1
|
Perkins DO, Jeffries CD, Clark SR, Upthegrove R, Wannan CMJ, Wray NR, Li QS, Do KQ, Walker E, Paul Amminger G, Anticevic A, Cotter D, Ellman LM, Mongan D, Phassouliotis C, Barbee J, Roth S, Billah T, Corcoran C, Calkins ME, Cerrato F, Khadimallah I, Klauser P, Winter-van Rossum I, Nunez AR, Bleggi RS, Martin AR, Bouix S, Pasternak O, Shah JL, Toben C, Wolf DH, Accelerating Medicines Partnership® Schizophrenia (AMP® SCZ), Kahn RS, Kane JM, McGorry PD, Bearden CE, Nelson B, Shenton ME, Woods SW. Body fluid biomarkers and psychosis risk in The Accelerating Medicines Partnership® Schizophrenia Program: design considerations. SCHIZOPHRENIA (HEIDELBERG, GERMANY) 2025; 11:78. [PMID: 40399418 PMCID: PMC12095529 DOI: 10.1038/s41537-025-00610-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 02/11/2025] [Indexed: 05/23/2025]
Abstract
Advances in proteomic assay methodologies and genomics have significantly improved our understanding of the blood proteome. Schizophrenia and psychosis risk are linked to polygenic scores for schizophrenia and other mental disorders, as well as to altered blood and saliva levels of biomarkers involved in hormonal signaling, redox balance, and chronic systemic inflammation. The Accelerating Medicines Partnership® Schizophrenia (AMP®SCZ) aims to ascertain biomarkers that both predict clinical outcomes and provide insights into the biological processes driving clinical outcomes in persons meeting CHR criteria. AMP®SCZ will follow almost 2000 CHR and 640 community study participants for two years, assessing biomarkers at baseline and two-month follow-up including the collection of blood and saliva samples. The following provides the rationale and methods for plans to utilize polygenic risk scores for schizophrenia and other disorders, salivary cortisol levels, and a discovery-based proteomic platform for plasma analyses. We also provide details about the standardized methods used to collect and store these biological samples, as well as the study participant metadata and quality control measures related to preanalytical factors that could influence the values of the biomarkers. Finally, we discuss our plans for analyzing the results of blood- and saliva-based biomarkers. Watch Dr. Perkins discuss their work and this article: https://vimeo.com/1062879582?share=copy#t=0 .
Collapse
Affiliation(s)
- Diana O Perkins
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Clark D Jeffries
- Rennaisance Computing Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Scott R Clark
- Discipline of Psychiatry, University of Adelaide, Adelaide, SA, Australia
- Basil Hetzel Institute, Woodville, SA, Australia
| | - Rachel Upthegrove
- Institute for Mental Health, University of Birmingham, Birmingham, UK
- Birmingham Womens and Childrens, NHS Foundation Trust, Birmingham, UK
| | - Cassandra M J Wannan
- Centre for Youth Mental Health, The University of Melbourne, Parkville, VIC, Australia
- Orygen, Parkville, VIC, Australia
| | - Naomi R Wray
- Department of Psychiatry, University of Oxford, Oxford, UK
- Institute for Molecular Biosciences, University of Queensland, Queensland, Australia
| | - Qingqin S Li
- JRD Data Science, Janssen Research & Development, LLC, Titusville, NJ, USA
| | - Kim Q Do
- Department of Psychiatry, Center for Psychiatric Neuroscience, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience King's College London, London, UK
| | - Elaine Walker
- Departments of Psychology and Psychiatry, Emory University, Atlanta, GA, United States of America
| | - G Paul Amminger
- Centre for Youth Mental Health, The University of Melbourne, Parkville, VIC, Australia
- Orygen, Parkville, VIC, Australia
| | - Alan Anticevic
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - David Cotter
- Department of Psychiatry, Royal College of Surgeons in Ireland, Dublin, Ireland
- School of Computing and Information Systems, The University of Melbourne, Parkville, VIC, Australia
| | - Lauren M Ellman
- Department of Psychology and Neuroscience, Temple University, Philadelphia, PA, USA
| | - David Mongan
- Department of Psychiatry, Royal College of Surgeons in Ireland, Dublin, Ireland
- Centre for Public Health, Queen's University Belfast, Belfast, United Kingdom
| | - Christina Phassouliotis
- Centre for Youth Mental Health, The University of Melbourne, Parkville, VIC, Australia
- Orygen, Parkville, VIC, Australia
| | - Jenna Barbee
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sharin Roth
- Genomics and Biomarker Research, Otsuka Pharmaceutical Development & Commercialization, Inc, Rockville, MD, USA
| | - Tashrif Billah
- Department of Psychiatry, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Cheryl Corcoran
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Monica E Calkins
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Felecia Cerrato
- Stanley Center for Psychiatric Research and Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ines Khadimallah
- Department of Psychiatry, Center for Psychiatric Neuroscience, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience King's College London, London, UK
| | - Paul Klauser
- Department of Psychiatry, Center for Psychiatric Neuroscience, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Department of Psychiatry, Service of Child and Adolescent Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | | | - Angela R Nunez
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Connecticut Mental Health Center, New Haven, CT, USA
| | - Rachel S Bleggi
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Alicia R Martin
- Stanley Center for Psychiatric Research and Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Sylvain Bouix
- Department of Psychiatry, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Brigham and Women's Hospital, Boston, MA, USA
- Department of Psychiatry, MGB, Massachusetts General Hospital, Boston, MA, USA
| | - Ofer Pasternak
- Department of Psychiatry, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Brigham and Women's Hospital, Boston, MA, USA
- Department of Psychiatry, MGB, Massachusetts General Hospital, Boston, MA, USA
| | - Jai L Shah
- Douglas Research Centre, Montreal, Quebec, Canada
- Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| | - Catherine Toben
- Discipline of Psychiatry, University of Adelaide, Adelaide, SA, Australia
| | - Daniel H Wolf
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Rene S Kahn
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John M Kane
- Department of Psychiatry, Donald and Barbara Zucker School of Medicine, Hempstead, N.Y, USA
- Institute for Behavioral Science, Feinstein Institute for Medical Research, Manhasset, NY, USA
| | - Patrick D McGorry
- Centre for Youth Mental Health, The University of Melbourne, Parkville, VIC, Australia
- Orygen, Parkville, VIC, Australia
| | - Carrie E Bearden
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, USA
| | - Barnaby Nelson
- Centre for Youth Mental Health, The University of Melbourne, Parkville, VIC, Australia
- Orygen, Parkville, VIC, Australia
| | - Martha E Shenton
- Department of Psychiatry, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Brigham and Women's Hospital, Boston, MA, USA
- Department of Psychiatry, MGB, Massachusetts General Hospital, Boston, MA, USA
| | - Scott W Woods
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Connecticut Mental Health Center, New Haven, CT, USA
| |
Collapse
|
2
|
Wang J, Li F, Ren F, Ma W, Liu Y, Zhang W, Li X, Bi Y, Xin Q, Gao F. Association of GRM7 Polymorphisms with Bilateral Auditory Regions Glutamate and Coupling with Glutathione in ARHL Patients. Mol Neurobiol 2025:10.1007/s12035-025-05011-3. [PMID: 40360955 DOI: 10.1007/s12035-025-05011-3] [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: 10/29/2024] [Accepted: 04/29/2025] [Indexed: 05/15/2025]
Abstract
This study aimed to investigate the relationship between GRM7 polymorphisms and the levels of glutamate (Glu) and glutathione (GSH) in bilateral auditory regions (ARs) of ARHL patients. Seventy-eight ARHL patients (mean age, 65.94 years ± 3.37 [SD]; 44 men) and 46 normal hearing (NH) controls (mean age, 65.72 years ± 2.32 [SD]; 28 men) were enrolled. Glu and GSH levels in bilateral ARs of all participants were measured and estimated by using magnetic resonance spectroscopy (MRS) and LCModel. In addition, we collected peripheral venous blood samples from all participants for DNA extraction and investigated polymorphisms in the GRM7 gene using TaqMan SNP genotyping. The results showed that Glu and GSH levels in bilateral ARs were significantly lower in GRM7 high-risk group compared with GRM7 low-risk group, regardless of disease status (all pglu < 0.001; all pgsh = 0.001). Furthermore, GRM7 low-risk ARHL group had lower Glu levels in bilateral ARs than GRM7 low-risk NH group, whereas no difference was observed between NH and ARHL groups in high-risk (all pglu < 0.05; all pglu > 0.05). Finally, we found that Glu and GSH levels were positively correlated only in the low-risk NH group (rleft = 0.536 p = 0.007; rright = 0.545 p = 0.006). The glutamatergic dysfunction in ARs may be associated with GRM7 polymorphisms, and redox reactions are involved in regulating the glutamatergic abnormalities. The TT genotype of GRM7 rs11928865 SNP is more vulnerable to damage from the antioxidant and the glutamatergic system.
Collapse
Affiliation(s)
- Jing Wang
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jing-Wu Road, Jinan, 250021, China
| | - Fuyan Li
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jing-Wu Road, Jinan, 250021, China
| | - Fuxin Ren
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jing-Wu Road, Jinan, 250021, China
| | - Wen Ma
- Department of Otolaryngology, The Central Hospital of Jinan City, Shandong University, Jinan, China
| | - Yuxi Liu
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jing-Wu Road, Jinan, 250021, China
| | - Wenjing Zhang
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jing-Wu Road, Jinan, 250021, China
| | - Xi Li
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jing-Wu Road, Jinan, 250021, China
| | - Yanfei Bi
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jing-Wu Road, Jinan, 250021, China
| | - Qian Xin
- Central Laboratory, The Second Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, China.
| | - Fei Gao
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jing-Wu Road, Jinan, 250021, China.
| |
Collapse
|
3
|
Chen T, Yin S, Yue R, Pi C, Zuo Y, Jiang Y, Zheng W, Jiang J, Yang Y, Chu S, Liu K, Wei Y, Zhao L. Novel chlorine-containing curcumin analogue CAK06 promotes neuroprotection and rapid antidepressant through Nrf2-induced anti-inflammatory and antioxidant effects. Neurotherapeutics 2025:e00600. [PMID: 40307166 DOI: 10.1016/j.neurot.2025.e00600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Revised: 04/07/2025] [Accepted: 04/18/2025] [Indexed: 05/02/2025] Open
Abstract
Currently, conventional antidepressants are often limited by poor efficacy and delayed onset, and there is an urgent need for the development of rapid acting antidepressant alternatives. The aim of this study was to develop novel antidepressants with a rapid onset of action by inhibiting inflammation and oxidative stress. A series of chlorine-containing curcumin analogues were designed and synthesized. Among them, compound CAK06 exhibited the highest potency and most robust antidepressant activity, as demonstrated through ABTS free radical ion scavenging assays, relative proliferation rate measurements, and both hydrogen peroxide and corticosterone injury models. In the lipopolysaccharide-induced BV2 cell stress model, treatment with CAK06 resulted in a 69 % decrease in nitric oxide levels and a 52 % reduction in the fluorescence intensity of reactive oxygen species compared to the model group. qRT-PCR results showed that CAK06 upregulated the expression of anti-inflammatory and antioxidant genes while downregulating the expression of pro-inflammatory genes. In the CUMS depression model, CAK06 exerted rapid antidepressant effects after 14 days of oral administration. Notably, after 28 days, CAK06 produced a more pronounced improvement in depression-like behaviors compared to the widely used antidepressant fluoxetine. Mechanistically, molecular docking, Western blot, and Elisa results indicated that CAK06 may alleviate oxidative stress, inflammatory responses, and enhance synaptic plasticity in CUMS mice via the Nrf2-HO-1/BDNF-TrkB pathway. These results suggest that the new compound CAK06 exhibits rapid antidepressant effects, positioning it as a promising novel antidepressant candidate.
Collapse
Affiliation(s)
- Tao Chen
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, 646000, PR China; Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, PR China; Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, PR China; Department of Psychiatry, Fundamental and Clinical Research on Mental Disorders Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, PR China; Pharmacy Department, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing 400014, China
| | - Suyu Yin
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, 646000, PR China; Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, PR China; Department of Psychiatry, Fundamental and Clinical Research on Mental Disorders Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, PR China
| | - Ruxu Yue
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, 646000, PR China; Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, PR China; Department of Psychiatry, Fundamental and Clinical Research on Mental Disorders Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, PR China
| | - Chao Pi
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, 646000, PR China; Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, PR China; Department of Psychiatry, Fundamental and Clinical Research on Mental Disorders Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, PR China
| | - Ying Zuo
- Department of Comprehensive Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, PR China
| | - Yongqiang Jiang
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, 646000, PR China; Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, PR China; Department of Psychiatry, Fundamental and Clinical Research on Mental Disorders Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, PR China
| | - Wenwu Zheng
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Jun Jiang
- Department of Thyroid Surgery, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, PR China
| | - Yan Yang
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, 646000, PR China
| | - Shifeng Chu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Kezhi Liu
- Department of Psychiatry, Fundamental and Clinical Research on Mental Disorders Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, PR China.
| | - Yumeng Wei
- Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, PR China.
| | - Ling Zhao
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, PR China; Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, PR China; Department of Psychiatry, Fundamental and Clinical Research on Mental Disorders Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, PR China.
| |
Collapse
|
4
|
Baidoe-Ansah D, Mirzapourdelavar H, Aleshin S, Schott BH, Seidenbecher C, Kaushik R, Dityatev A. Neurocan regulates axon initial segment organization and neuronal activity. Matrix Biol 2025; 136:22-35. [PMID: 39788215 DOI: 10.1016/j.matbio.2025.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 12/31/2024] [Accepted: 01/06/2025] [Indexed: 01/12/2025]
Abstract
The neural extracellular matrix (ECM) accumulates in the form of perineuronal nets (PNNs), particularly around fast-spiking GABAergic interneurons in the cortex and hippocampus, but also around synapses and in association with the axon initial segments (AIS) and nodes of Ranvier. Increasing evidence highlights the role of Neurocan (Ncan), a brain-specific component of ECM, in the pathophysiology of neuropsychiatric disorders like bipolar disorder and schizophrenia. Ncan localizes at PNNs, perisynaptically, and at the nodes of Ranvier and the AIS, highlighting its potential role in regulating axonal excitability. Here, we used knockdown and knockout approaches in mouse primary cortical neurons in combination with immunocytochemistry, Western blotting and electrophysiological techniques to characterize the role of Ncan in the organization of PNNs and AISs and regulation of neuronal activity. We found that reduced Ncan levels led to remodeling of PNNs around neurons via upregulation of aggrecan mRNA and protein levels, increased expression of activity-dependent c-Fos and FosB genes and elevated spontaneous synaptic activity. The latter correlated with increased levels of ankyrin-G in the AIS, particularly in excitatory neurons, and with the elevated expression of Nav1.6 channels. Our results suggest that Ncan regulates the expression of key proteins in PNNs and AISs and provide new insights into its role in fine-tuning neuronal functions.
Collapse
Affiliation(s)
- David Baidoe-Ansah
- German Center for Neurodegenerative Diseases (DZNE), Helmholtz Association of German Research Centers, Magdeburg, Germany
| | - Hadi Mirzapourdelavar
- German Center for Neurodegenerative Diseases (DZNE), Helmholtz Association of German Research Centers, Magdeburg, Germany
| | - Stepan Aleshin
- German Center for Neurodegenerative Diseases (DZNE), Helmholtz Association of German Research Centers, Magdeburg, Germany
| | - Björn Hendrik Schott
- Leibniz Institute for Neurobiology (LIN), Magdeburg, Germany; Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany; Department of Psychiatry and Psychotherapy, University Medical Center, Göttingen, Germany; German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| | - Constanze Seidenbecher
- Leibniz Institute for Neurobiology (LIN), Magdeburg, Germany; Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany; Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), Jena-Magdeburg-Halle, Germany
| | - Rahul Kaushik
- German Center for Neurodegenerative Diseases (DZNE), Helmholtz Association of German Research Centers, Magdeburg, Germany
| | - Alexander Dityatev
- German Center for Neurodegenerative Diseases (DZNE), Helmholtz Association of German Research Centers, Magdeburg, Germany; Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany; Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany.
| |
Collapse
|
5
|
Santos-Silva T, Lopes CFB, Hazar Ülgen D, Guimarães DA, Guimarães FS, Alberici LC, Sandi C, Gomes FV. Adolescent Stress-Induced Ventral Hippocampus Redox Dysregulation Underlies Behavioral Deficits and Excitatory/Inhibitory Imbalance Related to Schizophrenia. Schizophr Bull 2025; 51:501-512. [PMID: 38525594 PMCID: PMC11908863 DOI: 10.1093/schbul/sbae033] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/26/2024]
Abstract
BACKGROUND AND HYPOTHESIS Redox dysregulation has been proposed as a convergent point of childhood trauma and the emergence of psychiatric disorders, such as schizophrenia (SCZ). A critical region particularly vulnerable to environmental insults during adolescence is the ventral hippocampus (vHip). However, the impact of severe stress on vHip redox states and their functional consequences, including behavioral and electrophysiological changes related to SCZ, are not entirely understood. STUDY DESIGN After exposing adolescent animals to physical stress (postnatal day, PND31-40), we explored social and cognitive behaviors (PND47-49), the basal activity of pyramidal glutamate neurons, the number of parvalbumin (PV) interneurons, and the transcriptomic signature of the vHip (PND51). We also evaluated the impact of stress on the redox system, including mitochondrial respiratory function, reactive oxygen species (ROS) production, and glutathione (GSH) levels in the vHip and serum. STUDY RESULTS Adolescent-stressed animals exhibited loss of sociability, cognitive impairment, and vHip excitatory/inhibitory (E/I) imbalance. Genome-wide transcriptional profiling unveiled the impact of stress on redox system- and synaptic-related genes. Stress impacted mitochondrial respiratory function and changes in ROS levels in the vHip. GSH and glutathione disulfide (GSSG) levels were elevated in the serum of stressed animals, while GSSG was also increased in the vHip and negatively correlated with sociability. Additionally, PV interneuron deficits in the vHip caused by adolescent stress were associated with oxidative stress. CONCLUSIONS Our results highlight the negative impact of adolescent stress on vHip redox regulation and mitochondrial function, which are partially associated with E/I imbalance and behavioral abnormalities related to SCZ.
Collapse
Affiliation(s)
- Thamyris Santos-Silva
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Caio Fábio Baeta Lopes
- Department of Biomolecular Sciences, Ribeirão Preto Pharmaceutical Sciences School, University of São Paulo, Ribeirão Preto, Brazil
| | - Doğukan Hazar Ülgen
- Brain Mind Institute, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Danielle A Guimarães
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Francisco S Guimarães
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Luciane Carla Alberici
- Department of Biomolecular Sciences, Ribeirão Preto Pharmaceutical Sciences School, University of São Paulo, Ribeirão Preto, Brazil
| | - Carmen Sandi
- Brain Mind Institute, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Felipe V Gomes
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
6
|
Jiang S, Jia Q, Peng Z, Zhou Q, An Z, Chen J, Yi Q. Can artificial intelligence be the future solution to the enormous challenges and suffering caused by Schizophrenia? SCHIZOPHRENIA (HEIDELBERG, GERMANY) 2025; 11:32. [PMID: 40021674 PMCID: PMC11871033 DOI: 10.1038/s41537-025-00583-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Accepted: 02/11/2025] [Indexed: 03/03/2025]
Abstract
This study evaluated the potential of artificial intelligence (AI) in the diagnosis, treatment, and prognostic assessment of schizophrenia (SZ) and explored collaborative directions for AI applications in future medical innovations. SZ is a severe mental disorder that causes significant suffering and imposes challenges on patients. With the rapid advancement of machine learning and deep learning technologies, AI has demonstrated notable advantages in the early diagnosis of high-risk populations. By integrating multidimensional biomarkers and linguistic behavior data of patients, AI can provide further objective and precise diagnostic criteria. Moreover, it aids in formulating personalized treatment plans, enhancing therapeutic outcomes, and offering new therapeutic strategies for patients with treatment-resistant SZ. Furthermore, AI excels in developing individualized prognostic plans, which enables the rapid identification of disease progression, accurate prediction of disease trajectory, and timely adjustment of treatment strategies, thereby improving prognosis and facilitating recovery. Despite the immense potential of AI in SZ management, its role as an auxiliary tool must be emphasized, with clinical judgment and compassionate care from healthcare professionals remaining crucial. Future research should focus on optimizing human-machine interactions to achieve efficient AI application in SZ management. The in-depth integration of AI technology into clinical practice will advance the field of SZ, ultimately improving the quality of life and treatment outcomes of patients.
Collapse
Affiliation(s)
- Shijie Jiang
- Department of Medical Psychology, the first Affiliated Hospital of Xinjiang Medical University, Xinjiang Clinical Research Center for Mental Health, Urumqi, 830011, Xinjiang, China
| | - Qiyu Jia
- Department of Trauma Orthopaedics, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, Xinjiang, China
| | - Zhenlei Peng
- Department of Medical Psychology, the first Affiliated Hospital of Xinjiang Medical University, Xinjiang Clinical Research Center for Mental Health, Urumqi, 830011, Xinjiang, China
| | - Qixuan Zhou
- Department of Medical Psychology, the first Affiliated Hospital of Xinjiang Medical University, Xinjiang Clinical Research Center for Mental Health, Urumqi, 830011, Xinjiang, China
| | - Zhiguo An
- Department of Medical Psychology, the first Affiliated Hospital of Xinjiang Medical University, Xinjiang Clinical Research Center for Mental Health, Urumqi, 830011, Xinjiang, China.
| | - Jianhua Chen
- Shanghai Institute of Traditional Chinese Medicine for Mental Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
| | - Qizhong Yi
- Department of Medical Psychology, the first Affiliated Hospital of Xinjiang Medical University, Xinjiang Clinical Research Center for Mental Health, Urumqi, 830011, Xinjiang, China.
| |
Collapse
|
7
|
Hagen JM, Cornelissen A, Veeneman RR, van der Heijden HS, Sutterland AL, Vermeulen JM, Tan HL, de Haan L. Skin advanced glycation end products, indicating cumulative oxidative stress, associated with schizophrenia but not with psychosis-like experiences. Schizophr Res 2025; 276:237-242. [PMID: 39938246 DOI: 10.1016/j.schres.2025.01.027] [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: 10/14/2024] [Revised: 01/17/2025] [Accepted: 01/31/2025] [Indexed: 02/14/2025]
Abstract
BACKGROUND Excessive oxidative stress is one of the proposed pathophysiological mechanisms in the development of schizophrenia. As a result of oxidative stress, advanced glycation end products (AGEs) are formed. Skin autofluorescence measures of AGEs (SAF-AGEs) serve as indicator of cumulative oxidative stress and are associated with occurrence of psychotic disorders. Here, the association between SAF-AGE level as a proxy for cumulative oxidative stress and subsequent psychosis-like experiences (PLEs) in the general population is investigated. METHODS Participants of the Lifelines study, a population-based cohort study in the Netherlands, who had completed a SAF-AGE measurement at baseline and the Psychosis Screening Questionnaire (PSQ) at 10 year follow-up were included. Association between SAF-AGE level and PSQ was assessed in a linear mixed model, adjusted for family ties and sex. Sociodemographic factors and polygenic score for schizophrenia (PGS-SZ) were added to the model hierarchically. RESULTS Of the 30,862 included participants (59 % female, mean age of 46), 1318 (4.3 %) experienced PLEs. Age-adjusted Z-score of SAF-AGE level (n = 83,016) and self-reported schizophrenia diagnosis showed a statistically significant association (estimate = 0.65 [95 % CI 0.40, - 0.89], p < .001). SAF-AGE level was not significantly associated to PSQ (estimate = 2.88-3 [95 % CI -4.98-51, 5.81-3], p = .05). The adjusted model showed similar results. CONCLUSIONS Results indicate that excessive oxidative stress is not involved in development of PLEs, as opposed to in case of clinical psychosis.
Collapse
Affiliation(s)
- J M Hagen
- Arkin Mental Health Institute, Amsterdam, the Netherlands; Amsterdam UMC, University of Amsterdam, Department of Psychiatry, Amsterdam, the Netherlands.
| | - A Cornelissen
- Amsterdam UMC, University of Amsterdam, Department of Psychiatry, Amsterdam, the Netherlands
| | - R R Veeneman
- Amsterdam UMC, University of Amsterdam, Department of Psychiatry, Amsterdam, the Netherlands; Amsterdam UMC location University of Amsterdam, Department of Psychiatry, Genetic Epidemiology, Amsterdam, the Netherlands
| | - H S van der Heijden
- Amsterdam UMC, University of Amsterdam, Department of Psychiatry, Amsterdam, the Netherlands
| | - A L Sutterland
- Amsterdam UMC, University of Amsterdam, Department of Psychiatry, Amsterdam, the Netherlands
| | - J M Vermeulen
- Amsterdam UMC, University of Amsterdam, Department of Psychiatry, Amsterdam, the Netherlands
| | - H L Tan
- Amsterdam UMC, University of Amsterdam, Department of Cardiology, Heart Center, Amsterdam, the Netherlands; Netherlands Heart Institute, Utrecht, the Netherlands
| | - L de Haan
- Amsterdam UMC, University of Amsterdam, Department of Psychiatry, Amsterdam, the Netherlands
| |
Collapse
|
8
|
Gee A, Dazzan P, Grace AA, Modinos G. Corticolimbic circuitry as a druggable target in schizophrenia spectrum disorders: a narrative review. Transl Psychiatry 2025; 15:21. [PMID: 39856031 PMCID: PMC11760974 DOI: 10.1038/s41398-024-03221-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 12/06/2024] [Accepted: 12/27/2024] [Indexed: 01/27/2025] Open
Abstract
Schizophrenia spectrum disorders (SSD) involve disturbances in the integration of perception, emotion and cognition. The corticolimbic system is an interacting set of cortical and subcortical brain regions critically involved in this process. Understanding how neural circuitry and molecular mechanisms within this corticolimbic system may contribute to the development of not only positive symptoms but also negative and cognitive deficits in SSD has been a recent focus of intense research, as the latter are not adequately treated by current antipsychotic medications and are more strongly associated with poorer functioning and long-term outcomes. This review synthesises recent developments examining corticolimbic dysfunction in the pathophysiology of SSD, with a focus on neuroimaging advances and related novel methodologies that enable the integration of data across different scales. We then integrate how these findings may inform the identification of novel therapeutic and preventive targets for SSD symptomatology. A range of pharmacological interventions have shown initial promise in correcting corticolimbic dysfunction and improving negative, cognitive and treatment-resistant symptoms. We discuss current challenges and opportunities for improving the still limited translation of these research findings into clinical practice. We argue how our knowledge of the role of corticolimbic dysfunction can be improved by combining multiple research modalities to examine hypotheses across different spatial and temporal scales, combining neuroimaging with experimental interventions and utilising large-scale consortia to advance biomarker identification. Translation of these findings into clinical practice will be aided by consideration of optimal intervention timings, biomarker-led patient stratification, and the development of more selective medications.
Collapse
Affiliation(s)
- Abigail Gee
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Paola Dazzan
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Anthony A Grace
- Departments of Neuroscience, Psychiatry and Psychology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Gemma Modinos
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom.
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK.
| |
Collapse
|
9
|
Suschana E, Anderson T, Hong C, Narikatte A, Silverberg J, Sharma MS. The role of anti-inflammatory diets and supplementation in metabolic syndrome and symptom remission in adults with schizophrenia: a systematic review. Front Psychiatry 2025; 15:1506353. [PMID: 39839138 PMCID: PMC11747649 DOI: 10.3389/fpsyt.2024.1506353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 12/02/2024] [Indexed: 01/23/2025] Open
Abstract
Introduction Immune dysregulation and chronic inflammation have been hypothesized as potential pathways in metabolic syndrome and schizophrenia. Anti-inflammatory diets have the potential not only to treat metabolic syndrome but also to reduce the symptom burden in schizophrenia. The aim of this systematic review was to investigate the role of anti-inflammatory diets and vitamin supplementation in the management of metabolic syndrome and in symptom remission in people with schizophrenia. Methods This systematic review included research articles from PubMed, EMBASE, Scopus, PsycINFO, and the Cochrane Central Register for Controlled Trials. The primary outcomes were markers of metabolic syndrome and symptoms of psychosis. Results Our search identified 2,124 potential studies, of which 1,559 were screened based on the title and abstract, resulting in 81 full-text articles assessed for eligibility. A total of 17 studies were included, which demonstrated mixed findings on the impacts of anti-inflammatory diet interventions on metabolic markers and symptom remission in schizophrenia. Prebiotic, probiotic, and fish oil supplementation showed improvements in metabolic markers. Fish oil and vitamin D supplementation demonstrated symptom remission in some trials. Conclusion It is important to consider that people with schizophrenia may experience common external barriers that hinder adherence to dietary interventions. These findings underscore the need for larger trials with standardized dietary protocols and consistent metabolic and symptom outcome measures in order to better understand the potential role of anti-inflammatory interventions in this population. Systematic review registration https://www.crd.york.ac.uk/prospero/, identifier CRD42024511596.
Collapse
Affiliation(s)
- Elizabeth Suschana
- University of Connecticut School of Medicine, Farmington, CT, United States
| | - Thea Anderson
- University of Connecticut School of Medicine, Farmington, CT, United States
| | - Catriona Hong
- University of Connecticut School of Medicine, Farmington, CT, United States
| | - Arun Narikatte
- Frank H. Netter MD School of Medicine, Quinnipiac University, North Haven, CT, United States
| | - Jillian Silverberg
- Library Services, University of Connecticut School of Medicine, Farmington, CT, United States
| | - Manu Suresh Sharma
- Department of Psychiatry, Institute of Living, Hartford, CT, United States
| |
Collapse
|
10
|
Camporesi S, Xin L, Golay P, Eap CB, Cleusix M, Cuenod M, Fournier M, Hashimoto K, Jenni R, Ramain J, Restellini R, Solida A, Conus P, Do KQ, Khadimallah I. Neurocognition and NMDAR co-agonists pathways in individuals with treatment resistant first-episode psychosis: a 3-year follow-up longitudinal study. Mol Psychiatry 2024; 29:3669-3679. [PMID: 38849515 PMCID: PMC11541217 DOI: 10.1038/s41380-024-02631-4] [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: 01/09/2024] [Revised: 05/15/2024] [Accepted: 05/28/2024] [Indexed: 06/09/2024]
Abstract
This study aims to determine whether 1) individuals with treatment-resistant schizophrenia display early cognitive impairment compared to treatment-responders and healthy controls and 2) N-methyl-D-aspartate-receptor hypofunction is an underlying mechanism of cognitive deficits in treatment-resistance. In this case‒control 3-year-follow-up longitudinal study, n = 697 patients with first-episode psychosis, aged 18 to 35, were screened for Treatment Response and Resistance in Psychosis criteria through an algorithm that assigns patients to responder, limited-response or treatment-resistant category (respectively resistant to 0, 1 or 2 antipsychotics). Assessments at baseline: MATRICS Consensus Cognitive Battery; N-methyl-D-aspartate-receptor co-agonists biomarkers in brain by MRS (prefrontal glutamate levels) and plasma (D-serine and glutamate pathways key markers). Patients were compared to age- and sex-matched healthy controls (n = 114). Results: patient mean age 23, 27% female. Treatment-resistant (n = 51) showed lower scores than responders (n = 183) in processing speed, attention/vigilance, working memory, verbal learning and visual learning. Limited responders (n = 59) displayed an intermediary phenotype. Treatment-resistant and limited responders were merged in one group for the subsequent D-serine and glutamate pathway analyses. This group showed D-serine pathway dysregulation, with lower levels of the enzymes serine racemase and serine-hydroxymethyltransferase 1, and higher levels of the glutamate-cysteine transporter 3 than in responders. Better cognition was associated with higher D-serine and lower glutamate-cysteine transporter 3 levels only in responders; this association was disrupted in the treatment resistant group. Treatment resistant patients and limited responders displayed early cognitive and persistent functioning impairment. The dysregulation of NMDAR co-agonist pathways provides underlying molecular mechanisms for cognitive deficits in treatment-resistant first-episode psychosis. If replicated, our findings would open ways to mechanistic biomarkers guiding response-based patient stratification and targeting cognitive improvement in clinical trials.
Collapse
Affiliation(s)
- Sara Camporesi
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
- Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
- Department of psychiatry and Emergency Department, Geneva University Hospital, Geneva, Switzerland
| | - Lijing Xin
- Center for Biomedical Imaging, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Philippe Golay
- Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
| | - Chin Bin Eap
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, Geneva, Switzerland
- Center for Research and Innovation in Clinical Pharmaceutical Sciences, University of Lausanne, Lausanne, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, University of Lausanne, Geneva, Switzerland
| | - Martine Cleusix
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
| | - Michel Cuenod
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
| | - Margot Fournier
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
| | - Kenji Hashimoto
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan
| | - Raoul Jenni
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
| | - Julie Ramain
- Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
- Training and Research Institute in Mental Health (IFRSM), Neuchâtel Centre of Psychiatry, Neuchâtel, Switzerland
| | - Romeo Restellini
- Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
- Emergency medicine department, Geneva University Hospital, Geneva, Switzerland
| | - Alessandra Solida
- Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
- Psychiatry Department for Adults 2, Neuchâtel Centre of Psychiatry, Prefargier, Switzerland
| | - Philippe Conus
- Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
| | - Kim Q Do
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
| | - Ines Khadimallah
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland.
- Translational Research Center, University Hospital of Psychiatry, University of Bern, Bern, Switzerland.
| |
Collapse
|
11
|
Mendez-Victoriano G, Zhu Y, Middleton F, Massa PT, Ajulu K, Webster MJ, Weickert CS. Increased Parenchymal Macrophages are associated with decreased Tyrosine Hydroxylase mRNA levels in the Substantia Nigra of people with Schizophrenia and Bipolar Disorder. Psychiatry Res 2024; 340:116141. [PMID: 39153291 DOI: 10.1016/j.psychres.2024.116141] [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: 10/11/2023] [Revised: 07/09/2024] [Accepted: 08/10/2024] [Indexed: 08/19/2024]
Abstract
Increased activation of inflammatory macrophages and altered expression of dopamine markers are found in the midbrains of people with schizophrenia (SZ). The relationship of midbrain macrophages to dopamine neurons has not been explored, nor is it known if changes in midbrain macrophages are also present in bipolar disorder (BD) or major depressive disorder (MDD). Herein, we determined whether there were differences in CD163+ cell density in the Substantia Nigra (SN), and cerebral peduncles (CP) of SZ, BD, and MDD compared to controls (CTRL). We also analyzed whether CD163 protein and dopamine-synthesizing enzyme tyrosine hydroxylase (TH) mRNA levels differed among diagnostic groups and if they correlated with the density of macrophages. Overall, perivascular CD163+ cell density was higher in the gray matter (SN) than in the white matter (CP). Compared to CTRL, we found increased density of parenchymal CD163+ cells in the SN of the three psychiatric groups and increased CD163 protein levels in SZ. CD163 protein was positively correlated with density of perivascular CD163+ cells. TH mRNA was reduced in SZ and BD and negatively correlated with parenchymal CD163+ cell density. We provide the first quantitative and molecular evidence of an increase in the density of parenchymal macrophages in the midbrain of major mental illnesses and show that the presence of these macrophages may negatively impact dopaminergic neurons.
Collapse
Affiliation(s)
- Gerardo Mendez-Victoriano
- Department of Neuroscience and Physiology, Upstate Medical University, Syracuse, NY, USA; Neuroscience Research Australia, Sydney, NSW, Australia; School of Clinical Medicine, Faculty of Medicine & Health, University of New South Wales, Sydney, NSW 2052, Australia
| | - Yunting Zhu
- Department of Neuroscience and Physiology, Upstate Medical University, Syracuse, NY, USA
| | - Frank Middleton
- Department of Neuroscience and Physiology, Upstate Medical University, Syracuse, NY, USA
| | - Paul T Massa
- Department of Neuroscience and Physiology, Upstate Medical University, Syracuse, NY, USA
| | - Kachikwulu Ajulu
- Department of Neuroscience and Physiology, Upstate Medical University, Syracuse, NY, USA
| | - Maree J Webster
- Laboratory of Brain Research, Stanley Medical Research Institute, 9800 Medical Center Drive, Rockville, MD, USA
| | - Cynthia S Weickert
- Department of Neuroscience and Physiology, Upstate Medical University, Syracuse, NY, USA; Neuroscience Research Australia, Sydney, NSW, Australia; School of Clinical Medicine, Faculty of Medicine & Health, University of New South Wales, Sydney, NSW 2052, Australia.
| |
Collapse
|
12
|
Uliana DL, Lisboa JRF, Gomes FV, Grace AA. The excitatory-inhibitory balance as a target for the development of novel drugs to treat schizophrenia. Biochem Pharmacol 2024; 228:116298. [PMID: 38782077 PMCID: PMC11410545 DOI: 10.1016/j.bcp.2024.116298] [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: 02/01/2024] [Revised: 05/13/2024] [Accepted: 05/16/2024] [Indexed: 05/25/2024]
Abstract
The intricate balance between excitation and inhibition (E/I) in the brain plays a crucial role in normative information processing. Dysfunctions in the E/I balance have been implicated in various psychiatric disorders, including schizophrenia (SCZ). In particular, abnormalities in GABAergic signaling, specifically in parvalbumin (PV)-containing interneurons, have been consistently observed in SCZ pathophysiology. PV interneuron function is vital for maintaining an ideal E/I balance, and alterations in PV interneuron-mediated inhibition contribute to circuit deficits observed in SCZ, including hippocampus hyperactivity and midbrain dopamine system overdrive. While current antipsychotic medications primarily target D2 dopamine receptors and are effective primarily in treating positive symptoms, novel therapeutic strategies aiming to restore the E/I balance could potentially mitigate not only positive symptoms but also negative symptoms and cognitive deficits. This could involve, for instance, increasing the inhibitory drive onto excitatory neurons or decreasing the putative enhanced pyramidal neuron activity due to functional loss of PV interneurons. Compounds targeting the glycine site at glutamate NMDA receptors and muscarinic acetylcholine receptors on PV interneurons that can increase PV interneuron drive, as well as drugs that increase the postsynaptic action of GABA, such as positive allosteric modulators of α5-GABA-A receptors, and decrease glutamatergic output, such as mGluR2/3 agonists, represent promising approaches. Preventive strategies aiming at E/I balance also represent a path to reduce the risk of transitioning to SCZ in high-risk individuals. Therefore, compounds with novel mechanisms targeting E/I balance provide optimism for more effective and tailored interventions in the management of SCZ.
Collapse
Affiliation(s)
- Daniela L Uliana
- Departments of Neuroscience, Psychiatry and Psychology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Joao Roberto F Lisboa
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Felipe V Gomes
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Anthony A Grace
- Departments of Neuroscience, Psychiatry and Psychology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
13
|
Li X, Wang X, Yang Y, Zhou J, Wu X, Zhao J, Zhang J, Guo X, Shao M, Song M, Su X, Han Y, Liu Q, Chen T, Zhang L, Liu B, Yue W, Lv L, Li W. Elevated plasma matrix metalloproteinase 9 in schizophrenia patients associated with poor antipsychotic treatment response and white matter density deficits. SCHIZOPHRENIA (HEIDELBERG, GERMANY) 2024; 10:71. [PMID: 39191778 DOI: 10.1038/s41537-024-00494-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 08/09/2024] [Indexed: 08/29/2024]
Abstract
Oxidative stress and neuroinflammation contribute to schizophrenia (SCZ) pathology and may influence treatment efficacy. Matrix metalloproteinase 9 (MMP9) is a critical molecular node mediating the interaction between oxidative stress and inflammation, and so may influence treatment efficacy. Here we examined the associations of plasma MMP9 concentration with antipsychotic drug responses, clinical symptoms, and brain structure. A total of 129 healthy controls and 124 patients with SCZ were included in this study. Patients were monitored clinically during 8 weeks of antipsychotic treatment and classified as poor responders (n = 49) or good responders (n = 75). We then compared plasma MMP9 concentrations in healthy controls at baseline and both SCZ responder groups at baseline and after the 8-week antipsychotic treatment regimen. Cognitive function was also examined using the MATRICS Consensus Cognitive Battery. In addition, we extracted regional white matter density from magnetic resonance images of patients. Compared to healthy controls, plasma MMP9 levels were significantly elevated in poor responders at baseline and negatively correlated with both white matter density in the right superior temporal gyrus and the change in cognitive symptoms after treatment. Conversely, there was no significant difference in plasma MMP9 between good responders and healthy controls, and no associations of plasma MMP9 with cognitive symptoms or regional white matter density among good responders. Elevated plasma MMP9 is associated with poor antipsychotic drug efficacy and white matter deficits in SCZ patients, and so may be a useful biomarker to guide personalized treatment.
Collapse
Affiliation(s)
- Xiaojing Li
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453002, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, 453002, China
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, 453002, China
- Henan Collaborative Innovation Center of Prevention and Treatment of Mental Disorder, Xinxiang, 453002, China
| | - Xiujuan Wang
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453002, China
| | - Yongfeng Yang
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453002, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, 453002, China
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, 453002, China
| | - Jiahui Zhou
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453002, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, 453002, China
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, 453002, China
| | - Xufei Wu
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, 453002, China
| | - Jingyuan Zhao
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453002, China
| | - Jianhong Zhang
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453002, China
| | - Xiaoge Guo
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453002, China
| | - Minglong Shao
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453002, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, 453002, China
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, 453002, China
| | - Meng Song
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453002, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, 453002, China
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, 453002, China
| | - Xi Su
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453002, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, 453002, China
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, 453002, China
| | - Yong Han
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453002, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, 453002, China
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, 453002, China
| | - Qing Liu
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453002, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, 453002, China
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, 453002, China
| | - Tengfei Chen
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453002, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, 453002, China
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, 453002, China
| | - Luwen Zhang
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453002, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, 453002, China
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, 453002, China
| | - Bing Liu
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, 100875, Beijing, China
- Chinese Institute for Brain Research, 102206, Beijing, China
| | - Weihua Yue
- Institute of Mental Health, Peking University, 100191, Beijing, China
- Key Laboratory for Mental Health, Ministry of Health, 100191, Beijing, China
| | - Luxian Lv
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453002, China.
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, 453002, China.
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, 453002, China.
| | - Wenqiang Li
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453002, China.
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, 453002, China.
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, 453002, China.
- Henan Collaborative Innovation Center of Prevention and Treatment of Mental Disorder, Xinxiang, 453002, China.
| |
Collapse
|
14
|
Lorenc-Koci E, Górny M, Chwatko G, Kamińska K, Iciek M, Rogóż Z. The effect of phencyclidine-mediated blockade of NMDA receptors in the early postnatal period on glutathione and sulfur amino acid levels in the rat brain as a potential causative factor of schizophrenia-like behavior in adulthood. Pharmacol Rep 2024; 76:863-877. [PMID: 38904712 PMCID: PMC11294273 DOI: 10.1007/s43440-024-00607-3] [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: 03/12/2024] [Revised: 05/21/2024] [Accepted: 05/22/2024] [Indexed: 06/22/2024]
Abstract
BACKGROUND Phencyclidine, an NMDA receptor antagonist, is frequently used to model behavioral and neurochemical changes correlated with schizophrenia in laboratory animals. The present study aimed to examine the effects of repeated administration of phencyclidine during early postnatal development on the contents of glutathione and sulfur-containing amino acids, as well as the activity of antioxidant enzymes in the brain of 12-day-old rats, and schizophrenia-like symptoms in adulthood. METHODS Male Sprague-Dawley pups were administered phencyclidine (10 mg/kg) or saline subcutaneously on the postnatal days p2, p6, p9 and p12. In 12-day-old pups, 4 h after the last dose of phencyclidine, the levels of glutathione, cysteine, methionine, and homocysteine, and the enzymatic activity of superoxide dismutase (SOD), glutathione peroxidase (GPx), and glutathione reductase (GR) were measured in the frontal cortex, hippocampus, and striatum. In 70-72-day-old rats, schizophrenia-like symptoms were assessed using behavioral tests. RESULTS Biochemical data showed that perinatal phencyclidine treatment significantly reduced glutathione and cysteine levels in all brain structures studied, methionine was diminished in the striatum, and homocysteine in both the frontal cortex and striatum. GR activity was increased in the frontal cortex while SODactivity was decreased in the hippocampus. Behaviorally, perinatal phencyclidine induced long-term deficits in social and cognitive function and a decrease in locomotor activity assessed as the time of walking. Finally, perinatal treatment with phencyclidine resulted in a significant reduction in body weight gain over time. CONCLUSION Our research provides further evidence for the usefulness of the phencyclidine-induced neurodevelopmental model of schizophrenia for studying the pathogenesis of schizophrenia.
Collapse
Affiliation(s)
- Elżbieta Lorenc-Koci
- Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna Street, Kraków, 31-343, Poland.
| | - Magdalena Górny
- The Chair of Medical Biochemistry, Jagiellonian University Medical College, 7 Kopernika Street, Kraków, 31-034, Poland
| | - Grażyna Chwatko
- Department of Environmental Chemistry, University of Łódź, 163 Pomorska Street, Łódź, 90-236, Poland
| | - Kinga Kamińska
- Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna Street, Kraków, 31-343, Poland
| | - Małgorzata Iciek
- The Chair of Medical Biochemistry, Jagiellonian University Medical College, 7 Kopernika Street, Kraków, 31-034, Poland
| | - Zofia Rogóż
- Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna Street, Kraków, 31-343, Poland
| |
Collapse
|
15
|
Idotta C, Pagano MA, Tibaldi E, Cadamuro M, Saetti R, Silvestrini M, Pigato G, Leanza L, Peruzzo R, Meneghetti L, Piazza S, Meneguzzo P, Favaro A, Grassi L, Toffanin T, Brunati AM. Neural stem/progenitor cells from olfactory neuroepithelium collected by nasal brushing as a cell model reflecting molecular and cellular dysfunctions in schizophrenia. World J Biol Psychiatry 2024; 25:317-329. [PMID: 38869228 DOI: 10.1080/15622975.2024.2357096] [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: 01/11/2024] [Revised: 05/02/2024] [Accepted: 05/15/2024] [Indexed: 06/14/2024]
Abstract
OBJECTIVES Neural stem/progenitor cells derived from olfactory neuroepithelium (hereafter olfactory neural stem/progenitor cells, ONSPCs) are emerging as a potential tool in the exploration of psychiatric disorders. The present study intended to assess whether ONSPCs could help discern individuals with schizophrenia (SZ) from non-schizophrenic (NS) subjects by exploring specific cellular and molecular features. METHODS ONSPCs were collected from 19 in-patients diagnosed with SZ and 31 NS individuals and propagated in basal medium. Mitochondrial ATP production, expression of β-catenin and cell proliferation, which are described to be altered in SZ, were examined in freshly isolated or newly thawed ONSPCs after a few culture passages. RESULTS SZ-ONSPCs exhibited a lower mitochondrial ATP production and insensitivity to agents capable of positively or negatively affecting β-catenin expression with respect to NS-ONSPCs. As to proliferation, it declined in SZ-ONSPCs as the number of culture passages increased compared to a steady level of growth shown by NS-ONSPCs. CONCLUSIONS The ease and safety of sample collection as well as the differences observed between NS- and SZ-ONSPCs, may lay the groundwork for a new approach to obtain biological material from a large number of living individuals and gain a better understanding of the mechanisms underlying SZ pathophysiology.
Collapse
Affiliation(s)
- Carlo Idotta
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | - Mario Angelo Pagano
- Department of Molecular Medicine, University of Padua, Padua, Italy
- Department of Neuroscience and Rehabilitation, Institute of Psychiatry, University of Ferrara, Ferrara, Italy
| | - Elena Tibaldi
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | | | - Roberto Saetti
- Department of Otolaryngology, San Bortolo Hospital, ULSS 8 Berica, Vicenza, Italy
| | - Marina Silvestrini
- Department of Otolaryngology, San Bortolo Hospital, ULSS 8 Berica, Vicenza, Italy
| | | | - Luigi Leanza
- Department of Biology, University of Padua, Padua, Italy
| | - Roberta Peruzzo
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | | | - Stefano Piazza
- Department of Mental Health, ULSS 8 Berica, Vicenza, Italy
| | - Paolo Meneguzzo
- Department of Neuroscience, University of Padua, Padua, Italy
- Padova Neuroscience Center, University of Padua, Padua, Italy
| | - Angela Favaro
- Department of Neuroscience, University of Padua, Padua, Italy
- Padova Neuroscience Center, University of Padua, Padua, Italy
| | - Luigi Grassi
- Department of Neuroscience and Rehabilitation, Institute of Psychiatry, University of Ferrara, Ferrara, Italy
| | - Tommaso Toffanin
- Department of Neuroscience and Rehabilitation, Institute of Psychiatry, University of Ferrara, Ferrara, Italy
| | | |
Collapse
|
16
|
Kim E, Redwood S, Liu F, Roche DJO, Chen S, Bentley WE, Eaton WW, Čiháková D, Talor MV, Kelly DL, Payne GF. Pilot study indicates that a gluten-free diet lowers oxidative stress for gluten-sensitive persons with schizophrenia. Schizophr Res 2024; 269:71-78. [PMID: 38749320 PMCID: PMC11215979 DOI: 10.1016/j.schres.2024.05.001] [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: 01/22/2024] [Revised: 04/19/2024] [Accepted: 05/02/2024] [Indexed: 05/22/2024]
Abstract
One-third of people with schizophrenia have elevated levels of anti-gliadin antibodies (AGA IgG). A 5-week randomized double-blind pilot study was performed in 2014-2017 in an inpatient setting to test the effect of a gluten-free diet (GFD) on participants with schizophrenia or schizoaffective disorder who also had elevated AGA IgG (≥ 20 U) but were negative for celiac disease. This earlier pilot study reported that the GFD-group showed improved gastrointestinal and psychiatric symptoms, and also improvements in TNF-α and the inflammatory cytokine IL-23. Here, we performed measurements of these banked plasma samples to detect levels of oxidative stress (OxSt) using a recently developed iridium (Ir)-reducing capacity assay. Triplicate measurements of these samples showed an Intraclass Correlation Coefficient of 0.84 which indicates good reproducibility. Further, a comparison of the OxSt measurements at the baseline and 5-week end-point for this small sample size shows that the GFD-group (N = 7) had lowered OxSt levels compared to the gluten-containing diet group (GCD; N = 9; p = 0.05). Finally, we showed that improvements in OxSt over these 5 weeks were correlated to improvements in gastrointestinal (r = +0.64, p = 0.0073) and psychiatric (r = +0.52, p = 0.039) symptoms. Also, we showed a possible association between the decrease in OxSt and the lowered levels of IL-23 (r = +0.44, p = 0.087), although without statistical significance. Thus, the Ir-reducing capacity assay provides a simple, objective measure of OxSt with the results providing further evidence that inflammation, redox dysregulation and OxSt may mediate interactions between the gut and brain.
Collapse
Affiliation(s)
- Eunkyoung Kim
- Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, MD 20742, United States; Robert E. Fischell Institute for Biomedical Devices, University of Maryland, College Park, MD 20742, United States
| | - Sidney Redwood
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, United States
| | - Fang Liu
- Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD 21228, United States
| | - Daniel J O Roche
- Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD 21228, United States
| | - Shuo Chen
- Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD 21228, United States
| | - William E Bentley
- Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, MD 20742, United States; Robert E. Fischell Institute for Biomedical Devices, University of Maryland, College Park, MD 20742, United States; Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, United States
| | - William W Eaton
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, United States
| | - Daniela Čiháková
- Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, United States; Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD 21205, United States
| | - Monica V Talor
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD 21205, United States
| | - Deanna L Kelly
- Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD 21228, United States.
| | - Gregory F Payne
- Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, MD 20742, United States; Robert E. Fischell Institute for Biomedical Devices, University of Maryland, College Park, MD 20742, United States.
| |
Collapse
|
17
|
Hajizadeh Moghaddam A, Malekzadeh Estalkhi F, Khanjani Jelodar S, Ahmed Hasan T, Farhadi-Pahnedari S, Karimian M. Neuroprotective effects of alpha-pinene against behavioral deficits in ketamine-induced mice model of schizophrenia: Focusing on oxidative stress status. IBRO Neurosci Rep 2024; 16:182-189. [PMID: 38318342 PMCID: PMC10839590 DOI: 10.1016/j.ibneur.2023.12.012] [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: 08/02/2023] [Revised: 12/11/2023] [Accepted: 12/30/2023] [Indexed: 02/07/2024] Open
Abstract
Schizophrenia (SCZ) is a profound neurological disorder that affects approximately 1% of the global population. Alpha-pinene (α-pinene) is a natural and active monoterpene found in coniferous tree oil, primarily pine, with diverse pharmacological characteristics, including antioxidative, anxiolytic, and antidepressant properties. This research study delves into the neuroprotective effects of α-pinene on oxidative stress, memory deficits, and depressive and anxiety-like behaviors in a ketamine-induced mice model of SCZ using male mice. The mice were randomly divided into six groups: vehicle, control, positive control, ketamine, α-pinene at 50 mg/kg, and α-pinene at 100 mg/kg. Treatment of the ketamine-induced mice model of SCZ with α-pinene yielded significant improvements in depressive and anxiety-like behaviors and cognitive impairments. Furthermore, it significantly elevated glutathione (GSH) levels, total antioxidant capacity (TAC), dopamine levels, catalase (CAT), and superoxide dismutase (SOD) activities while markedly reducing malondialdehyde (MDA) levels. The current study establishes that α-pinene treatment effectively mitigates oxidative damage, cognitive deficits, and depressive and anxiogenic-like behaviors in the brains of ketamine-treated mice. Therefore, α-pinene treatment is an efficacious approach to forestall the neurobehavioral and neurobiochemical adverse effects of the ketamine-induced SCZ model of mice.
Collapse
Affiliation(s)
| | | | | | - Tabarek Ahmed Hasan
- Department of Animal Biology, Faculty of Basic Sciences, University of Mazandaran, Babolsar, Iran
| | | | - Mohammad Karimian
- Department of Molecular and Cell Biology, Faculty of Basic Sciences, University of Mazandaran, Babolsar, Iran
| |
Collapse
|
18
|
Li H, Huang Y, Liang L, Li H, Li S, Feng Y, Feng S, Wu K, Wu F. The relationship between the gut microbiota and oxidative stress in the cognitive function of schizophrenia: A pilot study in China. Schizophr Res 2024; 267:444-450. [PMID: 38643725 DOI: 10.1016/j.schres.2024.03.053] [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: 03/22/2024] [Accepted: 03/31/2024] [Indexed: 04/23/2024]
Abstract
Cognitive impairment is a core symptom of schizophrenia. The gut microbiota (GM) and oxidative stress may play important roles in the pathophysiological mechanisms of cognitive impairment. This study aimed to explore the relationship between GM and oxidative stress in the cognitive function of schizophrenia. GM obtained by 16S RNA sequencing and serum superoxide dismutase (SOD) levels from schizophrenia patients (N = 68) and healthy controls (HCs, N = 72) were analyzed. All psychiatric symptoms were assessed using the Positive and Negative Syndrome Scale (PANSS). Cognitive function was assessed using the MATRICS Consensus Cognitive Battery (MCCB). Correlation analysis was used to explore the relationship between GM, SOD, and cognitive function. Machine learning models were used to identify potential biomarkers. Compared to HCs, the relative abundances of Collinsella, undefined Ruminococcus, Lactobacillus, Eubacterium, Mogibacterium, Desulfovibrio, Bulleidia, Succinivibrio, Corynebacterium, and Atopobium were higher in patients with schizophrenia, but Faecalibacterium, Anaerostipes, Turicibacter, and Ruminococcus were lower. In patients with schizophrenia, the positive factor, general factor, and total score of MCCB positively correlated with Lactobacillus, Collinsella, and Lactobacillus, respectively; SOD negatively correlated with Eubacterium, Collinsella, Lactobacillus, Corynebacterium, Bulleidia, Mogibacterium, and Succinivibrio, but positively correlated with Faecalibacterium, Ruminococcus, and MCCB verbal learning index scores; Faecalibacterium and Turicibacter were positively correlated with MCCB visual learning index scores and speed of processing index scores, respectively. Our findings revealed a correlation between SOD and GM and confirmed that cognitive dysfunction in patients with schizophrenia involves abnormal SOD levels and GM changes.
Collapse
Affiliation(s)
- Hehua Li
- The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yuanyuan Huang
- The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Liqin Liang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, China
| | - Hanqiu Li
- The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Shijia Li
- Swammerdam Institute for Life Sciences (SILS)-University of Amsterdam, Amsterdam, the Netherlands
| | - Yangdong Feng
- The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Shixuan Feng
- The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Kai Wu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China.
| | - Fengchun Wu
- The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
19
|
Bahnamiri PJ, Hajizadeh Moghaddam A, Ranjbar M, Nazifi E. Effects of Nostoc commune extract on the cerebral oxidative and neuroinflammatory status in a mice model of schizophrenia. Biochem Biophys Rep 2024; 37:101594. [PMID: 38371525 PMCID: PMC10873873 DOI: 10.1016/j.bbrep.2023.101594] [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: 08/27/2023] [Revised: 11/19/2023] [Accepted: 11/22/2023] [Indexed: 02/20/2024] Open
Abstract
Cyanobacterium Nostoc commune has long been used to alleviate various diseases. This research examines the effects of Nostoc commune extract (NCE) against behavioral disorders, cerebral oxidative stress, and inflammatory damage in the ketamine-induced schizophrenia model. Oral NCE administration (70 and 150 mg/kg/d) is performed after intraperitoneal ketamine injection (20 mg/kg) for 14 consecutive days. The forced swimming and open field tests are used to assess schizophrenia-like behaviors. After the behavioral test, dopamine (DA) level, oxidative stress markers, as well as the interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) expression are measured in the cerebral cortex. The results show that NCE treatment ameliorates KET-induced anxiety and depressive-like behaviors in OFT and FST, respectively. NCE considerably decreases the malondialdehyde (MDA) and DA levels and IL-6 and TNF-α expressions in mice with schizophrenia-like symptoms. Also, a significant increase is observed in the glutathione (GSH) level and catalase (CAT), superoxide dismutase (SOD), and glutathione reductase (GRx) activity in cerebral tissue. The present study shows that NCE treatment effectively improves KET-induced schizophrenia-like behaviors and oxidative and inflammatory damage. Therefore, NCE, via its bioactive constituents, could have strong neuroprotective effects in the schizophrenia-like model.
Collapse
Affiliation(s)
| | | | - Mojtaba Ranjbar
- Faculty of Biotechnology, Amol University of Special Modern Technologies, Amol, Iran
| | - Ehsan Nazifi
- Department of Plant Sciences, University of Mazandaran, Babolsar, Iran
| |
Collapse
|
20
|
Guan X, Chen Y, Wang X, Xiu M, Wu F, Zhang X. Total antioxidant capacity, obesity and clinical correlates in first-episode and drug-naïve patients with schizophrenia. Schizophr Res 2024; 264:81-86. [PMID: 38113675 DOI: 10.1016/j.schres.2023.12.004] [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: 06/21/2023] [Revised: 09/02/2023] [Accepted: 12/05/2023] [Indexed: 12/21/2023]
Abstract
BACKGROUND Overweight/obesity is a growing concern in schizophrenia (SZ). A few studies have shown that excessive oxidative stress and abnormal antioxidants were associated with pathogenesis and psychiatric symptoms in first episode antipsychotics naïve (FEAN) patients with SZ. However, there is no study has explored the interrelationships between total antioxidant status (TAS) and the severity of psychiatric symptoms in the early stage of SZ. This study aimed to evaluate the impact of overweight/obesity on psychiatric symptoms in FEAN patients with SZ. METHODS A total of 241 patients with FEAN SZ and 119 healthy controls were recruited and symptoms were evaluated by the Positive and Negative Syndrome Scale (PANSS). TAS levels were also measured in patients and healthy controls. RESULTS We found a significant negative association between body mass index (BMI) and TAS in FEAN patients, but not in controls. In addition, BMI and TAS were negatively associated with psychiatric symptoms. Interestingly, further regression analysis revealed that the interaction between BMI and TAS was associated with the negative symptoms in the early stage of SZ. CONCLUSIONS Our study indicates that abnormal TAS levels interacting with overweight/obesity may be involved in the pathophysiology of SZ, in particular negative symptoms.
Collapse
Affiliation(s)
- Xiaoni Guan
- Peking University HuiLongGuan Clinical Medical School, Beijing HuiLongGuan Hospital, Beijing, China
| | - Yuping Chen
- Qingdao Mental Health Center, Qingdao, China
| | - Xin Wang
- Qingdao Mental Health Center, Qingdao, China
| | - Meihong Xiu
- Peking University HuiLongGuan Clinical Medical School, Beijing HuiLongGuan Hospital, Beijing, China
| | - Fengchun Wu
- Department of Psychiatry, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China; Department of Biomedical Engineering, Guangzhou Medical University, Guangzhou, China.
| | - Xiangyang Zhang
- Department of Psychiatry, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China; CAS Key Laboratory of Mental Health, Institute of Psychology, Beijing, China.
| |
Collapse
|
21
|
Prokhorova TA, Androsova LV, Tereshkina EB, Boksha IS, Savushkina OK, Pochueva VV, Sheshenin VS, Burbaeva GS, Klyushnik TP. [Clinical and psychopathological characteristics of patients with late-onset schizophrenia and schizophrenia-like psychoses in clusters identified by biological parameters]. Zh Nevrol Psikhiatr Im S S Korsakova 2024; 124:137-144. [PMID: 38676688 DOI: 10.17116/jnevro2024124041137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2024]
Abstract
OBJECTIVE To assess clinical and psychopathological characteristics of late-aged female patients with late-onset psychoses in clusters formed on the basis of biochemical and immunological blood parameters. MATERIAL AND METHODS We examined 59 women with schizophrenia and schizophrenia-like psychoses with onset after 40 years (ICD-10 F20, F22.8, F25, F23, F06.2), including 34 women with late-onset (40-60 years) and 25 with very late onset psychoses (after 60 years). At the time of hospitalization, a clinical/ psychopathological study was carried out using CGI-S, PANSS, CDSS, and HAMD-17, as well as the activities of glutathione reductase (GR) and glutathione-S-transferase (GT) have been determined in erythrocyte hemolysates, and the activities of leukocyte elastase (LE) and α1-proteinase inhibitor (α1-PI) have been assessed in blood plasma. Biochemical and immunological parameters have been also determined in 34 age-matched mentally healthy women. RESULTS Clustering by signs such as GR, GT, LE and α1-PI has yielded two clusters of objects (patients) significantly different in GT (p<0.0001), LE (p<0.0001), and α1-PI (p<0.001) activities. Relatively to the controls, in the cluster 1 patients, the activities of GST and α1-PI are increased, the activity of LE is decreased, whereas, in the cluster 2 patients, the activity of GR is decreased, and the activities of LE and α1-PI are increased. Cluster 1 patients differ from cluster 2 patients in greater severity of the condition (CGI-S, p=0.04) and higher total scores on PANSS subscales' items. Cluster 1 includes 76% of patients with very late onset. Different correlations between clinical and biological signs are found in two clusters. CONCLUSION The identified clusters have different clinical and psychopathological characteristics. Dividing patients into subgroups according to biochemical and immunological parameters is promising for the search for differentiated therapeutic approaches.
Collapse
Affiliation(s)
| | | | | | - I S Boksha
- Mental Health Research Center, Moscow, Russia
| | | | | | | | | | | |
Collapse
|
22
|
Wasserthal S, Muthesius A, Hurlemann R, Ruhrmann S, Schmidt SJ, Hellmich M, Schultze-Lutter F, Klosterkötter J, Müller H, Meyer-Lindenberg A, Poeppl TB, Walter H, Hirjak D, Koutsouleris N, Fallgatter AJ, Bechdolf A, Brockhaus-Dumke A, Mulert C, Philipsen A, Kambeitz J. N-Acetylcysteine and a Specialized Preventive Intervention for Individuals at High Risk for Psychosis: A Randomized Double-Blind Multicenter Trial. SCHIZOPHRENIA BULLETIN OPEN 2024; 5:sgae005. [PMID: 39144108 PMCID: PMC11207905 DOI: 10.1093/schizbullopen/sgae005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/16/2024]
Abstract
Background and Hypothesis Clinical high risk for psychosis (CHR-P) offers a window of opportunity for early intervention and recent trials have shown promising results for the use of N-acetylcysteine (NAC) in schizophrenia. Moreover, integrated preventive psychological intervention (IPPI), applies social-cognitive remediation to aid in preventing the transition to the psychosis of CHR-P patients. Study Design In this double-blind, randomized, controlled multicenter trial, a 2 × 2 factorial design was applied to investigate the effects of NAC compared to placebo (PLC) and IPPI compared to psychological stress management (PSM). The primary endpoint was the transition to psychosis or deterioration of CHR-P symptoms after 18 months. Study Results While insufficient recruitment led to early trial termination, a total of 48 participants were included in the study. Patients receiving NAC showed numerically higher estimates of event-free survival probability (IPPI + NAC: 72.7 ± 13.4%, PSM + NAC: 72.7 ± 13.4%) as compared to patients receiving PLC (IPPI + PLC: 56.1 ± 15.3%, PSM + PLC: 39.0 ± 17.4%). However, a log-rank chi-square test in Kaplan-Meier analysis revealed no significant difference of survival probability for NAC vs control (point hazard ratio: 0.879, 95% CI 0.281-2.756) or IPPI vs control (point hazard ratio: 0.827, 95% CI 0.295-2.314). The number of adverse events (AE) did not differ significantly between the four groups. Conclusions The superiority of NAC or IPPI in preventing psychosis in patients with CHR-P compared to controls could not be statistically validated in this trial. However, results indicate a consistent pattern that warrants further testing of NAC as a promising and well-tolerated intervention for CHR patients in future trials with adequate statistical power.
Collapse
Affiliation(s)
- Sven Wasserthal
- Division of Medical Psychology, Department of Psychiatry and Psychotherapy, University Hospital of Bonn, Bonn, Germany
| | - Ana Muthesius
- Department of Psychiatry and Psychotherapy, University of Cologne and University Hospital Cologne, Cologne, Germany
| | - René Hurlemann
- Department of Psychiatry, School of Medicine and Health Sciences, University of Oldenburg, Oldenburg, Germany
| | - Stephan Ruhrmann
- Department of Psychiatry and Psychotherapy, University of Cologne and University Hospital Cologne, Cologne, Germany
| | - Stefanie J Schmidt
- Division of Clinical Child and Adolescent Psychology, University of Bern, Bern, Switzerland
| | - Martin Hellmich
- Faculty of Medicine and University Hospital Cologne, Institute of Medical Statistics and Computational Biology, University of Cologne, Cologne, Germany
| | - Frauke Schultze-Lutter
- Department of Psychiatry and Psychotherapy, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
- Department of Psychology, Faculty of Psychology, Airlangga University, Surabaya, Indonesia
- University Hospital of Child and Adolescent Psychiatry and Psychotherapy, University of Bern, Bern, Switzerland
| | - Joachim Klosterkötter
- Department of Psychiatry and Psychotherapy, University of Cologne and University Hospital Cologne, Cologne, Germany
| | - Hendrik Müller
- Department of Psychiatry and Psychotherapy, University of Cologne and University Hospital Cologne, Cologne, Germany
| | - Andreas Meyer-Lindenberg
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, University of Heidelberg/Medical Faculty Mannheim, Mannheim, Germany
| | - Timm B Poeppl
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany
- Department of Psychiatry, Psychotherapy and Psychosomatics, RWTH Aachen University, Aachen, Germany
| | - Henrik Walter
- Department of Psychiatry and Psychotherapy CCM, Charité, Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Dusan Hirjak
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, University of Heidelberg/Medical Faculty Mannheim, Mannheim, Germany
| | - Nikolaos Koutsouleris
- Department of Psychiatry and Psychotherapy, Ludwig Maximilian University of Munich, Munich, Germany
| | - Andreas J Fallgatter
- Department of Psychiatry and Psychotherapy, Tübingen Center for Mental Health, University of Tübingen, Tübingen, Germany
- German Center for Mental Health (DZPG), Partner Site Tübingen, Tübingen, Germany
| | - Andreas Bechdolf
- Department of Psychiatry and Psychotherapy CCM, Charité, Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Department of Psychiatry, Psychotherapy and Psychosomatic Medicine with Early Intervention and Recognition Center (FRITZ), Vivantes Klinikum Am Urban, Berlin, Germany
| | | | - Christoph Mulert
- Center of Psychiatry, Justus-Liebig University, Giessen, Germany
| | - Alexandra Philipsen
- Department of Psychiatry and Psychotherapy, University Hospital of Bonn, Bonn, Germany
| | - Joseph Kambeitz
- Department of Psychiatry and Psychotherapy, University of Cologne and University Hospital Cologne, Cologne, Germany
| |
Collapse
|
23
|
Stein A, Zhu C, Du F, Öngür D. Magnetic Resonance Spectroscopy Studies of Brain Energy Metabolism in Schizophrenia: Progression from Prodrome to Chronic Psychosis. Curr Psychiatry Rep 2023; 25:659-669. [PMID: 37812338 DOI: 10.1007/s11920-023-01457-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/05/2023] [Indexed: 10/10/2023]
Abstract
PURPOSE OF REVIEW Schizophrenia (SZ) is a debilitating mental illness; existing treatments are partially effective and associated with significant side effect burden, largely due to our limited understanding of disease mechanisms and the trajectory of disease progression. Accumulating evidence suggests that metabolic changes associated with glucose metabolism, mitochondrial dysfunction, and redox imbalance play an important role in the pathophysiology of schizophrenia. However, the molecular mechanisms associated with these abnormalities in the brains of schizophrenia patients and the ways in which they change over time remain unclear. This paper aims to review the current literature on molecular mechanisms and in vivo magnetic resonance spectroscopy (MRS) studies of impaired energy metabolism in patients at clinical high risk for psychosis, with first-episode SZ, and with chronic SZ. Our review covers research related to high-energy phosphate metabolism, lactate, intracellular pH, redox ratio, and the antioxidant glutathione. RECENT FINDINGS Both first-episode and chronic SZ patients display a significant reduction in creatine kinase reaction activity and redox (NAD + /NADH) ratio in the prefrontal cortex. Chronic, but not first-episode, SZ patients also show a trend toward increased lactate levels and decreased pH value. These findings suggest a progressive shift from oxidative phosphorylation to glycolysis for energy production over the course of SZ, which is associated with redox imbalance and mitochondrial dysfunction. Accumulating evidence indicates that aberrant brain energy metabolism associated with mitochondrial dysfunction and redox imbalance plays a critical role in SZ and will be a promising target for future treatments.
Collapse
Affiliation(s)
- Abigail Stein
- Psychotic Disorders Division, McLean Hospital, Belmont, 02478, USA
- McLean Imaging Center, McLean Hospital, Belmont, 02478, USA
| | - Chenyanwen Zhu
- Psychotic Disorders Division, McLean Hospital, Belmont, 02478, USA
- McLean Imaging Center, McLean Hospital, Belmont, 02478, USA
| | - Fei Du
- Psychotic Disorders Division, McLean Hospital, Belmont, 02478, USA.
- McLean Imaging Center, McLean Hospital, Belmont, 02478, USA.
- Harvard Medical School, Boston, MA, 02115, USA.
| | - Dost Öngür
- Psychotic Disorders Division, McLean Hospital, Belmont, 02478, USA.
- Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
24
|
Carceller H, Gramuntell Y, Klimczak P, Nacher J. Perineuronal Nets: Subtle Structures with Large Implications. Neuroscientist 2023; 29:569-590. [PMID: 35872660 DOI: 10.1177/10738584221106346] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/25/2023]
Abstract
Perineuronal nets (PNNs) are specialized structures of the extracellular matrix that surround the soma and proximal dendrites of certain neurons in the central nervous system, particularly parvalbumin-expressing interneurons. Their appearance overlaps the maturation of neuronal circuits and the closure of critical periods in different regions of the brain, setting their connectivity and abruptly reducing their plasticity. As a consequence, the digestion of PNNs, as well as the removal or manipulation of their components, leads to a boost in this plasticity and can play a key role in the functional recovery from different insults and in the etiopathology of certain neurologic and psychiatric disorders. Here we review the structure, composition, and distribution of PNNs and their variation throughout the evolutive scale. We also discuss methodological approaches to study these structures. The function of PNNs during neurodevelopment and adulthood is discussed, as well as the influence of intrinsic and extrinsic factors on these specialized regions of the extracellular matrix. Finally, we review current data on alterations in PNNs described in diseases of the central nervous system (CNS), focusing on psychiatric disorders. Together, all the data available point to the PNNs as a promising target to understand the physiology and pathologic conditions of the CNS.
Collapse
Affiliation(s)
- Héctor Carceller
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, Spain
- CIBERSAM, Spanish National Network for Research in Mental Health, Instituto de Salud Carlos III, Madrid, Spain
- Biomedical Imaging Unit FISABIO-CIPF, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana, Valencia, Spain
| | - Yaiza Gramuntell
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, Spain
| | - Patrycja Klimczak
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, Spain
- CIBERSAM, Spanish National Network for Research in Mental Health, Instituto de Salud Carlos III, Madrid, Spain
| | - Juan Nacher
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, Spain
- CIBERSAM, Spanish National Network for Research in Mental Health, Instituto de Salud Carlos III, Madrid, Spain
- Fundación Investigación Hospital Clínico de Valencia, INCLIVA, Valencia, Spain
| |
Collapse
|
25
|
Karanikas E. The Gordian knot of the immune-redox systems' interactions in psychosis. Int Clin Psychopharmacol 2023; 38:285-296. [PMID: 37351570 DOI: 10.1097/yic.0000000000000481] [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] [Indexed: 06/24/2023]
Abstract
During the last decades the attempt to enlighten the pathobiological substrate of psychosis, from merely focusing on neurotransmitters, has expanded into new areas like the immune and redox systems. Indeed, the inflammatory hypothesis concerning psychosis etiopathology has exponentially grown with findings reflecting dysfunction/aberration of the immune/redox systems' effector components namely cytokines, chemokines, CRP, complement system, antibodies, pro-/anti-oxidants, oxidative stress byproducts just to name a few. Yet, we still lie far from comprehending the underlying cellular mechanisms, their causality directions, and the moderating/mediating parameters affecting these systems; let alone the inter-systemic (between immune and redox) interactions. Findings from preclinical studies on the stress field have provided evidence indicative of multifaceted interactions among the immune and redox components so tightly intertwined as a Gordian knot. Interestingly the literature concerning the interactions between these same systems in the context of psychosis appears minimal (if not absent) and ambiguous. This review attempts to draw a frame of the immune-redox systems' interactions starting from basic research on the stress field and expanding on clinical studies with cohorts with psychosis, hoping to instigate new avenues of research.
Collapse
Affiliation(s)
- Evangelos Karanikas
- Department of Psychiatry, 424 General Military Hospital, Ring Road, Nea Efkarpia, Thessaloniki, Greece
| |
Collapse
|
26
|
Skupienski R, Steullet P, Do KQ, Xin L. Developmental changes in cerebral NAD and neuroenergetics of an antioxidant compromised mouse model of schizophrenia. Transl Psychiatry 2023; 13:275. [PMID: 37543592 PMCID: PMC10404265 DOI: 10.1038/s41398-023-02568-2] [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: 03/19/2022] [Revised: 07/20/2023] [Accepted: 07/24/2023] [Indexed: 08/07/2023] Open
Abstract
Defects in essential metabolic regulation for energy supply, increased oxidative stress promoting excitatory/inhibitory imbalance and phospholipid membrane dysfunction have been implicated in the pathophysiology of schizophrenia (SZ). The knowledge about the developmental trajectory of these key pathophysiological components and their interplay is important to develop new preventive and treatment strategies. However, this assertion is so far limited. To investigate the developmental regulations of these key components in the brain, we assessed, for the first time, in vivo redox state from the oxidized (NAD+) and reduced (NADH) form of Nicotinamide Adenine Dinucleotide (NAD), energy and membrane metabolites, inhibitory and excitatory neurotransmitters by 31P and 1H MRS during the neurodevelopment of an SZ animal model with genetically compromised glutathione synthesis (gclm-KO mice). When compared to age-matched wild type (WT), an increase in NAD+/NADH redox ratio was found in gclm-KO mice until early adulthood, followed by a decrease in full adults as observed in patients. Especially, in early postnatal life (P20, corresponding to childhood), levels of several metabolites were altered in gclm-KO mice, including NAD+, NAD+/NADH, ATP, and glutamine + glutamate, suggesting an interactive compensation for redox dysregulation between NAD, energy metabolism, and neurotransmission. The identified temporal neurometabolic regulations under deficits in redox regulation provide insights into preventive treatment targets for at-risk individuals, and other neurodevelopmental disorders involving oxidative stress and energetic dysfunction.
Collapse
Affiliation(s)
- Radek Skupienski
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), Prilly, Switzerland
- CIBM Center for Biomedical Imaging, Lausanne, Switzerland
| | - Pascal Steullet
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), Prilly, Switzerland
| | - Kim Q Do
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), Prilly, Switzerland
| | - Lijing Xin
- CIBM Center for Biomedical Imaging, Lausanne, Switzerland.
- Animal Imaging and Technology, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
| |
Collapse
|
27
|
Jekabsone A, Jankeviciute S, Pampuscenko K, Borutaite V, Morkuniene R. The Role of Intracellular Ca 2+ and Mitochondrial ROS in Small Aβ 1-42 Oligomer-Induced Microglial Death. Int J Mol Sci 2023; 24:12315. [PMID: 37569690 PMCID: PMC10418347 DOI: 10.3390/ijms241512315] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/27/2023] [Accepted: 07/29/2023] [Indexed: 08/13/2023] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia worldwide, and it contributes up to 70% of cases. AD pathology involves abnormal amyloid beta (Aβ) accumulation, and the link between the Aβ1-42 structure and toxicity is of major interest. NMDA receptors (NMDAR) are thought to be essential in Aβ-affected neurons, but the role of this receptor in glial impairment is still unclear. In addition, there is insufficient knowledge about the role of Aβ species regarding mitochondrial redox states in neurons and glial cells, which may be critical in developing Aβ-caused neurotoxicity. In this study, we investigated whether different Aβ1-42 species-small oligomers, large oligomers, insoluble fibrils, and monomers-were capable of producing neurotoxic effects via microglial NMDAR activation and changes in mitochondrial redox states in primary rat brain cell cultures. Small Aβ1-42 oligomers induced a concentration- and time-dependent increase in intracellular Ca2+ and necrotic microglial death. These changes were partially prevented by the NMDAR inhibitors MK801, memantine, and D-2-amino-5-phosphopentanoic acid (DAP5). Neither microglial intracellular Ca2+ nor viability was significantly affected by larger Aβ1-42 species or monomers. In addition, the small Aβ1-42 oligomers caused mitochondrial reactive oxygen species (mtROS)-mediated mitochondrial depolarization, glutamate release, and neuronal cell death. In microglia, the Aβ1-42-induced mtROS overproduction was mediated by intracellular calcium ions and Aβ-binding alcohol dehydrogenase (ABAD). The data suggest that the pharmacological targeting of microglial NMDAR and mtROS may be a promising strategy for AD therapy.
Collapse
Affiliation(s)
- Aiste Jekabsone
- Neuroscience Institute, Lithuanian University of Health Sciences, LT-50162 Kaunas, Lithuania; (A.J.); (S.J.); (K.P.); (V.B.)
- Faculty of Pharmacy, Lithuanian University of Health Sciences, LT-50162 Kaunas, Lithuania
| | - Silvija Jankeviciute
- Neuroscience Institute, Lithuanian University of Health Sciences, LT-50162 Kaunas, Lithuania; (A.J.); (S.J.); (K.P.); (V.B.)
| | - Katryna Pampuscenko
- Neuroscience Institute, Lithuanian University of Health Sciences, LT-50162 Kaunas, Lithuania; (A.J.); (S.J.); (K.P.); (V.B.)
| | - Vilmante Borutaite
- Neuroscience Institute, Lithuanian University of Health Sciences, LT-50162 Kaunas, Lithuania; (A.J.); (S.J.); (K.P.); (V.B.)
| | - Ramune Morkuniene
- Neuroscience Institute, Lithuanian University of Health Sciences, LT-50162 Kaunas, Lithuania; (A.J.); (S.J.); (K.P.); (V.B.)
- Faculty of Pharmacy, Lithuanian University of Health Sciences, LT-50162 Kaunas, Lithuania
| |
Collapse
|
28
|
Schallmo MP, Weldon KB, Kamath RS, Moser HR, Montoya SA, Killebrew KW, Demro C, Grant AN, Marjańska M, Sponheim SR, Olman CA. The psychosis human connectome project: Design and rationale for studies of visual neurophysiology. Neuroimage 2023; 272:120060. [PMID: 36997137 PMCID: PMC10153004 DOI: 10.1016/j.neuroimage.2023.120060] [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: 01/06/2023] [Revised: 03/22/2023] [Accepted: 03/27/2023] [Indexed: 04/01/2023] Open
Abstract
Visual perception is abnormal in psychotic disorders such as schizophrenia. In addition to hallucinations, laboratory tests show differences in fundamental visual processes including contrast sensitivity, center-surround interactions, and perceptual organization. A number of hypotheses have been proposed to explain visual dysfunction in psychotic disorders, including an imbalance between excitation and inhibition. However, the precise neural basis of abnormal visual perception in people with psychotic psychopathology (PwPP) remains unknown. Here, we describe the behavioral and 7 tesla MRI methods we used to interrogate visual neurophysiology in PwPP as part of the Psychosis Human Connectome Project (HCP). In addition to PwPP (n = 66) and healthy controls (n = 43), we also recruited first-degree biological relatives (n = 44) in order to examine the role of genetic liability for psychosis in visual perception. Our visual tasks were designed to assess fundamental visual processes in PwPP, whereas MR spectroscopy enabled us to examine neurochemistry, including excitatory and inhibitory markers. We show that it is feasible to collect high-quality data across multiple psychophysical, functional MRI, and MR spectroscopy experiments with a sizable number of participants at a single research site. These data, in addition to those from our previously described 3 tesla experiments, will be made publicly available in order to facilitate further investigations by other research groups. By combining visual neuroscience techniques and HCP brain imaging methods, our experiments offer new opportunities to investigate the neural basis of abnormal visual perception in PwPP.
Collapse
Affiliation(s)
- Michael-Paul Schallmo
- Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis, MN, USA.
| | - Kimberly B Weldon
- Department of Radiology, Center for Magnetic Resonance Research, University of Minnesota, Minneapolis, MN, USA; Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis, MN, USA
| | - Rohit S Kamath
- Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis, MN, USA
| | - Hannah R Moser
- Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis, MN, USA
| | - Samantha A Montoya
- Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis, MN, USA
| | - Kyle W Killebrew
- Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis, MN, USA
| | - Caroline Demro
- Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis, MN, USA; Department of Psychology, University of Minnesota, Minneapolis, MN, USA
| | - Andrea N Grant
- Department of Radiology, Center for Magnetic Resonance Research, University of Minnesota, Minneapolis, MN, USA
| | - Małgorzata Marjańska
- Department of Radiology, Center for Magnetic Resonance Research, University of Minnesota, Minneapolis, MN, USA
| | - Scott R Sponheim
- Veterans Affairs Medical Center, Minneapolis, MN, USA; Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis, MN, USA
| | - Cheryl A Olman
- Department of Psychology, University of Minnesota, Minneapolis, MN, USA; Department of Radiology, Center for Magnetic Resonance Research, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
29
|
Lotan A, Luza S, Opazo CM, Ayton S, Lane DJR, Mancuso S, Pereira A, Sundram S, Weickert CS, Bousman C, Pantelis C, Everall IP, Bush AI. Perturbed iron biology in the prefrontal cortex of people with schizophrenia. Mol Psychiatry 2023; 28:2058-2070. [PMID: 36750734 PMCID: PMC10575779 DOI: 10.1038/s41380-023-01979-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 01/10/2023] [Accepted: 01/20/2023] [Indexed: 02/09/2023]
Abstract
Despite loss of grey matter volume and emergence of distinct cognitive deficits in young adults diagnosed with schizophrenia, current treatments for schizophrenia do not target disruptions in late maturational reshaping of the prefrontal cortex. Iron, the most abundant transition metal in the brain, is essential to brain development and function, but in excess, it can impair major neurotransmission systems and lead to lipid peroxidation, neuroinflammation and accelerated aging. However, analysis of cortical iron biology in schizophrenia has not been reported in modern literature. Using a combination of inductively coupled plasma-mass spectrometry and western blots, we quantified iron and its major-storage protein, ferritin, in post-mortem prefrontal cortex specimens obtained from three independent, well-characterised brain tissue resources. Compared to matched controls (n = 85), among schizophrenia cases (n = 86) we found elevated tissue iron, unlikely to be confounded by demographic and lifestyle variables, by duration, dose and type of antipsychotic medications used or by copper and zinc levels. We further observed a loss of physiologic age-dependent iron accumulation among people with schizophrenia, in that the iron level among cases was already high in young adulthood. Ferritin, which stores iron in a redox-inactive form, was paradoxically decreased in individuals with the disorder. Such iron-ferritin uncoupling could alter free, chemically reactive, tissue iron in key reasoning and planning areas of the young-adult schizophrenia cortex. Using a prediction model based on iron and ferritin, our data provide a pathophysiologic link between perturbed cortical iron biology and schizophrenia and indicate that achievement of optimal cortical iron homeostasis could offer a new therapeutic target.
Collapse
Affiliation(s)
- Amit Lotan
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, 3010, Australia
- Department of Psychiatry and the Biological Psychiatry Laboratory, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Sandra Luza
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, 3010, Australia
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne & Melbourne Health, Carlton, VIC, Australia
| | - Carlos M Opazo
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, 3010, Australia.
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne & Melbourne Health, Carlton, VIC, Australia.
| | - Scott Ayton
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Darius J R Lane
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Serafino Mancuso
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne & Melbourne Health, Carlton, VIC, Australia
| | - Avril Pereira
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, 3010, Australia
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne & Melbourne Health, Carlton, VIC, Australia
| | - Suresh Sundram
- Department of Psychiatry, School of Clinical Sciences, Monash University, Melbourne, VIC, Australia
- Mental Health Program, Monash Health, Melbourne, VIC, Australia
| | - Cynthia Shannon Weickert
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Randwick, NSW, Australia
- School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Chad Bousman
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne & Melbourne Health, Carlton, VIC, Australia
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Departments of Medical Genetics, Psychiatry, Physiology & Pharmacology, University of Calgary, Calgary, AB, Canada
- The Cooperative Research Centre (CRC) for Mental Health, Melbourne, VIC, Australia
| | - Christos Pantelis
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne & Melbourne Health, Carlton, VIC, Australia
- North Western Mental Health, Melbourne, VIC, Australia
| | - Ian P Everall
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne & Melbourne Health, Carlton, VIC, Australia
- North Western Mental Health, Melbourne, VIC, Australia
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Ashley I Bush
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, 3010, Australia.
- The Cooperative Research Centre (CRC) for Mental Health, Melbourne, VIC, Australia.
| |
Collapse
|
30
|
Li Z, Sun X, He J, Kong D, Wang J, Wang L. Identification of a Hypoxia-Related Signature as Candidate Detector for Schizophrenia Based on Genome-Wide Gene Expression. Hum Hered 2023; 88:18-28. [PMID: 36913932 PMCID: PMC10124753 DOI: 10.1159/000529902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 02/15/2023] [Indexed: 03/15/2023] Open
Abstract
INTRODUCTION Schizophrenia (SCZ), a severe neuropsychiatric disorder with high genetic susceptibility, has high rates of misdiagnosis due to the unavoidably subjective factors and heterogeneous clinical presentations. Hypoxia has been identified as an importantly risk factor that participates in the development of SCZ. Therefore, development of a hypoxia-related biomarker for SCZ diagnosis is promising. Therefore, we dedicated to develop a biomarker that could contribute to distinguishing healthy controls and SCZ patients. METHODS GSE17612, GSE21935, and GSE53987 datasets, consisting of 97 control samples and 99 SCZ samples, were involved in our study. The hypoxia score was calculated based on the single-sample gene-set enrichment analysis using the hypoxia-related differentially expressed genes to quantify the expression levels of these genes for each SCZ patient. Patients in high-score groups were defined if their hypoxia score was in the upper half of all hypoxia scores and patients in low-score groups if their hypoxia score was in the lower half. GSEA was applied to detect the functional pathway of these differently expressed genes. CIBERSORT algorithm was utilized to evaluate the tumor-infiltrating immune cells of SCZ patients. RESULTS In this study, we developed and validated a biomarker consisting of 12 hypoxia-related genes that could distinguish healthy controls and SCZ patients robustly. We found that the metabolism reprogramming might be activated in the patient with high hypoxia score. Finally, CIBERSORT analysis illustrated that lower composition of naive B cells and higher composition of memory B cells might be observed in low-score groups of SCZ patients. CONCLUSION These findings revealed that the hypoxia-related signature was acceptable as a detector for SCZ, providing further insight into effective diagnosis and treatment strategies for SCZ.
Collapse
Affiliation(s)
- Zhitao Li
- Department of Psychiatry and Psychological Clinic, Affiliated Quanzhou First Hospital, Fujian Medical University, Quanzhou, China
| | - Xinyu Sun
- Department of Psychiatry and Psychological Clinic, Affiliated Quanzhou First Hospital, Fujian Medical University, Quanzhou, China
| | - Jia He
- Department of Psychiatry and Psychological Clinic, Affiliated Quanzhou First Hospital, Fujian Medical University, Quanzhou, China
| | - Dongyan Kong
- Department of Psychiatry and Psychological Clinic, Affiliated Quanzhou First Hospital, Fujian Medical University, Quanzhou, China
| | - Jinyi Wang
- Department of Psychiatry, Quanzhou Third Hospital, Quanzhou, China
| | - Lili Wang
- Department of Psychiatry, Quanzhou Third Hospital, Quanzhou, China
| |
Collapse
|
31
|
The Effect of Clozapine and Novel Glutamate Modulator JNJ-46356479 on Nitrosative Stress in a Postnatal Murine Ketamine Model of Schizophrenia. Int J Mol Sci 2023; 24:ijms24021022. [PMID: 36674542 PMCID: PMC9866372 DOI: 10.3390/ijms24021022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/26/2022] [Accepted: 12/26/2022] [Indexed: 01/06/2023] Open
Abstract
Schizophrenia (SZ) is a heterogeneous mental disorder, affecting ~1% of the worldwide population. One of the main pathophysiological theories of SZ is the imbalance of excitatory glutamatergic pyramidal neurons and inhibitory GABAergic interneurons, involving N-methyl-D-aspartate receptors (NMDAr). This may lead to local glutamate storms coupled with excessive dendritic pruning and subsequent cellular stress, including nitrosative stress, during a critical period of neurodevelopment, such as adolescence. Nitrosative stress is mediated by nitric oxide (NO), which is released by NO synthases (NOS) and has emerged as a key signaling molecule implicated in SZ. Regarding glutamatergic models of SZ, the administration of NMDAr antagonists has been found to increase NOS levels in the prefrontal cortex (PFC) and ventral hippocampus (HPC). We hypothesized that suboptimal NOS function in adolescence could be a target for early treatments, including clozapine (CLZ) and the novel metabotropic glutamate receptor modulator JNJ-46356479 (JNJ). We analyzed the protein levels of NOS isoforms in adult PFC and HPC of a postnatal ketamine induced murine model of SZ receiving CLZ or JNJ during adolescence by western blot. Endothelial NOS and neuronal NOS increased under ketamine administration in PFC and decreased in CLZ or JNJ treatments. The same trends were found in the HPC in neuronal NOS. In contrast, inducible NOS was increased under JNJ treatment with respect to ketamine induction in the HPC, and the same trends were found in the PFC. Taken together, our findings suggest a misbalance of the NOS system following NMDAr antagonist administration, which was then modulated under early CLZ and JNJ treatments.
Collapse
|
32
|
Griffiths SL, Upthegrove R, Corsi-Zuelli F, Deakin B. Rethinking Immunity and Cognition in Clinical High Risk for Psychosis. Curr Top Behav Neurosci 2023; 63:475-497. [PMID: 36409457 DOI: 10.1007/7854_2022_399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
It is well known that schizophrenia is associated with cognitive impairment, reduced cortical grey matter and increased circulating concentrations of inflammatory cytokines. However, the relationship between these findings is not clear. We outline the influential neuroinflammatory hypotheses that raised cytokines provoke a damaging immune response in microglia that results in reduced grey matter and associated cognitive performance. We investigated whether such an interaction might be detectable in the prodromal period as illness emerges from the Clinical High Risk for Psychosis (CHR-P). Meta-analyses suggest that compared with controls, impaired cognition and reduced grey matter are already present by the prodrome and that greater decrements are present in those who later develop symptoms. In contrast, the few cytokine studies report no abnormalities in CHR-P except that interleukin-6 (IL-6) levels were raised versus controls and to a greater extent in the future patients, in one study. We noted that cognitive impairment and less cortical grey matter are more severe in schizophrenia than in affective disorders, but that increased cytokine levels are similarly prevalent across disorders. We found no studies correlating cytokine levels with cognitive impairment in CHR-P but such correlations seem unlikely given the minimal relationship reported in a recent meta-analysis of the many cytokine-cognition studies in established illness. From this and other evidence, we conclude that neuroinflammation is not a core feature of schizophrenia nor a substrate for reduced grey matter volume or cognitive function. We draw attention instead to the emerging evidence that brain-resident immune cells and signalling molecules such as Tregs and IL-6, which are influenced by schizophrenia risk genes, regulate and are necessary for the development and function of neuron-glia interaction. We suggest that cognitive impairment in schizophrenia can be seen as a convergence of genetic and immune-neurodevelopmental dysregulation whereas raised cytokines are a consequence of impaired Tregs control of systemic inflammation.
Collapse
Affiliation(s)
| | - Rachel Upthegrove
- Institute for Mental Health, University of Birmingham, Birmingham, UK
| | - Fabiana Corsi-Zuelli
- Institute for Mental Health, University of Birmingham, Birmingham, UK
- Division of Psychiatry, Department of Neuroscience and Behaviour, Ribeirao Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Bill Deakin
- Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, School of Biological Sciences, University of Manchester, Manchester, UK.
| |
Collapse
|
33
|
Vinogradov S, Chafee MV, Lee E, Morishita H. Psychosis spectrum illnesses as disorders of prefrontal critical period plasticity. Neuropsychopharmacology 2023; 48:168-185. [PMID: 36180784 PMCID: PMC9700720 DOI: 10.1038/s41386-022-01451-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 08/17/2022] [Accepted: 08/21/2022] [Indexed: 01/05/2023]
Abstract
Emerging research on neuroplasticity processes in psychosis spectrum illnesses-from the synaptic to the macrocircuit levels-fill key gaps in our models of pathophysiology and open up important treatment considerations. In this selective narrative review, we focus on three themes, emphasizing alterations in spike-timing dependent and Hebbian plasticity that occur during adolescence, the critical period for prefrontal system development: (1) Experience-dependent dysplasticity in psychosis emerges from activity decorrelation within neuronal ensembles. (2) Plasticity processes operate bidirectionally: deleterious environmental and experiential inputs shape microcircuits. (3) Dysregulated plasticity processes interact across levels of scale and time and include compensatory mechanisms that have pathogenic importance. We present evidence that-given the centrality of progressive dysplastic changes, especially in prefrontal cortex-pharmacologic or neuromodulatory interventions will need to be supplemented by corrective learning experiences for the brain if we are to help people living with these illnesses to fully thrive.
Collapse
Affiliation(s)
- Sophia Vinogradov
- Department of Psychiatry & Behavioral Science, University of Minnesota Medical School, Minneapolis, MN, USA.
| | - Matthew V Chafee
- Department of Neuroscience, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Erik Lee
- Masonic Institute for the Developing Brain, University of Minnesota Medical School, Minneapolis, MN, USA
- University of Minnesota Informatics Institute, University of Minnesota, Minneapolis, MN, USA
| | - Hirofumi Morishita
- Department of Psychiatry, Neuroscience, & Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
34
|
Morén C, Treder N, Martínez-Pinteño A, Rodríguez N, Arbelo N, Madero S, Gómez M, Mas S, Gassó P, Parellada E. Systematic Review of the Therapeutic Role of Apoptotic Inhibitors in Neurodegeneration and Their Potential Use in Schizophrenia. Antioxidants (Basel) 2022; 11:2275. [PMID: 36421461 PMCID: PMC9686909 DOI: 10.3390/antiox11112275] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 11/02/2022] [Accepted: 11/03/2022] [Indexed: 09/15/2023] Open
Abstract
Schizophrenia (SZ) is a deleterious brain disorder affecting cognition, emotion and reality perception. The most widely accepted neurochemical-hypothesis is the imbalance of neurotransmitter-systems. Depleted GABAergic-inhibitory function might produce a regionally-located dopaminergic and glutamatergic-storm in the brain. The dopaminergic-release may underlie the positive psychotic-symptoms while the glutamatergic-release could prompt the primary negative symptoms/cognitive deficits. This may occur due to excessive synaptic-pruning during the neurodevelopmental stages of adolescence/early adulthood. Thus, although SZ is not a neurodegenerative disease, it has been suggested that exaggerated dendritic-apoptosis could explain the limited neuroprogression around its onset. This apoptotic nature of SZ highlights the potential therapeutic action of anti-apoptotic drugs, especially at prodromal stages. If dysregulation of apoptotic mechanisms underlies the molecular basis of SZ, then anti-apoptotic molecules could be a prodromal therapeutic option to halt or prevent SZ. In fact, risk alleles related in apoptotic genes have been recently associated to SZ and shared molecular apoptotic changes are common in the main neurodegenerative disorders and SZ. PRISMA-guidelines were considered. Anti-apoptotic drugs are commonly applied in classic neurodegenerative disorders with promising results. Despite both the apoptotic-hallmarks of SZ and the widespread use of anti-apoptotic targets in neurodegeneration, there is a strikingly scarce number of studies investigating anti-apoptotic approaches in SZ. We analyzed the anti-apoptotic approaches conducted in neurodegeneration and the potential applications of such anti-apoptotic therapies as a promising novel therapeutic strategy, especially during early stages.
Collapse
Affiliation(s)
- Constanza Morén
- Barcelona Clínic Schizophrenia Unit (BCSU), Institute of Neuroscience, Psychiatry and Psychology Service, Hospital Clínic of Barcelona, University of Barcelona, 08036 Barcelona, Spain
- Clinical and Experimental Neuroscience Area, The August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
- U722 Group, Centro de Investigación Biomédica en Red de Enfermedades Raras, CIBERER, Carlos III Health Institute, 28029 Madrid, Spain
- Department of Basic Clinical Practice, Pharmacology Unit, University of Barcelona, 08036 Barcelona, Spain
| | - Nina Treder
- Faculty of Psychology and Neuroscience, Maastricht University, 6211 LK Maastricht, The Netherlands
| | - Albert Martínez-Pinteño
- Department of Basic Clinical Practice, Pharmacology Unit, University of Barcelona, 08036 Barcelona, Spain
| | - Natàlia Rodríguez
- Department of Basic Clinical Practice, Pharmacology Unit, University of Barcelona, 08036 Barcelona, Spain
| | - Néstor Arbelo
- Barcelona Clínic Schizophrenia Unit (BCSU), Institute of Neuroscience, Psychiatry and Psychology Service, Hospital Clínic of Barcelona, University of Barcelona, 08036 Barcelona, Spain
- G04 Group, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Carlos III Health Institute, 28029 Madrid, Spain
| | - Santiago Madero
- Barcelona Clínic Schizophrenia Unit (BCSU), Institute of Neuroscience, Psychiatry and Psychology Service, Hospital Clínic of Barcelona, University of Barcelona, 08036 Barcelona, Spain
- G04 Group, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Carlos III Health Institute, 28029 Madrid, Spain
| | - Marta Gómez
- Barcelona Clínic Schizophrenia Unit (BCSU), Institute of Neuroscience, Psychiatry and Psychology Service, Hospital Clínic of Barcelona, University of Barcelona, 08036 Barcelona, Spain
- G04 Group, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Carlos III Health Institute, 28029 Madrid, Spain
- Department of Psychiatry, Servizo Galego de Saúde (SERGAS), 36001 Pontevedra, Spain
| | - Sergi Mas
- Clinical and Experimental Neuroscience Area, The August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
- Department of Basic Clinical Practice, Pharmacology Unit, University of Barcelona, 08036 Barcelona, Spain
- G04 Group, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Carlos III Health Institute, 28029 Madrid, Spain
| | - Patricia Gassó
- Clinical and Experimental Neuroscience Area, The August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
- Department of Basic Clinical Practice, Pharmacology Unit, University of Barcelona, 08036 Barcelona, Spain
- G04 Group, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Carlos III Health Institute, 28029 Madrid, Spain
| | - Eduard Parellada
- Barcelona Clínic Schizophrenia Unit (BCSU), Institute of Neuroscience, Psychiatry and Psychology Service, Hospital Clínic of Barcelona, University of Barcelona, 08036 Barcelona, Spain
- Clinical and Experimental Neuroscience Area, The August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
- Department of Basic Clinical Practice, Pharmacology Unit, University of Barcelona, 08036 Barcelona, Spain
- G04 Group, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Carlos III Health Institute, 28029 Madrid, Spain
| |
Collapse
|
35
|
Czekus C, Steullet P, Orero López A, Bozic I, Rusterholz T, Bandarabadi M, Do KQ, Gutierrez Herrera C. Alterations in TRN-anterodorsal thalamocortical circuits affect sleep architecture and homeostatic processes in oxidative stress vulnerable Gclm -/- mice. Mol Psychiatry 2022; 27:4394-4406. [PMID: 35902628 PMCID: PMC9734061 DOI: 10.1038/s41380-022-01700-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 06/22/2022] [Accepted: 07/01/2022] [Indexed: 12/14/2022]
Abstract
Schizophrenia is associated with alterations of sensory integration, cognitive processing and both sleep architecture and sleep oscillations in mouse models and human subjects, possibly through changes in thalamocortical dynamics. Oxidative stress (OxS) damage, including inflammation and the impairment of fast-spiking gamma-aminobutyric acid neurons have been hypothesized as a potential mechanism responsible for the onset and development of schizophrenia. Yet, the link between OxS and perturbation of thalamocortical dynamics and sleep remains unclear. Here, we sought to investigate the effects of OxS on sleep regulation by characterizing the dynamics of thalamocortical networks across sleep-wake states in a mouse model with a genetic deletion of the modifier subunit of glutamate-cysteine ligase (Gclm knockout, KO) using high-density electrophysiology in freely-moving mice. We found that Gcml KO mice exhibited a fragmented sleep architecture and impaired sleep homeostasis responses as revealed by the increased NREM sleep latencies, decreased slow-wave activities and spindle rate after sleep deprivation. These changes were associated with altered bursting activity and firing dynamics of neurons from the thalamic reticularis nucleus, anterior cingulate and anterodorsal thalamus. Administration of N-acetylcysteine (NAC), a clinically relevant antioxidant, rescued the sleep fragmentation and spindle rate through a renormalization of local neuronal dynamics in Gclm KO mice. Collectively, these findings provide novel evidence for a link between OxS and the deficits of frontal TC network dynamics as a possible mechanism underlying sleep abnormalities and impaired homeostatic responses observed in schizophrenia.
Collapse
Affiliation(s)
- Christina Czekus
- Center for Experimental Neurology, Department of Neurology, Inselspital University Hospital, Bern, Switzerland
| | - Pascal Steullet
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital, Site de Cery, CH-1008, Prilly-Lausanne, Switzerland
| | - Albert Orero López
- Center for Experimental Neurology, Department of Neurology, Inselspital University Hospital, Bern, Switzerland
| | - Ivan Bozic
- Department for Biomedical Research, University of Bern, Bern, Switzerland
| | - Thomas Rusterholz
- Center for Experimental Neurology, Department of Neurology, Inselspital University Hospital, Bern, Switzerland
| | - Mojtaba Bandarabadi
- Center for Experimental Neurology, Department of Neurology, Inselspital University Hospital, Bern, Switzerland
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Kim Q Do
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital, Site de Cery, CH-1008, Prilly-Lausanne, Switzerland
| | - Carolina Gutierrez Herrera
- Center for Experimental Neurology, Department of Neurology, Inselspital University Hospital, Bern, Switzerland.
- Department for Biomedical Research, University of Bern, Bern, Switzerland.
| |
Collapse
|
36
|
Knight S, McCutcheon R, Dwir D, Grace AA, O'Daly O, McGuire P, Modinos G. Hippocampal circuit dysfunction in psychosis. Transl Psychiatry 2022; 12:344. [PMID: 36008395 PMCID: PMC9411597 DOI: 10.1038/s41398-022-02115-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 08/09/2022] [Accepted: 08/11/2022] [Indexed: 11/09/2022] Open
Abstract
Despite strong evidence of the neurodevelopmental origins of psychosis, current pharmacological treatment is not usually initiated until after a clinical diagnosis is made, and is focussed on antagonising striatal dopamine receptors. These drugs are only partially effective, have serious side effects, fail to alleviate the negative and cognitive symptoms of the disorder, and are not useful as a preventive treatment. In recent years, attention has turned to upstream brain regions that regulate striatal dopamine function, such as the hippocampus. This review draws together these recent data to discuss why the hippocampus may be especially vulnerable in the pathophysiology of psychosis. First, we describe the neurodevelopmental trajectory of the hippocampus and its susceptibility to dysfunction, exploring this region's proneness to structural and functional imbalances, metabolic pressures, and oxidative stress. We then examine mechanisms of hippocampal dysfunction in psychosis and in individuals at high-risk for psychosis and discuss how and when hippocampal abnormalities may be targeted in these groups. We conclude with future directions for prospective studies to unlock the discovery of novel therapeutic strategies targeting hippocampal circuit imbalances to prevent or delay the onset of psychosis.
Collapse
Affiliation(s)
- Samuel Knight
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK.
| | - Robert McCutcheon
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Daniella Dwir
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Anthony A Grace
- Departments of Neuroscience, Psychiatry and Psychology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Owen O'Daly
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Philip McGuire
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
- NIHR Maudsley Biomedical Research Centre, London, UK
| | - Gemma Modinos
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
| |
Collapse
|
37
|
Chronic N-Acetylcysteine Treatment Prevents Amphetamine-Induced Hyperactivity in Heterozygous Disc1 Mutant Mice, a Putative Prodromal Schizophrenia Animal Model. Int J Mol Sci 2022; 23:ijms23169419. [PMID: 36012679 PMCID: PMC9408838 DOI: 10.3390/ijms23169419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/14/2022] [Accepted: 08/17/2022] [Indexed: 11/17/2022] Open
Abstract
Symptoms of schizophrenia (SZ) typically emerge during adolescence to young adulthood, which gives a window before full-blown psychosis for early intervention. Strategies for preventing the conversion from the prodromal phase to the psychotic phase are warranted. Heterozygous (Het) Disc1 mutant mice are considered a prodromal model of SZ, suitable for studying psychotic conversion. We evaluated the preventive effect of chronic N-acetylcysteine (NAC) administration, covering the prenatal era to adulthood, on the reaction following the Amph challenge, which mimics the outbreak or conversion of psychosis, in adult Het Disc1 mice. Biochemical and morphological features were examined in the striatum of NAC-treated mice. Chronic NAC treatment normalized the Amph-induced activity in the Het Disc1 mice. Furthermore, the striatal phenotypes of Het Disc1 mice were rescued by NAC including dopamine receptors, the expression of GSK3s, MSN dendritic impairments, and striatal PV density. The current study demonstrated a potent preventive effect of chronic NAC treatment in Disc1 Het mice on the acute Amph test, which mimics the outbreak of psychosis. Our findings not only support the benefit of NAC as a dietary supplement for SZ prodromes, but also advance our knowledge of striatal dopamine receptors, PV neurons, and GSK3 signaling pathways as therapeutic targets for treating or preventing the pathogenesis of mental disorders.
Collapse
|
38
|
Liu H, Liu H, Jiang S, Su L, Lu Y, Chen Z, Li X, Li X, Wang X, Xiu M, Zhang X. Sex-Specific Association between Antioxidant Defense System and Therapeutic Response to Risperidone in Schizophrenia: A Prospective Longitudinal Study. Curr Neuropharmacol 2022; 20:1793-1803. [PMID: 34766896 PMCID: PMC9881066 DOI: 10.2174/1570159x19666211111123918] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/14/2021] [Accepted: 09/20/2021] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND There are various differences in response to different antipsychotics and antioxidant defense systems (ADS) by sex. Previous studies have shown that several ADS enzymes are closely related to the treatment response of patients with antipsychotics-naïve first-episode (ANFE) schizophrenia. OBJECTIVE Therefore, the main goal of this study was to assess the sex difference in the relationship between changes in ADS enzyme activities and risperidone response. METHODS The plasma activities of glutathione peroxidase (GPx), catalase (CAT), superoxide dismutase (SOD), and total antioxidant status (TAS) were measured in 218 patients and 125 healthy controls. Patients were treated with risperidone for 3 months, and we measured PANSS for psychopathological symptoms and ADS biomarkers at baseline and at the end of 3 months of treatment. We compared sex-specific group differences between 50 non-responders and 168 responders at baseline and at the end of the three months of treatment. RESULTS We found that female patients responded better to risperidone treatment than male patients. At baseline and 3-month follow-up, there were no significant sex differences in TAS levels and three ADS enzyme activities. Interestingly, only in female patients, after 12 weeks of risperidone treatment, the GPx activity of responders was higher than that of non-responders. CONCLUSION These results indicate that after treatment with risperidone, changes in GPx activity were associated with treatment response, suggesting that changes in GPx may be a predictor of response to risperidone treatment in female patients with ANFE schizophrenia.
Collapse
Affiliation(s)
- Haixia Liu
- Department of Psychiatry, Shandong Mental Health Center, Jinan, China;
| | - Hua Liu
- Qingdao Mental Health Center, Qingdao University, Qingdao, China;
| | - Shuling Jiang
- Department of Neurology, Linyi Central Hospital, Shandong, China;
| | - Lei Su
- Department of Psychiatry, Shandong Mental Health Center, Jinan, China;
| | - Yi Lu
- Department of Psychiatry, Shandong Mental Health Center, Jinan, China;
| | - Zhenli Chen
- Department of Psychiatry, Shandong Mental Health Center, Jinan, China;
| | - Xiaojing Li
- Department of Psychiatry, Shandong Mental Health Center, Jinan, China;
| | - Xirong Li
- Department of Psychiatry, Shandong Mental Health Center, Jinan, China;
| | - Xuemei Wang
- Department of Psychiatry, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China; ,Address correspondence to these authors at the CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China; E-mail: ; Department of Psychiatry, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China; E-mail: ; Linyin Road, Qixing District, Suzhou, Jiangsu, 215006, China; E-mail:
| | - Meihong Xiu
- Peking University HuiLongGuan Clinical Medical School, Beijing HuiLongGuan Hospital, Beijing, China; ,Address correspondence to these authors at the CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China; E-mail: ; Department of Psychiatry, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China; E-mail: ; Linyin Road, Qixing District, Suzhou, Jiangsu, 215006, China; E-mail:
| | - Xiangyang Zhang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China,Address correspondence to these authors at the CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China; E-mail: ; Department of Psychiatry, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China; E-mail: ; Linyin Road, Qixing District, Suzhou, Jiangsu, 215006, China; E-mail:
| |
Collapse
|
39
|
Alameda L, Trotta G, Quigley H, Rodriguez V, Gadelrab R, Dwir D, Dempster E, Wong CCY, Forti MD. Can epigenetics shine a light on the biological pathways underlying major mental disorders? Psychol Med 2022; 52:1645-1665. [PMID: 35193719 PMCID: PMC9280283 DOI: 10.1017/s0033291721005559] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 11/30/2021] [Accepted: 12/29/2021] [Indexed: 12/27/2022]
Abstract
A significant proportion of the global burden of disease can be attributed to mental illness. Despite important advances in identifying risk factors for mental health conditions, the biological processing underlying causal pathways to disease onset remain poorly understood. This represents a limitation to implement effective prevention and the development of novel pharmacological treatments. Epigenetic mechanisms have emerged as mediators of environmental and genetic risk factors which might play a role in disease onset, including childhood adversity (CA) and cannabis use (CU). Particularly, human research exploring DNA methylation has provided new and promising insights into the role of biological pathways implicated in the aetio-pathogenesis of psychiatric conditions, including: monoaminergic (Serotonin and Dopamine), GABAergic, glutamatergic, neurogenesis, inflammatory and immune response and oxidative stress. While these epigenetic changes have been often studied as disease-specific, similarly to the investigation of environmental risk factors, they are often transdiagnostic. Therefore, we aim to review the existing literature on DNA methylation from human studies of psychiatric diseases (i) to identify epigenetic modifications mapping onto biological pathways either transdiagnostically or specifically related to psychiatric diseases such as Eating Disorders, Post-traumatic Stress Disorder, Bipolar and Psychotic Disorder, Depression, Autism Spectrum Disorder and Anxiety Disorder, and (ii) to investigate a convergence between some of these epigenetic modifications and the exposure to known risk factors for psychiatric disorders such as CA and CU, as well as to other epigenetic confounders in psychiatry research.
Collapse
Affiliation(s)
- Luis Alameda
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Departamento de Psiquiatría, Centro Investigación Biomedica en Red de Salud Mental (CIBERSAM), Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Sevilla, Spain
| | - Giulia Trotta
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, King's College London, London, UK
| | - Harriet Quigley
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Victoria Rodriguez
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Romayne Gadelrab
- Centre for Affective Disorders, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Daniella Dwir
- Department of Psychiatry, Center for Psychiatric Neuroscience, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Emma Dempster
- University of Exeter Medical School, University of Exeter, Barrack Road, Exeter, UK
| | - Chloe C. Y. Wong
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, King's College London, London, UK
| | - Marta Di Forti
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, King's College London, London, UK
- South London and Maudsley NHS Foundation Trust, London, UK
| |
Collapse
|
40
|
Monji A, Mizoguchi Y. Neuroinflammation in Late-Onset Schizophrenia: Viewing from the Standpoint of the Microglia Hypothesis. Neuropsychobiology 2022; 81:98-103. [PMID: 34515181 DOI: 10.1159/000517861] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 06/14/2021] [Indexed: 11/19/2022]
Abstract
Schizophrenia develops mainly in adolescence, but late-onset schizophrenia (LOS) is not uncommon. According to the international consensus, schizophrenia which develops over 40 years old is called LOS and psychosis which develops over 60 years old is called very late-onset schizophrenia-like psychosis (VLOS). Compared to early-onset schizophrenia (EOS) that develops before the age of 40 years, LOS and VLOS are reported to be more common in women, and there are clinically clear differences such as less involvement of genetic factors than EOS. This review outlines the abnormalities of the neuroimmune system in the pathophysiology of LOS, especially focusing on the role of microglia.
Collapse
Affiliation(s)
- Akira Monji
- Department of Psychiatry, Faculty of Medicine, Saga University, Saga, Japan
| | - Yoshito Mizoguchi
- Department of Psychiatry, Faculty of Medicine, Saga University, Saga, Japan
| |
Collapse
|
41
|
Cuenod M, Steullet P, Cabungcal JH, Dwir D, Khadimallah I, Klauser P, Conus P, Do KQ. Caught in vicious circles: a perspective on dynamic feed-forward loops driving oxidative stress in schizophrenia. Mol Psychiatry 2022; 27:1886-1897. [PMID: 34759358 PMCID: PMC9126811 DOI: 10.1038/s41380-021-01374-w] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 10/18/2021] [Accepted: 10/20/2021] [Indexed: 12/18/2022]
Abstract
A growing body of evidence has emerged demonstrating a pathological link between oxidative stress and schizophrenia. This evidence identifies oxidative stress as a convergence point or "central hub" for schizophrenia genetic and environmental risk factors. Here we review the existing experimental and translational research pinpointing the complex dynamics of oxidative stress mechanisms and their modulation in relation to schizophrenia pathophysiology. We focus on evidence supporting the crucial role of either redox dysregulation, N-methyl-D-aspartate receptor hypofunction, neuroinflammation or mitochondria bioenergetics dysfunction, initiating "vicious circles" centered on oxidative stress during neurodevelopment. These processes would amplify one another in positive feed-forward loops, leading to persistent impairments of the maturation and function of local parvalbumin-GABAergic neurons microcircuits and myelinated fibers of long-range macrocircuitry. This is at the basis of neural circuit synchronization impairments and cognitive, emotional, social and sensory deficits characteristic of schizophrenia. Potential therapeutic approaches that aim at breaking these different vicious circles represent promising strategies for timely and safe interventions. In order to improve early detection and increase the signal-to-noise ratio for adjunctive trials of antioxidant, anti-inflammatory and NMDAR modulator drugs, a reverse translation of validated circuitry approach is needed. The above presented processes allow to identify mechanism based biomarkers guiding stratification of homogenous patients groups and target engagement required for successful clinical trials, paving the way towards precision medicine in psychiatry.
Collapse
Affiliation(s)
- Michel Cuenod
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), Prilly, Lausanne, Switzerland
| | - Pascal Steullet
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), Prilly, Lausanne, Switzerland
| | - Jan-Harry Cabungcal
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), Prilly, Lausanne, Switzerland
| | - Daniella Dwir
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), Prilly, Lausanne, Switzerland
| | - Ines Khadimallah
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), Prilly, Lausanne, Switzerland
| | - Paul Klauser
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), Prilly, Lausanne, Switzerland
- Service of Child and Adolescent Psychiatry, Department of Psychiatry, Lausanne University Hospital, Prilly, Lausanne, Switzerland
| | - Philippe Conus
- Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital, Prilly, Lausanne, Switzerland
| | - Kim Q Do
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), Prilly, Lausanne, Switzerland.
| |
Collapse
|
42
|
Uehara T, Kurachi M, Kondo T, Abe H, Itoh H, Sumiyoshi T, Suzuki M. Apocynin-Tandospirone Derivatives Suppress Methamphetamine-Induced Hyperlocomotion in Rats with Neonatal Exposure to Dizocilpine. J Pers Med 2022; 12:jpm12030366. [PMID: 35330366 PMCID: PMC8951253 DOI: 10.3390/jpm12030366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/24/2022] [Accepted: 02/25/2022] [Indexed: 12/10/2022] Open
Abstract
Accumulating evidence implicates oxidative stress as a potential pathophysiological mechanism of schizophrenia. Accordingly, we synthesized new chemicals using apocynin and tandospirone as lead compounds (A-2, A-3 and A-4). These novel compounds decreased reactive oxygen species (ROS) concentrations in vitro and reversed decreases in glutathione levels in the medial prefrontal cortex of rats transiently exposed to MK-801, an N-methyl-d-aspartate receptor antagonist, in the neonatal period. To determine whether A-2, A-3 and A-4 show behavioral effects associated with antipsychotic properties, the effects of these compounds on methamphetamine (MAP)-induced locomotor and vertical activity were examined in the model rats. A-2 and A-3, administered for 14 days around the puberty period, ameliorated MAP-induced hyperlocomotion in MK-801-treated rats in the post-puberty period, while A-4 suppressed MAP-induced vertical activity. These findings indicate that apocynin-tandospirone derivatives present anti-dopaminergic effects and may alleviate psychotic symptoms of schizophrenia.
Collapse
Affiliation(s)
- Takashi Uehara
- Department of Neuropsychiatry, Kanazawa Medical University, Uchinada 920-0293, Japan
- Correspondence: ; Tel.: +81-76-286-2211 (ext. 3437); Fax: +81-76-286-3341
| | - Masayoshi Kurachi
- Department of Neuropsychiatry, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan; (M.K.); (H.I.); (M.S.)
| | - Takashi Kondo
- Center for Low-Temperature Plasma Sciences, Nagoya University, Nagoya 464-8603, Japan;
| | - Hitoshi Abe
- Department of Applied Chemistry, Faculty of Engineering, University of Toyama, Toyama 930-8555, Japan;
| | - Hiroko Itoh
- Department of Neuropsychiatry, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan; (M.K.); (H.I.); (M.S.)
| | - Tomiki Sumiyoshi
- Department of Preventive Intervention for Psychiatric Disorders, National Institute of Mental Health, National Center of Neurology and Psychiatry, Tokyo 187-8551, Japan;
| | - Michio Suzuki
- Department of Neuropsychiatry, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan; (M.K.); (H.I.); (M.S.)
| |
Collapse
|
43
|
Hei G, Smith RC, Li R, Ou J, Song X, Zheng Y, He Y, Arriaza J, Fahey JW, Cornblatt B, Kang D, Yang Y, Huang J, Wang X, Cadenhead K, Zhang M, Davis JM, Zhao J, Jin H, Wu R. Sulforaphane Effects on Cognition and Symptoms in First and Early Episode Schizophrenia: A Randomized Double-Blind Trial. SCHIZOPHRENIA BULLETIN OPEN 2022; 3:sgac024. [PMID: 39144775 PMCID: PMC11205988 DOI: 10.1093/schizbullopen/sgac024] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/16/2024]
Abstract
Objective Cognitive symptoms are associated with significant dysfunction in schizophrenia. Oxidative stress and inflammation involving histone deacetylase (HDAC) have been implicated in the pathophysiology of schizophrenia. Sulforaphane has antioxidant properties and is an HDAC inhibitor. The objective of this study was to determine the efficacy of sulforaphane on cognition dysfunction for patients with schizophrenia. Methods This double-blind randomized 22-week trial of patients with first-episode schizophrenia was conducted in four psychiatric institutions in China. Patients were randomized to three groups (two doses of sulforaphane vs. placebo) and symptomatic and cognitive assessments were completed at multiple times. The primary outcome measure was change in the MATRICS Composite score. The secondary outcomes were change in MATRICS Domain scores, PANSS Total Scores and change in side-effects. Results A total of 172 patients were randomized and 151 patients had at least one follow up evaluation. There were no significant effects of sulforaphane, on the primary outcome, MATRICS overall composite score. However, on secondary outcomes, sulforaphane did significantly improve performance scores on MATRICS battery Domains of spatial working memory (F = 5.68, P = 0.004), reasoning-problem solving (F = 2.82, P = 0.063), and verbal learning (F = 3.56, P = 0.031). There were no effects on PANSS symptom scores. Sulforaphane was well tolerated. Conclusion Although the primary outcome was not significant, improvement in three domains of the MATRICS battery, suggests a positive cognitive effect on some cognitive functions, which warrants further clinical trials to further assess whether sulforaphane may be a useful adjunct for treating some types of cognitive deficits in schizophrenia.
Collapse
Affiliation(s)
- Gangrui Hei
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders,the Second Xiangya Hospital of Central South University; China National Clinical Research Center on Mental Disorders; Hunan Medical Center for Mental Health, China National Technology Institute on Mental Disorders; Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, Hunan 410011, China
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Robert C Smith
- Nathan S. Kline Institute for Psychiatric Research, Orangeburg, New York, USA
- Department of Psychiatry, New York University School of Medicine, New York, NY, USA
| | - Ranran Li
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders,the Second Xiangya Hospital of Central South University; China National Clinical Research Center on Mental Disorders; Hunan Medical Center for Mental Health, China National Technology Institute on Mental Disorders; Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, Hunan 410011, China
| | - Jianjun Ou
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders,the Second Xiangya Hospital of Central South University; China National Clinical Research Center on Mental Disorders; Hunan Medical Center for Mental Health, China National Technology Institute on Mental Disorders; Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, Hunan 410011, China
| | - Xueqing Song
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Yingjun Zheng
- The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510000, China
| | - Yiqun He
- The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453002, China
| | - Jen Arriaza
- School of Professional Studies, New York University, New York, NY, USA
| | - Jed W Fahey
- Center for Human Nutrition, International Health. School of Medicine, John Hopkins University, Baltimore, Maryland, USA
| | | | - Dongyu Kang
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders,the Second Xiangya Hospital of Central South University; China National Clinical Research Center on Mental Disorders; Hunan Medical Center for Mental Health, China National Technology Institute on Mental Disorders; Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, Hunan 410011, China
| | - Ye Yang
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders,the Second Xiangya Hospital of Central South University; China National Clinical Research Center on Mental Disorders; Hunan Medical Center for Mental Health, China National Technology Institute on Mental Disorders; Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, Hunan 410011, China
| | - Jing Huang
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders,the Second Xiangya Hospital of Central South University; China National Clinical Research Center on Mental Disorders; Hunan Medical Center for Mental Health, China National Technology Institute on Mental Disorders; Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, Hunan 410011, China
| | - Xiaoyi Wang
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders,the Second Xiangya Hospital of Central South University; China National Clinical Research Center on Mental Disorders; Hunan Medical Center for Mental Health, China National Technology Institute on Mental Disorders; Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, Hunan 410011, China
| | - Kristin Cadenhead
- Department of Psychiatry, University of California San Diego and Psychiatric Service, VA San Diego Healthcare System, San Diego, CA, USA
| | - Mimei Zhang
- Columbia University Mailman School of Public Health, New York City, NY, USA
| | - John M Davis
- Department of Psychiatry, University of Illinois, Chicago, IL, USA
| | - Jingping Zhao
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders,the Second Xiangya Hospital of Central South University; China National Clinical Research Center on Mental Disorders; Hunan Medical Center for Mental Health, China National Technology Institute on Mental Disorders; Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, Hunan 410011, China
| | - Hua Jin
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders,the Second Xiangya Hospital of Central South University; China National Clinical Research Center on Mental Disorders; Hunan Medical Center for Mental Health, China National Technology Institute on Mental Disorders; Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, Hunan 410011, China
- Department of Psychiatry, University of California San Diego and Psychiatric Service, VA San Diego Healthcare System, San Diego, CA, USA
| | - Renrong Wu
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders,the Second Xiangya Hospital of Central South University; China National Clinical Research Center on Mental Disorders; Hunan Medical Center for Mental Health, China National Technology Institute on Mental Disorders; Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, Hunan 410011, China
| |
Collapse
|
44
|
Walther S, Lefebvre S, Conring F, Gangl N, Nadesalingam N, Alexaki D, Wüthrich F, Rüter M, Viher PV, Federspiel A, Wiest R, Stegmayer K. Limbic links to paranoia: increased resting-state functional connectivity between amygdala, hippocampus and orbitofrontal cortex in schizophrenia patients with paranoia. Eur Arch Psychiatry Clin Neurosci 2022; 272:1021-1032. [PMID: 34636951 PMCID: PMC9388427 DOI: 10.1007/s00406-021-01337-w] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 09/22/2021] [Indexed: 11/24/2022]
Abstract
Paranoia is a frequent and highly distressing experience in psychosis. Models of paranoia suggest limbic circuit pathology. Here, we tested whether resting-state functional connectivity (rs-fc) in the limbic circuit was altered in schizophrenia patients with current paranoia. We collected MRI scans in 165 subjects including 89 patients with schizophrenia spectrum disorders (schizophrenia, schizoaffective disorder, brief psychotic disorder, schizophreniform disorder) and 76 healthy controls. Paranoia was assessed using a Positive And Negative Syndrome Scale composite score. We tested rs-fc between bilateral nucleus accumbens, hippocampus, amygdala and orbitofrontal cortex between groups and as a function of paranoia severity. Patients with paranoia had increased connectivity between hippocampus and amygdala compared to patients without paranoia. Likewise, paranoia severity was linked to increased connectivity between hippocampus and amygdala. Furthermore, paranoia was associated with increased connectivity between orbitofrontal and medial prefrontal cortex. In addition, patients with paranoia had increased functional connectivity within the frontal hubs of the default mode network compared to healthy controls. These results demonstrate that current paranoia is linked to aberrant connectivity within the core limbic circuit and prefrontal cortex reflecting amplified threat processing and impaired emotion regulation. Future studies will need to explore the association between limbic hyperactivity, paranoid ideation and perceived stress.
Collapse
Affiliation(s)
- Sebastian Walther
- Translational Research Center, University Hospital of Psychiatry and Psychotherapy, University of Bern, Bern, Switzerland
| | - Stephanie Lefebvre
- Translational Research Center, University Hospital of Psychiatry and Psychotherapy, University of Bern, Bern, Switzerland.
| | - Frauke Conring
- Translational Research Center, University Hospital of Psychiatry and Psychotherapy, University of Bern, Bern, Switzerland
| | - Nicole Gangl
- Translational Research Center, University Hospital of Psychiatry and Psychotherapy, University of Bern, Bern, Switzerland
| | - Niluja Nadesalingam
- Translational Research Center, University Hospital of Psychiatry and Psychotherapy, University of Bern, Bern, Switzerland
| | - Danai Alexaki
- Translational Research Center, University Hospital of Psychiatry and Psychotherapy, University of Bern, Bern, Switzerland
| | - Florian Wüthrich
- Translational Research Center, University Hospital of Psychiatry and Psychotherapy, University of Bern, Bern, Switzerland
| | - Maximilian Rüter
- Translational Research Center, University Hospital of Psychiatry and Psychotherapy, University of Bern, Bern, Switzerland
| | - Petra V. Viher
- Translational Research Center, University Hospital of Psychiatry and Psychotherapy, University of Bern, Bern, Switzerland
| | - Andrea Federspiel
- Translational Research Center, University Hospital of Psychiatry and Psychotherapy, University of Bern, Bern, Switzerland
| | - Roland Wiest
- Institute of Diagnostic and Interventional Neuroradiology, Inselspital, University of Bern, Bern, Switzerland
| | - Katharina Stegmayer
- Translational Research Center, University Hospital of Psychiatry and Psychotherapy, University of Bern, Bern, Switzerland
| |
Collapse
|
45
|
Susai SR, Mongan D, Healy C, Cannon M, Nelson B, Markulev C, Schäfer MR, Berger M, Mossaheb N, Schlögelhofer M, Smesny S, Hickie IB, Berger GE, Chen EYH, de Haan L, Nieman DH, Nordentoft M, Riecher-Rössler A, Verma S, Thompson A, Yung AR, McGorry PD, Föcking M, Cotter D, Amminger GP. The association of plasma inflammatory markers with omega-3 fatty acids and their mediating role in psychotic symptoms and functioning: An analysis of the NEURAPRO clinical trial. Brain Behav Immun 2022; 99:147-156. [PMID: 34624483 DOI: 10.1016/j.bbi.2021.09.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/17/2021] [Accepted: 09/25/2021] [Indexed: 12/12/2022] Open
Abstract
There is increasing evidence that dysregulation of polyunsaturated fatty acids (FAs) mediated membrane function plays a role in the pathophysiology of schizophrenia. Even though preclinical findings have supported the anti-inflammatory properties of omega-3 FAs on brain health, their biological roles as anti-inflammatory agents and their therapeutic role on clinical symptoms of psychosis risk are not well understood. In the current study, we investigated the relationship of erythrocyte omega-3 FAs with plasma immune markers in a clinical high risk for psychosis (CHR) sample. In addition, a mediation analysis was performed to examine whether previously reported associations between omega-3 FAs and clinical outcomes were mediated via plasma immune markers. Clinical outcomes for CHR participants in the NEURAPRO clinical trial were measured using the Brief Psychiatric Rating Scale (BPRS), Schedule for the Scale of Assessment of Negative Symptoms (SANS) and Social and Occupational Functioning Assessment Scale (SOFAS) scales. The erythrocyte omega-3 index [eicosapentaenoic acid (EPA) + docosahexaenoic acid (DHA)] and plasma concentrations of inflammatory markers were quantified at baseline (n = 268) and 6 month follow-up (n = 146) by gas chromatography and multiplex immunoassay, respectively. In linear regression models, the baseline plasma concentrations of Interleukin (IL)-15, Intercellular adhesion molecule (ICAM)-1 and Vascular cell adhesion molecule (VCAM)-1 were negatively associated with baseline omega-3 index. In addition, 6-month change in IL-12p40 and TNF-α showed a negative association with change in omega-3 index. In longitudinal analyses, the baseline and 6 month change in omega-3 index was negatively associated with VCAM-1 and TNF-α respectively at follow-up. Mediation analyses provided little evidence for mediating effects of plasma immune markers on the relationship between omega-3 FAs and clinical outcomes (psychotic symptoms and functioning) in CHR participants. Our results indicate a predominantly anti-inflammatory relationship of omega-3 FAs on plasma inflammatory status in CHR individuals, but this did not appear to convey clinical benefits at 6 month and 12 month follow-up. Both immune and non-immune biological effects of omega-3 FAs would be resourceful in understanding the clinical benefits of omega-3 FAs in CHR papulation.
Collapse
Affiliation(s)
- Subash Raj Susai
- Department of Psychiatry, RCSI University of Medicine and Health Sciences, Dublin, Ireland.
| | - David Mongan
- Department of Psychiatry, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Colm Healy
- Department of Psychiatry, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Mary Cannon
- Department of Psychiatry, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Barnaby Nelson
- Centre for Youth Mental Health, The University of Melbourne, Melbourne, Victoria, Australia; Orygen, 35 Poplar Rd, Parkville 3052, Australia
| | - Connie Markulev
- Centre for Youth Mental Health, The University of Melbourne, Melbourne, Victoria, Australia; Orygen, 35 Poplar Rd, Parkville 3052, Australia
| | - Miriam R Schäfer
- Centre for Youth Mental Health, The University of Melbourne, Melbourne, Victoria, Australia; Orygen, 35 Poplar Rd, Parkville 3052, Australia
| | - Maximus Berger
- Centre for Youth Mental Health, The University of Melbourne, Melbourne, Victoria, Australia; Orygen, 35 Poplar Rd, Parkville 3052, Australia
| | - Nilufar Mossaheb
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | - Monika Schlögelhofer
- BioPsyC-Biopsychosocial Corporation - Non-Profit Association for Research Funding, Austria; Department of Child and Adolescent Psychiatry, Medical University Vienna, Austria
| | - Stefan Smesny
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Jena, Germany
| | - Ian B Hickie
- Brain and Mind Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Gregor E Berger
- Child and Adolescent Psychiatric Service of the Canton of Zurich, Zürich, Switzerland
| | - Eric Y H Chen
- Department of Psychiatry, University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Lieuwe de Haan
- Department of Psychiatry, Academic Medical Center, Amsterdam, the Netherlands
| | - Dorien H Nieman
- Department of Psychiatry, Academic Medical Center, Amsterdam, the Netherlands
| | - Merete Nordentoft
- Mental Health Center Copenhagen, Department of Clinical Medicine, Copenhagen University Hospital, Denmark
| | | | - Swapna Verma
- Institute of Mental Health, Singapore, Singapore
| | - Andrew Thompson
- Centre for Youth Mental Health, The University of Melbourne, Melbourne, Victoria, Australia; Orygen, 35 Poplar Rd, Parkville 3052, Australia
| | - Alison Ruth Yung
- Centre for Youth Mental Health, The University of Melbourne, Melbourne, Victoria, Australia; Orygen, 35 Poplar Rd, Parkville 3052, Australia; Institute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong, Australia; School of Health Sciences, University of Manchester, UK
| | - Patrick D McGorry
- Centre for Youth Mental Health, The University of Melbourne, Melbourne, Victoria, Australia; Orygen, 35 Poplar Rd, Parkville 3052, Australia
| | - Melanie Föcking
- Department of Psychiatry, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - David Cotter
- Department of Psychiatry, RCSI University of Medicine and Health Sciences, Dublin, Ireland.
| | - G Paul Amminger
- Centre for Youth Mental Health, The University of Melbourne, Melbourne, Victoria, Australia; Orygen, 35 Poplar Rd, Parkville 3052, Australia; Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
46
|
Li Z, Zhang T, Xu L, Wei Y, Cui H, Tang Y, Liu X, Qian Z, Zhang H, Liu P, Li C, Wang J. Plasma metabolic alterations and potential biomarkers in individuals at clinical high risk for psychosis. Schizophr Res 2022; 239:19-28. [PMID: 34800912 DOI: 10.1016/j.schres.2021.11.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 10/21/2021] [Accepted: 11/08/2021] [Indexed: 01/10/2023]
Abstract
BACKGROUND Early identification and treatment of clinical high-risk for psychosis (CHRP) are critical to prevent the onset of psychosis, but there is no objective biomarker for CHR-P diagnosis. METHODS Ninety medication naïve CHR-P subjects and eighty-six healthy controls (HCs) were recruited. The metabolic profiles of plasma samples were acquired using an untargeted metabolomics approach based on ultra-high-performance liquid chromatography equipped with quadrupole time-of-flight mass spectrometry. The obtained data were further mapped on the Kyoto Encyclopedia of Genes and Genomes for pathway analysis, and an ensemble learning method was applied to identify diagnostic biomarkers. Bayesian linear regression model was then used to explore predicative biomarkers of conversion to psychosis. Receiver-operating characteristic (ROC) curve analysis was performed to evaluate the diagnostic or predicative value of potential biomarkers. RESULTS A total of one hundred and four differential metabolites and forty-eight differential pathways were identified. A panel of five metabolites was found that could effectively discriminate CHR-P from HCs with area under the ROC curve of 1 in the training set (70% of the samples) and 0.997 in the testing set (30% of the samples). The biosynthesis of unsaturated fatty acids pathway perturbed most significantly in CHR-P subjects. Twenty-three CHR-P subjects converted to psychotic disorders during two-year follow-up, and increased 1-stearoyl-2-arachidonoyl-sn-glycerol in plasma was potentially associated with the higher risk of conversion to psychosis. CONCLUSIONS These findings demonstrate the alterations of plasma metabolic profiles in CHR-P population, which may deliver valuable biomarkers for early identification and outcome prediction of CHR-P.
Collapse
Affiliation(s)
- Zhixing Li
- Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai Key Laboratory of Psychotic Disorders, Shanghai 200030, PR China
| | - Tianhong Zhang
- Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai Key Laboratory of Psychotic Disorders, Shanghai 200030, PR China.
| | - Lihua Xu
- Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai Key Laboratory of Psychotic Disorders, Shanghai 200030, PR China
| | - Yanyan Wei
- Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai Key Laboratory of Psychotic Disorders, Shanghai 200030, PR China
| | - Huiru Cui
- Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai Key Laboratory of Psychotic Disorders, Shanghai 200030, PR China
| | - Yingying Tang
- Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai Key Laboratory of Psychotic Disorders, Shanghai 200030, PR China
| | - Xiaohua Liu
- Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai Key Laboratory of Psychotic Disorders, Shanghai 200030, PR China
| | - Zhenying Qian
- Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai Key Laboratory of Psychotic Disorders, Shanghai 200030, PR China
| | - Hu Zhang
- School of Pharmacy, Brain Health Research Centre, Brain Research New Zealand, University of Otago, Dunedin, New Zealand
| | - Ping Liu
- Department of Anatomy, School of Biomedical Sciences, Brain Health Research Centre, Brain Research New Zealand, University of Otago, Dunedin, New Zealand.
| | - Chunbo Li
- Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai Key Laboratory of Psychotic Disorders, Shanghai 200030, PR China
| | - Jijun Wang
- Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai Key Laboratory of Psychotic Disorders, Shanghai 200030, PR China; CAS Center for Excellence in Brain Science and Intelligence Technology (CEBSIT), Chinese Academy of Science, Shanghai 200031, PR China; Institute of Psychology and Behavioral Science, Shanghai Jiao Tong University, Shanghai 200030, PR China.
| |
Collapse
|
47
|
Khadimallah I, Jenni R, Cabungcal JH, Cleusix M, Fournier M, Beard E, Klauser P, Knebel JF, Murray MM, Retsa C, Siciliano M, Spencer KM, Steullet P, Cuenod M, Conus P, Do KQ. Mitochondrial, exosomal miR137-COX6A2 and gamma synchrony as biomarkers of parvalbumin interneurons, psychopathology, and neurocognition in schizophrenia. Mol Psychiatry 2022; 27:1192-1204. [PMID: 34686767 PMCID: PMC9054672 DOI: 10.1038/s41380-021-01313-9] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 09/01/2021] [Accepted: 09/22/2021] [Indexed: 12/16/2022]
Abstract
Early detection and intervention in schizophrenia requires mechanism-based biomarkers that capture neural circuitry dysfunction, allowing better patient stratification, monitoring of disease progression and treatment. In prefrontal cortex and blood of redox dysregulated mice (Gclm-KO ± GBR), oxidative stress induces miR-137 upregulation, leading to decreased COX6A2 and mitophagy markers (NIX, Fundc1, and LC3B) and to accumulation of damaged mitochondria, further exacerbating oxidative stress and parvalbumin interneurons (PVI) impairment. MitoQ, a mitochondria-targeted antioxidant, rescued all these processes. Translating to early psychosis patients (EPP), blood exosomal miR-137 increases and COX6A2 decreases, combined with mitophagy markers alterations, suggest that observations made centrally and peripherally in animal model were reflected in patients' blood. Higher exosomal miR-137 and lower COX6A2 levels were associated with a reduction of ASSR gamma oscillations in EEG. As ASSR requires proper PVI-related networks, alterations in miR-137/COX6A2 plasma exosome levels may represent a proxy marker of PVI cortical microcircuit impairment. EPP can be stratified in two subgroups: (a) a patients' group with mitochondrial dysfunction "Psy-D", having high miR-137 and low COX6A2 levels in exosomes, and (b) a "Psy-ND" subgroup with no/low mitochondrial impairment, including patients having miR-137 and COX6A2 levels in the range of controls. Psy-D patients exhibited more impaired ASSR responses in association with worse psychopathological status, neurocognitive performance, and global and social functioning, suggesting that impairment of PVI mitochondria leads to more severe disease profiles. This stratification would allow, with high selectivity and specificity, the selection of patients for treatments targeting brain mitochondria dysregulation and capture the clinical and functional efficacy of future clinical trials.
Collapse
Affiliation(s)
- Ines Khadimallah
- grid.8515.90000 0001 0423 4662Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Raoul Jenni
- grid.8515.90000 0001 0423 4662Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), Lausanne, Switzerland ,grid.9851.50000 0001 2165 4204Service of General Psychiatry, Department of Psychiatry, University Hospital Center and University of Lausanne, Prilly Lausanne, Switzerland
| | - Jan-Harry Cabungcal
- grid.8515.90000 0001 0423 4662Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Martine Cleusix
- grid.8515.90000 0001 0423 4662Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), Lausanne, Switzerland ,grid.9851.50000 0001 2165 4204Service of General Psychiatry, Department of Psychiatry, University Hospital Center and University of Lausanne, Prilly Lausanne, Switzerland
| | - Margot Fournier
- grid.8515.90000 0001 0423 4662Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Elidie Beard
- grid.8515.90000 0001 0423 4662Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Paul Klauser
- grid.8515.90000 0001 0423 4662Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), Lausanne, Switzerland ,grid.9851.50000 0001 2165 4204Service of General Psychiatry, Department of Psychiatry, University Hospital Center and University of Lausanne, Prilly Lausanne, Switzerland
| | - Jean-François Knebel
- grid.8515.90000 0001 0423 4662The LINE (Laboratory for Investigative Neurophysiology), Radiodiagnostic Service, University Hospital Center and University of Lausanne, 1011 Lausanne, Switzerland ,grid.8515.90000 0001 0423 4662Sensory, Perceptual and Cognitive Neuroscience Section, Center for Biomedical Imaging (CIBM), University Hospital Center and University of Lausanne, 1011 Lausanne, Switzerland
| | - Micah M. Murray
- grid.8515.90000 0001 0423 4662The LINE (Laboratory for Investigative Neurophysiology), Radiodiagnostic Service, University Hospital Center and University of Lausanne, 1011 Lausanne, Switzerland ,grid.8515.90000 0001 0423 4662Sensory, Perceptual and Cognitive Neuroscience Section, Center for Biomedical Imaging (CIBM), University Hospital Center and University of Lausanne, 1011 Lausanne, Switzerland ,grid.428685.50000 0004 0627 5427Ophthalmology Department, Fondation Asile des Aveugles and University of Lausanne, Lausanne, Switzerland
| | - Chrysa Retsa
- grid.8515.90000 0001 0423 4662The LINE (Laboratory for Investigative Neurophysiology), Radiodiagnostic Service, University Hospital Center and University of Lausanne, 1011 Lausanne, Switzerland ,grid.8515.90000 0001 0423 4662Sensory, Perceptual and Cognitive Neuroscience Section, Center for Biomedical Imaging (CIBM), University Hospital Center and University of Lausanne, 1011 Lausanne, Switzerland
| | - Milena Siciliano
- grid.8515.90000 0001 0423 4662Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Kevin M. Spencer
- grid.410370.10000 0004 4657 1992Neural Dynamics Laboratory, Research Service, Veterans Affairs Boston Healthcare System, Boston, MA USA ,grid.38142.3c000000041936754XDepartment of Psychiatry, Harvard Medical School, Boston, MA USA
| | - Pascal Steullet
- grid.8515.90000 0001 0423 4662Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Michel Cuenod
- grid.8515.90000 0001 0423 4662Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Philippe Conus
- grid.9851.50000 0001 2165 4204Service of General Psychiatry, Department of Psychiatry, University Hospital Center and University of Lausanne, Prilly Lausanne, Switzerland
| | - Kim Q. Do
- grid.8515.90000 0001 0423 4662Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| |
Collapse
|
48
|
Raj A, Kaushal A, Datta I. Impact of monomeric and aggregated wild-type and A30P/A53T double-mutant α-synuclein on antioxidant mechanism and glutamate metabolic profile of cultured astrocytes. J Neurosci Res 2021; 100:681-706. [PMID: 34904280 DOI: 10.1002/jnr.24994] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 10/28/2021] [Accepted: 11/15/2021] [Indexed: 12/17/2022]
Abstract
Serving as a source of glutathione and up-taking and metabolizing glutamate are the primary supportive role of astrocytes for the adjacent neurons. Despite the clear physical association between astrocytes and α-synuclein, the effect of extracellular α-synuclein on these astrocytic functions has not yet been elucidated. Hence, we aim to assess the effect of various forms of α-synuclein on antioxidant mechanism and glutamate metabolism. Wild-type and A53T/A30P double-mutant α-synuclein, both in monomeric and aggregated forms, were added extracellularly to media of midbrain rat astrocyte culture, with their survival, oxidative, and nitrative stress, glutathione and glutamate content, expression of enzymes associated with oxidative stress and glutamate metabolism, glutamate and glutathione transporters being assessed along with the association/engulfment of these peptides by astrocytes. A30P/A53T peptide associated more with astrocytes, and low-extracellular K+ concentration showed prominent reduction in the engulfment of the monomeric forms, suggesting that the association of the aggregated forms was greater with the membrane. The peptide-associated astrocytes showed lower survival and increased oxidative stress generation, owing to the decrease in nuclear localization of Nrf2 and increase in iNOS, and further aggravated by the decrease in glutathione content and related enzymes like glutathione synthetase, glutathione peroxidase, and glutathione reductase. Glutamate uptake increased in aggregate-treated cells due to the increase in GLAST1 expression, de novo synthesis of glutamate by pyruvate carboxylase, and/or glutamine synthase, bolstered by the differential glutamate dehydrogenase enzyme activity. We thus show for the first time that extracellular α-synuclein exposure leads to astrocytic dysfunction with respect to the antioxidant mechanism and glutamate metabolic profile. The impact was higher in the case of the aggregated and mutated peptide, with the highest dysfunction for the mutant aggregated α-synuclein treatment.
Collapse
Affiliation(s)
- Aishwarya Raj
- Department of Biophysics, National Institute of Mental Health and Neurosciences, Institute of National Importance, Bengaluru, India
| | - Alka Kaushal
- Department of Biophysics, National Institute of Mental Health and Neurosciences, Institute of National Importance, Bengaluru, India
| | - Indrani Datta
- Department of Biophysics, National Institute of Mental Health and Neurosciences, Institute of National Importance, Bengaluru, India
| |
Collapse
|
49
|
Thalamic reticular nucleus impairments and abnormal prefrontal control of dopamine system in a developmental model of schizophrenia: prevention by N-acetylcysteine. Mol Psychiatry 2021; 26:7679-7689. [PMID: 34193975 PMCID: PMC8716611 DOI: 10.1038/s41380-021-01198-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 06/10/2021] [Indexed: 02/06/2023]
Abstract
Recent evidence showed thalamic abnormalities in schizophrenia involving disruptions to the parvalbumin neurons in the thalamic reticular nucleus (TRN). However, their functional consequences, as well as a potential linkage to oxidative stress, are unclear. The TRN is posited to gate prefrontal control of dopamine neuron activity in the ventral tegmental area (VTA). Thus, we hypothesized that schizophrenia-related TRN abnormalities might contribute to dopamine dysregulation, a well-known feature of the disorder. To test this, in adult rats exposed prenatally to methylazoxymethanol acetate (MAM rats), oxidative impairments to the parvalbumin neurons in the anterior TRN were assessed by immunohistochemistry. Using in vivo electrophysiology, we investigated whether inactivation of the prefrontal cortex would produce differential effects on VTA dopamine neurons in MAM rats. We show that MAM rats displayed reduced markers of parvalbumin and wisteria floribunda agglutinin-labeled perineuronal nets, correlating with increased markers of oxidative stress (8-oxo-7, 8-dihydro-20-deoxyguanosine, and 3-nitrotyrosine). Moreover, MAM rats displayed heightened baseline and abnormal prefrontal control of VTA dopamine neuron activity, as tetrodotoxin-induced inactivation of the infralimbic prefrontal cortex decreased the dopamine population activity, contrary to the normal increase in controls. Such dopamine neuron dysregulation was recapitulated by enzymatic perineuronal net digestion in the TRN of normal rats. Furthermore, juvenile (postnatal day 11-25) antioxidant treatment (N-acetyl-cysteine, 900 mg/L drinking water) prevented all these impairments in MAM rats. Our findings suggest that early accumulation of oxidative stress in the TRN may shape the later onset of schizophrenia pathophysiology, highlighting redox regulation as a potential target for early intervention.
Collapse
|
50
|
Palaniyappan L, Sabesan P, Li X, Luo Q. Schizophrenia Increases Variability of the Central Antioxidant System: A Meta-Analysis of Variance From MRS Studies of Glutathione. Front Psychiatry 2021; 12:796466. [PMID: 34916980 PMCID: PMC8669304 DOI: 10.3389/fpsyt.2021.796466] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 11/02/2021] [Indexed: 11/13/2022] Open
Abstract
Patients with schizophrenia diverge in their clinical trajectories. Such diverge outcomes may result from the resilience provided by antioxidant response system centered on glutathione (GSH). Proton Magnetic Resonance Spectroscopy (1H-MRS) has enabled the precise in vivo measurement of intracortical GSH; but individual studies report highly variable results even when GSH levels are measured from the same brain region. This inconsistency could be due to the presence of distinct subgroups of schizophrenia with varying GSH-levels. At present, we do not know if schizophrenia increases the interindividual variability of intracortical GSH relative to matched healthy individuals. We reviewed all 1H-MRS GSH studies in schizophrenia focused on the Anterior Cingulate Cortex published until August 2021. We estimated the relative variability of ACC GSH levels in patients compared to control groups using the variability ratio (VR) and coefficient of variation ratio (CVR). The presence of schizophrenia significantly increases the variability of intracortical GSH in the ACC (logVR = 0.12; 95% CI: 0.03-0.21; log CVR = 0.15; 95% CI = 0.06-0.23). Insofar as increased within-group variability (heterogeneity) could result from the existence of subtypes, our results call for a careful examination of intracortical GSH distribution in schizophrenia to seek redox-deficient and redox-sufficient subgroups. An increase in GSH variability among patients also indicate that the within-group predictability of adaptive response to oxidative stress may be lower in schizophrenia. Uncovering the origins of this illness-related reduction in the redox system stability may provide novel treatment targets in schizophrenia.
Collapse
Affiliation(s)
- Lena Palaniyappan
- Department of Psychiatry, University of Western Ontario, London, ON, Canada
- Robarts Research Institute, London, ON, Canada
- Lawson Health Research Institute, London, ON, Canada
- Department of Medical Biophysics, University of Western Ontario, London, ON, Canada
| | | | - Xuan Li
- MOE-Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
| | - Qiang Luo
- MOE-Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
- National Clinical Research Center for Aging and Medicine at Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science and Human Phenome Institute, Fudan University, Shanghai, China
| |
Collapse
|