1
|
Blasco MB, Nisha Aji K, Ramos-Jiménez C, Leppert IR, Tardif CL, Cohen J, Rusjan PM, Mizrahi R. Synaptic Density in Early Stages of Psychosis and Clinical High Risk. JAMA Psychiatry 2025; 82:171-180. [PMID: 39535765 PMCID: PMC11561726 DOI: 10.1001/jamapsychiatry.2024.3608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 09/09/2024] [Indexed: 11/16/2024]
Abstract
Importance Synaptic dysfunction is involved in schizophrenia pathophysiology. However, whether in vivo synaptic density is reduced in early stages of psychosis, including its high-risk states, remains unclear. Objective To investigate whether synaptic density (synaptic vesicle glycoprotein 2A [SV2A] binding potential) is reduced in first-episode psychosis (FEP) and in clinical high risk (CHR) and investigate the effect of cannabis use on synaptic density and examine its relationship with psychotic symptoms and gray matter microstructure across groups. Design, Setting, and Participants This cross-sectional study was performed in a tertiary care psychiatric hospital from July 2021 to October 2023. Participants were patients with antipsychotic-free or minimally exposed FEP or CHR and healthy controls with a clean urine drug screen (except cannabis). Main Outcomes and Measures Synaptic density was quantified with dynamic 90-minute [18F]SynVesT-1 positron emission tomography (PET) scans across prioritized brain regions of interest (ROIs) delineated in individual magnetic resonance images (MRIs). Cannabis use was confirmed with urine drug screens. Gray matter microstructure was assessed using diffusion-weighted MRI to estimate neurite density. Results A total of 49 participants were included, including 16 patients with FEP (mean [SD] age, 26.1 [4.6] years; 9 males and 7 females), 17 patients at CHR (mean [SD] age, 21.2 [3.5] years; 8 males and 9 females), and 16 healthy controls (mean [SD] age, 23.4 [3.6] years; 7 males and 9 females). Synaptic density was significantly different between groups (F2,273 = 4.02, P = .02, Cohen F = 0.17; ROI: F5,273 = 360.18, P < .01, Cohen F = 2.55) with a group × ROI interaction (F10,273 = 2.67, P < .01, Cohen F = 0.32). Synaptic density was lower in cannabis users (F1,272 = 5.31, P = .02, Cohen F = 0.14). Lower synaptic density across groups was associated with more negative symptoms (Positive and Negative Syndrome Scale negative scores: F1,81 = 4.31, P = .04, Cohen F = 0.23; Scale of Psychosis-Risk Symptoms negative scores: F1,90 = 4.12, P = .04, Cohen F = 0.21). SV2A binding potential was significantly associated with neurite density index (F1,138 = 6.76, P = .01, Cohen F = 0.22). Conclusions and Relevance This study found that synaptic density reductions were present during the early stages of psychosis and its risk states and associated with negative symptoms. The implications of SV2A for negative symptoms in psychosis and CHR warrant further investigation. Future studies should investigate the impact of cannabis use on synaptic density in CHR longitudinally.
Collapse
Affiliation(s)
- M. Belen Blasco
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada
- Douglas Research Centre, Clinical and Translational Sciences Lab, Montreal, Quebec, Canada
| | - Kankana Nisha Aji
- Douglas Research Centre, Clinical and Translational Sciences Lab, Montreal, Quebec, Canada
| | - Christian Ramos-Jiménez
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada
- Douglas Research Centre, Clinical and Translational Sciences Lab, Montreal, Quebec, Canada
| | - Ilana Ruth Leppert
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Christine Lucas Tardif
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
- Department of Biomedical Engineering, McGill University, Montreal, Quebec, Canada
| | - Johan Cohen
- Douglas Mental Health University Institute, McGill University, Verdun, Quebec, Canada
| | - Pablo M. Rusjan
- Douglas Research Centre, Clinical and Translational Sciences Lab, Montreal, Quebec, Canada
- Douglas Mental Health University Institute, McGill University, Verdun, Quebec, Canada
- Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| | - Romina Mizrahi
- Douglas Research Centre, Clinical and Translational Sciences Lab, Montreal, Quebec, Canada
- Douglas Mental Health University Institute, McGill University, Verdun, Quebec, Canada
- Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
2
|
Nisha Aji K, Lalang N, Ramos-Jiménez C, Rahimian R, Mechawar N, Turecki G, Chartrand D, Boileau I, Meyer JH, Rusjan PM, Mizrahi R. Evidence of altered monoamine oxidase B, an astroglia marker, in early psychosis and high-risk state. Mol Psychiatry 2024:10.1038/s41380-024-02816-x. [PMID: 39511452 DOI: 10.1038/s41380-024-02816-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 10/22/2024] [Accepted: 10/25/2024] [Indexed: 11/15/2024]
Abstract
A novel radiotracer, [11C]SL25.1188, targets monoamine oxidase-B (MAO-B) enzyme, found primarily in astrocytes, which metabolizes monoamines (including dopamine), particularly in subcortical regions. Altered astrocyte function in schizophrenia is supported by convergent evidence from post-mortem, genetic, transcriptomic, peripheral and preclinical findings. We aimed to test whether levels of MAO-B, an index of astrocyte function are low in the living brains of early psychosis and their high-risk states. Thirty-eight participants including antipsychotic-free/minimally exposed clinical participants with first-episode psychosis (FEP), clinical high-risk (CHR) individuals and healthy volunteers (HVs) underwent a 90-min positron emission tomography (PET) scan with [11C]SL25.1188, to measure MAO-B VT, an index of MAO-B concentration. Participants were excluded if tested positive on urine drug screen (except for cannabis). This study of 14 FEP (mean[SD] age, 25.7[5.7] years; 6 F), 7 CHR (mean[SD] age, 20.9[3.7] years; 4 F) and 17 HV (mean[SD] age, 31.2[13.9] years; 9 F) demonstrated significant group differences in regional MAO-B VT (F(2,37.42) = 4.56, p = 0.02, Cohen's f = 0.49), controlling for tobacco (F (1,37.42) = 5.37, p = 0.03) and cannabis use (F(1,37.42) = 5.11, p = 0.03) with significantly lower MAO-B VT in CHR compared to HV (Cohen's d = 0.99). We report a significant cannabis effect on MAO-B VT (F(1,39.19) = 12.57, p = 0.001, Cohen's f = 0.57), with a significant group-by-cannabis interaction (F(2,37.30) = 3.82, p = 0.03, Cohen's f = 0.45), indicating lower MAO-B VT in cannabis-using clinical groups. Lower MAO-B VT levels were more robust in striatal than cortical regions, in both clinical groups (F(12,46.84) = 2.08, p = 0.04, Cohen's f = 0.73) and in cannabis users (F(6,46.84) = 6.42, p < 0.001, Cohen's f = 0.91). Lower MAO-B concentration supports astrocyte dysfunction in cannabis-using CHR and FEP clinical populations. Lower MAO-B is consistent with replicated striatal dopamine elevation in psychosis, as well as astrocyte dysfunction in schizophrenia.
Collapse
Affiliation(s)
- Kankana Nisha Aji
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON, Canada
- Clinical and Translational Sciences Lab, Douglas Research Centre, Montreal, QC, Canada
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Nittha Lalang
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON, Canada
- Vertex Pharmaceuticals, Boston, MA, USA
| | - Christian Ramos-Jiménez
- Clinical and Translational Sciences Lab, Douglas Research Centre, Montreal, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Reza Rahimian
- Douglas Mental Health University Institute, McGill Group for Suicide Studies, Verdun, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Naguib Mechawar
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
- Douglas Mental Health University Institute, McGill Group for Suicide Studies, Verdun, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Gustavo Turecki
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
- Douglas Mental Health University Institute, McGill Group for Suicide Studies, Verdun, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Daniel Chartrand
- Department of Anesthesia, McGill University, Montreal, QC, Canada
- Department of Anesthesia, Montreal Neurological Institute, Montreal, QC, Canada
| | - Isabelle Boileau
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Jeffrey H Meyer
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON, Canada
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Pablo M Rusjan
- Clinical and Translational Sciences Lab, Douglas Research Centre, Montreal, QC, Canada.
- Department of Psychiatry, McGill University, Montreal, QC, Canada.
| | - Romina Mizrahi
- Clinical and Translational Sciences Lab, Douglas Research Centre, Montreal, QC, Canada.
- Department of Psychiatry, McGill University, Montreal, QC, Canada.
| |
Collapse
|
3
|
Koskuvi M, Pörsti E, Hewitt T, Räsänen N, Wu YC, Trontti K, McQuade A, Kalyanaraman S, Ojansuu I, Vaurio O, Cannon TD, Lönnqvist J, Therman S, Suvisaari J, Kaprio J, Blurton-Jones M, Hovatta I, Lähteenvuo M, Rolova T, Lehtonen Š, Tiihonen J, Koistinaho J. Genetic contribution to microglial activation in schizophrenia. Mol Psychiatry 2024; 29:2622-2633. [PMID: 38519640 PMCID: PMC11420079 DOI: 10.1038/s41380-024-02529-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 03/06/2024] [Accepted: 03/13/2024] [Indexed: 03/25/2024]
Abstract
Several lines of evidence indicate the involvement of neuroinflammatory processes in the pathophysiology of schizophrenia (SCZ). Microglia are brain resident immune cells responding toward invading pathogens and injury-related products, and additionally, have a critical role in improving neurogenesis and synaptic functions. Aberrant activation of microglia in SCZ is one of the leading hypotheses for disease pathogenesis, but due to the lack of proper human cell models, the role of microglia in SCZ is not well studied. We used monozygotic twins discordant for SCZ and healthy individuals to generate human induced pluripotent stem cell-derived microglia to assess the transcriptional and functional differences in microglia between healthy controls, affected twins and unaffected twins. The microglia from affected twins had increased expression of several common inflammation-related genes compared to healthy individuals. Microglia from affected twins had also reduced response to interleukin 1 beta (IL1β) treatment, but no significant differences in migration or phagocytotic activity. Ingenuity Pathway Analysis (IPA) showed abnormalities related to extracellular matrix signaling. RNA sequencing predicted downregulation of extracellular matrix structure constituent Gene Ontology (GO) terms and hepatic fibrosis pathway activation that were shared by microglia of both affected and unaffected twins, but the upregulation of major histocompatibility complex (MHC) class II receptors was observed only in affected twin microglia. Also, the microglia of affected twins had heterogeneous response to clozapine, minocycline, and sulforaphane treatments. Overall, despite the increased expression of inflammatory genes, we observed no clear functional signs of hyperactivation in microglia from patients with SCZ. We conclude that microglia of the patients with SCZ have gene expression aberrations related to inflammation response and extracellular matrix without contributing to increased microglial activation.
Collapse
Affiliation(s)
- Marja Koskuvi
- Neuroscience Center, University of Helsinki, Helsinki, Finland
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Elina Pörsti
- Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - Tristen Hewitt
- Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - Noora Räsänen
- Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - Ying-Chieh Wu
- Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - Kalevi Trontti
- Neuroscience Center, University of Helsinki, Helsinki, Finland
- SleepWell Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Psychology and Logopedics, University of Helsinki, Helsinki, Finland
| | - Amanda McQuade
- Department of Neurobiology & Behavior, UC Irvine, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, UC Irvine, Irvine, CA, USA
- Institute for Memory Impairments and Neurological Disorders, UC Irvine, Irvine, CA, USA
| | | | - Ilkka Ojansuu
- Department of Forensic Psychiatry, University of Eastern Finland, Niuvanniemi Hospital, Kuopio, Finland
| | - Olli Vaurio
- Department of Forensic Psychiatry, University of Eastern Finland, Niuvanniemi Hospital, Kuopio, Finland
| | - Tyrone D Cannon
- Department of Psychology and Psychiatry, Yale University, New Haven, CT, USA
| | - Jouko Lönnqvist
- Mental Health Unit, Department of Public Health Solutions, National Institute for Health and Welfare, Helsinki, Finland
- Department of Psychiatry, University of Helsinki, Helsinki, Finland
| | - Sebastian Therman
- Mental Health Unit, Department of Public Health Solutions, National Institute for Health and Welfare, Helsinki, Finland
| | - Jaana Suvisaari
- Mental Health Unit, Department of Public Health Solutions, National Institute for Health and Welfare, Helsinki, Finland
| | - Jaakko Kaprio
- Institute for Molecular Medicine FIMM, University of Helsinki, Helsinki, Finland
| | - Mathew Blurton-Jones
- Department of Neurobiology & Behavior, UC Irvine, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, UC Irvine, Irvine, CA, USA
- Institute for Memory Impairments and Neurological Disorders, UC Irvine, Irvine, CA, USA
| | - Iiris Hovatta
- Neuroscience Center, University of Helsinki, Helsinki, Finland
- SleepWell Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Psychology and Logopedics, University of Helsinki, Helsinki, Finland
| | - Markku Lähteenvuo
- Department of Forensic Psychiatry, University of Eastern Finland, Niuvanniemi Hospital, Kuopio, Finland
| | - Taisia Rolova
- Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - Šárka Lehtonen
- Neuroscience Center, University of Helsinki, Helsinki, Finland
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Jari Tiihonen
- Neuroscience Center, University of Helsinki, Helsinki, Finland
- Department of Forensic Psychiatry, University of Eastern Finland, Niuvanniemi Hospital, Kuopio, Finland
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Jari Koistinaho
- Neuroscience Center, University of Helsinki, Helsinki, Finland.
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
4
|
Gangadin SS, Enthoven AD, van Beveren NJM, Laman JD, Sommer IEC. Immune Dysfunction in Schizophrenia Spectrum Disorders. Annu Rev Clin Psychol 2024; 20:229-257. [PMID: 38996077 DOI: 10.1146/annurev-clinpsy-081122-013201] [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: 07/14/2024]
Abstract
Evidence from epidemiological, clinical, and biological research resulted in the immune hypothesis: the hypothesis that immune system dysfunction is involved in the pathophysiology of schizophrenia spectrum disorders (SSD). The promising implication of this hypothesis is the potential to use existing immunomodulatory treatment for innovative interventions for SSD. Here, we provide a selective historical review of important discoveries that have shaped our understanding of immune dysfunction in SSD. We first explain the basic principles of immune dysfunction, after which we travel more than a century back in time. Starting our journey with neurosyphilis-associated psychosis in the nineteenth century, we continue by evaluating the role of infections and autoimmunity in SSD and findings from assessment of immune function using new techniques, such as cytokine levels, microglia density, neuroimaging, and gene expression. Drawing from these findings, we discuss anti-inflammatory interventions for SSD, and we conclude with a look into the future.
Collapse
Affiliation(s)
- S S Gangadin
- Department of Psychiatry, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands;
| | - A D Enthoven
- Department of Psychiatry, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands;
| | - N J M van Beveren
- Department of Neuroscience, Erasmus MC, Rotterdam, The Netherlands
- Parnassia Group for Mental Health Care, The Hague and Rotterdam, The Netherlands
| | - J D Laman
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - I E C Sommer
- Department of Psychiatry, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands;
| |
Collapse
|
5
|
Nisha Aji K, Cisbani G, Weidenauer A, Koppel A, Hafizi S, Da Silva T, Kiang M, Rusjan PM, Bazinet RP, Mizrahi R. Neurofilament light-chain (NfL) and 18 kDa translocator protein in early psychosis and its putative high-risk. Brain Behav Immun Health 2024; 37:100742. [PMID: 38495956 PMCID: PMC10940889 DOI: 10.1016/j.bbih.2024.100742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 12/27/2023] [Accepted: 02/15/2024] [Indexed: 03/19/2024] Open
Abstract
Evidence of elevated peripheral Neurofilament light-chain (NfL) as a biomarker of neuronal injury can be utilized to reveal nonspecific axonal damage, which could reflect altered neuroimmune function. To date, only a few studies have investigated NfL as a fluid biomarker in schizophrenia primarily, though none in its putative prodrome (Clinical High-Risk, CHR) or in untreated first-episode psychosis (FEP). Further, it is unknown whether peripheral NfL is associated with 18 kDa translocator protein (TSPO), a validated neuroimmune marker. In this secondary study, we investigated for the first time (1) serum NfL in early stages of psychosis including CHR and FEP as compared to healthy controls, and (2) examined its association with brain TSPO, using [18F]FEPPA positron emission tomography (PET). Further, in the exploratory analyses, we aimed to assess associations between serum NfL and symptom severity in patient group and cognitive impairment in the combined cohort. A large cohort of 84 participants including 27 FEP (24 antipsychotic-naive), 41 CHR (34 antipsychotic-naive) and 16 healthy controls underwent structural brain MRI and [18F]FEPPA PET scan and their blood samples were obtained and assessed for serum NfL concentrations. We found no significant differences in serum NfL levels across clinical groups, controlling for age. We also found no significant association between NfL levels and brain TSPO in the entire cohort. We observed a negative association between serum NfL and negative symptom severity in CHR. Our findings suggest that neither active neuroaxonal deterioration as measured with NfL nor associated neuroimmune activation (TSPO) is clearly identifiable in an early mostly untreated psychosis sample including its putative high-risk.
Collapse
Affiliation(s)
- Kankana Nisha Aji
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada
- Douglas Research Centre, Clinical and Translational Sciences Lab, Montreal, Quebec, Canada
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Giulia Cisbani
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Ana Weidenauer
- Division of General Psychiatry, Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - Alex Koppel
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Sina Hafizi
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Tania Da Silva
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Michael Kiang
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Pablo M. Rusjan
- Douglas Research Centre, Clinical and Translational Sciences Lab, Montreal, Quebec, Canada
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| | - Richard P. Bazinet
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Romina Mizrahi
- Douglas Research Centre, Clinical and Translational Sciences Lab, Montreal, Quebec, Canada
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
6
|
Liu J, Liu TT, Mou L, Zhang Y, Chen X, Wang Q, Deng BL, Liu J. P2X7 receptor: a potential target for treating comorbid anxiety and depression. Purinergic Signal 2024:10.1007/s11302-024-10007-0. [PMID: 38642324 DOI: 10.1007/s11302-024-10007-0] [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: 01/09/2024] [Accepted: 04/03/2024] [Indexed: 04/22/2024] Open
Abstract
In clinical practice, depression and anxiety frequently coexist, and they are both comorbid with somatic diseases. The P2X7R is an adenosine 5'-triphosphate (ATP)-gated non-selective cation channel that is widely expressed in immune-related cells. Under conditions of stress, chronic pain, and comorbid chronic physical illness, P2X7R activation in glial cells leads to neuroinflammation. This could contribute to the development of anxiety and depression-related emotional disturbances. Previous studies have shown that the P2X7 receptor (P2X7R) plays an important role in the pathogenesis of both anxiety and depression. Thus, the P2X7R may play a role in the comorbidity of anxiety and depression. Positron emission tomography can be used to assess the degree and location of neuroinflammation by monitoring functional and expression-related changes in P2X7R, which can facilitate clinical diagnoses and guide the treatment of patients with anxiety and depression. Moreover, single nucleotide polymorphisms (SNPs) in the P2X7R gene are associated with susceptibility to different types of psychiatric disorders. Thus, evaluating the SNPs of the P2X7R gene could enable personalized mood disorder diagnoses and treatments. If the P2X7R were set as a therapeutic target, selective P2X7R antagonists may modulate P2X7R function, thereby altering the balance of intra- and extra-cellular ATP. This could have therapeutic implications for treating anxiety and depression, as well as for pain management. According to in vitro and in vivo studies, the P2X7R plays an important role in anxiety and depression. In this review, we consider the potential of the P2X7R as a therapeutic target for comorbid anxiety and depression, and discuss the potential diagnostic and therapeutic value of this receptor.
Collapse
Affiliation(s)
- Jun Liu
- Department of Neurology, School of Clinical Medicine, Southwest Medical University, Luzhou, China
- Department of Geriatric Neurology, Qinglongchang Ward, Chengdu Sixth People's Hospital, Chengdu, China
| | - Ting-Ting Liu
- Department of Neurology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Lan Mou
- Department of Neurology, School of Clinical Medicine, Southwest Medical University, Luzhou, China
| | - Yuwen Zhang
- Department of Neurology, School of Clinical Medicine, Southwest Medical University, Luzhou, China
| | - Xiang Chen
- Department of Neurology, School of Clinical Medicine, Southwest Medical University, Luzhou, China
| | - Qi Wang
- Department of Neurology, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, Chengdu, China
| | - Bin-Lu Deng
- Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Jie Liu
- Department of Neurology, School of Clinical Medicine, Southwest Medical University, Luzhou, China.
- Department of Neurology, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, Chengdu, China.
- Department of Neurology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
7
|
Liu D, Jin Z, Wei H, Zhu C, Liu K, You P, Ju J, Xu J, Zhu W, Xu Q. Anti-SFT2D2 autoantibodies alter dendrite spine and cause psychotic behavior in mice. J Psychiatr Res 2024; 171:99-107. [PMID: 38262166 DOI: 10.1016/j.jpsychires.2024.01.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/06/2023] [Accepted: 01/09/2024] [Indexed: 01/25/2024]
Abstract
BACKGROUND Autoimmunity plays an important role in schizophrenia (SCZ). Autoantibodies against SFT2D2 have been reported in patients with SCZ; however, the specific mechanism remains unclear. This study aimed to describe an autoimmune model, namely, mice immunized against SFT2D2-peptides. METHODS ApoE-/- and WT mice (C57BL/6) were immunized four times (day 0, day 14, day 21, day 35) with SFT2D2 peptide or KLH via subcutaneous injection. Behavioral tests were conducted after the third immunization, and immunochemistry of brain tissue were performed after the sacrifice of the mice. RESULTS Active immunization with KLH-coupled SFT2D2-derived peptides in both WT and ApoE-/- (compromised blood-brain barrier) mice led to high circulating levels of anti-SFT2D2 IgG. While there was no detectable deficit in WT mice, impaired pre-pulse inhibition, motor impairments, and reduced cognition in ApoE-/- mice, without signs of anxiety and depression were observed. In addition, immunohistochemical assays demonstrated that activated microglia and astrocytes were increased but neuronal dendritic spine densities were decreased, accompanied by increased expression of complement molecule C4 across brain regions in ApoE-/- mice. CONCLUSIONS In model mice with compromised blood-brain barrier, endogenous anti-SFT2D2 IgG can activate glial cells and modulate synaptic plasticity, and induce a series of psychosis-like changes. These antibodies may reveal valuable therapeutic targets, which may improve the treatment strategies for a subgroup of SCZ patients.
Collapse
Affiliation(s)
- Duilin Liu
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Zhongman Jin
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Hui Wei
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China; Neuroscience Center, Chinese Academy of Medical Sciences, Beijing, China
| | - Caiyun Zhu
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China; Neuroscience Center, Chinese Academy of Medical Sciences, Beijing, China
| | - Kejiang Liu
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Pengsheng You
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Jiahang Ju
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Jinming Xu
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Wanwan Zhu
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China; Neuroscience Center, Chinese Academy of Medical Sciences, Beijing, China
| | - Qi Xu
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China; Neuroscience Center, Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
8
|
Gangadin SS, Mandl RCW, de Witte LD, van Haren NEM, Schutte MJL, Begemann MJH, Kahn RS, Sommer IEC. Lower fractional anisotropy without evidence for neuro-inflammation in patients with early-phase schizophrenia spectrum disorders. Schizophr Res 2024; 264:557-566. [PMID: 36577563 DOI: 10.1016/j.schres.2022.12.009] [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: 06/09/2022] [Revised: 12/05/2022] [Accepted: 12/07/2022] [Indexed: 12/28/2022]
Abstract
Various lines of research suggest immune dysregulation as a potential therapeutic target for negative and cognitive symptoms in schizophrenia spectrum disorders (SSD). Immune dysregulation would lead to higher extracellular free-water (EFW) in cerebral white matter (WM), which may partially underlie the frequently reported lower fractional anisotropy (FA) in SSD. We aim to investigate differences in EFW concentrations - a presumed proxy for neuro-inflammation - between early-phase SSD patients (n = 55) and healthy controls (HC; n = 37), and to explore immunological and cognitive correlates. To increase specificity for EFW, we study several complementary magnetic resonance imaging contrasts that are sensitive to EFW. FA, mean diffusivity (MD), magnetization transfer ratio (MTR), myelin water fraction (MWF) and quantitative T1 and T2 were calculated from diffusion-weighted imaging (DWI), magnetization transfer imaging (MTI) and multicomponent driven equilibrium single-pulse observation of T1/T2 (mcDESPOT). For each measure, WM skeletons were constructed with tract-based spatial statistics. Multivariate SSD-HC comparisons with WM skeletons and their average values (i.e. global WM) were not statistically significant. In voxel-wise analyses, FA was significantly lower in SSD in the genu of the corpus callosum and in the left superior longitudinal fasciculus (p < 0.04). Global WM measures did not correlate with immunological markers (i.e. IL1-RA, IL-6, IL-8, IL-10 and CRP) or cognition in HC and SSD after corrections for multiple comparisons. We confirmed lower FA in early-phase SSD patients. However, nonFA measures did not provide additional evidence for immune dysregulation or for higher EFW as the primary mechanism underlying the reported lower FA values in SSD.
Collapse
Affiliation(s)
- Shiral S Gangadin
- Section Cognitive Neuroscience, Department of Biomedical Sciences of Cells & Systems, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| | - René C W Mandl
- Section Cognitive Neuroscience, Department of Biomedical Sciences of Cells & Systems, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Lot D de Witte
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York City, NY, USA.
| | - Neeltje E M van Haren
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus University Medical Center-Sophia Children's Hospital, Rotterdam, the Netherlands.
| | - Maya J L Schutte
- Section Cognitive Neuroscience, Department of Biomedical Sciences of Cells & Systems, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| | - Marieke J H Begemann
- Section Cognitive Neuroscience, Department of Biomedical Sciences of Cells & Systems, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| | - René S Kahn
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York City, NY, USA.
| | - Iris E C Sommer
- Section Cognitive Neuroscience, Department of Biomedical Sciences of Cells & Systems, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| |
Collapse
|
9
|
Qurashi I, Chaudhry IB, Khoso AB, Omair Husain M, Hafeez D, Kiran T, Lane S, Naqvi HA, Minhas FA, Tamizuddin Nizami A, Razzaque B, Qambar Bokhari S, Yung AR, Deakin B, Husain N. A randomised double-blind placebo-controlled trial of minocycline and/or omega-3 fatty acids added to treatment as usual for at risk Mental States: The NAYAB study. Brain Behav Immun 2024; 115:609-616. [PMID: 37924960 DOI: 10.1016/j.bbi.2023.10.025] [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: 03/05/2023] [Revised: 10/03/2023] [Accepted: 10/28/2023] [Indexed: 11/06/2023] Open
Abstract
BACKGROUND Inflammatory mechanisms are thought to contribute to the onset of psychosis in persons with an at-risk mental state (ARMS). We investigated whether the anti-inflammatory properties of minocycline and omega-3 polyunsaturated fatty acids (omega-3), alone or synergistically, would prevent transition to psychosis in ARMS in a randomised, double-blind, placebo-controlled trial in Pakistan. METHODS 10,173 help-seeking individuals aged 16-35 years were screened using the Prodromal Questionaire-16. Individuals scoring 6 and over were interviewed using the Comprehensive Assessment of At-Risk Mental States (CAARMS) to confirm ARMS. Participants (n = 326) were randomised to minocycline, omega-3, combined minocycline and omega-3 or to double placebo for 6 months. The primary outcome was transition to psychosis at 12 months. FINDINGS Forty-five (13.8 %) participants transitioned to psychosis. The risk of transition was greater in those randomised to omega-3 alone or in combination with minocycline (17.3.%), compared to 10.4 % in those not exposed to omega-3; a risk-ratio (RR) of 1.67, 95 % CI [0.95, 2.92] p = 0.07. The RR for transitions on minocycline vs. no minocycline was 0.86, 95 % CI [0.50, 1.49] p > 0.10. In participants who did not become psychotic, CAARMS and depression symptom scores were reduced at six and twelve months (mean CAARMS difference = 1.43; 95 % CI [0.33, 1.76] p < 0.01 in those exposed to omega-3. Minocycline did not affect CAARMS or depression scores. INTERPRETATION In keeping with other studies, omega-3 appears to have beneficial effects on ARMS and mood symptom severity but it increased transition to psychosis, which may reflect metabolic or developmental consequences of chronic poor nutrition in the population. Transition to psychosis was too rare to reveal a preventative effect of minocycline but minocycline did not improve symptom severity. ARMS symptom severity and transition to psychosis appear to have distinct pathogeneses which are differentially modulated by omega-3 supplementation. FUNDING The study was funded by the Stanley Research Medical Institute.
Collapse
Affiliation(s)
- Inti Qurashi
- Institute of Population Health, University of Liverpool, Liverpool, UK; Mersey Care NHS Foundation Trust, Prescott, UK
| | - Imran B Chaudhry
- Division of Psychology and Mental Health, University of Manchester, Manchester, UK; Department of Psychiatry, Ziauddin University, Karachi, Pakistan; Pakistan Institute of Living and Learning, Karachi, Pakistan
| | - Ameer B Khoso
- Pakistan Institute of Living and Learning, Karachi, Pakistan
| | - Muhammad Omair Husain
- Centre for Addiction and Mental Health, Toronto, Canada; Department of Psychiatry, University of Toronto, Toronto, Canada
| | | | - Tayyeba Kiran
- Pakistan Institute of Living and Learning, Karachi, Pakistan
| | - Steven Lane
- Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Haider A Naqvi
- Department of Psychiatry, Dow University of Health Sciences, Karachi, Pakistan
| | - Fareed A Minhas
- Institute of Psychiatry, Rawalpindi Medical University, Rawalpindi, Pakistan
| | | | - Bushra Razzaque
- Institute of Psychiatry, Rawalpindi Medical University, Rawalpindi, Pakistan
| | - Sumira Qambar Bokhari
- Department of Psychiatry & Behavioural Sciences, Services Institute of Medical Sciences, Lahore, Pakistan
| | - Alison R Yung
- Centre for Youth Mental Health, The University of Melbourne, Parkville, Victoria, Australia; Orygen, Parkville, Victoria, Australia
| | - Bill Deakin
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, University of Manchester, Manchester, UK.
| | - Nusrat Husain
- Division of Psychology and Mental Health, University of Manchester, Manchester, UK; Mersey Care NHS Foundation Trust, Prescott, UK
| |
Collapse
|
10
|
Matuleviciute R, Akinluyi ET, Muntslag TAO, Dewing JM, Long KR, Vernon AC, Tremblay ME, Menassa DA. Microglial contribution to the pathology of neurodevelopmental disorders in humans. Acta Neuropathol 2023; 146:663-683. [PMID: 37656188 PMCID: PMC10564830 DOI: 10.1007/s00401-023-02629-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 08/25/2023] [Accepted: 08/26/2023] [Indexed: 09/02/2023]
Abstract
Microglia are the brain's resident macrophages, which guide various developmental processes crucial for brain maturation, activity, and plasticity. Microglial progenitors enter the telencephalic wall by the 4th postconceptional week and colonise the fetal brain in a manner that spatiotemporally tracks key neurodevelopmental processes in humans. However, much of what we know about how microglia shape neurodevelopment comes from rodent studies. Multiple differences exist between human and rodent microglia warranting further focus on the human condition, particularly as microglia are emerging as critically involved in the pathological signature of various cognitive and neurodevelopmental disorders. In this article, we review the evidence supporting microglial involvement in basic neurodevelopmental processes by focusing on the human species. We next concur on the neuropathological evidence demonstrating whether and how microglia contribute to the aetiology of two neurodevelopmental disorders: autism spectrum conditions and schizophrenia. Next, we highlight how recent technologies have revolutionised our understanding of microglial biology with a focus on how these tools can help us elucidate at unprecedented resolution the links between microglia and neurodevelopmental disorders. We conclude by reviewing which current treatment approaches have shown most promise towards targeting microglia in neurodevelopmental disorders and suggest novel avenues for future consideration.
Collapse
Affiliation(s)
- Rugile Matuleviciute
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Elizabeth T Akinluyi
- Division of Medical Sciences, University of Victoria, Victoria, Canada
- Department of Pharmacology and Therapeutics, Afe Babalola University, Ado Ekiti, Nigeria
| | - Tim A O Muntslag
- Princess Maxima Centre for Paediatric Oncology, Utrecht, The Netherlands
| | | | - Katherine R Long
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Anthony C Vernon
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | | | - David A Menassa
- Department of Neuropathology & The Queen's College, University of Oxford, Oxford, UK.
- Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden.
| |
Collapse
|
11
|
Klein HC, Guest PC, Dobrowolny H, Steiner J. Inflammation and viral infection as disease modifiers in schizophrenia. Front Psychiatry 2023; 14:1231750. [PMID: 37850104 PMCID: PMC10577328 DOI: 10.3389/fpsyt.2023.1231750] [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: 05/30/2023] [Accepted: 09/12/2023] [Indexed: 10/19/2023] Open
Abstract
Numerous studies have now implicated a role for inflammation in schizophrenia. However, many aspects surrounding this aspect of the disease are still controversial. This controversy has been driven by conflicting evidence on the role of both pro-and anti-inflammatory factors and by often contentious findings concerning cytokine and immune cell profiles in the central nervous system and periphery. Current evidence supports the point that interleukin-6 is elevated in CSF, but does not support activation of microglia, resident macrophage-like cells in the brain. Furthermore, the mechanisms involving transit of the peripheral immune system factors across the blood brain barrier to central parenchyma have still not been completely elucidated. This process appears to involve perivascular macrophages and accompanying dendritic cells retained in the parenchyma by the chemokine and cytokine composition of the surrounding milieu. In addition, a number of studies have shown that this can be modulated by infection with viruses such as herpes simplex virus type I which may disrupt antigen presentation in the perivascular space, with long-lasting consequences. In this review article, we discuss the role of inflammation and viral infection as potential disease modifiers in schizophrenia. The primary viral hit may occur in the fetus in utero, transforming the immune response regulatory T-cells or the virus may secondarily remain latent in immune cells or neurons and modify further immune responses in the developing individual. It is hoped that unraveling this pathway further and solidifying our understanding of the pathophysiological mechanisms involved will pave the way for future studies aimed at identification and implementation of new biomarkers and drug targets. This may facilitate the development of more effective personalized therapies for individuals suffering with schizophrenia.
Collapse
Affiliation(s)
- Hans C. Klein
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
- Research and Education Department Addiction Care Northern Netherlands, Groningen, Netherlands
| | - Paul C. Guest
- Department of Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
- Laboratory of Translational Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Henrik Dobrowolny
- Department of Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
- Laboratory of Translational Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Johann Steiner
- Department of Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
- Laboratory of Translational Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
- Center for Health and Medical Prevention (CHaMP), Magdeburg, Germany
- German Center for Mental Health (DZPG), Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), Halle-Jena-Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
| |
Collapse
|
12
|
De Picker LJ, Morrens M, Branchi I, Haarman BCM, Terada T, Kang MS, Boche D, Tremblay ME, Leroy C, Bottlaender M, Ottoy J. TSPO PET brain inflammation imaging: A transdiagnostic systematic review and meta-analysis of 156 case-control studies. Brain Behav Immun 2023; 113:415-431. [PMID: 37543251 DOI: 10.1016/j.bbi.2023.07.023] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 06/26/2023] [Accepted: 07/30/2023] [Indexed: 08/07/2023] Open
Abstract
INTRODUCTION The 18-kDa translocator protein (TSPO) is increasingly recognized as a molecular target for PET imaging of inflammatory responses in various central nervous system (CNS) disorders. However, the reported sensitivity and specificity of TSPO PET to identify brain inflammatory processes appears to vary greatly across disorders, disease stages, and applied quantification methods. To advance TSPO PET as a potential biomarker to evaluate brain inflammation and anti-inflammatory therapies, a better understanding of its applicability across disorders is needed. We conducted a transdiagnostic systematic review and meta-analysis of all in vivo human TSPO PET imaging case-control studies in the CNS. Specifically, we investigated the direction, strength, and heterogeneity associated with the TSPO PET signal across disorders in pre-specified brain regions, and explored the demographic and methodological sources of heterogeneity. METHODS We searched for English peer-reviewed articles that reported in vivo human case-control TSPO PET differences. We extracted the demographic details, TSPO PET outcomes, and technical variables of the PET procedure. A random-effects meta-analysis was applied to estimate case-control standardized mean differences (SMD) of the TSPO PET signal in the lobar/whole-brain cortical grey matter (cGM), thalamus, and cortico-limbic circuitry between different illness categories. Heterogeneity was evaluated with the I2 statistic and explored using subgroup and meta-regression analyses for radioligand generation, PET quantification method, age, sex, and publication year. Significance was set at the False Discovery Rate (FDR)-corrected P < 0.05. RESULTS 156 individual case-control studies were included in the systematic review, incorporating data for 2381 healthy controls and 2626 patients. 139 studies documented meta-analysable data and were grouped into 11 illness categories. Across all the illness categories, we observed a significantly higher TSPO PET signal in cases compared to controls for the cGM (n = 121 studies, SMD = 0.358, PFDR < 0.001, I2 = 68%), with a significant difference between the illness categories (P = 0.004). cGM increases were only significant for Alzheimer's disease (SMD = 0.693, PFDR < 0.001, I2 = 64%) and other neurodegenerative disorders (SMD = 0.929, PFDR < 0.001, I2 = 73%). Cortico-limbic increases (n = 97 studies, SMD = 0.541, P < 0.001, I2 = 67%) were most prominent for Alzheimer's disease, mild cognitive impairment, other neurodegenerative disorders, mood disorders and multiple sclerosis. Thalamic involvement (n = 79 studies, SMD = 0.393, P < 0.001, I2 = 71%) was observed for Alzheimer's disease, other neurodegenerative disorders, multiple sclerosis, and chronic pain and functional disorders (all PFDR < 0.05). Main outcomes for systemic immunological disorders, viral infections, substance use disorders, schizophrenia and traumatic brain injury were not significant. We identified multiple sources of between-study variance to the TSPO PET signal including a strong transdiagnostic effect of the quantification method (explaining 25% of between-study variance; VT-based SMD = 0.000 versus reference tissue-based studies SMD = 0.630; F = 20.49, df = 1;103, P < 0.001), patient age (9% of variance), and radioligand generation (5% of variance). CONCLUSION This study is the first overarching transdiagnostic meta-analysis of case-control TSPO PET findings in humans across several brain regions. We observed robust increases in the TSPO signal for specific types of disorders, which were widespread or focal depending on illness category. We also found a large and transdiagnostic horizontal (positive) shift of the effect estimates of reference tissue-based compared to VT-based studies. Our results can support future studies to optimize experimental design and power calculations, by taking into account the type of disorder, brain region-of-interest, radioligand, and quantification method.
Collapse
Affiliation(s)
- Livia J De Picker
- Collaborative Antwerp Psychiatric Research Institute (CAPRI), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium; Scientific Initiative of Neuropsychiatric and Psychopharmacological Studies (SINAPS), University Psychiatric Centre Campus Duffel, Duffel, Belgium.
| | - Manuel Morrens
- Collaborative Antwerp Psychiatric Research Institute (CAPRI), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium; Scientific Initiative of Neuropsychiatric and Psychopharmacological Studies (SINAPS), University Psychiatric Centre Campus Duffel, Duffel, Belgium
| | - Igor Branchi
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Roma, Italy
| | - Bartholomeus C M Haarman
- Department of Psychiatry, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Tatsuhiro Terada
- Department of Biofunctional Imaging, Preeminent Medical Photonics Education & Research Center, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Min Su Kang
- LC Campbell Cognitive Neurology Unit, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| | - Delphine Boche
- Clinical Neurosciences, Clinical and Experimental Sciences School, Faculty of Medicine, University of Southampton, UK
| | - Marie-Eve Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada; Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, BC, Canada; Neurology and Neurosurgery Department, McGill University, Montréal, QC, Canada; Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Claire Leroy
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d'Imagerie Biomédicale Multimodale Paris-Saclay (BioMaps), Orsay, France
| | - Michel Bottlaender
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d'Imagerie Biomédicale Multimodale Paris-Saclay (BioMaps), Orsay, France; Université Paris-Saclay, UNIACT, Neurospin, CEA, Gif-sur-Yvette, France
| | - Julie Ottoy
- LC Campbell Cognitive Neurology Unit, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
13
|
Kübler R, Ormel PR, Sommer IEC, Kahn RS, de Witte LD. Gene expression profiling of monocytes in recent-onset schizophrenia. Brain Behav Immun 2023; 111:334-342. [PMID: 37149105 DOI: 10.1016/j.bbi.2023.04.019] [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: 08/04/2022] [Revised: 04/26/2023] [Accepted: 04/30/2023] [Indexed: 05/08/2023] Open
Abstract
Immune-related mechanisms have been suggested to be involved in schizophrenia. Various studies have shown changes in monocytes isolated from the blood of schizophrenia patients, including changes in monocyte numbers, as well as altered protein and transcript levels of important markers. However, validation of these findings and understanding how these results are related to immune-related changes in the brain and schizophrenia genetic risk factors, is limited. The goal of this study was to better understand changes observed in monocytes of patients with early-onset schizophrenia. Using RNA sequencing, we analyzed gene expression profiles of monocytes isolated from twenty patients with early-onset schizophrenia and seventeen healthy controls. We validated expression changes of 7 out of 29 genes that were differentially expressed in previous studies including TNFAIP3, DUSP2, and IL6. At a transcriptome-wide level, we found 99 differentially expressed genes. Effect sizes of differentially expressed genes were moderately correlated with differential expression in brain tissue (Pearson's r = 0.49). Upregulated genes were enriched for genes in NF-κB and LPS signaling pathways. Downregulated genes were enriched for glucocorticoid response pathways. These pathways have been implicated in schizophrenia before and play a role in regulating the activation of myeloid cells. Interestingly, they are also involved in several non-inflammatory processes in the central nervous system, such as neurogenesis and neurotransmission. Future studies are needed to better understand how dysregulation of the NF-κB and glucocorticoid pathways affects inflammatory and non-inflammatory processes in schizophrenia. The fact that dysregulation of these pathways is also seen in brain tissue, provides potential possibilities for biomarker development.
Collapse
Affiliation(s)
- Raphael Kübler
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Paul R Ormel
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, the Netherlands
| | - Iris E C Sommer
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, the Netherlands; Department of Neuroscience, University Medical Center Groningen, Groningen, the Netherlands
| | - René S Kahn
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, the Netherlands
| | - Lot D de Witte
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, the Netherlands.
| |
Collapse
|
14
|
Nisha Aji K, Hafizi S, Da Silva T, Kiang M, Rusjan PM, Weickert CS, Mizrahi R. Interaction between peripheral and central immune markers in clinical high risk for psychosis. Brain Behav Immun Health 2023; 30:100636. [PMID: 37293440 PMCID: PMC10244662 DOI: 10.1016/j.bbih.2023.100636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 05/07/2023] [Indexed: 06/10/2023] Open
Abstract
Neuroinflammatory events prior to the diagnosis of schizophrenia may play a role in transition to illness. To date only one in-vivo study has investigated this association between peripheral proinflammatory cytokines and brain markers of inflammation (e.g., mitochondrial 18 kDa translocator protein, TSPO) in schizophrenia, but none in its putative prodrome. In this study, we primarily aimed to (Barron et al., 2017) test study group (clinical high-risk (CHR) and healthy controls) differences in peripheral inflammatory markers and test for any associations with symptom measures, (Hafizi et al., 2017a) investigate the interaction between brain TSPO levels (dorsolateral prefrontal cortex (DLPFC) and hippocampus) and peripheral inflammatory clusters (entire cohort and (CHR) group independently) within a relatively large group of individuals at CHR for psychosis (N = 38) and healthy controls (N = 20). Participants underwent structural brain magnetic resonance imaging (MRI) and TSPO [18F]FEPPA positron emission tomography (PET) scans. Serum samples were assessed for peripheral inflammatory markers (i.e., CRP and interleukins). For exploratory analysis, we aimed to examine cluster differences for symptom measures and identify independent peripheral predictors of brain TSPO expression. Here, we report increased IL-8 levels that are positively correlated with prodromal general symptom severity and showed trend-level association with apathy in CHR. We identified distinct inflammatory clusters characterized by inflammatory markers (IL-1 β, IL-2, IFN-γ) that were comparable between entire cohort and CHR. TSPO levels did not differ between inflammatory clusters (entire cohort or CHR). Finally, we show that CRP, IL-1 β, TNF-α, and IFN-γ levels were the independent peripheral predictors of brain TSPO expression. Thus, alterations in brain TSPO expression in response to inflammatory processes are not evident in CHR. Taken together, clustering by inflammatory status is a promising strategy to characterize the interaction between brain TSPO and peripheral markers of inflammation.
Collapse
Affiliation(s)
- Kankana Nisha Aji
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Douglas Research Centre, Clinical and Translational Sciences Lab, Montreal, Quebec, Canada
| | - Sina Hafizi
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Department of Psychiatry, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Tania Da Silva
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Michael Kiang
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Pablo M. Rusjan
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Douglas Research Centre, Clinical and Translational Sciences Lab, Montreal, Quebec, Canada
- Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| | | | - Romina Mizrahi
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Douglas Research Centre, Clinical and Translational Sciences Lab, Montreal, Quebec, Canada
- Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
15
|
Serpa M, Doshi J, Joaquim HPG, Vieira ELM, Erus G, Chaim-Avancini TM, Cavallet M, Guglielmi LG, Sallet PC, Talib L, Teixeira AL, van de Bilt MT, McGuire P, Gattaz WF, Davatzikos C, Busatto GF, Zanetti MV. Inflammatory cytokines and white matter microstructure in the acute phase of first-episode psychosis: A longitudinal study. Schizophr Res 2023; 257:5-18. [PMID: 37230043 DOI: 10.1016/j.schres.2023.05.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 01/14/2023] [Accepted: 05/06/2023] [Indexed: 05/27/2023]
Abstract
OBJECTIVES Schizophrenia-related psychosis is associated with abnormalities in white matter (WM) microstructure and structural brain dysconnectivity. However, the pathological process underlying such changes is unknown. We sought to investigate the potential association between peripheral cytokine levels and WM microstructure during the acute phase of first-episode psychosis (FEP) in a cohort of drug-naïve patients. METHODS Twenty-five non-affective FEP patients and 69 healthy controls underwent MRI scanning and blood collection at study entry. After achieving clinical remission, 21 FEP were reassessed; 38 age and biological sex-matched controls also had a second assessment. We measured fractional anisotropy (FA) of selected WM regions-of-interest (ROIs) and plasma levels of four cytokines (IL-6, IL-10, IFN-γ, and TNF-α). RESULTS At baseline (acute psychosis), the FEP group showed reduced FA relative to controls in half the examined ROIs. Within the FEP group, IL-6 levels were negatively correlated with FA values. Longitudinally, patients showed increments of FA in several ROIs affected at baseline, and such changes were associated with reductions in IL-6 levels. CONCLUSIONS A state-dependent process involving an interplay between a pro-inflammatory cytokine and brain WM might be associated with the clinical manifestation of FEP. This association suggests a deleterious effect of IL-6 on WM tracts during the acute phase of psychosis.
Collapse
Affiliation(s)
- Mauricio Serpa
- Laboratory of Psychiatric Neuroimaging (LIM21), Department and Institute of Psychiatry, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil; Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK; Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), Universidade de Sao Paulo, Sao Paulo, SP, Brazil; Laboratory of Neuroscience (LIM27), Department and Institute of Psychiatry, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil.
| | - Jimit Doshi
- Section of Biomedical Image Analysis (SBIA), Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Helena P G Joaquim
- Laboratory of Neuroscience (LIM27), Department and Institute of Psychiatry, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Erica L M Vieira
- Universidade Federal de Minas Gerais, Faculdade de Medicina, Belo Horizonte, MG, Brazil
| | - Guray Erus
- Section of Biomedical Image Analysis (SBIA), Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Tiffany M Chaim-Avancini
- Laboratory of Psychiatric Neuroimaging (LIM21), Department and Institute of Psychiatry, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil; Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Mikael Cavallet
- Laboratory of Psychiatric Neuroimaging (LIM21), Department and Institute of Psychiatry, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Luiza Guilherme Guglielmi
- Laboratory of Immunology, Instituto do Coracao (INCOR), Hospital das Clinicas FMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Paulo C Sallet
- Laboratory of Neuroscience (LIM27), Department and Institute of Psychiatry, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Leda Talib
- Laboratory of Neuroscience (LIM27), Department and Institute of Psychiatry, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Antonio L Teixeira
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Martinus T van de Bilt
- Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), Universidade de Sao Paulo, Sao Paulo, SP, Brazil; Laboratory of Neuroscience (LIM27), Department and Institute of Psychiatry, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Philip McGuire
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK; NIHR Biomedical Research Centre, South London and Maudsley NHS Foundation Trust, London, United Kingdom
| | - Wagner F Gattaz
- Laboratory of Neuroscience (LIM27), Department and Institute of Psychiatry, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Christos Davatzikos
- Section of Biomedical Image Analysis (SBIA), Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Geraldo F Busatto
- Laboratory of Psychiatric Neuroimaging (LIM21), Department and Institute of Psychiatry, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil; Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | | |
Collapse
|
16
|
Foley ÉM, Griffiths SL, Murray A, Rogers J, Corsi-Zuelli F, Hickinbotham H, Warwick E, Wilson M, Kaser M, Murray GK, Deakin B, Jadon D, Suckling J, Barnes NM, Upthegrove R, Khandaker GM. Protocol for the Psychosis Immune Mechanism Stratified Medicine (PIMS) trial: a randomised double-blind placebo-controlled trial of single-dose tocilizumab in patients with psychosis. BMJ Open 2023; 13:e067944. [PMID: 36963796 PMCID: PMC10040013 DOI: 10.1136/bmjopen-2022-067944] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 02/22/2023] [Indexed: 03/26/2023] Open
Abstract
INTRODUCTION Evidence suggests a potentially causal role of interleukin 6 (IL-6), a pleiotropic cytokine that generally promotes inflammation, in the pathogenesis of psychosis. However, no interventional studies in patients with psychosis, stratified using inflammatory markers, have been conducted to assess the therapeutic potential of targeting IL-6 in psychosis and to elucidate potential mechanism of effect. Tocilizumab is a humanised monoclonal antibody targeting the IL-6 receptor to inhibit IL-6 signalling, licensed in the UK for treatment of rheumatoid arthritis. The primary objective of this study is to test whether IL-6 contributes to the pathogenesis of first episode psychosis and to examine potential mechanisms by which IL-6 affects psychotic symptoms. A secondary objective is to examine characteristics of inflammation-associated psychosis. METHODS AND ANALYSIS A proof-of-concept study employing a randomised, parallel-group, double-blind, placebo-controlled design testing the effect of IL-6 inhibition on anhedonia in patients with psychosis. Approximately 60 participants with a diagnosis of schizophrenia and related psychotic disorders (ICD-10 codes F20, F22, F25, F28, F29) with evidence of low-grade inflammation (IL-6≥0.7 pg/mL) will receive either one intravenous infusion of tocilizumab (4.0 mg/kg; max 800 mg) or normal saline. Psychiatric measures and blood samples will be collected at baseline, 7, 14 and 28 days post infusion. Cognitive and neuroimaging data will be collected at baseline and 14 days post infusion. In addition, approximately 30 patients with psychosis without evidence of inflammation (IL-6<0.7 pg/mL) and 30 matched healthy controls will be recruited to complete identical baseline assessments to allow for comparison of the characteristic features of inflammation-associated psychosis. ETHICS AND DISSEMINATION The study is sponsored by the University of Bristol and has been approved by the Cambridge East Research Ethics Committee (reference: 22/EE/0010; IRAS project ID: 301682). Study findings will be published in peer-review journals. Findings will also be disseminated by scientific presentation and other means. TRIAL REGISTRATION NUMBER ISRCTN23256704.
Collapse
Affiliation(s)
- Éimear M Foley
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- Centre for Academic Mental Health, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Sian Lowri Griffiths
- Institute for Mental Health and Centre for Human Brain Health, University of Birmingham, Birmingham, UK
| | - Alexander Murray
- Institute for Mental Health and Centre for Human Brain Health, University of Birmingham, Birmingham, UK
| | - Jack Rogers
- Institute for Mental Health and Centre for Human Brain Health, University of Birmingham, Birmingham, UK
| | - Fabiana Corsi-Zuelli
- Institute for Mental Health and Centre for Human Brain Health, University of Birmingham, Birmingham, UK
- Department of Neuroscience and Behaviour, Division of Psychiatry, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
- Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | | | - Ella Warwick
- Institute for Mental Health and Centre for Human Brain Health, University of Birmingham, Birmingham, UK
| | - Martin Wilson
- Institute for Mental Health and Centre for Human Brain Health, University of Birmingham, Birmingham, UK
| | - Muzaffer Kaser
- Department of Psychiatry, University of Cambridge, Cambridge, UK
- Cambridgeshire and Peterborough NHS Foundation Trust, Fulbourn, UK
| | - Graham K Murray
- Department of Psychiatry, University of Cambridge, Cambridge, UK
- Cambridgeshire and Peterborough NHS Foundation Trust, Fulbourn, UK
| | - Bill Deakin
- Faculty of Biology, Medicine and Health, Division of Neuroscience and Experimental Psychology, School of Biological Sciences, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Deepak Jadon
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - John Suckling
- Department of Psychiatry, University of Cambridge, Cambridge, UK
- Cambridgeshire and Peterborough NHS Foundation Trust, Fulbourn, UK
| | - Nicholas M Barnes
- Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Rachel Upthegrove
- Institute for Mental Health and Centre for Human Brain Health, University of Birmingham, Birmingham, UK
- Early Intervention Service, Birmingham Women's and Children's NHS Foundation Trust, Birmingham, UK
| | - Golam M Khandaker
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- Centre for Academic Mental Health, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- National Institute for Health and Care Research, Bristol Biomedical Research Centre, Bristol, UK
- Avon and Wiltshire Mental Health Partnership NHS Trust, Bristol, UK
| |
Collapse
|
17
|
Raval NR, Wetherill RR, Wiers CE, Dubroff JG, Hillmer AT. Positron Emission Tomography of Neuroimmune Responses in Humans: Insights and Intricacies. Semin Nucl Med 2023; 53:213-229. [PMID: 36270830 PMCID: PMC11261531 DOI: 10.1053/j.semnuclmed.2022.08.008] [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: 08/15/2022] [Accepted: 08/30/2022] [Indexed: 11/06/2022]
Abstract
The brain's immune system plays a critical role in responding to immune challenges and maintaining homeostasis. However, dysregulated neuroimmune function contributes to neurodegenerative disease and neuropsychiatric conditions. In vivo positron emission tomography (PET) imaging of the neuroimmune system has facilitated a greater understanding of its physiology and the pathology of some neuropsychiatric conditions. This review presents an in-depth look at PET findings from human neuroimmune function studies, highlighting their importance in current neuropsychiatric research. Although the majority of human PET studies feature radiotracers targeting the translocator protein 18 kDa (TSPO), this review also considers studies with other neuroimmune targets, including monoamine oxidase B, cyclooxygenase-1 and cyclooxygenase-2, nitric oxide synthase, and the purinergic P2X7 receptor. Promising new targets, such as colony-stimulating factor 1, Sphingosine-1-phosphate receptor 1, and the purinergic P2Y12 receptor, are also discussed. The significance of validating neuroimmune targets and understanding their function and expression is emphasized in this review to better identify and interpret PET results.
Collapse
Affiliation(s)
- Nakul R Raval
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT; Yale PET Center, Yale University, New Haven, CT
| | - Reagan R Wetherill
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Corinde E Wiers
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA; Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Jacob G Dubroff
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Ansel T Hillmer
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT; Yale PET Center, Yale University, New Haven, CT; Department of Psychiatry, Yale University, New Haven, CT.
| |
Collapse
|
18
|
Rantala MJ, Luoto S, Borráz-León JI, Krams I. Schizophrenia: the new etiological synthesis. Neurosci Biobehav Rev 2022; 142:104894. [PMID: 36181926 DOI: 10.1016/j.neubiorev.2022.104894] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 08/25/2022] [Accepted: 09/25/2022] [Indexed: 10/31/2022]
Abstract
Schizophrenia has been an evolutionary paradox: it has high heritability, but it is associated with decreased reproductive success. The causal genetic variants underlying schizophrenia are thought to be under weak negative selection. To unravel this paradox, many evolutionary explanations have been suggested for schizophrenia. We critically discuss the constellation of evolutionary hypotheses for schizophrenia, highlighting the lack of empirical support for most existing evolutionary hypotheses-with the exception of the relatively well supported evolutionary mismatch hypothesis. It posits that evolutionarily novel features of contemporary environments, such as chronic stress, low-grade systemic inflammation, and gut dysbiosis, increase susceptibility to schizophrenia. Environmental factors such as microbial infections (e.g., Toxoplasma gondii) can better predict the onset of schizophrenia than polygenic risk scores. However, researchers have not been able to explain why only a small minority of infected people develop schizophrenia. The new etiological synthesis of schizophrenia indicates that an interaction between host genotype, microbe infection, and chronic stress causes schizophrenia, with neuroinflammation and gut dysbiosis mediating this etiological pathway. Instead of just alleviating symptoms with drugs, the parasite x genotype x stress model emphasizes that schizophrenia treatment should focus on detecting and treating possible underlying microbial infection(s), neuroinflammation, gut dysbiosis, and chronic stress.
Collapse
Affiliation(s)
- Markus J Rantala
- Department of Biology, University of Turku, FIN-20014 Turku, Finland.
| | - Severi Luoto
- School of Population Health, University of Auckland, 1023 Auckland, New Zealand
| | | | - Indrikis Krams
- Institute of Ecology and Earth Sciences, University of Tartu, 51014 Tartu, Estonia; Department of Zoology and Animal Ecology, Faculty of Biology, University of Latvia, 1004, Rīga, Latvia
| |
Collapse
|
19
|
Eggerstorfer B, Kim JH, Cumming P, Lanzenberger R, Gryglewski G. Meta-analysis of molecular imaging of translocator protein in major depression. Front Mol Neurosci 2022; 15:981442. [PMID: 36226319 PMCID: PMC9549359 DOI: 10.3389/fnmol.2022.981442] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 09/01/2022] [Indexed: 11/13/2022] Open
Abstract
Molecular neuroimaging studies provide mounting evidence that neuroinflammation plays a contributory role in the pathogenesis of major depressive disorder (MDD). This has been the focus of a number of positron emission tomography (PET) studies of the 17-kDa translocator protein (TSPO), which is expressed by microglia and serves as a marker of neuroinflammation. In this meta-analysis, we compiled and analyzed all available molecular imaging studies comparing cerebral TSPO binding in MDD patients with healthy controls. Our systematic literature search yielded eight PET studies encompassing 238 MDD patients and 164 healthy subjects. The meta-analysis revealed relatively increased TSPO binding in several cortical regions (anterior cingulate cortex: Hedges' g = 0.6, 95% CI: 0.36, 0.84; hippocampus: g = 0.54, 95% CI: 0.26, 0.81; insula: g = 0.43, 95% CI: 0.17, 0.69; prefrontal cortex: g = 0.36, 95% CI: 0.14, 0.59; temporal cortex: g = 0.39, 95% CI: -0.04, 0.81). While the high range of effect size in the temporal cortex might reflect group-differences in body mass index (BMI), exploratory analyses failed to reveal any relationship between elevated TSPO availability in the other four brain regions and depression severity, age, BMI, radioligand, or the binding endpoint used, or with treatment status at the time of scanning. Taken together, this meta-analysis indicates a widespread ∼18% increase of TSPO availability in the brain of MDD patients, with effect sizes comparable to those in earlier molecular imaging studies of serotonin transporter availability and monoamine oxidase A binding.
Collapse
Affiliation(s)
- Benjamin Eggerstorfer
- Department of Psychiatry and Psychotherapy, Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna, Vienna, Austria
| | - Jong-Hoon Kim
- Department of Psychiatry, Gachon University College of Medicine, Gil Medical Center, Neuroscience Research Institute, GAIHST, Gachon University, Incheon, South Korea
| | - Paul Cumming
- Department of Nuclear Medicine, Inselspital, Bern University, Bern, Switzerland
- School of Psychology and Counselling, Queensland University of Technology, Brisbane, QLD, Australia
| | - Rupert Lanzenberger
- Department of Psychiatry and Psychotherapy, Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna, Vienna, Austria
| | - Gregor Gryglewski
- Department of Psychiatry and Psychotherapy, Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH), Medical University of Vienna, Vienna, Austria
| |
Collapse
|
20
|
Yue W, Huang H, Duan J. Potential diagnostic biomarkers for schizophrenia. MEDICAL REVIEW (BERLIN, GERMANY) 2022; 2:385-416. [PMID: 37724326 PMCID: PMC10388817 DOI: 10.1515/mr-2022-0009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 06/20/2022] [Indexed: 09/20/2023]
Abstract
Schizophrenia (SCH) is a complex and severe mental disorder with high prevalence, disability, mortality and carries a heavy disease burden, the lifetime prevalence of SCH is around 0.7%-1.0%, which has a profound impact on the individual and society. In the clinical practice of SCH, key problems such as subjective diagnosis, experiential treatment, and poor overall prognosis are still challenging. In recent years, some exciting discoveries have been made in the research on objective biomarkers of SCH, mainly focusing on genetic susceptibility genes, metabolic indicators, immune indices, brain imaging, electrophysiological characteristics. This review aims to summarize the biomarkers that may be used for the prediction and diagnosis of SCH.
Collapse
Affiliation(s)
- Weihua Yue
- Institute of Mental Health, Peking University Sixth Hospital, Beijing, China
- National Clinical Research Center for Mental Disorders & NHC Key Laboratory of Mental Health (Peking University) and Chinese Academy of Medical Sciences Research Unit, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China
- Chinese Institute for Brain Research, Beijing, China
| | - Hailiang Huang
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Jubao Duan
- Center for Psychiatric Genetics, NorthShore University Health System, Evanston, IL, USA
- Department of Psychiatry and Behavioral Neurosciences, University of Chicago, Chicago, IL, USA
| |
Collapse
|
21
|
Cervenka S, Frick A, Bodén R, Lubberink M. Application of positron emission tomography in psychiatry-methodological developments and future directions. Transl Psychiatry 2022; 12:248. [PMID: 35701411 PMCID: PMC9198063 DOI: 10.1038/s41398-022-01990-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 05/20/2022] [Accepted: 05/25/2022] [Indexed: 11/09/2022] Open
Abstract
Mental disorders represent an increasing source of disability and high costs for societies globally. Molecular imaging techniques such as positron emission tomography (PET) represent powerful tools with the potential to advance knowledge regarding disease mechanisms, allowing the development of new treatment approaches. Thus far, most PET research on pathophysiology in psychiatric disorders has focused on the monoaminergic neurotransmission systems, and although a series of discoveries have been made, the results have not led to any material changes in clinical practice. We outline areas of methodological development that can address some of the important obstacles to fruitful progress. First, we point towards new radioligands and targets that can lead to the identification of processes upstream, or parallel to disturbances in monoaminergic systems. Second, we describe the development of new methods of PET data quantification and PET systems that may facilitate research in psychiatric populations. Third, we review the application of multimodal imaging that can link molecular imaging data to other aspects of brain function, thus deepening our understanding of disease processes. Fourth, we highlight the need to develop imaging study protocols to include longitudinal and interventional paradigms, as well as frameworks to assess dimensional symptoms such that the field can move beyond cross-sectional studies within current diagnostic boundaries. Particular effort should be paid to include also the most severely ill patients. Finally, we discuss the importance of harmonizing data collection and promoting data sharing to reach the desired sample sizes needed to fully capture the phenotype of psychiatric conditions.
Collapse
Affiliation(s)
- Simon Cervenka
- Department of Medical Sciences, Psychiatry, Uppsala University, Uppsala, Sweden. .,Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden.
| | - Andreas Frick
- grid.8993.b0000 0004 1936 9457Department of Medical Sciences, Psychiatry, Uppsala University, Uppsala, Sweden
| | - Robert Bodén
- grid.8993.b0000 0004 1936 9457Department of Medical Sciences, Psychiatry, Uppsala University, Uppsala, Sweden
| | - Mark Lubberink
- grid.8993.b0000 0004 1936 9457Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
22
|
Monji A, Mizoguchi Y. Neuroinflammation in Late-Onset Schizophrenia: Viewing from the Standpoint of the Microglia Hypothesis. Neuropsychobiology 2022; 81:98-103. [PMID: 34515181 DOI: 10.1159/000517861] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 06/14/2021] [Indexed: 11/19/2022]
Abstract
Schizophrenia develops mainly in adolescence, but late-onset schizophrenia (LOS) is not uncommon. According to the international consensus, schizophrenia which develops over 40 years old is called LOS and psychosis which develops over 60 years old is called very late-onset schizophrenia-like psychosis (VLOS). Compared to early-onset schizophrenia (EOS) that develops before the age of 40 years, LOS and VLOS are reported to be more common in women, and there are clinically clear differences such as less involvement of genetic factors than EOS. This review outlines the abnormalities of the neuroimmune system in the pathophysiology of LOS, especially focusing on the role of microglia.
Collapse
Affiliation(s)
- Akira Monji
- Department of Psychiatry, Faculty of Medicine, Saga University, Saga, Japan
| | - Yoshito Mizoguchi
- Department of Psychiatry, Faculty of Medicine, Saga University, Saga, Japan
| |
Collapse
|
23
|
Guo J, Qiu T, Wang L, Shi L, Ai M, Xia Z, Peng Z, Zheng A, Li X, Kuang L. Microglia Loss and Astrocyte Activation Cause Dynamic Changes in Hippocampal [18F]DPA-714 Uptake in Mouse Models of Depression. Front Cell Neurosci 2022; 16:802192. [PMID: 35250485 PMCID: PMC8896346 DOI: 10.3389/fncel.2022.802192] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 01/31/2022] [Indexed: 12/18/2022] Open
Abstract
Major depression is a serious and chronic mental illness. However, its etiology is poorly understood. Although glial cells have been increasingly implicated in the pathogenesis of depression, the specific role of microglia and astrocytes in stress-induced depression remains unclear. Translocator protein (TSPO) has long been considered a marker of neuroinflammation and microglial activation. However, this protein is also present on astrocytes. Thus, it is necessary to explore the relationships between TSPO, microglia, and astrocytes in the context of depression. In this study, C57BL/6J male mice were subjected to chronic unpredictable stress (CUS) for 5 weeks. Subsequently, sucrose preference and tail suspension tests (TSTs) were performed to assess anhedonia and despair in these mice. [18F]DPA-714 positron emission tomography (PET) was adopted to dynamically assess the changes in glial cells before and 2, 4, or 5 weeks after CUS exposure. The numbers of TSPO+ cells, ionized calcium-binding adaptor molecule (Iba)-1+ microglial cells, TSPO+/Iba-1+ cells, glial fibrillary acidic protein (GFAP)+ astrocytes, TSPO+/GFAP+ cells, and TUNEL-stained microglia were quantified using immunofluorescence staining. Real-time PCR was used to evaluate interleukin (IL)-1β, IL-4, and IL-18 expression in the hippocampus. We observed that hippocampal [18F]DPA-714 uptake significantly increased after 2 weeks of CUS. However, the signal significantly decreased after 5 weeks of CUS. CUS significantly reduced the number of Iba-1+, TSPO+, and TSPO+/Iba-1+ cells in the hippocampus, especially in the CA1 and dentate gyrus (DG) subregions. However, this intervention increased the number of GFAP+ astrocytes in the CA2/CA3 subregions of the hippocampus. In addition, microglial apoptosis in the early stage of CUS appeared to be involved in microglia loss. Further, the expression of pro-inflammatory cytokines (IL-1β and IL-18) was significantly decreased after CUS. In contrast, the expression of the anti-inflammatory cytokine IL-4 was significantly increased after 2 weeks of CUS. These results suggested that the CUS-induced dynamic changes in hippocampal [18F]DPA-714 uptake and several cytokines may be due to combined microglial and astrocyte action. These findings provide a theoretical reference for the future clinical applications of TSPO PET.
Collapse
Affiliation(s)
- Jiamei Guo
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Tian Qiu
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lixia Wang
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lei Shi
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ming Ai
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhu Xia
- Department of Nuclear Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhiping Peng
- College of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Anhai Zheng
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiao Li
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Li Kuang
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- *Correspondence: Li Kuang,
| |
Collapse
|
24
|
Abstract
ABSTRACT Inflammatory phenomena are found in many psychiatric disorders-notably, depression, schizophrenia, and posttraumatic stress disorder. Inflammation has been linked to severity and treatment resistance, and may both contribute to, and result from, the pathophysiology of some psychiatric illnesses. Emerging research suggests that inflammation may contribute to symptom domains of reward, motor processing, and threat reactivity across different psychiatric diagnoses. Reward-processing deficits contribute to motivational impairments in depression and schizophrenia, and motor-processing deficits contribute to psychomotor slowing in both depression and schizophrenia. A number of experimental models and clinical trials suggest that inflammation produces deficits in reward and motor processing through common pathways connecting the cortex and the striatum, which includes the nucleus accumbens, caudate nucleus, and putamen.The observed effects of inflammation on psychiatric disorders may cut across traditional conceptualizations of psychiatric diagnoses. Further study may lead to targeted immunomodulating treatments that address difficult-to-treat symptoms in a number of psychiatric disorders. In this review, we use a Research Domain Criteria framework to discuss proposed mechanisms for inflammation and its effects on the domains of reward processing, psychomotor slowing, and threat reactivity. We also discuss data that support contributing roles of metabolic dysregulation and sex differences on the behavioral outcomes of inflammation. Finally, we discuss ways that future studies can help disentangle this complex topic to yield fruitful results that will help advance the field of psychoneuroimmunology.
Collapse
Affiliation(s)
- David S Thylur
- From the Department of Psychiatry and Behavioral Sciences, Emory University
| | | |
Collapse
|
25
|
Morrens M, Overloop C, Coppens V, Loots E, Van Den Noortgate M, Vandenameele S, Leboyer M, De Picker L. The relationship between immune and cognitive dysfunction in mood and psychotic disorder: a systematic review and a meta-analysis. Mol Psychiatry 2022; 27:3237-3246. [PMID: 35484245 PMCID: PMC9708549 DOI: 10.1038/s41380-022-01582-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 04/11/2022] [Accepted: 04/12/2022] [Indexed: 12/18/2022]
Abstract
BACKGROUND In psychotic and mood disorders, immune alterations are hypothesized to underlie cognitive symptoms, as they have been associated with elevated blood levels of inflammatory cytokines, kynurenine metabolites, and markers of microglial activation. The current meta-analysis synthesizes all available clinical evidence on the associations between immunomarkers (IMs) and cognition in these psychiatric illnesses. METHODS Pubmed, Web of Science, and Psycinfo were searched for peer-reviewed studies on schizophrenia spectrum disorder (SZ), bipolar disorder (BD), or major depressive disorder (MDD) including an association analysis between at least one baseline neuropsychological outcome measure (NP) and one IM (PROSPERO ID:CRD42021278371). Quality assessment was performed using BIOCROSS. Correlation meta-analyses, and random effect models, were conducted in Comprehensive Meta-Analysis version 3 investigating the association between eight cognitive domains and pro-inflammatory and anti-inflammatory indices (PII and AII) as well as individual IM. RESULTS Seventy-five studies (n = 29,104) revealed global cognitive performance (GCP) to be very weakly associated to PII (r = -0.076; p = 0.003; I2 = 77.4) or AII (r = 0.067; p = 0.334; I2 = 38.0) in the combined patient sample. Very weak associations between blood-based immune markers and global or domain-specific GCP were found, either combined or stratified by diagnostic subgroup (GCP x PII: SZ: r = -0.036, p = 0.370, I2 = 70.4; BD: r = -0.095, p = 0.013, I2 = 44.0; MDD: r = -0.133, p = 0.040, I2 = 83.5). We found evidence of publication bias. DISCUSSION There is evidence of only a weak association between blood-based immune markers and cognition in mood and psychotic disorders. Significant publication and reporting biases were observed and most likely underlie the inflation of such associations in individual studies.
Collapse
Affiliation(s)
- M. Morrens
- grid.5284.b0000 0001 0790 3681Faculty of Medicine and Health Sciences, Collaborative Antwerp Psychiatric Research Institute (CAPRI), University of Antwerp, Antwerp, Belgium ,Scientific Initiative of Neuropsychiatric and Psychopharmacological Studies (SINAPS), University Psychiatric Centre Duffel, Duffel, Belgium
| | - C. Overloop
- Scientific Initiative of Neuropsychiatric and Psychopharmacological Studies (SINAPS), University Psychiatric Centre Duffel, Duffel, Belgium
| | - V. Coppens
- grid.5284.b0000 0001 0790 3681Faculty of Medicine and Health Sciences, Collaborative Antwerp Psychiatric Research Institute (CAPRI), University of Antwerp, Antwerp, Belgium ,Scientific Initiative of Neuropsychiatric and Psychopharmacological Studies (SINAPS), University Psychiatric Centre Duffel, Duffel, Belgium
| | - E. Loots
- grid.5284.b0000 0001 0790 3681Faculty of Medicine and Health Sciences, Nursing and obstetrics, University of Antwerp, Antwerp, Belgium
| | - M. Van Den Noortgate
- grid.5284.b0000 0001 0790 3681Faculty of Medicine and Health Sciences, Collaborative Antwerp Psychiatric Research Institute (CAPRI), University of Antwerp, Antwerp, Belgium
| | - S. Vandenameele
- grid.5284.b0000 0001 0790 3681Faculty of Medicine and Health Sciences, Collaborative Antwerp Psychiatric Research Institute (CAPRI), University of Antwerp, Antwerp, Belgium ,grid.411326.30000 0004 0626 3362University Hospital Brussels, Brussels Health Campus, Jette, Belgium
| | - M. Leboyer
- grid.462410.50000 0004 0386 3258INSERM U955, Equipe Psychiatrie Translationnelle, Créteil, France ,grid.484137.d0000 0005 0389 9389Fondation FondaMental, Créteil, France ,grid.412116.10000 0001 2292 1474AP-HP, Hôpitaux Universitaires Henri Mondor, DHU Pepsy, Pôle de Psychiatrie et d’Addictologie, Créteil, France ,grid.410511.00000 0001 2149 7878Université Paris Est Créteil, Faculté de Médecine, Creteil, France
| | - L. De Picker
- grid.5284.b0000 0001 0790 3681Faculty of Medicine and Health Sciences, Collaborative Antwerp Psychiatric Research Institute (CAPRI), University of Antwerp, Antwerp, Belgium ,Scientific Initiative of Neuropsychiatric and Psychopharmacological Studies (SINAPS), University Psychiatric Centre Duffel, Duffel, Belgium
| |
Collapse
|
26
|
Nutma E, Ceyzériat K, Amor S, Tsartsalis S, Millet P, Owen DR, Papadopoulos V, Tournier BB. Cellular sources of TSPO expression in healthy and diseased brain. Eur J Nucl Med Mol Imaging 2021; 49:146-163. [PMID: 33433698 PMCID: PMC8712293 DOI: 10.1007/s00259-020-05166-2] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 12/13/2020] [Indexed: 12/11/2022]
Abstract
The 18 kDa translocator protein (TSPO) is a highly conserved protein located in the outer mitochondrial membrane. TSPO binding, as measured with positron emission tomography (PET), is considered an in vivo marker of neuroinflammation. Indeed, TSPO expression is altered in neurodegenerative, neuroinflammatory, and neuropsychiatric diseases. In PET studies, the TSPO signal is often viewed as a marker of microglial cell activity. However, there is little evidence in support of a microglia-specific TSPO expression. This review describes the cellular sources and functions of TSPO in animal models of disease and human studies, in health, and in central nervous system diseases. A discussion of methods of analysis and of quantification of TSPO is also presented. Overall, it appears that the alterations of TSPO binding, their cellular underpinnings, and the functional significance of such alterations depend on many factors, notably the pathology or the animal model under study, the disease stage, and the involved brain regions. Thus, further studies are needed to fully determine how changes in TSPO binding occur at the cellular level with the ultimate goal of revealing potential therapeutic pathways.
Collapse
Affiliation(s)
- Erik Nutma
- Department of Pathology, Amsterdam UMC, VUmc, Amsterdam, The Netherlands
| | - Kelly Ceyzériat
- Division of Adult Psychiatry, Department of Psychiatry, University Hospitals of Geneva, Avenue de la Roseraie, 64, 1206, Geneva, Switzerland
- Division of Nuclear medicine and Molecular Imaging, University Hospitals of Geneva, Geneva, Switzerland
- Division of Radiation Oncology, Department of Oncology, University Hospitals of Geneva, Geneva, Switzerland
| | - Sandra Amor
- Department of Pathology, Amsterdam UMC, VUmc, Amsterdam, The Netherlands
- Centre for Neuroscience and Trauma, Blizard Institute, Barts and the London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Stergios Tsartsalis
- Division of Adult Psychiatry, Department of Psychiatry, University Hospitals of Geneva, Avenue de la Roseraie, 64, 1206, Geneva, Switzerland
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - Philippe Millet
- Division of Adult Psychiatry, Department of Psychiatry, University Hospitals of Geneva, Avenue de la Roseraie, 64, 1206, Geneva, Switzerland
- Department of Psychiatry, University of Geneva, Geneva, Switzerland
| | - David R Owen
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - Vassilios Papadopoulos
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, USA
| | - Benjamin B Tournier
- Division of Adult Psychiatry, Department of Psychiatry, University Hospitals of Geneva, Avenue de la Roseraie, 64, 1206, Geneva, Switzerland.
- Department of Psychiatry, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
27
|
De Picker LJ, Haarman BCM. Applicability, potential and limitations of TSPO PET imaging as a clinical immunopsychiatry biomarker. Eur J Nucl Med Mol Imaging 2021; 49:164-173. [PMID: 33735406 DOI: 10.1007/s00259-021-05308-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 03/08/2021] [Indexed: 02/08/2023]
Abstract
PURPOSE TSPO PET imaging may hold promise as a single-step diagnostic work-up for clinical immunopsychiatry. This review paper on the clinical applicability of TSPO PET for primary psychiatric disorders discusses if and why TSPO PET imaging might become the first clinical immunopsychiatry biomarker and the investment prerequisites and scientific advancements needed to accommodate this transition from bench to bedside. METHODS We conducted a systematic search of the literature to identify clinical studies of TSPO PET imaging in patients with primary psychiatric disorders. We included both original case-control studies as well as longitudinal cohort studies of patients with a primary psychiatric diagnosis. RESULTS Thirty-one original studies met our inclusion criteria. In the field of immunopsychiatry, TSPO PET has until now mostly been studied in schizophrenia and related psychotic disorders, and to a lesser extent in mood disorders and neurodevelopmental disorders. Quantitative TSPO PET appears most promising as a predictive biomarker for the transdiagnostic identification of subgroups or disease stages that could benefit from immunological treatments, or as a prognostic biomarker forecasting patients' illness course. Current scanning protocols are still too unreliable, impractical and invasive for clinical use in symptomatic psychiatric patients. CONCLUSION TSPO PET imaging in its present form does not yet offer a sufficiently attractive cost-benefit ratio to become a clinical immunopsychiatry biomarker. Its translation to psychiatric clinical practice will depend on the prioritising of longitudinal research and the establishment of a uniform protocol rendering clinically meaningful TSPO uptake quantification at the shortest possible scan duration without arterial cannulation.
Collapse
Affiliation(s)
- Livia J De Picker
- University Psychiatric Hospital Campus Duffel, Stationsstraat 22C, 2570, Duffel, Belgium.
- Collaborative Antwerp Psychiatric Research Institute, University of Antwerp, Wilrijkstraat 1, 2650, Edegem, Belgium.
| | - Benno C M Haarman
- Department of Psychiatry, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9700RA, Groningen, The Netherlands
- Rob Giel Research Center (RGOc), University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9700RA, Groningen, The Netherlands
| |
Collapse
|
28
|
Corsi-Zuelli F, Deakin B, de Lima MHF, Qureshi O, Barnes NM, Upthegrove R, Louzada-Junior P, Del-Ben CM. T regulatory cells as a potential therapeutic target in psychosis? Current challenges and future perspectives. Brain Behav Immun Health 2021; 17:100330. [PMID: 34661175 PMCID: PMC7611834 DOI: 10.1016/j.bbih.2021.100330] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 08/15/2021] [Accepted: 08/16/2021] [Indexed: 12/12/2022] Open
Abstract
Many studies have reported that patients with psychosis, even before drug treatment, have mildly raised levels of blood cytokines relative to healthy controls. In contrast, there is a remarkable scarcity of studies investigating the cellular basis of immune function and cytokine changes in psychosis. The few flow-cytometry studies have been limited to counting the proportion of the major classes of monocyte and lymphocytes without distinguishing their pro- and anti-inflammatory subsets. Moreover, most of the investigations are cross-sectional and conducted with patients on long-term medication. These features make it difficult to eliminate confounding of illness-related changes by lifestyle factors, disease duration, and long exposure to antipsychotics. This article focuses on regulatory T cells (Tregs), cornerstone immune cells that regulate innate and adaptive immune forces and neuro-immune interactions between astrocytes and microglia. Tregs are also implicated in cardio-metabolic disorders that are common comorbidities of psychosis. We have recently proposed that Tregs are hypofunctional ('h-Tregs') in psychosis driven by our clinical findings and other independent research. Our h-Treg-glial imbalance hypothesis offers a new account for the co-occurrence of systemic immune dysregulation and mechanisms of psychosis development. This article extends our recent review, the h-Treg hypothesis, to cover new discoveries on Treg-based therapies from pre-clinical findings and their clinical implications. We provide a detailed characterisation of Treg studies in psychosis, identifying important methodological limitations and perspectives for scientific innovation. The outcomes presented in this article reaffirms our proposed h-Treg state in psychosis and reveals emerging preclinical research suggesting the potential benefit of Treg-enhancing therapies. There is a clear need for longitudinal studies conducted with drug-naïve or minimally treated patients using more sophisticated techniques of flow-cytometry, CyTOF expression markers, and in vitro co-culture assays to formally test the suppressive capacity of Tregs. Investment in Treg research offers major potential benefits in targeting emerging immunomodulatory treatment modalities on person-specific immune dysregulations.
Collapse
Affiliation(s)
- Fabiana Corsi-Zuelli
- Department of Neuroscience and Behaviour, Division of Psychiatry, Ribeirão Preto Medical School, University of São Paulo, 14048-900, Brazil
- Center for Research on Inflammatory Diseases – CRID, Ribeirão Preto Medical School, University of São Paulo, São Paulo, 14048-900, Brazil
| | - Bill Deakin
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PT, UK
| | - Mikhael Haruo Fernandes de Lima
- Center for Research on Inflammatory Diseases – CRID, Ribeirão Preto Medical School, University of São Paulo, São Paulo, 14048-900, Brazil
- Department of Internal Medicine, Division of Clinical Immunology, Ribeirão Preto Medical School, University of São Paulo, 14048-900, Brazil
| | - Omar Qureshi
- Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, Vincent Drive Edgbaston, Birmingham, B15 2TT, UK
- Celentyx Ltd, Birmingham Research Park, Vincent Drive, Edgbaston, Birmingham, B15 2SQ, UK
| | - Nicholas M. Barnes
- Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, Vincent Drive Edgbaston, Birmingham, B15 2TT, UK
| | - Rachel Upthegrove
- Institute for Mental Health, University of Birmingham, Prichatts Rd, Edgbaston, B152TT, UK
- Birmingham Early Intervention Service, Birmingham Women's and Children's NHS Foundation Trust, B4 6NH, UK
| | - Paulo Louzada-Junior
- Center for Research on Inflammatory Diseases – CRID, Ribeirão Preto Medical School, University of São Paulo, São Paulo, 14048-900, Brazil
- Department of Internal Medicine, Division of Clinical Immunology, Ribeirão Preto Medical School, University of São Paulo, 14048-900, Brazil
| | - Cristina Marta Del-Ben
- Department of Neuroscience and Behaviour, Division of Psychiatry, Ribeirão Preto Medical School, University of São Paulo, 14048-900, Brazil
| |
Collapse
|
29
|
Da Silva T, Guma E, Hafizi S, Koppel A, Rusjan P, Kennedy JL, Chakravarty MM, Mizrahi R. Genetically Predicted Brain C4A Expression Is Associated With TSPO and Hippocampal Morphology. Biol Psychiatry 2021; 90:652-660. [PMID: 34456009 DOI: 10.1016/j.biopsych.2021.06.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 06/27/2021] [Accepted: 06/28/2021] [Indexed: 01/18/2023]
Abstract
BACKGROUND Alterations in the immune system, particularly C4A, have been implicated in the pathophysiology of schizophrenia. C4A promotes synapse elimination by microglia in preclinical models; however, it is unknown whether this process is also present in living humans and how it affects brain morphology. METHODS Participants (N = 111; 33 patients with psychosis, 37 individuals at clinical high risk, and 41 healthy control subjects) underwent a TSPO [18F]FEPPA positron emission tomography scan and a magnetic resonance imaging scan. Brain C4A expression was genetically predicted as a function of the dosage of each of 4 structural elements (C4AL, C4BL, C4AS, C4BS). RESULTS Higher genetically predicted brain C4A expression was associated with higher brain microglial marker (TSPO) and altered hippocampal morphology, including reduced surface area and medial displacement in the CA1 area. This study is the first to quantify genetically predicted brain C4A expression in individuals at clinical high risk, showing significantly lower C4A in individuals at clinical high risk compared with healthy control subjects. We also showed a robust effect of sex on genetically predicted brain C4A expression and effects of both sex and cannabis use on brain TSPO. CONCLUSIONS This study shows for the first time complement system (C4A) coupling with a microglial marker (TSPO) and hippocampal morphology in living human brain. These findings pave the way for future research on the interaction between C4A and glial cell function, which has the potential to inform the disease mechanism underlying psychosis and schizophrenia.
Collapse
Affiliation(s)
- Tania Da Silva
- Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Elisa Guma
- Computational Brain Anatomy Laboratory, Montreal, Quebec, Canada; Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada
| | - Sina Hafizi
- Department of Psychiatry, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Alex Koppel
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Pablo Rusjan
- Cerebral Imaging Center, Douglas Mental Health University Institute, Montreal, Quebec, Canada; Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| | - James L Kennedy
- Tanenbaum Centre for Pharmacogenetics, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Mallar M Chakravarty
- Cerebral Imaging Center, Douglas Mental Health University Institute, Montreal, Quebec, Canada; Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada; Department of Psychiatry, McGill University, Montreal, Quebec, Canada; Department of Biological and Biomedical Engineering, McGill University, Montreal, Quebec, Canada
| | - Romina Mizrahi
- Cerebral Imaging Center, Douglas Mental Health University Institute, Montreal, Quebec, Canada; Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada; Department of Psychiatry, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
30
|
De Picker LJ, Victoriano GM, Richards R, Gorvett AJ, Lyons S, Buckland GR, Tofani T, Norman JL, Chatelet DS, Nicoll JAR, Boche D. Immune environment of the brain in schizophrenia and during the psychotic episode: A human post-mortem study. Brain Behav Immun 2021; 97:319-327. [PMID: 34339805 PMCID: PMC8475749 DOI: 10.1016/j.bbi.2021.07.017] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 07/22/2021] [Accepted: 07/24/2021] [Indexed: 02/05/2023] Open
Abstract
A causal relationship between immune dysregulation and schizophrenia has been supported by genome-wide association studies and epidemiological evidence. It remains unclear to what extent the brain immune environment is implicated in this hypothesis. We investigated the immunophenotype of microglia and the presence of perivascular macrophages and T lymphocytes in post-mortem brain tissue. Dorsal prefrontal cortex of 40 controls (22F:18M) and 37 (10F:27M) schizophrenia cases, of whom 16 had active psychotic symptoms at the time of death, was immunostained for seven markers of microglia (CD16, CD32a, CD64, CD68, HLA-DR, Iba1 and P2RY12), two markers for perivascular macrophages (CD163 and CD206) and T-lymphocytes (CD3). Automated quantification was blinded to the case designation and performed separately on the grey and white matter. 3D reconstruction of Iba1-positive microglia was performed in selected cases. An increased cortical expression of microglial Fcγ receptors (CD64 F = 7.92, p = 0.007; CD64/HLA-DR ratio F = 5.02, p = 0.029) highlights the importance of communication between the central and peripheral immune systems in schizophrenia. Patients in whom psychotic symptoms were present at death demonstrated an age-dependent increase of Iba1 and increased CD64/HLA-DR ratios relative to patients without psychotic symptoms. Microglia in schizophrenia demonstrated a primed/reactive morphology. A potential role for T-lymphocytes was observed, but we did not confirm the presence of recruited macrophages in the brains of schizophrenia patients. Taking in account the limitations of a post-mortem study, our findings support the hypothesis of an alteration of the brain immune environment in schizophrenia, with symptomatic state- and age-dependent effects.
Collapse
Affiliation(s)
- Livia J De Picker
- Collaborative Antwerp Psychiatric Research Institute, University of Antwerp, Antwerp, Belgium; University Psychiatric Department Campus Duffel, Duffel, Belgium
| | - Gerardo Mendez Victoriano
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Rhys Richards
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Alexander J Gorvett
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Simeon Lyons
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - George R Buckland
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Tommaso Tofani
- Psychiatry Unit, Health Science Department, University of Florence, Florence, Italy
| | - Jeanette L Norman
- Histochemistry Research Unit, Clinical and Experimental Sciences, Faculty of Medicine University of Southampton, Southampton, UK
| | - David S Chatelet
- Biomedical Imaging Unit, Southampton General Hospital, University of Southampton, Southampton, UK
| | - James A R Nicoll
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK; Department of Cellular Pathology, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Delphine Boche
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK.
| |
Collapse
|
31
|
Corsi-Zuelli F, Deakin B. Impaired regulatory T cell control of astroglial overdrive and microglial pruning in schizophrenia. Neurosci Biobehav Rev 2021; 125:637-653. [PMID: 33713699 DOI: 10.1016/j.neubiorev.2021.03.004] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 02/16/2021] [Accepted: 03/06/2021] [Indexed: 02/07/2023]
Abstract
It is widely held that schizophrenia involves an active process of peripheral inflammation that induces or reflects brain inflammation with activation of microglia, the brain's resident immune cells. However, recent in vivo radioligand binding studies and large-scale transcriptomics in post-mortem brain report reduced markers of microglial inflammation. The findings suggest a contrary hypothesis; that microglia are diverted into their non-inflammatory synaptic remodelling phenotype that interferes with neurodevelopment and perhaps contributes to the relapsing nature of schizophrenia. Recent discoveries on the regulatory interactions between micro- and astroglial cells and immune regulatory T cells (Tregs) cohere with clinical omics data to suggest that: i) disinhibited astrocytes mediate the shift in microglial phenotype via the production of transforming growth factor-beta, which also contributes to the disturbances of dopamine and GABA function in schizophrenia, and ii) systemically impaired functioning of Treg cells contributes to the dysregulation of glial function, the low-grade peripheral inflammation, and the hitherto unexplained predisposition to auto-immunity and reduced life-expectancy in schizophrenia, including greater COVID-19 mortality.
Collapse
Affiliation(s)
- Fabiana Corsi-Zuelli
- Department of Neuroscience and Behaviour, Division of Psychiatry, Ribeirão Preto Medical School, University of São Paulo, 14048-900, Ribeirão Preto, São Paulo, Brazil
| | - Bill Deakin
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PT, UK.
| |
Collapse
|
32
|
Sommer IE, Gangadin SS, de Witte LD, Koops S, van Baal C, Bahn S, Drexhage H, van Haren NEM, Veling W, Bruggeman R, Martens P, Wiersma S, Veerman SRT, Grootens KP, van Beveren N, Kahn RS, Begemann MJH. Simvastatin Augmentation for Patients With Early-Phase Schizophrenia-Spectrum Disorders: A Double-Blind, Randomized Placebo-Controlled Trial. Schizophr Bull 2021; 47:1108-1115. [PMID: 33608711 PMCID: PMC8266622 DOI: 10.1093/schbul/sbab010] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Schizophrenia-spectrum disorders (SSD) are associated with increased inflammatory markers, both in brain and periphery. Augmentation with drugs that lower this pro-inflammatory status may improve clinical presentation. Simvastatin crosses the blood-brain barrier, has anti- inflammatory and neuroprotective effects and reduces metabolic syndrome. In this study, we investigated if 12 months of simvastatin augmentation can improve symptoms and cognition in patients with early SSD. This double-blind placebo-controlled trial included 127 SSD patients across the Netherlands, <3 years after their diagnosis. From these, 119 were randomly assigned 1:1 to simvastatin 40 mg (n = 61) or placebo (n = 58), stratified for sex and study site. Primary outcomes were symptom severity and cognition after 12 months of treatment. Depression, symptom subscores, general functioning, metabolic syndrome, movement disorders, and safety were secondary outcomes. Intention to treat analyses were performed using linear mixed models and ANCOVA. No main effect of simvastatin treatment was found on total symptom severity after 12 months of treatment as compared to placebo (X2(1) = 0.01, P = .90). Group differences varied over time (treatment*time X2(4) = 11.2; P = .025), with significantly lower symptom severity in the simvastatin group after 6 months (mean difference = -4.8; P = .021; 95% CI: -8.8 to -0.7) and at 24 months follow-up (mean difference = -4.7; P = .040; 95% CI: -9.3 to -0.2). No main treatment effect was found for cognition (F(1,0.1) = 0.37, P = .55) or secondary outcomes. SAEs occurred more frequently with placebo (19%) than with simvastatin (6.6%). This negative finding corroborates other large scale studies on aspirin, minocycline, and celecoxib that could not replicate positive findings of smaller studies, and suggests that anti-inflammatory augmentation does not improve the clinical presentation of SSD.
Collapse
Affiliation(s)
- Iris E Sommer
- Department of Biomedical Sciences of Cells & Systems, University Medical Center Groningen (UMCG), University of Groningen, Groningen, the Netherlands,Department of Psychiatry, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands,To whom correspondence should be addressed; Department of Biomedical Sciences of Cells & Systems, University Medical Center Groningen (UMCG), University of Groningen, Groningen, the Netherlands; tel: +31625647485, e-mail:
| | - Shiral S Gangadin
- Department of Biomedical Sciences of Cells & Systems, University Medical Center Groningen (UMCG), University of Groningen, Groningen, the Netherlands
| | - Lot D de Witte
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Sanne Koops
- Department of Biomedical Sciences of Cells & Systems, University Medical Center Groningen (UMCG), University of Groningen, Groningen, the Netherlands
| | - C van Baal
- Department of Biostatics and Research Support, University Medical Center Utrecht, Julius Center, Utrecht, the Netherlands
| | - Sabine Bahn
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| | - Hemmo Drexhage
- Department of Immunology Research, Erasmus MC, Rotterdam, the Netherlands
| | - N E M van Haren
- Department of Child Psychiatry, Erasmus MC, Rotterdam, the Netherlands
| | - Wim Veling
- Department of Psychiatry, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - R Bruggeman
- Department of Psychiatry, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Peter Martens
- Reinier van Arkel Institute for Mental Health Care, ‘s Hertogenbosch, the Netherlands
| | - Sybren Wiersma
- Early Intervention Psychosis Team, GGZ inGeest Specialized Mental Health Care, Hoofddorp, the Netherlands
| | - Selene R T Veerman
- Community Mental Health, Mental Health Service Noord-Holland Noord, Alkmaar, The Netherlands
| | - Koen P Grootens
- Reinier van Arkel Institute for Mental Health Care, ‘s Hertogenbosch, the Netherlands
| | - Nico van Beveren
- Antes Center for Mental Health Care, Department of Neuroscience, and Department of Psychiatry, Erasmus MC, Rotterdam, the Netherlands
| | - Rene S Kahn
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY,Department of Psychiatry, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Marieke J H Begemann
- Department of Biomedical Sciences of Cells & Systems, University Medical Center Groningen (UMCG), University of Groningen, Groningen, the Netherlands
| |
Collapse
|
33
|
Iliopoulou SM, Tsartsalis S, Kaiser S, Millet P, Tournier BB. Dopamine and Neuroinflammation in Schizophrenia - Interpreting the Findings from Translocator Protein (18kDa) PET Imaging. Neuropsychiatr Dis Treat 2021; 17:3345-3357. [PMID: 34819729 PMCID: PMC8608287 DOI: 10.2147/ndt.s334027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 10/09/2021] [Indexed: 12/22/2022] Open
Abstract
Schizophrenia is a complex disease whose pathophysiology is not yet fully understood. In addition to the long prevailing dopaminergic hypothesis, the evidence suggests that neuroinflammation plays a role in the pathophysiology of the disease. Recent studies using positron emission tomography (PET) that target a 18kDa translocator protein (TSPO) in activated microglial cells in an attempt to measure neuroinflammation in patients have shown a decrease or a lack of an increase in TSPO binding. Many biological and methodological considerations have been formulated to explain these findings. Although dopamine has been described as an immunomodulatory molecule, its potential role in neuroinflammation has not been explored in the aforementioned studies. In this review, we discuss the interactions between dopamine and neuroinflammation in psychotic states. Dopamine may inhibit neuroinflammation in activated microglia. Proinflammatory molecules released from microglia may decrease dopaminergic transmission. This could potentially explain why the levels of neuroinflammation in the brain of patients with schizophrenia seem to be unchanged or decreased compared to those in healthy subjects. However, most data are indirect and are derived from animal studies or from studies performed outside the field of schizophrenia. Further studies are needed to combine TSPO and dopamine imaging to study the association between microglial activation and dopamine system function.
Collapse
Affiliation(s)
- Sotiria Maria Iliopoulou
- Adult Psychiatry Division, Department of Psychiatry, Geneva University Hospitals (HUG), Geneva, 1225, Switzerland
| | | | - Stefan Kaiser
- Adult Psychiatry Division, Department of Psychiatry, Geneva University Hospitals (HUG), Geneva, 1225, Switzerland.,Faculty of Medicine, University of Geneva, Geneva, 1204, Switzerland
| | - Philippe Millet
- Adult Psychiatry Division, Department of Psychiatry, Geneva University Hospitals (HUG), Geneva, 1225, Switzerland.,Faculty of Medicine, University of Geneva, Geneva, 1204, Switzerland
| | - Benjamin B Tournier
- Adult Psychiatry Division, Department of Psychiatry, Geneva University Hospitals (HUG), Geneva, 1225, Switzerland.,Faculty of Medicine, University of Geneva, Geneva, 1204, Switzerland
| |
Collapse
|
34
|
Meyer JH, Cervenka S, Kim MJ, Kreisl WC, Henter ID, Innis RB. Neuroinflammation in psychiatric disorders: PET imaging and promising new targets. Lancet Psychiatry 2020; 7:1064-1074. [PMID: 33098761 PMCID: PMC7893630 DOI: 10.1016/s2215-0366(20)30255-8] [Citation(s) in RCA: 151] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 04/30/2020] [Accepted: 05/01/2020] [Indexed: 01/14/2023]
Abstract
Neuroinflammation is a multifaceted physiological and pathophysiological response of the brain to injury and disease. Given imaging findings of 18 kDa translocator protein (TSPO) and the development of radioligands for other inflammatory targets, PET imaging of neuroinflammation is at a particularly promising stage. This Review critically evaluates PET imaging results of inflammation in psychiatric disorders, including major depressive disorder, schizophrenia and psychosis disorders, substance use, and obsessive-compulsive disorder. We also consider promising new targets that can be measured in the brain, such as monoamine oxidase B, cyclooxygenase-1 and cyclooxygenase-2, colony stimulating factor 1 receptor, and the purinergic P2X7 receptor. Thus far, the most compelling TSPO imaging results have arguably been found in major depressive disorder, for which consistent increases have been observed, and in schizophrenia and psychosis, for which patients show reduced TSPO levels. This pattern highlights the importance of validating brain biomarkers of neuroinflammation for each condition separately before moving on to patient stratification and treatment monitoring trials.
Collapse
Affiliation(s)
- Jeffrey H Meyer
- Campbell Family Mental Health Research Institute, Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Simon Cervenka
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet and Stockholm Health Care Services, Stockholm, Sweden
| | - Min-Jeong Kim
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, MD, USA
| | - William C Kreisl
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Ioline D Henter
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, MD, USA
| | - Robert B Innis
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, MD, USA.
| |
Collapse
|