Science, Monash University, Clayton, Australia; Monash Biomedical Imaging, Monash University, Clayton, Australia; Department of Psychology, Yale University, New Haven, Connecticut; Orygen, Parkville, Victoria, Australia; Centre for Youth Mental Health, The University of Melbourne, Melbourne, Victoria, Australia.
| | - Priscila T Levi
- Turner Institute for Brain and Mental Health, School of Psychological Science, Monash University, Clayton, Australia; Monash Biomedical Imaging, Monash University, Clayton, Australia
| | - Alexander Holmes
- Turner Institute for Brain and Mental Health, School of Psychological Science, Monash University, Clayton, Australia; Monash Biomedical Imaging, Monash University, Clayton, Australia
| | - Edwina R Orchard
- Yale Child Study Centre, Yale University, New Haven, Connecticut
| | - Ashlea Segal
- Turner Institute for Brain and Mental Health, School of Psychological Science, Monash University, Clayton, Australia; Monash Biomedical Imaging, Monash University, Clayton, Australia; Wu Tsai Institute, Department of Neuroscience, Yale University, New Haven, Connecticut; Department of Neuroscience, Yale School of Medicine, Yale University, New Haven, Connecticut
| | - Shona M Francey
- Orygen, Parkville, Victoria, Australia; Centre for Youth Mental Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Brian O'Donoghue
- Centre for Youth Mental Health, The University of Melbourne, Melbourne, Victoria, Australia; St. Vincent's University Hospital, Dublin 4, Ireland; Department of Psychiatry, University College Dublin, Dublin 4, Ireland
| | - Vanessa L Cropley
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, University of Melbourne, Melbourne, Victoria, Australia
| | - Barnaby Nelson
- Orygen, Parkville, Victoria, Australia; Centre for Youth Mental Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Jessica Graham
- Orygen, Parkville, Victoria, Australia; Centre for Youth Mental Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Lara Baldwin
- Orygen, Parkville, Victoria, Australia; Centre for Youth Mental Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Hok Pan Yuen
- Orygen, Parkville, Victoria, Australia; Centre for Youth Mental Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Kelly Allott
- Orygen, Parkville, Victoria, Australia; Centre for Youth Mental Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Mario Alvarez-Jimenez
- Orygen, Parkville, Victoria, Australia; Centre for Youth Mental Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Susy Harrigan
- Orygen, Parkville, Victoria, Australia; Centre for Youth Mental Health, The University of Melbourne, Melbourne, Victoria, Australia; Centre for Mental Health, Melbourne School of Global and Population Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Christos Pantelis
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, University of Melbourne, Melbourne, Victoria, Australia; Western Hospital Sunshine, St. Albans, Victoria, Australia
| | - Stephen J Wood
- Orygen, Parkville, Victoria, Australia; Centre for Youth Mental Health, The University of Melbourne, Melbourne, Victoria, Australia; School of Psychology, University of Birmingham, Edgbaston, United Kingdom
| | - Patrick McGorry
- Orygen, Parkville, Victoria, Australia; Centre for Youth Mental Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Alex Fornito
- Turner Institute for Brain and Mental Health, School of Psychological Science, Monash University, Clayton, Australia; Monash Biomedical Imaging, Monash University, Clayton, Australia
| |
Collapse
|
3
|
Lieberman JA, Mendelsohn A, Goldberg TE, Emsley R. Preventing disease progression in schizophrenia: What are we waiting for. J Psychiatr Res 2025; 181:716-727. [PMID: 39754992 DOI: 10.1016/j.jpsychires.2024.12.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 12/09/2024] [Accepted: 12/26/2024] [Indexed: 01/06/2025]
Abstract
Despite research advances and progress in health care, schizophrenia remains a debilitating and costly disease. Onset occurs typically during youth and can lead to a relapsing and ultimately chronic course with persistent symptoms and functional impairment if not promptly and properly treated. Consequently, over time, schizophrenia causes substantial distress and disability for patients, their families and accrues to a collective burden to society. Recent research has revealed much about the pathophysiology that underlies the progressive nature of schizophrenia. Additionally, treatment strategies for disease management have been developed that have the potential to not just control psychotic symptoms but limit the cumulative morbidity of the illness. Given the evidence for their effectiveness and feasibility for their application, it is perplexing that this model of care has not yet become the standard of care and widely implemented to reduce the burden of illness on patients and society. This begs the question of whether the failure of implementation of a potentially disease-modifying strategy is due to the lack of evidence of efficacy (or belief in it) and readiness for implementation, or whether it's the lack of motivation and political will to support their utilization. To address this question, we reviewed and summarized the literature describing the natural history, pathophysiology and therapeutic strategies that can alleviate symptoms, prevent relapse, and potentially modify the course of schizophrenia. We conclude that, while we await further advances in mental health care from research, we must fully appreciate and take advantage of the effectiveness of existing treatments and overcome the attitudinal, policy, and infrastructural barriers to providing optimal mental health care capable of providing a disease-modifying treatment to patients with schizophrenia.
Collapse
Affiliation(s)
- Jeffrey A Lieberman
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA.
| | - Alana Mendelsohn
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Terry E Goldberg
- Division of Geriatric Psychiatry, Department of Psychiatry, Columbia University Medical Center, New York, NY, USA
| | - Robin Emsley
- Department of Psychiatry, Stellenbosch University, Cape Town, South Africa
| |
Collapse
|
4
|
Leucht S, Priller J, Davis JM. Antipsychotic Drugs: A Concise Review of History, Classification, Indications, Mechanism, Efficacy, Side Effects, Dosing, and Clinical Application. Am J Psychiatry 2024; 181:865-878. [PMID: 39350614 DOI: 10.1176/appi.ajp.20240738] [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: 10/23/2024]
Abstract
The introduction of the first antipsychotic drug, chlorpromazine, was a milestone for psychiatry. The authors review the history, classification, indications, mechanism, efficacy, side effects, dosing, drug initiation, switching, and other practical issues and questions related to antipsychotics. Classifications such as first-generation/typical versus second-generation/atypical antipsychotics are neither valid nor useful; these agents should be described according to the Neuroscience-based Nomenclature (NbN). Antipsychotic drugs are not specific for treating schizophrenia. They reduce psychosis regardless of the underlying diagnosis, and they go beyond nonspecific sedation. All currently available antipsychotic drugs are dopamine blockers or dopamine partial agonists. In schizophrenia, effect sizes for relapse prevention are larger than for acute treatment. A major unresolved problem is the implausible increase in placebo response in antipsychotic drug trials over the decades. Differences in side effects, which can be objectively measured, such as weight gain, are less equivocal than differences in rating-scale-measured (subjective) efficacy. The criteria for choosing among antipsychotics are mainly pragmatic and include factors such as available formulations, metabolism, half-life, efficacy, and side effects in previous illness episodes. Plasma levels help to detect nonadherence, and once-daily dosing at night (which is possible with many antipsychotics) and long-acting injectable formulations are useful when adherence is a problem. Dose-response curves for both acute treatment and relapse prevention follow a hyperbolic pattern, with maximally efficacious average dosages for schizophrenia of around 5 mg/day risperidone equivalents. Computer apps facilitating the choice between drugs are available. Future drug development should include pharmacogenetics and focus on drugs for specific aspects of psychosis.
Collapse
Affiliation(s)
- Stefan Leucht
- Technical University of Munich, TUM School of Medicine and Health, Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar, Munich (Leucht, Priller); German Center for Mental Health, Munich (Leucht, Priller); Neuropsychiatry, Charité-Universitätsmedizin Berlin, and German Center for Neurodegenerative Disorders, Berlin (Priller); University of Edinburgh and UK Dementia Research Institute, Edinburgh (Priller); Department of Psychiatry, University of Illinois at Chicago (Davis)
| | - Josef Priller
- Technical University of Munich, TUM School of Medicine and Health, Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar, Munich (Leucht, Priller); German Center for Mental Health, Munich (Leucht, Priller); Neuropsychiatry, Charité-Universitätsmedizin Berlin, and German Center for Neurodegenerative Disorders, Berlin (Priller); University of Edinburgh and UK Dementia Research Institute, Edinburgh (Priller); Department of Psychiatry, University of Illinois at Chicago (Davis)
| | - John M Davis
- Technical University of Munich, TUM School of Medicine and Health, Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar, Munich (Leucht, Priller); German Center for Mental Health, Munich (Leucht, Priller); Neuropsychiatry, Charité-Universitätsmedizin Berlin, and German Center for Neurodegenerative Disorders, Berlin (Priller); University of Edinburgh and UK Dementia Research Institute, Edinburgh (Priller); Department of Psychiatry, University of Illinois at Chicago (Davis)
| |
Collapse
|
5
|
Chung AN, Huang MC, Liu TH, Chang HM, Chen PY, Liu YL, Bavato F. Ketamine-dependent patients with persistent psychosis have higher neurofilament light chain levels than patients with schizophrenia. Asian J Psychiatr 2024; 100:104167. [PMID: 39111088 DOI: 10.1016/j.ajp.2024.104167] [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] [Received: 11/27/2023] [Revised: 07/21/2024] [Accepted: 07/22/2024] [Indexed: 09/13/2024]
Abstract
OBJECTIVES Ketamine can induce persisting psychosis in a subset of individuals who use it chronically and heavily. Previously, we found that the psychopathology and cognitive impairments in patients with ketamine dependence (KD) exhibiting persistent psychosis (KPP) bear resemblances with schizophrenia, albeit with less severity in those with no persistent psychosis (KNP). Furthermore, we also showed that patients with KD had higher blood levels of neurofilament light chain (NFL), a biomarker for neuroaxonal injury, compared to healthy controls. In this study, we aimed to investigate the differences in NFL levels between patients with KPP and KNP while comparing the levels of individuals with schizophrenia and healthy controls. METHODS We enrolled 64 treatment-seeking ketamine-dependent patients (53 with KNP and 11 with KPP), 37 medication-free patients with schizophrenia, and 80 healthy controls. Blood NFL levels were measured by single molecule array immunoassay. RESULTS NFL levels were highest in the KPP subgroup, followed by the KNP subgroup, and then the schizophrenia and control groups (mean ± SD: 24.5 ± 24.7, 12.9 ± 10.9, 9.2 ± 12.2, and 6.2 ± 2.2 pg/mL, respectively), with no significant difference observed between the schizophrenia and control groups. CONCLUSIONS We found that KD is associated with higher NFL levels compared to schizophrenia, with the KPP subgroup showing the most consistent alterations. The observation of accentuated neuroaxonal pathology in individuals with KPP implies that this clinical manifestation is associated with a specific neurobiological phenotype, despite prior evidence suggesting syndromal similarity between schizophrenia and KPP.
Collapse
Affiliation(s)
- An-Nie Chung
- Department of Psychiatry, Taipei City Psychiatric Center, Taipei City Hospital, Taipei, Taiwan; Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, Miaoli County, Taiwan
| | - Ming-Chyi Huang
- Department of Psychiatry, Taipei City Psychiatric Center, Taipei City Hospital, Taipei, Taiwan; Department of Psychiatry, School of Medicine, College of Medicine, Taipei Medical University, Taiwan; Psychiatric Research Center, Taipei Medical University Hospital., 250 Wuxing St, Taipei, Taiwan; Psychiatric Research Center, Wang-Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Tung-Hsia Liu
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, Miaoli County, Taiwan
| | - Hu-Ming Chang
- Department of Psychiatry, Taipei City Psychiatric Center, Taipei City Hospital, Taipei, Taiwan
| | - Po-Yu Chen
- Department of Psychiatry, Taipei City Psychiatric Center, Taipei City Hospital, Taipei, Taiwan; Department of Psychiatry, School of Medicine, College of Medicine, Taipei Medical University, Taiwan.
| | - Yu-Li Liu
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, Miaoli County, Taiwan.
| | - Francesco Bavato
- Experimental and Clinical Pharmacopsychology, Department of Psychiatry, Psychotherapy, and Psychosomatics, Psychiatric Hospital of the University of Zurich, Zurich, Switzerland
| |
Collapse
|
6
|
Tang B, Yao L, Strawn JR, Zhang W, Lui S. Neurostructural, Neurofunctional, and Clinical Features of Chronic, Untreated Schizophrenia: A Narrative Review. Schizophr Bull 2024:sbae152. [PMID: 39212651 DOI: 10.1093/schbul/sbae152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Studies of individuals with chronic, untreated schizophrenia (CUS) can provide important insights into the natural course of schizophrenia and how antipsychotic pharmacotherapy affects neurobiological aspects of illness course and progression. We systematically review 17 studies on the neuroimaging, cognitive, and epidemiological aspects of CUS individuals. These studies were conducted at the Shanghai Mental Health Center, Institute of Mental Health at Peking University, and Huaxi MR Research Center between 2013 and 2021. CUS is associated with cognitive impairment, severe symptoms, and specific demographic characteristics and is different significantly from those observed in antipsychotic-treated individuals. Furthermore, CUS individuals have neurostructural and neurofunctional alterations in frontal and temporal regions, corpus callosum, subcortical, and visual processing areas, as well as default-mode and somatomotor networks. As the disease progresses, significant structural deteriorations occur, such as accelerated cortical thinning in frontal and temporal lobes, greater reduction in fractional anisotropy in the genu of corpus callosum, and decline in nodal metrics of gray mater network in thalamus, correlating with worsening cognitive deficits and clinical outcomes. In addition, striatal hypertrophy also occurs, independent of antipsychotic treatment. Contrasting with the negative neurostructural and neurofunctional effects of short-term antipsychotic treatment, long-term therapy frequently results in significant improvements. It notably enhances white matter integrity and the functions of key subcortical regions such as the amygdala, hippocampus, and striatum, potentially improving cognitive functions. This narrative review highlights the progressive neurobiological sequelae of CUS, the importance of early detection, and long-term treatment of schizophrenia, particularly because treatment may attenuate neurobiological deterioration and improve clinical outcomes.
Collapse
Affiliation(s)
- Biqiu Tang
- Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital of Sichuan University, Chengdu, China
- Department of Psychiatry & Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH
| | - Li Yao
- Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital of Sichuan University, Chengdu, China
| | - Jeffrey R Strawn
- Department of Psychiatry & Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH
| | - Wenjing Zhang
- Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital of Sichuan University, Chengdu, China
| | - Su Lui
- Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
7
|
Hua JPY, Abram SV, Loewy RL, Stuart B, Fryer SL, Vinogradov S, Mathalon DH. Brain Age Gap in Early Illness Schizophrenia and the Clinical High-Risk Syndrome: Associations With Experiential Negative Symptoms and Conversion to Psychosis. Schizophr Bull 2024; 50:1159-1170. [PMID: 38815987 PMCID: PMC11349027 DOI: 10.1093/schbul/sbae074] [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] [Indexed: 06/01/2024]
Abstract
BACKGROUND AND HYPOTHESIS Brain development/aging is not uniform across individuals, spawning efforts to characterize brain age from a biological perspective to model the effects of disease and maladaptive life processes on the brain. The brain age gap represents the discrepancy between estimated brain biological age and chronological age (in this case, based on structural magnetic resonance imaging, MRI). Structural MRI studies report an increased brain age gap (biological age > chronological age) in schizophrenia, with a greater brain age gap related to greater negative symptom severity. Less is known regarding the nature of this gap early in schizophrenia (ESZ), if this gap represents a psychosis conversion biomarker in clinical high-risk (CHR-P) individuals, and how altered brain development and/or aging map onto specific symptom facets. STUDY DESIGN Using structural MRI, we compared the brain age gap among CHR-P (n = 51), ESZ (n = 78), and unaffected comparison participants (UCP; n = 90), and examined associations with CHR-P psychosis conversion (CHR-P converters n = 10; CHR-P non-converters; n = 23) and positive and negative symptoms. STUDY RESULTS ESZ showed a greater brain age gap relative to UCP and CHR-P (Ps < .010). CHR-P individuals who converted to psychosis showed a greater brain age gap (P = .043) relative to CHR-P non-converters. A larger brain age gap in ESZ was associated with increased experiential (P = .008), but not expressive negative symptom severity. CONCLUSIONS Consistent with schizophrenia pathophysiological models positing abnormal brain maturation, results suggest abnormal brain development is present early in psychosis. An increased brain age gap may be especially relevant to motivational and functional deficits in schizophrenia.
Collapse
Affiliation(s)
- Jessica P Y Hua
- Sierra Pacific Mental Illness Research Education and Clinical Centers, San Francisco VA Medical Center, University of California, San Francisco, CA, USA
- Mental Health Service, San Francisco VA Health Care System, San Francisco, CA, USA
- Department of Psychiatry and Behavioral Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Samantha V Abram
- Mental Health Service, San Francisco VA Health Care System, San Francisco, CA, USA
- Department of Psychiatry and Behavioral Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Rachel L Loewy
- Department of Psychiatry and Behavioral Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Barbara Stuart
- Department of Psychiatry and Behavioral Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Susanna L Fryer
- Mental Health Service, San Francisco VA Health Care System, San Francisco, CA, USA
- Department of Psychiatry and Behavioral Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Sophia Vinogradov
- Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis, MN, USA
| | - Daniel H Mathalon
- Mental Health Service, San Francisco VA Health Care System, San Francisco, CA, USA
- Department of Psychiatry and Behavioral Sciences, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
8
|
Kim WS, Heo DW, Maeng J, Shen J, Tsogt U, Odkhuu S, Zhang X, Cheraghi S, Kim SW, Ham BJ, Rami FZ, Sui J, Kang CY, Suk HI, Chung YC. Deep Learning-based Brain Age Prediction in Patients With Schizophrenia Spectrum Disorders. Schizophr Bull 2024; 50:804-814. [PMID: 38085061 PMCID: PMC11283195 DOI: 10.1093/schbul/sbad167] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/29/2024]
Abstract
BACKGROUND AND HYPOTHESIS The brain-predicted age difference (brain-PAD) may serve as a biomarker for neurodegeneration. We investigated the brain-PAD in patients with schizophrenia (SCZ), first-episode schizophrenia spectrum disorders (FE-SSDs), and treatment-resistant schizophrenia (TRS) using structural magnetic resonance imaging (sMRI). STUDY DESIGN We employed a convolutional network-based regression (SFCNR), and compared its performance with models based on three machine learning (ML) algorithms. We pretrained the SFCNR with sMRI data of 7590 healthy controls (HCs) selected from the UK Biobank. The parameters of the pretrained model were transferred to the next training phase with a new set of HCs (n = 541). The brain-PAD was analyzed in independent HCs (n = 209) and patients (n = 233). Correlations between the brain-PAD and clinical measures were investigated. STUDY RESULTS The SFCNR model outperformed three commonly used ML models. Advanced brain aging was observed in patients with SCZ, FE-SSDs, and TRS compared to HCs. A significant difference in brain-PAD was observed between FE-SSDs and TRS with ridge regression but not with the SFCNR model. Chlorpromazine equivalent dose and cognitive function were correlated with the brain-PAD in SCZ and FE-SSDs. CONCLUSIONS Our findings indicate that there is advanced brain aging in patients with SCZ and higher brain-PAD in SCZ can be used as a surrogate marker for cognitive dysfunction. These findings warrant further investigations on the causes of advanced brain age in SCZ. In addition, possible psychosocial and pharmacological interventions targeting brain health should be considered in early-stage SCZ patients with advanced brain age.
Collapse
Affiliation(s)
- Woo-Sung Kim
- Department of Psychiatry, Jeonbuk National University, Medical School, Jeonju, Korea
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Korea
| | - Da-Woon Heo
- Department of Artificial Intelligence, Korea University, Seoul, Korea
| | - Junyeong Maeng
- Department of Artificial Intelligence, Korea University, Seoul, Korea
| | - Jie Shen
- Department of Psychiatry, Jeonbuk National University, Medical School, Jeonju, Korea
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Korea
- Department of Psychiatry, Yanbian University, Medical School, Yanji, China
| | - Uyanga Tsogt
- Department of Psychiatry, Jeonbuk National University, Medical School, Jeonju, Korea
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Korea
| | - Soyolsaikhan Odkhuu
- Department of Psychiatry, Jeonbuk National University, Medical School, Jeonju, Korea
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Korea
| | - Xuefeng Zhang
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Korea
| | - Sahar Cheraghi
- Department of Psychiatry, Jeonbuk National University, Medical School, Jeonju, Korea
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Korea
| | - Sung-Wan Kim
- Department of Psychiatry, Chonnam National University Medical School, Gwangju, Korea
| | - Byung-Joo Ham
- Department of Psychiatry, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Korea
| | - Fatima Zahra Rami
- Department of Psychiatry, Jeonbuk National University, Medical School, Jeonju, Korea
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Korea
| | - Jing Sui
- Tri-Institutional Center for Translational Research in Neuroimaging and Data Science (TReNDS), Georgia State University, Georgia Institute of Technology, Emory University, Atlanta, GA, USA
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China
| | - Chae Yeong Kang
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Korea
| | - Heung-Il Suk
- Department of Artificial Intelligence, Korea University, Seoul, Korea
- Department of Brain and Cognitive Engineering, Korea University, Seoul, Korea
| | - Young-Chul Chung
- Department of Psychiatry, Jeonbuk National University, Medical School, Jeonju, Korea
- Department of Psychiatry, Jeonbuk National University Hospital, Jeonju, Korea
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Korea
| |
Collapse
|
9
|
Jezsó B, Kálmán S, Farkas KG, Hathy E, Vincze K, Kovács-Schoblocher D, Lilienberg J, Tordai C, Nemoda Z, Homolya L, Apáti Á, Réthelyi JM. Haloperidol, Olanzapine, and Risperidone Induce Morphological Changes in an In Vitro Model of Human Hippocampal Neurogenesis. Biomolecules 2024; 14:688. [PMID: 38927091 PMCID: PMC11201986 DOI: 10.3390/biom14060688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/04/2024] [Accepted: 06/09/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND Induced pluripotent stem cell (iPSC) based neuronal differentiation is valuable for studying neuropsychiatric disorders and pharmacological mechanisms at the cellular level. We aimed to examine the effects of typical and atypical antipsychotics on human iPSC-derived neural progenitor cells (NPCs). METHODS Proliferation and neurite outgrowth were measured by live cell imaging, and gene expression levels related to neuronal identity were analyzed by RT-QPCR and immunocytochemistry during differentiation into hippocampal dentate gyrus granule cells following treatment of low- and high-dose antipsychotics (haloperidol, olanzapine, and risperidone). RESULTS Antipsychotics did not modify the growth properties of NPCs after 3 days of treatment. However, the characteristics of neurite outgrowth changed significantly in response to haloperidol and olanzapine. After three weeks of differentiation, mRNA expression levels of the selected neuronal markers increased (except for MAP2), while antipsychotics caused only subtle changes. Additionally, we found no changes in MAP2 or GFAP protein expression levels as a result of antipsychotic treatment. CONCLUSIONS Altogether, antipsychotic medications promoted neurogenesis in vitro by influencing neurite outgrowth rather than changing cell survival or gene expression. This study provides insights into the effects of antipsychotics on neuronal differentiation and highlights the importance of considering neurite outgrowth as a potential target of action.
Collapse
Affiliation(s)
- Bálint Jezsó
- Institute of Molecular Life Sciences, HUN-REN RCNS, Magyar tudósok körútja 2., H-1117 Budapest, Hungary; (B.J.)
- Doctoral School of Biology, Institute of Biology, Eötvös Loránd University, Pázmány Péter sétány 1/c., H-1117 Budapest, Hungary
- ELTE-MTA “Momentum” Motor Enzymology Research Group, Department of Biochemistry, Eötvös Loránd University, Pázmány Péter sétány 1/c., H-1117 Budapest, Hungary
| | - Sára Kálmán
- Albert Szent-Györgyi Health Centre, Department of Psychiatry, University of Szeged, Szentháromság utca 5., H-6722 Szeged, Hungary;
| | - Kiara Gitta Farkas
- Institute of Molecular Life Sciences, HUN-REN RCNS, Magyar tudósok körútja 2., H-1117 Budapest, Hungary; (B.J.)
| | - Edit Hathy
- Institute of Molecular Life Sciences, HUN-REN RCNS, Magyar tudósok körútja 2., H-1117 Budapest, Hungary; (B.J.)
- Department of Psychiatry and Psychotherapy, Semmelweis University, Balassa utca 6., H-1083 Budapest, Hungary
| | - Katalin Vincze
- Institute of Molecular Life Sciences, HUN-REN RCNS, Magyar tudósok körútja 2., H-1117 Budapest, Hungary; (B.J.)
- Department of Psychiatry and Psychotherapy, Semmelweis University, Balassa utca 6., H-1083 Budapest, Hungary
| | | | - Julianna Lilienberg
- Institute of Molecular Life Sciences, HUN-REN RCNS, Magyar tudósok körútja 2., H-1117 Budapest, Hungary; (B.J.)
| | - Csongor Tordai
- Institute of Molecular Life Sciences, HUN-REN RCNS, Magyar tudósok körútja 2., H-1117 Budapest, Hungary; (B.J.)
- Department of Psychiatry and Psychotherapy, Semmelweis University, Balassa utca 6., H-1083 Budapest, Hungary
| | - Zsófia Nemoda
- Department of Psychiatry and Psychotherapy, Semmelweis University, Balassa utca 6., H-1083 Budapest, Hungary
| | - László Homolya
- Institute of Molecular Life Sciences, HUN-REN RCNS, Magyar tudósok körútja 2., H-1117 Budapest, Hungary; (B.J.)
| | - Ágota Apáti
- Institute of Molecular Life Sciences, HUN-REN RCNS, Magyar tudósok körútja 2., H-1117 Budapest, Hungary; (B.J.)
| | - János M. Réthelyi
- Department of Psychiatry and Psychotherapy, Semmelweis University, Balassa utca 6., H-1083 Budapest, Hungary
| |
Collapse
|
10
|
Konttajärvi T, Haapea M, Huhtaniska S, Björnholm L, Miettunen J, Isohanni M, Penttilä M, Murray GK, Koponen H, Vernon AC, Jääskeläinen E, Lieslehto J. The contribution of first-episode illness characteristics and cumulative antipsychotic usage to progressive structural brain changes over a long-term follow-up in schizophrenia. Psychiatry Res Neuroimaging 2024; 339:111790. [PMID: 38354478 DOI: 10.1016/j.pscychresns.2024.111790] [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: 03/08/2023] [Revised: 11/26/2023] [Accepted: 01/08/2024] [Indexed: 02/16/2024]
Abstract
Exposure to antipsychotics as well as certain first-episode illness characteristics have been associated with greater gray matter (GM) deficits in the early phase of schizophrenia. Whether the first-episode illness characteristics affect the long-term progression of the structural brain changes remain unexplored. We therefore assessed the role of first-episode illness characteristics and life-time antipsychotic use in relation to long-term structural brain GM changes in schizophrenia. Individuals with schizophrenia (SZ, n = 29) and non-psychotic controls (n = 61) from the Northern Finland Birth Cohort 1966 underwent structural MRI at the ages of 34 (baseline) and 43 (follow-up) years. At follow-up, the average duration of illness was 19.8 years. Voxel-based morphometry was used to assess the effects of predictors on longitudinal GM changes in schizophrenia-relevant brain areas. Younger age of onset (AoO), higher cumulative antipsychotic dose and severity of symptoms were associated with greater GM deficits in the SZ group at follow-up. None of the first-episode illness characteristics were associated with longitudinal GM changes during 9-year follow-up period. We conclude that a younger AoO and high life-time antipsychotic use may contribute to progression of structural brain changes in schizophrenia. Apart from AoO, other first-episode illness characteristics may not contribute to longitudinal GM changes in midlife.
Collapse
Affiliation(s)
| | - Marianne Haapea
- Research Unit of Population Health, University of Oulu, Oulu, Finland; Medical Research Center Oulu, Oulu University of Hospital and University of Oulu, Finland; Department of Psychiatry, Oulu University of Hospital, Finland; Department of Diagnostic Radiology, Oulu University Hospital, Oulu, Finland
| | - Sanna Huhtaniska
- Research Unit of Population Health, University of Oulu, Oulu, Finland
| | - Lassi Björnholm
- Department of Psychiatry, Oulu University of Hospital, Finland; Research Unit of Clinical Neuroscience, University of Oulu, Finland
| | - Jouko Miettunen
- Research Unit of Population Health, University of Oulu, Oulu, Finland; Medical Research Center Oulu, Oulu University of Hospital and University of Oulu, Finland
| | - Matti Isohanni
- Research Unit of Population Health, University of Oulu, Oulu, Finland
| | - Matti Penttilä
- Research Unit of Population Health, University of Oulu, Oulu, Finland
| | - Graham K Murray
- Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
| | - Hannu Koponen
- University of Helsinki, Helsinki University Hospital, Psychiatry, Helsinki, Finland
| | - Anthony C Vernon
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom; MRC Centre for Neurodevelopmental Disorders, King's College London, London,United Kingdom
| | - Erika Jääskeläinen
- Research Unit of Population Health, University of Oulu, Oulu, Finland; Medical Research Center Oulu, Oulu University of Hospital and University of Oulu, Finland; Department of Psychiatry, Oulu University of Hospital, Finland
| | - Johannes Lieslehto
- Research Unit of Population Health, University of Oulu, Oulu, Finland; Medical Research Center Oulu, Oulu University of Hospital and University of Oulu, Finland; Department of Forensic Psychiatry, University of Eastern Finland, Niuvanniemi Hospital, Kuopio, Finland; Department of Clinical Neuroscience, Division of Insurance Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
11
|
Emsley R. Antipsychotics and structural brain changes: could treatment adherence explain the discrepant findings? Ther Adv Psychopharmacol 2023; 13:20451253231195258. [PMID: 37701891 PMCID: PMC10493054 DOI: 10.1177/20451253231195258] [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: 03/15/2023] [Accepted: 07/11/2023] [Indexed: 09/14/2023] Open
Abstract
Progressive structural brain changes are well documented in schizophrenia and have been linked to both illness progression and the extent of antipsychotic treatment exposure. Literature reporting longitudinal changes in brain structure in individuals with schizophrenia is selectively reviewed to assess the roles of illness, antipsychotic treatment, adherence and other factors in the genesis of these changes. This narrative review considers literature investigating longitudinal changes in brain structure in individuals with schizophrenia. The review focusses on structural changes in the cortex, basal ganglia and white matter. It also examines effects of medication non-adherence and relapse on the clinical course of the illness and on structural brain changes. Studies investigating structural magnetic resonance imaging changes in patients treated with long-acting injectable antipsychotics are reviewed. Temporal changes in brain structure in schizophrenia can be divided into those that are associated with antipsychotic treatment and those that are not. Changes associated with treatment include increases in basal ganglia and white matter volumes. Relapse episodes may be a critical factor in illness progression and brain volume reductions. Medication adherence may be an important factor that could explain the findings that brain volume reductions are associated with poor treatment response, higher intensity of antipsychotic treatment exposure and more time spent in relapse. Improved adherence via long-acting injectable antipsychotics and adherence focussed psychosocial interventions could maximize protective effects of antipsychotics against illness progression.
Collapse
Affiliation(s)
- Robin Emsley
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, PO Box 241, Tygerberg Campus, Cape Town 8000, South Africa
| |
Collapse
|
12
|
Hua JPY, Loewy RL, Stuart B, Fryer SL, Niendam TA, Carter CS, Vinogradov S, Mathalon DH. Cortical and subcortical brain morphometry abnormalities in youth at clinical high-risk for psychosis and individuals with early illness schizophrenia. Psychiatry Res Neuroimaging 2023; 332:111653. [PMID: 37121090 PMCID: PMC10362971 DOI: 10.1016/j.pscychresns.2023.111653] [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: 01/23/2023] [Revised: 03/27/2023] [Accepted: 04/18/2023] [Indexed: 05/02/2023]
Abstract
Neuroimaging studies have documented morphometric brain abnormalities in schizophrenia, but less is known about them in individuals at clinical high-risk for psychosis (CHR-P), including how they compare with those observed in early schizophrenia (ESZ). Accordingly, we implemented multivariate profile analysis of regional morphometric profiles in CHR-P (n = 89), ESZ (n = 93) and healthy controls (HC; n = 122). ESZ profiles differed from HC and CHR-P profiles, including 1) cortical thickness: significant level reduction and regional non-parallelism reflecting widespread thinning, except for entorhinal and pericalcarine cortex, 2) basal ganglia volume: significant level increase and regional non-parallelism reflecting larger caudate and pallidum, and 3) ventricular volume: significant level increase with parallel regional profiles. CHR-P and ESZ cerebellar profiles showed significant non-parallelism with HC profiles. Regional profiles did not significantly differ between groups for cortical surface area or subcortical volume. Compared to CHR-P followed for ≥18 months without psychosis conversion (n = 31), CHR-P converters (n = 17) showed significant non-parallel ventricular volume expansion reflecting specific enlargement of lateral and inferolateral regions. Antipsychotic dosage in ESZ was significantly correlated with frontal cortical thinning. Results suggest that morphometric abnormalities in ESZ are not present in CHR-P, except for ventricular enlargement, which was evident in CHR-P who developed psychosis.
Collapse
Affiliation(s)
- Jessica P Y Hua
- Sierra Pacific Mental Illness Research Education and Clinical Centers, San Francisco VA Medical Center, and the University of California, San Francisco, CA, United States; Mental Health Service, San Francisco VA Medical Center, San Francisco, 94121, CA, United States; Department of Psychiatry and Behavioral Sciences, University of California San Francisco, San Francisco, 94143, CA, United States; Department of Psychological Sciences, University of Missouri, Columbia, 65211, MO, United States
| | - Rachel L Loewy
- Department of Psychiatry and Behavioral Sciences, University of California San Francisco, San Francisco, 94143, CA, United States
| | - Barbara Stuart
- Department of Psychiatry and Behavioral Sciences, University of California San Francisco, San Francisco, 94143, CA, United States
| | - Susanna L Fryer
- Mental Health Service, San Francisco VA Medical Center, San Francisco, 94121, CA, United States
| | - Tara A Niendam
- Department of Psychiatry and Behavioral Sciences, University of California Davis, Davis, 95616, CA, United States
| | - Cameron S Carter
- Department of Psychiatry and Behavioral Sciences, University of California Davis, Davis, 95616, CA, United States
| | - Sophia Vinogradov
- Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis, 55455, MN, United States
| | - Daniel H Mathalon
- Mental Health Service, San Francisco VA Medical Center, San Francisco, 94121, CA, United States; Department of Psychiatry and Behavioral Sciences, University of California San Francisco, San Francisco, 94143, CA, United States.
| |
Collapse
|
13
|
Sharma K, Dev KK. The Effects of Antipsychotics in Experimental Models of Krabbe Disease. Biomedicines 2023; 11:biomedicines11051313. [PMID: 37238985 DOI: 10.3390/biomedicines11051313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 04/13/2023] [Accepted: 04/21/2023] [Indexed: 05/28/2023] Open
Abstract
The role of altered myelin in the onset and development of schizophrenia and changes in myelin due to antipsychotics remains unclear. Antipsychotics are D2 receptor antagonists, yet D2 receptor agonists increase oligodendrocyte progenitor numbers and limit oligodendrocyte injury. Conflicting studies suggest these drugs promote the differentiation of neural progenitors to oligodendrocyte lineage, while others report antipsychotics inhibit the proliferation and differentiation of oligodendrocyte precursors. Here, we utilised in-vitro (human astrocytes), ex-vivo (organotypic slice cultures) and in-vivo (twitcher mouse model) experimental study designs of psychosine-induced demyelination, a toxin that accumulates in Krabbe disease (KD), to investigate direct effects of antipsychotics on glial cell dysfunction and demyelination. Typical and atypical antipsychotics, and selective D2 and 5HT2A receptor antagonists, attenuated psychosine-induced cell viability, toxicity, and morphological aberrations in human astrocyte cultures. Haloperidol and clozapine reduced psychosine-induced demyelination in mouse organotypic cerebellar slices. These drugs also attenuated the effects of psychosine on astrocytes and microglia and restored non-phosphorylated neurofilament levels, indicating neuroprotective effects. In the demyelinating twitcher mouse model of KD, haloperidol improved mobility and significantly increased the survival of these animals. Overall, this study suggests that antipsychotics directly regulate glial cell dysfunction and exert a protective effect on myelin loss. This work also points toward the potential use of these pharmacological agents in KD.
Collapse
Affiliation(s)
- Kapil Sharma
- Drug Development Research Group, Department of Physiology, School of Medicine, Trinity College Dublin, D02 R590 Dublin, Ireland
| | - Kumlesh K Dev
- Drug Development Research Group, Department of Physiology, School of Medicine, Trinity College Dublin, D02 R590 Dublin, Ireland
| |
Collapse
|
14
|
Emsley R, du Plessis S, Phahladira L, Luckhoff HK, Scheffler F, Kilian S, Smit R, Buckle C, Chiliza B, Asmal L. Antipsychotic treatment effects and structural MRI brain changes in schizophrenia. Psychol Med 2023; 53:2050-2059. [PMID: 35441587 PMCID: PMC10106303 DOI: 10.1017/s0033291721003809] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 07/21/2021] [Accepted: 09/01/2021] [Indexed: 11/06/2022]
Abstract
BACKGROUND Progressive brain structural MRI changes are described in schizophrenia and have been ascribed to both illness progression and antipsychotic treatment. We investigated treatment effects, in terms of total cumulative antipsychotic dose, efficacy and tolerability, on brain structural changes over the first 24 months of treatment in schizophrenia. METHODS A prospective, 24-month, single-site cohort study in 99 minimally treated patients with first-episode schizophrenia, schizophreniform and schizoaffective disorder, and 98 matched healthy controls. We treated the patients according to a fixed protocol with flupenthixol decanoate, a long-acting injectable antipsychotic. We assessed psychopathology, cognition, extrapyramidal symptoms and BMI, and acquired MRI scans at months 0, 12 and 24. We selected global cortical thickness, white matter volume and basal ganglia volume as the regions of interest. RESULTS The only significant group × time interaction was for basal ganglia volumes. However, patients, but not controls, displayed cortical thickness reductions and increases in white matter and basal ganglia volumes. Cortical thickness reductions were unrelated to treatment. White matter volume increases were associated with lower cumulative antipsychotic dose, greater improvements in psychopathology and cognition, and more extrapyramidal symptoms. Basal ganglia volume increases were associated with greater improvements in psychopathology, greater increases in BMI and more extrapyramidal symptoms. CONCLUSIONS We provide evidence for plasticity in white matter and basal ganglia associated with antipsychotic treatment in schizophrenia, most likely linked to the dopamine blocking actions of these agents. Cortical changes may be more closely related to the neurodevelopmental, non-dopaminergic aspects of the illness.
Collapse
Affiliation(s)
- Robin Emsley
- Department of Psychiatry, Stellenbosch University, Tygerberg Campus, Cape Town, South Africa
| | - Stefan du Plessis
- Department of Psychiatry, Stellenbosch University, Tygerberg Campus, Cape Town, South Africa
| | - Lebogang Phahladira
- Department of Psychiatry, Stellenbosch University, Tygerberg Campus, Cape Town, South Africa
| | - Hilmar K. Luckhoff
- Department of Psychiatry, Stellenbosch University, Tygerberg Campus, Cape Town, South Africa
| | - Frederika Scheffler
- Department of Psychiatry, Stellenbosch University, Tygerberg Campus, Cape Town, South Africa
| | - Sanja Kilian
- Department of Psychiatry, Stellenbosch University, Tygerberg Campus, Cape Town, South Africa
| | - Retha Smit
- Department of Psychiatry, Stellenbosch University, Tygerberg Campus, Cape Town, South Africa
| | - Chanelle Buckle
- Department of Psychiatry, Stellenbosch University, Tygerberg Campus, Cape Town, South Africa
| | - Bonginkosi Chiliza
- Department of Psychiatry, Nelson R Mandela School of Medicine, University of Kwazulu-Natal, Durban, South Africa
| | - Laila Asmal
- Department of Psychiatry, Stellenbosch University, Tygerberg Campus, Cape Town, South Africa
| |
Collapse
|
15
|
Affiliation(s)
- Martin Harrow
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, USA
| | - Thomas H Jobe
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, USA
| | - Liping Tong
- Advocate Aurora Health, Downers Grove, IL, USA
| |
Collapse
|
16
|
Altered expression of microglial markers of phagocytosis in schizophrenia. Schizophr Res 2023; 251:22-29. [PMID: 36527956 DOI: 10.1016/j.schres.2022.12.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 09/07/2022] [Accepted: 12/06/2022] [Indexed: 12/23/2022]
Abstract
BACKGROUND Cognitive disturbances in schizophrenia have been linked to a lower density of dendritic spines on pyramidal neurons in the prefrontal cortex (PFC). Complement component C4, which has previously been found at higher levels in schizophrenia, marks synapses for phagocytosis by microglia. Thus, elevated consumption of dendritic spines by microglia mediated through excessive complement activity may play a role in lower spine density in schizophrenia. However, it is unclear if microglia themselves have the molecular capacity for enhanced phagocytosis of spines in schizophrenia. METHODS Transcript levels for complement components and microglia-specific phagocytic markers were quantified using quantitative PCR in the PFC of 62 matched pairs of schizophrenia and unaffected comparison subjects and in antipsychotic-exposed monkeys. RESULTS Relative to comparison subjects, schizophrenia subjects had higher mRNA levels for C4 (+154 %); C1q (+69 %), which initiates the classical complement pathway that includes C4; and for microglia-specific markers that enable phagocytic activity including TAM receptor tyrosine kinases Axl (+27 %) and MerTK (+27 %) and lysosome-associated glycoprotein CD68 (+27 %) (all p ≤ .042). Transcript levels for microglial phagocytic markers were correlated with C4 mRNA levels in schizophrenia subjects (all r ≥ 0.31, p ≤ .015). We also found further evidence consistent with microglial activation in schizophrenia, including higher mRNA levels for THIK1 (TWIK-related halothane-inhibited potassium channel: +30 %) and lower mRNA levels for the purinergic receptor P2Y12 (-27 %) (all p ≤ .016). Transcript levels were unchanged in antipsychotic-exposed monkeys. CONCLUSIONS These results are consistent with the presence of increased complement activity and an elevated molecular capacity of microglia for phagocytosis in the same schizophrenia subjects.
Collapse
|
17
|
Kruse AO, Bustillo JR. Glutamatergic dysfunction in Schizophrenia. Transl Psychiatry 2022; 12:500. [PMID: 36463316 PMCID: PMC9719533 DOI: 10.1038/s41398-022-02253-w] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 11/05/2022] [Accepted: 11/09/2022] [Indexed: 12/04/2022] Open
Abstract
The NMDA-R hypofunction model of schizophrenia started with the clinical observation of the precipitation of psychotic symptoms in patients with schizophrenia exposed to PCP or ketamine. Healthy volunteers exposed to acute low doses of ketamine experienced mild psychosis but also negative and cognitive type symptoms reminiscent of the full clinical picture of schizophrenia. In rodents, acute systemic ketamine resulted in a paradoxical increase in extracellular frontal glutamate as well as of dopamine. Similar increase in prefrontal glutamate was documented with acute ketamine in healthy volunteers with 1H-MRS. Furthermore, sub-chronic low dose PCP lead to reductions in frontal dendritic tree density in rodents. In post-mortem ultrastructural studies in schizophrenia, a broad reduction in dendritic complexity and somal volume of pyramidal cells has been repeatedly described. This most likely accounts for the broad, subtle progressive cortical thinning described with MRI in- vivo. Additionally, prefrontal reductions in the obligatory GluN1 subunit of the NMDA-R has been repeatedly found in post-mortem tissue. The vast 1H-MRS literature in schizophrenia has documented trait-like small increases in glutamate concentrations in striatum very early in the illness, before antipsychotic treatment (the same structure where increased pre-synaptic release of dopamine has been reported with PET). The more recent genetic literature has reliably detected very small risk effects for common variants involving several glutamate-related genes. The pharmacological literature has followed two main tracks, directly informed by the NMDA-R hypo model: agonism at the glycine site (as mostly add-on studies targeting negative and cognitive symptoms); and pre-synaptic modulation of glutamatergic release (as single agents for acute psychosis). Unfortunately, both approaches have failed so far. There is little doubt that brain glutamatergic abnormalities are present in schizophrenia and that some of these are related to the etiology of the illness. The genetic literature directly supports a non- specific etiological role for glutamatergic dysfunction. Whether NMDA-R hypofunction as a specific mechanism accounts for any important component of the illness is still not evident. However, a glutamatergic model still has heuristic value to guide future research in schizophrenia. New tools to jointly examine brain glutamatergic, GABA-ergic and dopaminergic systems in-vivo, early in the illness, may lay the ground for a next generation of clinical trials that go beyond dopamine D2 blockade.
Collapse
Affiliation(s)
- Andreas O Kruse
- Department of Psychiatry and Behavioral Sciences, University of New Mexico, Albuquerque, NM, 87131, USA.
| | - Juan R Bustillo
- Department of Psychiatry and Behavioral Sciences, University of New Mexico, Albuquerque, NM, 87131, USA
| |
Collapse
|
18
|
Stehr M, Kiderlen M, Dorph‐Petersen K. Improving Cavalieri volume estimation based on non-equidistant planar sections: The trapezoidal estimator. J Microsc 2022; 288:40-53. [PMID: 36095148 PMCID: PMC9828659 DOI: 10.1111/jmi.13141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 09/07/2022] [Accepted: 09/08/2022] [Indexed: 01/12/2023]
Abstract
The Cavalieri estimator allows one to infer the volume of an object from area measurements in equidistant planar sections. It is known that applying this estimator in the non-equidistant case may inflate the coefficient of error considerably. We therefore consider a newly introduced variant, the trapezoidal estimator, and make it available to practitioners. Its typical variance behaviour for natural objects is comparable to the equidistant case. We state this unbiased estimator, describe variance estimates and explain how the latter can be simplified under rather general but realistic models for the gaps between sections. Simulations and an application to a synthetic area function based on parietal lobes of 18 monkeys illustrate the new methods.
Collapse
Affiliation(s)
- Mads Stehr
- Department of FinanceCopenhagen Business SchoolFrederiksbergDenmark
| | | | - Karl‐Anton Dorph‐Petersen
- Translational Neuropsychiatry Unit, Department of Clinical MedicineAarhus UniversityAarhusDenmark,Translational Neuroscience Program, Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| |
Collapse
|
19
|
Zhou R, He M, Fan J, Li R, Zuo Y, Li B, Gao G, Sun T. The role of hypothalamic endoplasmic reticulum stress in schizophrenia and antipsychotic-induced weight gain: A narrative review. Front Neurosci 2022; 16:947295. [PMID: 36188456 PMCID: PMC9523121 DOI: 10.3389/fnins.2022.947295] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 08/29/2022] [Indexed: 11/21/2022] Open
Abstract
Schizophrenia (SCZ) is a serious mental illness that affects 1% of people worldwide. SCZ is associated with a higher risk of developing metabolic disorders such as obesity. Antipsychotics are the main treatment for SCZ, but their side effects include significant weight gain/obesity. Despite extensive research, the underlying mechanisms by which SCZ and antipsychotic treatment induce weight gain/obesity remain unclear. Hypothalamic endoplasmic reticulum (ER) stress is one of the most important pathways that modulates inflammation, neuronal function, and energy balance. This review aimed to investigate the role of hypothalamic ER stress in SCZ and antipsychotic-induced weight gain/obesity. Preliminary evidence indicates that SCZ is associated with reduced dopamine D2 receptor (DRD2) signaling, which significantly regulates the ER stress pathway, suggesting the importance of ER stress in SCZ and its related metabolic disorders. Antipsychotics such as olanzapine activate ER stress in hypothalamic neurons. These effects may induce decreased proopiomelanocortin (POMC) processing, increased neuropeptide Y (NPY) and agouti-related protein (AgRP) expression, autophagy, and leptin and insulin resistance, resulting in hyperphagia, decreased energy expenditure, and central inflammation, thereby causing weight gain. By activating ER stress, antipsychotics such as olanzapine activate hypothalamic astrocytes and Toll-like receptor 4 signaling, thereby causing inflammation and weight gain/obesity. Moreover, evidence suggests that antipsychotic-induced ER stress may be related to their antagonistic effects on neurotransmitter receptors such as DRD2 and the histamine H1 receptor. Taken together, ER stress inhibitors could be a potential effective intervention against SCZ and antipsychotic-induced weight gain and inflammation.
Collapse
Affiliation(s)
- Ruqin Zhou
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, China
| | - Meng He
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, China
- *Correspondence: Meng He,
| | - Jun Fan
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, China
| | - Ruoxi Li
- School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yufeng Zuo
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, China
| | - Benben Li
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, China
| | - Guanbin Gao
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan, China
- Guanbin Gao,
| | - Taolei Sun
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, China
- Taolei Sun,
| |
Collapse
|
20
|
Larsen NY, Vihrs N, Møller J, Sporring J, Tan X, Li X, Ji G, Rajkowska G, Sun F, Nyengaard JR. Layer III pyramidal cells in the prefrontal cortex reveal morphological changes in subjects with depression, schizophrenia, and suicide. Transl Psychiatry 2022; 12:363. [PMID: 36064829 PMCID: PMC9445178 DOI: 10.1038/s41398-022-02128-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 08/15/2022] [Accepted: 08/18/2022] [Indexed: 11/09/2022] Open
Abstract
Brodmann Area 46 (BA46) has long been regarded as a hotspot of disease pathology in individuals with schizophrenia (SCH) and major depressive disorder (MDD). Pyramidal neurons in layer III of the Brodmann Area 46 (BA46) project to other cortical regions and play a fundamental role in corticocortical and thalamocortical circuits. The AutoCUTS-LM pipeline was used to study the 3-dimensional structural morphology and spatial organization of pyramidal cells. Using quantitative light microscopy, we used stereology to calculate the entire volume of layer III in BA46 and the total number and density of pyramidal cells. Volume tensors estimated by the planar rotator quantified the volume, shape, and nucleus displacement of pyramidal cells. All of these assessments were carried out in four groups of subjects: controls (C, n = 10), SCH (n = 10), MDD (n = 8), and suicide subjects with a history of depression (SU, n = 11). SCH subjects had a significantly lower somal volume, total number, and density of pyramidal neurons when compared to C and tended to show a volume reduction in layer III of BA46. When comparing MDD subjects with C, the measured parameters were inclined to follow SCH, although there was only a significant reduction in pyramidal total cell number. While no morphometric differences were observed between SU and MDD, SU had a significantly higher total number of pyramidal cells and nucleus displacement than SCH. Finally, no differences in the spatial organization of pyramidal cells were found among groups. These results suggest that despite significant morphological alterations in layer III of BA46, which may impair prefrontal connections in people with SCH and MDD, the spatial organization of pyramidal cells remains the same across the four groups and suggests no defects in neuronal migration. The increased understanding of pyramidal cell biology may provide the cellular basis for symptoms and neuroimaging observations in SCH and MDD patients.
Collapse
Affiliation(s)
- Nick Y. Larsen
- grid.7048.b0000 0001 1956 2722Core Centre for Molecular Morphology, Section for Stereology and Microscopy, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark ,grid.7048.b0000 0001 1956 2722Center of Functionally Integrative Neuroscience, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark ,Sino-Danish Center for Education and Research, Aarhus, Denmark ,grid.410726.60000 0004 1797 8419University of the Chinese Academy of Sciences, Beijing, China ,grid.5117.20000 0001 0742 471XCentre for Stochastic Geometry and Advanced Bioimaging, Aalborg University, Aarhus University and University of Copenhagen, Aarhus, Denmark
| | - Ninna Vihrs
- grid.5117.20000 0001 0742 471XDepartment of Mathematical Sciences, Aalborg University, Aalborg, Denmark
| | - Jesper Møller
- grid.5117.20000 0001 0742 471XCentre for Stochastic Geometry and Advanced Bioimaging, Aalborg University, Aarhus University and University of Copenhagen, Aarhus, Denmark ,grid.5117.20000 0001 0742 471XDepartment of Mathematical Sciences, Aalborg University, Aalborg, Denmark
| | - Jon Sporring
- grid.5117.20000 0001 0742 471XCentre for Stochastic Geometry and Advanced Bioimaging, Aalborg University, Aarhus University and University of Copenhagen, Aarhus, Denmark ,grid.5254.60000 0001 0674 042XDepartment of Computer Science, University of Copenhagen, Copenhagen, Denmark
| | - Xueke Tan
- grid.418856.60000 0004 1792 5640National Key Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China ,grid.418856.60000 0004 1792 5640Center for Biological Imaging, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Xixia Li
- grid.5254.60000 0001 0674 042XDepartment of Computer Science, University of Copenhagen, Copenhagen, Denmark ,grid.418856.60000 0004 1792 5640National Key Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Gang Ji
- grid.5254.60000 0001 0674 042XDepartment of Computer Science, University of Copenhagen, Copenhagen, Denmark ,grid.418856.60000 0004 1792 5640National Key Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Grazyna Rajkowska
- grid.410721.10000 0004 1937 0407Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS USA
| | - Fei Sun
- Sino-Danish Center for Education and Research, Aarhus, Denmark ,grid.410726.60000 0004 1797 8419University of the Chinese Academy of Sciences, Beijing, China ,grid.418856.60000 0004 1792 5640National Key Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China ,grid.418856.60000 0004 1792 5640Center for Biological Imaging, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Jens R. Nyengaard
- grid.7048.b0000 0001 1956 2722Core Centre for Molecular Morphology, Section for Stereology and Microscopy, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark ,Sino-Danish Center for Education and Research, Aarhus, Denmark ,grid.5117.20000 0001 0742 471XCentre for Stochastic Geometry and Advanced Bioimaging, Aalborg University, Aarhus University and University of Copenhagen, Aarhus, Denmark ,grid.154185.c0000 0004 0512 597XDepartment of Pathology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
21
|
Das J, Beyaztas BH, Mac-Ocloo MK, Majumdar A, Mandal A. Testing Equality of Multiple Population Means under Contaminated Normal Model Using the Density Power Divergence. ENTROPY (BASEL, SWITZERLAND) 2022; 24:1189. [PMID: 36141075 PMCID: PMC9497527 DOI: 10.3390/e24091189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/18/2022] [Accepted: 08/21/2022] [Indexed: 06/16/2023]
Abstract
This paper considers the problem of comparing several means under the one-way Analysis of Variance (ANOVA) setup. In ANOVA, outliers and heavy-tailed error distribution can seriously hinder the treatment effect, leading to false positive or false negative test results. We propose a robust test of ANOVA using an M-estimator based on the density power divergence. Compared with the existing robust and non-robust approaches, the proposed testing procedure is less affected by data contamination and improves the analysis. The asymptotic properties of the proposed test are derived under some regularity conditions. The finite-sample performance of the proposed test is examined via a series of Monte-Carlo experiments and two empirical data examples-bone marrow transplant dataset and glucose level dataset. The results produced by the proposed testing procedure are favorably compared with the classical ANOVA and robust tests based on Huber's M-estimator and Tukey's MM-estimator.
Collapse
Affiliation(s)
- Jagannath Das
- Department of Mathematical Sciences, University of Texas at El Paso, El Paso, TX 79968, USA
| | | | | | - Arunabha Majumdar
- Department of Mathematics, Indian Institute of Technology Hyderabad, Kandi 502284, Telangana, India
| | - Abhijit Mandal
- Department of Mathematical Sciences, University of Texas at El Paso, El Paso, TX 79968, USA
| |
Collapse
|
22
|
Liu C, Kim WS, Shen J, Tsogt U, Kang NI, Lee KH, Chung YC. Altered Neuroanatomical Signatures of Patients With Treatment-Resistant Schizophrenia Compared to Patients With Early-Stage Schizophrenia and Healthy Controls. Front Psychiatry 2022; 13:802025. [PMID: 35664476 PMCID: PMC9158464 DOI: 10.3389/fpsyt.2022.802025] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 04/12/2022] [Indexed: 11/25/2022] Open
Abstract
Background The relationship between brain structural changes and cognitive dysfunction in schizophrenia is strong. However, few studies have investigated both neuroanatomical abnormalities and cognitive dysfunction in treatment-resistant schizophrenia (TRS). We examined neuroanatomical markers and cognitive function between patients with TRS or early-stage schizophrenia (ES-S) and healthy controls (HCs). Relationships between neuroanatomical markers and cognitive function in the patient groups were also investigated. Methods A total of 46 and 45 patients with TRS and ES-S and 61 HCs underwent structural magnetic resonance imaging (MRI) brain scanning and comprehensive cognitive tests. MRI scans were analyzed using the FreeSurfer to investigate differences in cortical surface area (CSA), cortical thickness (CT), cortical volume (CV), and subcortical volume (SCV) among the groups. Four cognitive domains (attention, verbal memory, executive function, and language) were assessed. Comparisons of neuroanatomical and cognitive function results among the three groups were performed. Results A widespread reduction in CT was observed in patients with TRS compared to HCs, but differences in cortical thinning between TRS and ES-S patients were mainly limited to the inferior frontal gyrus and insula. Several subcortical structures (accumbens, amygdala, hippocampus, putamen, thalamus and ventricles) were significantly altered in TRS patients compared to both ES-S patients and HCs. Performance in the verbal memory domain was significantly worse in TRS patients compared to ES-S patients. A positive relationship between the thickness of the left middle temporal gyrus and the composite score for language was identified in patients with ES-S. Conclusions Our findings suggest significant cognitive impairment and reductions in CT and SCV in individuals with TRS compared to those with ES-S and HCs. These abnormalities could act as biomarkers for earlier identification of TRS.
Collapse
Affiliation(s)
- Congcong Liu
- Department of Psychiatry, Medical School, Jeonbuk National University, Jeonju, South Korea
| | - Woo-Sung Kim
- Department of Psychiatry, Medical School, Jeonbuk National University, Jeonju, South Korea
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, South Korea
| | - Jie Shen
- Department of Psychiatry, Medical School, Jeonbuk National University, Jeonju, South Korea
| | - Uyanga Tsogt
- Department of Psychiatry, Medical School, Jeonbuk National University, Jeonju, South Korea
| | - Nam-In Kang
- Department of Psychiatry, Maeumsarang Hospital, Wanju, South Korea
| | - Keon-Hak Lee
- Department of Psychiatry, Maeumsarang Hospital, Wanju, South Korea
| | - Young-Chul Chung
- Department of Psychiatry, Medical School, Jeonbuk National University, Jeonju, South Korea
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, South Korea
- Department of Psychiatry, Jeonbuk National University Hospital, Jeonju, South Korea
| |
Collapse
|
23
|
Rothschild AJ. Should Antipsychotic Medications Be Prescribed to Patients With Nonpsychotic Depression? J Clin Psychopharmacol 2022; 42:231-233. [PMID: 35475846 DOI: 10.1097/jcp.0000000000001550] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Affiliation(s)
- Anthony J Rothschild
- From the Department of Psychiatry, University of Massachusetts Chan Medical School and UMass Memorial Health Care, Worcester, MA
| |
Collapse
|
24
|
Murray RM, Bora E, Modinos G, Vernon A. Schizophrenia: A developmental disorder with a risk of non-specific but avoidable decline. Schizophr Res 2022; 243:181-186. [PMID: 35390609 DOI: 10.1016/j.schres.2022.03.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 03/08/2022] [Accepted: 03/11/2022] [Indexed: 12/31/2022]
Abstract
The onset of schizophrenia is determined by biological and social risk factors operating predominantly during development. These result in subtle deviations in brain structure and cognitive function. Striatal dopamine dysregulation follows, causing abnormal salience and resultant psychotic symptoms. Most people diagnosed as having schizophrenia do not progressively deteriorate; many improve or recover. However, poor care can allow a cycle of deterioration to be established, stress increasing dopamine dysregulation, leading to more stress consequent on continuing psychotic experiences, and so further dopamine release. Additionally, long-term antipsychotics can induce dopamine supersensitivity with resultant relapse and eventually treatment resistance. Some patients suffer loss of social and cognitive function, but this is a consequence of the hazards that afflict the person with schizophrenia, not a direct consequence of genetic predisposition. Thus, brain health and cognition can be further impaired by chronic medication effects, cardiovascular and cerebrovascular events, obesity, poor diet, and lack of exercise; drug use, especially of tobacco and cannabis, are likely to contribute. Poverty, homelessness and poor nutrition which become the lot of some people with schizophrenia, can also affect cognition. Regrettably, the model of progressive deterioration provides psychiatry and its funders with an alibi for the effects of poor care.
Collapse
Affiliation(s)
- R M Murray
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, United Kingdom.
| | - E Bora
- Dokuz Eylül Üniversitesi, Izmir, Izmir, Turkey
| | - G Modinos
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, United Kingdom
| | - A Vernon
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, United Kingdom
| |
Collapse
|
25
|
Fountoulakis KN, Stahl SM. The effect of first- and second-generation antipsychotics on brain morphology in schizophrenia: A systematic review of longitudinal magnetic resonance studies with a randomized allocation to treatment arms. J Psychopharmacol 2022; 36:428-438. [PMID: 35395911 DOI: 10.1177/02698811221087645] [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] [Indexed: 11/16/2022]
Abstract
Schizophrenia manifests as loss of brain volume in specific areas in a progressive nature and an important question concerns whether long-term treatment with medications contributes to this. The aim of the current PRISMA systematic review was to search for prospective studies involving randomization to treatment. PROSPERO ID: CRD42020197874. The MEDLINE/PUBMED was searched and it returned 2638 articles; 3 were fulfilling the inclusion criteria. A fourth was published later; they included 359 subjects, of whom 86 were healthy controls, while the rest were first-episode patients, with 91 under olanzapine, 93 under haloperidol, 48 under risperidone, 5 under paliperidone, 6 under ziprasidone, and 30 under placebo. Probably one-third of patients were suffering from a psychotic disorder other than schizophrenia. The consideration of their results suggested that there is no significant difference between these medications concerning their effects on brain structure and also in comparison to healthy subjects. There does not seem to be any strong support to the opinion that medications that treat psychosis cause loss of brain volume in patients with schizophrenia. On the contrary, the data might imply the possible presence of a protective effect for D2, 5-HT2, and NE alpha-2 antagonists (previously called SGAs). However, the literature is limited and focused research in large study samples is essential to clarify the issue, since important numerical differences do exist. The possibility of the results and their heterogeneity to be artifacts secondary to a modification of magnetic resonance imaging (MRI) signal by antipsychotics should not be easily rejected until relevant data are available.
Collapse
Affiliation(s)
- Konstantinos N Fountoulakis
- 3rd Department of Psychiatry, Faculty of Medicine, School of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Stephen M Stahl
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA.,Department of Psychiatry, Cambridge University, Cambridge, UK
| |
Collapse
|
26
|
Krajner F, Hadaya L, McQueen G, Sendt KV, Gillespie A, Avila A, Lally J, Hedges EP, Diederen K, Howes OD, Barker GJ, Lythgoe DJ, Kempton MJ, McGuire P, MacCabe JH, Egerton A. Subcortical volume reduction and cortical thinning 3 months after switching to clozapine in treatment resistant schizophrenia. SCHIZOPHRENIA (HEIDELBERG, GERMANY) 2022; 8:13. [PMID: 35236831 PMCID: PMC8891256 DOI: 10.1038/s41537-022-00230-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 12/06/2021] [Indexed: 11/29/2022]
Abstract
The neurobiological effects of clozapine are under characterised. We examined the effects clozapine treatment on subcortical volume and cortical thickness and investigated whether macrostructural changes were linked to alterations in glutamate or N-acetylaspartate (NAA). Data were acquired in 24 patients with treatment-resistant schizophrenia before and 12 weeks after switching to clozapine. During clozapine treatment we observed reductions in caudate and putamen volume, lateral ventricle enlargement (P < 0.001), and reductions in thickness of the left inferior temporal cortex, left caudal middle frontal cortex, and the right temporal pole. Reductions in right caudate volume were associated with local reductions in NAA (P = 0.002). None of the morphometric changes were associated with changes in glutamate levels. These results indicate that clozapine treatment is associated with subcortical volume loss and cortical thinning and that at least some of these effects are linked to changes in neuronal or metabolic integrity.
Collapse
Affiliation(s)
- Fanni Krajner
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London, UK
| | - Laila Hadaya
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London, UK
| | - 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
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, 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
| | - Emily P Hedges
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London, UK
| | - Kelly Diederen
- 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
- South London and Maudsley NHS Trust, London, UK
| | - Gareth J Barker
- Department of Neuroimaging, Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London, UK
| | - David J Lythgoe
- Department of Neuroimaging, Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London, UK
| | - Matthew J Kempton
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London, UK
| | - Philip McGuire
- 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
| | - James H MacCabe
- 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
| | - Alice Egerton
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London, UK.
| |
Collapse
|
27
|
Smucny J, Dienel SJ, Lewis DA, Carter CS. Mechanisms underlying dorsolateral prefrontal cortex contributions to cognitive dysfunction in schizophrenia. Neuropsychopharmacology 2022; 47:292-308. [PMID: 34285373 PMCID: PMC8617156 DOI: 10.1038/s41386-021-01089-0] [Citation(s) in RCA: 106] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 06/25/2021] [Accepted: 06/28/2021] [Indexed: 02/07/2023]
Abstract
Kraepelin, in his early descriptions of schizophrenia (SZ), characterized the illness as having "an orchestra without a conductor." Kraepelin further speculated that this "conductor" was situated in the frontal lobes. Findings from multiple studies over the following decades have clearly implicated pathology of the dorsolateral prefrontal cortex (DLPFC) as playing a central role in the pathophysiology of SZ, particularly with regard to key cognitive features such as deficits in working memory and cognitive control. Following an overview of the cognitive mechanisms associated with DLPFC function and how they are altered in SZ, we review evidence from an array of neuroscientific approaches addressing how these cognitive impairments may reflect the underlying pathophysiology of the illness. Specifically, we present evidence suggesting that alterations of the DLPFC in SZ are evident across a range of spatial and temporal resolutions: from its cellular and molecular architecture, to its gross structural and functional integrity, and from millisecond to longer timescales. We then present an integrative model based upon how microscale changes in neuronal signaling in the DLPFC can influence synchronized patterns of neural activity to produce macrocircuit-level alterations in DLPFC activation that ultimately influence cognition and behavior. We conclude with a discussion of initial efforts aimed at targeting DLPFC function in SZ, the clinical implications of those efforts, and potential avenues for future development.
Collapse
Affiliation(s)
- Jason Smucny
- Department of Psychiatry and Behavioral Sciences, University of California Davis Medical Center, Sacramento, CA, USA
- Center for Neuroscience, University of California Davis, Davis, CA, USA
| | - Samuel J Dienel
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| | - David A Lewis
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Cameron S Carter
- Department of Psychiatry and Behavioral Sciences, University of California Davis Medical Center, Sacramento, CA, USA.
- Center for Neuroscience, University of California Davis, Davis, CA, USA.
| |
Collapse
|
28
|
Kanahara N, Yamanaka H, Shiko Y, Kawasaki Y, Iyo M. The effects of cumulative antipsychotic dose on brain structures in patients with schizophrenia: Observational study of multiple CT scans over a long-term clinical course. Psychiatry Res Neuroimaging 2022; 319:111422. [PMID: 34856453 DOI: 10.1016/j.pscychresns.2021.111422] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 10/23/2021] [Accepted: 10/28/2021] [Indexed: 10/19/2022]
Abstract
Multiple lines of evidence indicate that antipsychotic agents could affect brain structures of schizophrenia patients. However, the effect of antipsychotic dosage or type on brain structure is uncertain. The present study retrospectively analyzed brain computed tomography (CT) images from a psychiatric hospital to examine the relationship between cumulative dose of antipsychotics and brain volume reduction in schizophrenia patients. A total of 43 patients with repeated relapse episode of psychosis were included and CT scans that were performed an average of 3.2 times per patient during nearly 13 years of follow-up were analyzed. The results revealed significant positive relationships of expansion of cerebrospinal fluid space with cumulative dosage of all antipsychotics and that of typical antipsychotics. Patients treated with antipsychotics including typical antipsychotics exhibited a greater volume reduction compared to patients treated with only atypical antipsychotics. The present study was one of the longest longitudinal studies examining the effects of antipsychotics on brain volume in schizophrenia patients. These results suggest a relation between cumulative lifetime antipsychotic dosage and progressive brain volume reduction in patients with schizophrenia. However, the effects of specific agent on brain structure are still uncertain, and more detailed analysis is needed.
Collapse
Affiliation(s)
- Nobuhisa Kanahara
- Division of Medical Treatment and Rehabilitation, Center for Forensic Mental Health, Chiba University, Chiba, Japan; Department of Psychiatry, Chiba University Graduate School of Medicine, Chiba, Japan.
| | - Hiroshi Yamanaka
- Department of Psychiatry, Chiba Psychiatric Medical Center, Chiba, Japan
| | - Yuki Shiko
- Biostatistics Section, Clinical Research Center, Chiba University Hospital, Chiba, Japan
| | - Yohei Kawasaki
- Biostatistics Section, Clinical Research Center, Chiba University Hospital, Chiba, Japan
| | - Masaomi Iyo
- Department of Psychiatry, Chiba University Graduate School of Medicine, Chiba, Japan
| |
Collapse
|
29
|
Seney ML, Glausier J, Sibille E. Large-Scale Transcriptomics Studies Provide Insight Into Sex Differences in Depression. Biol Psychiatry 2022; 91:14-24. [PMID: 33648716 PMCID: PMC8263802 DOI: 10.1016/j.biopsych.2020.12.025] [Citation(s) in RCA: 39] [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: 10/24/2020] [Revised: 12/17/2020] [Accepted: 12/18/2020] [Indexed: 12/11/2022]
Abstract
Major depressive disorder (MDD) is a leading cause of disability, affecting more than 300 million people worldwide. We first review the well-known sex difference in incidence of MDD, with women being twice as likely to be diagnosed as men, and briefly summarize how the impact of MDD varies between men and women, with sex differences in symptoms, severity, and antidepressant drug response. We then attempt to deconstruct the biological bases for MDD and discuss implications for sex differences research. Next, we review findings from human postmortem studies, both from selected candidate gene studies and from well-powered, unbiased transcriptomics studies, which suggest distinct, and possibly opposite, molecular changes in the brains of depressed men and women. We then discuss inherent challenges of research on the human postmortem brain and suggest paths forward that rely on thoughtful cohort design. Although studies indicate that circulating gonadal hormones might underlie the observed sex differences in MDD, we discuss how additional sex-specific factors, such as genetic sex and developmental exposure to gonadal hormones, may also contribute to altered vulnerability, and we highlight various nuances that we believe should be considered when determining mechanisms underlying observed sex differences. Altogether, this review highlights not only how various sex-specific factors might influence susceptibility or resilience to depression, but also how those sex-specific factors might result in divergent pathology in men and women.
Collapse
Affiliation(s)
- Marianne L Seney
- Department of Psychiatry, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania; Translational Neuroscience Program, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania.
| | - Jill Glausier
- Department of Psychiatry, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania; Translational Neuroscience Program, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania
| | - Etienne Sibille
- Campbell Family Mental Health Research Institute at the Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
30
|
Gutman BA, van Erp TG, Alpert K, Ching CRK, Isaev D, Ragothaman A, Jahanshad N, Saremi A, Zavaliangos‐Petropulu A, Glahn DC, Shen L, Cong S, Alnæs D, Andreassen OA, Doan NT, Westlye LT, Kochunov P, Satterthwaite TD, Wolf DH, Huang AJ, Kessler C, Weideman A, Nguyen D, Mueller BA, Faziola L, Potkin SG, Preda A, Mathalon DH, Bustillo J, Calhoun V, Ford JM, Walton E, Ehrlich S, Ducci G, Banaj N, Piras F, Piras F, Spalletta G, Canales‐Rodríguez EJ, Fuentes‐Claramonte P, Pomarol‐Clotet E, Radua J, Salvador R, Sarró S, Dickie EW, Voineskos A, Tordesillas‐Gutiérrez D, Crespo‐Facorro B, Setién‐Suero E, van Son JM, Borgwardt S, Schönborn‐Harrisberger F, Morris D, Donohoe G, Holleran L, Cannon D, McDonald C, Corvin A, Gill M, Filho GB, Rosa PGP, Serpa MH, Zanetti MV, Lebedeva I, Kaleda V, Tomyshev A, Crow T, James A, Cervenka S, Sellgren CM, Fatouros‐Bergman H, Agartz I, Howells F, Stein DJ, Temmingh H, Uhlmann A, de Zubicaray GI, McMahon KL, Wright M, Cobia D, Csernansky JG, Thompson PM, Turner JA, Wang L. A meta-analysis of deep brain structural shape and asymmetry abnormalities in 2,833 individuals with schizophrenia compared with 3,929 healthy volunteers via the ENIGMA Consortium. Hum Brain Mapp 2022; 43:352-372. [PMID: 34498337 PMCID: PMC8675416 DOI: 10.1002/hbm.25625] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/13/2021] [Accepted: 07/14/2021] [Indexed: 01/06/2023] Open
Abstract
Schizophrenia is associated with widespread alterations in subcortical brain structure. While analytic methods have enabled more detailed morphometric characterization, findings are often equivocal. In this meta-analysis, we employed the harmonized ENIGMA shape analysis protocols to collaboratively investigate subcortical brain structure shape differences between individuals with schizophrenia and healthy control participants. The study analyzed data from 2,833 individuals with schizophrenia and 3,929 healthy control participants contributed by 21 worldwide research groups participating in the ENIGMA Schizophrenia Working Group. Harmonized shape analysis protocols were applied to each site's data independently for bilateral hippocampus, amygdala, caudate, accumbens, putamen, pallidum, and thalamus obtained from T1-weighted structural MRI scans. Mass univariate meta-analyses revealed more-concave-than-convex shape differences in the hippocampus, amygdala, accumbens, and thalamus in individuals with schizophrenia compared with control participants, more-convex-than-concave shape differences in the putamen and pallidum, and both concave and convex shape differences in the caudate. Patterns of exaggerated asymmetry were observed across the hippocampus, amygdala, and thalamus in individuals with schizophrenia compared to control participants, while diminished asymmetry encompassed ventral striatum and ventral and dorsal thalamus. Our analyses also revealed that higher chlorpromazine dose equivalents and increased positive symptom levels were associated with patterns of contiguous convex shape differences across multiple subcortical structures. Findings from our shape meta-analysis suggest that common neurobiological mechanisms may contribute to gray matter reduction across multiple subcortical regions, thus enhancing our understanding of the nature of network disorganization in schizophrenia.
Collapse
Affiliation(s)
- Boris A. Gutman
- Department of Biomedical EngineeringIllinois Institute of TechnologyChicagoIllinoisUSA
- Institute for Information Transmission Problems (Kharkevich Institute)MoscowRussia
| | - Theo G.M. van Erp
- Clinical Translational Neuroscience Laboratory, Department of Psychiatry and Human BehaviorUniversity of California IrvineIrvineCaliforniaUSA
- Center for the Neurobiology of Learning and MemoryUniversity of California IrvineIrvineCaliforniaUSA
| | - Kathryn Alpert
- Department of Psychiatry and Behavioral SciencesNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Christopher R. K. Ching
- Imaging Genetics Center, Mark & Mary Stevens Neuroimaging & Informatics Institute, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Dmitry Isaev
- Department of Biomedical EngineeringDuke UniversityDurhamNorth CarolinaUSA
| | - Anjani Ragothaman
- Department of biomedical engineeringOregon Health and Science universityPortlandOregonUSA
| | - Neda Jahanshad
- Imaging Genetics Center, Mark & Mary Stevens Neuroimaging & Informatics Institute, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Arvin Saremi
- Imaging Genetics Center, Mark & Mary Stevens Neuroimaging & Informatics Institute, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Artemis Zavaliangos‐Petropulu
- Imaging Genetics Center, Mark & Mary Stevens Neuroimaging & Informatics Institute, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - David C. Glahn
- Department of PsychiatryBoston Children's Hospital and Harvard Medical SchoolBostonMassachusettsUSA
| | - Li Shen
- Department of Biostatistics, Epidemiology and InformaticsUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Shan Cong
- Department of Biostatistics, Epidemiology and InformaticsUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Dag Alnæs
- NORMENT, Division of Mental Health and AddictionOslo University Hospital & Institute of Clinical Medicine, University of OsloOsloNorway
| | - Ole Andreas Andreassen
- NORMENT, Division of Mental Health and AddictionOslo University Hospital & Institute of Clinical Medicine, University of OsloOsloNorway
| | - Nhat Trung Doan
- NORMENT, Division of Mental Health and AddictionOslo University Hospital & Institute of Clinical Medicine, University of OsloOsloNorway
| | - Lars T. Westlye
- NORMENT, Division of Mental Health and AddictionOslo University Hospital & Institute of Clinical Medicine, University of OsloOsloNorway
- Department of PsychologyUniversity of OsloOsloNorway
| | - Peter Kochunov
- Department of PsychiatryUniversity of Maryland School of MedicineBaltimoreMarylandUSA
| | - Theodore D. Satterthwaite
- Department of PsychiatryUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUSA
| | - Daniel H. Wolf
- Department of PsychiatryUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUSA
| | - Alexander J. Huang
- Clinical Translational Neuroscience Laboratory, Department of Psychiatry and Human BehaviorUniversity of California IrvineIrvineCaliforniaUSA
| | - Charles Kessler
- Clinical Translational Neuroscience Laboratory, Department of Psychiatry and Human BehaviorUniversity of California IrvineIrvineCaliforniaUSA
| | - Andrea Weideman
- Clinical Translational Neuroscience Laboratory, Department of Psychiatry and Human BehaviorUniversity of California IrvineIrvineCaliforniaUSA
| | - Dana Nguyen
- Department of PediatricsUniversity of California IrvineIrvineCaliforniaUSA
| | - Bryon A. Mueller
- Department of Psychiatry and Behavioral SciencesUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Lawrence Faziola
- Department of Psychiatry and Human BehaviorUniversity of California IrvineIrvineCaliforniaUSA
| | - Steven G. Potkin
- Department of Psychiatry and Human BehaviorUniversity of California IrvineIrvineCaliforniaUSA
| | - Adrian Preda
- Department of Psychiatry and Human BehaviorUniversity of California IrvineIrvineCaliforniaUSA
| | - Daniel H. Mathalon
- Department of Psychiatry and Weill Institute for NeurosciencesUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Judith Ford Mental HealthVA San Francisco Healthcare SystemSan FranciscoCaliforniaUSA
| | - Juan Bustillo
- Departments of Psychiatry & NeuroscienceUniversity of New MexicoAlbuquerqueNew MexicoUSA
| | - Vince Calhoun
- Tri‐institutional Center for Translational Research in Neuroimaging and Data Science (TReNDS) [Georgia State University, Georgia Institute of Technology]Emory UniversityAtlantaGeorgiaUSA
- Department of Electrical and Computer EngineeringThe University of New MexicoAlbuquerqueNew MexicoUSA
| | - Judith M. Ford
- Judith Ford Mental HealthVA San Francisco Healthcare SystemSan FranciscoCaliforniaUSA
- Department of Psychiatry and Behavioral SciencesUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | | | - Stefan Ehrlich
- Division of Psychological & Social Medicine and Developmental NeurosciencesFaculty of Medicine, TU‐DresdenDresdenGermany
| | | | - Nerisa Banaj
- Laboratory of NeuropsychiatryIRCCS Santa Lucia FoundationRomeItaly
| | - Fabrizio Piras
- Laboratory of NeuropsychiatryIRCCS Santa Lucia FoundationRomeItaly
| | - Federica Piras
- Laboratory of NeuropsychiatryIRCCS Santa Lucia FoundationRomeItaly
| | - Gianfranco Spalletta
- Laboratory of NeuropsychiatryIRCCS Santa Lucia FoundationRomeItaly
- Menninger Department of Psychiatry and Behavioral SciencesBaylor College of MedicineHoustonTexasUSA
| | | | | | | | - Joaquim Radua
- FIDMAG Germanes Hospitalàries Research FoundationCIBERSAMBarcelonaSpain
- Institut d'Investigacions Biomdiques August Pi i Sunyer (IDIBAPS)BarcelonaSpain
| | - Raymond Salvador
- FIDMAG Germanes Hospitalàries Research FoundationCIBERSAMBarcelonaSpain
| | - Salvador Sarró
- FIDMAG Germanes Hospitalàries Research FoundationCIBERSAMBarcelonaSpain
| | - Erin W. Dickie
- Centre for Addiction and Mental Health (CAMH)TorontoCanada
| | | | | | | | | | | | - Stefan Borgwardt
- Department of PsychiatryUniversity of BaselBaselSwitzerland
- Department of Psychiatry and PsychotherapyUniversity of LübeckLübeckGermany
| | | | - Derek Morris
- Centre for Neuroimaging and Cognitive Genomics, Discipline of BiochemistryNational University of Ireland GalwayGalwayIreland
| | - Gary Donohoe
- Centre for Neuroimaging and Cognitive Genomics, School of PsychologyNational University of Ireland GalwayGalwayIreland
| | - Laurena Holleran
- Centre for Neuroimaging and Cognitive Genomics, School of PsychologyNational University of Ireland GalwayGalwayIreland
| | - Dara Cannon
- Clinical Neuroimaging Laboratory, Centre for Neuroimaging and Cognitive GenomicsNational University of Ireland GalwayGalwayIreland
| | - Colm McDonald
- Clinical Neuroimaging Laboratory, Centre for Neuroimaging and Cognitive GenomicsNational University of Ireland GalwayGalwayIreland
| | - Aiden Corvin
- Neuropsychiatric Genetics Research Group, Department of PsychiatryTrinity College DublinDublinIreland
- Trinity College Institute of NeuroscienceTrinity College DublinDublinIreland
| | - Michael Gill
- Neuropsychiatric Genetics Research Group, Department of PsychiatryTrinity College DublinDublinIreland
- Trinity College Institute of NeuroscienceTrinity College DublinDublinIreland
| | - Geraldo Busatto Filho
- Laboratory of Psychiatric Neuroimaging (LIM‐21), Departamento e Instituto de PsiquiatriaHospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao PauloSao PauloSPBrazil
| | - Pedro G. P. Rosa
- Laboratory of Psychiatric Neuroimaging (LIM‐21), Departamento e Instituto de PsiquiatriaHospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao PauloSao PauloSPBrazil
| | - Mauricio H. Serpa
- Laboratory of Psychiatric Neuroimaging (LIM‐21), Departamento e Instituto de PsiquiatriaHospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao PauloSao PauloSPBrazil
| | - Marcus V. Zanetti
- Laboratory of Psychiatric Neuroimaging (LIM‐21), Departamento e Instituto de PsiquiatriaHospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao PauloSao PauloSPBrazil
- Hospital Sirio‐LibanesSao PauloSPBrazil
| | - Irina Lebedeva
- Laboratory of Neuroimaging and Multimodal AnalysisMental Health Research CenterMoscowRussia
| | - Vasily Kaleda
- Department of Endogenous Mental DisordersMental Health Research CenterMoscowRussia
| | - Alexander Tomyshev
- Laboratory of Neuroimaging and Multimodal AnalysisMental Health Research CenterMoscowRussia
| | - Tim Crow
- Department of PsychiatryUniversity of OxfordOxfordUK
| | - Anthony James
- Department of PsychiatryUniversity of OxfordOxfordUK
| | - Simon Cervenka
- Centre for Psychiatry Reserach, Department of Clinical NeuroscienceKarolinska Institutet, & Stockholm Health Care Services, Region StockholmStockholmSweden
| | - Carl M Sellgren
- Department of Physiology and PharmacologyKarolinska InstitutetStockholmSweden
| | - Helena Fatouros‐Bergman
- Centre for Psychiatry Reserach, Department of Clinical NeuroscienceKarolinska Institutet, & Stockholm Health Care Services, Region StockholmStockholmSweden
| | - Ingrid Agartz
- NORMENT, Division of Mental Health and AddictionOslo University Hospital & Institute of Clinical Medicine, University of OsloOsloNorway
| | - Fleur Howells
- Department of Psychiatry and Mental Health, Faculty of Health SciencesUniversity of Cape TownCape TownWCSouth Africa
- Neuroscience InstituteUniversity of Cape Town, Cape TownWCSouth Africa
| | - Dan J. Stein
- Department of Psychiatry and Mental Health, Faculty of Health SciencesUniversity of Cape TownCape TownWCSouth Africa
- Neuroscience InstituteUniversity of Cape Town, Cape TownWCSouth Africa
- SA MRC Unit on Risk & Resilience in Mental DisordersUniversity of Cape TownCape TownWCSouth Africa
| | - Henk Temmingh
- Department of Psychiatry and Mental Health, Faculty of Health SciencesUniversity of Cape TownCape TownWCSouth Africa
| | - Anne Uhlmann
- Department of Psychiatry and Mental Health, Faculty of Health SciencesUniversity of Cape TownCape TownWCSouth Africa
- Department of Child and Adolescent PsychiatryTU DresdenGermany
| | - Greig I. de Zubicaray
- School of Psychology, Faculty of HealthQueensland University of Technology (QUT)BrisbaneQLDAustralia
| | - Katie L. McMahon
- School of Clinical SciencesQueensland University of Technology (QUT)BrisbaneQLDAustralia
| | - Margie Wright
- Queensland Brain InstituteUniversity of QueenslandBrisbaneQLDAustralia
| | - Derin Cobia
- Department of Psychiatry and Behavioral SciencesNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
- Department of Psychology and Neuroscience CenterBrigham Young UniversityProvoUtahUSA
| | - John G. Csernansky
- Department of Psychiatry and Behavioral SciencesNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Paul M. Thompson
- Imaging Genetics Center, Mark & Mary Stevens Neuroimaging & Informatics Institute, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | | | - Lei Wang
- Department of Psychiatry and Behavioral SciencesNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
- Department of Psychiatry and Behavioral HealthOhio State University Wexner Medical CenterColumbusOhioUSA
| |
Collapse
|
31
|
Mach J, Gemikonakli G, Logan C, Vander Wyk B, Allore H, Ekambareshwar S, Kane AE, Howlett SE, de Cabo R, Le Couteur DG, Hilmer SN. Chronic Polypharmacy with Increasing Drug Burden Index Exacerbates Frailty and Impairs Physical Function, with Effects Attenuated by Deprescribing, in Aged Mice. J Gerontol A Biol Sci Med Sci 2021; 76:1010-1018. [PMID: 32147704 DOI: 10.1093/gerona/glaa060] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Indexed: 12/15/2022] Open
Abstract
Polypharmacy (use of ≥5 medications) and increasing Drug Burden Index (DBI) score (measure of person's total exposure to anticholinergic/sedative medications) are associated with impaired physical function in observational studies of older adults. Deprescribing, the supervised withdrawal of medications for which harms outweigh benefits for an individual, may be a useful intervention. Current knowledge is limited to clinical observational studies that are unable to determine causality. Here, we establish a preclinical model that investigates the effects of chronic polypharmacy, increasing DBI, and deprescribing on global health outcomes in aging. In a longitudinal study, middle-aged (12 months) male C57BL/6J (B6) mice were administered control feed or feed and/or water containing polypharmacy or monotherapy with different DBI scores. At 21 months, each treatment group was subdivided (stratified by frailty at 21 months) to either continue on treatment for life or to have treatment withdrawn (deprescribed). Frailty and physical function were evaluated at 12, 15, 18, and 24 months, and were analyzed using a mixed modeling approach. Polypharmacy with increasing DBI and monotherapy with citalopram caused mice to become frailer, less mobile, and impaired their strength and functional activities. Critically, deprescribing in old age reversed a number of these outcomes. This is the first preclinical study to demonstrate that chronic polypharmacy with increasing DBI augments frailty and impairs function in old age, and that drug withdrawal in old age reversed these outcomes. It was not the number of drugs (polypharmacy) but the type and dose of drugs (DBI) that caused adverse geriatric outcomes.
Collapse
Affiliation(s)
- John Mach
- Laboratory of Ageing and Pharmacology, Kolling Institute of Medical Research, Royal North Shore Hospital and University of Sydney, St Leonards, New South Wales, Australia
- Departments of Clinical Pharmacology and Aged Care, Royal North Shore Hospital, St Leonards, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, New South Wales, Australia
| | - Gizem Gemikonakli
- Laboratory of Ageing and Pharmacology, Kolling Institute of Medical Research, Royal North Shore Hospital and University of Sydney, St Leonards, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, New South Wales, Australia
| | - Caitlin Logan
- Laboratory of Ageing and Pharmacology, Kolling Institute of Medical Research, Royal North Shore Hospital and University of Sydney, St Leonards, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, New South Wales, Australia
| | - Brent Vander Wyk
- Department of Internal Medicine, Yale University, New Haven, Connecticut
| | - Heather Allore
- Department of Internal Medicine, Yale University, New Haven, Connecticut
- Department of Biostatistics, Yale School of Public Health, New Haven, Connecticut
| | - Swathi Ekambareshwar
- Laboratory of Ageing and Pharmacology, Kolling Institute of Medical Research, Royal North Shore Hospital and University of Sydney, St Leonards, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, New South Wales, Australia
| | - Alice E Kane
- Faculty of Medicine and Health, The University of Sydney, New South Wales, Australia
- School of Life and Environmental Sciences, Harvard Medical School, Boston, Massachusetts
| | - Susan E Howlett
- Departments of Pharmacology and Medicine (Geriatric Medicine), Dalhousie University, Halifax, Canada
| | - Rafael de Cabo
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - David G Le Couteur
- Faculty of Medicine and Health, The University of Sydney, New South Wales, Australia
- Ageing and Alzhiemers Institute (AAAI), Centre for Education and Research on Ageing (CERA) and ANZAC Research Institute, Concord Hospital, Sydney, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, New South Wales, Australia
| | - Sarah N Hilmer
- Laboratory of Ageing and Pharmacology, Kolling Institute of Medical Research, Royal North Shore Hospital and University of Sydney, St Leonards, New South Wales, Australia
- Departments of Clinical Pharmacology and Aged Care, Royal North Shore Hospital, St Leonards, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, New South Wales, Australia
| |
Collapse
|
32
|
Xiao Y, Liao W, Long Z, Tao B, Zhao Q, Luo C, Tamminga CA, Keshavan MS, Pearlson GD, Clementz BA, Gershon ES, Ivleva EI, Keedy SK, Biswal BB, Mechelli A, Lencer R, Sweeney JA, Lui S, Gong Q. Subtyping Schizophrenia Patients Based on Patterns of Structural Brain Alterations. Schizophr Bull 2021; 48:241-250. [PMID: 34508358 PMCID: PMC8781382 DOI: 10.1093/schbul/sbab110] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Schizophrenia is a complex and heterogeneous syndrome. Whether quantitative imaging biomarkers can identify discrete subgroups of patients as might be used to foster personalized medicine approaches for patient care remains unclear. Cross-sectional structural MR images of 163 never-treated first-episode schizophrenia patients (FES) and 133 chronically ill patients with midcourse schizophrenia from the Bipolar and Schizophrenia Network for Intermediate Phenotypes (B-SNIP) consortium and a total of 403 healthy controls were recruited. Morphometric measures (cortical thickness, surface area, and subcortical structures) were extracted for each subject and then the optimized subtyping results were obtained with nonsupervised cluster analysis. Three subgroups of patients defined by distinct patterns of regional cortical and subcortical morphometric features were identified in FES. A similar three subgroup pattern was identified in the independent dataset of patients from the multi-site B-SNIP consortium. Similarities of classification patterns across these two patient cohorts suggest that the 3-group typology is relatively stable over the course of illness. Cognitive functions were worse in subgroup 1 with midcourse schizophrenia than those in subgroup 3. These findings provide novel insight into distinct subgroups of patients with schizophrenia based on structural brain features. Findings of different cognitive functions among the subgroups support clinical differences in the MRI-defined illness subtypes. Regardless of clinical presentation and stage of illness, anatomic MR subgrouping biomarkers can separate neurobiologically distinct subgroups of schizophrenia patients, which represent an important and meaningful step forward in differentiating subtypes of patients for studies of illness neurobiology and potentially for clinical trials.
Collapse
Affiliation(s)
- Yuan Xiao
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, Sichuan, China,Department of Psychiatry, University of Münster, Münster, Germany
| | - Wei Liao
- Center for Information in BioMedicine, School of Life Science and Technology, University of Electronic Science and Technology, Chengdu, Sichuan, China
| | - Zhiliang Long
- Center for Information in BioMedicine, School of Life Science and Technology, University of Electronic Science and Technology, Chengdu, Sichuan, China
| | - Bo Tao
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Qiannan Zhao
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Chunyan Luo
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Carol A Tamminga
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Matcheri S Keshavan
- Department of Psychiatry, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Godfrey D Pearlson
- Departments of Psychiatry and Neurobiology, Yale University and Olin Neuropsychiatric Research Center, Hartford, CT, USA
| | - Brett A Clementz
- Department of Psychology, University of Georgia, Athens, GA, USA
| | - Elliot S Gershon
- Department of Psychiatry, University of Chicago, Chicago, IL, USA
| | - Elena I Ivleva
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Sarah K Keedy
- Department of Psychiatry, University of Chicago, Chicago, IL, USA
| | - Bharat B Biswal
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Andrea Mechelli
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
| | - Rebekka Lencer
- Department of Psychiatry, University of Münster, Münster, Germany
| | - John A Sweeney
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, Sichuan, China,Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH, USA
| | - Su Lui
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, Sichuan, China,To whom correspondence should be addressed; #37 GuoXue Xiang, Chengdu 610041, China; Tel: 86-28-85423960, Fax: 86-28-85423503; e-mail:
| | - Qiyong Gong
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
33
|
Affiliation(s)
| | - Bente Pakkenberg
- Research Laboratory for Stereology and Neuroscience, Bispebjerg Hospital, Copenhagen, Denmark
| | - Adrian Baddeley
- School of Electrical Engineering, Computing and Mathematical Sciences, Curtin University, Perth, Australia
| |
Collapse
|
34
|
Ochoa ELM. Lithium as a Neuroprotective Agent for Bipolar Disorder: An Overview. Cell Mol Neurobiol 2021; 42:85-97. [PMID: 34357564 DOI: 10.1007/s10571-021-01129-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 07/14/2021] [Indexed: 11/28/2022]
Abstract
Lithium (Li+) is a first option treatment for adult acute episodes of Bipolar Disorder (BD) and for the prophylaxis of new depressed or manic episodes. It is also the preferred choice as maintenance treatment. Numerous studies have shown morphological abnormalities in the brains of BD patients, suggesting that this highly heritable disorder may exhibit progressive and deleterious changes in brain structure. Since treatment with Li+ ameliorates these abnormalities, it has been postulated that Li+ is a neuroprotective agent in the same way atypical antipsychotics are neuroprotective in patients diagnosed with schizophrenia spectrum disorders. Li+'s neuroprotective properties are related to its modulation of nerve growth factors, inflammation, mitochondrial function, oxidative stress, and programmed cell death mechanisms such as autophagy and apoptosis. Notwithstanding, it is not known whether Li+-induced neuroprotection is related to the inhibition of its putative molecular targets in a BD episode: the enzymes inositol-monophosphatase, (IMPase), glycogen-synthase-kinase 3β (GSK3), and Protein kinase C (PKC). Furthermore, it is uncertain whether these neuroprotective mechanisms are correlated with Li+'s clinical efficacy in maintaining mood stability. It is expected that in a nearby future, precision medicine approaches will improve diagnosis and expand treatment options. This will certainly contribute to ameliorating the medical and economic burden created by this devastating mood disorder.
Collapse
Affiliation(s)
- Enrique L M Ochoa
- Department of Psychiatry and Behavioral Sciences, Volunteer Clinical Faculty, University of California at Davis, 2230 Stockton Boulevard, Sacramento, CA, 95817, USA.
| |
Collapse
|
35
|
Jiang Y, Wang Y, Huang H, He H, Tang Y, Su W, Xu L, Wei Y, Zhang T, Hu H, Wang J, Yao D, Wang J, Luo C. Antipsychotics Effects on Network-Level Reconfiguration of Cortical Morphometry in First-Episode Schizophrenia. Schizophr Bull 2021; 48:231-240. [PMID: 34313782 PMCID: PMC8781340 DOI: 10.1093/schbul/sbab082] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Cortical thickness reductions are evident in schizophrenia (SZ). Associations between antipsychotic medications (APMs) and cortical morphometry have been explored in SZ patients. This raises the question of whether the reconfiguration of morphological architecture by APM plays potential compensatory roles for abnormalities in the cerebral cortex. Structural magnetic resonance imaging was obtained from 127 medication-naive first-episode SZ patients and 133 matched healthy controls. Patients received 12 weeks of APM and were categorized as responders (n = 75) or nonresponders (NRs, n = 52) at follow-up. Using surface-based morphometry and structural covariance (SC) analysis, this study investigated the short-term effects of antipsychotics on cortical thickness and cortico-cortical covariance. Global efficiency was computed to characterize network integration of the large-scale structural connectome. The relationship between covariance and cortical thinning was examined by SC analysis among the top-n regions with thickness reduction. Widespread cortical thickness reductions were observed in pre-APM patients. Post-APM patients showed more reductions in cortical thickness, even in the frontotemporal regions without baseline reductions. Covariance analysis revealed strong cortico-cortical covariance and higher network integration in responders than in NRs. For the NRs, some of the prefrontal and temporal nodes were not covariant between the top-n regions with cortical thickness reduction. Antipsychotic effects are not restricted to a single brain region but rather exhibit a network-level covariance pattern. Neuroimaging connectomics highlights the positive effects of antipsychotics on the reconfiguration of brain architecture, suggesting that abnormalities in regional morphology may be compensated by increasing interregional covariance when symptoms are controlled by antipsychotics.
Collapse
Affiliation(s)
- Yuchao Jiang
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, Center for Information in Medicine, University of Electronic Science and Technology of China, Chengdu, PR China,High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, PR China
| | - Yingchan Wang
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Huan Huang
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, Center for Information in Medicine, University of Electronic Science and Technology of China, Chengdu, PR China,High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, PR China
| | - Hui He
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, Center for Information in Medicine, University of Electronic Science and Technology of China, Chengdu, PR China
| | - Yingying Tang
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Wenjun Su
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Lihua Xu
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Yanyan Wei
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Tianhong Zhang
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Hao Hu
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Jinhong Wang
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Dezhong Yao
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, Center for Information in Medicine, University of Electronic Science and Technology of China, Chengdu, PR China,Research Unit of NeuroInformation (2019RU035), Chinese Academy of Medical Sciences, Chengdu, PR China
| | - Jijun Wang
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China,CAS Center for Excellence in Brain Science and Intelligence Technology (CEBSIT), Chinese Academy of Science, Shanghai, PR China,Institute of Psychology and Behavioral Science, Shanghai Jiao Tong University, Shanghai, PR China
| | - Cheng Luo
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, Center for Information in Medicine, University of Electronic Science and Technology of China, Chengdu, PR China,High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, PR China,Research Unit of NeuroInformation (2019RU035), Chinese Academy of Medical Sciences, Chengdu, PR China,To whom correspondence should be addressed; University of Electronic Science and Technology of China, Second North Jianshe Road, Chengdu 610054, PR China; tel: 86-28-83201018, fax: 86-28-83208238, e-mail:
| |
Collapse
|
36
|
Chopra S, Fornito A, Francey SM, O'Donoghue B, Cropley V, Nelson B, Graham J, Baldwin L, Tahtalian S, Yuen HP, Allott K, Alvarez-Jimenez M, Harrigan S, Sabaroedin K, Pantelis C, Wood SJ, McGorry P. Differentiating the effect of antipsychotic medication and illness on brain volume reductions in first-episode psychosis: A Longitudinal, Randomised, Triple-blind, Placebo-controlled MRI Study. Neuropsychopharmacology 2021; 46:1494-1501. [PMID: 33637835 PMCID: PMC8209146 DOI: 10.1038/s41386-021-00980-0] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 01/17/2021] [Accepted: 01/20/2021] [Indexed: 01/31/2023]
Abstract
Changes in brain volume are a common finding in Magnetic Resonance Imaging (MRI) studies of people with psychosis and numerous longitudinal studies suggest that volume deficits progress with illness duration. However, a major unresolved question concerns whether these changes are driven by the underlying illness or represent iatrogenic effects of antipsychotic medication. In this study, 62 antipsychotic-naïve patients with first-episode psychosis (FEP) received either a second-generation antipsychotic (risperidone or paliperidone) or a placebo pill over a treatment period of 6 months. Both FEP groups received intensive psychosocial therapy. A healthy control group (n = 27) was also recruited. Structural MRI scans were obtained at baseline, 3 months and 12 months. Our primary aim was to differentiate illness-related brain volume changes from medication-related changes within the first 3 months of treatment. We secondarily investigated long-term effects at the 12-month timepoint. From baseline to 3 months, we observed a significant group x time interaction in the pallidum (p < 0.05 FWE-corrected), such that patients receiving antipsychotic medication showed increased volume, patients on placebo showed decreased volume, and healthy controls showed no change. Across the entire patient sample, a greater increase in pallidal grey matter volume over 3 months was associated with a greater reduction in symptom severity. Our findings indicate that psychotic illness and antipsychotic exposure exert distinct and spatially distributed effects on brain volume. Our results align with prior work in suggesting that the therapeutic efficacy of antipsychotic medications may be primarily mediated through their effects on the basal ganglia.
Collapse
Affiliation(s)
- Sidhant Chopra
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Clayton, VIC, Australia.
- Monash Biomedical Imaging, Monash University, Clayton, VIC, Australia.
| | - Alex Fornito
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Clayton, VIC, Australia
- Monash Biomedical Imaging, Monash University, Clayton, VIC, Australia
| | - Shona M Francey
- Orygen, Parkville, VIC, Australia
- Centre for Youth Mental Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Brian O'Donoghue
- Orygen, Parkville, VIC, Australia
- Centre for Youth Mental Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Vanessa Cropley
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, University of Melbourne & Melbourne Health, Melbourne, VIC, Australia
| | - Barnaby Nelson
- Orygen, Parkville, VIC, Australia
- Centre for Youth Mental Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Jessica Graham
- Orygen, Parkville, VIC, Australia
- Centre for Youth Mental Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Lara Baldwin
- Orygen, Parkville, VIC, Australia
- Centre for Youth Mental Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Steven Tahtalian
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, University of Melbourne & Melbourne Health, Melbourne, VIC, Australia
| | - Hok Pan Yuen
- Orygen, Parkville, VIC, Australia
- Centre for Youth Mental Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Kelly Allott
- Orygen, Parkville, VIC, Australia
- Centre for Youth Mental Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Mario Alvarez-Jimenez
- Orygen, Parkville, VIC, Australia
- Centre for Youth Mental Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Susy Harrigan
- Department of Social Work, Monash University, Clayton, VIC, Australia
- Melbourne School of Population and Global Health, University of Melbourne, Parkville, VIC, Australia
| | - Kristina Sabaroedin
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Clayton, VIC, Australia
- Monash Biomedical Imaging, Monash University, Clayton, VIC, Australia
| | - Christos Pantelis
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, University of Melbourne & Melbourne Health, Melbourne, VIC, Australia
- The Florey Institute for Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, Australia
| | - Stephen J Wood
- Orygen, Parkville, VIC, Australia
- Centre for Youth Mental Health, The University of Melbourne, Melbourne, VIC, Australia
- School of Psychology, University Birmingham, Edgbaston, UK
| | - Patrick McGorry
- Orygen, Parkville, VIC, Australia
- Centre for Youth Mental Health, The University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
37
|
De Peri L, Deste G, Vita A. Strucutural brain imaging at the onset of schizophrenia:What have we learned and what have we missed. Psychiatry Res 2021; 301:113962. [PMID: 33945963 DOI: 10.1016/j.psychres.2021.113962] [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: 04/12/2021] [Accepted: 04/19/2021] [Indexed: 11/28/2022]
Abstract
Over the past 50 years, the application of structural neuroimaging techniques to schizophrenia research has added relevant information about the pathophysiology of the disorder. Several lines of investigation gave strong evidence that schizophrenia is associated with multiple subtle brain abnormalities that involve both cerebral grey and white matter volumes and structure. The time of onset and longitudinal course of brain morphological abnormalities support the notion that brain pathology of schizophrenia has a neurodevelopmental component and a progressive course, although several confounders of brain changes should be carefully taken into account. Brain anomalies detected before and close to the onset of schizophrenia are likely to be unrelated to confounders of brain changes such as antipsychotic drug treatment, duration of illness or illicit substance abuse, i.e. they related to the pathological process of the disorder per se. Nonetheless, clinically useful diagnostic or prognostic biomarkers have not derived from neuroimaging studies and this is likely related to the neurobiological heterogeneity of the disorder. Thus, there is the compelling need to set new methodological standards for developing innovative hypothesis-driven studies to overcome what we have missed to date in neuroimaging research in schizophrenia.
Collapse
Affiliation(s)
- Luca De Peri
- Cantonal Psychiatric Clinic, Cantonal Socio-Psychiatric Association, Mendrisio, Switzerland
| | - Giacomo Deste
- Department of Mental Health, Spedali Civili Hospital, Brescia, Italy
| | - Antonio Vita
- Department of Mental Health, Spedali Civili Hospital, Brescia, Italy; Department of Clinical and Experimentale Sciences, University of Brescia, Italy.
| |
Collapse
|
38
|
Bustillo JR, Mayer EG, Upston J, Jones T, Garcia C, Sheriff S, Maudsley A, Tohen M, Gasparovic C, Lenroot R. Increased Glutamate Plus Glutamine in the Right Middle Cingulate in Early Schizophrenia but Not in Bipolar Psychosis: A Whole Brain 1H-MRS Study. Front Psychiatry 2021; 12:660850. [PMID: 34163382 PMCID: PMC8215955 DOI: 10.3389/fpsyt.2021.660850] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 04/19/2021] [Indexed: 01/11/2023] Open
Abstract
Proton magnetic resonance spectroscopy (1H-MRS) studies have examined glutamatergic abnormalities in schizophrenia and bipolar-I disorders, mostly in single voxels. Though the critical nodes remain unknown, schizophrenia and bipolar-I involve brain networks with broad abnormalities. To provide insight on the biochemical differences that may underlie these networks, the combined glutamine and glutamate signal (Glx) and other metabolites were examined in patients in early psychosis with whole brain 1H-MRS imaging (1H-MRSI). Data were acquired in young schizophrenia subjects (N = 48), bipolar-I subjects (N = 21) and healthy controls (N = 51). Group contrasts for Glx, as well as for N-acetyl aspartate, choline, myo-inositol and creatine, from all voxels that met spectral quality criteria were analyzed in standardized brain space, followed by cluster-corrected level alpha-value (CCLAV ≤ 0.05) analysis. Schizophrenia subjects had higher Glx in the right middle cingulate gyrus (19 voxels, CCLAV = 0.05) than bipolar-I subjects. Healthy controls had intermediate Glx values, though not significant. Schizophrenia subjects also had higher N-acetyl aspartate (three clusters, left occipital, left frontal, right frontal), choline (two clusters, left and right frontal) and myo-inositol (one cluster, left frontal) than bipolar-I, with healthy controls having intermediate values. These increases were likely accounted for by antipsychotic medication effects in the schizophrenia subgroup for N-acetyl aspartate and choline. Likewise, creatine was increased in two clusters in treated vs. antipsychotic-naïve schizophrenia, supporting a medication effect. Conversely, the increments in Glx in right cingulate were not driven by antipsychotic medication exposure. We conclude that increments in Glx in the cingulate may be critical to the pathophysiology of schizophrenia and are consistent with the NMDA hypo-function model. This model however may be more specific to schizophrenia than to psychosis in general. Postmortem and neuromodulation schizophrenia studies focusing on right cingulate, may provide critical mechanistic and therapeutic advancements, respectively.
Collapse
Affiliation(s)
- Juan R. Bustillo
- Department of Psychiatry and Behavioral Sciences, University of New Mexico, Albuquerque, NM, United States
| | - Elizabeth G. Mayer
- Department of Psychiatry and Behavioral Sciences, University of New Mexico, Albuquerque, NM, United States
| | - Joel Upston
- Department of Psychiatry and Behavioral Sciences, University of New Mexico, Albuquerque, NM, United States
- Department of Mathematics and Statistics, University of New Mexico, Albuquerque, NM, United States
| | - Thomas Jones
- Department of Psychiatry and Behavioral Sciences, University of New Mexico, Albuquerque, NM, United States
| | - Crystal Garcia
- Department of Psychiatry and Behavioral Sciences, University of New Mexico, Albuquerque, NM, United States
| | - Sulaiman Sheriff
- Department of Radiology, University of Miami, Miami, FL, United States
| | - Andrew Maudsley
- Department of Radiology, University of Miami, Miami, FL, United States
| | - Mauricio Tohen
- Department of Psychiatry and Behavioral Sciences, University of New Mexico, Albuquerque, NM, United States
| | | | - Rhoshel Lenroot
- Department of Psychiatry and Behavioral Sciences, University of New Mexico, Albuquerque, NM, United States
| |
Collapse
|
39
|
Slomianka L. Basic quantitative morphological methods applied to the central nervous system. J Comp Neurol 2021; 529:694-756. [PMID: 32639600 PMCID: PMC7818269 DOI: 10.1002/cne.24976] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 06/15/2020] [Accepted: 06/16/2020] [Indexed: 12/19/2022]
Abstract
Generating numbers has become an almost inevitable task associated with studies of the morphology of the nervous system. Numbers serve a desire for clarity and objectivity in the presentation of results and are a prerequisite for the statistical evaluation of experimental outcomes. Clarity, objectivity, and statistics make demands on the quality of the numbers that are not met by many methods. This review provides a refresher of problems associated with generating numbers that describe the nervous system in terms of the volumes, surfaces, lengths, and numbers of its components. An important aim is to provide comprehensible descriptions of the methods that address these problems. Collectively known as design-based stereology, these methods share two features critical to their application. First, they are firmly based in mathematics and its proofs. Second and critically underemphasized, an understanding of their mathematical background is not necessary for their informed and productive application. Understanding and applying estimators of volume, surface, length or number does not require more of an organizational mastermind than an immunohistochemical protocol. And when it comes to calculations, square roots are the gravest challenges to overcome. Sampling strategies that are combined with stereological probes are efficient and allow a rational assessment if the numbers that have been generated are "good enough." Much may be unfamiliar, but very little is difficult. These methods can no longer be scapegoats for discrepant results but faithfully produce numbers on the material that is assessed. They also faithfully reflect problems that associated with the histological material and the anatomically informed decisions needed to generate numbers that are not only valid in theory. It is within reach to generate practically useful numbers that must integrate with qualitative knowledge to understand the function of neural systems.
Collapse
Affiliation(s)
- Lutz Slomianka
- University of Zürich, Institute of AnatomyZürichSwitzerland
| |
Collapse
|
40
|
Matsche MA, Blazer VS, Pulster EL, Mazik PM. Biological and anthropogenic influences on macrophage aggregates in white perch Morone americana from Chesapeake Bay, USA. DISEASES OF AQUATIC ORGANISMS 2021; 143:79-100. [PMID: 33570042 DOI: 10.3354/dao03555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The response of macrophage aggregates in fish to a variety of environmental stressors has been useful as a biomarker of exposure to habitat degradation. Total volume of macrophage aggregates (MAV) was estimated in the liver and spleen of white perch Morone americana from Chesapeake Bay using stereological approaches. Hepatic and splenic MAV were compared between fish populations from the rural Choptank River (n = 122) and the highly urbanized Severn River (n = 131). Hepatic and splenic MAV increased with fish age, were greater in females from the Severn River only, and were significantly greater in fish from the more polluted Severn River (higher concentrations of polycyclic aromatic hydrocarbons, organochlorine pesticides, and brominated diphenyl ethers). Water temperature and dissolved oxygen had a significant effect on organ volumes, but not on MAV. Age and river were most influential on hepatic and splenic MAV, suggesting that increased MAV in Severn River fish resulted from chronic exposures to higher concentrations of environmental contaminants and other stressors. Hemosiderin was abundant in 97% of spleens and was inversely related to fish condition and positively related to fish age and trematode infections. Minor amounts of hemosiderin were detected in 30% of livers and positively related to concentrations of benzo[a]pyrene metabolite equivalents in the bile. This study demonstrated that hepatic and splenic MAV were useful indicators in fish from the 2 tributaries with different land use characteristics and concentrations of environmental contaminants. More data are needed from additional tributaries with a wider gradient of environmental impacts to validate our results in this species.
Collapse
Affiliation(s)
- Mark A Matsche
- Maryland Department of Natural Resources, Cooperative Oxford Laboratory, 904 South Morris Street, Oxford, Maryland 21654, USA
| | | | | | | |
Collapse
|
41
|
Lyall AE, Nägele FL, Pasternak O, Gallego JA, Malhotra AK, McNamara RK, Kubicki M, Peters BD, Robinson DG, Szeszko PR. A 16-week randomized placebo-controlled trial investigating the effects of omega-3 polyunsaturated fatty acid treatment on white matter microstructure in recent-onset psychosis patients concurrently treated with risperidone. Psychiatry Res Neuroimaging 2021; 307:111219. [PMID: 33221631 PMCID: PMC8127861 DOI: 10.1016/j.pscychresns.2020.111219] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 10/21/2020] [Accepted: 11/03/2020] [Indexed: 11/19/2022]
Abstract
We examined the impact of treatment with fish oil (FO), a rich source of omega-3 polyunsaturated fatty acids (n-3 PUFA), on white matter in 37 recent-onset psychosis patients receiving risperidone in a double-blind placebo-controlled randomized clinical trial. Patients were scanned at baseline and randomly assigned to receive 16-weeks of treatment with risperidone + FO or risperidone + placebo. Eighteen patients received follow-up MRIs (FO, n = 10/Placebo, n = 8). Erythrocyte levels of n-3 PUFAs eicosapentaenoic acid (EPA), docosahexaenoic acid (DHA), and docosapentaenoic acid (DPA) were obtained at both time points. We employed Free Water Imaging metrics representing the extracellular free water fraction (FW) and fractional anisotropy of the tissue (FA-t). Analyses were conducted using Tract-Based-Spatial-Statistics and nonparametric permutation-based tests with family-wise error correction. There were significant positive correlations of FA-t with DHA and DPA among all patients at baseline. Patients treated with risperidone + placebo demonstrated reductions in FA-t and increases in FW, whereas patients treated with risperidone + FO exhibited no significant changes in FW and FA-t reductions were largely attenuated. The correlations of DPA and DHA with baseline FA-t support the hypothesis that n-3 PUFA intake or biosynthesis are associated with white matter abnormalities in psychosis. Adjuvant FO treatment may partially mitigate against white matter alterations observed in recent-onset psychosis patients following risperidone treatment.
Collapse
Affiliation(s)
- Amanda E Lyall
- Psychiatry Neuroimaging Laboratory, Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States; Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States.
| | - Felix L Nägele
- Psychiatry Neuroimaging Laboratory, Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States; Psychiatry Neuroimaging Branch, Department of Psychiatry and Psychotherapy, University Medical Center Hamburg-Eppendorf, University of Hamburg, Hamburg, Germany
| | - Ofer Pasternak
- Psychiatry Neuroimaging Laboratory, Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States; Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Juan A Gallego
- Departments of Psychiatry and of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States; Feinstein Institutes for Medical Research, Manhasset, NY, United States
| | - Anil K Malhotra
- Departments of Psychiatry and of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States; Feinstein Institutes for Medical Research, Manhasset, NY, United States
| | - Robert K McNamara
- Department of Psychiatry and Behavioral Neuroscience, Lipidomics Research Program, University of Cincinnati, United States
| | - Marek Kubicki
- Psychiatry Neuroimaging Laboratory, Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States; Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States; Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Bart D Peters
- Feinstein Institutes for Medical Research, Manhasset, NY, United States
| | - Delbert G Robinson
- Departments of Psychiatry and of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States; Feinstein Institutes for Medical Research, Manhasset, NY, United States
| | - Philip R Szeszko
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Mental Illness Research, Education and Clinical Center, James J. Peters VA Medical Center, Bronx, NY, United States
| |
Collapse
|
42
|
Kimura H, Kanahara N, Iyo M. Rationale and neurobiological effects of treatment with antipsychotics in patients with chronic schizophrenia considering dopamine supersensitivity. Behav Brain Res 2021; 403:113126. [PMID: 33460681 DOI: 10.1016/j.bbr.2021.113126] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 12/29/2020] [Accepted: 01/04/2021] [Indexed: 12/12/2022]
Abstract
The long-term treatment of patients with schizophrenia often involves the management of relapses for most patients and the development of treatment resistance in some patients. To stabilize the clinical course and allow as many patients as possible to recover, clinicians need to recognize dopamine supersensitivity, which can be provoked by administration of high dosages of antipsychotics, and deal with it properly. However, no treatment guidelines have addressed this issue. The present review summarized the characteristics of long-acting injectable antipsychotics, dopamine partial agonists, and clozapine in relation to dopamine supersensitivity from the viewpoints of receptor profiles and pharmacokinetics. The potential merits and limitations of these medicines are discussed, as well as the risks of treating patients with established dopamine supersensitivity with these classes of drugs. Finally, the review discussed the biological influence of antipsychotic treatment on the human brain based on findings regarding the relationship between the hippocampus and antipsychotics.
Collapse
Affiliation(s)
- Hiroshi Kimura
- Department of Psychiatry, School of Medicine, International University of Health and Welfare, Chiba, Japan; Department of Psychiatry, Chiba University Graduate School of Medicine, Chiba, Japan; Department of Psychiatry, Gakuji-kai Kimura Hospital, Chiba, Japan.
| | - Nobuhisa Kanahara
- Department of Psychiatry, Chiba University Graduate School of Medicine, Chiba, Japan; Division of Medical Treatment and Rehabilitation, Chiba University Center for Forensic Mental Health, Chiba, Japan
| | - Masaomi Iyo
- Department of Psychiatry, Chiba University Graduate School of Medicine, Chiba, Japan
| |
Collapse
|
43
|
Sonnenschein SF, Grace AA. Emerging therapeutic targets for schizophrenia: a framework for novel treatment strategies for psychosis. Expert Opin Ther Targets 2021; 25:15-26. [PMID: 33170748 PMCID: PMC7855878 DOI: 10.1080/14728222.2021.1849144] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 11/05/2020] [Indexed: 01/10/2023]
Abstract
Introduction: Antipsychotic drugs are central to the treatment of schizophrenia, but their limitations necessitate improved treatment strategies. Multiple lines of research have implicated glutamatergic dysfunction in the hippocampus as an early source of pathophysiology in schizophrenia. Novel compounds have been designed to treat glutamatergic dysfunction, but they have produced inconsistent results in clinical trials. Areas covered: This review discusses how the hippocampus is thought to drive psychotic symptoms through its influence on the dopamine system. It offers the reader an evaluation of proposed treatment strategies including direct modulation of GABA or glutamate neurotransmission or reducing the deleterious impact of stress on circuit development. Finally, we offer a perspective on aspects of future research that will advance our knowledge and may create new therapeutic opportunities. PubMed was searched for relevant literature between 2010 and 2020 and related studies. Expert opinion: Targeting aberrant excitatory-inhibitory neurotransmission in the hippocampus and its related circuits has the potential to alleviate symptoms and reduce the risk of transition to psychosis if implemented as an early intervention. Longitudinal multimodal brain imaging combined with mechanistic theories generated from animal models can be used to better understand the progression of hippocampal-dopamine circuit dysfunction and heterogeneity in treatment response.
Collapse
Affiliation(s)
| | - Anthony A. Grace
- Departments of Neuroscience, Psychiatry and Psychology, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
44
|
Brocos-Mosquera I, Gabilondo AM, Meana JJ, Callado LF, Erdozain AM. Spinophilin expression in postmortem prefrontal cortex of schizophrenic subjects: Effects of antipsychotic treatment. Eur Neuropsychopharmacol 2021; 42:12-21. [PMID: 33257116 DOI: 10.1016/j.euroneuro.2020.11.011] [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: 04/15/2020] [Revised: 11/04/2020] [Accepted: 11/11/2020] [Indexed: 10/22/2022]
Abstract
Schizophrenia has been associated with alterations in neurotransmission and synaptic dysfunction. Spinophilin is a multifunctional scaffold protein that modulates excitatory synaptic transmission and dendritic spine morphology. Spinophilin can also directly interact with and regulate several receptors for neurotransmitters, such as dopamine D2 receptors, which play a role in the pathophysiology of schizophrenia and are targets of antipsychotics. Several studies have thus suggested an implication of spinophilin in schizophrenia. In the present study spinophilin protein expression was determined by western blot in the postmortem dorsolateral prefrontal cortex of 24 subjects with schizophrenia (12 antipsychotic-free and 12 antipsychotic-treated subjects) and 24 matched controls. Experiments were performed in synaptosomal membranes (SPM) and in postsynaptic density fractions (PSD). As previously reported, two specific bands for this protein were observed: an upper 120-130 kDa band and a lower 80-95 kDa band. The spinophilin lower band showed a significant decrease in schizophrenia subjects compared to matched controls, both in SPM and PSD fractions (-15%, p = 0.007 and -15%, p = 0.039, respectively). When schizophrenia subjects were divided by the presence or absence of antipsychotics in blood at death, the lower band showed a significant decrease in antipsychotic-treated schizophrenia subjects (-24%, p = 0.003 for SPM and -26%, p = 0.014 for PSD), but not in antipsychotic-free subjects, compared to their matched controls. These results suggest that antipsychotics could produce alterations in spinophilin expression that do not seem to be related to schizophrenia per se. These changes may underlie some of the side effects of antipsychotics.
Collapse
Affiliation(s)
- Iria Brocos-Mosquera
- Department of Pharmacology, University of the Basque Country, UPV/EHU, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain
| | - Ane M Gabilondo
- Department of Pharmacology, University of the Basque Country, UPV/EHU, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain; Biocruces Bizkaia Health Research Institute, Barakaldo, Bizkaia, Spain
| | - J Javier Meana
- Department of Pharmacology, University of the Basque Country, UPV/EHU, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain; Biocruces Bizkaia Health Research Institute, Barakaldo, Bizkaia, Spain
| | - Luis F Callado
- Department of Pharmacology, University of the Basque Country, UPV/EHU, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain; Biocruces Bizkaia Health Research Institute, Barakaldo, Bizkaia, Spain
| | - Amaia M Erdozain
- Department of Pharmacology, University of the Basque Country, UPV/EHU, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain.
| |
Collapse
|
45
|
Hatzipantelis C, Langiu M, Vandekolk TH, Pierce TL, Nithianantharajah J, Stewart GD, Langmead CJ. Translation-Focused Approaches to GPCR Drug Discovery for Cognitive Impairments Associated with Schizophrenia. ACS Pharmacol Transl Sci 2020; 3:1042-1062. [PMID: 33344888 PMCID: PMC7737210 DOI: 10.1021/acsptsci.0c00117] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Indexed: 01/07/2023]
Abstract
There are no effective therapeutics for cognitive impairments associated with schizophrenia (CIAS), which includes deficits in executive functions (working memory and cognitive flexibility) and episodic memory. Compounds that have entered clinical trials are inadequate in terms of efficacy and/or tolerability, highlighting a clear translational bottleneck and a need for a cohesive preclinical drug development strategy. In this review we propose hippocampal-prefrontal-cortical (HPC-PFC) circuitry underlying CIAS-relevant cognitive processes across mammalian species as a target source to guide the translation-focused discovery and development of novel, procognitive agents. We highlight several G protein-coupled receptors (GPCRs) enriched within HPC-PFC circuitry as therapeutic targets of interest, including noncanonical approaches (biased agonism and allosteric modulation) to conventional clinical targets, such as dopamine and muscarinic acetylcholine receptors, along with prospective novel targets, including the orphan receptors GPR52 and GPR139. We also describe the translational limitations of popular preclinical cognition tests and suggest touchscreen-based assays that probe cognitive functions reliant on HPC-PFC circuitry and reflect tests used in the clinic, as tests of greater translational relevance. Combining pharmacological and behavioral testing strategies based in HPC-PFC circuit function creates a cohesive, translation-focused approach to preclinical drug development that may improve the translational bottleneck currently hindering the development of treatments for CIAS.
Collapse
Affiliation(s)
- Cassandra
J. Hatzipantelis
- Drug
Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Monica Langiu
- Drug
Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Teresa H. Vandekolk
- Drug
Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Tracie L. Pierce
- Drug
Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Jess Nithianantharajah
- Florey
Institute of Neuroscience
and Mental Health, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Gregory D. Stewart
- Drug
Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Christopher J. Langmead
- Drug
Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| |
Collapse
|
46
|
Feng R, Womer FY, Edmiston EK, Chen Y, Wang Y, Chang M, Yin Z, Wei Y, Duan J, Ren S, Li C, Liu Z, Jiang X, Wei S, Li S, Zhang X, Zuo XN, Tang Y, Wang F. Antipsychotic Effects on Cortical Morphology in Schizophrenia and Bipolar Disorders. Front Neurosci 2020; 14:579139. [PMID: 33362453 PMCID: PMC7758211 DOI: 10.3389/fnins.2020.579139] [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: 07/01/2020] [Accepted: 11/10/2020] [Indexed: 11/30/2022] Open
Abstract
Background: Previous studies of atypical antipsychotic effects on cortical structures in schizophrenia (SZ) and bipolar disorder (BD) have findings that vary between the short and long term. In particular, there has not been a study exploring the effects of atypical antipsychotics on age-related cortical structural changes in SZ and BD. This study aimed to determine whether mid- to long-term atypical antipsychotic treatment (mean duration = 20 months) is associated with cortical structural changes and whether age-related cortical structural changes are affected by atypical antipsychotics. Methods: Structural magnetic resonance imaging images were obtained from 445 participants consisting of 88 medicated patients (67 with SZ, 21 with BD), 84 unmedicated patients (50 with SZ, 34 with BD), and 273 healthy controls (HC). Surface-based analyses were employed to detect differences in thickness and area among the three groups. We examined the age-related effects of atypical antipsychotics after excluding the potential effects of illness duration. Results: Significant differences in cortical thickness were observed in the frontal, temporal, parietal, and insular areas and the isthmus of the cingulate gyrus. The medicated group showed greater cortical thinning in these regions than the unmediated group and HC; furthermore, there were age-related differences in the effects of atypical antipsychotics, and these effects did not relate to illness duration. Moreover, cortical thinning was significantly correlated with lower symptom scores and Wisconsin Card Sorting Test (WCST) deficits in patients. After false discovery rate correction, cortical thinning in the right middle temporal gyrus in patients was significantly positively correlated with lower HAMD scores. The unmedicated group showed only greater frontotemporal thickness than the HC group. Conclusion: Mid- to long-term atypical antipsychotic use may adversely affect cortical thickness over the course of treatment and ageing and may also result in worsening cognitive function.
Collapse
Affiliation(s)
- Ruiqi Feng
- Department of Radiology, The First Affiliated Hospital of China Medical University, Shenyang, China
- Brain Function Research Section, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Fay Y. Womer
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, United States
| | - E. Kale Edmiston
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Yifan Chen
- Brain Function Research Section, The First Affiliated Hospital of China Medical University, Shenyang, China
- Department of Psychiatry, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Yinshan Wang
- CAS Key Laboratory of Behavioral Science and Research Center for Lifespan Development of Mind and Brain (CLIMB), Institute of Psychology, Beijing, China
| | - Miao Chang
- Department of Radiology, The First Affiliated Hospital of China Medical University, Shenyang, China
- Brain Function Research Section, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Zhiyang Yin
- Brain Function Research Section, The First Affiliated Hospital of China Medical University, Shenyang, China
- Department of Psychiatry, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Yange Wei
- Brain Function Research Section, The First Affiliated Hospital of China Medical University, Shenyang, China
- Department of Psychiatry, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Jia Duan
- Brain Function Research Section, The First Affiliated Hospital of China Medical University, Shenyang, China
- Department of Psychiatry, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Sihua Ren
- Department of Radiology, The First Affiliated Hospital of China Medical University, Shenyang, China
- Brain Function Research Section, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Chao Li
- Department of Radiology, The First Affiliated Hospital of China Medical University, Shenyang, China
- Brain Function Research Section, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Zhuang Liu
- School of Public Health, China Medical University, Shenyang, China
| | - Xiaowei Jiang
- Department of Radiology, The First Affiliated Hospital of China Medical University, Shenyang, China
- Brain Function Research Section, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Shengnan Wei
- Department of Radiology, The First Affiliated Hospital of China Medical University, Shenyang, China
- Brain Function Research Section, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Songbai Li
- Department of Radiology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Xizhe Zhang
- School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, China
| | - Xi-Nian Zuo
- Key Laboratory of Brain and Education Sciences, School of Education Sciences, Nanning Normal University, Nanning, China
| | - Yanqing Tang
- Brain Function Research Section, The First Affiliated Hospital of China Medical University, Shenyang, China
- Department of Psychiatry, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Fei Wang
- Department of Radiology, The First Affiliated Hospital of China Medical University, Shenyang, China
- Brain Function Research Section, The First Affiliated Hospital of China Medical University, Shenyang, China
- Department of Psychiatry, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
47
|
Glausier JR, Enwright JF, Lewis DA. Diagnosis- and Cell Type-Specific Mitochondrial Functional Pathway Signatures in Schizophrenia and Bipolar Disorder. Am J Psychiatry 2020; 177:1140-1150. [PMID: 33115248 PMCID: PMC8195258 DOI: 10.1176/appi.ajp.2020.19111210] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
OBJECTIVE The shared risk factors and clinical features in schizophrenia and bipolar disorder may be linked via mitochondrial dysfunction. However, the severity of mitochondrial dysfunction, and/or the specific mitochondrial functional pathways affected, may differ between diagnoses, especially at the level of individual cell types. METHODS Transcriptomic profiling data for a gene set indexing mitochondrial functional pathways were obtained for dorsolateral prefrontal cortex (DLPFC) gray matter and layer 3 and layer 5 pyramidal neurons of subjects with schizophrenia or bipolar disorder. Analyses were conducted using a dual strategy: identification of differentially expressed genes (DEGs) and their functional pathway enrichment, and application of weighted gene coexpression network analysis. These analyses were repeated in monkeys chronically exposed to antipsychotic drugs to determine their effect on mitochondrial-related gene expression. RESULTS In DLPFC gray matter, 41% of mitochondrial-related genes were differentially expressed in the schizophrenia group, whereas 8% were differentially expressed in the bipolar group. In the schizophrenia group, 83% of DEGs showed lower expression, and these were significantly enriched for three functional pathways, each indexing energy production. DEGs in the bipolar disorder group were not enriched for functional pathways. This disease-related pattern of findings was also identified in pyramidal neurons. None of the gene expression alterations disrupted coexpression modules, and DEGs were not attributable to antipsychotic medications. CONCLUSIONS Schizophrenia and bipolar disorder do not appear to share similar mitochondrial alterations in the DLPFC. The selective and coordinated down-regulation of energy production genes in schizophrenia is consistent with the effects of chronic reductions in pyramidal neuron firing, and enhancement of this activity may serve as a therapeutic target.
Collapse
Affiliation(s)
- Jill R Glausier
- Department of Psychiatry, University of Pittsburgh (all authors)
| | - John F Enwright
- Department of Psychiatry, University of Pittsburgh (all authors)
| | - David A Lewis
- Department of Psychiatry, University of Pittsburgh (all authors)
| |
Collapse
|
48
|
Boz Z, Hu M, Yu Y, Huang XF. N-acetylcysteine prevents olanzapine-induced oxidative stress in mHypoA-59 hypothalamic neurons. Sci Rep 2020; 10:19185. [PMID: 33154380 PMCID: PMC7644715 DOI: 10.1038/s41598-020-75356-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 09/25/2020] [Indexed: 12/30/2022] Open
Abstract
Olanzapine is a second-generation antipsychotic (AP) drug commonly prescribed for the treatment of schizophrenia. Recently, olanzapine has been found to cause brain tissue volume loss in rodent and primate studies; however, the underlying mechanism remains unknown. Abnormal autophagy and oxidative stress have been implicated to have a role in AP-induced neurodegeneration, while N-acetylcysteine (NAC) is a potent antioxidant, shown to be beneficial in the treatment of schizophrenia. Here, we investigate the role of olanzapine and NAC on cell viability, oxidative stress, mitochondrial mass and mitophagy in hypothalamic cells. Firstly, cell viability was assessed in mHypoA-59 and mHypoA NPY/GFP cells using an MTS assay and flow cytometric analyses. Olanzapine treated mHypoA-59 cells were then assessed for mitophagy markers and oxidative stress; including quantification of lysosomes, autophagosomes, LC3B-II, p62, superoxide anion (O2–) and mitochondrial mass. NAC (10 mM) was used to reverse the effects of olanzapine (100 µM) on O2−, mitochondrial mass and LC3B-II. We found that olanzapine significantly impacted cell viability in mHypoA-59 hypothalamic cells in a dose and time-dependent manner. Olanzapine inhibited mitophagy, instigated oxidative stress and prompted mitochondrial abnormalities. NAC was able to mitigate olanzapine-induced effects. These findings suggest that high doses of olanzapine may cause neurotoxicity of hypothalamic neurons via increased production of reactive oxygen species (ROS), mitochondrial damage and mitophagy inhibition. This could in part explain data suggesting that APs may reduce brain volume.
Collapse
Affiliation(s)
- Zehra Boz
- Illawarra Health and Medical Research Institute and School of Medicine, University of Wollongong, Wollongong, NSW, 2522, Australia
| | - Minmin Hu
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Yinghua Yu
- Illawarra Health and Medical Research Institute and School of Medicine, University of Wollongong, Wollongong, NSW, 2522, Australia.,Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Xu-Feng Huang
- Illawarra Health and Medical Research Institute and School of Medicine, University of Wollongong, Wollongong, NSW, 2522, Australia.
| |
Collapse
|
49
|
Zito MF, Marder SR. Rethinking the risks and benefits of long-term maintenance in schizophrenia. Schizophr Res 2020; 225:77-81. [PMID: 31806525 DOI: 10.1016/j.schres.2019.10.057] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 10/28/2019] [Accepted: 10/30/2019] [Indexed: 11/26/2022]
Abstract
This review addresses the risks and benefits of long-term maintenance antipsychotic treatment for patients that extends beyond two years. It focuses on framing discussions with patients who are recovering from a first episode. For these patients the evidence strongly supports the benefits over the risk for the first two years. However, both the clinical side effects of antipsychotics and the possible long-term effects of dopamine blocking drugs on the brain require a more nuanced discussion beyond this initial period. In most cases, the decision will be to continue antipsychotics but to consider strategies for mitigating the risks of drugs. This review provides information about the relative risks of dose reduction and intermittent treatment.
Collapse
Affiliation(s)
- Michael F Zito
- Semel Institute for Neuroscience at UCLA and the VA Desert Pacific Mental Illness Research, Education, and Clinical Center, Los Angeles, USA
| | - Stephen R Marder
- Semel Institute for Neuroscience at UCLA and the VA Desert Pacific Mental Illness Research, Education, and Clinical Center, Los Angeles, USA.
| |
Collapse
|
50
|
Palaniyappan L, Sukumar N. Reconsidering brain tissue changes as a mechanistic focus for early intervention in psychiatry. J Psychiatry Neurosci 2020; 45. [PMID: 33119489 PMCID: PMC7595740 DOI: 10.1503/jpn.200172] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Affiliation(s)
- Lena Palaniyappan
- From the Robarts Research Institute, Western University (Palaniyappan); the Department of Psychiatry, Western University (Palaniyappan, Sukumar); the Lawson Health Research Institute, Imaging Division (Palaniyappan); and the Department of Medical Biophysics, Western University (Palaniyappan), London, Ont., Canada
| | - Niron Sukumar
- From the Robarts Research Institute, Western University (Palaniyappan); the Department of Psychiatry, Western University (Palaniyappan, Sukumar); the Lawson Health Research Institute, Imaging Division (Palaniyappan); and the Department of Medical Biophysics, Western University (Palaniyappan), London, Ont., Canada
| |
Collapse
|