1
|
Fischer C, Thomas D, Gurke R, Tegeder I. Brain region specific regulation of anandamide (down) and sphingosine-1-phosphate (up) in association with anxiety (AEA) and resilience (S1P) in a mouse model of chronic unpredictable mild stress. Pflugers Arch 2024; 476:1863-1880. [PMID: 39177699 PMCID: PMC11582197 DOI: 10.1007/s00424-024-03012-0] [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: 05/12/2024] [Revised: 05/12/2024] [Accepted: 08/13/2024] [Indexed: 08/24/2024]
Abstract
Chronic unpredictable and unavoidable stress is associated with mental health problems such as depression and anxiety, whereas cycles of stress and stress relief strengthen resilience. It has been suggested that increased breakdown of brain endocannabinoids (eCB) promotes a feeling of adversity. To assess the impact of stress on bioactive lipid homeostasis, we analyzed eCB, sphingolipids, and ceramides in seven brain regions and plasma in a mouse model of chronic unpredictable mild stress. Chronic unpredictable mild stress (CUMS) was associated with low levels of anandamide in hippocampus and prefrontal cortex in association with indicators of anxiety (elevated plus maze). Oppositely, CUMS caused elevated levels of sphingosine-1-phosphate (S1P d18:1) and sphinganine-1-phosphate (S1P d18:0) in the midbrain and thalamus, which was associated with readouts of increased stress resilience, i.e., marble burying and struggling in the tail suspension tests. In the periphery, elevated plasma levels of ceramides revealed similarities with human major depression and suggested unfavorable effects of stress on metabolism, but plasma lipids were not associated with body weight, sucrose consumption, or behavioral features of depression or anxiety. The observed brain site-specific lipid changes suggest that the forebrain succumbs to adverse stress effects while the midbrain takes up defensive adjustments.
Collapse
Affiliation(s)
- Caroline Fischer
- Goethe-University Frankfurt, Faculty of Medicine, Institute of Clinical Pharmacology, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Dominique Thomas
- Goethe-University Frankfurt, Faculty of Medicine, Institute of Clinical Pharmacology, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Robert Gurke
- Goethe-University Frankfurt, Faculty of Medicine, Institute of Clinical Pharmacology, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Irmgard Tegeder
- Goethe-University Frankfurt, Faculty of Medicine, Institute of Clinical Pharmacology, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany.
| |
Collapse
|
2
|
Du J, Baranova A, Zhang G, Zhang F. The causal relationship between immune cell traits and schizophrenia: a Mendelian randomization analysis. Front Immunol 2024; 15:1452214. [PMID: 39399496 PMCID: PMC11466782 DOI: 10.3389/fimmu.2024.1452214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 09/09/2024] [Indexed: 10/08/2024] Open
Abstract
Introduction The complex and unresolved pathogenesis of schizophrenia has posed significant challenges to its diagnosis and treatment. While recent research has established a clear association between immune function and schizophrenia, the causal relationship between the two remains elusive. Methods We employed a bidirectional two-sample Mendelian randomization approach to investigate the causal relationship between schizophrenia and 731 immune cell traits by utilizing public GWAS data. We further validated the causal relationship between schizophrenia and six types of white cell measures. Results We found the overall causal effects of schizophrenia on immune cell traits were significantly higher than the reverse ones (0.011 ± 0.049 vs 0.001 ± 0.016, p < 0.001), implying that disease may lead to an increase in immune cells by itself. We also identified four immune cell traits that may increase the risk of schizophrenia: CD11c+ monocyte %monocyte (odds ratio (OR): 1.06, 95% confidence interval (CI): 1.03~1.09, FDR = 0.027), CD11c+ CD62L- monocyte %monocyte (OR:1.06, 95% CI: 1.03~1.09, FDR = 0.027), CD25 on IgD+ CD38- naive B cell (OR:1.03, 95% CI:1.01~1.06, FDR = 0.042), and CD86 on monocyte (OR = 1.04, 95% CI:1.01~1.06, FDR = 0.042). However, we did not detect any significant causal effects of schizophrenia on immune cell traits. Using the white blood cell traits data, we identified that schizophrenia increases the lymphocyte counts (OR:1.03, 95%CI: 1.01-1.04, FDR = 0.007), total white blood cell counts (OR:1.02, 95%CI: 1.01-1.04, FDR = 0.021) and monocyte counts (OR:1.02, 95%CI: 1.00-1.03, FDR = 0.034). The lymphocyte counts were nominally associated with the risk of schizophrenia (OR:1.08,95%CI:1.01-1.16, P=0.019). Discussion Our study found that the causal relationship between schizophrenia and the immune system is complex, enhancing our understanding of the role of immune regulation in the development of this disorder. These findings offer new insights for exploring diagnostic and therapeutic options for schizophrenia.
Collapse
Affiliation(s)
- Jianbin Du
- Department of Geriatric Psychiatry, The Affiliated Mental Health Center of Jiangnan University, Wuxi Central Rehabilitation Hospital, Wuxi, Jiangsu, China
| | - Ancha Baranova
- School of Systems Biology, George Mason University, Fairfax, VA, United States
- Research Centre for Medical Genetics, Moscow, Russia
| | - Guofu Zhang
- Department of Geriatric Psychiatry, The Affiliated Mental Health Center of Jiangnan University, Wuxi Central Rehabilitation Hospital, Wuxi, Jiangsu, China
| | - Fuquan Zhang
- Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
- Institute of Neuropsychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
3
|
Hartmann SM, Heider J, Wüst R, Fallgatter AJ, Volkmer H. Microglia-neuron interactions in schizophrenia. Front Cell Neurosci 2024; 18:1345349. [PMID: 38510107 PMCID: PMC10950997 DOI: 10.3389/fncel.2024.1345349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 02/21/2024] [Indexed: 03/22/2024] Open
Abstract
Multiple lines of evidence implicate increased neuroinflammation mediated by glial cells to play a key role in neurodevelopmental disorders such as schizophrenia. Microglia, which are the primary innate immune cells of the brain, are crucial for the refinement of the synaptic circuitry during early brain development by synaptic pruning and the regulation of synaptic plasticity during adulthood. Schizophrenia risk factors as genetics or environmental influences may further be linked to increased activation of microglia, an increase of pro-inflammatory cytokine levels and activation of the inflammasome resulting in an overall elevated neuroinflammatory state in patients. Synaptic loss, one of the central pathological hallmarks of schizophrenia, is believed to be due to excess removal of synapses by activated microglia, primarily affecting glutamatergic neurons. Therefore, it is crucial to investigate microglia-neuron interactions, which has been done by multiple studies focusing on post-mortem brain tissues, brain imaging, animal models and patient iPSC-derived 2D culture systems. In this review, we summarize the major findings in patients and in vivo and in vitro models in the context of neuron-microglia interactions in schizophrenia and secondly discuss the potential of anti-inflammatory treatments for the alleviation of positive, negative, and cognitive symptoms.
Collapse
Affiliation(s)
- Sophia-Marie Hartmann
- Molecular Neurobiology, Department of Pharma and Biotech, NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Johanna Heider
- Molecular Neurobiology, Department of Pharma and Biotech, NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Richard Wüst
- Department of Psychiatry, Tübingen Center for Mental Health (TüCMH), University of Tübingen, Tübingen, Germany
| | - Andreas J. Fallgatter
- Department of Psychiatry, Tübingen Center for Mental Health (TüCMH), University of Tübingen, Tübingen, Germany
| | - Hansjürgen Volkmer
- Molecular Neurobiology, Department of Pharma and Biotech, NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| |
Collapse
|
4
|
Rittmannsberger H, Barth M, Lamprecht B, Malik P, Yazdi-Zorn K. [Interaction of somatic findings and psychiatric symptoms in COVID-19. A scoping review]. NEUROPSYCHIATRIE : KLINIK, DIAGNOSTIK, THERAPIE UND REHABILITATION : ORGAN DER GESELLSCHAFT OSTERREICHISCHER NERVENARZTE UND PSYCHIATER 2024; 38:1-23. [PMID: 38055146 DOI: 10.1007/s40211-023-00487-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 10/24/2023] [Indexed: 12/07/2023]
Abstract
An infection with SARS-CoV‑2 can affect the central nervous system, leading to neurological as well as psychiatric symptoms. In this respect, mechanisms of inflammation seem to be of much greater importance than the virus itself. This paper deals with the possible contributions of organic changes to psychiatric symptomatology and deals especially with delirium, cognitive symptoms, depression, anxiety, posttraumatic stress disorder and psychosis. Processes of neuroinflammation with infection of capillary endothelial cells and activation of microglia and astrocytes releasing high amounts of cytokines seem to be of key importance in all kinds of disturbances. They can lead to damage in grey and white matter, impairment of cerebral metabolism and loss of connectivity. Such neuroimmunological processes have been described as a organic basis for many psychiatric disorders, as affective disorders, psychoses and dementia. As the activation of the glia cells can persist for a long time after the offending agent has been cleared, this can contribute to long term sequalae of the infection.
Collapse
Affiliation(s)
- Hans Rittmannsberger
- Abteilung Psychiatrie und psychotherapeutische Medizin, Pyhrn-Eisenwurzen Klinikum Steyr, Steyr, Österreich.
| | - Martin Barth
- Abteilung Psychiatrie und psychotherapeutische Medizin, Pyhrn-Eisenwurzen Klinikum Steyr, Steyr, Österreich
| | - Bernd Lamprecht
- Med Campus III, Universitätsklinik für Innere Medizin mit Schwerpunkt Pneumologie, Kepler Universitätsklinikum GmbH, Linz, Österreich
- Medizinische Fakultät, Johannes Kepler Universität Linz, Linz, Österreich
| | - Peter Malik
- Abteilung Psychiatrie und psychotherapeutische Medizin, Pyhrn-Eisenwurzen Klinikum Steyr, Steyr, Österreich
| | - Kurosch Yazdi-Zorn
- Neuromed Campus, Klinik für Psychiatrie mit Schwerpunkt Suchtmedizin, Kepler Universitätsklinikum GmbH, Linz, Österreich
- Medizinische Fakultät, Johannes Kepler Universität Linz, Linz, Österreich
| |
Collapse
|
5
|
Xenaki LA, Dimitrakopoulos S, Selakovic M, Stefanis N. Stress, Environment and Early Psychosis. Curr Neuropharmacol 2024; 22:437-460. [PMID: 37592817 PMCID: PMC10845077 DOI: 10.2174/1570159x21666230817153631] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 01/02/2023] [Accepted: 01/04/2023] [Indexed: 08/19/2023] Open
Abstract
Existing literature provides extended evidence of the close relationship between stress dysregulation, environmental insults, and psychosis onset. Early stress can sensitize genetically vulnerable individuals to future stress, modifying their risk for developing psychotic phenomena. Neurobiological substrate of the aberrant stress response to hypothalamic-pituitary-adrenal axis dysregulation, disrupted inflammation processes, oxidative stress increase, gut dysbiosis, and altered brain signaling, provides mechanistic links between environmental risk factors and the development of psychotic symptoms. Early-life and later-life exposures may act directly, accumulatively, and repeatedly during critical neurodevelopmental time windows. Environmental hazards, such as pre- and perinatal complications, traumatic experiences, psychosocial stressors, and cannabis use might negatively intervene with brain developmental trajectories and disturb the balance of important stress systems, which act together with recent life events to push the individual over the threshold for the manifestation of psychosis. The current review presents the dynamic and complex relationship between stress, environment, and psychosis onset, attempting to provide an insight into potentially modifiable factors, enhancing resilience and possibly influencing individual psychosis liability.
Collapse
Affiliation(s)
- Lida-Alkisti Xenaki
- First Department of Psychiatry, School of Medicine, Eginition Hospital, National and Kapodistrian University of Athens, 72 Vas. Sophias Ave., Athens, 115 28, Greece
| | - Stefanos Dimitrakopoulos
- First Department of Psychiatry, School of Medicine, Eginition Hospital, National and Kapodistrian University of Athens, 72 Vas. Sophias Ave., Athens, 115 28, Greece
| | - Mirjana Selakovic
- First Department of Psychiatry, School of Medicine, Eginition Hospital, National and Kapodistrian University of Athens, 72 Vas. Sophias Ave., Athens, 115 28, Greece
| | - Nikos Stefanis
- First Department of Psychiatry, School of Medicine, Eginition Hospital, National and Kapodistrian University of Athens, 72 Vas. Sophias Ave., Athens, 115 28, Greece
| |
Collapse
|
6
|
Weyer MP, Strehle J, Schäfer MKE, Tegeder I. Repurposing of pexidartinib for microglia depletion and renewal. Pharmacol Ther 2024; 253:108565. [PMID: 38052308 DOI: 10.1016/j.pharmthera.2023.108565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 11/20/2023] [Accepted: 11/22/2023] [Indexed: 12/07/2023]
Abstract
Pexidartinib (PLX3397) is a small molecule receptor tyrosine kinase inhibitor of colony stimulating factor 1 receptor (CSF1R) with moderate selectivity over other members of the platelet derived growth factor receptor family. It is approved for treatment of tenosynovial giant cell tumors (TGCT). CSF1R is highly expressed by microglia, which are macrophages of the central nervous system (CNS) that defend the CNS against injury and pathogens and contribute to synapse development and plasticity. Challenged by pathogens, apoptotic cells, debris, or inflammatory molecules they adopt a responsive state to propagate the inflammation and eventually return to a homeostatic state. The phenotypic switch may fail, and disease-associated microglia contribute to the pathophysiology in neurodegenerative or neuropsychiatric diseases or long-lasting detrimental brain inflammation after brain, spinal cord or nerve injury or ischemia/hemorrhage. Microglia also contribute to the growth permissive tumor microenvironment of glioblastoma (GBM). In rodents, continuous treatment for 1-2 weeks via pexidartinib food pellets leads to a depletion of microglia and subsequent repopulation from the remaining fraction, which is aided by peripheral monocytes that search empty niches for engraftment. The putative therapeutic benefit of such microglia depletion or forced renewal has been assessed in almost any rodent model of CNS disease or injury or GBM with heterogeneous outcomes, but a tendency of partial beneficial effects. So far, microglia monitoring e.g. via positron emission imaging is not standard of care for patients receiving Pexidartinib (e.g. for TGCT), so that the depletion and repopulation efficiency in humans is still largely unknown. Considering the virtuous functions of microglia, continuous depletion is likely no therapeutic option but short-lasting transient partial depletion to stimulate microglia renewal or replace microglia in genetic disease in combination with e.g. stem cell transplantation or as part of a multimodal concept in treatment of glioblastoma appears feasible. The present review provides an overview of the preclinical evidence pro and contra microglia depletion as a therapeutic approach.
Collapse
Affiliation(s)
- Marc-Philipp Weyer
- Institute of Clinical Pharmacology, Goethe-University Frankfurt, Faculty of Medicine, Frankfurt, Germany
| | - Jenny Strehle
- Department of Anesthesiology, University Medical Center Johannes Gutenberg-University Mainz, Germany
| | - Michael K E Schäfer
- Department of Anesthesiology, University Medical Center Johannes Gutenberg-University Mainz, Germany
| | - Irmgard Tegeder
- Institute of Clinical Pharmacology, Goethe-University Frankfurt, Faculty of Medicine, Frankfurt, Germany.
| |
Collapse
|
7
|
Bansal S, Bae GY, Robinson BM, Dutterer J, Hahn B, Luck SJ, Gold JM. Qualitatively Different Delay-Dependent Working Memory Distortions in People With Schizophrenia and Healthy Control Participants. BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2023; 8:1218-1227. [PMID: 37459911 PMCID: PMC10792108 DOI: 10.1016/j.bpsc.2023.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 06/30/2023] [Accepted: 07/08/2023] [Indexed: 07/28/2023]
Abstract
BACKGROUND Impairments in working memory (WM) have been well documented in people with schizophrenia (PSZ). However, these quantitative WM impairments can often be explained by nonspecific factors, such as impaired goal maintenance. Here, we used a spatial orientation delayed response task to explore a qualitative difference in WM dynamics between PSZ and healthy control participants (HCs). More specifically, we took advantage of the discovery that WM representations may drift either toward or away from previous trial targets (serial dependence). We tested the hypothesis that WM representations would drift toward the previous trial target in HCs but away from the previous trial target in PSZ. METHODS We assessed serial dependence in PSZ (n = 31) and HCs (n = 25) using orientation as the to-be-remembered feature and memory delays lasting from 0 to 8 seconds. Participants were asked to remember the orientation of a teardrop-shaped object and reproduce the orientation after a delay period of varying length. RESULTS Consistent with prior studies, we found that current trial memory representations were less precise in PSZ than in HCs. We also found that WM for the current trial orientation drifted toward the previous trial orientation in HCs (representational attraction) but drifted away from the previous trial orientation in PSZ (representational repulsion). CONCLUSIONS These results demonstrate a qualitative difference in WM dynamics between PSZ and HCs that cannot be easily explained by nuisance factors such as reduced effort. Most computational neuroscience models also fail to explain these results because they maintain information solely by means of sustained neural firing, which does not extend across trials. The results suggest a fundamental difference between PSZ and HCs in longer-term memory mechanisms that persist across trials, such as short-term potentiation and neuronal adaptation.
Collapse
Affiliation(s)
- Sonia Bansal
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland, School of Medicine, Baltimore, Maryland.
| | - Gi-Yeul Bae
- Department of Psychology, Arizona State University, Tempe, Arizona
| | - Benjamin M Robinson
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland, School of Medicine, Baltimore, Maryland
| | - Jenna Dutterer
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland, School of Medicine, Baltimore, Maryland
| | - Britta Hahn
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland, School of Medicine, Baltimore, Maryland
| | - Steven J Luck
- Center for Mind & Brain and Department of Psychology, University of California, Davis, Davis, California
| | - James M Gold
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland, School of Medicine, Baltimore, Maryland
| |
Collapse
|
8
|
Jonker I, Doorduin J, Knegtering H, van't Hag E, Dierckx RA, de Vries EFJ, Schoevers RA, Klein HC. Antiviral treatment in schizophrenia: a randomized pilot PET study on the effects of valaciclovir on neuroinflammation. Psychol Med 2023; 53:7087-7095. [PMID: 37016791 PMCID: PMC10719624 DOI: 10.1017/s0033291723000430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 01/22/2023] [Accepted: 02/03/2023] [Indexed: 04/06/2023]
Abstract
BACKGROUND Patients with schizophrenia experience cognitive impairment, which could be related to neuroinflammation in the hippocampus. The cause for such hippocampal inflammation is still unknown, but it has been suggested that herpes virus infection is involved. This study therefore aimed to determine whether add-on treatment of schizophrenic patients with the anti- viral drug valaciclovir would reduce hippocampal neuroinflammation and consequently improve cognitive symptoms. METHODS We performed a double-blind monocenter study in 24 male and female patients with schizophrenia, experiencing active psychotic symptoms. Patients were orally treated with the anti-viral drug valaciclovir for seven consecutive days (8 g/day). Neuroinflammation was measured with Positron Emission Tomography using the translocator protein ligand [11C]-PK11195, pre-treatment and at seven days post-treatment, as were psychotic symptoms and cognition. RESULTS Valaciclovir treatment resulted in reduced TSPO binding (39%) in the hippocampus, as well as in the brainstem, frontal lobe, temporal lobe, parahippocampal gyrus, amygdala, parietal lobe, occipital lobe, insula and cingulate gyri, nucleus accumbens and thalamus (31-40%) when using binding potential (BPND) as an outcome. With total distribution volume (VT) as outcome we found essentially the same results, but associations only approached statistical significance (p = 0.050 for hippocampus). Placebo treatment did not affect neuroinflammation. No effects of valaciclovir on psychotic symptoms or cognitive functioning were found. CONCLUSION We found a decreased TSPO binding following antiviral treatment, which could suggest a viral underpinning of neuroinflammation in psychotic patients. Whether this reduced neuroinflammation by treatment with valaciclovir has clinical implications and is specific for schizophrenia warrants further research.
Collapse
Affiliation(s)
- Iris Jonker
- Department of Psychiatry, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Janine Doorduin
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Henderikus Knegtering
- Department of Psychiatry, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Lentis Mental Health Institution, Groningen, The Netherlands
| | - Erna van't Hag
- Department of Psychiatry, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Rudi A. Dierckx
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Erik F. J. de Vries
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Robert A. Schoevers
- Department of Psychiatry, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Hans C. Klein
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
9
|
Litke R, Vicari J, Huang BT, Shapiro L, Roh KH, Silver A, Talreja P, Palacios N, Yoon Y, Kellner C, Kaniskan H, Vangeti S, Jin J, Ramos-Lopez I, Mobbs C. Novel small molecules inhibit proteotoxicity and inflammation: Mechanistic and therapeutic implications for Alzheimer's Disease, healthspan and lifespan- Aging as a consequence of glycolysis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.12.544352. [PMID: 37398396 PMCID: PMC10312632 DOI: 10.1101/2023.06.12.544352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Inflammation drives many age-related, especially neurological, diseases, and likely mediates age-related proteotoxicity. For example, dementia due to Alzheimer's Disease (AD), cerebral vascular disease, many other neurodegenerative conditions is increasingly among the most devastating burdens on the American (and world) health system and threatens to bankrupt the American health system as the population ages unless effective treatments are developed. Dementia due to either AD or cerebral vascular disease, and plausibly many other neurodegenerative and even psychiatric conditions, is driven by increased age-related inflammation, which in turn appears to mediate Abeta and related proteotoxic processes. The functional significance of inflammation during aging is also supported by the fact that Humira, which is simply an antibody to the pro-inflammatory cytokine TNF-a, is the best-selling drug in the world by revenue. These observations led us to develop parallel high-throughput screens to discover small molecules which inhibit age-related Abeta proteotoxicity in a C. elegans model of AD AND LPS-induced microglial TNF-a. In the initial screen of 2560 compounds (Microsource Spectrum library) to delay Abeta proteotoxicity, the most protective compounds were, in order, phenylbutyrate, methicillin, and quetiapine, which belong to drug classes (HDAC inhibitors, beta lactam antibiotics, and tricyclic antipsychotics, respectably) already robustly implicated as promising to protect in neurodegenerative diseases, especially AD. RNAi and chemical screens indicated that the protective effects of HDAC inhibitors to reduce Abeta proteotoxicity are mediated by inhibition of HDAC2, also implicated in human AD, dependent on the HAT Creb binding protein (Cbp), which is also required for the protective effects of both dietary restriction and the daf-2 mutation (inactivation of IGF-1 signaling) during aging. In addition to methicillin, several other beta lactam antibiotics also delayed Abeta proteotoxicity and reduced microglial TNF-a. In addition to quetiapine, several other tricyclic antipsychotic drugs also delayed age-related Abeta proteotoxicity and increased microglial TNF-a, leading to the synthesis of a novel congener, GM310, which delays Abeta as well as Huntingtin proteotoxicity, inhibits LPS-induced mouse and human microglial and monocyte TNF-a, is highly concentrated in brain after oral delivery with no apparent toxicity, increases lifespan, and produces molecular responses highly similar to those produced by dietary restriction, including induction of Cbp inhibition of inhibitors of Cbp, and genes promoting a shift away from glycolysis and toward metabolism of alternate (e.g., lipid) substrates. GM310, as well as FDA-approved tricyclic congeners, prevented functional impairments and associated increase in TNF-a in a mouse model of stroke. Robust reduction of glycolysis by GM310 was functionally corroborated by flux analysis, and the glycolytic inhibitor 2-DG inhibited microglial TNF-a and other markers of inflammation, delayed Abeta proteotoxicity, and increased lifespan. These results support the value of phenotypic screens to discover drugs to treat age-related, especially neurological and even psychiatric diseases, including AD and stroke, and to clarify novel mechanisms driving neurodegeneration (e.g., increased microglial glycolysis drives neuroinflammation and subsequent neurotoxicity) suggesting novel treatments (selective inhibitors of microglial glycolysis).
Collapse
|
10
|
Pignon B, Decio V, Pirard P, Bouaziz O, Corruble E, Geoffroy PA, Kovess-Masfety V, Leboyer M, Lemogne C, Messika J, Perduca V, Schürhoff F, Regnault N, Tebeka S. The risk of hospitalization for psychotic disorders following hospitalization for COVID-19: a French nationwide longitudinal study. Mol Psychiatry 2023; 28:3293-3304. [PMID: 37537285 DOI: 10.1038/s41380-023-02207-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 07/24/2023] [Accepted: 07/26/2023] [Indexed: 08/05/2023]
Abstract
COVID-19, like other infectious diseases, may be a risk factor for psychotic disorders. We aimed to compare the proportions of hospitalizations for psychotic disorders in the 12 months following discharge from hospital for either COVID-19 or for another reason in the adult general population in France during the first wave of the pandemic. We conducted a retrospective longitudinal nationwide study using the national French administrative healthcare database. Psychotic disorders were first studied as a whole, and then chronic and acute disorders separately. The role of several adjustment factors, including sociodemographics, a history of psychotic disorder, the duration of the initial hospitalization, and the level of care received during that hospitalization, were also analyzed. Between 1 January 2020 and 30 June 2020, a total of 14,622 patients were hospitalized for psychotic disorders in the 12 months following discharge from hospital for either COVID-19 or another reason. Initial hospitalization for COVID-19 (vs. another reason) was associated with a lower rate of subsequent hospitalization for psychotic disorders (0.31% vs. 0.51%, odds ratio (OR) = 0.60, 95% confidence interval (CI) [0.53-0.67]). This was true for both chronic and acute disorders, even after adjusting for the various study variables. Importantly, a history of psychotic disorder was a major determinant of hospitalization for psychotic disorders (adjusted OR = 126.56, 95% CI [121.85-131.46]). Our results suggest that, in comparison to individuals initially hospitalized for another reason, individuals initially hospitalized for COVID-19 present a lower risk of hospitalization for first episodes of psychotic symptoms/disorders or for psychotic relapse in the 12 months following discharge. This finding contradicts the hypothesis that there is a higher risk of psychotic disorders after a severe COVID-19.
Collapse
Affiliation(s)
- Baptiste Pignon
- Univ Paris-Est-Créteil (UPEC), AP-HP, Hôpitaux Universitaires « H. Mondor », DMU IMPACT, INSERM, IMRB, translational Neuropsychiatry, Fondation FondaMental, F-94010, Creteil, France.
| | - Valentina Decio
- Santé publique France, French National Public Health Agency, Non Communicable Diseases and Trauma Division, F-94415, Saint-Maurice, France
| | - Philippe Pirard
- Santé publique France, French National Public Health Agency, Non Communicable Diseases and Trauma Division, F-94415, Saint-Maurice, France
| | | | - Emmanuelle Corruble
- CESP, MOODS Team, INSERM UMR 1018, Faculté de Médecine, Univ Paris-Saclay, Le Kremlin Bicêtre, F-94275, France
- Service Hospitalo-Universitaire de Psychiatrie de Bicêtre, Hôpitaux Universitaires Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Hôpital de Bicêtre, Le Kremlin Bicêtre, F-94275, France
| | - Pierre A Geoffroy
- Département de psychiatrie et d'addictologie, AP-HP, GHU Paris Nord, DMU Neurosciences, Hopital Bichat - Claude Bernard, F-75018, Paris, France
- GHU Paris - Psychiatry & Neurosciences, 1 rue Cabanis, 75014, Paris, France
- Université Paris Cité, NeuroDiderot, Inserm, FHU I2-D2, F-75019, Paris, France
| | | | - Marion Leboyer
- Univ Paris-Est-Créteil (UPEC), AP-HP, Hôpitaux Universitaires « H. Mondor », DMU IMPACT, INSERM, IMRB, translational Neuropsychiatry, Fondation FondaMental, F-94010, Creteil, France
| | - Cédric Lemogne
- Université Paris Cité and Université Sorbonne Paris Nord, Inserm, INRAE, Center for Research in Epidemiology and StatisticS (CRESS), F-75004, Paris, France
- Service de Psychiatrie de l'adulte, AP-HP, Hôpital Hôtel-Dieu, F-75004, Paris, France
| | - Jonathan Messika
- APHP.Nord-Université Paris Cité, Hôpital Bichat-Claude Bernard, Service de Pneumologie B et Transplantation Pulmonaire, Paris, France
- Physiopathology and Epidemiology of Respiratory Diseases, UMR1152 INSERM and Université de Paris, Paris, France
| | | | - Franck Schürhoff
- Univ Paris-Est-Créteil (UPEC), AP-HP, Hôpitaux Universitaires « H. Mondor », DMU IMPACT, INSERM, IMRB, translational Neuropsychiatry, Fondation FondaMental, F-94010, Creteil, France
| | - Nolwenn Regnault
- Santé publique France, French National Public Health Agency, Non Communicable Diseases and Trauma Division, F-94415, Saint-Maurice, France
| | - Sarah Tebeka
- Santé publique France, French National Public Health Agency, Non Communicable Diseases and Trauma Division, F-94415, Saint-Maurice, France
| |
Collapse
|
11
|
Long Y, Wang Y, Shen Y, Huang J, Li Y, Wu R, Zhao J. Minocycline and antipsychotics inhibit inflammatory responses in BV-2 microglia activated by LPS via regulating the MAPKs/ JAK-STAT signaling pathway. BMC Psychiatry 2023; 23:514. [PMID: 37464316 DOI: 10.1186/s12888-023-05014-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 07/08/2023] [Indexed: 07/20/2023] Open
Abstract
BACKGROUND Abnormal activation of microglia is involved in the pathogenesis of schizophrenia. Minocycline and antipsychotics have been reported to be effective in inhibiting the activation of microglia and thus alleviating the negative symptoms of patients with schizophrenia. However, the specific molecular mechanism by which minocycline and antipsychotics inhibit microglial activation is not clear. In this study, we aimed to explore the molecular mechanism of treatment effect of minocycline and antipsychotics on schizophrenia. METHODS Microglia cells were activated by lipopolysaccharide (LPS) and further treated with minocycline, haloperidol, and risperidone. Then cell morphology, specific marker, cytokines, and nitric oxide production process, and the proteins in related molecular signaling pathways in LPS-activated microglia were compared among groups. RESULTS The study found that minocycline, risperidone, and haloperidol significantly inhibited morphological changes and reduced the expression of OX-42 protein induced by LPS. Minocycline significantly decreased the production of interleukin-6 (IL-6), tumor necrosis factor-alpha (TNF-α), and interleukin-1beta (IL-1β). Risperidone also showed significant decrease in the production of IL-6 and TNF-α, while haloperidol only showed significant decrease in the production of IL-6. Minocycline, risperidone, and haloperidol were found to significantly inhibit nitric oxide (NO) expression, but had no effect on inducible nitric oxide synthase (iNOS) expression. Both minocycline and risperidone were effective in decreasing the activity of c‑Jun N‑terminal kinase (JNK) and extracellular signal-regulated kinase (ERK) in the mitogen-activated protein kinases (MAPKs) signal pathway. Additionally, minocycline and risperidone were found to increase the activity of phosphorylated-p38. In contrast, haloperidol only suppressed the activity of ERK. Minocycline also suppressed the activation of janus kinase 2 (JAK2) and signal transducer and activator of transcription 3 (STAT3), while risperidone and haloperidol only suppressed the activation of STAT3. CONCLUSIONS The results demonstrated that minocycline and risperidone exert stronger anti-inflammatory and neuroprotective effects stronger than haloperidol, through MAPKs and Janus kinase-signal transducer and activator of transcription (JAK-STAT) signaling pathways in BV2 cells stimulated with LPS, revealing the underlying mechanisms of minocycline and atypical antipsychotics in the treatment of negative schizophrenia symptoms.
Collapse
Affiliation(s)
- Yujun Long
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Ying Wang
- Mental Health Center of Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yidong Shen
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Jing Huang
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Yamin Li
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Renrong Wu
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China.
| | - Jingping Zhao
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China.
| |
Collapse
|
12
|
Delorme TC, Ozell-Landry W, Cermakian N, Srivastava LK. Behavioral and cellular responses to circadian disruption and prenatal immune activation in mice. Sci Rep 2023; 13:7791. [PMID: 37179433 PMCID: PMC10182998 DOI: 10.1038/s41598-023-34363-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 04/28/2023] [Indexed: 05/15/2023] Open
Abstract
Most individuals with neurodevelopmental disorders (NDDs), including schizophrenia and autism spectrum disorders, experience disruptions in sleep and circadian rhythms. Epidemiological studies indicate that exposure to prenatal infection increases the risk of developing NDDs. We studied how environmental circadian disruption contributes to NDDs using maternal immune activation (MIA) in mice, which models prenatal infection. Pregnant dams were injected with viral mimetic poly IC (or saline) at E9.5. Adult poly IC- and saline-exposed offspring were subjected to 4 weeks of each of the following: standard lighting (LD1), constant light (LL) and standard lighting again (LD2). Behavioral tests were conducted in the last 12 days of each condition. Poly IC exposure led to significant behavioral differences, including reduced sociability (males only) and deficits in prepulse inhibition. Interestingly, poly IC exposure led to reduced sociability specifically when males were tested after LL exposure. Mice were exposed again to either LD or LL for 4 weeks and microglia were characterized. Notably, poly IC exposure led to increased microglial morphology index and density in dentate gyrus, an effect attenuated by LL exposure. Our findings highlight interactions between circadian disruption and prenatal infection, which has implications in informing the development of circadian-based therapies for individuals with NDDs.
Collapse
Affiliation(s)
- Tara C Delorme
- Douglas Mental Health University Institute, 6875 Boulevard LaSalle, Montréal, QC, H4H 1R3, Canada
- Integrated Program in Neuroscience, McGill University, Montréal, QC, H3A 2B4, Canada
| | - William Ozell-Landry
- Douglas Mental Health University Institute, 6875 Boulevard LaSalle, Montréal, QC, H4H 1R3, Canada
| | - Nicolas Cermakian
- Douglas Mental Health University Institute, 6875 Boulevard LaSalle, Montréal, QC, H4H 1R3, Canada.
- Department of Psychiatry, McGill University, Montréal, QC, H3A 1A1, Canada.
| | - Lalit K Srivastava
- Douglas Mental Health University Institute, 6875 Boulevard LaSalle, Montréal, QC, H4H 1R3, Canada.
- Department of Psychiatry, McGill University, Montréal, QC, H3A 1A1, Canada.
| |
Collapse
|
13
|
Jensen SB, Sheikh MA, Akkouh IA, Szabo A, O’Connell KS, Lekva T, Engh JA, Agartz I, Elvsåshagen T, Ormerod MBEG, Weibell MA, Johnsen E, Kroken RA, Melle I, Drange OK, Nærland T, Vaaler AE, Westlye LT, Aukrust P, Djurovic S, Eiel Steen N, Andreassen OA, Ueland T. Elevated Systemic Levels of Markers Reflecting Intestinal Barrier Dysfunction and Inflammasome Activation Are Correlated in Severe Mental Illness. Schizophr Bull 2023; 49:635-645. [PMID: 36462169 PMCID: PMC10154716 DOI: 10.1093/schbul/sbac191] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/07/2022]
Abstract
BACKGROUND AND HYPOTHESIS Gut microbiota alterations have been reported in severe mental illness (SMI) but fewer studies have probed for signs of gut barrier disruption and inflammation. We hypothesized that gut leakage of microbial products due to intestinal inflammation could contribute to systemic inflammasome activation in SMI. STUDY DESIGN We measured plasma levels of the chemokine CCL25 and soluble mucosal vascular addressin cell adhesion molecule-1 (sMAdCAM-1) as markers of T cell homing, adhesion and inflammation in the gut, lipopolysaccharide binding protein (LBP) and intestinal fatty acid binding protein (I-FABP) as markers of bacterial translocation and gut barrier dysfunction, in a large SMI cohort (n = 567) including schizophrenia (SCZ, n = 389) and affective disorder (AFF, n = 178), relative to healthy controls (HC, n = 418). We assessed associations with plasma IL-18 and IL-18BPa and leukocyte mRNA expression of NLRP3 and NLRC4 as markers of inflammasome activation. STUDY RESULTS Our main findings were: (1) higher levels of sMAdCAM-1 (P = .002), I-FABP (P = 7.6E-11), CCL25 (P = 9.6E-05) and LBP (P = 2.6E-04) in SMI compared to HC in age, sex, BMI, CRP and freezer storage time adjusted analysis; (2) the highest levels of sMAdCAM-1 and CCL25 (both P = 2.6E-04) were observed in SCZ and I-FABP (P = 2.5E-10) and LBP (3) in AFF; and (3), I-FABP correlated with IL-18BPa levels and LBP correlated with NLRC4. CONCLUSIONS Our findings support that intestinal barrier inflammation and dysfunction in SMI could contribute to systemic inflammation through inflammasome activation.
Collapse
Affiliation(s)
- Søren B Jensen
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
| | - Mashhood A Sheikh
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
| | - Ibrahim A Akkouh
- Division of Mental Health and Addiction, Norwegian Centre for Mental Disorders Research, NORMENT, Oslo University Hospital, Oslo, Norway
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Attila Szabo
- Division of Mental Health and Addiction, Norwegian Centre for Mental Disorders Research, NORMENT, Oslo University Hospital, Oslo, Norway
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
- K.G. Jebsen Center for Neurodevelopmental disorders, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Kevin S O’Connell
- Division of Mental Health and Addiction, Norwegian Centre for Mental Disorders Research, NORMENT, Oslo University Hospital, Oslo, Norway
| | - Tove Lekva
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
| | - John A Engh
- Division of Mental Health and Addiction, Norwegian Centre for Mental Disorders Research, NORMENT, Oslo University Hospital, Oslo, Norway
- Division of Mental health and Addiction, Vestfold Hospital Trust, Tønsberg, Norway
| | - Ingrid Agartz
- K.G. Jebsen Center for Neurodevelopmental disorders, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm Health Care Services, Stockholm County Council, Stockholm, Sweden
- Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
- Norwegian Centre for Mental Disorders Research, NORMENT, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Torbjørn Elvsåshagen
- Division of Mental Health and Addiction, Norwegian Centre for Mental Disorders Research, NORMENT, Oslo University Hospital, Oslo, Norway
| | - Monica B E G Ormerod
- Division of Mental Health and Addiction, Norwegian Centre for Mental Disorders Research, NORMENT, Oslo University Hospital, Oslo, Norway
- Norwegian Centre for Mental Disorders Research, NORMENT, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Melissa A Weibell
- Division of Psychiatry, Network for Clinical Psychosis Research, Stavanger University Hospital, Stavanger, Norway
- Network for Medical Sciences, Faculty of Health, University of Stavanger, Stavanger, Norway
| | - Erik Johnsen
- Division of Psychiatry, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
- NORMENT Center of Excellence, University of Bergen and Haukeland University Hospital, Bergen, Norway
| | - Rune A Kroken
- Division of Psychiatry, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
- NORMENT Center of Excellence, University of Bergen and Haukeland University Hospital, Bergen, Norway
| | - Ingrid Melle
- Division of Mental Health and Addiction, Norwegian Centre for Mental Disorders Research, NORMENT, Oslo University Hospital, Oslo, Norway
- Norwegian Centre for Mental Disorders Research, NORMENT, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ole K Drange
- Department of Mental Health, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Østmarka, Division of Mental Health, St. Olavs University Hospital, Trondheim, Norway
- Department of Psychiatry, Sørlandet Hospital, Kristiansand, Norway
| | - Terje Nærland
- K.G. Jebsen Center for Neurodevelopmental disorders, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Rare Disorders, Division of Child and Adolescent medicine, Oslo University Hospital, Oslo, Norway
| | - Arne E Vaaler
- Department of Mental Health, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Østmarka, Division of Mental Health, St. Olavs University Hospital, Trondheim, Norway
| | - Lars T Westlye
- Division of Mental Health and Addiction, Norwegian Centre for Mental Disorders Research, NORMENT, Oslo University Hospital, Oslo, Norway
- K.G. Jebsen Center for Neurodevelopmental disorders, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Psychology, University of Oslo, Oslo, Norway
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
- Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Srdjan Djurovic
- Division of Mental Health and Addiction, Norwegian Centre for Mental Disorders Research, NORMENT, Oslo University Hospital, Oslo, Norway
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
- K.G. Jebsen Center for Neurodevelopmental disorders, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Psychology, University of Oslo, Oslo, Norway
| | - Nils Eiel Steen
- Division of Mental Health and Addiction, Norwegian Centre for Mental Disorders Research, NORMENT, Oslo University Hospital, Oslo, Norway
- Norwegian Centre for Mental Disorders Research, NORMENT, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ole A Andreassen
- Division of Mental Health and Addiction, Norwegian Centre for Mental Disorders Research, NORMENT, Oslo University Hospital, Oslo, Norway
- K.G. Jebsen Center for Neurodevelopmental disorders, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Norwegian Centre for Mental Disorders Research, NORMENT, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Thor Ueland
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- K.G. Jebsen Thrombosis Research and Expertise Center, University of Tromsø, Tromsø, Norway
| |
Collapse
|
14
|
Markiewicz-Gospodarek A, Markiewicz R, Borowski B, Dobrowolska B, Łoza B. Self-Regulatory Neuronal Mechanisms and Long-Term Challenges in Schizophrenia Treatment. Brain Sci 2023; 13:brainsci13040651. [PMID: 37190616 DOI: 10.3390/brainsci13040651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 04/04/2023] [Accepted: 04/11/2023] [Indexed: 05/17/2023] Open
Abstract
Schizophrenia is a chronic and relapsing disorder that is characterized not only by delusions and hallucinations but also mainly by the progressive development of cognitive and social deficits. These deficits are related to impaired synaptic plasticity and impaired neurotransmission in the nervous system. Currently, technological innovations and medical advances make it possible to use various self-regulatory methods to improve impaired synaptic plasticity. To evaluate the therapeutic effect of various rehabilitation methods, we reviewed methods that modify synaptic plasticity and improve the cognitive and executive processes of patients with a diagnosis of schizophrenia. PubMed, Scopus, and Google Scholar bibliographic databases were searched with the keywords mentioned below. A total of 555 records were identified. Modern methods of schizophrenia therapy with neuroplastic potential, including neurofeedback, transcranial magnetic stimulation, transcranial direct current stimulation, vagus nerve stimulation, virtual reality therapy, and cognitive remediation therapy, were reviewed and analyzed. Since randomized controlled studies of long-term schizophrenia treatment do not exceed 2-3 years, and the pharmacological treatment itself has an incompletely estimated benefit-risk ratio, treatment methods based on other paradigms, including neuronal self-regulatory and neural plasticity mechanisms, should be considered. Methods available for monitoring neuroplastic effects in vivo (e.g., fMRI, neuropeptides in serum), as well as unfavorable parameters (e.g., features of the metabolic syndrome), enable individualized monitoring of the effectiveness of long-term treatment of schizophrenia.
Collapse
Affiliation(s)
| | - Renata Markiewicz
- Department of Neurology, Neurological and Psychiatric Nursing, Medical University of Lublin, 20-093 Lublin, Poland
| | - Bartosz Borowski
- Students Scientific Association at the Department of Human Anatomy, Medical University of Lublin, 20-090 Lublin, Poland
| | - Beata Dobrowolska
- Department of Holistic Care and Management in Nursing, Medical University of Lublin, 20-081 Lublin, Poland
| | - Bartosz Łoza
- Department of Psychiatry, Medical University of Warsaw, 02-091 Warsaw, Poland
| |
Collapse
|
15
|
Bansal S, Bae GY, Robinson BM, Dutterer J, Hahn B, Luck SJ, Gold JM. Qualitatively different delay-dependent working memory distortions in people with schizophrenia and healthy control subjects. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.04.535597. [PMID: 37066149 PMCID: PMC10104073 DOI: 10.1101/2023.04.04.535597] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
Background Impairments in working memory(WM) have been well-documented in people with schizophrenia(PSZ). However, these quantitative WM impairments can often be explained by nonspecific factors, such as impaired goal maintenance. Here, we used a spatial orientation delayed-response task to explore a qualitative difference in WM dynamics between PSZ and healthy control subjects(HCS). Specifically, we took advantage of the discovery that WM representations may drift either toward or away from previous-trial targets(serial dependence). We tested the hypothesis that WM representations drift toward the previous-trial target in HCS but away from the previous-trial target in PSZ. Methods We assessed serial dependence in PSZ(N=31) and HCS(N=25), using orientation as the to-be-remembered feature and memory delays from 0 to 8s. Participants were asked to remember the orientation of a teardrop-shaped object and reproduce the orientation after a varying delay period. Results Consistent with prior studies, we found that current-trial memory representations were less precise in PSZ than in HCS. We also found that WM for the current-trial orientation drifted toward the previous-trial orientation in HCS(representational attraction) but drifted away from the previous-trial orientation in PSZ(representational repulsion). Conclusions These results demonstrate a qualitative difference in WM dynamics between PSZ and HCS that cannot easily be explained by nuisance factors such as reduced effort. Most computational neuroscience models also fail to explain these results, because they maintain information solely by means of sustained neural firing, which does not extend across trials. The results suggest a fundamental difference between PSZ and HCS in longer-term memory mechanisms that persist across trials, such as short-term potentiation and neuronal adaptation.
Collapse
Affiliation(s)
- Sonia Bansal
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland
| | - Gi-Yeul Bae
- Department of Psychology, Arizona State University, Tempe, Arizona
| | - Benjamin M Robinson
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland
| | - Jenna Dutterer
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland
| | - Britta Hahn
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland
| | - Steven J Luck
- Center for Mind & Brain and Department of Psychology, University of California, Davis, Davis, California
| | - James M Gold
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
16
|
da Silva FER, Cordeiro RC, de Carvalho Lima CN, Cardozo PL, Vasconcelos GS, Monte AS, Sanders LLO, Vasconcelos SMM, de Lucena DF, Cruz BF, Nicolato R, Seeman MV, Ribeiro FM, Macedo DS. Sex and the Estrous-Cycle Phase Influence the Expression of G Protein-Coupled Estrogen Receptor 1 (GPER) in Schizophrenia: Translational Evidence for a New Target. Mol Neurobiol 2023; 60:3650-3663. [PMID: 36917419 DOI: 10.1007/s12035-023-03295-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 02/24/2023] [Indexed: 03/16/2023]
Abstract
Schizophrenia is a mental disorder with sex bias in disease onset and symptom severity. Recently, it was observed that females present more severe symptoms in the perimenstrual phase of the menstrual cycle. The administration of estrogen also alleviates schizophrenia symptoms. Despite this, little is known about symptom fluctuation over the menstrual cycle and the underlying mechanisms. To address this issue, we worked with the two-hit schizophrenia animal model induced by neonatal exposure to a virus-like particle, Poly I:C, associated with peripubertal unpredictable stress exposure. Prepulse inhibition of the startle reflex (PPI) in male and female mice was considered analogous to human schizophrenia-like behavior. Female mice were studied in the proestrus (high-estrogen estrous cycle phase) and diestrus (low-estrogen phase). Additionally, we evaluated the hippocampal mRNA expression of estrogen synthesis proteins; TSPO and aromatase; and estrogen receptors ERα, ERβ, and GPER. We also collected peripheral blood mononuclear cells (PBMCs) from male and female patients with schizophrenia and converted them to induced microglia-like cells (iMGs) to evaluate the expression of GPER. We observed raised hippocampal expression of GPER in two-hit female mice at the proestrus phase without PPI deficits and higher levels of proteins related to estrogen synthesis, TSPO, and aromatase. In contrast, two-hit adult males with PPI deficits presented lower hippocampal mRNA expression of TSPO, aromatase, and GPER. iMGs from male and female patients with schizophrenia showed lower mRNA expression of GPER than controls. Therefore, our results suggest that GPER alterations constitute an underlying mechanism for sex influence in schizophrenia.
Collapse
Affiliation(s)
- Francisco Eliclécio Rodrigues da Silva
- Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceara, Rua Cel. Nunes de Melo 1000, 60430-275, CE, Fortaleza, Brazil
| | - Rafaela Carneiro Cordeiro
- Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceara, Rua Cel. Nunes de Melo 1000, 60430-275, CE, Fortaleza, Brazil.,University of Texas Health Science Center at Houston, Houston, USA
| | - Camila N de Carvalho Lima
- Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceara, Rua Cel. Nunes de Melo 1000, 60430-275, CE, Fortaleza, Brazil.,University of Texas Health Science Center at Houston, Houston, USA
| | - Pablo Leal Cardozo
- Department of Biochemistry and Immunology, Institute of Biological Sciences (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Germana Silva Vasconcelos
- Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceara, Rua Cel. Nunes de Melo 1000, 60430-275, CE, Fortaleza, Brazil
| | - Aline Santos Monte
- Health Science Institute, University of International Integration of Afro-Brazilian Lusophony UNILAB, Redenção, Brazil
| | - Lia Lira Olivier Sanders
- Course of Medicine, Centro Universitário Christus-Unichristus, Fortaleza, Brazil.,Department of Clinical Medicine, Federal University of Ceara, Fortaleza, Brazil
| | - Silvânia Maria Mendes Vasconcelos
- Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceara, Rua Cel. Nunes de Melo 1000, 60430-275, CE, Fortaleza, Brazil
| | - David Freitas de Lucena
- Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceara, Rua Cel. Nunes de Melo 1000, 60430-275, CE, Fortaleza, Brazil
| | - Breno Fiuza Cruz
- Department of Mental Health, Faculty of Medicine, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Rodrigo Nicolato
- Department of Mental Health, Faculty of Medicine, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Mary V Seeman
- Department of Psychiatry, University of Toronto, Toronto, Canada
| | - Fabíola Mara Ribeiro
- Department of Biochemistry and Immunology, Institute of Biological Sciences (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Danielle S Macedo
- Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceara, Rua Cel. Nunes de Melo 1000, 60430-275, CE, Fortaleza, Brazil. .,National Institute for Translational Medicine (INCT-TM, CNPq), São Paulo, Brazil.
| |
Collapse
|
17
|
Cell Adhesion Molecules in Schizophrenia Patients with Metabolic Syndrome. Metabolites 2023; 13:metabo13030376. [PMID: 36984816 PMCID: PMC10058418 DOI: 10.3390/metabo13030376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 02/20/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023] Open
Abstract
Metabolic syndrome (MetS) is a common comorbidity of schizophrenia and significantly shortens life expectancy of the patients. Intercellular (ICAM), vascular (VCAM), and neural (NCAM) cell adhesion molecules (CAMs) mediate neuroinflammatory processes, and their soluble forms (e.g., sICAM) in plasma are present in parallel with their cell-bound forms. In this study, their serum levels were examined in 211 white Siberian patients with paranoid schizophrenia (82 patients with and 129 without MetS according to the 2005 International Diabetes Federation criteria). Serum levels of CAMs were determined with Magpix and Luminex 200 (Luminex, Austin, TX, USA) using xMAP Technology. The level of sICAM-1 was significantly higher and that of sVCAM-1 significantly lower in patients with MetS compared to patients without MetS. Levels of NCAM did not differ between the groups. More pronounced Spearman’s correlations between CAMs, age, duration of schizophrenia, and body–mass index were observed among patients without MetS than among patients with MetS. Our results are consistent with MetS’s being associated with endothelial dysfunction along with other components of inflammation. Through these endothelial components of peripheral inflammatory processes, MetS might induce intracerebral neuroinflammatory changes, but further investigation is needed to confirm this.
Collapse
|
18
|
Inflammation and cognition in severe mental illness: patterns of covariation and subgroups. Mol Psychiatry 2023; 28:1284-1292. [PMID: 36577840 PMCID: PMC10005942 DOI: 10.1038/s41380-022-01924-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 12/06/2022] [Accepted: 12/13/2022] [Indexed: 12/29/2022]
Abstract
A potential relationship between dysregulation of immune/inflammatory pathways and cognitive impairment has been suggested in severe mental illnesses (SMI), such as schizophrenia (SZ) and bipolar (BD) spectrum disorders. However, multivariate relationships between peripheral inflammatory/immune-related markers and cognitive domains are unclear, and many studies do not account for inter-individual variance in both cognitive functioning and inflammatory/immune status. This study aimed to investigate covariance patterns between inflammatory/immune-related markers and cognitive domains and further elucidate heterogeneity in a large SMI and healthy control (HC) cohort (SZ = 343, BD = 289, HC = 770). We applied canonical correlation analysis (CCA) to identify modes of maximum covariation between a comprehensive selection of cognitive domains and inflammatory/immune markers. We found that poor verbal learning and psychomotor processing speed was associated with higher levels of interleukin-18 system cytokines and beta defensin 2, reflecting enhanced activation of innate immunity, a pattern augmented in SMI compared to HC. Applying hierarchical clustering on covariance patterns identified by the CCA revealed a high cognition-low immune dysregulation subgroup with predominantly HC (24% SZ, 45% BD, 74% HC) and a low cognition-high immune dysregulation subgroup predominantly consisting of SMI patients (76% SZ, 55% BD, 26% HC). These subgroups differed in IQ, years of education, age, CRP, BMI (all groups), level of functioning, symptoms and defined daily dose (DDD) of antipsychotics (SMI cohort). Our findings suggest a link between cognitive impairment and innate immune dysregulation in a subset of individuals with severe mental illness.
Collapse
|
19
|
Skorobogatov K, Autier V, Foiselle M, Richard JR, Boukouaci W, Wu CL, Raynal S, Carbonne C, Laukens K, Meysman P, Coppens V, le Corvoisier P, Barau C, De Picker L, Morrens M, Tamouza R, Leboyer M. Kynurenine pathway abnormalities are state-specific but not diagnosis-specific in schizophrenia and bipolar disorder. Brain Behav Immun Health 2023; 27:100584. [PMID: 36685639 PMCID: PMC9852293 DOI: 10.1016/j.bbih.2022.100584] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 12/29/2022] [Indexed: 01/04/2023] Open
Abstract
Schizophrenia (SCZ) and bipolar disorder (BD) are associated with immunological dysfunctions that have been hypothesized to lead to clinical symptomatology in particular through kynurenine pathway abnormalities. The aim of this study was thus to investigate the impact of serum kynurenine metabolite levels on diagnosis, clinical state, symptom severity and clinical course in a large French transdiagnostic cohort of SCZ and BD patients. Four patient groups (total n = 507) were included in a cross-sectional observational study: 1) hospitalized acute bipolar patients (n = 205); 2) stable bipolar outpatients (n = 116); 3) hospitalized acute schizophrenia patients (n = 111) and 4) stable schizophrenia outpatients (n = 75), in addition to healthy controls (HC) (n = 185). The quantitative determination of serum kynurenine metabolites was performed using liquid chromatography-tandem mass spectrometry. Kynurenine levels were lower in all patients combined compared to HC while ANCOVA analyses did not reveal inter-diagnostic difference between SCZ and BD. Interestingly, hospitalized patients of both diagnostic groups combined displayed significantly lower kynurenine levels than stabilized outpatients. Psychotic symptoms were associated with lower quinaldic acid (F = 9.18, p=<.001), which is KAT-driven, whereas a longer duration of illness contributed to abnormalities in tryptophan (F = 5.41, p = .023), kynurenine (F = 16.93, p=<.001), xanthurenic acid (F = 9.34, p = .002), quinolinic acid (F = 9.18, p = .003) and picolinic acid (F = 4.15, p = .043), metabolized through the KMO-branch. These data confirm illness state rather than diagnosis to drive KP alterations in SCZ and BD. Lower levels of KP metabolites can thus be viewed as a transdiagnostic feature of SCZ and BD, independently associated with acute symptomatology and a longer duration of illness. Quinaldic acid has seldomly been investigated by previous studies and appears an important state marker in SCZ and BD. As serum samples are used in this study, it is not possible to extrapolate these findings to the brain.
Collapse
Affiliation(s)
- Katrien Skorobogatov
- Scientific Initiative for Neuropsychiatric and Psychopharmacological Studies (SINAPS), University Psychiatric Hospital Campus Duffel (UPCD), Duffel, Belgium
- Collaborative Antwerp Psychiatric Research Institute (CAPRI), University of Antwerp, Antwerp, Belgium
| | | | - Marianne Foiselle
- Université Paris Est Creteil, Inserm U955, IMRB Translational Neuropsychiatry Laboratory, Creteil, France
- AP-HP, Hôpitaux Universitaires H Mondor, DMU IMPACT, FHU ADAPT, Créteil, France
- Fondation FondaMental, Creteil, France
| | - Jean-Romain Richard
- Université Paris Est Creteil, Inserm U955, IMRB Translational Neuropsychiatry Laboratory, Creteil, France
- AP-HP, Hôpitaux Universitaires H Mondor, DMU IMPACT, FHU ADAPT, Créteil, France
- Fondation FondaMental, Creteil, France
| | - Wahid Boukouaci
- Université Paris Est Creteil, Inserm U955, IMRB Translational Neuropsychiatry Laboratory, Creteil, France
- AP-HP, Hôpitaux Universitaires H Mondor, DMU IMPACT, FHU ADAPT, Créteil, France
- Fondation FondaMental, Creteil, France
| | - Ching-Lien Wu
- Université Paris Est Creteil, Inserm U955, IMRB Translational Neuropsychiatry Laboratory, Creteil, France
- AP-HP, Hôpitaux Universitaires H Mondor, DMU IMPACT, FHU ADAPT, Créteil, France
- Fondation FondaMental, Creteil, France
| | | | | | - Kris Laukens
- Biomedical Informatics Research Center Antwerp (BIOMINA), University of Antwerp/Antwerp University Hospital, Antwerp, Belgium
- Department of Mathematics and Computer Science, University of Antwerp, Antwerp, Belgium
| | - Pieter Meysman
- Biomedical Informatics Research Center Antwerp (BIOMINA), University of Antwerp/Antwerp University Hospital, Antwerp, Belgium
- Department of Mathematics and Computer Science, University of Antwerp, Antwerp, Belgium
| | - Violette Coppens
- Scientific Initiative for Neuropsychiatric and Psychopharmacological Studies (SINAPS), University Psychiatric Hospital Campus Duffel (UPCD), Duffel, Belgium
- Collaborative Antwerp Psychiatric Research Institute (CAPRI), University of Antwerp, Antwerp, Belgium
| | - Philippe le Corvoisier
- Inserm, Centre d'Investigation Clinique 1430, AP-HP, Hôpital Henri Mondor, Université Paris Est Créteil, Créteil, France
| | - Caroline Barau
- Plateforme de Ressources Biologiques, HU Henri Mondor, F94010, France
| | - Livia De Picker
- Scientific Initiative for Neuropsychiatric and Psychopharmacological Studies (SINAPS), University Psychiatric Hospital Campus Duffel (UPCD), Duffel, Belgium
- Collaborative Antwerp Psychiatric Research Institute (CAPRI), University of Antwerp, Antwerp, Belgium
| | - Manuel Morrens
- Scientific Initiative for Neuropsychiatric and Psychopharmacological Studies (SINAPS), University Psychiatric Hospital Campus Duffel (UPCD), Duffel, Belgium
- Collaborative Antwerp Psychiatric Research Institute (CAPRI), University of Antwerp, Antwerp, Belgium
| | - Ryad Tamouza
- Université Paris Est Creteil, Inserm U955, IMRB Translational Neuropsychiatry Laboratory, Creteil, France
- AP-HP, Hôpitaux Universitaires H Mondor, DMU IMPACT, FHU ADAPT, Créteil, France
- Fondation FondaMental, Creteil, France
| | - Marion Leboyer
- Université Paris Est Creteil, Inserm U955, IMRB Translational Neuropsychiatry Laboratory, Creteil, France
- AP-HP, Hôpitaux Universitaires H Mondor, DMU IMPACT, FHU ADAPT, Créteil, France
- Fondation FondaMental, Creteil, France
| |
Collapse
|
20
|
Sheikh MA, O'Connell KS, Lekva T, Szabo A, Akkouh IA, Osete JR, Agartz I, Engh JA, Andreou D, Boye B, Bøen E, Elvsåshagen T, Hope S, Frogner Werner MC, Joa I, Johnsen E, Kroken RA, Lagerberg TV, Melle I, Drange OK, Morken G, Nærland T, Sørensen K, Vaaler AE, Weibell MA, Westlye LT, Aukrust P, Djurovic S, Steen NE, Andreassen OA, Ueland T. Systemic Cell Adhesion Molecules in Severe Mental Illness: Potential Role of Intercellular CAM-1 in Linking Peripheral and Neuroinflammation. Biol Psychiatry 2023; 93:187-196. [PMID: 36182530 DOI: 10.1016/j.biopsych.2022.06.029] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 06/14/2022] [Accepted: 06/14/2022] [Indexed: 12/23/2022]
Abstract
BACKGROUND Cell adhesion molecules (CAMs) orchestrate leukocyte trafficking and could link peripheral and neuroinflammation in patients with severe mental illness (SMI), by promoting inflammatory and immune-mediated responses and mediating signals across blood-brain barrier. We hypothesized that CAMs would be dysregulated in SMI and evaluated plasma levels of different vascular and neural CAMs. Dysregulated CAMs in plasma were further evaluated in vivo in leukocytes and brain tissue and in vitro in induced pluripotent stem cells. METHODS We compared plasma soluble levels of different vascular (VCAM-1, ICAM-1, P-SEL) and neural (JAM-A, NCAD) CAMs in circulating leukocytes in a large SMI sample of schizophrenia (SCZ) spectrum disorder (n = 895) and affective disorder (n = 737) and healthy control participants (n = 1070) controlling for age, sex, body mass index, C-reactive protein, and freezer storage time. We also evaluated messenger RNA expression of ICAM1 and related genes encoding ICAM-1 receptors in leukocytes using microarray (n = 842) and in available RNA sequencing data from the CommonMind Consortium (CMC) in postmortem samples from the dorsolateral prefrontal cortex (n = 474). The regulation of soluble ICAM-1 in induced pluripotent stem cell-derived neurons and astrocytes was assessed in patients with SCZ and healthy control participants (n = 8 of each). RESULTS Our major findings were 1) increased soluble ICAM-1 in patients with SMI compared with healthy control participants; 2) increased ITGB2 messenger RNA, encoding the beta chain of the ICAM-1 receptor, in circulating leukocytes from patients with SMI and increased prefrontal cortex messenger RNA expression of ICAM1 in SCZ; and 3) enhanced soluble ICAM-1 release in induced pluripotent stem cell-derived neurons from patients with SCZ. CONCLUSIONS Our results support a systemic and cerebral dysregulation of soluble ICAM-1 expression in SMI and especially in patients with SCZ.
Collapse
Affiliation(s)
- Mashhood A Sheikh
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
| | - Kevin S O'Connell
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Tove Lekva
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
| | - Attila Szabo
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Ibrahim A Akkouh
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Jordi Requena Osete
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Ingrid Agartz
- NORMENT, University of Oslo, Oslo, Norway; Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway; Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet & Stockholm Health Care Services, Stockholm County Council, Stockholm, Sweden
| | - John A Engh
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Vestfold Hospital Trust, Division of Mental Health and Addiction, Tønsberg, Norway
| | - Dimitrios Andreou
- NORMENT, University of Oslo, Oslo, Norway; Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway; Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet & Stockholm Health Care Services, Stockholm County Council, Stockholm, Sweden
| | | | | | - Torbjørn Elvsåshagen
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Sigrun Hope
- Department of Neuro Habilitation, Oslo University Hospital Ullevål, Oslo, Norway
| | - Maren Caroline Frogner Werner
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Inge Joa
- Network for Clinical Psychosis Research, Division of Psychiatry, Stavanger University Hospital, Stavanger, Norway; Network for Medical Sciences, Faculty of Health, University of Stavanger, Stavanger, Norway
| | - Erik Johnsen
- Division of Psychiatry, Haukeland University Hospital, Bergen, Norway; Department of Clinical Medicine, University of Bergen, Bergen, Norway; NORMENT Centre of Excellence, Bergen, Norway
| | - Rune A Kroken
- Division of Psychiatry, Haukeland University Hospital, Bergen, Norway; Department of Clinical Medicine, University of Bergen, Bergen, Norway; NORMENT Centre of Excellence, Bergen, Norway
| | - Trine Vik Lagerberg
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Ingrid Melle
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; NORMENT, University of Oslo, Oslo, Norway
| | - Ole Kristian Drange
- Department of Mental Health, Norwegian University of Science and Technology, Trondheim, Norway; Department of Østmarka, Division of Mental Health, St. Olavs University Hospital, Trondheim, Norway; Department of Psychiatry, Sørlandet Hospital HF, Kristiansand, Norway
| | - Gunnar Morken
- Department of Mental Health, Norwegian University of Science and Technology, Trondheim, Norway
| | | | - Kjetil Sørensen
- Department of Mental Health, Norwegian University of Science and Technology, Trondheim, Norway
| | - Arne E Vaaler
- Department of Mental Health, Norwegian University of Science and Technology, Trondheim, Norway; Department of Østmarka, Division of Mental Health, St. Olavs University Hospital, Trondheim, Norway
| | - Melissa Authen Weibell
- Network for Clinical Psychosis Research, Division of Psychiatry, Stavanger University Hospital, Stavanger, Norway; Network for Medical Sciences, Faculty of Health, University of Stavanger, Stavanger, Norway
| | - Lars T Westlye
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway; Faculty of Medicine, University of Oslo, Oslo, Norway; Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Srdjan Djurovic
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; K.G. Jebsen Center for Neurodevelopmental Disorders, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Nils Eiel Steen
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; NORMENT, University of Oslo, Oslo, Norway
| | - Ole A Andreassen
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway; NORMENT, University of Oslo, Oslo, Norway
| | - Thor Ueland
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway; Faculty of Medicine, University of Oslo, Oslo, Norway; K.G. Jebsen Thrombosis Research and Expertise Center, University of Tromsø, Tromsø, Norway.
| |
Collapse
|
21
|
Fessel J. Formulating treatment of major psychiatric disorders: algorithm targets the dominantly affected brain cell-types. DISCOVER MENTAL HEALTH 2023; 3:3. [PMID: 37861813 PMCID: PMC10501034 DOI: 10.1007/s44192-022-00029-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 12/21/2022] [Indexed: 10/21/2023]
Abstract
BACKGROUND Pharmacotherapy for most psychiatric conditions was developed from serendipitous observations of benefit from drugs prescribed for different reasons. An algorithmic approach to formulating pharmacotherapy is proposed, based upon which combination of changed activities by brain cell-types is dominant for any particular condition, because those cell-types contain and surrogate for genetic, metabolic and environmental information, that has affected their function. The algorithm performs because functions of some or all the affected cell-types benefit from several available drugs: clemastine, dantrolene, erythropoietin, fingolimod, fluoxetine, lithium, memantine, minocycline, pioglitazone, piracetam, and riluzole PROCEDURES/FINDINGS: Bipolar disorder, major depressive disorder, schizophrenia, Alzheimer's disease, and post-traumatic stress disorder, illustrate the algorithm; for them, literature reviews show that no single combination of altered cell-types accounts for all cases; but they identify, for each condition, which combination occurs most frequently, i.e., dominates, as compared with other possible combinations. Knowing the dominant combination of altered cell-types in a particular condition, permits formulation of therapy with combinations of drugs taken from the above list. The percentage of patients who might benefit from that therapy, depends upon the frequency with which the dominant combination occurs in patients with that particular condition. CONCLUSIONS Knowing the dominant combination of changed cell types in psychiatric conditions, permits an algorithmically formulated, rationally-based treatment. Different studies of the same condition often produce discrepant results; all might be correct, because identical clinical phenotypes result from different combinations of impaired cell-types, thus producing different results. Clinical trials would validate both the proposed concept and choice of drugs.
Collapse
Affiliation(s)
- Jeffrey Fessel
- Department of Medicine, University of California, 2069 Filbert Street, San Francisco, CA, 94123, USA.
| |
Collapse
|
22
|
Keshri N, Nandeesha H. Dysregulation of Synaptic Plasticity Markers in Schizophrenia. Indian J Clin Biochem 2023; 38:4-12. [PMID: 36684500 PMCID: PMC9852406 DOI: 10.1007/s12291-022-01068-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 07/05/2022] [Indexed: 01/25/2023]
Abstract
Schizophrenia is a mental disorder characterized by cognitive impairment resulting in compromised quality of life. Since the regulation of synaptic plasticity has functional implications in various aspects of cognition such as learning, memory, and neural circuit maturation, the dysregulation of synaptic plasticity is considered as a pathobiological feature of schizophrenia. The findings from our recently concluded studies indicate that there is an alteration in levels of synaptic plasticity markers such as neural cell adhesion molecule-1 (NCAM-1), Neurotropin-3 (NT-3) and Matrix-mettaloproteinase-9 (MMP-9) in schizophrenia patients. The objective of the present article is to review the role of markers of synaptic plasticity in schizophrenia. PubMed database (http;//www.ncbi.nlm.nih.gov/pubmed) was used to perform an extensive literature search using the keywords schizophrenia and synaptic plasticity. We conclude that markers of synaptic plasticity are altered in schizophrenia and may lead to complications of schizophrenia including cognitive dysfunction.
Collapse
Affiliation(s)
- Neha Keshri
- Department of Biochemistry, JIPMER, Puducherry, 605006 India
| | | |
Collapse
|
23
|
The Role of Gut Dysbiosis in the Pathophysiology of Neuropsychiatric Disorders. Cells 2022; 12:cells12010054. [PMID: 36611848 PMCID: PMC9818777 DOI: 10.3390/cells12010054] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/09/2022] [Accepted: 12/14/2022] [Indexed: 12/25/2022] Open
Abstract
Mounting evidence shows that the complex gut microbial ecosystem in the human gastrointestinal (GI) tract regulates the physiology of the central nervous system (CNS) via microbiota and the gut-brain (MGB) axis. The GI microbial ecosystem communicates with the brain through the neuroendocrine, immune, and autonomic nervous systems. Recent studies have bolstered the involvement of dysfunctional MGB axis signaling in the pathophysiology of several neurodegenerative, neurodevelopmental, and neuropsychiatric disorders (NPDs). Several investigations on the dynamic microbial system and genetic-environmental interactions with the gut microbiota (GM) have shown that changes in the composition, diversity and/or functions of gut microbes (termed "gut dysbiosis" (GD)) affect neuropsychiatric health by inducing alterations in the signaling pathways of the MGB axis. Interestingly, both preclinical and clinical evidence shows a positive correlation between GD and the pathogenesis and progression of NPDs. Long-term GD leads to overstimulation of hypothalamic-pituitary-adrenal (HPA) axis and the neuroimmune system, along with altered neurotransmitter levels, resulting in dysfunctional signal transduction, inflammation, increased oxidative stress (OS), mitochondrial dysfunction, and neuronal death. Further studies on the MGB axis have highlighted the significance of GM in the development of brain regions specific to stress-related behaviors, including depression and anxiety, and the immune system in the early life. GD-mediated deregulation of the MGB axis imbalances host homeostasis significantly by disrupting the integrity of the intestinal and blood-brain barrier (BBB), mucus secretion, and gut immune and brain immune functions. This review collates evidence on the potential interaction between GD and NPDs from preclinical and clinical data. Additionally, we summarize the use of non-therapeutic modulators such as pro-, pre-, syn- and post-biotics, and specific diets or fecal microbiota transplantation (FMT), which are promising targets for the management of NPDs.
Collapse
|
24
|
Nunes FDD, Ferezin LP, Pereira SC, Figaro-Drumond FV, Pinheiro LC, Menezes IC, Baes CVW, Coeli-Lacchini FB, Tanus-Santos JE, Juruena MF, Lacchini R. The Association of Biochemical and Genetic Biomarkers in VEGF Pathway with Depression. Pharmaceutics 2022; 14:pharmaceutics14122757. [PMID: 36559251 PMCID: PMC9785844 DOI: 10.3390/pharmaceutics14122757] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/29/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022] Open
Abstract
VEGF is an important neurotrophic and vascular factor involved in mental disorders. The objective of this study was to verify the effect of genetic polymorphisms in the VEGF pathway on the risk for depression, symptom intensity, and suicide attempts. To examine the association between the VEGF pathway and depression, we genotyped polymorphisms and measured the plasma concentrations of VEGF, KDR, and FLT1 proteins. The participants were 160 patients with depression and 114 healthy controls. The questionnaires that assessed the clinical profile of the patients were the MINI-International Neuropsychiatric Interview, GRID-HAMD21, CTQ, BSI, and the number of suicide attempts. Genotyping of participants was performed using the real-time PCR and protein measurements were performed using the enzyme-linked immunosorbent assay (ELISA). VEGF and its inhibitors were reduced in depression. Individuals with depression and displaying the homozygous AA of the rs699947 polymorphism had higher plasma concentrations of VEGF (p-value = 0.006) and were associated with a greater number of suicide attempts (p-value = 0.041). Individuals with depression that were homozygous for the G allele of the FLT1 polymorphism rs7993418 were associated with lower symptom severity (p-value = 0.040). Our results suggest that VEGF pathway polymorphisms are associated with the number of suicide attempts and the severity of depressive symptoms.
Collapse
Affiliation(s)
- Fernanda Daniela Dornelas Nunes
- Department of Psychiatric Nursing and Human Sciences, Ribeirão Preto College of Nursing, University of Sao Paolo, Sao Paulo 14040-902, Brazil
| | - Letícia Perticarrara Ferezin
- Department of Psychiatric Nursing and Human Sciences, Ribeirão Preto College of Nursing, University of Sao Paolo, Sao Paulo 14040-902, Brazil
| | - Sherliane Carla Pereira
- Department of Pharmacology, Faculty of Medicine of Ribeirao Preto, University of Sao Paulo, Sao Paolo 14049-900, Brazil
| | - Fernanda Viana Figaro-Drumond
- Department of Psychiatric Nursing and Human Sciences, Ribeirão Preto College of Nursing, University of Sao Paolo, Sao Paulo 14040-902, Brazil
| | - Lucas Cézar Pinheiro
- Department of Psychiatric Nursing and Human Sciences, Ribeirão Preto College of Nursing, University of Sao Paolo, Sao Paulo 14040-902, Brazil
| | - Itiana Castro Menezes
- Department of Neuroscience and Behavior, Ribeirao Preto Medical School, University of Sao Paulo, Sao Paulo 14049-900, Brazil
| | - Cristiane von Werne Baes
- Department of Neuroscience and Behavior, Ribeirao Preto Medical School, University of Sao Paulo, Sao Paulo 14049-900, Brazil
| | - Fernanda Borchers Coeli-Lacchini
- Blood Center Foundation, Clinics Hospital of the Faculty of Medicine of Ribeirao Preto, University of Sao Paulo, Sao Paolo 14051-060, Brazil
| | - José Eduardo Tanus-Santos
- Department of Pharmacology, Faculty of Medicine of Ribeirao Preto, University of Sao Paulo, Sao Paolo 14049-900, Brazil
| | - Mário Francisco Juruena
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King’s College London and South London and Maudsley NHS Foundation Trust, Bethlem Royal Hospital, Monks Orchard Road, Beckenham BR3 3BX, UK
| | - Riccardo Lacchini
- Department of Psychiatric Nursing and Human Sciences, Ribeirão Preto College of Nursing, University of Sao Paolo, Sao Paulo 14040-902, Brazil
- Correspondence: ; Tel.: +16-33153447
| |
Collapse
|
25
|
First-episode psychotic disorders in the wake of the COVID-19 pandemic: a descriptive review of casereports. Acta Neuropsychiatr 2022; 34:289-310. [PMID: 35357298 DOI: 10.1017/neu.2022.11] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Since the onset of COVID-19 pandemic, many case reports and case series dealt with new-onset psychotic disorders in patients either infected with SARS-CoV-2 or thematically linked to the pandemic, but without an infection. Our aim was to provide a comprehensive collection of these reports to illustrate the nature of these psychoses. METHODS We conducted a literature search in MEDLINE, Embase, PsycINFO, using search terms regarding first-episode psychotic disorders in the context of corona. RESULTS 96 case reports or case series covering 146 patients (62 without and 84 with SARS-CoV-2 infection) were found. Compared to patients without infection, patients with infection showed significantly more often visual hallucinations (28.6% vs 8.1%), confusion (36.9% vs 11.3%), an acute onset of illness (88.5% vs 59.6%) and less often depression (13.1% vs 35.5%) and a delusional content related to the pandemic (29.5% vs 78.3%). Both groups had an equally favourable outcome with a duration of psychosis ≤2 weeks in half and full remission in two-thirds of patients. In patients with infection, signs of inflammation were reported in 78.3% and increased CRP in 58.6%. While reports on patients with infection are continuously published, no report about patients without infection was found after July 2020. CONCLUSION Cases without infection were considered reactive and originated all from the first wave of the corona pandemic. In cases with infection, inflammation was considered as the main pathogenetic factor but was not found in all patients. Diagnosis was impeded by the overlap of psychosis with delirium.
Collapse
|
26
|
Elkjaer Greenwood Ormerod MB, Ueland T, Frogner Werner MC, Hjell G, Rødevand L, Sæther LS, Lunding SH, Johansen IT, Ueland T, Lagerberg TV, Melle I, Djurovic S, Andreassen OA, Steen NE. Composite immune marker scores associated with severe mental disorders and illness course. Brain Behav Immun Health 2022; 24:100483. [PMID: 35856063 PMCID: PMC9287150 DOI: 10.1016/j.bbih.2022.100483] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 06/28/2022] [Indexed: 12/29/2022] Open
Abstract
Background Low-grade inflammation has been implicated in the pathophysiology of severe mental disorders (SMDs) and a link between immune activation and clinical characteristics is suggested. However, few studies have investigated how patterns across immune markers are related to diagnosis and illness course. Methods A total of 948 participants with a diagnosis of schizophrenia (SCZ, N = 602) or bipolar (BD, N = 346) spectrum disorder, and 814 healthy controls (HC) were included. Twenty-five immune markers comprising cell adhesion molecules (CAMs), interleukin (IL)-18-system factors, defensins, chemokines and other markers, related to neuroinflammation, blood-brain barrier (BBB) function, inflammasome activation and immune cell orchestration were analyzed. Eight immune principal component (PC) scores were constructed by PC Analysis (PCA) and applied in general linear models with diagnosis and illness course characteristics. Results Three PC scores were significantly associated with a SCZ and/or BD diagnosis (HC reference), with largest, however small, effect sizes of scores based on CAMs, BBB markers and defensins (p < 0.001, partial η2 = 0.02-0.03). Number of psychotic episodes per year in SCZ was associated with a PC score based on IL-18 system markers and the potential neuroprotective cytokine A proliferation-inducing ligand (p = 0.006, partial η2 = 0.071). Conclusion Analyses of composite immune markers scores identified specific patterns suggesting CAMs-mediated BBB dysregulation pathways associated with SMDs and interrelated pro-inflammatory and neuronal integrity processes associated with severity of illness course. This suggests a complex pattern of immune pathways involved in SMDs and SCZ illness course.
Collapse
Affiliation(s)
| | - Thor Ueland
- NORMENT Centre, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- KG Jebsen Inflammatory Research Center, University of Oslo, Oslo, Norway
| | - Maren Caroline Frogner Werner
- NORMENT Centre, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Gabriela Hjell
- NORMENT Centre, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Psychiatry, Østfold Hospital, Graalum, Norway
| | - Linn Rødevand
- NORMENT Centre, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Linn Sofie Sæther
- NORMENT Centre, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Department of Psychology, University of Oslo, Oslo, Norway
| | - Synve Hoffart Lunding
- NORMENT Centre, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ingrid Torp Johansen
- NORMENT Centre, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Torill Ueland
- NORMENT Centre, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Psychology, University of Oslo, Oslo, Norway
| | - Trine Vik Lagerberg
- NORMENT Centre, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Ingrid Melle
- NORMENT Centre, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Srdjan Djurovic
- Department of Medical Genetics, Oslo University Hospital and University of Oslo, Oslo, Norway
- NORMENT, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Ole Andreas Andreassen
- NORMENT Centre, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Nils Eiel Steen
- NORMENT Centre, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
27
|
Brisch R, Wojtylak S, Saniotis A, Steiner J, Gos T, Kumaratilake J, Henneberg M, Wolf R. The role of microglia in neuropsychiatric disorders and suicide. Eur Arch Psychiatry Clin Neurosci 2022; 272:929-945. [PMID: 34595576 PMCID: PMC9388452 DOI: 10.1007/s00406-021-01334-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 09/09/2021] [Indexed: 02/08/2023]
Abstract
This narrative review examines the possible role of microglial cells, first, in neuroinflammation and, second, in schizophrenia, depression, and suicide. Recent research on the interactions between microglia, astrocytes and neurons and their involvement in pathophysiological processes of neuropsychiatric disorders is presented. This review focuses on results from postmortem, positron emission tomography (PET) imaging studies, and animal models of schizophrenia and depression. Third, the effects of antipsychotic and antidepressant drug therapy, and of electroconvulsive therapy on microglial cells are explored and the upcoming development of therapeutic drugs targeting microglia is described. Finally, there is a discussion on the role of microglia in the evolutionary progression of human lineage. This view may contribute to a new understanding of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Ralf Brisch
- Department of Forensic Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | - Szymon Wojtylak
- Department of Pathomorphology, Medical University of Gdańsk, Gdańsk, Poland
| | - Arthur Saniotis
- Department of Anthropology, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
- Department of Pharmacy, Knowledge University, Erbil, Kurdistan Region, Iraq
| | - Johann Steiner
- Department of Psychiatry and Psychotherapy, Otto-von-Guericke-University, Magdeburg, Germany
| | - Tomasz Gos
- Department of Forensic Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | - Jaliya Kumaratilake
- Biological Anthropology and Comparative Anatomy Research Unit, Medical School, The University of Adelaide, Adelaide, Australia
| | - Maciej Henneberg
- Biological Anthropology and Comparative Anatomy Research Unit, Medical School, The University of Adelaide, Adelaide, Australia
- Institute of Evolutionary Medicine, University of Zurich, Zurich, Switzerland
| | - Rainer Wolf
- Department of Nursing and Health, Hochschule Fulda, University of Applied Sciences, Fulda, Germany.
| |
Collapse
|
28
|
Koutsouleris N, Pantelis C, Velakoulis D, McGuire P, Dwyer DB, Urquijo-Castro MF, Paul R, Dong S, Popovic D, Oeztuerk O, Kambeitz J, Salokangas RKR, Hietala J, Bertolino A, Brambilla P, Upthegrove R, Wood SJ, Lencer R, Borgwardt S, Maj C, Nöthen M, Degenhardt F, Polyakova M, Mueller K, Villringer A, Danek A, Fassbender K, Fliessbach K, Jahn H, Kornhuber J, Landwehrmeyer B, Anderl-Straub S, Prudlo J, Synofzik M, Wiltfang J, Riedl L, Diehl-Schmid J, Otto M, Meisenzahl E, Falkai P, Schroeter ML. Exploring Links Between Psychosis and Frontotemporal Dementia Using Multimodal Machine Learning: Dementia Praecox Revisited. JAMA Psychiatry 2022; 79:907-919. [PMID: 35921104 PMCID: PMC9350851 DOI: 10.1001/jamapsychiatry.2022.2075] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 06/12/2022] [Indexed: 12/04/2022]
Abstract
Importance The behavioral and cognitive symptoms of severe psychotic disorders overlap with those seen in dementia. However, shared brain alterations remain disputed, and their relevance for patients in at-risk disease stages has not been explored so far. Objective To use machine learning to compare the expression of structural magnetic resonance imaging (MRI) patterns of behavioral-variant frontotemporal dementia (bvFTD), Alzheimer disease (AD), and schizophrenia; estimate predictability in patients with bvFTD and schizophrenia based on sociodemographic, clinical, and biological data; and examine prognostic value, genetic underpinnings, and progression in patients with clinical high-risk (CHR) states for psychosis or recent-onset depression (ROD). Design, Setting, and Participants This study included 1870 individuals from 5 cohorts, including (1) patients with bvFTD (n = 108), established AD (n = 44), mild cognitive impairment or early-stage AD (n = 96), schizophrenia (n = 157), or major depression (n = 102) to derive and compare diagnostic patterns and (2) patients with CHR (n = 160) or ROD (n = 161) to test patterns' prognostic relevance and progression. Healthy individuals (n = 1042) were used for age-related and cohort-related data calibration. Data were collected from January 1996 to July 2019 and analyzed between April 2020 and April 2022. Main Outcomes and Measures Case assignments based on diagnostic patterns; sociodemographic, clinical, and biological data; 2-year functional outcomes and genetic separability of patients with CHR and ROD with high vs low pattern expression; and pattern progression from baseline to follow-up MRI scans in patients with nonrecovery vs preserved recovery. Results Of 1870 included patients, 902 (48.2%) were female, and the mean (SD) age was 38.0 (19.3) years. The bvFTD pattern comprising prefrontal, insular, and limbic volume reductions was more expressed in patients with schizophrenia (65 of 157 [41.2%]) and major depression (22 of 102 [21.6%]) than the temporo-limbic AD patterns (28 of 157 [17.8%] and 3 of 102 [2.9%], respectively). bvFTD expression was predicted by high body mass index, psychomotor slowing, affective disinhibition, and paranoid ideation (R2 = 0.11). The schizophrenia pattern was expressed in 92 of 108 patients (85.5%) with bvFTD and was linked to the C9orf72 variant, oligoclonal banding in the cerebrospinal fluid, cognitive impairment, and younger age (R2 = 0.29). bvFTD and schizophrenia pattern expressions forecasted 2-year psychosocial impairments in patients with CHR and were predicted by polygenic risk scores for frontotemporal dementia, AD, and schizophrenia. Findings were not associated with AD or accelerated brain aging. Finally, 1-year bvFTD/schizophrenia pattern progression distinguished patients with nonrecovery from those with preserved recovery. Conclusions and Relevance Neurobiological links may exist between bvFTD and psychosis focusing on prefrontal and salience system alterations. Further transdiagnostic investigations are needed to identify shared pathophysiological processes underlying the neuroanatomical interface between the 2 disease spectra.
Collapse
Affiliation(s)
- Nikolaos Koutsouleris
- Department of Psychiatry and Psychotherapy, Ludwig Maximilian University, Munich, Germany
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
- Max-Planck Institute of Psychiatry, Munich, Germany
| | - Christos Pantelis
- Melbourne Neuropsychiatry Centre, University of Melbourne, Melbourne, Australia
| | - Dennis Velakoulis
- Melbourne Neuropsychiatry Centre, University of Melbourne, Melbourne, Australia
| | - Philip McGuire
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Dominic B. Dwyer
- Department of Psychiatry and Psychotherapy, Ludwig Maximilian University, Munich, Germany
| | | | - Riya Paul
- Department of Psychiatry and Psychotherapy, Ludwig Maximilian University, Munich, Germany
| | - Sen Dong
- Department of Psychiatry and Psychotherapy, Ludwig Maximilian University, Munich, Germany
| | - David Popovic
- Department of Psychiatry and Psychotherapy, Ludwig Maximilian University, Munich, Germany
| | - Oemer Oeztuerk
- Department of Psychiatry and Psychotherapy, Ludwig Maximilian University, Munich, Germany
| | - Joseph Kambeitz
- Department of Psychiatry and Psychotherapy, University of Cologne, Cologne, Germany
| | | | - Jarmo Hietala
- Department of Psychiatry, University of Turku, Turku, Finland
| | - Alessandro Bertolino
- Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari Aldo Moro, Bari, Italy
| | - Paolo Brambilla
- Department of Neurosciences and Mental Health, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Rachel Upthegrove
- Institute of Mental Health, University of Birmingham, Birmingham, United Kingdom
- Early Intervention Service, Birmingham Women’s and Children’s NHS Foundation Trust, Birmingham, United Kingdom
| | - Stephen J. Wood
- School of Psychology, University of Birmingham, Birmingham, United Kingdom
- Centre for Youth Mental Health, University of Melbourne, Melbourne, Australia
- Orygen, Melbourne, Australia
| | - Rebekka Lencer
- Department of Psychiatry and Psychotherapy, University of Lübeck, Lübeck, Germany
- Institute for Translational Psychiatry, University Muenster, Muenster, Germany
| | - Stefan Borgwardt
- Department of Psychiatry and Psychotherapy, University of Lübeck, Lübeck, Germany
- Department of Psychiatry, University Psychiatric Clinics (UPK), University of Basel, Basel, Switzerland
| | - Carlo Maj
- Institute for Genomic Statistics and Bioinformatics, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Markus Nöthen
- Institute of Human Genetics, School of Medicine, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Franziska Degenhardt
- Institute of Human Genetics, School of Medicine, University Hospital Bonn, University of Bonn, Bonn, Germany
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Essen, University of Duisburg-Essen, Duisburg, Germany
| | - Maryna Polyakova
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
- Clinic for Cognitive Neurology, University Hospital Leipzig, Leipzig, Germany
| | - Karsten Mueller
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Arno Villringer
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Adrian Danek
- Department of Neurology, Ludwig Maximilian University Munich, Munich, Germany
| | - Klaus Fassbender
- Department of Neurology, Saarland University Hospital, Homburg, Germany
| | - Klaus Fliessbach
- Department of Psychiatry and Psychotherapy, University Hospital Bonn, Bonn, Germany
- German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| | - Holger Jahn
- Department of Psychiatry and Psychotherapy, University Hospital Hamburg, Hamburg, Germany
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, Friedrich Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | | | | | - Johannes Prudlo
- Department of Neurology, University Medicine Rostock, Rostock, Germany
| | - Matthis Synofzik
- German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
- Department of Neurodegenerative Diseases, Center of Neurology, Hertie Institute for Clinical Brain Research, Tübingen, Germany
| | - Jens Wiltfang
- German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
- Department of Psychiatry and Psychotherapy, Medical University Göttingen, Göttingen, Germany
| | - Lina Riedl
- Department of Psychiatry and Psychotherapy, Technical University of Munich, Munich, Germany
| | - Janine Diehl-Schmid
- Department of Psychiatry and Psychotherapy, Technical University of Munich, Munich, Germany
| | - Markus Otto
- Department of Neurology, University of Ulm, Ulm, Germany
| | - Eva Meisenzahl
- Department of Psychiatry and Psychotherapy, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Peter Falkai
- Department of Psychiatry and Psychotherapy, Ludwig Maximilian University, Munich, Germany
| | - Matthias L. Schroeter
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
- Clinic for Cognitive Neurology, University Hospital Leipzig, Leipzig, Germany
| |
Collapse
|
29
|
Gober R, Ardalan M, Shiadeh SMJ, Duque L, Garamszegi SP, Ascona M, Barreda A, Sun X, Mallard C, Vontell RT. Microglia activation in postmortem brains with schizophrenia demonstrates distinct morphological changes between brain regions. Brain Pathol 2022; 32:e13003. [PMID: 34297453 PMCID: PMC8713533 DOI: 10.1111/bpa.13003] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 05/11/2021] [Accepted: 06/21/2021] [Indexed: 12/18/2022] Open
Abstract
Schizophrenia (SCZ) is a psychiatric disorder that can include symptoms of disorganized speech and thoughts with uncertain underlying mechanisms possibly linked to over-activated microglia. In this study, we used brain samples from sixteen donors with SCZ and thirteen control donors to assess the differential activation of microglia by quantifying density and 3D reconstruction of microglia stained with ionized calcium-binding adaptor molecule-1 (Iba1). Our samples consisted of sections from the frontal, temporal, and cingulate cortical gray matter, subcortical white matter regions (SCWM), and included the anterior corpus callosum. In the first series of studies, we performed a density analysis followed by a spatial analysis to ascertain the microglial density, distribution, and soma size in SCZ brains. Second, we performed a series of morphological quantification techniques to investigate the arborization patterns of the microglia in SCZ. The results demonstrated an increase in microglia density in the cortical gray matter regions in SCZ cases, while in the SCWM, there was a significant increase in microglia density in the frontal and temporal, but not in the other brain regions of interest (ROIs). Spatial analysis using the "nearest neighbor" demonstrated that there was no effect in "clustering", but there were shorter distances between microglia seen in the SCZ cases. The morphological measures showed that there was a region-dependent increase in the microglia soma size in the SCZ cases while the Sholl analysis revealed a significant decrease in the microglia arborization in the SCZ cases across all the ROI's studied. An in-depth 3D reconstruction of microglia in Brodmann area 9 cortical region found that there was a significant association between age and reduced microglial arborization in the SCZ cases. This region-dependent age association can help determine whether longitudinal changes in microglial activation across age are brain region-dependent, which may point to potential therapeutic targets.
Collapse
Affiliation(s)
- Ryan Gober
- Brain Endowment BankUniversity of MiamiMiamiFLUSA
| | - Maryam Ardalan
- Centre for Perinatal Medicine and HealthInstitute of Neuroscience and PhysiologySahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Department of Clinical MedicineTranslational Neuropsychiatry UnitAarhus UniversityAarhusDenmark
| | - Seyedeh Marziyeh Jabbari Shiadeh
- Centre for Perinatal Medicine and HealthInstitute of Neuroscience and PhysiologySahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Department of Clinical MedicineTranslational Neuropsychiatry UnitAarhus UniversityAarhusDenmark
| | - Linda Duque
- Brain Endowment BankUniversity of MiamiMiamiFLUSA
| | | | | | | | - Xiaoyan Sun
- Brain Endowment BankUniversity of MiamiMiamiFLUSA
| | - Carina Mallard
- Centre for Perinatal Medicine and HealthInstitute of Neuroscience and PhysiologySahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | | |
Collapse
|
30
|
The association of matrix metalloproteinase 9 (MMP9) with hippocampal volume in schizophrenia: a preliminary MRI study. Neuropsychopharmacology 2022; 47:524-530. [PMID: 33833403 PMCID: PMC8674225 DOI: 10.1038/s41386-021-00997-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/13/2021] [Accepted: 03/05/2021] [Indexed: 02/02/2023]
Abstract
Matrix metalloproteinases 9 (MMP9) are enzymes involved in regulating neuroplasticity in the hippocampus. This, combined with evidence for disrupted hippocampal structure and function in schizophrenia, has prompted our current investigation into the relationship between MMP9 and hippocampal volumes in schizophrenia. 34 healthy individuals (mean age = 32.50, male = 21, female = 13) and 30 subjects with schizophrenia (mean age = 33.07, male = 19, female = 11) underwent a blood draw and T1-weighted magnetic resonance imaging. The hippocampus was automatically segmented utilizing FreeSurfer. MMP9 plasma levels were measured with ELISA. ANCOVAs were conducted to compare MMP9 plasma levels (corrected for age and sex) and hippocampal volumes between groups (corrected for age, sex, total intracranial volume). Spearman correlations were utilized to investigate the relationship between symptoms, medication, duration of illness, number of episodes, and MMP9 plasma levels in patients. Last, we explored the correlation between MMP9 levels and hippocampal volumes in patients and healthy individuals separately. Patients displayed higher MMP9 plasma levels than healthy individuals (F(1, 60) = 21.19, p < 0.0001). MMP9 levels correlated with negative symptoms in patients (R = 0.39, p = 0.035), but not with medication, duration of illness, or the number of episodes. Further, patients had smaller left (F(1,59) = 9.12, p = 0.0040) and right (F(1,59) = 6.49, p = 0.013) hippocampal volumes. Finally, left (R = -0.39, p = 0.034) and right (R = -0.37, p = 0.046) hippocampal volumes correlated negatively with MMP9 plasma levels in patients. We observe higher MMP9 plasma levels in SCZ, associated with lower hippocampal volumes, suggesting involvement of MMP9 in the pathology of SCZ. Future studies are needed to investigate how MMP9 influences the pathology of SCZ over the lifespan, whether the observed associations are specific for schizophrenia, and if a therapeutic modulation of MMP9 promotes neuroprotective effects in SCZ.
Collapse
|
31
|
Xu H, Wei Y, Zheng L, Zhang H, Luo T, Li H, Ma J, Chen J. Relation Between Unconjugated Bilirubin and Peripheral Biomarkers of Inflammation Derived From Complete Blood Counts in Patients With Acute Stage of Schizophrenia. Front Psychiatry 2022; 13:843985. [PMID: 35463529 PMCID: PMC9022903 DOI: 10.3389/fpsyt.2022.843985] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 01/20/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Inflammation and oxidative stress are the major leading hypothetical causes of schizophrenia. Unconjugated bilirubin (UCB) is an efficient endogenous plasma antioxidant. Inflammation is closely linked to oxidative stress. The relationship between UCB and inflammatory markers should be paid close attention in schizophrenia acute stage. In this paper, combined UCB and inflammatory markers were evaluated for their capability in predicting schizophrenia in the acute stage to find an easy and effective indicator to identify acute schizophrenia. METHODS A total of 6,937 acute schizophrenia patients and 6,404 healthy controls (HCs) were enrolled. UCB and peripheral biomarkers of inflammation derived from complete blood counts (CBC) were investigated in the subjects with acute schizophrenia, and the results were compared with HCs. Simultaneously, Spearman test was employed to assess the correlation between the variables, while logistic regression was adopted to determine the combined equation and receiver operating characteristic curve was used to evaluate the combined value of UCB and peripheral biomarkers of inflammation derived from CBC to predict schizophrenia in the acute stage. RESULTS The study indicates that white blood cells, neutrophil, monocyte, mean platelet volume (MPV), red cell distribution width (RDW), neutrophil/lymphocyte ratio (NLR), and monocyte/lymphocyte ratio (MLR) have significantly increased in schizophrenia (p < 0.05 for all), while platelet, lymphocyte, and platelet/lymphocyte ratio (PLR) in schizophrenia have significantly decreased (p < 0.05 for all). UCB exhibits negative correlation with MPV significantly (r = 0.121, p < 0.001), and no correlation with neutrophil and monocyte. The correlations between UCB and other peripheral biomarkers of inflammation derived from CBC are very weak. MPV, RDW, NLR, MLR, PLR, and UCB were taken as independent variables for a logistic regression analysis. The model was as follows: Logit ( P 1 ) = - 6 . 141 + 0 . 827 MPV + 5 . 613 MLR - 0 . 005 PLR - 0 . 346 UBC . The combination demonstrates better effectiveness in predicting schizophrenia in the acute stage (AUC 0.831, 95% CI 0.825 to 0.837). CONCLUSION UCB has a protective effect on acute stage of schizophrenia, which is weak and indirect by affecting the proinflammatory processes. Our findings suggest that a combination of MLR, MPV, PLR, and UBC could be used to predict acute stage of schizophrenia.
Collapse
Affiliation(s)
- Haiting Xu
- Peking University HuiLongGuan Clinical Medical School, Beijing HuiLongGuan Hospital, Beijing, China
| | - Yanyan Wei
- Peking University HuiLongGuan Clinical Medical School, Beijing HuiLongGuan Hospital, Beijing, China
| | - Lina Zheng
- Liaocheng People's Hospital, Liaocheng, China
| | - Hua Zhang
- Dongying People's Hospital, Dongying, China
| | - Tangren Luo
- The Third Hospital of Longyan, Longyan, China
| | - Hongjuan Li
- Peking University HuiLongGuan Clinical Medical School, Beijing HuiLongGuan Hospital, Beijing, China
| | - Jinbao Ma
- Capital Medical University Beijing TongRen Hospital, Beijing, China
| | - Jingxu Chen
- Peking University HuiLongGuan Clinical Medical School, Beijing HuiLongGuan Hospital, Beijing, China
| |
Collapse
|
32
|
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
|
33
|
Lin A, Forsyth JK, Hoftman GD, Kushan-Wells L, Jalbrzikowski M, Dokuru D, Coppola G, Fiksinski A, Zinkstok J, Vorstman J, Nachun D, Bearden CE. Transcriptomic profiling of whole blood in 22q11.2 reciprocal copy number variants reveals that cell proportion highly impacts gene expression. Brain Behav Immun Health 2021; 18:100386. [PMID: 34841284 PMCID: PMC8607166 DOI: 10.1016/j.bbih.2021.100386] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 10/31/2021] [Indexed: 11/24/2022] Open
Abstract
22q11.2 reciprocal copy number variants (CNVs) offer a powerful quasi-experimental "reverse-genetics" paradigm to elucidate how gene dosage (i.e., deletions and duplications) disrupts the transcriptome to cause further downstream effects. Clinical profiles of 22q11.2 CNV carriers indicate that disrupted gene expression causes alterations in neuroanatomy, cognitive function, and psychiatric disease risk. However, interpreting transcriptomic signal in bulk tissue requires careful consideration of potential changes in cell composition. We first characterized transcriptomic dysregulation in peripheral blood from reciprocal 22q11.2 CNV carriers using differential expression analysis and weighted gene co-expression network analysis (WGCNA) to identify modules of co-expressed genes. We also assessed for group differences in cell composition and re-characterized transcriptomic differences after accounting for cell type proportions and medication usage. Finally, to explore whether CNV-related transcriptomic changes relate to downstream phenotypes associated with 22q11.2 CNVs, we tested for associations of gene expression with neuroimaging measures and behavioral traits, including IQ and psychosis or ASD diagnosis. 22q11.2 deletion carriers (22qDel) showed widespread expression changes at the individual gene as well as module eigengene level compared to 22q11.2 duplication carriers (22qDup) and controls. 22qDup showed increased expression of 5 genes within the 22q11.2 locus, and CDH6 located outside of the locus. Downregulated modules in 22qDel implicated altered immune and inflammatory processes. Celltype deconvolution analyses revealed significant differences between CNV and control groups in T-cell, mast cell, and macrophage proportions; differential expression of individual genes between groups was substantially attenuated after adjusting for cell composition. Individual gene, module eigengene, and cell proportions were not significantly associated with psychiatric or neuroanatomic traits. Our findings suggest broad immune-related dysfunction in 22qDel and highlight the importance of understanding differences in cell composition when interpreting transcriptomic changes in clinical populations. Results also suggest novel directions for future investigation to test whether 22q11.2 CNV effects on macrophages have implications for brain-related microglial function that may contribute to psychiatric phenotypes in 22q11.2 CNV carriers.
Collapse
Affiliation(s)
- Amy Lin
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, USA
- Neuroscience Interdepartmental Program, University of California at Los Angeles, Los Angeles, CA, USA
| | - Jennifer K. Forsyth
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Psychology, University of Washington, WA, USA
| | - Gil D. Hoftman
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, USA
| | - Leila Kushan-Wells
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, USA
| | | | - Deepika Dokuru
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, USA
| | - Giovanni Coppola
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, USA
| | - Ania Fiksinski
- Wilhelmina Children's Hospital & University Medical Center Utrecht, Brain Center, the Netherlands
- Maastricht University, Department of Psychiatry and Neuropsychology, Division of Mental Health, MHeNS, the Netherlands
| | - Janneke Zinkstok
- Department of Psychiatry and Brain Center, University Medical Center Utrecht, the Netherlands
| | - Jacob Vorstman
- Program in Genetics and Genome Biology, Research Institute, The Hospital for Sick Children, Toronto, Canada; Department of Psychiatry, University of Toronto, Toronto, Canada
| | - Daniel Nachun
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Carrie E. Bearden
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Psychology, University of California, Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
34
|
Chauveau F, Becker G, Boutin H. Have (R)-[ 11C]PK11195 challengers fulfilled the promise? A scoping review of clinical TSPO PET studies. Eur J Nucl Med Mol Imaging 2021; 49:201-220. [PMID: 34387719 PMCID: PMC8712292 DOI: 10.1007/s00259-021-05425-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 05/19/2021] [Indexed: 12/19/2022]
Abstract
PURPOSE The prototypical TSPO radiotracer (R)-[11C]PK11195 has been used in humans for more than thirty years to visualize neuroinflammation in several pathologies. Alternative radiotracers have been developed to improve signal-to-noise ratio and started to be tested clinically in 2008. Here we examined the scientific value of these "(R)-[11C]PK11195 challengers" in clinical research to determine if they could supersede (R)-[11C]PK11195. METHODS A systematic MEDLINE (PubMed) search was performed (up to end of year 2020) to extract publications reporting TSPO PET in patients with identified pathologies, excluding studies in healthy subjects and methodological studies. RESULTS Of the 288 publications selected, 152 used 13 challengers, and 142 used (R)-[11C]PK11195. Over the last 20 years, the number of (R)-[11C]PK11195 studies remained stable (6 ± 3 per year), but was surpassed by the total number of challenger studies for the last 6 years. In total, 3914 patients underwent a TSPO PET scan, and 47% (1851 patients) received (R)-[11C]PK11195. The 2 main challengers were [11C]PBR28 (24%-938 patients) and [18F]FEPPA (11%-429 patients). Only one-in-ten patients (11%-447) underwent 2 TSPO scans, among whom 40 (1%) were scanned with 2 different TSPO radiotracers. CONCLUSIONS Generally, challengers confirmed disease-specific initial (R)-[11C]PK11195 findings. However, while their better signal-to-noise ratio seems particularly useful in diseases with moderate and widespread neuroinflammation, most challengers present an allelic-dependent (Ala147Thr polymorphism) TSPO binding and genetic stratification is hindering their clinical implementation. As new challengers, insensitive to TSPO human polymorphism, are about to enter clinical evaluation, we propose this systematic review to be regularly updated (living review).
Collapse
Affiliation(s)
- Fabien Chauveau
- University of Lyon, Lyon Neuroscience Research Center (CRNL), CNRS UMR5292, INSERM U1028, University Lyon 1, Lyon, France.
| | - Guillaume Becker
- GIGA - CRC In Vivo Imaging, University Liege, Liege, Belgium
- University of Lyon, CarMeN Laboratory, INSERM U1060, University Lyon 1, Hospices Civils Lyon, Lyon, France
| | - Hervé Boutin
- Faculty of Biology Medicine and Health, Wolfson Molecular Imaging Centre, University of Manchester, Manchester, UK.
- Wolfson Molecular Imaging Centre, University of Manchester, Manchester, UK.
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK.
| |
Collapse
|
35
|
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
|
36
|
Stein H, Barbosa J, Compte A. Towards biologically constrained attractor models of schizophrenia. Curr Opin Neurobiol 2021; 70:171-181. [PMID: 34839146 DOI: 10.1016/j.conb.2021.10.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 10/19/2021] [Accepted: 10/27/2021] [Indexed: 12/31/2022]
Abstract
Alterations in neuromodulation or synaptic transmission in biophysical attractor network models, as proposed by the dominant dopaminergic and glutamatergic theories of schizophrenia, successfully mimic working memory (WM) deficits in people with schizophrenia (PSZ). Yet, multiple, often opposing alterations in memory circuits can lead to the same behavioral patterns in these network models. Here, we critically revise the computational and experimental literature that links NMDAR hypofunction to WM precision loss in PSZ. We show in network simulations that currently available experimental evidence cannot set apart competing biophysical accounts. Critical points to resolve are the effects of increases vs. decreases in E/I ratio (e.g. through NMDAR blockade) on firing rate tuning and shared noise modulations and possible concomitant deficits in short-term plasticity. We argue that these concerted experimental and computational efforts will lead to a better understanding of the neurobiology underlying cognitive deficits in PSZ.
Collapse
Affiliation(s)
- Heike Stein
- Laboratoire de Neurosciences Cognitives et Computationnelles, Département d'Études Cognitives, École Normale Supérieure, INSERM U960, PSL University, Paris, France
| | - Joao Barbosa
- Laboratoire de Neurosciences Cognitives et Computationnelles, Département d'Études Cognitives, École Normale Supérieure, INSERM U960, PSL University, Paris, France
| | - Albert Compte
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
| |
Collapse
|
37
|
Skorobogatov K, De Picker L, Verkerk R, Coppens V, Leboyer M, Müller N, Morrens M. Brain Versus Blood: A Systematic Review on the Concordance Between Peripheral and Central Kynurenine Pathway Measures in Psychiatric Disorders. Front Immunol 2021; 12:716980. [PMID: 34630391 PMCID: PMC8495160 DOI: 10.3389/fimmu.2021.716980] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 09/08/2021] [Indexed: 12/19/2022] Open
Abstract
Objective Disturbances in the kynurenine pathway have been implicated in the pathophysiology of psychotic and mood disorders, as well as several other psychiatric illnesses. It remains uncertain however to what extent metabolite levels detectable in plasma or serum reflect brain kynurenine metabolism and other disease-specific pathophysiological changes. The primary objective of this systematic review was to investigate the concordance between peripheral and central (CSF or brain tissue) kynurenine metabolites. As secondary aims we describe their correlation with illness course, treatment response, and neuroanatomical abnormalities in psychiatric diseases. Methods We performed a systematic literature search until February 2021 in PubMed. We included 27 original research articles describing a correlation between peripheral and central kynurenine metabolite measures in preclinical studies and human samples from patients suffering from neuropsychiatric disorders and other conditions. We also included 32 articles reporting associations between peripheral KP markers and symptom severity, CNS pathology or treatment response in schizophrenia, bipolar disorder or major depressive disorder. Results For kynurenine and 3-hydroxykynurenine, moderate to strong concordance was found between peripheral and central concentrations not only in psychiatric disorders, but also in other (patho)physiological conditions. Despite discordant findings for other metabolites (mainly tryptophan and kynurenic acid), blood metabolite levels were associated with clinical symptoms and treatment response in psychiatric patients, as well as with observed neuroanatomical abnormalities and glial activity. Conclusion Only kynurenine and 3-hydroxykynurenine demonstrated a consistent and reliable concordance between peripheral and central measures. Evidence from psychiatric studies on kynurenine pathway concordance is scarce, and more research is needed to determine the validity of peripheral kynurenine metabolite assessment as proxy markers for CNS processes. Peripheral kynurenine and 3-hydroxykynurenine may nonetheless represent valuable predictive and prognostic biomarker candidates for psychiatric disorders.
Collapse
Affiliation(s)
- Katrien Skorobogatov
- Faculty 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
| | - Livia De Picker
- Faculty 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
| | - Robert Verkerk
- Laboratory of Medical Biochemistry, University of Antwerp, Antwerp, Belgium
| | - Violette Coppens
- Faculty 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
| | - Marion Leboyer
- INSERM U955, Equipe Psychiatrie Translationnelle, Créteil, France.,Fondation FondaMental - Hôpital Albert Chenevier - Pôle Psychiatrie, Créteil, France.,AP-HP, Hôpitaux Universitaires Henri Mondor, DHU Pepsy, Pôle de Psychiatrie et d'Addictologie, Créteil, France.,Université Paris Est Créteil, Faculté de Médecine, Creteil, France
| | - Norbert Müller
- Department of Psychiatry and Psychotherapy, Ludwig-Maximilians-University, München, Germany
| | - Manuel Morrens
- Faculty 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
|
38
|
Rigney G, Ayubcha C, Werner TJ, Alavi A, Revheim ME. The utility of PET imaging in the diagnosis and management of psychosis: a brief review. Clin Transl Imaging 2021. [DOI: 10.1007/s40336-021-00466-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Abstract
Purpose
Advances in the pathophysiological characterization of psychosis has led to a newfound role of biomarkers in diagnostic and prognostic contexts. Further, advances in the accuracy and sensitivity of nuclear medicine imaging techniques, and specifically positron emission tomography (PET), have improved the ability to diagnose and manage individuals experiencing first-episode psychosis or those at greater risk for developing psychosis.
Methods
Literature searches were performed in PubMed, Google Scholar, and Web of Science to identify papers related to the use of PET imaging in the diagnosis or management of psychosis. Search terms used included “positron emission tomography”, “PET imaging”, “psychosis”, “disorders of psychosis”, “schizophrenia”, “biomarkers”, “diagnostic biomarkers”, “prognostic biomarker”, “monitoring biomarker”, “outcome biomarker”, and “predictive biomarker.”
Results
Studies included fell into three categories: those examining microglia, those studying dopamine synthesis capacity, and those examining acetylcholine receptor activity. Microglial imaging has been shown to be ineffective in all patients with psychosis, but some believe it shows promise in a subset of patients with psychosis, although no defining characteristics of said subset have been postulated. Studies of dopamine synthesis capacity suggest that presynaptic dopamine is reliably elevated in patients with psychosis, but levels of dopamine active transporter are not. Further, positron emission tomography (PET) with [18F]fluoro-l-dihydroxyphenylalanine ([18F]FDOPA)-PET has been recently used successfully as a predictive biomarker of dopaminergic treatment response, although more work is needed to validate such findings. Finally, existing studies have also documented lower levels of binding to the α7 nicotinic cholinergic receptor (α7-nAChR) via [18F]-ASEM PET in patients with psychosis, however there is a dearth of prospective, randomized studies evaluating the efficacy of [18F]-ASEM as a diagnostic or monitoring biomarker of any kind.
Conclusion
Molecular imaging has become a useful tool in the diagnosis and management of psychosis. Further work must be done to improve the comparative prognostic value and diagnostic accuracy of different radiotracers.
Collapse
|
39
|
De Picker L. The future of immunopsychiatry: Three milestones to clinical innovation. Brain Behav Immun Health 2021; 16:100314. [PMID: 34589805 PMCID: PMC8474175 DOI: 10.1016/j.bbih.2021.100314] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 07/28/2021] [Accepted: 07/28/2021] [Indexed: 12/30/2022] Open
Abstract
Psychoneuroimmunology, the area of research dedicated to understanding the fundamental interactions between the central nervous system and the immune system, has given rise to the development of Immunopsychiatry, a new discipline which harnesses the immune system to produce beneficial outcomes for mental health problems. Immunopsychiatry has the potential to become a clinically relevant specialty area in psychiatric practice, but has not yet been adopted by the wider mental health community. This paper aims to map out the future trajectory of Immunopsychiatry on its road towards science-to-policy knowledge translation and clinical implementation. Three critical milestones which will need to be reached in order for Immunopsychiatry to fulfil its promise for clinical innovation are discussed: a clear definition of patients who fall within the immunopsychiatric continuum; demonstration of well-defined clinical benefit and incorporation in clinical guidelines; and convergence with other paradigms in biological psychiatry.
Collapse
Affiliation(s)
- L.J. De Picker
- University Psychiatric Hospital Campus Duffel, Duffel, Belgium
- Collaborative Antwerp Psychiatric Research Institute, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
40
|
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
|
41
|
The Influence of Virus Infection on Microglia and Accelerated Brain Aging. Cells 2021; 10:cells10071836. [PMID: 34360004 PMCID: PMC8303900 DOI: 10.3390/cells10071836] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 07/15/2021] [Accepted: 07/16/2021] [Indexed: 12/12/2022] Open
Abstract
Microglia are the resident immune cells of the central nervous system contributing substantially to health and disease. There is increasing evidence that inflammatory microglia may induce or accelerate brain aging, by interfering with physiological repair and remodeling processes. Many viral infections affect the brain and interfere with microglia functions, including human immune deficiency virus, flaviviruses, SARS-CoV-2, influenza, and human herpes viruses. Especially chronic viral infections causing low-grade neuroinflammation may contribute to brain aging. This review elucidates the potential role of various neurotropic viruses in microglia-driven neurocognitive deficiencies and possibly accelerated brain aging.
Collapse
|
42
|
Romero-Miguel D, Casquero-Veiga M, MacDowell KS, Torres-Sanchez S, Garcia-Partida JA, Lamanna-Rama N, Romero-Miranda A, Berrocoso E, Leza JC, Desco M, Soto-Montenegro ML. A Characterization of the Effects of Minocycline Treatment During Adolescence on Structural, Metabolic, and Oxidative Stress Parameters in a Maternal Immune Stimulation Model of Neurodevelopmental Brain Disorders. Int J Neuropsychopharmacol 2021; 24:734-748. [PMID: 34165516 PMCID: PMC8453277 DOI: 10.1093/ijnp/pyab036] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 06/01/2021] [Accepted: 06/18/2021] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Minocycline (MIN) is a tetracycline with antioxidant, anti-inflammatory, and neuroprotective properties. Given the likely involvement of inflammation and oxidative stress (IOS) in schizophrenia, MIN has been proposed as a potential adjuvant treatment in this pathology. We tested an early therapeutic window, during adolescence, as prevention of the schizophrenia-related deficits in the maternal immune stimulation (MIS) animal model. METHODS On gestational day 15, Poly I:C or vehicle was injected in pregnant Wistar rats. A total 93 male offspring received MIN (30 mg/kg) or saline from postnatal day (PND) 35-49. At PND70, rats were submitted to the prepulse inhibition test. FDG-PET and T2-weighted MRI brain studies were performed at adulthood. IOS markers were evaluated in frozen brain tissue. RESULTS MIN treatment did not prevent prepulse inhibition test behavioral deficits in MIS offspring. However, MIN prevented morphometric abnormalities in the third ventricle but not in the hippocampus. Additionally, MIN reduced brain metabolism in cerebellum and increased it in nucleus accumbens. Finally, MIN reduced the expression of iNOS (prefrontal cortex, caudate-putamen) and increased the levels of KEAP1 (prefrontal cortex), HO1 and NQO1 (amygdala, hippocampus), and HO1 (caudate-putamen). CONCLUSIONS MIN treatment during adolescence partially counteracts volumetric abnormalities and IOS deficits in the MIS model, likely via iNOS and Nrf2-ARE pathways, also increasing the expression of cytoprotective enzymes. However, MIN treatment during this peripubertal stage does not prevent sensorimotor gating deficits. Therefore, even though it does not prevent all the MIS-derived abnormalities evaluated, our results suggest the potential utility of early treatment with MIN in other schizophrenia domains.
Collapse
Affiliation(s)
| | - Marta Casquero-Veiga
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain,CIBER de Salud Mental (CIBERSAM), Madrid, Spain
| | - Karina S MacDowell
- CIBER de Salud Mental (CIBERSAM), Madrid, Spain,Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense (UCM), IIS Imas12, IUIN, Madrid, Spain
| | - Sonia Torres-Sanchez
- CIBER de Salud Mental (CIBERSAM), Madrid, Spain,Neuropsychopharmacology and Psychobiology Research Group, Psychobiology Area, Department of Psychology, Universidad de Cádiz, Puerto Real (Cádiz), Spain,Instituto de Investigación e Innovación en Ciencias Biomédicas de Cádiz, INiBICA, Hospital Universitario Puerta del Mar, Cádiz, Spain
| | - José Antonio Garcia-Partida
- CIBER de Salud Mental (CIBERSAM), Madrid, Spain,Neuropsychopharmacology and Psychobiology Research Group, Psychobiology Area, Department of Psychology, Universidad de Cádiz, Puerto Real (Cádiz), Spain,Instituto de Investigación e Innovación en Ciencias Biomédicas de Cádiz, INiBICA, Hospital Universitario Puerta del Mar, Cádiz, Spain
| | | | | | - Esther Berrocoso
- CIBER de Salud Mental (CIBERSAM), Madrid, Spain,Neuropsychopharmacology and Psychobiology Research Group, Psychobiology Area, Department of Psychology, Universidad de Cádiz, Puerto Real (Cádiz), Spain,Instituto de Investigación e Innovación en Ciencias Biomédicas de Cádiz, INiBICA, Hospital Universitario Puerta del Mar, Cádiz, Spain
| | - Juan C Leza
- CIBER de Salud Mental (CIBERSAM), Madrid, Spain,Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense (UCM), IIS Imas12, IUIN, Madrid, Spain
| | - Manuel Desco
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain,CIBER de Salud Mental (CIBERSAM), Madrid, Spain,Departamento de Bioingeniería e Ingeniería Aeroespacial, Universidad Carlos III de Madrid, Leganés, Spain,Centro Nacional de Investigaciones Cardiovasculares, CNIC, Madrid, Spain,Correspondence: Manuel Desco, PhD, Laboratorio de Imagen Médica, Unidad de Medicina y Cirugía Experimental, Hospital General Universitario Gregorio Marañón, Dr. Esquerdo, 46. E-28007 Madrid, Spain ()
| | - María Luisa Soto-Montenegro
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain,CIBER de Salud Mental (CIBERSAM), Madrid, Spain,High Performance Research Group in Physiopathology and Pharmacology of the Digestive System (NeuGut), University Rey Juan Carlos (URJC), Alcorcón, Spain
| |
Collapse
|
43
|
Hebbrecht K, Skorobogatov K, Giltay EJ, Coppens V, De Picker L, Morrens M. Tryptophan Catabolites in Bipolar Disorder: A Meta-Analysis. Front Immunol 2021; 12:667179. [PMID: 34093561 PMCID: PMC8170319 DOI: 10.3389/fimmu.2021.667179] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 04/13/2021] [Indexed: 02/02/2023] Open
Abstract
Objective Tryptophan catabolites (TRYCATs) are implicated in the pathophysiology of mood disorders by mediating immune-inflammation and neurodegenerative processes. We performed a meta-analysis of TRYCAT levels in bipolar disorder (BD) patients compared to healthy controls. Methods A systematic literature search in seven electronic databases (PubMed, Embase, Web of Science, Cochrane, Emcare, PsycINFO, Academic Search Premier) was conducted on TRYCAT levels in cerebrospinal fluid or peripheral blood according to the PRISMA statement. A minimum of three studies per TRYCAT was required for inclusion. Standardized mean differences (SMD) were computed using random effect models. Subgroup analyses were performed for BD patients in a different mood state (depressed, manic). The methodological quality of the studies was rated using the modified Newcastle-Ottawa Quality assessment Scale. Results Twenty-one eligible studies were identified. Peripheral levels of tryptophan (SMD = -0.44; p < 0.001), kynurenine (SMD = - 0.3; p = 0.001) and kynurenic acid (SMD = -.45; p = < 0.001) were lower in BD patients versus healthy controls. In the only three eligible studies investigating TRP in cerebrospinal fluid, tryptophan was not significantly different between BD and healthy controls. The methodological quality of the studies was moderate. Subgroup analyses revealed no significant difference in TRP and KYN values between manic and depressed BD patients, but these results were based on a limited number of studies. Conclusion The TRYCAT pathway appears to be downregulated in BD patients. There is a need for more and high-quality studies of peripheral and central TRYCAT levels, preferably using longitudinal designs.
Collapse
Affiliation(s)
- Kaat Hebbrecht
- Faculty 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
| | - Katrien Skorobogatov
- Faculty 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
| | - Erik J. Giltay
- Faculty of Medicine and Health Sciences, Collaborative Antwerp Psychiatric Research Institute (CAPRI), University of Antwerp, Antwerp, Belgium
- Department of Psychiatry, Leiden University Medical Center, Leiden, Netherlands
| | - Violette Coppens
- Faculty 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
| | - Livia De Picker
- Faculty 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
| | - Manuel Morrens
- Faculty 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
|
44
|
The Potential Use of Peripheral Blood Mononuclear Cells as Biomarkers for Treatment Response and Outcome Prediction in Psychiatry: A Systematic Review. Mol Diagn Ther 2021; 25:283-299. [PMID: 33978935 DOI: 10.1007/s40291-021-00516-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/10/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND Psychiatric disorders have a major impact on the global burden of disease while therapeutic interventions remain insufficient to adequately treat a large number of patients. Regrettably, the efficacy of several psychopharmacological treatment regimens becomes apparent only after 4-6 weeks, and at this point, a significant number of patients present as non-responsive. As such, many patients go weeks/months without appropriate treatment or symptom management. Adequate biomarkers for treatment success and outcome prediction are thus urgently needed. OBJECTIVE With this systematic review, we provide an overview of the use of peripheral blood mononuclear cells (PBMCs) and their signaling pathways in evaluating and/or predicting the effectiveness of different treatment regimens in the course of psychiatric illnesses. We highlight PBMC characteristics that (i) reflect treatment presence, (ii) allow differentiation of responders from non-responders, and (iii) prove predictive at baseline with regard to treatment outcome for a broad range of psychiatric intervention strategies. REVIEW METHODS A PubMed database search was performed to extract papers investigating the relation between any type of PBMC characteristic and treatment presence and/or outcome in patients suffering from severe mental illness. Criteria for eligibility were: written in English; psychiatric diagnosis based on DSM-III-R or newer; PBMC isolation via gradient centrifugation; comparison between treated and untreated patients via PBMC features; sample size ≥ n = 5 per experimental group. Papers not researching in vivo treatment effects between patients and healthy controls, non-clinical trials, and non-hypothesis-/data-driven (e.g., -omics designs) approaches were excluded. DATA SYNTHESIS Twenty-nine original articles were included and qualitatively summarized. Antidepressant and antipsychotic treatments were mostly reflected by intracellular inflammatory markers while intervention with mood stabilizers was evidenced through cell maturation pathways. Lastly, cell viability parameters mirrored predominantly non-pharmacological therapeutic strategies. As for response prediction, PBMC (subtype) counts and telomerase activity seemed most promising for antidepressant treatment outcome determination; full length brain-derived neurotrophic factor (BDNF)/truncated BDNF were shown to be most apt to prognosticate antipsychotic treatment. CONCLUSIONS We conclude that, although inherent limitations to and heterogeneity in study designs in combination with the scarce number of original studies hamper unambiguous identification, several PBMC characteristics-mostly related to inflammatory pathways and cell viability-indeed show promise towards establishment as clinically relevant treatment biomarkers.
Collapse
|
45
|
Mekori-Domachevsky E, Taler M, Weinberger R, Guri Y, Dar S, Shani S, Dekel I, Weizman A, Gothelf D. Neutrophils to lymphocytes ratio and psychosis in 22q11.2 deletion syndrome - Clinical and scientific implications. Schizophr Res 2021; 231:164-169. [PMID: 33866261 DOI: 10.1016/j.schres.2021.03.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 03/13/2021] [Accepted: 03/30/2021] [Indexed: 11/25/2022]
Abstract
BACKGROUND Individuals with 22q11.2 deletion syndrome (22q11.2DS) are at risk for having both psychotic and immune disorders, thus, implying a possible link between the two. The aim of the current study was to evaluate the usefulness of the neutrophiles to leukocytes ratio (NLR), an inflammatory marker, as a bio-marker for overt and prodromal psychotic symptoms in 22q11.2DS. METHODS Forty-nine individuals with 22q11.2DS (13 with psychotic disorders and 36 without psychotic disorders) and 30 age- and sex-matched healthy controls underwent psychiatric evaluation using a structured psychiatric interview, the Scale of Prodromal Symptoms (SOPS) and the Global Assessment of Functioning (GAF) scale. Blood samples were collected from all participants on the day of assessment. NLR was calculated, compared among the study groups and correlated with SOPS and GAF scores. The non-psychotic 22q11.2DS group was further divided into high- and low-inflammation groups by NLR values and the analyses were done again. RESULTS NLR was higher in the psychotic- and the high-inflammation non-psychotic 22q11.2DS groups compared to the low-inflammation non-psychotic 22q11.2DS group and controls. In the high-inflammation non-psychotic 22q11.2DS group NLR increase was associated with an increase of total negative symptoms scores on SOPS and a decrease in GAF scores. CONCLUSION Our results suggest the potential utility of NLR as a bio-marker for psychotic disorders and subthreshold prodromal symptoms in 22q11.2DS. Furthermore, they imply that a disequilibrium between the innate and adaptive arms of the immune system facilitates the progression of psychosis in at risk populations. Further longitudinal studies are warranted to validate our findings, as this was a cross sectional observation.
Collapse
Affiliation(s)
- Ehud Mekori-Domachevsky
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; The Child and Adolescent Psychiatry Division, Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel.
| | - Michal Taler
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Laboratory of Biological Psychiatry, Felsenstein Medical Research Center, Petah Tikva, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | | | - Yael Guri
- Eitanim Mental Health Center, Jerusalem, Israel
| | - Shira Dar
- Laboratory of Biological Psychiatry, Felsenstein Medical Research Center, Petah Tikva, Israel
| | - Shachar Shani
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Idit Dekel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; The Child and Adolescent Psychiatry Division, Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel
| | - Abraham Weizman
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel; Geha Mental Health Center, Petach Tikva, Israel
| | - Doron Gothelf
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; The Child and Adolescent Psychiatry Division, Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
46
|
Multimodal assessment of white matter microstructure in antipsychotic-naïve schizophrenia patients and confounding effects of recreational drug use. Brain Imaging Behav 2021; 15:36-48. [PMID: 31909444 DOI: 10.1007/s11682-019-00230-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Cerebral white matter (WM) aberrations in schizophrenia have been linked to multiple neurobiological substrates but the underlying mechanisms remain unknown. Moreover, antipsychotic treatment and substance use constitute potential confounders. Multimodal studies using diffusion tensor imaging (DTI) and magnetization transfer imaging (MTI) may provide deeper insight into the whole brain WM pathophysiology in schizophrenia. We combined DTI and MTI to investigate WM integrity in 51 antipsychotic-naïve, first-episode schizophrenia patients and 55 matched healthy controls, using 3 T magnetic resonance imaging (MRI). Psychopathology was assessed with the positive and negative syndrome scale (PANSS). A whole brain partial least squares correlation (PLSC) method was used to conjointly analyze DTI-derived measures (fractional anisotropy (FA), axial diffusivity (AD), radial diffusivity (RD), mode of anisotropy (MO)) and the magnetization transfer ratio (MTR) to identify group differences, and associations with psychopathology. In secondary analyses, we excluded recreational substance users from both groups resulting in 34 patients and 51 healthy controls. The primary PLSC group difference analysis identified a significant pattern of lower FA, AD, MO and higher RD in patients (p = 0.04). This pattern suggests disorganized WM microstructure in patients. The secondary PLSC group difference analysis without recreational substance users revealed a significant pattern of lower FA and higher AD, RD, MO, MTR in patients (p = 0.04). This pattern in the substance free patients is consistent with higher extracellular free-water concentrations, which may reflect neuroinflammation. No significant associations with psychopathology were observed. Recreational substance use appears to be a confounding issue, which calls for attention in future WM studies.
Collapse
|
47
|
Racki V, Marcelic M, Stimac I, Petric D, Kucic N. Effects of Haloperidol, Risperidone, and Aripiprazole on the Immunometabolic Properties of BV-2 Microglial Cells. Int J Mol Sci 2021; 22:4399. [PMID: 33922377 PMCID: PMC8122792 DOI: 10.3390/ijms22094399] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/18/2021] [Accepted: 04/19/2021] [Indexed: 02/07/2023] Open
Abstract
Microglial cells are resident macrophages in the brain that have been implicated in the pathophysiology of schizophrenia. There is a lack of studies covering the effects of antipsychotics on microglial cells. The current literature points to a possible anti-inflammatory action without clear mechanisms of action. The aim of this study is to characterize the effects of haloperidol, risperidone and aripiprazole on BV-2 microglial cells in in vitro conditions. We have used immunofluorescence and flow cytometry to analyze the classical pro and anti-inflammatory markers, while a real-time metabolic assay (Seahorse) was used to assess metabolic function. We analyzed the expression of p70S6K to evaluate the mTOR pathway activity with Western blot. In this study, we demonstrate the varying effects of haloperidol, risperidone and aripiprazole administration in BV-2 microglial cells. All three tested antipsychotics were successful in reducing the pro-inflammatory action of microglial cells, although only aripiprazole increased the expression of anti-inflammatory markers. Most significant differences in the possible mechanisms of action were seen in the real-time metabolic assays and in the mTORC1 signaling pathway activity, with aripiprazole being the only antipsychotic to reduce the mTORC1 activity. Our results shed some new light on the effects of haloperidol, risperidone and aripiprazole action in microglial cells, and reveal a novel possible mechanism of action for aripiprazole.
Collapse
Affiliation(s)
- Valentino Racki
- Department of Neurology, Medical Faculty, University of Rijeka, 51000 Rijeka, Croatia
| | - Marina Marcelic
- Department of Physiology and Immunology, Medical Faculty, University of Rijeka, 51000 Rijeka, Croatia; (M.M.); (I.S.); (N.K.)
| | - Igor Stimac
- Department of Physiology and Immunology, Medical Faculty, University of Rijeka, 51000 Rijeka, Croatia; (M.M.); (I.S.); (N.K.)
| | - Daniela Petric
- Department of Psychiatry, Medical Faculty, University of Rijeka, 51000 Rijeka, Croatia;
| | - Natalia Kucic
- Department of Physiology and Immunology, Medical Faculty, University of Rijeka, 51000 Rijeka, Croatia; (M.M.); (I.S.); (N.K.)
| |
Collapse
|
48
|
Morrens M, Coppens V, Walther S. Do Immune Dysregulations and Oxidative Damage Drive Mood and Psychotic Disorders? Neuropsychobiology 2021; 79:251-254. [PMID: 30991415 DOI: 10.1159/000496622] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 01/07/2019] [Indexed: 12/18/2022]
Affiliation(s)
- Manuel Morrens
- Collaborative Antwerp Psychiatric Research Institute (CAPRI), Faculty of Medicine and Health Sciences, Campus Drie Eiken, University of Antwerp, Antwerp, Belgium, .,University Department of Psychiatry, Campus Duffel, Duffel, Belgium,
| | - Violette Coppens
- Collaborative Antwerp Psychiatric Research Institute (CAPRI), Faculty of Medicine and Health Sciences, Campus Drie Eiken, University of Antwerp, Antwerp, Belgium.,University Department of Psychiatry, Campus Duffel, Duffel, Belgium
| | - Sebastian Walther
- Translational Research Center, University Hospital of Psychiatry, University of Bern, Bern, Switzerland
| |
Collapse
|
49
|
Coppens V, De Wachter O, Goossens J, Hendrix J, Maudsley S, Azmi A, van Gastel J, Van Saet A, Lauwers T, Morrens M. Profiling of the Peripheral Blood Mononuclear Cell Proteome in Schizophrenia and Mood Disorders for the Discovery of Discriminatory Biomarkers: A Proof-of-Concept Study. Neuropsychobiology 2021; 79:324-334. [PMID: 32392557 DOI: 10.1159/000507631] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 03/29/2020] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Current diagnoses in psychiatry are solely based on the evaluation of clinical presentation by the treating psychiatrist. This results in a high percentage of misdiagnosis and consequential inefficient treatment; especially regarding major depressive disorder (MDD), depression in the context of bipolar disorder (BD-D), bipolar disorder with manic symptoms (BD-M), and psychosis in the context of schizophrenia (SZ). Objective biomarkers allowing for accurate discriminatory diagnostics are therefore urgently needed. METHODS Peripheral blood mononuclear cell (PBMC) proteomes of patients with MDD (n = 5) , BD-D (n = 3), BD-M (n = 4), and SZ (n = 4), and also of healthy controls (HC; n = 6) were analyzed by state-of-the-art mass spectrometry. Proteins with a differential expression of a >2 standard deviation (SD) expression fold change from that of the HC and between either MDD versus BD-D or BD-M versus SZ were subsequently identified as potential discriminatory biomarkers. RESULTS In total, 4,271 individual proteins were retrieved from the HC. Of these, about 2,800 were detected in all patient and HC samples. For objective discrimination between MDD and BD-D, 66 candidate biomarkers were found. In parallel, 72 proteins might harbor a biomarker capacity for differential diagnostics of BD-M and SZ. A single biomarker was contraregulated versus HC in each pair of comparisons. DISCUSSION With this work, we provide a register of candidate biomarkers with the potential to objectively discriminate MDD from BD-D, and BD-M from SZ. Although concerning a proof-of-concept study with limited sample size, these data provide a stepping-stone for follow-up research on the validation of the true discriminatory potential and feasibility of clinical implementation of the discovered biomarker candidates.
Collapse
Affiliation(s)
- Violette Coppens
- Scientific Initiative for Neuropsychiatric and Psychopharmacological Studies (SINAPS), Collaborative Antwerp Psychiatric Research Institute (CAPRI), University of Antwerp, Antwerp, Belgium, .,Scientific Initiative for Neuropsychiatric and Psychopharmacological Studies (SINAPS), University Psychiatric Center Duffel, Duffel, Belgium,
| | - Oskar De Wachter
- Scientific Initiative for Neuropsychiatric and Psychopharmacological Studies (SINAPS), Collaborative Antwerp Psychiatric Research Institute (CAPRI), University of Antwerp, Antwerp, Belgium.,Scientific Initiative for Neuropsychiatric and Psychopharmacological Studies (SINAPS), University Psychiatric Center Duffel, Duffel, Belgium
| | - Jobbe Goossens
- Scientific Initiative for Neuropsychiatric and Psychopharmacological Studies (SINAPS), Collaborative Antwerp Psychiatric Research Institute (CAPRI), University of Antwerp, Antwerp, Belgium.,Scientific Initiative for Neuropsychiatric and Psychopharmacological Studies (SINAPS), University Psychiatric Center Duffel, Duffel, Belgium
| | - Jolien Hendrix
- Scientific Initiative for Neuropsychiatric and Psychopharmacological Studies (SINAPS), Collaborative Antwerp Psychiatric Research Institute (CAPRI), University of Antwerp, Antwerp, Belgium
| | - Stuart Maudsley
- Receptor Biology Lab, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Abdelkrim Azmi
- Center for Molecular Neurology, VIB, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Jaana van Gastel
- Receptor Biology Lab, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Alysia Van Saet
- Scientific Initiative for Neuropsychiatric and Psychopharmacological Studies (SINAPS), Collaborative Antwerp Psychiatric Research Institute (CAPRI), University of Antwerp, Antwerp, Belgium.,Scientific Initiative for Neuropsychiatric and Psychopharmacological Studies (SINAPS), University Psychiatric Center Duffel, Duffel, Belgium
| | - Tina Lauwers
- Scientific Initiative for Neuropsychiatric and Psychopharmacological Studies (SINAPS), Collaborative Antwerp Psychiatric Research Institute (CAPRI), University of Antwerp, Antwerp, Belgium.,Scientific Initiative for Neuropsychiatric and Psychopharmacological Studies (SINAPS), University Psychiatric Center Duffel, Duffel, Belgium
| | - Manuel Morrens
- Scientific Initiative for Neuropsychiatric and Psychopharmacological Studies (SINAPS), Collaborative Antwerp Psychiatric Research Institute (CAPRI), University of Antwerp, Antwerp, Belgium.,Scientific Initiative for Neuropsychiatric and Psychopharmacological Studies (SINAPS), University Psychiatric Center Duffel, Duffel, Belgium
| |
Collapse
|
50
|
Shelton HW, Gabbita SP, Gill WD, Burgess KC, Whicker WS, Brown RW. The effects of a novel inhibitor of tumor necrosis factor (TNF) alpha on prepulse inhibition and microglial activation in two distinct rodent models of schizophrenia. Behav Brain Res 2021; 406:113229. [PMID: 33684425 DOI: 10.1016/j.bbr.2021.113229] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 01/14/2021] [Accepted: 03/02/2021] [Indexed: 12/17/2022]
Abstract
Increased neuroinflammation has been shown in individuals diagnosed with schizophrenia (SCHZ). This study evaluated a novel immune modulator (PD2024) that targets the pro-inflammatory cytokine tumor necrosis factor-alpha (TNFα) to alleviate sensorimotor gating deficits and microglial activation employing two different rodent models of SCHZ. In Experiment 1, rats were neonatally treated with saline or the dopamine D2-like agonist quinpirole (NQ; 1 mg/kg) from postnatal day (P) 1-21 which produces increases of dopamine D2 receptor sensitivity throughout the animal's lifetime. In Experiment 2, rats were neonatally treated with saline or the immune system stimulant polyinosinic:polycytidylic acid (Poly I:C) from P5-7. Neonatal Poly I:C treatment mimics immune system activation associated with SCHZ. In both experiments, rats were raised to P30 and administered a control diet or a novel TNFα inhibitor PD2024 (10 mg/kg) in the diet from P30 until P67. At P45-46 and from P60-67, animals were behaviorally tested on auditory sensorimotor gating as measured through prepulse inhibition (PPI). NQ or Poly I:C treatment resulted in PPI deficits, and PD2024 treatment alleviated PPI deficits in both models. Results also revealed that increased hippocampal and prefrontal cortex microglial activation produced by neonatal Poly I:C was significantly reduced to control levels by PD2024. In addition, a separate group of animals neonatally treated with saline or Poly I:C from P5-7 demonstrated increased TNFα protein levels in the hippocampus but not prefrontal cortex, verifying increased TNFα in the brain produced by Poly I:C. Results from this study suggests that that brain TNFα is a viable pharmacological target to treat the neuroinflammation known to be associated with SCHZ.
Collapse
Affiliation(s)
- Heath W Shelton
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, United States
| | | | - W Drew Gill
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, United States
| | - Katherine C Burgess
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, United States
| | - Wyatt S Whicker
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, United States
| | - Russell W Brown
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, United States.
| |
Collapse
|