1
|
Chang S, Maeng S, Kim Y, Bae JN, Lee JS, Kim WH. Factors Affecting Continuation of Aripiprazole Long-acting Injection in the Real World. CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE : THE OFFICIAL SCIENTIFIC JOURNAL OF THE KOREAN COLLEGE OF NEUROPSYCHOPHARMACOLOGY 2025; 23:219-226. [PMID: 40223256 PMCID: PMC12000674 DOI: 10.9758/cpn.24.1232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 10/18/2024] [Accepted: 11/14/2024] [Indexed: 04/15/2025]
Abstract
Objective Aripiprazole long-acting injection (LAI) is both effective and widely used in clinical practice. Notably, no previous studies have examined the factors affecting the continuation of aripiprazole LAI within the Korean population. This study aims to identify real-world factors contributing to the continuation of aripiprazole LAI. Methods A 1-year retrospective cohort study was conducted on 68 patients initiating aripiprazole LAI at Inha University Hospital, Korea. Patient medical records were reviewed to assess continuation rates, the duration from initiation to discontinuation, and reasons for discontinuation. Results Overall, 27.9% of patients discontinued aripiprazole LAI within 12 months. The predominant reasons for discontinuation were insufficient efficacy (42.1%), adverse effects (31.6%), and preference for oral medication (21.1%). Univariate analysis revealed associations between discontinuation and patient compliance. After adjustment for sociodemographic factors, patient compliance and transitioning from paliperidone LAI to aripiprazole LAI were associated with the continuation of aripiprazole LAI. Multiple logistic regression analysis, adjusted for sociodemographic variables and compliance, identified significant associations between discontinuation and compliance. Conclusion This study supports the clinical effectiveness of aripiprazole LAI, demonstrated by a reduced one-year discontinuation rate compared to that observed in a paliperidone LAI study with a similar design. Large-scale, long-term prospective studies are essential to definitively ascertain the factors associated with LAI discontinuation.
Collapse
Affiliation(s)
- Soyeon Chang
- Department of Psychiatry, Inha University Hospital, Inha University College of Medicine, Incheon, Korea
| | - Seri Maeng
- Department of Psychiatry, Inha University Hospital, Inha University College of Medicine, Incheon, Korea
| | - Yangsik Kim
- Department of Psychiatry, Inha University Hospital, Inha University College of Medicine, Incheon, Korea
| | - Jae-Nam Bae
- Department of Psychiatry, Inha University Hospital, Inha University College of Medicine, Incheon, Korea
| | - Jeong-Seop Lee
- Department of Psychiatry, Inha University Hospital, Inha University College of Medicine, Incheon, Korea
| | - Won-Hyoung Kim
- Department of Psychiatry, Inha University Hospital, Inha University College of Medicine, Incheon, Korea
| |
Collapse
|
2
|
Wong TY, Moore TM, Hillman N, Calkins ME, Shahriar S, Dietterich T, Ruparel K, Roalf DR, Wolf DH, Satterthwaite TD, Ered A, Gur RE, Gur RC. Longitudinal Development of Neurocognitive Functioning and Gray Matter Volume in Youths With Recurrent Psychosis Spectrum Symptoms. Schizophr Bull 2025:sbaf049. [PMID: 40382716 DOI: 10.1093/schbul/sbaf049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/20/2025]
Abstract
BACKGROUND AND HYPOTHESIS Neurodevelopmental risk-factor models of psychosis highlight the importance of early developmental deviations in the emergence of psychosis. However, few longitudinal studies map neurodevelopment and neurocognitive trajectories across age in preclinical psychosis. We investigated longitudinal trajectories in neurocognition and brain volume in a community cohort of adolescents with recurrent psychosis spectrum (PS) symptoms, tracking their development into young adulthood compared to their typically developing (TD) peers. STUDY DESIGN Utilizing the Philadelphia Neurodevelopmental Cohort, we analyzed data of 231 youths aged 8-30 with at least one follow-up assessment, including 88 with PS. STUDY RESULTS Individuals with PS showed similar developmental trajectories but demonstrated significant impairments in executive functioning (t = -2.81, q = 0.010), memory (t = -2.34, q = 0.019), complex cognition (t = -3.72, q = 0.001), social cognition (t = -2.73, q = 0.010), motor (t = -2.50, q = 0.015), and general cognition (t = -3.20, q = 0.004). Lower cortical (t = -2.46, P = .014) and subcortical (t = -2.41, P = .016) gray matter volume in the recurrent PS group compared to the TD group were documented with age-related group differences becoming less pronounced by young adulthood. Further analyses revealed age-by-group interactions (qs < 0.05) observed in a few temporal and frontal regions, with differences between groups at earlier ages. CONCLUSIONS These findings suggest that recurrent PS symptoms are linked to early neurocognitive and brain structure deficits, highlighting the need for interventions to reduce psychosis risk and support healthy neurodevelopment.
Collapse
Affiliation(s)
- Ting Yat Wong
- Department of Psychiatry, Neurodevelopment and Psychosis Section, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States
- Department of Psychology, The Education University of Hong Kong, Hong Kong Special Administrative Region, 999077, China
- Center of Psychosocial Health, The Education University of Hong, Hong Kong Special Administrative Region, 999077, China
| | - Tyler M Moore
- Department of Psychiatry, Neurodevelopment and Psychosis Section, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States
- Lifespan Brain Institute of the Children's Hospital of Philadelphia and Penn Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States
| | - Noah Hillman
- Department of Psychiatry, Neurodevelopment and Psychosis Section, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States
| | - Monica E Calkins
- Department of Psychiatry, Neurodevelopment and Psychosis Section, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States
- Lifespan Brain Institute of the Children's Hospital of Philadelphia and Penn Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States
| | - Sarah Shahriar
- Department of Psychiatry, Neurodevelopment and Psychosis Section, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States
| | - Tyler Dietterich
- Department of Psychiatry, Neurodevelopment and Psychosis Section, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, 90095, United States
| | - Kosha Ruparel
- Department of Psychiatry, Neurodevelopment and Psychosis Section, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States
- Lifespan Brain Institute of the Children's Hospital of Philadelphia and Penn Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States
| | - David R Roalf
- Department of Psychiatry, Neurodevelopment and Psychosis Section, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States
- Lifespan Brain Institute of the Children's Hospital of Philadelphia and Penn Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States
| | - Daniel H Wolf
- Department of Psychiatry, Neurodevelopment and Psychosis Section, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States
- Lifespan Brain Institute of the Children's Hospital of Philadelphia and Penn Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States
| | - Theodore D Satterthwaite
- Department of Psychiatry, Neurodevelopment and Psychosis Section, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States
- Lifespan Brain Institute of the Children's Hospital of Philadelphia and Penn Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States
| | - Arielle Ered
- Department of Psychiatry, Neurodevelopment and Psychosis Section, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States
| | - Raquel E Gur
- Department of Psychiatry, Neurodevelopment and Psychosis Section, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States
- Lifespan Brain Institute of the Children's Hospital of Philadelphia and Penn Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States
| | - Ruben C Gur
- Department of Psychiatry, Neurodevelopment and Psychosis Section, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States
- Lifespan Brain Institute of the Children's Hospital of Philadelphia and Penn Medicine, University of Pennsylvania, Philadelphia, PA, 19104, United States
| |
Collapse
|
3
|
Moran PM, Granger KT. IUPHAR review: Moving beyond dopamine-based therapeutic strategies for schizophrenia. Pharmacol Res 2025; 216:107727. [PMID: 40320224 DOI: 10.1016/j.phrs.2025.107727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Revised: 03/27/2025] [Accepted: 04/01/2025] [Indexed: 05/12/2025]
Abstract
In the following we comprehensively review approaches to treating schizophrenia that do not primarily involve dopamine antagonism or partial agonism. Following 70 years of broadly similar dopamine D2 receptor antagonist/partial agonist drugs, Cobenfy™ was approved as a novel antipsychotic in September 2024. Cobenfy™ is a combination formulation of xanomeline, a muscarinic cholinergic M1/M4 receptor agonist and trospium, a peripherally restricted muscarinic antagonist included to offset peripheral side effects of xanomeline. This approval has reinvigorated optimism in the field and raised important questions for the future direction of antipsychotic drug development. We review therapeutic strategies beyond dopamine that have been and are currently being investigated to address whether there are a sufficient number of novel approaches to maintain the momentum of this breakthrough and question why it has taken so long. The current pipeline of late-stage compounds is low and potentially constrained by historical setbacks and challenges in clinical trial design for schizophrenia. This success rate has future potential to improve given the range of biomarkers in development designed to enable greater precision in future clinical trials. Cobenfy™ approval demonstrates that with combination formulations designed to improve side effect profiles and optimised clinical trial design it is possible to generate tolerable and efficacious treatment options for patients beyond a solely dopaminergic framework. We conclude that advances in understanding the neurobiology of schizophrenia, while not complete, has generated a diverse and well justified pool of potentially novel and repurpose-ready approaches, with mechanisms beyond simple dopamine D2 antagonism/partial agonism.
Collapse
Affiliation(s)
- Paula M Moran
- School of Psychology, University of Nottingham, University Park, NG72RD, UK.
| | - Kiri T Granger
- Monument Therapeutics Ltd., Alderley Park, Congleton Road, Cheshire, Macclesfield SK10 4TG, UK.
| |
Collapse
|
4
|
Speers LJ, Bilkey DK. Inflammation in Schizophrenia: The Role of Disordered Oscillatory Mechanisms. Cells 2025; 14:650. [PMID: 40358174 PMCID: PMC12071425 DOI: 10.3390/cells14090650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2025] [Revised: 04/16/2025] [Accepted: 04/24/2025] [Indexed: 05/15/2025] Open
Abstract
Schizophrenia is a chronic, debilitating disorder with diverse symptomatology, including disorganised cognition and behaviour. Despite considerable research effort, we have only a limited understanding of the underlying brain dysfunction. A significant proportion of individuals with schizophrenia exhibit high levels of inflammation, and inflammation associated with maternal immune system activation is a risk factor for the disorder. In this review, we outline the potential role of inflammation in the disorder, with a particular focus on how cytokine release might affect the development and function of GABAergic interneurons. One consequence of this change in inhibitory control is a disruption in oscillatory processes in the brain. These changes disrupt the spatial and temporal synchrony of neural activity in the brain, which, by disturbing representations of time and space, may underlie some of the disorganisation symptoms observed in the disorder.
Collapse
Affiliation(s)
| | - David K. Bilkey
- Psychology Department, University of Otago, Dunedin 9016, New Zealand
| |
Collapse
|
5
|
Ghiyamihoor F, Peymani P, Perron J, Asemi‐Rad A, Marzban M, Mohite A, Ardila K, Aljada B, Marzban A, Toback M, Eltonsy S, Ko JH, Siddiqui TJ, Steele CJ, Kong J, Manto M, MacDonald ME, Gill JS, Sillitoe RV, Balcı F, Beheshti I, Marzban H. Volumetric Changes in Cerebellar Transverse Zones: Age and Sex Effects in Health and Neurological Disorders. Hum Brain Mapp 2025; 46:e70214. [PMID: 40241499 PMCID: PMC12003958 DOI: 10.1002/hbm.70214] [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: 01/04/2025] [Revised: 03/05/2025] [Accepted: 04/02/2025] [Indexed: 04/18/2025] Open
Abstract
Cerebellar volumetric changes are intricately linked to aging, with distinct patterns across its transverse zones, the functional subdivisions characterized by unique cytoarchitectural and connectivity profiles. Despite research efforts, the cerebellar aging process in health and neurological disorders remains poorly understood. In this study, we investigated the effects of age and sex on total cerebellum, transverse zone, and lobule volumes using MRI data from over 45,000 participants compiled from six neuroimaging datasets. We also propose a framework for estimating cerebellum age as an indicator of cerebellar health. Significant age-dependent volume reductions were observed across transverse zones, with the central zone (CZ; lobules VI and VII) exhibiting the steepest decline in both health and neurological disorders. This finding highlights the CZ's vulnerability to aging and its critical role in cognitive and emotional processing. We also found prominent sex differences in age-dependent volumetric changes. Males exhibited smaller total intracranial volume (TIV)-adjusted cerebellum volume and faster age-dependent volume reduction than females in both health and mild cognitive impairment (MCI), Alzheimer disease (AD), and Parkinson disease (PD). In contrast, females with schizophrenia (SZ) and cocaine use disorder (CUD) revealed faster age-dependent cerebellar volume reduction than males. Patients with MCI, AD, and PD experienced more pronounced atrophy in the posterior (PZ) and nodular (NZ) zones compared to age-matched healthy controls, while SZ patients were characterized by a more prominent reduction in CZ. In CUD, a non-significant volume decline was observed in all zones compared to the controls. Moreover, our framework for estimating cerebellum age revealed a notable difference in cerebellar aging between healthy individuals and neurological patients. Finally, we charted age-dependent changes in cerebellar volume in healthy individuals, focusing on transverse zones capturing the functional subdivisions. These findings underscore the potential of cerebellar volumetric analysis as a biomarker for early detection and monitoring of neurodegenerative and neuropsychiatric disorders. Our novel approach complements and enhances MRI-based analyses, providing essential insights into the pathogenesis of aging, neurodegeneration, and chronic neuropsychiatric conditions.
Collapse
Affiliation(s)
- Farshid Ghiyamihoor
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health SciencesUniversity of ManitobaWinnipegManitobaCanada
- The Children's Hospital Research Institute of Manitoba (CHRIM), Rady Faculty of Health ScienceUniversity of ManitobaWinnipegManitobaCanada
| | - Payam Peymani
- College of Pharmacy, Rady Faculty of Health SciencesUniversity of ManitobaWinnipegManitobaCanada
| | - Jarrad Perron
- Graduate Program in Biomedical Engineering, Price Faculty of EngineeringUniversity of ManitobaWinnipegManitobaCanada
| | - Azam Asemi‐Rad
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health SciencesUniversity of ManitobaWinnipegManitobaCanada
- The Children's Hospital Research Institute of Manitoba (CHRIM), Rady Faculty of Health ScienceUniversity of ManitobaWinnipegManitobaCanada
| | - Mehdi Marzban
- Department of Electrical & Software Engineering, Schulich School of EngineeringUniversity of CalgaryCalgaryAlbertaCanada
| | - Aashka Mohite
- Department of Biomedical Engineering, Schulich School of EngineeringUniversity of CalgaryCalgaryAlbertaCanada
- Hotchkiss Brain Institute, Cumming School of MedicineUniversity of CalgaryCalgaryAlbertaCanada
| | - Karen Ardila
- Department of Biomedical Engineering, Schulich School of EngineeringUniversity of CalgaryCalgaryAlbertaCanada
- Hotchkiss Brain Institute, Cumming School of MedicineUniversity of CalgaryCalgaryAlbertaCanada
| | - Bara Aljada
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health SciencesUniversity of ManitobaWinnipegManitobaCanada
| | - Asghar Marzban
- Department of Pediatrics, School of MedicineZanjan University of Medical SciencesZanjanIran
| | - Mehnosh Toback
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health SciencesUniversity of ManitobaWinnipegManitobaCanada
| | - Sherif Eltonsy
- The Children's Hospital Research Institute of Manitoba (CHRIM), Rady Faculty of Health ScienceUniversity of ManitobaWinnipegManitobaCanada
- College of Pharmacy, Rady Faculty of Health SciencesUniversity of ManitobaWinnipegManitobaCanada
| | - Ji Hyun Ko
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health SciencesUniversity of ManitobaWinnipegManitobaCanada
- Graduate Program in Biomedical Engineering, Price Faculty of EngineeringUniversity of ManitobaWinnipegManitobaCanada
| | - Tabrez J. Siddiqui
- The Children's Hospital Research Institute of Manitoba (CHRIM), Rady Faculty of Health ScienceUniversity of ManitobaWinnipegManitobaCanada
- Graduate Program in Biomedical Engineering, Price Faculty of EngineeringUniversity of ManitobaWinnipegManitobaCanada
- Department of Physiology and Pathophysiology, Max Rady College of Medicine, Rady Faculty of Health ScienceUniversity of ManitobaWinnipegManitobaCanada
| | - Christopher J. Steele
- Department of Psychology and School of HealthConcordia UniversityMontrealQuebecCanada
- Department of NeurologyMax Planck Institute for Human Cognitive and Brain SciencesLeipzigGermany
| | - Jiming Kong
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health SciencesUniversity of ManitobaWinnipegManitobaCanada
| | - Mario Manto
- Service des NeurosciencesUniversité de MonsMonsBelgium
| | - M. Ethan MacDonald
- Department of Biomedical Engineering, Schulich School of EngineeringUniversity of CalgaryCalgaryAlbertaCanada
- Hotchkiss Brain Institute, Cumming School of MedicineUniversity of CalgaryCalgaryAlbertaCanada
| | - Jason S. Gill
- Jan and Dan Duncan Neurological Research Institute at Texas Children's HospitalHoustonTexasUSA
- Department of Pediatrics, Division of Neurology and Developmental NeuroscienceBaylor College of MedicineHoustonTexasUSA
| | - Roy V. Sillitoe
- Department of Pathology & ImmunologyBaylor College of MedicineHoustonTexasUSA
- Department of NeuroscienceBaylor College of MedicineHoustonTexasUSA
| | - Fuat Balcı
- The Children's Hospital Research Institute of Manitoba (CHRIM), Rady Faculty of Health ScienceUniversity of ManitobaWinnipegManitobaCanada
- Department of Biological Sciences, Faculty of ScienceUniversity of ManitobaWinnipegManitobaCanada
| | - Iman Beheshti
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health SciencesUniversity of ManitobaWinnipegManitobaCanada
| | - Hassan Marzban
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health SciencesUniversity of ManitobaWinnipegManitobaCanada
- The Children's Hospital Research Institute of Manitoba (CHRIM), Rady Faculty of Health ScienceUniversity of ManitobaWinnipegManitobaCanada
| |
Collapse
|
6
|
Carpentier A, Zampetas D, Durand A, Naassila M, Bralet M. Relationships Between Immune-Inflammatory Features and Social Cognitive Impairments in Patients With Schizophrenia Spectrum Disorders: A Systematic Review. Brain Behav 2025; 15:e70384. [PMID: 40159715 PMCID: PMC11955717 DOI: 10.1002/brb3.70384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 01/29/2025] [Accepted: 02/11/2025] [Indexed: 04/02/2025] Open
Abstract
INTRODUCTION Patients with schizophrenia spectrum disorders (SSD), particularly patients with schizophrenia, have social cognitive impairments characterized by difficulties in emotion recognition, the ability to attribute mental states, explaining the causes of events, and identifying and utilizing social cues. These impairments appear from early life and are associated with poor functional and social prognosis. The origin of these impairments is not fully understood. The inflammatory hypothesis is one of the pathophysiological hypotheses of schizophrenia. Inflammatory marker abnormalities are also present in the early stages of schizophrenia and are associated with neuronal degeneration. Following our main hypothesis, the aim of this work was to conduct a review to explore the relationship between social cognition and inflammatory markers in SSD. METHODS The review included original studies reporting measures of social cognition and plasma levels of inflammatory markers in patients with SSD using the Pubmed, PsycINFO, and Embase databases. The PRISMA methodology was followed. RESULTS Eleven studies were selected and analyzed. They showed significant correlations between plasma cytokine levels and theory of mind and facial emotion recognition abilities. CONCLUSION The correlations do not seem to be specific to social cognitive impairments, but our results support the hypothesis of a link between pro- and anti-inflammatory markers and cognition in SSD. In the future, other studies should be conducted to clarify this link from a diagnostic and therapeutic perspective: identification of inflammatory trait factors and patient subgroups and personalized anti-inflammatory therapies.
Collapse
Affiliation(s)
- Alexandre Carpentier
- Service Pathologies Résistantes (SPR), Pôle Ressource Évaluation en Réhabilitation PsychoSociale (PRERPS)Centre Hospitalier Isarien––EPSM de l'OiseClermont de l'OiseFrance
- Université Picardie Jules VerneAmiensFrance
- Institut de Psychiatrie (CNRS GDR 3557)ParisFrance
- INSERM UMR 1247––Groupe de Recherche sur l'Alcool et les Pharmacodépendances (GRAP)Université de Picardie Jules Verne, Centre Universitaire de Recherche en SantéAmiensFrance
| | - Dimitrios Zampetas
- Service Pathologies Résistantes (SPR), Pôle Ressource Évaluation en Réhabilitation PsychoSociale (PRERPS)Centre Hospitalier Isarien––EPSM de l'OiseClermont de l'OiseFrance
- Université Picardie Jules VerneAmiensFrance
| | - Alexandre Durand
- Service Pathologies Résistantes (SPR), Pôle Ressource Évaluation en Réhabilitation PsychoSociale (PRERPS)Centre Hospitalier Isarien––EPSM de l'OiseClermont de l'OiseFrance
- Université Picardie Jules VerneAmiensFrance
- INSERM UMR 1247––Groupe de Recherche sur l'Alcool et les Pharmacodépendances (GRAP)Université de Picardie Jules Verne, Centre Universitaire de Recherche en SantéAmiensFrance
| | - Mickael Naassila
- Université Picardie Jules VerneAmiensFrance
- Institut de Psychiatrie (CNRS GDR 3557)ParisFrance
- INSERM UMR 1247––Groupe de Recherche sur l'Alcool et les Pharmacodépendances (GRAP)Université de Picardie Jules Verne, Centre Universitaire de Recherche en SantéAmiensFrance
| | - Marie‐Cécile Bralet
- Institut de Psychiatrie (CNRS GDR 3557)ParisFrance
- INSERM UMR 1247––Groupe de Recherche sur l'Alcool et les Pharmacodépendances (GRAP)Université de Picardie Jules Verne, Centre Universitaire de Recherche en SantéAmiensFrance
- Service CRISALID‐HDF, Centre support de réhabilitation psychosociale et de remédiation cognitive, PRERPSCentre Hospitalier Isarien––EPSM de l'OiseClermont de l'OiseFrance
| |
Collapse
|
7
|
Delorme TC, Arcego DM, Penichet D, O'Toole N, Huebener N, Silveira PP, Srivastava LK, Cermakian N. Large-scale effects of prenatal inflammation and early life circadian disruption in mice: Implications for neurodevelopmental disorders. Brain Behav Immun 2025; 127:409-422. [PMID: 40118225 DOI: 10.1016/j.bbi.2025.03.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 02/17/2025] [Accepted: 03/17/2025] [Indexed: 03/23/2025] Open
Abstract
Around 80 % of individuals with neurodevelopmental disorders such as schizophrenia and autism spectrum disorders experience disruptions in sleep/circadian rhythms. We explored whether environmental circadian disruption interacts with prenatal infection, a risk factor for neurodevelopmental disorders, to induce sex-specific deficits in mice. A maternal immune activation (MIA) protocol was used by injecting pregnant mice with viral mimic poly IC or saline at E9.5. Juvenile/adolescent male and female offspring (3-7 weeks old) were then subjected to a standard light:dark cycle (12:12LD) or to constant light (LL). Significant interactions between treatment (MIA, control) and lighting (12:12LD, LL) were evident in behaviors related to cognition, anxiety, and sociability. This pattern persisted in our RNA sequencing analysis of the dorsal hippocampus, where poly IC exposure resulted in numerous differentially expressed genes (DEGs) in males, while exposure to both poly IC and LL led to a marked reduction in DEGs. Through WGCNA analysis, many significant gene modules were found to be positively associated with poly IC (vs. saline) and LL (vs. LD) in males (fewer in females). Many of the identified hub-bottleneck genes were homologous to human genes associated with sleep/circadian rhythms and neurodevelopmental disorders as revealed by GWA studies. The MIA- and LL-associated modules were enriched in microglia gene signatures, which was paralleled by trends of effects of each of the factors on microglia morphology. In conclusion, in a mouse model of prenatal infection, circadian disruption induced by LL during adolescence acts as a modulator of the effects of MIA at behavioral, cellular, and molecular levels.
Collapse
Affiliation(s)
- Tara C Delorme
- Douglas Mental Health University Institute, Montréal, Québec H4H 1R3, Canada; Integrated Program in Neuroscience, McGill University, Montréal, Québec H3A 2B4, Canada
| | - Danusa M Arcego
- Douglas Mental Health University Institute, Montréal, Québec H4H 1R3, Canada; Department of Psychiatry, McGill University, Montréal, Québec H3A 1A1, Canada
| | - Danae Penichet
- Douglas Mental Health University Institute, Montréal, Québec H4H 1R3, Canada; Integrated Program in Neuroscience, McGill University, Montréal, Québec H3A 2B4, Canada
| | - Nicholas O'Toole
- Douglas Mental Health University Institute, Montréal, Québec H4H 1R3, Canada
| | - Nikki Huebener
- Douglas Mental Health University Institute, Montréal, Québec H4H 1R3, Canada
| | - Patrícia P Silveira
- Douglas Mental Health University Institute, Montréal, Québec H4H 1R3, Canada; Department of Psychiatry, McGill University, Montréal, Québec H3A 1A1, Canada
| | - Lalit K Srivastava
- Douglas Mental Health University Institute, Montréal, Québec H4H 1R3, Canada; Department of Psychiatry, McGill University, Montréal, Québec H3A 1A1, Canada.
| | - Nicolas Cermakian
- Douglas Mental Health University Institute, Montréal, Québec H4H 1R3, Canada; Department of Psychiatry, McGill University, Montréal, Québec H3A 1A1, Canada.
| |
Collapse
|
8
|
Faustmann TJ, Corvace F, Faustmann PM, Ismail FS. Influence of antipsychotic drugs on microglia-mediated neuroinflammation in schizophrenia: perspectives in an astrocyte-microglia co-culture model. Front Psychiatry 2025; 16:1522128. [PMID: 40171306 PMCID: PMC11959008 DOI: 10.3389/fpsyt.2025.1522128] [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: 11/04/2024] [Accepted: 02/12/2025] [Indexed: 04/03/2025] Open
Abstract
Schizophrenia is a severe mental disorder with a strong lifetime impact on patients' health and wellbeing. Usually, symptomatic treatment includes typical or atypical antipsychotics. Study findings show an involvement of low-grade inflammation (blood, brain parenchyma, and cerebrospinal fluid) in schizophrenia. Moreover, experimental and neuropathological evidence suggests that reactive microglia, which are the main resident immune cells of the central nervous system (CNS), have a negative impact on the differentiation and function of oligodendrocytes, glial progenitor cells, and astrocytes, which results in the disruption of neuronal networks and dysregulated synaptic transmission, contributing to the pathophysiology of schizophrenia. Here, the role of microglial cells related to neuroinflammation in schizophrenia was discussed to be essential. This review aims to summarize the evidence for the influence of antipsychotics on microglial inflammatory mechanisms in schizophrenia. Furthermore, we propose an established astrocyte-microglia co-culture model for testing regulatory mechanisms and examining the effects of antipsychotics on glia-mediated neuroinflammation. This could lead to a better understanding of how typical and atypical antipsychotics can be used to address positive and negative symptoms in schizophrenia and comorbidities like inflammatory diseases or the status of low-grade inflammation.
Collapse
Affiliation(s)
- Timo Jendrik Faustmann
- Department of Psychiatry and Psychotherapy, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Franco Corvace
- Department of Neuroanatomy and Molecular Brain Research, Medical Faculty, Ruhr University Bochum, Bochum, Germany
| | - Pedro M. Faustmann
- Department of Neuroanatomy and Molecular Brain Research, Medical Faculty, Ruhr University Bochum, Bochum, Germany
| | - Fatme Seval Ismail
- Department of Neurology, Klinikum Vest, Academic Teaching Hospital of the Ruhr University Bochum, Recklinghausen, Germany
| |
Collapse
|
9
|
Hojjati SH, Chen K, Chiang GC, Kuceyeski A, Wang XH, Razlighi QR, Pahlajani S, Glodzik L, Tanzi EB, Reinhardt M, Butler TA. Utilizing structural MRI and unsupervised clustering to differentiate schizophrenia and Alzheimer's disease in late-onset psychosis. Behav Brain Res 2025; 480:115386. [PMID: 39644998 PMCID: PMC12040479 DOI: 10.1016/j.bbr.2024.115386] [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: 09/30/2024] [Revised: 11/14/2024] [Accepted: 12/04/2024] [Indexed: 12/09/2024]
Abstract
Late-onset psychosis (LOP) represents a highly heterogeneous and understudied condition, with potential origins ranging from atypically late onset of schizophrenia (SCZ) to Alzheimer's Disease (AD). Despite the clinical necessity of differentiating these conditions to guide effective treatment, achieving an accurate diagnosis remains challenging. This study aimed to utilize data-driven analyses of structural magnetic resonance imaging (MRI) to distinguish between these diagnostic possibilities. Utilizing publicly available datasets of MRI scans from 699 healthy control (HC) participants and 469 patients diagnosed with SCZ or AD, our analysis focused on bilateral subcortical volumetric measures in the caudate, hippocampus, putamen, and amygdala. We first trained an unsupervised K-means clustering algorithm based on SCZ and AD patients and achieved a clustering accuracy of 81 % and an area under curvature (AUC) of 0.79 in distinguishing between these two groups. Subsequently, we calculated the Euclidean distance between the AD and SCZ cluster centroids for each of ten patients with unexplained onset of psychosis after age 45 from a clinical MRI registry. Six patients were classified as AD and four as SCZ. Our findings revealed that among LOP participants, those classified in the SCZ cluster exhibited significantly greater right putamen volumes compared to those in the AD cluster (p < 0.0025). There were also intriguing clinical differences. While we do not have diagnostic biomarker information to confirm these classifications, this study sheds light on the heterogeneity of psychoses in late life and illustrates the potential use of widely available structural MRI and data-driven methods to enhance diagnostic accuracy and treatment outcomes for LOP patients.
Collapse
Affiliation(s)
- Seyed Hani Hojjati
- Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, New York, NY, USA.
| | - Kewei Chen
- College of Health Solutions, Arizona State University, Phoenix, AZ, USA; School of Mathematics and Statistics, Arizona State University, Phoenix, AZ, USA; Department of Neurology, University of Arizona College of Medicine, Phoenix, AZ, USA; Banner Alzheimer's Institute, Phoenix, AZ, USA
| | - Gloria C Chiang
- Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, New York, NY, USA
| | - Amy Kuceyeski
- Department of Computational Biology, Cornell University, Ithaca, NY, USA; Department of Psychiatry, Brain Health Institute, Rutgers University, Piscataway, NJ, USA
| | - Xiuyuan H Wang
- Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, New York, NY, USA
| | - Qolamreza R Razlighi
- Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, New York, NY, USA
| | - Silky Pahlajani
- Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, New York, NY, USA
| | - Lidia Glodzik
- Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, New York, NY, USA
| | - Emily B Tanzi
- Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, New York, NY, USA
| | - Michael Reinhardt
- Department of Psychiatry, SUNY Downstate Medical Center, Brooklyn, NY, USA
| | - Tracy A Butler
- Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
10
|
Ji L, Huang B, Ren D, Wei X, Liu L, Bi Y, Li Z, Yuan F, Han K, Li K, Yang F, Li X, Yu T, Shi Y, He L, Lu Q, He G. Identification of Schizophrenia-Risk Regulatory Variant rs1399178 in the Non-coding Region With Its Impact on NRF1 Binding. CNS Neurosci Ther 2025; 31:e70275. [PMID: 40019038 PMCID: PMC11868986 DOI: 10.1111/cns.70275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/28/2024] [Accepted: 01/31/2025] [Indexed: 03/01/2025] Open
Abstract
AIMS The challenges in identifying functional variants from genome-wide association studies (GWAS) and unraveling regulatory mechanisms in schizophrenia research persist, particularly in intronic regions. A non-coding regulatory variant, rs1399178, associated with schizophrenia risk, is identified and its impact on NRF1 binding is investigated. METHODS This study focuses on schizophrenia GWAS risk loci, using functional genomics, expression analyses and structural analysis to identify 736 schizophrenia risk single-nucleotide polymorphisms (SNPs) that disrupt transcription factor (TF) binding. RESULTS Among these SNPs, rs1399178 stands out as a bifunctional intergenic SNP that can switch between acting as a promoter and an enhancer, potentially influencing MLH1 and LRRFIP2 expression via expression quantitative trait loci analysis. Importantly, mutation of the G allele of rs1399178 to A significantly diminishes its binding affinity to nuclear respiratory factor 1 (NRF1). Structural analysis provides further insight into this alteration in the protein-nucleic acid complex formation. CONCLUSION Based on our data, a model is proposed in which rs1399178 confers schizophrenia risk by modifying NRF1 binding profiles, thereby regulating the abundance of target genes through promoter-enhancer switching. This study provides novel insights into the regulatory mechanisms of schizophrenia risk variants, highlighting the intricate nature of genetic interactions and potential therapeutic targets.
Collapse
Affiliation(s)
- Lei Ji
- Bio‐X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric DisordersShanghai Jiao Tong UniversityShanghaiChina
- The Collaborative Innovation Center for Brain Science, and Brain Science and Technology Research CenterShanghai Jiao Tong UniversityShanghaiChina
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health CenterShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Bojian Huang
- Bio‐X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric DisordersShanghai Jiao Tong UniversityShanghaiChina
- The Collaborative Innovation Center for Brain Science, and Brain Science and Technology Research CenterShanghai Jiao Tong UniversityShanghaiChina
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health CenterShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Decheng Ren
- Bio‐X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric DisordersShanghai Jiao Tong UniversityShanghaiChina
- The Collaborative Innovation Center for Brain Science, and Brain Science and Technology Research CenterShanghai Jiao Tong UniversityShanghaiChina
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health CenterShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xiaoxi Wei
- Bio‐X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric DisordersShanghai Jiao Tong UniversityShanghaiChina
- The Collaborative Innovation Center for Brain Science, and Brain Science and Technology Research CenterShanghai Jiao Tong UniversityShanghaiChina
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health CenterShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Liangjie Liu
- Bio‐X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric DisordersShanghai Jiao Tong UniversityShanghaiChina
- The Collaborative Innovation Center for Brain Science, and Brain Science and Technology Research CenterShanghai Jiao Tong UniversityShanghaiChina
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health CenterShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yan Bi
- Bio‐X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric DisordersShanghai Jiao Tong UniversityShanghaiChina
- The Collaborative Innovation Center for Brain Science, and Brain Science and Technology Research CenterShanghai Jiao Tong UniversityShanghaiChina
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health CenterShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Zhiqiang Li
- Bio‐X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric DisordersShanghai Jiao Tong UniversityShanghaiChina
- The Collaborative Innovation Center for Brain Science, and Brain Science and Technology Research CenterShanghai Jiao Tong UniversityShanghaiChina
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health CenterShanghai Jiao Tong University School of MedicineShanghaiChina
- Affiliated Hospital of Qingdao University and Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio‐X Institutes)Qingdao UniversityQingdaoChina
| | - Fan Yuan
- Bio‐X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric DisordersShanghai Jiao Tong UniversityShanghaiChina
- The Collaborative Innovation Center for Brain Science, and Brain Science and Technology Research CenterShanghai Jiao Tong UniversityShanghaiChina
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health CenterShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Ke Han
- Bio‐X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric DisordersShanghai Jiao Tong UniversityShanghaiChina
- The Collaborative Innovation Center for Brain Science, and Brain Science and Technology Research CenterShanghai Jiao Tong UniversityShanghaiChina
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health CenterShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Keyi Li
- Bio‐X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric DisordersShanghai Jiao Tong UniversityShanghaiChina
- The Collaborative Innovation Center for Brain Science, and Brain Science and Technology Research CenterShanghai Jiao Tong UniversityShanghaiChina
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health CenterShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Fengping Yang
- Bio‐X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric DisordersShanghai Jiao Tong UniversityShanghaiChina
- The Collaborative Innovation Center for Brain Science, and Brain Science and Technology Research CenterShanghai Jiao Tong UniversityShanghaiChina
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health CenterShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xingwang Li
- Bio‐X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric DisordersShanghai Jiao Tong UniversityShanghaiChina
- The Collaborative Innovation Center for Brain Science, and Brain Science and Technology Research CenterShanghai Jiao Tong UniversityShanghaiChina
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health CenterShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Tao Yu
- Bio‐X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric DisordersShanghai Jiao Tong UniversityShanghaiChina
- The Collaborative Innovation Center for Brain Science, and Brain Science and Technology Research CenterShanghai Jiao Tong UniversityShanghaiChina
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health CenterShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yi Shi
- Bio‐X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric DisordersShanghai Jiao Tong UniversityShanghaiChina
- The Collaborative Innovation Center for Brain Science, and Brain Science and Technology Research CenterShanghai Jiao Tong UniversityShanghaiChina
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health CenterShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Lin He
- Bio‐X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric DisordersShanghai Jiao Tong UniversityShanghaiChina
- The Collaborative Innovation Center for Brain Science, and Brain Science and Technology Research CenterShanghai Jiao Tong UniversityShanghaiChina
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health CenterShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Qing Lu
- Bio‐X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric DisordersShanghai Jiao Tong UniversityShanghaiChina
- Department of Otorhinolaryngology‐Head and Neck SurgeryChongqing General HospitalChongqingChina
- Ear InstituteShanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose DiseasesShanghaiChina
| | - Guang He
- Bio‐X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric DisordersShanghai Jiao Tong UniversityShanghaiChina
- The Collaborative Innovation Center for Brain Science, and Brain Science and Technology Research CenterShanghai Jiao Tong UniversityShanghaiChina
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health CenterShanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
11
|
Siafakas N, Anastassopoulou C, Pournaras S, Tsakris A, Alevizakis E, Kympouropoulos S, Spandidos DA, Rizos E. Viruses and psychiatric disorders: We have not crossed the borderline from hypothesis to proof yet (Review). Mol Med Rep 2025; 31:61. [PMID: 39749697 PMCID: PMC11711936 DOI: 10.3892/mmr.2024.13426] [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: 10/06/2023] [Accepted: 12/10/2024] [Indexed: 01/04/2025] Open
Abstract
Most psychiatric disorders are heterogeneous and are attributed to the synergistic action of a multitude of factors. It is generally accepted that psychiatric disorders are the outcome of interactions between genetic predisposition and environmental perturbations, which involve psychosocial stress, or alterations in the physiological state of the organism. A number of hypotheses have been presented on such environmental influences that may include direct insults such as injury, malnutrition and hostile living conditions, or indirect sequelae following infection from viruses such as influenza, arboviruses, enteroviruses and several herpesviruses, or the differential expression of human endogenous retroviruses. It is known that the concept of viruses is far more extensive than their perception as mere agents of acute infections, or chronic debilitating diseases, such as AIDS or some forms of cancer. Notably, an apparent causal connection between viruses and the pathophysiology of diseases has been suggested; however, it remains unclear as to how to establish this causal connection. There are inherent difficulties in answering this question with certainty, which may be due to the multitude of genetic and environmental influences that can lead to psychopathology; the latent state of chronic infection exhibited by a number of neurotropic viruses; the late onset of psychiatric disorders with respect to the acute phase of viral infection at which detection tests would be successful; the complexity of the virome; and the existence of thousands of viral species. The present review aims to provide an outline of the conclusions that have thus far been reached regarding a possible association between viral infection and psychiatric disease, and the obstacles confronted during the quest for the truth behind the role of viruses.
Collapse
Affiliation(s)
- Nikolaos Siafakas
- Clinical Microbiology Laboratory, National and Kapodistrian University of Athens, Attikon University Hospital, 12462 Athens, Greece
| | - Cleo Anastassopoulou
- Laboratory of Microbiology, National and Kapodistrian University of Athens, Medical School, 11527 Athens, Greece
| | - Spyridon Pournaras
- Clinical Microbiology Laboratory, National and Kapodistrian University of Athens, Attikon University Hospital, 12462 Athens, Greece
| | - Athanasios Tsakris
- Laboratory of Microbiology, National and Kapodistrian University of Athens, Medical School, 11527 Athens, Greece
| | - Evangelos Alevizakis
- 2nd Department of Psychiatry, National and Kapodistrian University of Athens, Attikon University Hospital, 12462 Athens, Greece
| | - Stylianos Kympouropoulos
- 2nd Department of Psychiatry, National and Kapodistrian University of Athens, Attikon University Hospital, 12462 Athens, Greece
| | - Demetrios A. Spandidos
- Laboratory of Clinical Virology, Medical School, University of Crete, 71003 Heraklion, Greece
| | - Emmanouil Rizos
- 2nd Department of Psychiatry, National and Kapodistrian University of Athens, Attikon University Hospital, 12462 Athens, Greece
| |
Collapse
|
12
|
Hall MB, Lemanski EA, Schwarz JM. Prenatal Maternal Immune Activation with Lipopolysaccharide Accelerates the Developmental Acquisition of Neonatal Reflexes in Rat Offspring Without Affecting Maternal Care Behaviors. Biomolecules 2025; 15:347. [PMID: 40149883 PMCID: PMC11940702 DOI: 10.3390/biom15030347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Revised: 02/18/2025] [Accepted: 02/24/2025] [Indexed: 03/29/2025] Open
Abstract
Maternal immune activation (MIA)-infection with an immunogen during pregnancy-is linked to an increased risk of neurodevelopmental disorders (NDDs) in offspring. Both MIA and NDDs are associated with developmental delays in offsprings' motor behavior. Therefore, the current study examined the effects of MIA on neonatal reflex development in male and female offspring. Sprague Dawley rats were administered lipopolysaccharide (LPS; 50 μg/mL/kg, i.p.) or saline on embryonic day (E)15 of gestation. The offspring were then tested daily from postnatal day (P)3-P21 to determine their neonatal reflex abilities. The maternal care behaviors of the dam were also quantified on P1-P5, P10, and P15. We found that, regardless of sex, the E15 LPS offspring were able to forelimb grasp, cliff avoid, and right with a correct posture at an earlier postnatal age than the E15 saline offspring did. The E15 LPS offspring also showed better performance of forelimb grasping, hindlimb grasping, righting with correct posture, and walking with correct posture than the E15 saline offspring did. There were no significant differences in maternal licking/grooming, arched-back nursing, non-arched-back nursing, or total nursing across the E15 groups. Overall, these findings suggest that MIA with LPS on E15 accelerates reflex development in offspring without affecting maternal care. This may be explained by the stress acceleration hypothesis, whereby early-life stress accelerates development to promote survival.
Collapse
Affiliation(s)
- Mary Beth Hall
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE 19716, USA;
- Interdisciplinary Neuroscience Graduate Program, University of Delaware, Newark, DE 19716, USA
| | - Elise A. Lemanski
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE 19716, USA;
- Interdisciplinary Neuroscience Graduate Program, University of Delaware, Newark, DE 19716, USA
| | - Jaclyn M. Schwarz
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE 19716, USA;
- Interdisciplinary Neuroscience Graduate Program, University of Delaware, Newark, DE 19716, USA
| |
Collapse
|
13
|
Joo SW, Lee J, Han J, Kim M, Kim Y, Lee H, Jo YT, Shin J, Lee J, Lee WH. Disparities in accelerated brain aging in recent-onset and chronic schizophrenia. Psychol Med 2025; 55:e60. [PMID: 39988480 PMCID: PMC12080660 DOI: 10.1017/s0033291725000285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/19/2025] [Accepted: 01/27/2025] [Indexed: 02/25/2025]
Abstract
BACKGROUND Patients with schizophrenia experience accelerated aging, accompanied by abnormalities in biomarkers such as shorter telomere length. Brain age prediction using neuroimaging data has gained attention in schizophrenia research, with consistently reported increases in brain-predicted age difference (brain-PAD). However, its associations with clinical symptoms and illness duration remain unclear. METHODS We developed brain age prediction models using structural magnetic resonance imaging (MRI) data from 10,938 healthy individuals. The models were validated on an independent test dataset comprising 79 healthy controls, 57 patients with recent-onset schizophrenia, and 71 patients with chronic schizophrenia. Group comparisons and the clinical associations of brain-PAD were analyzed using multiple linear regression. SHapley Additive exPlanations (SHAP) values estimated feature contributions to the model, and between-group differences in SHAP values and group-by-SHAP value interactions were also examined. RESULTS Patients with recent-onset schizophrenia and chronic schizophrenia exhibited increased brain-PAD values of 1.2 and 0.9 years, respectively. Between-group differences in SHAP values were identified in the right lateral prefrontal area (false discovery rate [FDR] p = 0.022), with group-by-SHAP value interactions observed in the left prefrontal area (FDR p = 0.049). A negative association between brain-PAD and Full-scale Intelligence Quotient scores in chronic schizophrenia was noted, which did not remain significant after correction for multiple comparisons. CONCLUSIONS Brain-PAD increases were pronounced in the early phase of schizophrenia. Regional brain abnormalities contributing to brain-PAD likely vary with illness duration. Future longitudinal studies are required to overcome limitations related to sample size, heterogeneity, and the cross-sectional design of this study.
Collapse
Affiliation(s)
- Sung Woo Joo
- Department of Psychiatry, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Junhyeok Lee
- Department of Software Convergence, Kyung Hee University, Yongin, Republic of Korea
| | - Juhyuk Han
- Department of Software Convergence, Kyung Hee University, Yongin, Republic of Korea
| | - Minjae Kim
- Department of Software Convergence, Kyung Hee University, Yongin, Republic of Korea
| | - Yeonwoo Kim
- Department of Software Convergence, Kyung Hee University, Yongin, Republic of Korea
| | - Howook Lee
- Department of Artificial Intelligence, Kyung Hee University, Yongin, Republic of Korea
| | - Young Tak Jo
- Department of Psychiatry, Kangdong Sacred Heart Hospital, Seoul, Republic of Korea
| | - Jaewook Shin
- Department of Medicine, CHA University School of Medicine, Seongnam, Republic of Korea
| | - Jungsun Lee
- Department of Psychiatry, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Won Hee Lee
- Department of Software Convergence, Kyung Hee University, Yongin, Republic of Korea
- Department of Artificial Intelligence, Kyung Hee University, Yongin, Republic of Korea
| |
Collapse
|
14
|
Zhen Y, Yang Y, Zheng Y, Zheng Z, Zheng H, Tang S. Aberrant Modular Dynamics of Functional Networks in Schizophrenia and Their Relationship with Neurotransmitter and Gene Expression Profiles. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.25.634845. [PMID: 39974915 PMCID: PMC11838238 DOI: 10.1101/2025.01.25.634845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Introduction Numerous studies have emphasized the time-varying modular architecture of functional brain networks and its relevance to cognitive functions in healthy participants. However, how brain modular dynamics change in schizophrenia and how these alterations relate to neurotransmitter and transcriptomic signatures have not been well elucidated. Methods We harmonized resting-state fMRI data from a multi-site sample including 223 patients and 279 healthy controls and applied the multilayer network method to estimate the regional module switching rate (flexibility) of functional brain connectomes. We examined aberrant flexibility in patients relative to controls and explored its relations to neurotransmitter systems and postmortem gene expression. Results Compared with controls, patients with schizophrenia had significantly higher flexibility in the somatomotor and right visual regions, and lower flexibility in the left parahippocampal gyrus, right supramarginal gyrus, right frontal-operculum-insula, bilateral precuneus posterior cingulate cortex, and bilateral inferior parietal gyrus. These alterations were associated with multiple neurotransmitter systems and weighted gene transcriptomic profiles. The most relevant genes were preferentially enriched for biological processes of transmembrane transport and brain development, specific cell types, and previously identified schizophrenia-related genes. Conclusions This study reveals aberrant modular dynamics in schizophrenia and its relations to neurotransmitter systems and schizophrenia-related transcriptomic profiles, providing insights into the understanding of the pathophysiology underlying schizophrenia.
Collapse
Affiliation(s)
- Yi Zhen
- School of Mathematical Sciences, Beihang University, Beijing 100191, China
- Key laboratory of Mathematics, Informatics and Behavioral Semantics, Beihang University, Beijing 100191, China
| | - Yaqian Yang
- Institute of Artificial Intelligence, Beihang University, Beijing 100191, China
- Key laboratory of Mathematics, Informatics and Behavioral Semantics, Beihang University, Beijing 100191, China
| | - Yi Zheng
- School of Mathematical Sciences, Beihang University, Beijing 100191, China
- Key laboratory of Mathematics, Informatics and Behavioral Semantics, Beihang University, Beijing 100191, China
| | - Zhiming Zheng
- Institute of Artificial Intelligence, Beihang University, Beijing 100191, China
- Key laboratory of Mathematics, Informatics and Behavioral Semantics, Beihang University, Beijing 100191, China
- Institute of Medical Artificial Intelligence, Binzhou Medical University, Yantai 264003, China
- Zhongguancun Laboratory, Beijing 100094, China
- Beijing Advanced Innovation Center for Future Blockchain and Privacy Computing, Beihang University, Beijing 100191, China
- State Key Laboratory of Complex & Critical Software Environment, Beihang University, Beijing 100191, China
| | - Hongwei Zheng
- Beijing Academy of Blockchain and Edge Computing, Beijing 100085, China
| | - Shaoting Tang
- Institute of Artificial Intelligence, Beihang University, Beijing 100191, China
- Key laboratory of Mathematics, Informatics and Behavioral Semantics, Beihang University, Beijing 100191, China
- Institute of Medical Artificial Intelligence, Binzhou Medical University, Yantai 264003, China
- Zhongguancun Laboratory, Beijing 100094, China
- Beijing Advanced Innovation Center for Future Blockchain and Privacy Computing, Beihang University, Beijing 100191, China
- State Key Laboratory of Complex & Critical Software Environment, Beihang University, Beijing 100191, China
| |
Collapse
|
15
|
Spoorthy MS, Tikka SK, Malathesh BC, Zaki SA. Rare Presentation of Schizophrenia in a Patient with Congenital Right Lung Agenesis: A Case Report. Indian J Psychol Med 2025; 47:93-95. [PMID: 39564253 PMCID: PMC11572402 DOI: 10.1177/02537176241245794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2024] Open
Affiliation(s)
- Mamidipalli Sai Spoorthy
- Dept. of Psychiatry, All India Institute of Medical Sciences Bibinagar (Hyderabad Metropolitan Region), Bibinagar, Telangana, India
| | - Sai Krishna Tikka
- Dept. of Psychiatry, All India Institute of Medical Sciences Bibinagar (Hyderabad Metropolitan Region), Bibinagar, Telangana, India
| | - Barikar C. Malathesh
- Dept. of Psychiatry, All India Institute of Medical Sciences Bibinagar (Hyderabad Metropolitan Region), Bibinagar, Telangana, India
| | - Syed Ahmed Zaki
- Dept. of Pediatrics, All India Institute of Medical Sciences, Bibinagar (Hyderabad Metropolitan Region), Bibinagar, Telangana, India
| |
Collapse
|
16
|
Yoshida Y, Shinomiya A, Oikawa M, Shimada T, Hanaki KI, Watanabe Y. [Neonatal Malnutrition Impacts Fibroblast Growth Factor 21-induced Neurite Outgrowth and Growth Hormone-releasing Hormone Secretion in Neonatal Mouse Brain]. YAKUGAKU ZASSHI 2025; 145:183-188. [PMID: 40024730 DOI: 10.1248/yakushi.24-00177-3] [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: 03/04/2025]
Abstract
Neonatal malnutrition has been suggested as a factor contributing to neurological and other disorders. However, the details of this mechanism remain unclear. We focused on fibroblast growth factor 21 (FGF21), an endocrine factor produced in the liver during lactation-the main source of nutrition during the neonatal period- and analyzed its role in the brain. From the RNA-seq analysis of mouse brains, we analyzed the genes whose expression was regulated by FGF21 and their respective functions. We found that FGF21 has two functions in the neonatal brain; FGF21 induces the production of growth hormone-releasing hormone (GHRH) in the hypothalamus and is involved in isoform determination of Kalirin, a Ras homologous guanine nucleotide exchange factor, and promotes neurite outgrowth in the brain. Furthermore, the above mechanism is regulated by SH2-containing tyrosine phosphatase (SHP2) activity downstream of the FGF receptor. Additionally, the conserved intron of the SHP2 gene, Ptpn11, shows altered activity in malnourished mouse brains. In summary, FGF21 functions in neurite outgrowth and GHRH production in the neonatal mouse brain, with the mechanism being regulated by SHP2. However, SHP2 activity depends on nutritional status. Our goal was to elucidate the mechanisms by which FGF21 is involved in the maintenance of the central nervous system during the neonatal period. This study provides new insights into the role of FGF21 in diseases caused by dysfunction due to malnutrition.
Collapse
Affiliation(s)
- Yuko Yoshida
- Laboratory of Biosecurity Management, Research Center for Biosafety, Laboratory Animal and Pathogen Bank, NIID
| | | | | | | | - Ken-Ichi Hanaki
- Laboratory of Biosecurity Management, Research Center for Biosafety, Laboratory Animal and Pathogen Bank, NIID
| | - Yoshifumi Watanabe
- Faculty of Pharmacy, Musashino University
- Research Institute of Pharmaceutical Sciences, Musashino University
| |
Collapse
|
17
|
Carril Pardo C, Oyarce Merino K, Vera-Montecinos A. Neuroinflammatory Loop in Schizophrenia, Is There a Relationship with Symptoms or Cognition Decline? Int J Mol Sci 2025; 26:310. [PMID: 39796167 PMCID: PMC11720417 DOI: 10.3390/ijms26010310] [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: 12/05/2024] [Revised: 12/19/2024] [Accepted: 12/24/2024] [Indexed: 01/13/2025] Open
Abstract
Schizophrenia (SZ), a complex psychiatric disorder of neurodevelopment, is characterised by a range of symptoms, including hallucinations, delusions, social isolation and cognitive deterioration. One of the hypotheses that underlie SZ is related to inflammatory events which could be partly responsible for symptoms. However, it is unknown how inflammatory molecules can contribute to cognitive decline in SZ. This review summarises and exposes the possible contribution of the imbalance between pro-inflammatory and anti-inflammatory interleukins like IL-1beta, IL-4 and TNFalfa among others on cognitive impairment. We discuss how this inflammatory imbalance affects microglia and astrocytes inducing the disruption of the blood-brain barrier (BBB) in SZ, which could impact the prefrontal cortex or associative areas involved in executive functions such as planning and working tasks. We also highlight that inflammatory molecules generated by intestinal microbiota alterations, due to dysfunctional microbial colonisers or the use of some anti-psychotics, could impact the central nervous system. Finally, the question arises as to whether it is possible to modulate or correct the inflammatory imbalance that characterises SZ, and if an immunomodulatory strategy can be incorporated into conventional clinical treatments, either alone or in complement, to be applied in specific phases, such as prodromal or in the first-episode psychosis.
Collapse
Affiliation(s)
- Claudio Carril Pardo
- Laboratorio de Neuroinmunología, Facultad de Medicina y Ciencia, Universidad San Sebastián, Sede Tres Pascualas, Concepción 4080871, Chile; (C.C.P.)
| | - Karina Oyarce Merino
- Laboratorio de Neuroinmunología, Facultad de Medicina y Ciencia, Universidad San Sebastián, Sede Tres Pascualas, Concepción 4080871, Chile; (C.C.P.)
| | - América Vera-Montecinos
- Departamento de Ciencias Biológicas y Químicas, Facultad De Medicina y Ciencia, Universidad San Sebastián, Sede Tres Pascualas Lientur 1457, Concepción 4080871, Chile
| |
Collapse
|
18
|
Dongre N, Shattar C, Ramdurg S, Kadakol GS. Expression Profiling of Reelin (RELN) Gene in Patients With Schizophrenia from North Karnataka. Cureus 2025; 17:e76850. [PMID: 39897195 PMCID: PMC11787944 DOI: 10.7759/cureus.76850] [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: 10/22/2024] [Accepted: 01/03/2025] [Indexed: 02/04/2025] Open
Abstract
Background Schizophrenia is a devastating neuropsychiatric condition of uncertain etiology with significant adverse effects on the affected people, their families, and society. India has a heterogeneous population with a high degree of inbreeding. Hence, it is necessary to screen Indian patients with psychotic disorders to get a true picture of the contribution of Reelin (RELN) mRNA expression in schizophrenia. Mental illness is a leading cause of several metabolic changes and other related complications. It is not clear whether these metabolic changes are due to alterations in the RELN gene expression or because of antipsychotic drug use. Therefore, it is necessary to study the link between RELN gene expression and the metabolic syndrome. Aim and objective The present study aims to study the expression profiling of the RELN gene in patients with schizophrenia and the occurrence of metabolic syndrome in these patients. Methodology Patients with clinically diagnosed schizophrenia were studied for RELN gene expression and the RELN protein was quantified using real-time reverse transcriptase-polymerase chain reaction (RT-PCR). Biochemical parameters like serum random blood sugar (RBS) and lipid profile were analyzed by standard biochemical methods on the semi-auto analyzer and the lipid ratios in the study population were calculated and compared with the age and sex-matched controls. The statistical analyses were performed using IBM SPSS Statistics for Windows, Version 20 (Released 2011; IBM Corp., Armonk, New York, United States). A p-value of <0.05 was considered statistically significant. All statistical tests were two-tailed. Results The present study involved 108 subjects, out of which 54 were patients with schizophrenia (study group) and the remaining 54 were healthy controls (control group). In the former group, 29 were female (53.7%) and 25 were male (46.3%) patients whereas in the latter group, 27 were female (50%) and 27 were male (50%) individuals. Majority of the participants in the study group showed moderate scores on the Brief Psychiatric Rating (BPRS) scale. The levels of RELN mRNA expression were decreased in schizophrenia. Compared to the controls, the levels of RBS, total cholesterol, triglycerides (TGs), and low-density lipoprotein cholesterol (LDL-C) were significantly increased and levels of high-density lipoprotein cholesterol (HDL-C) were significantly decreased in the patients with schizophrenia. Body mass index (BMI), waist circumference (WC), and lipid ratios were also significantly greater in these patients. Conclusion An analysis of the candidate genes in psychotic disorders can be helpful in designing targeted drugs to treat these patients. Early and regular monitoring of patients on antipsychotic drug treatment is needed to find and prevent the risk of developing metabolic syndrome, which is the major cause for atherosclerotic cardiovascular diseases in this group. Appropriate lifestyle modifications and early intervention can help in preventing early deaths in patients with psychotic disorders.
Collapse
Affiliation(s)
- Nilima Dongre
- Biochemistry, BLDE (Deemed to be University) Shri B M Patil Medical College, Hospital and Research Centre, Vijayapura, IND
| | - Chetan Shattar
- Biochemistry, BLDE (Deemed to be University) Shri B M Patil Medical College, Hospital and Research Centre, Vijayapura, IND
| | - Santosh Ramdurg
- Psychiatry, BLDE (Deemed to be University) Shri B M Patil Medical College, Hospital and Research Centre, Vijayapura, IND
| | - Gurushantappa S Kadakol
- Anatomy, Human Genetics Laboratory, BLDE (Deemed to be University) Shri B M Patil Medical College, Hospital and Research Centre, Vijayapura, IND
| |
Collapse
|
19
|
Senko D, Efimova O, Osetrova M, Anikanov N, Boyko M, Sharaev M, Morozova A, Zorkina Y, Kislov M, Kostyuk G, Stekolshchikova E, Khaitovich P. White matter lipidome alterations in the schizophrenia brain. SCHIZOPHRENIA (HEIDELBERG, GERMANY) 2024; 10:123. [PMID: 39725684 DOI: 10.1038/s41537-024-00542-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 11/25/2024] [Indexed: 12/28/2024]
Abstract
Numerous brain imaging studies have reported white matter alterations in schizophrenia, but the lipidome analysis of the corresponding tissue remains incomplete. In this study, we investigated the lipidome composition of six subcortical white matter regions corresponding to major axonal tracks in both control subjects and schizophrenia patients. All six regions exhibited a consistent pattern of quantitative lipidome alterations in schizophrenia, involving myelin-forming and mitochondria associated lipid classes. While alteration levels of myelin-forming lipids, particularly sphingolipids, aligned with the extent of the myelin changes reported in structural brain imaging studies, a significant decrease of mitochondria in the white matter, indicated by the lipidome alterations, was not previously investigated. To verify this effect, we performed lipidome analysis in a larger set of individuals and in the mitochondria-enriched membrane fraction, as well as directly quantified mitochondrial content. Our results suggest a substantial reduction of the mitochondrial quotient accompanied by the imbalance in myelin lipids in schizophrenia white matter.
Collapse
Affiliation(s)
- Dmitry Senko
- Skolkovo Institute of Science and Technology, Moscow, Russia.
| | - Olga Efimova
- Skolkovo Institute of Science and Technology, Moscow, Russia
| | - Maria Osetrova
- Skolkovo Institute of Science and Technology, Moscow, Russia
- Mental Health Clinic No.1 Named After N. A. Alexeev of Moscow Healthcare Department, Moscow, Russia
| | | | - Maria Boyko
- Skolkovo Institute of Science and Technology, Moscow, Russia
- BIMAI-lab, Sharjah, UAE
| | - Maksim Sharaev
- Skolkovo Institute of Science and Technology, Moscow, Russia
- Mental Health Clinic No.1 Named After N. A. Alexeev of Moscow Healthcare Department, Moscow, Russia
- BIMAI-lab, Sharjah, UAE
| | - Anna Morozova
- Mental Health Clinic No.1 Named After N. A. Alexeev of Moscow Healthcare Department, Moscow, Russia
- Serbsky National Medical Research Centre of Psychiatry and Narcology, Moscow, Russia
| | - Yana Zorkina
- Mental Health Clinic No.1 Named After N. A. Alexeev of Moscow Healthcare Department, Moscow, Russia
- Serbsky National Medical Research Centre of Psychiatry and Narcology, Moscow, Russia
| | - Maksim Kislov
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - Georgiy Kostyuk
- Mental Health Clinic No.1 Named After N. A. Alexeev of Moscow Healthcare Department, Moscow, Russia
- Lomonosov Moscow State University, Moscow, Russia
| | | | | |
Collapse
|
20
|
Liang J, Guan X, Sun Q, Hao Y, Xiu M. Neutrophil/lymphocyte ratio and cognitive performances in first-episode patients with schizophrenia and healthy controls. Prog Neuropsychopharmacol Biol Psychiatry 2024; 135:111092. [PMID: 39029649 DOI: 10.1016/j.pnpbp.2024.111092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 06/20/2024] [Accepted: 07/16/2024] [Indexed: 07/21/2024]
Abstract
Abnormal immune and inflammatory responses are considered to contribute to schizophrenia (SZ). The neutrophil/lymphocyte ratio (NLR) is an inexpensive and reproducible marker of systemic inflammatory responses. Accumulating studies have demonstrated that NLR values are increased in SZ compared to healthy controls and closely related to clinical symptoms in antipsychotic-naïve first-episode SZ (ANFES) patients. However, to our knowledge, only one study has examined NLR in relation to neurocognition in 27 first-episode psychosis patients and 27 controls. This study aimed to examine the relationship of NLR values with cognitive performances in ANFES patients with a larger sample size. Whole blood cell counts were measured in ninety-seven ANFES patients and fifty-six control subjects. The neurocognitive functions of all subjects were measured by the repeatable battery for the assessment of neuropsychological status (RBANS). ANFES patients performed worse on cognition and had increased NLR values relative to healthy controls. In addition, increased NLR was negatively associated with cognitive functions in ANFES patients. Lymphocyte count was positively correlated with cognitive functions in patients. These findings suggest that the abnormal immune and inflammation system indicated by NLR may be involved in the cognitive functions in ANFES patients.
Collapse
Affiliation(s)
- Jing Liang
- Qingdao Mental Health Center, Qingdao, China
| | - Xiaoni Guan
- Peking University HuiLongGuan Clinical Medical School, Beijing HuiLongGuan Hospital, Beijing, China
| | | | - Yong Hao
- Department of Neurology, Renji Hospital, School of Medicine Shanghai Jiao Tong University, Shanghai, China.
| | - Meihong Xiu
- Peking University HuiLongGuan Clinical Medical School, Beijing HuiLongGuan Hospital, Beijing, China.
| |
Collapse
|
21
|
de Miranda AS, Macedo DS, Sanders LLO, Monte AS, Soares MVR, Teixeira AL. Unraveling the role of the renin-angiotensin system in severe mental illnesses: An insight into psychopathology and cognitive deficits. Cell Signal 2024; 124:111429. [PMID: 39306262 DOI: 10.1016/j.cellsig.2024.111429] [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: 07/06/2024] [Revised: 09/16/2024] [Accepted: 09/19/2024] [Indexed: 09/26/2024]
Abstract
Severe mental illnesses (SMI), especially schizophrenia and bipolar disorder (BD), are associated with significant distress to patients, reduced life expectancy and a higher cost of care. There is growing evidence that SMI may increase the risk of dementia in later life, posing an additional challenge in the management of these patients. SMI present a complex and highly heterogeneous pathophysiology, which has hampered the understanding of its underlying pathological mechanisms and limited the success of the available therapies. Despite the advances in therapeutic approaches in psychiatry over the past decades, treatment resistance is still a common problem in clinical practice, highlighting the urgent need for novel therapeutic targets for SMI. The discovery that renin-angiotensin system (RAS) components are expressed in the central nervous system opened new possibilities for investigating a potential role for this system in the neurobiology of SMI. The safety and efficacy of AT1 receptor blockers and angiotensin-converting enzyme inhibitors in cardiovascular and metabolic diseases, common medical comorbidities among SMI patients and well-known risk factors for dementia, suggest the potential scalability of these strategies for the management of SMI outcomes including the risk of subsequent dementia. This review aimed to discuss the available evidence from animal models and human studies of the potential involvement of RAS in the pathophysiology of SMI. We also provided a reflection on drawbacks and perspectives that can foster the development of new related therapeutic strategies.
Collapse
Affiliation(s)
- Aline Silva de Miranda
- Laboratory of Neurobiology, Department of Morphology, Institute of Biological Science, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil.
| | - Danielle S Macedo
- Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceara, CE, Fortaleza, Brazil
| | - Lia Lira O Sanders
- Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceara, CE, Fortaleza, Brazil; Centro Universitário Christus-Unichristus, Fortaleza, Brazil
| | - Aline S Monte
- Health Science Institute, University of International Integration of Afro-Brazilian Lusophony - UNILAB, Redenção, Brazil
| | - Michelle Verde Ramo Soares
- Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceara, CE, Fortaleza, Brazil
| | - Antonio Lucio Teixeira
- The Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, Lozano Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
| |
Collapse
|
22
|
Zhang QX, Wu SS, Wang PJ, Zhang R, Valenzuela RK, Shang SS, Wan T, Ma J. Schizophrenia-Like Deficits and Impaired Glutamate/Gamma-aminobutyric acid Homeostasis in Zfp804a Conditional Knockout Mice. Schizophr Bull 2024; 50:1411-1426. [PMID: 38988003 PMCID: PMC11548938 DOI: 10.1093/schbul/sbae120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/12/2024]
Abstract
BACKGROUND AND HYPOTHESIS Zinc finger protein 804A (ZNF804A) was the first genome-wide associated susceptibility gene for schizophrenia (SCZ) and played an essential role in the pathophysiology of SCZ by influencing neurodevelopment regulation, neurite outgrowth, synaptic plasticity, and RNA translational control; however, the exact molecular mechanism remains unclear. STUDY DESIGN A nervous-system-specific Zfp804a (ZNF804A murine gene) conditional knockout (cKO) mouse model was generated using clustered regularly interspaced short palindromic repeat/Cas9 technology and the Cre/loxP method. RESULTS Multiple and complex SCZ-like behaviors, such as anxiety, depression, and impaired cognition, were observed in Zfp804a cKO mice. Molecular biological methods and targeted metabolomics assay validated that Zfp804a cKO mice displayed altered SATB2 (a cortical superficial neuron marker) expression in the cortex; aberrant NeuN, cleaved caspase 3, and DLG4 (markers of mature neurons, apoptosis, and postsynapse, respectively) expressions in the hippocampus and a loss of glutamate (Glu)/γ-aminobutyric acid (GABA) homeostasis with abnormal GAD67 (Gad1) expression in the hippocampus. Clozapine partly ameliorated some SCZ-like behaviors, reversed the disequilibrium of the Glu/GABA ratio, and recovered the expression of GAD67 in cKO mice. CONCLUSIONS Zfp804a cKO mice reproducing SCZ-like pathological and behavioral phenotypes were successfully developed. A novel mechanism was determined in which Zfp804a caused Glu/GABA imbalance and reduced GAD67 expression, which was partly recovered by clozapine treatment. These findings underscore the role of altered gene expression in understanding the pathogenesis of SCZ and provide a reliable SCZ model for future therapeutic interventions and biomarker discovery.
Collapse
Affiliation(s)
- Qiao-xia Zhang
- Department of Electron Microscope, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
| | - Shan-shan Wu
- Department of Electron Microscope, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
| | - Peng-jie Wang
- Department of Electron Microscope, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
| | - Rui Zhang
- Department of Electron Microscope, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
- Department of Biochemistry and Molecular Biology, College of Medical Technology, Guiyang Healthcare Vocational University, Guiyang, Guizhou, China
| | - Robert K Valenzuela
- JAX Center for Alzheimer’s and Dementia Research, The Jackson Laboratory, Bar Harbor, ME, USA
| | - Shan-shan Shang
- Department of Electron Microscope, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
| | - Ting Wan
- Department of Electron Microscope, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
| | - Jie Ma
- Department of Electron Microscope, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
| |
Collapse
|
23
|
Choudhary A, Peles D, Nayak R, Mizrahi L, Stern S. Current progress in understanding schizophrenia using genomics and pluripotent stem cells: A meta-analytical overview. Schizophr Res 2024; 273:24-38. [PMID: 36443183 DOI: 10.1016/j.schres.2022.11.001] [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: 06/23/2022] [Revised: 10/16/2022] [Accepted: 11/01/2022] [Indexed: 11/27/2022]
Abstract
Schizophrenia (SCZ) is a complex, heritable and polygenic neuropsychiatric disease, which disables the patients as well as decreases their life expectancy and quality of life. Common and rare variants studies on SCZ subjects have provided >100 genomic loci that hold importance in the context of SCZ pathophysiology. Transcriptomic studies from clinical samples have informed about the differentially expressed genes (DEGs) and non-coding RNAs in SCZ patients. Despite these advancements, no causative genes for SCZ were found and hence SCZ is difficult to recapitulate in animal models. In the last decade, induced Pluripotent Stem Cells (iPSCs)-based models have helped in understanding the neural phenotypes of SCZ by studying patient iPSC-derived 2D neuronal cultures and 3D brain organoids. Here, we have aimed to provide a simplistic overview of the current progress and advancements after synthesizing the enormous literature on SCZ genetics and SCZ iPSC-based models. Although further understanding of SCZ genetics and pathophysiological mechanisms using these technological advancements is required, the recent approaches have allowed to delineate important cellular mechanisms and biological pathways affected in SCZ.
Collapse
Affiliation(s)
- Ashwani Choudhary
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel
| | - David Peles
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel
| | - Ritu Nayak
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel
| | - Liron Mizrahi
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel
| | - Shani Stern
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel.
| |
Collapse
|
24
|
Santarriaga S, Gerlovin K, Layadi Y, Karmacharya R. Human stem cell-based models to study synaptic dysfunction and cognition in schizophrenia: A narrative review. Schizophr Res 2024; 273:78-97. [PMID: 36925354 PMCID: PMC10500041 DOI: 10.1016/j.schres.2023.02.029] [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: 10/21/2022] [Revised: 02/27/2023] [Accepted: 02/28/2023] [Indexed: 03/18/2023]
Abstract
Cognitive impairment is the strongest predictor of functional outcomes in schizophrenia and is hypothesized to result from synaptic dysfunction. However, targeting synaptic plasticity and cognitive deficits in patients remains a significant clinical challenge. A comprehensive understanding of synaptic plasticity and the molecular basis of learning and memory in a disease context can provide specific targets for the development of novel therapeutics targeting cognitive impairments in schizophrenia. Here, we describe the role of synaptic plasticity in cognition, summarize evidence for synaptic dysfunction in schizophrenia and demonstrate the use of patient derived induced-pluripotent stem cells for studying synaptic plasticity in vitro. Lastly, we discuss current advances and future technologies for bridging basic science research of synaptic dysfunction with clinical and translational research that can be used to predict treatment response and develop novel therapeutics.
Collapse
Affiliation(s)
- Stephanie Santarriaga
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA; Chemical Biology and Therapeutic Science Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Kaia Gerlovin
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA; Chemical Biology and Therapeutic Science Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Yasmine Layadi
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA; Chimie ParisTech, Université Paris Sciences et Lettres, Paris, France
| | - Rakesh Karmacharya
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA; Chemical Biology and Therapeutic Science Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Psychiatry, Harvard Medical School, Boston, MA, USA; Schizophrenia and Bipolar Disorder Program, McLean Hospital, Belmont, MA, USA.
| |
Collapse
|
25
|
Keshavan MS, Song SH. Neuroscience in Pictures: 3. Schizophrenia. Asian J Psychiatr 2024; 102:104278. [PMID: 39427364 DOI: 10.1016/j.ajp.2024.104278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 10/13/2024] [Accepted: 10/14/2024] [Indexed: 10/22/2024]
Abstract
Schizophrenia is a complex, heritable brain disorder characterized by psychotic, negative, cognitive, mood, and motor symptoms. This pictorial review explores the multifaceted nature of schizophrenia, from its etiology to prevention strategies. We discuss the interplay of genetic and environmental risk factors, neurobiological underpinnings, and stepwise progression. Recent advances in understanding circuit-level pathophysiology and neurotransmitter systems beyond dopamine are highlighted along with neuropathological findings, particularly the exaggerated synaptic pruning hypothesis. Based on these developments, we present an updated perspective on pharmacological interventions. Finally, we outline preventative strategies across different stages, emphasizing early intervention. This overview, designed as a teaching resource, aims to provide trainees, clinicians and researchers with a current understanding of schizophrenia's neurobiological underpinnings and the implications of such understanding to the evolving landscape of its diagnosis and management.
Collapse
Affiliation(s)
- Matcheri S Keshavan
- Department of Psychiatry, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA.
| | - Seo Ho Song
- Department of Psychiatry, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
26
|
Benster LL, Stapper N, Rodriguez K, Daniels H, Villodas M, Weissman CR, Daskalakis ZJ, Appelbaum LG. Developmental Predictors of Suicidality in Schizophrenia: A Systematic Review. Brain Sci 2024; 14:995. [PMID: 39452009 PMCID: PMC11506348 DOI: 10.3390/brainsci14100995] [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: 09/14/2024] [Revised: 09/28/2024] [Accepted: 09/28/2024] [Indexed: 10/26/2024] Open
Abstract
Background/Objectives: Schizophrenia (SZ) is a severe psychiatric disorder characterized by a complex interplay of genetic, developmental, and environmental factors that significantly increase the risk of suicidal ideation (SI) and suicide attempts (SAs). This systematic review synthesizes current research on the developmental predictors of SI in individuals with SZ, aiming to delineate the multifactorial etiology of suicide within this population. Methods: A comprehensive search across Medline, PsycINFO, and EMBASE databases identified 23 eligible studies, emphasizing the varied methodological approaches and the global distribution of research efforts. Results: These studies demonstrate a robust association between early life adversities, particularly childhood trauma such as physical neglect, emotional abuse, and sexual abuse, and the increased prevalence of SI and SAs in SZ. This review also highlights the significant genetic factors associated with the development of suicidality in SZ, raising the possibility that polymorphisms in inflammation-related genes and neurodevelopmental abnormalities may influence susceptibility to SI. Notably, family history of psychiatric conditions may exacerbate the risk of SI through both hereditary and environmental mechanisms. Environmental factors, including socioeconomic status and social support, are also implicated, underscoring the role of broader socio-environmental conditions influencing outcomes. Conclusions: This review supports the integration of biopsychosocial models in understanding SI in SZ, advocating for interventions addressing the complex interplay of risk factors and the need for longitudinal studies to elucidate the dynamic interactions between risk factors over time. This comprehensive understanding is crucial for developing targeted preventive strategies and enhancing the clinical management of SZ, aiming to reduce suicidality in this vulnerable population.
Collapse
Affiliation(s)
- Lindsay L. Benster
- Department of Psychiatry, UC San Diego, La Jolla, CA 92093, USA; (L.L.B.); (N.S.); (K.R.); (H.D.); (C.R.W.); (Z.J.D.)
- Department of Clinical Psychology, San Diego State University, San Diego, CA 92182, USA;
| | - Noah Stapper
- Department of Psychiatry, UC San Diego, La Jolla, CA 92093, USA; (L.L.B.); (N.S.); (K.R.); (H.D.); (C.R.W.); (Z.J.D.)
- Department of Clinical Psychology, San Diego State University, San Diego, CA 92182, USA;
| | - Katie Rodriguez
- Department of Psychiatry, UC San Diego, La Jolla, CA 92093, USA; (L.L.B.); (N.S.); (K.R.); (H.D.); (C.R.W.); (Z.J.D.)
| | - Hadley Daniels
- Department of Psychiatry, UC San Diego, La Jolla, CA 92093, USA; (L.L.B.); (N.S.); (K.R.); (H.D.); (C.R.W.); (Z.J.D.)
| | - Miguel Villodas
- Department of Clinical Psychology, San Diego State University, San Diego, CA 92182, USA;
| | - Cory R. Weissman
- Department of Psychiatry, UC San Diego, La Jolla, CA 92093, USA; (L.L.B.); (N.S.); (K.R.); (H.D.); (C.R.W.); (Z.J.D.)
| | - Zafiris J. Daskalakis
- Department of Psychiatry, UC San Diego, La Jolla, CA 92093, USA; (L.L.B.); (N.S.); (K.R.); (H.D.); (C.R.W.); (Z.J.D.)
| | - Lawrence G. Appelbaum
- Department of Psychiatry, UC San Diego, La Jolla, CA 92093, USA; (L.L.B.); (N.S.); (K.R.); (H.D.); (C.R.W.); (Z.J.D.)
| |
Collapse
|
27
|
Guo Z, Su Y, Huang WK, Yao XS, Hong Y, Gordin A, Nguyen HN, Wen Z, Ringeling FR, Chen G, Li S, Lu L, Xia M, Zheng W, Sawa A, Chen G, Christian KM, Song H, Ming GL. GABAergic neuron dysregulation in a human neurodevelopmental model for major psychiatric disorders. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.23.614564. [PMID: 39372772 PMCID: PMC11451812 DOI: 10.1101/2024.09.23.614564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
"GABA dysfunction" is a major hypothesis for the biological basis of schizophrenia with indirect supporting evidence from human post-mortem brain and genetic studies. Patient-derived induced pluripotent stem cells (iPSCs) have emerged as a valuable platform for modeling psychiatric disorders, and previous modeling has revealed glutamatergic synapse deficits. Whether GABAergic synapse properties are affected in patient-derived human neurons and how this impacts neuronal network activity remain poorly understood. Here we optimized a protocol to differentiate iPSCs into highly enriched ganglionic eminence-like neural progenitors and GABAergic neurons. Using a collection of iPSCs derived from patients of psychiatric disorders carrying a Disrupted-in-Schizophrenia 1 ( DISC1 ) mutation and their unaffected family member, together with respective isogenic lines, we identified mutation-dependent deficits in GABAergic synapse formation and function, a phenotype similar to that of mutant glutamatergic neurons. However, mutant glutamatergic and GABAergic neurons contribute differentially to neuronal network excitability and synchrony deficits. Finally, we showed that GABAergic synaptic transmission is also defective in neurons derived from several idiopathic schizophrenia patient iPSCs. Transcriptome analysis further showed some shared gene expression dysregulation, which is more prominent in DISC1 mutant neurons. Together, our study supports a functional GABAergic synaptic deficit in major psychiatric disorders.
Collapse
|
28
|
Wang C, Zhang Y, Chong JS, Zhang W, Zhang X, McIntyre RS, Li Z, Ho RCM, Tang TB, Lim LG. Altered functional connectivity subserving expressed emotion environments in schizophrenia: An fNIRS study. Schizophr Res 2024; 270:178-187. [PMID: 38917555 DOI: 10.1016/j.schres.2024.06.019] [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: 12/20/2023] [Revised: 04/16/2024] [Accepted: 06/15/2024] [Indexed: 06/27/2024]
Abstract
Living in high-expressed emotion (EE) environments, characterized by critical, hostile, or over-involved family attitudes, has been linked to increased relapse rates among individuals with schizophrenia (SZ). In our previous work (Wang et al., 2023), we conducted the first feasibility study of using functional near-infrared spectroscopy (fNIRS) with our developed EE stimuli to examine cortical hemodynamics in SZ. To better understand the neural mechanisms underlying EE environmental factors in SZ, we extended our investigation by employing functional connectivity (FC) analysis with a graph theory approach to fNIRS signals. Relative to healthy controls (N=40), individuals with SZ (N=37) exhibited altered connectivity across the medial prefrontal cortex (mPFC), left ventrolateral prefrontal cortex (vlPFC), and left superior temporal gyrus (STG) while exposed to EE environments. Notably, while individuals with SZ were exposed to high-EE environments, (i) reduced connectivity was observed in these brain regions and (ii) the left vlPFC-STG coupling was found to be associated with the negative symptom severity. Taken together, our FC findings suggest individuals with SZ experience a more extensive disruption in neural functioning and coordination, particularly indicating an increased susceptibility to high-EE environments. This further supports the potential utility of integrating fNIRS with the created EE stimuli for assessing EE environmental influences, paving the way for more targeted therapeutic interventions.
Collapse
Affiliation(s)
| | | | - Jie Sheng Chong
- Centre for Intelligent Signal and Imaging Research (CISIR), Universiti Teknologi PETRONAS, Seri Iskandar 32610, Perak, Malaysia
| | | | - Xi Zhang
- Huaibei Mental Health Center, China
| | - Roger S McIntyre
- Mood Disorders Psychopharmacology Unit, Poul Hansen Family Centre for Depression, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, Canada; Department of Psychiatry, University of Toronto, Toronto, Canada; Brain and Cognition Discovery Foundation, Toronto, Canada
| | - Zhifei Li
- Institute for Health Innovation and Technology (iHealthtech), National University of Singapore, 119077, Singapore
| | - Roger C M Ho
- Institute for Health Innovation and Technology (iHealthtech), National University of Singapore, 119077, Singapore; Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 119228, Singapore; Division of Life Science (LIFS), Hong Kong University of Science and Technology, Hong Kong
| | - Tong Boon Tang
- Centre for Intelligent Signal and Imaging Research (CISIR), Universiti Teknologi PETRONAS, Seri Iskandar 32610, Perak, Malaysia
| | - Lam Ghai Lim
- Department of Electrical and Robotics Engineering, School of Engineering, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway 47500, Selangor, Malaysia; Medical Engineering & Technology Hub, School of Engineering, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway 47500, Selangor, Malaysia.
| |
Collapse
|
29
|
Parsaei M, Sheipouri A, Partovifar P, Shahriarinamin M, Sani SM, Taebi M, Arvin A. Diffusion magnetic resonance imaging for treatment response prediction in schizophrenia spectrum disorders: A systematic review. Psychiatry Res Neuroimaging 2024; 342:111841. [PMID: 38870842 DOI: 10.1016/j.pscychresns.2024.111841] [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: 03/02/2024] [Revised: 05/11/2024] [Accepted: 06/04/2024] [Indexed: 06/15/2024]
Abstract
A substantial portion of schizophrenia spectrum disorder (SSD) patients exhibit resistance to antipsychotic treatments, emphasizing the need for reliable treatment response biomarkers. Previous magnetic resonance imaging (MRI) studies have identified various imaging predictors in SSD. This study focuses on evaluating the effectiveness of diffusion MRI sequences, diffusion tensor imaging (DTI) and diffusion-weighted imaging (DWI), in predicting antipsychotic response in SSD patients. A systematic search for relevant articles was conducted in PubMed, Embase, Scopus, and Web of Science on February 11, 2024. Twelve studies involving a total of 742 patients were systematically reviewed. The baseline DTI/DWI biomarkers revealed significant associations with antipsychotic treatment response. Notably a consistent negative link was found between response and baseline fractional anisotropy (FA) in fronto-temporo-limbic white matter tracts, specifically the superior longitudinal fasciculus, providing moderate-level evidence. In addition, weak-level evidence was found for the negative association between the treatment response and baseline FA in the corpus callosum, internal, and external capsule tracts. Collectively, this review demonstrated that obtaining pre-treatment brain diffusion MRI scans, particularly from white matter tracts of fronto-temporo-limbic network, can assist in delineating the treatment response trajectory in patients with SSD. However, additional larger randomized controlled trials are required to further substantiate these findings.
Collapse
Affiliation(s)
- Mohammadamin Parsaei
- Breastfeeding Research Center, Family Health Research Institute, Tehran University of Medical Sciences, Tehran, Iran; Maternal, Fetal & Neonatal Research Center, Family Health Research Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Amirmahdi Sheipouri
- NCweb Association, Students' Scientific Research Center (SSRC), Tehran University of Medical Sciences, Tehran, Iran; Neuroscience Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Paniz Partovifar
- Maternal, Fetal & Neonatal Research Center, Family Health Research Institute, Tehran University of Medical Sciences, Tehran, Iran; Iranian Center of Neurological Research, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Shahriarinamin
- NCweb Association, Students' Scientific Research Center (SSRC), Tehran University of Medical Sciences, Tehran, Iran; Department of Neuroscience, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Sheida Mobader Sani
- NCweb Association, Students' Scientific Research Center (SSRC), Tehran University of Medical Sciences, Tehran, Iran; Iranian Center of Neurological Research, Tehran University of Medical Sciences, Tehran, Iran
| | - Morvarid Taebi
- Center for Orthopedic Trans-Disciplinary Applied Research, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Arvin
- Center for Orthopedic Trans-Disciplinary Applied Research, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
30
|
Kim WS, Heo DW, Maeng J, Shen J, Tsogt U, Odkhuu S, Zhang X, Cheraghi S, Kim SW, Ham BJ, Rami FZ, Sui J, Kang CY, Suk HI, Chung YC. Deep Learning-based Brain Age Prediction in Patients With Schizophrenia Spectrum Disorders. Schizophr Bull 2024; 50:804-814. [PMID: 38085061 PMCID: PMC11283195 DOI: 10.1093/schbul/sbad167] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/29/2024]
Abstract
BACKGROUND AND HYPOTHESIS The brain-predicted age difference (brain-PAD) may serve as a biomarker for neurodegeneration. We investigated the brain-PAD in patients with schizophrenia (SCZ), first-episode schizophrenia spectrum disorders (FE-SSDs), and treatment-resistant schizophrenia (TRS) using structural magnetic resonance imaging (sMRI). STUDY DESIGN We employed a convolutional network-based regression (SFCNR), and compared its performance with models based on three machine learning (ML) algorithms. We pretrained the SFCNR with sMRI data of 7590 healthy controls (HCs) selected from the UK Biobank. The parameters of the pretrained model were transferred to the next training phase with a new set of HCs (n = 541). The brain-PAD was analyzed in independent HCs (n = 209) and patients (n = 233). Correlations between the brain-PAD and clinical measures were investigated. STUDY RESULTS The SFCNR model outperformed three commonly used ML models. Advanced brain aging was observed in patients with SCZ, FE-SSDs, and TRS compared to HCs. A significant difference in brain-PAD was observed between FE-SSDs and TRS with ridge regression but not with the SFCNR model. Chlorpromazine equivalent dose and cognitive function were correlated with the brain-PAD in SCZ and FE-SSDs. CONCLUSIONS Our findings indicate that there is advanced brain aging in patients with SCZ and higher brain-PAD in SCZ can be used as a surrogate marker for cognitive dysfunction. These findings warrant further investigations on the causes of advanced brain age in SCZ. In addition, possible psychosocial and pharmacological interventions targeting brain health should be considered in early-stage SCZ patients with advanced brain age.
Collapse
Affiliation(s)
- Woo-Sung Kim
- Department of Psychiatry, Jeonbuk National University, Medical School, Jeonju, Korea
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Korea
| | - Da-Woon Heo
- Department of Artificial Intelligence, Korea University, Seoul, Korea
| | - Junyeong Maeng
- Department of Artificial Intelligence, Korea University, Seoul, Korea
| | - Jie Shen
- Department of Psychiatry, Jeonbuk National University, Medical School, Jeonju, Korea
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Korea
- Department of Psychiatry, Yanbian University, Medical School, Yanji, China
| | - Uyanga Tsogt
- Department of Psychiatry, Jeonbuk National University, Medical School, Jeonju, Korea
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Korea
| | - Soyolsaikhan Odkhuu
- Department of Psychiatry, Jeonbuk National University, Medical School, Jeonju, Korea
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Korea
| | - Xuefeng Zhang
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Korea
| | - Sahar Cheraghi
- Department of Psychiatry, Jeonbuk National University, Medical School, Jeonju, Korea
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Korea
| | - Sung-Wan Kim
- Department of Psychiatry, Chonnam National University Medical School, Gwangju, Korea
| | - Byung-Joo Ham
- Department of Psychiatry, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Korea
| | - Fatima Zahra Rami
- Department of Psychiatry, Jeonbuk National University, Medical School, Jeonju, Korea
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Korea
| | - Jing Sui
- Tri-Institutional Center for Translational Research in Neuroimaging and Data Science (TReNDS), Georgia State University, Georgia Institute of Technology, Emory University, Atlanta, GA, USA
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China
| | - Chae Yeong Kang
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Korea
| | - Heung-Il Suk
- Department of Artificial Intelligence, Korea University, Seoul, Korea
- Department of Brain and Cognitive Engineering, Korea University, Seoul, Korea
| | - Young-Chul Chung
- Department of Psychiatry, Jeonbuk National University, Medical School, Jeonju, Korea
- Department of Psychiatry, Jeonbuk National University Hospital, Jeonju, Korea
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Korea
| |
Collapse
|
31
|
Yalcinbas EA, Ajanaku B, Nelson ED, Garcia-Flores R, Eagles NJ, Montgomery KD, Stolz JM, Wu J, Divecha HR, Chandra A, Bharadwaj RA, Bach S, Rajpurohit A, Tao R, Pertea G, Shin JH, Kleinman JE, Hyde TM, Weinberger DR, Huuki-Myers LA, Collado-Torres L, Maynard KR. Transcriptomic analysis of the human habenula in schizophrenia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.26.582081. [PMID: 38463979 PMCID: PMC10925152 DOI: 10.1101/2024.02.26.582081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Pathophysiology of many neuropsychiatric disorders, including schizophrenia (SCZD), is linked to habenula (Hb) function. While pharmacotherapies and deep brain stimulation targeting the Hb are emerging as promising therapeutic treatments, little is known about the cell type-specific transcriptomic organization of the human Hb or how it is altered in SCZD. Here we define the molecular neuroanatomy of the human Hb and identify transcriptomic changes in individuals with SCZD compared to neurotypical controls. Utilizing Hb-enriched postmortem human brain tissue, we performed single nucleus RNA-sequencing (snRNA-seq; n=7 neurotypical donors) and identified 17 molecularly defined Hb cell types across 16,437 nuclei, including 3 medial and 7 lateral Hb populations, several of which were conserved between rodents and humans. Single molecule fluorescent in situ hybridization (smFISH; n=3 neurotypical donors) validated snRNA-seq Hb cell types and mapped their spatial locations. Bulk RNA-sequencing and cell type deconvolution in Hb-enriched tissue from 35 individuals with SCZD and 33 neurotypical controls yielded 45 SCZD-associated differentially expressed genes (DEGs, FDR < 0.05), with 32 (71%) unique to Hb-enriched tissue. eQTL analysis identified 717 independent SNP-gene pairs (FDR < 0.05), where either the SNP is a SCZD risk variant (16 pairs) or the gene is a SCZD DEG (7 pairs). eQTL and SCZD risk colocalization analysis identified 16 colocalized genes. These results identify topographically organized cell types with distinct molecular signatures in the human Hb and demonstrate unique genetic changes associated with SCZD, thereby providing novel molecular insights into the role of Hb in neuropsychiatric disorders. One Sentence Summary Transcriptomic analysis of the human habenula and identification of molecular changes associated with schizophrenia risk and illness state.
Collapse
|
32
|
Deneault A, Dumais A, Désilets M, Hudon A. Natural Language Processing and Schizophrenia: A Scoping Review of Uses and Challenges. J Pers Med 2024; 14:744. [PMID: 39063998 PMCID: PMC11278236 DOI: 10.3390/jpm14070744] [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: 06/21/2024] [Revised: 07/04/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
(1) Background: Approximately 1% of the global population is affected by schizophrenia, a disorder marked by cognitive deficits, delusions, hallucinations, and language issues. It is associated with genetic, neurological, and environmental factors, and linked to dopaminergic hyperactivity and neurotransmitter imbalances. Recent research reveals that patients exhibit significant language impairments, such as reduced verbal output and fluency. Advances in machine learning and natural language processing show potential for early diagnosis and personalized treatments, but additional research is required for the practical application and interpretation of such technology. The objective of this study is to explore the applications of natural language processing in patients diagnosed with schizophrenia. (2) Methods: A scoping review was conducted across multiple electronic databases, including Medline, PubMed, Embase, and PsycInfo. The search strategy utilized a combination of text words and subject headings, focusing on schizophrenia and natural language processing. Systematically extracted information included authors, population, primary uses of the natural language processing algorithms, main outcomes, and limitations. The quality of the identified studies was assessed. (3) Results: A total of 516 eligible articles were identified, from which 478 studies were excluded based on the first analysis of titles and abstracts. Of the remaining 38 studies, 18 were selected as part of this scoping review. The following six main uses of natural language processing were identified: diagnostic and predictive modeling, followed by specific linguistic phenomena, speech and communication analysis, social media and online content analysis, clinical and cognitive assessment, and linguistic feature analysis. (4) Conclusions: This review highlights the main uses of natural language processing in the field of schizophrenia and the need for more studies to validate the effectiveness of natural language processing in diagnosing and treating schizophrenia.
Collapse
Affiliation(s)
- Antoine Deneault
- Department of Psychiatry and Addictology, Faculty of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada;
| | - Alexandre Dumais
- Department of Psychiatry, Institut Universitaire en santé Mentale de Montréal, Montreal, QC H1N 3M5, Canada; (A.D.); (M.D.)
| | - Marie Désilets
- Department of Psychiatry, Institut Universitaire en santé Mentale de Montréal, Montreal, QC H1N 3M5, Canada; (A.D.); (M.D.)
| | - Alexandre Hudon
- Department of Psychiatry, Institut Universitaire en santé Mentale de Montréal, Montreal, QC H1N 3M5, Canada; (A.D.); (M.D.)
| |
Collapse
|
33
|
Ghamri KA. Mutual effects of gestational diabetes and schizophrenia: how can one promote the other?: A review. Medicine (Baltimore) 2024; 103:e38677. [PMID: 38905391 PMCID: PMC11191934 DOI: 10.1097/md.0000000000038677] [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: 04/03/2024] [Accepted: 05/31/2024] [Indexed: 06/23/2024] Open
Abstract
Although the physical complications of gestational diabetes mellitus (GDM) are well known, emerging evidence suggests a significant link with psychiatric conditions such as schizophrenia (SCZ). This review aimed to explore the extent, nature, and implications of the association between GDM and SCZ, exploring how the 2 conditions may reciprocally influence each other. We conducted a comprehensive literature review and, analyzed clinical and mechanistic evidence supporting the mutual effects of GDM and SCZ. This review examined factors such as neurodevelopment and the impact of antipsychotics. The study found that Maternal GDM increases the risk of SCZ in offspring. Conversely, women with SCZ were more prone to hyperglycemic pregnancies. The research highlights significant regional variations in GDM prevalence, with the highest rate in the Middle East, North Africa, and South-East Asia regions. These regional variations may have an impact on the epidemiology of SCZ. Furthermore, this review identifies the potential biological and environmental mechanisms underlying these associations. There is a bidirectional relationship between GDM and SCZ, with each disorder potentially exacerbating the others. This relationship has significant implications for maternal and offspring health, particularly in regions with high GDM prevalence. These findings underline the need for integrated care approaches for women with SCZ during pregnancy and the importance of monitoring and managing GDM to mitigate the risk of SCZ in the offspring. Notably, this study recognizes the need for further research to fully understand these complex interactions and their implications for healthcare.
Collapse
Affiliation(s)
- Kholoud A. Ghamri
- Department of Internal Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
34
|
Karaglani M, Agorastos A, Panagopoulou M, Parlapani E, Athanasis P, Bitsios P, Tzitzikou K, Theodosiou T, Iliopoulos I, Bozikas VP, Chatzaki E. A novel blood-based epigenetic biosignature in first-episode schizophrenia patients through automated machine learning. Transl Psychiatry 2024; 14:257. [PMID: 38886359 PMCID: PMC11183091 DOI: 10.1038/s41398-024-02946-4] [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: 08/06/2023] [Revised: 05/15/2024] [Accepted: 05/17/2024] [Indexed: 06/20/2024] Open
Abstract
Schizophrenia (SCZ) is a chronic, severe, and complex psychiatric disorder that affects all aspects of personal functioning. While SCZ has a very strong biological component, there are still no objective diagnostic tests. Lately, special attention has been given to epigenetic biomarkers in SCZ. In this study, we introduce a three-step, automated machine learning (AutoML)-based, data-driven, biomarker discovery pipeline approach, using genome-wide DNA methylation datasets and laboratory validation, to deliver a highly performing, blood-based epigenetic biosignature of diagnostic clinical value in SCZ. Publicly available blood methylomes from SCZ patients and healthy individuals were analyzed via AutoML, to identify SCZ-specific biomarkers. The methylation of the identified genes was then analyzed by targeted qMSP assays in blood gDNA of 30 first-episode drug-naïve SCZ patients and 30 healthy controls (CTRL). Finally, AutoML was used to produce an optimized disease-specific biosignature based on patient methylation data combined with demographics. AutoML identified a SCZ-specific set of novel gene methylation biomarkers including IGF2BP1, CENPI, and PSME4. Functional analysis investigated correlations with SCZ pathology. Methylation levels of IGF2BP1 and PSME4, but not CENPI were found to differ, IGF2BP1 being higher and PSME4 lower in the SCZ group as compared to the CTRL group. Additional AutoML classification analysis of our experimental patient data led to a five-feature biosignature including all three genes, as well as age and sex, that discriminated SCZ patients from healthy individuals [AUC 0.755 (0.636, 0.862) and average precision 0.758 (0.690, 0.825)]. In conclusion, this three-step pipeline enabled the discovery of three novel genes and an epigenetic biosignature bearing potential value as promising SCZ blood-based diagnostics.
Collapse
Affiliation(s)
- Makrina Karaglani
- Laboratory of Pharmacology, Department of Medicine, Democritus University of Thrace, GR-68132, Alexandroupolis, Greece
- Institute of Agri-food and Life Sciences, University Research & Innovation Center, H.M.U.R.I.C., Hellenic Mediterranean University, GR-71003, Crete, Greece
| | - Agorastos Agorastos
- Institute of Agri-food and Life Sciences, University Research & Innovation Center, H.M.U.R.I.C., Hellenic Mediterranean University, GR-71003, Crete, Greece
- II. Department of Psychiatry, Faculty of Health Sciences, School of Medicine, Aristotle University of Thessaloniki, GR-56430, Thessaloniki, Greece
| | - Maria Panagopoulou
- Laboratory of Pharmacology, Department of Medicine, Democritus University of Thrace, GR-68132, Alexandroupolis, Greece
- Institute of Agri-food and Life Sciences, University Research & Innovation Center, H.M.U.R.I.C., Hellenic Mediterranean University, GR-71003, Crete, Greece
| | - Eleni Parlapani
- Ι. Department of Psychiatry, Faculty of Health Sciences, School of Medicine, Aristotle University of Thessaloniki, GR-56429, Thessaloniki, Greece
| | - Panagiotis Athanasis
- II. Department of Psychiatry, Faculty of Health Sciences, School of Medicine, Aristotle University of Thessaloniki, GR-56430, Thessaloniki, Greece
| | - Panagiotis Bitsios
- Department of Psychiatry and Behavioral Sciences, Faculty of Medicine, University of Crete, GR-71500, Heraklion, Greece
| | - Konstantina Tzitzikou
- Laboratory of Pharmacology, Department of Medicine, Democritus University of Thrace, GR-68132, Alexandroupolis, Greece
| | - Theodosis Theodosiou
- Laboratory of Pharmacology, Department of Medicine, Democritus University of Thrace, GR-68132, Alexandroupolis, Greece
- ABCureD P.C, GR-68131, Alexandroupolis, Greece
| | - Ioannis Iliopoulos
- Division of Basic Sciences, School of Medicine, University of Crete, GR-71003, Heraklion, Greece
| | - Vasilios-Panteleimon Bozikas
- II. Department of Psychiatry, Faculty of Health Sciences, School of Medicine, Aristotle University of Thessaloniki, GR-56430, Thessaloniki, Greece
| | - Ekaterini Chatzaki
- Laboratory of Pharmacology, Department of Medicine, Democritus University of Thrace, GR-68132, Alexandroupolis, Greece.
- Institute of Agri-food and Life Sciences, University Research & Innovation Center, H.M.U.R.I.C., Hellenic Mediterranean University, GR-71003, Crete, Greece.
- ABCureD P.C, GR-68131, Alexandroupolis, Greece.
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology, 70013, Heraklion, Greece.
| |
Collapse
|
35
|
Wang Y, Fan L, He Y, Yuan L, Li Z, Zheng W, Tang J, Li C, Jin K, Liu W, Chen X, Ouyang L, Ma X. Compensatory thickening of cortical thickness in early stage of schizophrenia. Cereb Cortex 2024; 34:bhae255. [PMID: 38897816 DOI: 10.1093/cercor/bhae255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 05/28/2024] [Accepted: 05/29/2024] [Indexed: 06/21/2024] Open
Abstract
Brain structural abnormality has been observed in the prodromal and early stages of schizophrenia, but the mechanism behind it is not clear. In this study, to explore the association between cortical abnormalities, metabolite levels, inflammation levels and clinical symptoms of schizophrenia, 51 drug-naive first-episode schizophrenia (FES) patients, 51 ultra-high risk for psychosis (UHR), and 51 healthy controls (HC) were recruited. We estimated gray matter volume (GMV), cortical thickness (CT), concentrations of different metabolites, and inflammatory marks among four groups (UHR converted to psychosis [UHR-C], UHR unconverted to psychosis [UHR-NC], FES, HC). UHR-C group had more CT in the right lateral occipital cortex and the right medial orbito-frontal cortex (rMOF), while a significant reduction in CT of the right fusiform cortex was observed in FES group. UHR-C group had significantly higher concentration of IL-6, while IL-17 could significantly predict CT of the right fusiform and IL-4 and IL-17 were significant predictors of CT in the rMOF. To conclude, it is reasonable to speculate that the increased CT in UHR-C group is related to the inflammatory response, and may participate in some compensatory mechanism, but might become exhaustive with the progress of the disease due to potential neurotoxic effects.
Collapse
Affiliation(s)
- Yujue Wang
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Furong District No. 139 Renmin Road, Changsha 410011, Hunan, China
| | - Lejia Fan
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Furong District No. 139 Renmin Road, Changsha 410011, Hunan, China
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, 6875 Bd LaSalle, Verdun, Montreal, QC H4H 1R3, Canada
| | - Ying He
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Furong District No. 139 Renmin Road, Changsha 410011, Hunan, China
- China National Technology Institute on Mental Disorders, Furong District No. 139 Renmin Road, Changsha 410011, Hunan, China
- Hunan Key Laboratory of Psychiatry and Mental Health, Furong District No. 139 Renmin Road, Changsha 410011, Hunan, China
- Institute of Mental Health, Furong District No. 139 Renmin Road, Changsha 410011, Hunan, China
- Hunan Medical Center for Mental Health, Furong District No. 139 Renmin Road, Changsha 410011, Hunan, China
| | - Liu Yuan
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Furong District No. 139 Renmin Road, Changsha 410011, Hunan, China
| | - Zongchang Li
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Furong District No. 139 Renmin Road, Changsha 410011, Hunan, China
| | - Wenxiao Zheng
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Furong District No. 139 Renmin Road, Changsha 410011, Hunan, China
| | - Jinsong Tang
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Furong District No. 139 Renmin Road, Changsha 410011, Hunan, China
| | - Chunwang Li
- Department of Radiology, Hunan Children's Hospital, Yuhua District catalpa garden road 86, Changsha 410007, Hunan, China
| | - Ke Jin
- Department of Radiology, Hunan Children's Hospital, Yuhua District catalpa garden road 86, Changsha 410007, Hunan, China
| | - Weiqing Liu
- Clinical Research Center for Mental Disorders, Shanghai Pudong New Area Mental Health Center, School of Medicine, Tongji University, #165 Sanlin road, Pudong New Area,Shanghai 200124, China
- Laboratory for Molecular Mechanisms of Brain Development, Center for Brain Science (CBS), 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Xiaogang Chen
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Furong District No. 139 Renmin Road, Changsha 410011, Hunan, China
- China National Technology Institute on Mental Disorders, Furong District No. 139 Renmin Road, Changsha 410011, Hunan, China
- Hunan Key Laboratory of Psychiatry and Mental Health, Furong District No. 139 Renmin Road, Changsha 410011, Hunan, China
- Institute of Mental Health, Furong District No. 139 Renmin Road, Changsha 410011, Hunan, China
- Hunan Medical Center for Mental Health, Furong District No. 139 Renmin Road, Changsha 410011, Hunan, China
| | - Lijun Ouyang
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Furong District No. 139 Renmin Road, Changsha 410011, Hunan, China
| | - Xiaoqian Ma
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Furong District No. 139 Renmin Road, Changsha 410011, Hunan, China
- China National Technology Institute on Mental Disorders, Furong District No. 139 Renmin Road, Changsha 410011, Hunan, China
- Hunan Key Laboratory of Psychiatry and Mental Health, Furong District No. 139 Renmin Road, Changsha 410011, Hunan, China
- Institute of Mental Health, Furong District No. 139 Renmin Road, Changsha 410011, Hunan, China
- Hunan Medical Center for Mental Health, Furong District No. 139 Renmin Road, Changsha 410011, Hunan, China
| |
Collapse
|
36
|
Georgiadis F, Larivière S, Glahn D, Hong LE, Kochunov P, Mowry B, Loughland C, Pantelis C, Henskens FA, Green MJ, Cairns MJ, Michie PT, Rasser PE, Catts S, Tooney P, Scott RJ, Schall U, Carr V, Quidé Y, Krug A, Stein F, Nenadić I, Brosch K, Kircher T, Gur R, Gur R, Satterthwaite TD, Karuk A, Pomarol-Clotet E, Radua J, Fuentes-Claramonte P, Salvador R, Spalletta G, Voineskos A, Sim K, Crespo-Facorro B, Tordesillas Gutiérrez D, Ehrlich S, Crossley N, Grotegerd D, Repple J, Lencer R, Dannlowski U, Calhoun V, Rootes-Murdy K, Demro C, Ramsay IS, Sponheim SR, Schmidt A, Borgwardt S, Tomyshev A, Lebedeva I, Höschl C, Spaniel F, Preda A, Nguyen D, Uhlmann A, Stein DJ, Howells F, Temmingh HS, Diaz Zuluaga AM, López Jaramillo C, Iasevoli F, Ji E, Homan S, Omlor W, Homan P, Kaiser S, Seifritz E, Misic B, Valk SL, Thompson P, van Erp TGM, Turner JA, Bernhardt B, Kirschner M. Connectome architecture shapes large-scale cortical alterations in schizophrenia: a worldwide ENIGMA study. Mol Psychiatry 2024; 29:1869-1881. [PMID: 38336840 PMCID: PMC11371638 DOI: 10.1038/s41380-024-02442-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 01/08/2024] [Accepted: 01/18/2024] [Indexed: 02/12/2024]
Abstract
Schizophrenia is a prototypical network disorder with widespread brain-morphological alterations, yet it remains unclear whether these distributed alterations robustly reflect the underlying network layout. We tested whether large-scale structural alterations in schizophrenia relate to normative structural and functional connectome architecture, and systematically evaluated robustness and generalizability of these network-level alterations. Leveraging anatomical MRI scans from 2439 adults with schizophrenia and 2867 healthy controls from 26 ENIGMA sites and normative data from the Human Connectome Project (n = 207), we evaluated structural alterations of schizophrenia against two network susceptibility models: (i) hub vulnerability, which examines associations between regional network centrality and magnitude of disease-related alterations; (ii) epicenter mapping, which identifies regions whose typical connectivity profile most closely resembles the disease-related morphological alterations. To assess generalizability and specificity, we contextualized the influence of site, disease stages, and individual clinical factors and compared network associations of schizophrenia with that found in affective disorders. Our findings show schizophrenia-related cortical thinning is spatially associated with functional and structural hubs, suggesting that highly interconnected regions are more vulnerable to morphological alterations. Predominantly temporo-paralimbic and frontal regions emerged as epicenters with connectivity profiles linked to schizophrenia's alteration patterns. Findings were robust across sites, disease stages, and related to individual symptoms. Moreover, transdiagnostic comparisons revealed overlapping epicenters in schizophrenia and bipolar, but not major depressive disorder, suggestive of a pathophysiological continuity within the schizophrenia-bipolar-spectrum. In sum, cortical alterations over the course of schizophrenia robustly follow brain network architecture, emphasizing marked hub susceptibility and temporo-frontal epicenters at both the level of the group and the individual. Subtle variations of epicenters across disease stages suggest interacting pathological processes, while associations with patient-specific symptoms support additional inter-individual variability of hub vulnerability and epicenters in schizophrenia. Our work outlines potential pathways to better understand macroscale structural alterations, and inter- individual variability in schizophrenia.
Collapse
Grants
- R01 MH118695 NIMH NIH HHS
- R01 NS114628 NINDS NIH HHS
- R21 MH097196 NIMH NIH HHS
- R01 EB015611 NIBIB NIH HHS
- RF1 NS114628 NINDS NIH HHS
- RF1 MH123163 NIMH NIH HHS
- R01 AA012207 NIAAA NIH HHS
- S10 OD023696 NIH HHS
- I01 CX000227 CSRD VA
- R01 MH112583 NIMH NIH HHS
- The Australian Schizophrenia Research Bank (ASRB) was supported by NHMRC (Enabling Grant, ID 386500), the Pratt Foundation, Ramsay Health Care, the Viertel Charitable Foundation and the Schizophrenia Research Institute. Chief Investigators for ASRB were Carr, V., Schall, U., Scott, R., Jablensky, A., Mowry, B., Michie, P., Catts, S., Henskens, F., Pantelis, C. We thank Loughland, C., the ASRB Manager, and acknowledge the help of Jason Bridge for ASRB database queries.
- The Australian Schizophrenia Research Bank (ASRB) was supported by NHMRC(Enabling Grant, ID 386500), \the Pratt Foundation, Ramsay Health Care, the Viertel Charitable Foundation and the Schizophrenia Research Institute. Chief Investigators for ASRB were Carr, V., Schall, U., Scott, R., Jablensky, A., Mowry, B., Michie, P., Catts, S., Henskens, F., Pantelis, C. We thank Loughland, C., the ASRB Manager, and acknowledge the help of Jason Bridge for ASRB database queries.
- NIMH Grant R01MH118695, NSF Grant 2112455
- Supported by the Ministry of Health of the Czech Republic, grant NU20-04-00393 and 17-31852A.
- Supported by the Instituto de Salud Carlos III, the Spanish Ministry of Science, Innovation, and Universities, the European Regional Development Fund (ERDF/FEDER), European Social Fund, “Investing in your future”, “A way of making Europe” (CPII19/00009)
- This work was funded by the German Research Foundation (DFG grant FOR2107, KI588/14-1 and FOR2107, KI588/14-2 to Tilo Kircher, Marburg, Germany).
- This work was supported by research grants from the National Healthcare Group, Singapore, and the Singapore Bioimaging Consortium research grants awarded to K.S.
- This work was supported by the awards by the Department of Veterans Affairs Clinical Science Research and Development Service (Grant No. I01CX000227) and the National Institute of Mental Health (Grant No. R01MH112583).
- This work was supported by the Italian Ministry of Health Grant RC21,22,23
- This work was in part supported by NIMH R21MH097196.
- This work was funded by the German Research Foundation (DFG, grant FOR2107 DA1151/5-1 and DA1151/5-2 to UD; SFB-TRR58, Projects C09 and Z02 to UD)
Collapse
Affiliation(s)
- Foivos Georgiadis
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital University of Zurich, Zurich, Switzerland.
| | - Sara Larivière
- McGill University, Montreal Neurological Institute, Montreal, QC, Canada
| | - David Glahn
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - L Elliot Hong
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, US
| | - Peter Kochunov
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, US
| | - Bryan Mowry
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD, Australia
| | - Carmel Loughland
- School of Medicine and Public Health, University of Newcastle, Newcastle, NSW, USA
| | - Christos Pantelis
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne, Carlton South, VIC, Australia
| | - Frans A Henskens
- School of Medicine and Public Health, University of Newcastle, Newcastle, NSW, Australia
| | - Melissa J Green
- School of Clinical Medicine, Discipline of Psychiatry and Mental Health, UNSW Sydney, Sydney, NSW, Australia
| | - Murray J Cairns
- School of Biomedical Science and Pharmacy, University of Newcastle, Newcastle, NSW, Australia
| | - Patricia T Michie
- School of Psychological Sciences, University of Newcastle, Newcastle, NSW, Australia
| | - Paul E Rasser
- School of Medicine and Public Health, College of Health, Medicine, and Wellbeing, The University of Newcastle, Callaghan, NSW, Australia
| | - Stanley Catts
- Faculty of Medicine, University of Queensland, St Lucia, QLD, Australia
| | - Paul Tooney
- School of Biomedical Science and Pharmacy, University of Newcastle, Newcastle, NSW, Australia
- Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Rodney J Scott
- School of Biomedical Science and Pharmacy, University of Newcastle, Newcastle, NSW, Australia
| | - Ulrich Schall
- Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Vaughan Carr
- School of Clinical Medicine, Discipline of Psychiatry, UNSW Sydney, Sydney, NSW, Australia
| | - Yann Quidé
- School of Clinical Medicine, Discipline of Psychiatry and Mental Health, UNSW Sydney, Sydney, NSW, Australia
| | - Axel Krug
- University Hospital Bonn, Department of Psychiatry and Psychotherapy, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Frederike Stein
- Department of Psychiatry, University of Marburg, Rudolf Bultmann Str. 8, 35039, Marburg, Germany
| | - Igor Nenadić
- Department. of Psychiatry and Psychotherapy, Philipps-University Marburg, Marburg, Germany
| | - Katharina Brosch
- Department of Psychiatry, University of Marburg, Rudolf Bultmann Str. 8, 35039, Marburg, Germany
| | - Tilo Kircher
- Department of Psychiatry, University of Marburg, Rudolf Bultmann Str. 8, 35039, Marburg, Germany
| | - Raquel Gur
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Ruben Gur
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | | | - Andriana Karuk
- FIDMAG Germanes Hospitalàries Research Foundation & CIBERSAM, ISCIII, Barcelona, Spain
| | - Edith Pomarol-Clotet
- FIDMAG Germanes Hospitalàries Research Foundation & CIBERSAM, ISCIII, Barcelona, Spain
| | - Joaquim Radua
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | | | - Raymond Salvador
- FIDMAG Germanes Hospitalàries Research Foundation & CIBERSAM, ISCIII, Barcelona, Spain
| | | | - Aristotle Voineskos
- School of Biomedical Science and Pharmacy, University of Newcastle, Newcastle, NSW, Australia
| | - Kang Sim
- West Region, Institute of Mental Health, Singapore, Singapore
| | | | - Diana Tordesillas Gutiérrez
- Department of Radiology, Marqués de Valdecilla University Hospital, Valdecilla Biomedical Research Institute IDIVAL, Santander, Spain
| | - Stefan Ehrlich
- Division of Psychological & Social Medicine and Developmental Neurosciences, Technischen Universität Dresden, Faculty of Medicine, University Hospital C.G. Carus, Dresden, Germany
| | - Nicolas Crossley
- Department of Psychiatry, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Dominik Grotegerd
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Jonathan Repple
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Rebekka Lencer
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Udo Dannlowski
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Vince Calhoun
- Tri-Institutional Center for Translational Research in Neuroimaging and Data Science (TReNDS), Georgia State, Georgia Tech, Emory, Atlanta, GA, USA
| | - Kelly Rootes-Murdy
- Tri-Institutional Center for Translational Research in Neuroimaging and Data Science (TReNDS), Georgia State, Georgia Tech, Emory, Atlanta, GA, USA
| | - Caroline Demro
- University of Minnesota Department of Psychology, Minneapolis, MN, USA
- Minneapolis VA Health Care System, Minneapolis, MN, USA
| | - Ian S Ramsay
- University of Minnesota Department of Psychiatry & Behavioral Sciences, Minneapolis, MN, USA
| | - Scott R Sponheim
- Minneapolis VA Health Care System, Minneapolis, MN, USA
- University of Minnesota Department of Psychiatry & Behavioral Sciences, Minneapolis, MN, USA
| | - Andre Schmidt
- University of Basel, Department of Psychiatry, Basel, Switzerland
| | | | | | - Irina Lebedeva
- Mental Health Research Center, Moscow, Russian Federation
| | - Cyril Höschl
- National Institute of Mental Health, Topolova 748, 250 67, Klecany, Czech Republic
| | - Filip Spaniel
- National Institute of Mental Health, Topolova 748, 250 67, Klecany, Czech Republic
| | - Adrian Preda
- Department of Psychiatry and Human Behavior, University of California Irvine, Irvine, CA, USA
| | - Dana Nguyen
- Department of Pediatric Neurology, University of California Irvine, Irvine, CA, USA
| | - Anne Uhlmann
- Department of child and adolescent psychiatry, TU Dresden, Dresden, Germany
| | - Dan J Stein
- Department of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa
| | - Fleur Howells
- Department of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa
| | - Henk S Temmingh
- Department of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa
| | - Ana M Diaz Zuluaga
- Research Group in Psychiatry, Department of Psychiatry, School of Medicine, Universidad de Antioquia, Medellin, Colombia
| | - Carlos López Jaramillo
- Research Group in Psychiatry, Department of Psychiatry, School of Medicine, Universidad de Antioquia, Medellin, Colombia
| | - Felice Iasevoli
- University of Naples, Department of Neuroscience, Naples, Italy
| | - Ellen Ji
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital University of Zurich, Zurich, Switzerland
| | - Stephanie Homan
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital University of Zurich, Zurich, Switzerland
| | - Wolfgang Omlor
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital University of Zurich, Zurich, Switzerland
| | - Philipp Homan
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital University of Zurich, Zurich, Switzerland
| | - Stefan Kaiser
- Division of Adult Psychiatry, Department of Psychiatry, Geneva University Hospitals, Geneva, Switzerland
| | - Erich Seifritz
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital University of Zurich, Zurich, Switzerland
| | - Bratislav Misic
- McGill University, Montreal Neurological Institute, Montreal, QC, Canada
| | - Sofie L Valk
- Forschungszentrum Jülich, Jülich, Germany
- Max Planck Institute for Cognitive and Brain Sciences, Leipzig, Germany
| | - Paul Thompson
- Imaging Genetics Center, Stevens Institute for Neuroimaging and Informatics, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Theo G M van Erp
- Department of Psychiatry and Human Behavior, University of California Irvine, Irvine, CA, USA
| | - Jessica A Turner
- Department of Psychiatry and Behavioral Health, the Ohio State University, Columbus, OH, USA
| | - Boris Bernhardt
- McGill University, Montreal Neurological Institute, Montreal, QC, Canada
| | - Matthias Kirschner
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital University of Zurich, Zurich, Switzerland.
- Division of Adult Psychiatry, Department of Psychiatry, Geneva University Hospitals, Geneva, Switzerland.
| |
Collapse
|
37
|
Sun T, Du YY, Zhang YQ, Tian QQ, Li X, Yu JY, Guo YY, Liu QQ, Yang L, Wu YM, Yang Q, Zhao MG. Activation of GPR55 Ameliorates Maternal Separation-Induced Learning and Memory Deficits by Augmenting 5-HT Synthesis in the Dorsal Raphe Nucleus of Juvenile Mice. ACS OMEGA 2024; 9:21838-21850. [PMID: 38799363 PMCID: PMC11112691 DOI: 10.1021/acsomega.3c08934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 04/16/2024] [Accepted: 04/24/2024] [Indexed: 05/29/2024]
Abstract
Maternal separation (MS) represents a profound early life stressor with enduring impacts on neuronal development and adult cognitive function in both humans and rodents. MS is associated with persistent dysregulations in neurotransmitter systems, including the serotonin (5-HT) pathway, which is pivotal for mood stabilization and stress-coping mechanisms. Although the novel cannabinoid receptor, GPR55, is recognized for its influence on learning and memory, its implications on the function and synaptic dynamics of 5-HT neurons within the dorsal raphe nucleus (DRN) remain to be elucidated. In this study, we sought to discern the repercussions of GPR55 activation on 5-HT synthesis within the DRN of adult C57BL/6J mice that experienced MS. Concurrently, we analyzed potential alterations in excitatory synaptic transmission, long-term synaptic plasticity, and relevant learning and memory outcomes. Our behavioral assessments indicated a marked amelioration in MS-induced learning and memory deficits following GPR55 activation. In conjunction with this, we noted a substantial decrease in 5-HT levels in the MS model, while GPR55 activation stimulated tryptophan hydroxylase 2 synthesis and fostered the release of 5-HT. Electrophysiological patch-clamp analyses highlighted the ability of GPR55 activation to alleviate MS-induced cognitive deficits by modulating the frequency and magnitude of miniature excitatory postsynaptic currents within the DRN. Notably, this cognitive enhancement was underpinned by the phosphorylation of both NMDA and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors. In summary, our findings underscore the capacity of GPR55 to elevate 5-HT synthesis and modify synaptic transmissions within the DRN of juvenile mice, positing GPR55 as a promising therapeutic avenue for ameliorating MS-induced cognitive impairment.
Collapse
Affiliation(s)
- Ting Sun
- Precision
Pharmacy & Drug Development Center, Department of Pharmacy, The Second Affiliated Hospital of Air Force Medical
University, Xi’an 710038, China
| | - Ya-Ya Du
- Precision
Pharmacy & Drug Development Center, Department of Pharmacy, The Second Affiliated Hospital of Air Force Medical
University, Xi’an 710038, China
| | - Yong-Qiang Zhang
- Department
of Chinese Materia Medica and Natural Medicines, School of Pharmacy, Air Force Medical University, Xi’an 710032, China
| | - Qin-Qin Tian
- Department
of Chemistry, School of Pharmacy, Air Force
Medical University, Xi’an 710032, China
| | - Xi Li
- Precision
Pharmacy & Drug Development Center, Department of Pharmacy, The Second Affiliated Hospital of Air Force Medical
University, Xi’an 710038, China
| | - Jiao-Yan Yu
- Precision
Pharmacy & Drug Development Center, Department of Pharmacy, The Second Affiliated Hospital of Air Force Medical
University, Xi’an 710038, China
| | - Yan-Yan Guo
- Precision
Pharmacy & Drug Development Center, Department of Pharmacy, The Second Affiliated Hospital of Air Force Medical
University, Xi’an 710038, China
| | - Qing-Qing Liu
- Precision
Pharmacy & Drug Development Center, Department of Pharmacy, The Second Affiliated Hospital of Air Force Medical
University, Xi’an 710038, China
| | - Le Yang
- Precision
Pharmacy & Drug Development Center, Department of Pharmacy, The Second Affiliated Hospital of Air Force Medical
University, Xi’an 710038, China
| | - Yu-Mei Wu
- Department
of Pharmacology, School of Pharmacy, Air
Force Medical University, Xi’an 710032, China
| | - Qi Yang
- Precision
Pharmacy & Drug Development Center, Department of Pharmacy, The Second Affiliated Hospital of Air Force Medical
University, Xi’an 710038, China
| | - Ming-Gao Zhao
- Precision
Pharmacy & Drug Development Center, Department of Pharmacy, The Second Affiliated Hospital of Air Force Medical
University, Xi’an 710038, China
| |
Collapse
|
38
|
Faris P, Pischedda D, Palesi F, D’Angelo E. New clues for the role of cerebellum in schizophrenia and the associated cognitive impairment. Front Cell Neurosci 2024; 18:1386583. [PMID: 38799988 PMCID: PMC11116653 DOI: 10.3389/fncel.2024.1386583] [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: 02/15/2024] [Accepted: 04/26/2024] [Indexed: 05/29/2024] Open
Abstract
Schizophrenia (SZ) is a complex neuropsychiatric disorder associated with severe cognitive dysfunction. Although research has mainly focused on forebrain abnormalities, emerging results support the involvement of the cerebellum in SZ physiopathology, particularly in Cognitive Impairment Associated with SZ (CIAS). Besides its role in motor learning and control, the cerebellum is implicated in cognition and emotion. Recent research suggests that structural and functional changes in the cerebellum are linked to deficits in various cognitive domains including attention, working memory, and decision-making. Moreover, cerebellar dysfunction is related to altered cerebellar circuit activities and connectivity with brain regions associated with cognitive processing. This review delves into the role of the cerebellum in CIAS. We initially consider the major forebrain alterations in CIAS, addressing impairments in neurotransmitter systems, synaptic plasticity, and connectivity. We then focus on recent findings showing that several mechanisms are also altered in the cerebellum and that cerebellar communication with the forebrain is impaired. This evidence implicates the cerebellum as a key component of circuits underpinning CIAS physiopathology. Further studies addressing cerebellar involvement in SZ and CIAS are warranted and might open new perspectives toward understanding the physiopathology and effective treatment of these disorders.
Collapse
Affiliation(s)
- Pawan Faris
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Doris Pischedda
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Fulvia Palesi
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Egidio D’Angelo
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
- Digital Neuroscience Center, IRCCS Mondino Foundation, Pavia, Italy
| |
Collapse
|
39
|
Sanz P, Tur N, Lana F. Mentalization-based approach for schizophrenia spectrum disorders: a psychotherapeutic proposal for evolved schizophrenic trajectories and serious mental disorders. Front Psychiatry 2024; 15:1240393. [PMID: 38779549 PMCID: PMC11109361 DOI: 10.3389/fpsyt.2024.1240393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 04/16/2024] [Indexed: 05/25/2024] Open
Abstract
There is a growing interest in psychotherapeutic approaches to pre-psychotic high-risk states or first-episode psychosis, where mentalization-based treatment has shown its utility. This article presents a mentalization-based approach for the treatment of those individuals diagnosed with an evolved schizophrenia spectrum disorder, whose characteristics make them especially inaccessible to reflective psychotherapeutic treatment. A synthesis of the conceptual frameworks that justify the needs for technical modification of the mentalization-based treatment foundational techniques is carried out, followed by the proposal of adaptations, with a focus in self-agency and patient-therapist dyad. Therapeutic interventions are outlined, including illustrative examples. The mentalizing approach presented here holds promise for future research and treatment opportunities for patients with evolved schizophrenia and other serious mental disorders.
Collapse
Affiliation(s)
- Pedro Sanz
- Programa de Trastorno Mental Grave, Area de Gestión Clínica de Psiquiatria y Salud Mental (AGCPSM), Hospital 12 de Octubre, Madrid, Spain
| | - Nuria Tur
- Servicio de Psiquiatria, Unidad del Niño y Adolescente, Hospital Clínico San Carlos, Madrid, Spain
| | - Fernando Lana
- Instituto de Neuropsiquiatría y Adiciones (INAD), Centro Emili Mira y Hospital del Mar, Parc de Salut Mar, Barcelona, Centro de Investigación en Red de Salud Mental (CIBERSAM), Departamento de Psiquiatría, Universidad Autónoma de Barcelona, EspañaIMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| |
Collapse
|
40
|
Olivares-Berjaga D, Martínez-Pinteño A, Rodríguez N, Madero S, Prohens L, Martínez-Serrano I, Mas S, Morén C, Parellada E, Gassó P. Effects of the PAM of mGluR2, JNJ-46356479, on brain apoptotic protein levels in a mouse model of schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry 2024; 131:110955. [PMID: 38296154 DOI: 10.1016/j.pnpbp.2024.110955] [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: 10/09/2023] [Revised: 01/25/2024] [Accepted: 01/28/2024] [Indexed: 02/12/2024]
Abstract
Current treatment for schizophrenia (SZ) ameliorates the positive symptoms, but is inefficient in treating the negative and cognitive symptoms. The SZ glutamatergic dysfunction hypothesis has opened new avenues in the development of novel drugs targeting the glutamate storm, an inducer of progressive neuropathological changes. Positive allosteric modulators of metabotropic glutamate receptor 2 (mGluR2), such as JNJ-46356479 (JNJ), reduce the presynaptic release of glutamate, which has previously been demonstrated to attenuate glutamate- and dopamine-induced apoptosis in human neuroblastoma cell cultures. We hypothesised that JNJ treatment would modify the brain levels of apoptotic proteins in a mouse model of ketamine (KET)-induced schizophrenia. We analysed the levels of proapoptotic (caspase-3 and Bax) and antiapoptotic (Bcl-2) proteins by western blot in the prefrontal cortex and hippocampus of JNJ-treated mice. JNJ attenuated apoptosis in the brain by partially restoring the levels of the antiapoptotic Bcl-2 protein, which is significantly reduced in animals exposed to KET. Additionally, a significant inverse correlation was observed between proapoptotic protein levels and behavioural deficits in the mice. Our findings suggest that JNJ may attenuate brain apoptosis in vivo, as previously described in cell cultures, providing a link between neuropathological deficits and SZ symptomatology.
Collapse
Affiliation(s)
| | - Albert Martínez-Pinteño
- Dept. of Basic Clinical Practice, University of Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
| | - Natalia Rodríguez
- Dept. of Basic Clinical Practice, University of Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
| | - Santiago Madero
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain; Barcelona Clínic Schizophrenia Unit (BCSU), Dpt. of Psychiatry, Institute of Neuroscience, Hospital Clínic of Barcelona, University of Barcelona, Spain.
| | - Llucía Prohens
- Dept. of Basic Clinical Practice, University of Barcelona, Spain.
| | | | - Sergi Mas
- Dept. of Basic Clinical Practice, University of Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain.
| | - Constanza Morén
- Dept. of Basic Clinical Practice, University of Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Barcelona Clínic Schizophrenia Unit (BCSU), Dpt. of Psychiatry, Institute of Neuroscience, Hospital Clínic of Barcelona, University of Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Spain.
| | - Eduard Parellada
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain; Barcelona Clínic Schizophrenia Unit (BCSU), Dpt. of Psychiatry, Institute of Neuroscience, Hospital Clínic of Barcelona, University of Barcelona, Spain.
| | - Patricia Gassó
- Dept. of Basic Clinical Practice, University of Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain.
| |
Collapse
|
41
|
Akkouh IA, Ueland T, Szabo A, Hughes T, Smeland OB, Andreassen OA, Osete JR, Djurovic S. Longitudinal Transcriptomic Analysis of Human Cortical Spheroids Identifies Axonal Dysregulation in the Prenatal Brain as a Mediator of Genetic Risk for Schizophrenia. Biol Psychiatry 2024; 95:687-698. [PMID: 37661009 DOI: 10.1016/j.biopsych.2023.08.017] [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: 01/25/2023] [Revised: 07/28/2023] [Accepted: 08/22/2023] [Indexed: 09/05/2023]
Abstract
BACKGROUND Schizophrenia (SCZ) has a known neurodevelopmental etiology, but limited access to human prenatal brain tissue hampers the investigation of basic disease mechanisms in early brain development. Here, we elucidate the molecular mechanisms contributing to SCZ risk in a disease-relevant model of the prenatal human brain. METHODS We generated induced pluripotent stem cell-derived organoids, termed human cortical spheroids (hCSs), from a large, genetically stratified sample of 14 SCZ cases and 14 age- and sex-matched controls. The hCSs were differentiated for 150 days, and comprehensive molecular characterization across 4 time points was carried out. RESULTS The transcriptional and cellular architecture of hCSs closely resembled that of fetal brain tissue at 10 to 24 postconception weeks, showing strongest spatial overlap with frontal regions of the cerebral cortex. A total of 3520 genes were differentially modulated between SCZ and control hCSs across organoid maturation, displaying a significant contribution of genetic loading, an overrepresentation of risk genes for autism spectrum disorder and SCZ, and the strongest enrichment for axonal processes in all hCS stages. The two axon guidance genes SEMA7A and SEMA5A, the first a promoter of synaptic functions and the second a repressor, were downregulated and upregulated, respectively, in SCZ hCSs. This expression pattern was confirmed at the protein level and replicated in a large postmortem sample. CONCLUSIONS Applying a disease-relevant model of the developing fetal brain, we identified consistent dysregulation of axonal genes as an early risk factor for SCZ, providing novel insights into the effects of genetic predisposition on the neurodevelopmental origins of the disorder.
Collapse
Affiliation(s)
- Ibrahim A Akkouh
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; Norwegian Centre for Mental Disorders Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway.
| | - Thor Ueland
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway; K.G. Jebsen Thrombosis Research and Expertise Center, University of Tromsø, Tromsø, Norway
| | - Attila Szabo
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; Norwegian Centre for Mental Disorders Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Timothy Hughes
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; Norwegian Centre for Mental Disorders Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Olav B Smeland
- Norwegian Centre for Mental Disorders Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Ole A Andreassen
- Norwegian Centre for Mental Disorders Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Jordi Requena Osete
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; Norwegian Centre for Mental Disorders Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Srdjan Djurovic
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; Norwegian Centre for Mental Disorders Research, Department of Clinical Science, University of Bergen, Bergen, Norway.
| |
Collapse
|
42
|
Zhou J, Li X, Wang X, Yang Y, Nai A, Shi H, Zhao J, Zhang J, Ding S, Han Y, Liu Q, Zhang L, Chen T, Liu B, Yue W, Lv L, Li W. Levels of neuronal pentraxin 2 in plasma is associated with cognitive function in patients with schizophrenia. Psychopharmacology (Berl) 2024; 241:865-874. [PMID: 38191677 DOI: 10.1007/s00213-023-06515-3] [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: 05/16/2023] [Accepted: 12/01/2023] [Indexed: 01/10/2024]
Abstract
RATIONALE The precise diagnosis and treatment of cognitive impairment remains a major challenge in the field of schizophrenia (SCZ) research. Synaptic dysfunction and loss are thought to be closely related to the occurrence and development of SCZ and may be involved in cognitive dysfunction. OBJECTIVES The purpose of this study was to investigate whether neuronal pentraxins (NPTXs) plays a role in the etiology of SCZ and provide evidence of its possible therapeutic value a new target for drug development. METHODS We recruited 275 participants, of whom 148 were SCZ from psychiatric hospital and 127 healthy control (HC) subjects from communities. Plasma concentrations of NPTXs were measured in HC and SCZ at baseline and after 8 weeks of antipsychotic treatment. The MATRICS Cognitive Consensus Battery was used to evaluate cognitive function. Furthermore, the brain is parcellated into 246 subregions using the Brainnetome atlas, and we extracted regional white matter volumes from magnetic resonance images of the SCZ groups. RESULTS Plasma NPTX2 levels were significantly lower in SCZ compared with HC subjects, but were significantly raised in SCZ after 8 weeks of antipsychotic treatment compared to baseline. In addition, baseline plasma NPTX2 levels were positively correlated with cognitive performance. CONCLUSIONS These findings indicate that NPTX2 may reveal novel aspects of disease etiology and act as a promising target for new drug development.
Collapse
Affiliation(s)
- Jiahui Zhou
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, China
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, China
| | - Xiaojing Li
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, China
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, China
| | - Xiujuan Wang
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Yongfeng Yang
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, China
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, China
| | - Aoyang Nai
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Han Shi
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Jingyuan Zhao
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Jianhong Zhang
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Shuang Ding
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Yong Han
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, China
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, China
| | - Qing Liu
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, China
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, China
| | - Luwen Zhang
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, China
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, China
| | - Tengfei Chen
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, China
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, China
| | - Bing Liu
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China
- Chinese Institute for Brain Research, Beijing, China
| | - Weihua Yue
- Institute of Mental Health, Peking University, Beijing, China
- Key Laboratory for Mental Health, Ministry of Health, Beijing, China
| | - Luxian Lv
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China.
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, China.
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, China.
| | - Wenqiang Li
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China.
- Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, China.
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, China.
| |
Collapse
|
43
|
Bris ÁG, MacDowell KS, Ulecia-Morón C, Martín-Hernández D, Moreno B, Madrigal JLM, García-Bueno B, Caso JR, Leza JC. Differential regulation of innate immune system in frontal cortex and hippocampus in a "double-hit" neurodevelopmental model in rats. Neurotherapeutics 2024; 21:e00300. [PMID: 38241165 PMCID: PMC10903097 DOI: 10.1016/j.neurot.2023.10.010] [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: 10/17/2023] [Accepted: 10/28/2023] [Indexed: 01/21/2024] Open
Abstract
Neurodevelopmental disorders (NDs) are neuropsychiatric conditions affecting central nervous system development, characterized by cognitive and behavioural alterations. Inflammation has been recently linked to NDs. Animal models are essential for understanding their pathophysiology and identifying therapeutic targets. Double-hit models can reproduce neurodevelopmental and neuroinflammatory impairments. Sixty-seven newborn rats were assigned to four groups: Control, Maternal deprivation (MD, 24-h-deprivation), Isolation (Iso, 5 weeks), and Maternal deprivation + Isolation (MD + Iso, also known as double-hit). Cognitive dysfunction was assessed using behavioural tests. Inflammasome, MAPKs, and TLRs inflammatory elements expression in the frontal cortex (FC) and hippocampus (HP) was analysed through western blot and qRT-PCR. Oxidative/nitrosative (O/N) evaluation and corticosterone levels were measured in plasma samples. Double-hit group was affected in executive and working memory. Most inflammasomes and TLRs inflammatory responses were increased in FC compared to the control group, whilst MAPKs were downregulated. Conversely, hippocampal inflammasome and inflammatory components were reduced after the double-hit exposure, while MAPKs were elevated. Our findings reveal differential regulation of innate immune system components in FC and HP in the double-hit group. Further investigations on MAPKs are necessary to understand their role in regulating HP neuroinflammatory status, potentially linking our MAPKs results to cognitive impairments through their proliferative and anti-inflammatory activity.
Collapse
Affiliation(s)
- Álvaro G Bris
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid. Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III (CIBERSAM, ISCIII). Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12) e Instituto Universitario de Investigación en Neuroquímica (IUIN), Spain
| | - Karina S MacDowell
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid. Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III (CIBERSAM, ISCIII). Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12) e Instituto Universitario de Investigación en Neuroquímica (IUIN), Spain
| | - Cristina Ulecia-Morón
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid. Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III (CIBERSAM, ISCIII). Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12) e Instituto Universitario de Investigación en Neuroquímica (IUIN), Spain
| | - David Martín-Hernández
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid. Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III (CIBERSAM, ISCIII). Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12) e Instituto Universitario de Investigación en Neuroquímica (IUIN), Spain
| | - Beatriz Moreno
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid. Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III (CIBERSAM, ISCIII). Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12) e Instituto Universitario de Investigación en Neuroquímica (IUIN), Spain
| | - José L M Madrigal
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid. Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III (CIBERSAM, ISCIII). Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12) e Instituto Universitario de Investigación en Neuroquímica (IUIN), Spain
| | - Borja García-Bueno
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid. Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III (CIBERSAM, ISCIII). Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12) e Instituto Universitario de Investigación en Neuroquímica (IUIN), Spain
| | - Javier R Caso
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid. Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III (CIBERSAM, ISCIII). Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12) e Instituto Universitario de Investigación en Neuroquímica (IUIN), Spain
| | - Juan C Leza
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense de Madrid. Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III (CIBERSAM, ISCIII). Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12) e Instituto Universitario de Investigación en Neuroquímica (IUIN), Spain.
| |
Collapse
|
44
|
Lv Y, Wen L, Hu WJ, Deng C, Ren HW, Bao YN, Su BW, Gao P, Man ZY, Luo YY, Li CJ, Xiang ZX, Wang B, Luan ZL. Schizophrenia in the genetic era: a review from development history, clinical features and genomic research approaches to insights of susceptibility genes. Metab Brain Dis 2024; 39:147-171. [PMID: 37542622 DOI: 10.1007/s11011-023-01271-x] [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: 04/20/2023] [Accepted: 07/27/2023] [Indexed: 08/07/2023]
Abstract
Schizophrenia is a devastating neuropsychiatric disorder affecting 1% of the world population and ranks as one of the disorders providing the most severe burden for society. Schizophrenia etiology remains obscure involving multi-risk factors, such as genetic, environmental, nutritional, and developmental factors. Complex interactions of genetic and environmental factors have been implicated in the etiology of schizophrenia. This review provides an overview of the historical origins, pathophysiological mechanisms, diagnosis, clinical symptoms and corresponding treatment of schizophrenia. In addition, as schizophrenia is a polygenic, genetic disorder caused by the combined action of multiple micro-effective genes, we further detail several approaches, such as candidate gene association study (CGAS) and genome-wide association study (GWAS), which are commonly used in schizophrenia genomics studies. A number of GWASs about schizophrenia have been performed with the hope to identify novel, consistent and influential risk genetic factors. Finally, some schizophrenia susceptibility genes have been identified and reported in recent years and their biological functions are also listed. This review may serve as a summary of past research on schizophrenia genomics and susceptibility genes (NRG1, DISC1, RELN, BDNF, MSI2), which may point the way to future schizophrenia genetics research. In addition, depending on the above discovery of susceptibility genes and their exact function, the development and application of antipsychotic drugs will be promoted in the future.
Collapse
Affiliation(s)
- Ye Lv
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Lin Wen
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Wen-Juan Hu
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Chong Deng
- Department of Neurosurgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, 116027, China
| | - Hui-Wen Ren
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Ya-Nan Bao
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Bo-Wei Su
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Ping Gao
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Zi-Yue Man
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Yi-Yang Luo
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Cheng-Jie Li
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Zhi-Xin Xiang
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Bing Wang
- Department of Endocrinology and Metabolism, The Central hospital of Dalian University of Technology, Dalian, 116000, China.
| | - Zhi-Lin Luan
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China.
| |
Collapse
|
45
|
Tranter MM, Faget L, Hnasko TS, Powell SB, Dillon DG, Barnes SA. Postnatal Phencyclidine-Induced Deficits in Decision Making Are Ameliorated by Optogenetic Inhibition of Ventromedial Orbitofrontal Cortical Glutamate Neurons. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2024; 4:264-274. [PMID: 38298783 PMCID: PMC10829674 DOI: 10.1016/j.bpsgos.2023.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 07/12/2023] [Accepted: 08/01/2023] [Indexed: 02/02/2024] Open
Abstract
Background The orbitofrontal cortex (OFC) is essential for decision making, and functional disruptions within the OFC are evident in schizophrenia. Postnatal phencyclidine (PCP) administration in rats is a neurodevelopmental manipulation that induces schizophrenia-relevant cognitive impairments. We aimed to determine whether manipulating OFC glutamate cell activity could ameliorate postnatal PCP-induced deficits in decision making. Methods Male and female Wistar rats (n = 110) were administered saline or PCP on postnatal days 7, 9, and 11. In adulthood, we expressed YFP (yellow fluorescent protein) (control), ChR2 (channelrhodopsin-2) (activation), or eNpHR 3.0 (enhanced halorhodopsin) (inhibition) in glutamate neurons within the ventromedial OFC (vmOFC). Rats were tested on the probabilistic reversal learning task once daily for 20 days while we manipulated the activity of vmOFC glutamate cells. Behavioral performance was analyzed using a Q-learning computational model of reinforcement learning. Results Compared with saline-treated rats expressing YFP, PCP-treated rats expressing YFP completed fewer reversals, made fewer win-stay responses, and had lower learning rates. We induced similar performance impairments in saline-treated rats by activating vmOFC glutamate cells (ChR2). Strikingly, PCP-induced performance deficits were ameliorated when the activity of vmOFC glutamate cells was inhibited (halorhodopsin). Conclusions Postnatal PCP-induced deficits in decision making are associated with hyperactivity of vmOFC glutamate cells. Thus, normalizing vmOFC activity may represent a potential therapeutic target for decision-making deficits in patients with schizophrenia.
Collapse
Affiliation(s)
- Michael M. Tranter
- Department of Psychiatry, University of California San Diego, La Jolla, California
- Research Service, VA San Diego Healthcare System, La Jolla, California
| | - Lauren Faget
- Department of Neurosciences, University of California San Diego, La Jolla, California
| | - Thomas S. Hnasko
- Research Service, VA San Diego Healthcare System, La Jolla, California
- Department of Neurosciences, University of California San Diego, La Jolla, California
| | - Susan B. Powell
- Department of Psychiatry, University of California San Diego, La Jolla, California
- Research Service, VA San Diego Healthcare System, La Jolla, California
| | - Daniel G. Dillon
- Center for Depression, Anxiety and Stress Research, McLean Hospital, Belmont, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Samuel A. Barnes
- Department of Psychiatry, University of California San Diego, La Jolla, California
- Research Service, VA San Diego Healthcare System, La Jolla, California
| |
Collapse
|
46
|
Sahay S, Henkel ND, Vargas CFA, McCullumsmith RE, O’Donovan SM. Activity of Protein Kinase A in the Frontal Cortex in Schizophrenia. Brain Sci 2023; 14:13. [PMID: 38248228 PMCID: PMC10813263 DOI: 10.3390/brainsci14010013] [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: 10/01/2023] [Revised: 11/21/2023] [Accepted: 12/02/2023] [Indexed: 01/23/2024] Open
Abstract
Schizophrenia is a serious cognitive disorder characterized by disruptions in neurotransmission, a process requiring the coordination of multiple kinase-mediated signaling events. Evidence suggests that the observed deficits in schizophrenia may be due to imbalances in kinase activity that propagate through an intracellular signaling network. Specifically, 3'-5'-cyclic adenosine monophosphate (cAMP)-associated signaling pathways are coupled to the activation of neurotransmitter receptors and modulate cellular functions through the activation of protein kinase A (PKA), an enzyme whose function is altered in the frontal cortex in schizophrenia. In this study, we measured the activity of PKA in human postmortem anterior cingulate cortex (ACC) and dorsolateral prefrontal cortex (DLPFC) tissue from schizophrenia and age- and sex-matched control subjects. No significant differences in PKA activity were observed in male and female individuals in either brain region; however, correlation analyses indicated that PKA activity in the ACC may be influenced by tissue pH in all subjects and by age and tissue pH in females. Our data provide novel insights into the function of PKA in the ACC and DLPFC in schizophrenia.
Collapse
Affiliation(s)
- Smita Sahay
- Department of Neurosciences, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; (S.S.); (N.D.H.); (C.F.-A.V.); (R.E.M.)
| | - Nicholas Daniel Henkel
- Department of Neurosciences, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; (S.S.); (N.D.H.); (C.F.-A.V.); (R.E.M.)
| | - Christina Flora-Anabelle Vargas
- Department of Neurosciences, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; (S.S.); (N.D.H.); (C.F.-A.V.); (R.E.M.)
| | - Robert Erne McCullumsmith
- Department of Neurosciences, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; (S.S.); (N.D.H.); (C.F.-A.V.); (R.E.M.)
- Neuroscience Institute, Promedica, Toledo, OH 43606, USA
| | - Sinead Marie O’Donovan
- Department of Neurosciences, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; (S.S.); (N.D.H.); (C.F.-A.V.); (R.E.M.)
| |
Collapse
|
47
|
Kyselicová K, Dukonyová D, Belica I, Ballová DS, Jankovičová V, Ostatníková D. Fingerprint patterns in relation to an altered neurodevelopment in patients with autism spectrum disorder. Dev Psychobiol 2023; 65:e22432. [PMID: 38010306 DOI: 10.1002/dev.22432] [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: 02/20/2023] [Revised: 09/28/2023] [Accepted: 09/29/2023] [Indexed: 11/29/2023]
Abstract
Dermatoglyphic patterns are permanently established and matured before the 24th week of gestation. Their frequencies and localization might be a good indicator of developmental instability in individuals with an altered neurodevelopment and show potential as biomarkers of autism spectrum disorder (ASD). In this study, fingerprint pattern counts and fluctuating asymmetry in the distribution of patterns are compared between 67 boys diagnosed with ASD (aged 5.11 ± 2.51 years) and 83 control boys (aged 8.58 ± 3.14 years). Boys with ASD had a higher rate of discordance in their fingerprint patterns (p = .0026), showing more often bilateral differences in the occurrence of certain patterns. A chi-square test revealed that the difference in pattern frequencies between boys with ASD and the control group is the most significant in frequencies of whorls, tented arches, and ulnar loops. Boys with ASD have significantly fewer ulnar loops, significantly more whorls, and tented arches in the right hand. The achieved results are in favor of the suggestion that prenatal influences, which play a role in the development of bilateral differences in fingerprint patterns up to the 24th week of gestation, may be a potential cause of an altered neurodevelopment in ASD individuals.
Collapse
Affiliation(s)
- Klaudia Kyselicová
- Academic Research Center for Autism, Institute of Physiology, Faculty of Medicine, Comenius University Bratislava, Bratislava, Slovakia
| | - Dóra Dukonyová
- Academic Research Center for Autism, Institute of Physiology, Faculty of Medicine, Comenius University Bratislava, Bratislava, Slovakia
| | - Ivan Belica
- Academic Research Center for Autism, Institute of Physiology, Faculty of Medicine, Comenius University Bratislava, Bratislava, Slovakia
| | - Dominika Sónak Ballová
- Faculty of Civil Engineering, Slovak University of Technology in Bratislava, Bratislava, Slovakia
| | - Viktória Jankovičová
- Faculty of Natural Sciences, Comenius University Bratislava, Bratislava, Slovakia
| | - Daniela Ostatníková
- Academic Research Center for Autism, Institute of Physiology, Faculty of Medicine, Comenius University Bratislava, Bratislava, Slovakia
| |
Collapse
|
48
|
Tsapakis EM, Mitkani CA, Fountoulakis KN. Neurological soft signs and schizophrenia. CNS Spectr 2023; 28:657-661. [PMID: 36924179 DOI: 10.1017/s1092852923001189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Abstract
Neurological soft signs (NSS) are likely to represent abnormal neurodevelopment and aberration in neural maturation and connectivity. They may not be unique to schizophrenia, but they appear to be a trait characteristic in psychosis and therefore could serve as an objective measure for the assessment of serious psychiatric disorder in the prodromal phase, at onset, and along the course of the disease. Evidence so far proposes that NSS are independent of antipsychotic treatment and therefore constitute a trait symptom, independent of the illness stage and medication. Somatomotor and somatosensory regions, spatial orientation, and visual processing areas, cerebellum, and basal ganglia are implicated as possible structural substrates of NSS. Several studies have examined the relationship between NSS and schizophrenia positive, negative symptoms and deficit syndrome; however, results have been so far ambiguous. Neurocognitive symptoms have been moderately related to NSS suggesting that neurocognitive deficits may contribute to the construct of NSS. Regardless of the fact that NSS are not unique to schizophrenia but extend across to the schizotypy continuum, they may help identify individuals at risk of developing schizophrenia later in life.
Collapse
Affiliation(s)
- Eva-Maria Tsapakis
- Agios Charalampos Mental Health Clinic, Heraklion, Greece
- Department of Psychiatry, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Calypso A Mitkani
- Department of Neurology, Agios Pavlos General Hospital of Thessaloniki, Kalamaria, Greece
| | - Konstantinos N Fountoulakis
- Department of Psychiatry, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
49
|
Yoshida Y, Oikawa M, Shimada T, Shinomiya A, Watanabe Y. Neonatal malnutrition impacts fibroblast growth factor 21-induced neuron neurite outgrowth and growth hormone-releasing hormone secretion in neonatal mouse brain. Biochem Biophys Res Commun 2023; 676:121-131. [PMID: 37506473 DOI: 10.1016/j.bbrc.2023.07.041] [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: 06/07/2023] [Revised: 07/12/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023]
Abstract
Neonatal malnutrition is one of the most common causes of neurological disorders. However, the mechanism of action of the factors associated with neonatal nutrition in the brain remains unclear. In this study, we focused on fibroblast growth factor (FGF) 21 to elucidate the effects of malnutrition on the neonatal brain. FGF21 is an endocrine factor produced by the liver during lactation which is the main source of nutrition during the neonatal period. In this study, malnourishment during nursing mice induced decreased levels of Fgf21 mRNA in the liver and decreased levels of FGF21 in the serum. RNA-seq analysis of neonatal mouse brain tissue revealed that FGF21 controlled the expression of Kalrn-201 in the neonatal mouse brain. Kalrn-201 is a transcript of Kalirin, a Ras homologous guanine nucleotide exchange factor at the synapse. In mouse neurons, FGF21 induced the expression of Kalirin-7 (a Kalirin isoform) by down-regulating Kalrn-201. FGF21-induced Kalirin-7 stimulated neurite outgrowth in Neuro-2a cells. FGF21 also induced Growth hormone-releasing hormone (GHRH) expression in Neuro-2a cells. Kalirin-7 and GHRH expression induced by FGF21 was altered by inhibiting the activity of SH2-containing tyrosine phosphatase (SHP2) which is located downstream of the FGF receptor (FGFR). Additionally, malnourished nursing induced intron retention of the SHP2 gene (Ptpn11), resulting in the alteration of Kalirin-7 and GHRH expression by FGF21 signaling. Ptpn11 intron retention is suggested to be involved in regulating SHP2 activity. Taken together, these results suggest that FGF21 plays a critical role in the induction of neuronal neurite outgrowth and GHRH secretion in the neonatal brain, and this mechanism is regulated by SHP2. Thus, Ptpn11 intron retention induced by malnourished nursing may be involved in SHP2 activity.
Collapse
Affiliation(s)
- Yuko Yoshida
- Faculty of Pharmacy, Musashino University, Tokyo, 202-8585, Japan; Research Institute of Pharmaceutical Sciences, Musashino University, Tokyo, 202-8585, Japan.
| | - Mana Oikawa
- Faculty of Pharmacy, Musashino University, Tokyo, 202-8585, Japan
| | - Taiga Shimada
- Faculty of Pharmacy, Musashino University, Tokyo, 202-8585, Japan
| | - Ai Shinomiya
- Faculty of Pharmacy, Musashino University, Tokyo, 202-8585, Japan
| | - Yoshifumi Watanabe
- Faculty of Pharmacy, Musashino University, Tokyo, 202-8585, Japan; Research Institute of Pharmaceutical Sciences, Musashino University, Tokyo, 202-8585, Japan
| |
Collapse
|
50
|
Eichenauer H, Ehlert U. The association between prenatal famine, DNA methylation and mental disorders: a systematic review and meta-analysis. Clin Epigenetics 2023; 15:152. [PMID: 37716973 PMCID: PMC10505322 DOI: 10.1186/s13148-023-01557-y] [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: 05/02/2023] [Accepted: 08/14/2023] [Indexed: 09/18/2023] Open
Abstract
BACKGROUND Undernutrition in pregnant women is an unfavorable environmental condition that can affect the intrauterine development via epigenetic mechanisms and thus have long-lasting detrimental consequences for the mental health of the offspring later in life. One epigenetic mechanism that has been associated with mental disorders and undernutrition is alterations in DNA methylation. The effect of prenatal undernutrition on the mental health of adult offspring can be analyzed through quasi-experimental studies such as famine studies. The present systematic review and meta-analysis aims to analyze the association between prenatal famine exposure, DNA methylation, and mental disorders in adult offspring. We further investigate whether altered DNA methylation as a result of prenatal famine exposure is prospectively linked to mental disorders. METHODS We conducted a systematic search of the databases PubMed and PsycINFO to identify relevant records up to September 2022 on offspring whose mothers experienced famine directly before and/or during pregnancy, examining the impact of prenatal famine exposure on the offspring's DNA methylation and/or mental disorders or symptoms. RESULTS The systematic review showed that adults who were prenatally exposed to famine had an increased risk of schizophrenia and depression. Several studies reported an association between prenatal famine exposure and hyper- or hypomethylation of specific genes. The largest number of studies reported differences in DNA methylation of the IGF2 gene. Altered DNA methylation of the DUSP22 gene mediated the association between prenatal famine exposure and schizophrenia in adult offspring. Meta-analysis confirmed the increased risk of schizophrenia following prenatal famine exposure. For DNA methylation, meta-analysis was not suitable due to different microarrays/data processing approaches and/or unavailable data. CONCLUSION Prenatal famine exposure is associated with an increased risk of mental disorders and DNA methylation changes. The findings suggest that changes in DNA methylation of genes involved in neuronal, neuroendocrine, and immune processes may be a mechanism that promotes the development of mental disorders such as schizophrenia and depression in adult offspring. Such findings are crucial given that undernutrition has risen worldwide, increasing the risk of famine and thus also of negative effects on mental health.
Collapse
Affiliation(s)
- Heike Eichenauer
- Clinical Psychology and Psychotherapy, University of Zurich, Binzmühlestrasse 14, 8050, Zurich, Switzerland
| | - Ulrike Ehlert
- Clinical Psychology and Psychotherapy, University of Zurich, Binzmühlestrasse 14, 8050, Zurich, Switzerland.
| |
Collapse
|