1
|
Santos AVS, Cardoso DS, Takada SH, Echeverry MB. Prenatal exposition to haloperidol: A preclinical narrative review. Neurosci Biobehav Rev 2023; 155:105470. [PMID: 37984569 DOI: 10.1016/j.neubiorev.2023.105470] [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: 09/04/2023] [Revised: 11/13/2023] [Accepted: 11/14/2023] [Indexed: 11/22/2023]
Abstract
Pre-existing maternal mental disorders may affect the early interactions between mother and baby, impacting the child's psychoemotional development. The typical antipsychotic haloperidol can be used during pregnancy, even with some restrictions. Its prescription is not limited to psychotic disorders, but also to other psychiatric conditions of high incidence and prevalence in the woman's fertile period. The present review focused on the preclinical available data regarding the biological and behavioral implications of embryonic exposure to haloperidol. The understanding of the effects of psychotropic drugs during neurodevelopment is important for its clinical aspect since there is limited evidence regarding the risks of antipsychotic drug treatment in pregnant women and their children. Moreover, a better comprehension of the mechanistic events that can be affected by antipsychotic treatment during the critical period of neurodevelopment may offer insights into the pathophysiology of neurodevelopmental disorders. The findings presented in this review converge to the existence of several risks associated with prenatal exposure to such medication and emphasize the need for further studies regarding its dimensions.
Collapse
Affiliation(s)
- Aline Valéria Sousa Santos
- Laboratory of Neuropharmacology and Motor Behavior, Center for Mathematics, Computation, and Cognition, Federal University of ABC, São Bernardo do Campo, SP, Brazil
| | - Débora Sterzeck Cardoso
- Neurogenetics Laboratory, Center for Mathematics, Computation, and Cognition, Federal University of ABC, São Bernardo do Campo, SP, Brazil
| | - Silvia Honda Takada
- Neurogenetics Laboratory, Center for Mathematics, Computation, and Cognition, Federal University of ABC, São Bernardo do Campo, SP, Brazil
| | - Marcela Bermúdez Echeverry
- Laboratory of Neuropharmacology and Motor Behavior, Center for Mathematics, Computation, and Cognition, Federal University of ABC, São Bernardo do Campo, SP, Brazil; Neuroscience Laboratory, School of Medicine, Universidad de Santander (UDES), Bucaramanga, Santander, Colombia.
| |
Collapse
|
2
|
Liao X, Chen M, Li Y. The glial perspective of autism spectrum disorder convergent evidence from postmortem brain and PET studies. Front Neuroendocrinol 2023; 70:101064. [PMID: 36889545 DOI: 10.1016/j.yfrne.2023.101064] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 02/12/2023] [Accepted: 03/02/2023] [Indexed: 03/08/2023]
Abstract
OBJECTIVE The present study aimed to systematically and quantitatively review evidence derived from both postmortem brain and PET studies to explore the pathological role of glia induced neuroinflammation in the pathogenesis of ASD, and discuss the implications of these findings in relation to disease pathogenesis and therapeutic strategies. METHOD An online databases search was performed to collate postmortem studies and PET studies regarding glia induced neuroinflammation in ASD as compared to controls. Two authors independently conducted the literature search, study selection and data extraction. The discrepancies generated in these processes was resolved through robust discussions among all authors. RESULT The literature search yielded the identification of 619 records, from which 22 postmortem studies and 3 PET studies were identified as eligible for the qualitative synthesis. Meta-analysis of postmortem studies reported increased microglial number and microglia density as well as increased GFAP protein expression and GFAP mRNA expression in ASD subjects as compared to controls. Three PET studies produced different outcomes and emphasized different details, with one reported increased and two reported decreased TSPO expression in ASD subjects as compared to controls. CONCLUSION Both postmortem evidences and PET studies converged to support the involvement of glia induced neuroinflammation in the pathogenesis of ASD. The limited number of included studies along with the considerable heterogeneity of these studies prevented the development of firm conclusions and challenged the explanation of variability. Future research should prioritize the replication of current studies and the validation of current observations.
Collapse
Affiliation(s)
- Xiaoli Liao
- Xiangya Nursing School, Central South University, Changsha, Hunan, China; Clinical Nursing Teaching and Research Section, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Miao Chen
- The First Affiliated Hospital, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Yamin Li
- Clinical Nursing Teaching and Research Section, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
3
|
Jankowski MM, Ignatowska-Jankowska BM, Glac W, Wiergowski M, Kazmierska-Grebowska P, Swiergiel AH. Intravenous haloperidol and cocaine alter the distribution of T CD3 + CD4 + , non-T/NK and NKT cells in rats. Clin Exp Pharmacol Physiol 2023; 50:453-462. [PMID: 36802086 DOI: 10.1111/1440-1681.13762] [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: 07/21/2022] [Revised: 02/02/2023] [Accepted: 02/14/2023] [Indexed: 02/20/2023]
Abstract
The modulation of dopamine transmission evokes strong behavioural effects that can be achieved by commonly used psychoactive drugs such as haloperidol or cocaine. Cocaine non-specifically increases dopamine transmission by blocking dopamine active transporter (DAT) and evokes behavioural arousal, whereas haloperidol is a non-specific D2-like dopamine receptor antagonist with sedative effects. Interestingly, dopamine has been found to affect immune cells in addition to its action in the central nervous system. Here, we address the possible interactions between haloperidol and cocaine and their effects on both immune cells and behaviour in freely moving rats. We use an intravenous model of haloperidol and binge cocaine administration to evaluate the drugs' impact on the distribution of lymphocyte subsets in both the peripheral blood and the spleen. We assess the drugs' behavioural effects by measuring locomotor activity. Cocaine evoked a pronounced locomotor response and stereotypic behaviours, both of which were completely blocked after pretreatment with haloperidol. The results suggest that blood lymphopenia, which was induced by haloperidol and cocaine (except for natural killer T cells), is independent of D2-like dopaminergic activity and most likely results from the massive secretion of corticosterone. Haloperidol pretreatment prevented the cocaine-induced decrease in NKT cell numbers. Moreover, the increased systemic D2-like dopaminergic activity after cocaine administration is a significant factor in retaining T CD3+ CD4+ lymphocytes and non-T/NK CD45RA+ cells in the spleen.
Collapse
Affiliation(s)
- Maciej M Jankowski
- Edmond and Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel.,Department of Animal and Human Physiology, Faculty of Biology, University of Gdansk, Gdansk, Poland
| | - Bogna M Ignatowska-Jankowska
- Department of Animal and Human Physiology, Faculty of Biology, University of Gdansk, Gdansk, Poland.,Neuronal Rhythms in Movement Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Wojciech Glac
- Department of Animal and Human Physiology, Faculty of Biology, University of Gdansk, Gdansk, Poland
| | - Marek Wiergowski
- Department of Forensic Medicine, Medical University of Gdansk, Gdansk, Poland
| | | | - Artur H Swiergiel
- Department of Animal and Human Physiology, Faculty of Biology, University of Gdansk, Gdansk, Poland
| |
Collapse
|
4
|
Real CC, Binda KH, Thomsen MB, Lillethorup TP, Brooks DJ, Landau AM. Selecting the Best Animal Model of Parkinson's Disease for Your Research Purpose: Insight from in vivo PET Imaging Studies. Curr Neuropharmacol 2023; 21:1241-1272. [PMID: 36797611 PMCID: PMC10286593 DOI: 10.2174/1570159x21666230216101659] [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: 08/17/2022] [Revised: 09/08/2022] [Accepted: 09/08/2022] [Indexed: 02/18/2023] Open
Abstract
Parkinson's disease (PD) is a debilitating neurodegenerative multisystem disorder leading to motor and non-motor symptoms in millions of individuals. Despite intense research, there is still no cure, and early disease biomarkers are lacking. Animal models of PD have been inspired by basic elements of its pathogenesis, such as dopamine dysfunction, alpha-synuclein accumulation, neuroinflammation and disruption of protein degradation, and these have been crucial for a deeper understanding of the mechanisms of pathology, the identification of biomarkers, and evaluation of novel therapies. Imaging biomarkers are non-invasive tools to assess disease progression and response to therapies; their discovery and validation have been an active field of translational research. Here, we highlight different considerations of animal models of PD that can be applied to future research, in terms of their suitability to answer different research questions. We provide the reader with important considerations of the best choice of model to use based on the disease features of each model, including issues related to different species. In addition, positron emission tomography studies conducted in PD animal models in the last 5 years are presented. With a variety of different species, interventions and genetic information, the choice of the most appropriate model to answer research questions can be daunting, especially since no single model recapitulates all aspects of this complex disorder. Appropriate animal models in conjunction with in vivo molecular imaging tools, if selected properly, can be a powerful combination for the assessment of novel therapies and developing tools for early diagnosis.
Collapse
Affiliation(s)
- Caroline Cristiano Real
- Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Aarhus, Denmark
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Karina Henrique Binda
- Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Aarhus, Denmark
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Majken Borup Thomsen
- Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Aarhus, Denmark
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Thea Pinholt Lillethorup
- Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Aarhus, Denmark
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - David James Brooks
- Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Aarhus, Denmark
- Institute of Translational and Clinical Research, University of Newcastle, Upon Tyne, UK
| | - Anne Marlene Landau
- Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Aarhus, Denmark
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
5
|
Bosch LFP, Kierdorf K. The Shape of μ—How Morphological Analyses Shape the Study of Microglia. Front Cell Neurosci 2022; 16:942462. [PMID: 35846562 PMCID: PMC9276927 DOI: 10.3389/fncel.2022.942462] [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: 05/12/2022] [Accepted: 06/13/2022] [Indexed: 11/14/2022] Open
Abstract
Microglia, the innate immune cells of the CNS parenchyma, serve as the first line of defense in a myriad of neurodevelopmental, neurodegenerative, and neuroinflammatory conditions. In response to the peripheral inflammation, circulating mediators, and other external signals that are produced by these conditions, microglia dynamically employ different transcriptional programs as well as morphological adaptations to maintain homeostasis. To understand these cells’ function, the field has established a number of essential analysis approaches, such as gene expression, cell quantification, and morphological reconstruction. Although high-throughput approaches are becoming commonplace in regard to other types of analyses (e.g., single-cell scRNA-seq), a similar standard for morphological reconstruction has yet to be established. In this review, we offer an overview of microglial morphological analysis methods, exploring the advantages and disadvantages of each, highlighting a number of key studies, and emphasizing how morphological analysis has significantly contributed to our understanding of microglial function in the CNS parenchyma. In doing so, we advocate for the use of unbiased, automated morphological reconstruction approaches in future studies, in order to capitalize on the valuable information embedded in the cellular structures microglia inhabit.
Collapse
Affiliation(s)
- Lance Fredrick Pahutan Bosch
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Katrin Kierdorf
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany
- CIBSS–Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
- *Correspondence: Katrin Kierdorf,
| |
Collapse
|
6
|
Bettegazzi B, Bellani S, Cattaneo S, Codazzi F, Grohovaz F, Zacchetti D. Gα13 Contributes to LPS-Induced Morphological Alterations and Affects Migration of Microglia. Mol Neurobiol 2021; 58:6397-6414. [PMID: 34529232 DOI: 10.1007/s12035-021-02553-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 08/26/2021] [Indexed: 11/24/2022]
Abstract
Microglia are the resident immune cells of the CNS that are activated in response to a variety of stimuli. This phenotypical change is aimed to maintain the local homeostasis, also by containing the insults and repair the damages. All these processes are tightly regulated and coordinated and a failure in restoring homeostasis by microglia can result in the development of neuroinflammation that can facilitate the progression of pathological conditions. Indeed, chronic microglia activation is commonly recognized as a hallmark of many neurological disorders, especially at an early stage. Many complex pathways, including cytoskeletal remodeling, are involved in the control of the microglial phenotypical and morphological changes that occur during activation. In this work, we focused on the small GTPase Gα13 and its role at the crossroad between RhoA and Rac1 signaling when microglia is exposed to pro-inflammatory stimulation. We propose the direct involvement of Gα13 in the cytoskeletal rearrangements mediated by FAK, LIMK/cofilin, and Rac1 during microglia activation. In fact, we show that Gα13 knockdown significantly inhibited LPS-induced microglial cell activation, in terms of both changes in morphology and migration, through the modulation of FAK and one of its downstream effectors, Rac1. In conclusion, we propose Gα13 as a critical factor in the regulation of morphological and functional properties of microglia during activation, which might become a target of intervention for the control of microglia inflammation.
Collapse
Affiliation(s)
- Barbara Bettegazzi
- IRCCS San Raffaele Scientific Institute, via Olgettina 60, 20132, Milan, Italy.
- Vita-Salute San Raffaele University, via Olgettina 58, 20132, Milan, Italy.
| | - Serena Bellani
- IRCCS San Raffaele Scientific Institute, via Olgettina 60, 20132, Milan, Italy
| | - Stefano Cattaneo
- IRCCS San Raffaele Scientific Institute, via Olgettina 60, 20132, Milan, Italy
- Vita-Salute San Raffaele University, via Olgettina 58, 20132, Milan, Italy
| | - Franca Codazzi
- IRCCS San Raffaele Scientific Institute, via Olgettina 60, 20132, Milan, Italy
- Vita-Salute San Raffaele University, via Olgettina 58, 20132, Milan, Italy
| | - Fabio Grohovaz
- IRCCS San Raffaele Scientific Institute, via Olgettina 60, 20132, Milan, Italy
- Vita-Salute San Raffaele University, via Olgettina 58, 20132, Milan, Italy
| | - Daniele Zacchetti
- IRCCS San Raffaele Scientific Institute, via Olgettina 60, 20132, Milan, Italy.
| |
Collapse
|
7
|
A high-fat diet, but not haloperidol or olanzapine administration, increases activated microglial expression in the rat brain. Neurosci Lett 2021; 757:135976. [PMID: 34023409 DOI: 10.1016/j.neulet.2021.135976] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 04/28/2021] [Accepted: 05/17/2021] [Indexed: 11/20/2022]
Abstract
This study examined the effects of chronic treatment of the antipsychotic drugs, haloperidol and olanzapine, on microglial activation in the brain. In addition, we explored the interaction of these antipsychotic drugs with normal and high-fat diet. In order to measure activated microglial expression, we used [3H] PK11195 in vitro autoradiography. Male Sprague Dawley rats were given a diet of either regular chow diet or a high-fat diet, and assigned either water, haloperidol drinking solution (1.5 mg/kg), or olanzapine drinking solution (10 mg/kg) for four weeks. Following treatment, rats were euthanized and brains extracted for [3H] PK11195 autoradiography. Rats on 4 weeks of a high-fat diet showed increased [3H] PK11195 binding compared to rats on a normal diet in the temporal association cortex (19 %), ectorhinal cortex (17 %), entorhinal cortex (18 %), and perirhinal cortex (18 %), irrespective of drug treatment. These are regions associated with memory, sensory, and visual processing. Rats treated with either haloperidol or olanzapine showed no differences in [3H] PK11195 binding compared to the control group. However, there were differences between the 2 different antipsychotic medications themselves. Haloperidol increased [3H] PK11195 binding in the amygdala (23 %), ectorhinal cortex (24 %), and perihinal cortex (29 %), compared to olanzapine. These results corroborate a known role of a high-fat diet and central inflammatory changes but suggest no role of these antipsychotic drugs in promoting neuroinflammation across 4 weeks compared to normal control rats.
Collapse
|
8
|
Snijders GJLJ, van Zuiden W, Sneeboer MAM, Berdenis van Berlekom A, van der Geest AT, Schnieder T, MacIntyre DJ, Hol EM, Kahn RS, de Witte LD. A loss of mature microglial markers without immune activation in schizophrenia. Glia 2021; 69:1251-1267. [PMID: 33410555 PMCID: PMC7986895 DOI: 10.1002/glia.23962] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 12/04/2020] [Accepted: 12/23/2020] [Indexed: 02/06/2023]
Abstract
Microglia, the immune cells of the brain, are important for neurodevelopment and have been hypothesized to play a role in the pathogenesis of schizophrenia (SCZ). Although previous postmortem studies pointed toward presence of microglial activation, this view has been challenged by more recent hypothesis-driven and hypothesis-free analyses. The aim of the present study is to further understand the observed microglial changes in SCZ. We first performed a detailed meta-analysis on studies that analyzed microglial cell density, microglial morphology, and expression of microglial-specific markers. We then further explored findings from the temporal cortex by performing immunostainings and qPCRs on an additional dataset. A random effect meta-analysis showed that the density of microglial cells was unaltered in SCZ (ES: 0.144 95% CI: 0.102 to 0.390, p = .250), and clear changes in microglial morphology were also absent. The expression of several microglial specific genes, such as CX3CR1, CSF1R, IRF8, OLR1, and TMEM119 was decreased in SCZ (ES: -0.417 95% CI: -0.417 to -0.546, p < .0001), consistent with genome-wide transcriptome meta-analysis results. These results indicate a change in microglial phenotype rather than density, which was validated with the use of TMEM119/Iba1 immunostainings on temporal cortex of a separate cohort. Changes in microglial gene expression were overlapping between SCZ and other psychiatric disorders, but largely opposite from changes reported in Alzheimer's disease. This distinct microglial phenotype provides a crucial molecular hallmark for future research into the role of microglia in SCZ and other psychiatric disorders.
Collapse
Affiliation(s)
- Gijsje J. L. J. Snijders
- Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht University, Brain Center Rudolf MagnusUniversity Medical Center Utrecht, Utrecht University (BCRM‐UMCU‐UU)UtrechtThe Netherlands
- Department of PsychiatryIcahn School of MedicineNew YorkNew YorkUSA
| | | | | | - Amber Berdenis van Berlekom
- Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht University, Brain Center Rudolf MagnusUniversity Medical Center Utrecht, Utrecht University (BCRM‐UMCU‐UU)UtrechtThe Netherlands
- Department of Translational Neuroscience (BCRM‐UMCU‐UU)UtrechtThe Netherlands
| | | | | | - Donald J. MacIntyre
- Division of Psychiatry, Centre for Clinical Brain SciencesUniversity of EdinburghEdinburghUK
| | - Elly M. Hol
- Department of Translational Neuroscience (BCRM‐UMCU‐UU)UtrechtThe Netherlands
- Neuroimmunology, Netherlands Institute for Neuroscience, An Institute of the Royal Academy of Arts and SciencesAmsterdamThe Netherlands
| | - René S. Kahn
- Department of PsychiatryIcahn School of MedicineNew YorkNew YorkUSA
- Mental Illness Research, Education and Clinical Center (MIRECC), James J Peters VA Medical CenterBronxNew YorkUSA
| | - Lot D. de Witte
- Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht University, Brain Center Rudolf MagnusUniversity Medical Center Utrecht, Utrecht University (BCRM‐UMCU‐UU)UtrechtThe Netherlands
- Department of PsychiatryIcahn School of MedicineNew YorkNew YorkUSA
- Mental Illness Research, Education and Clinical Center (MIRECC), James J Peters VA Medical CenterBronxNew YorkUSA
| |
Collapse
|
9
|
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: 11] [Impact Index Per Article: 3.7] [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
|
10
|
Ohene-Nyako M, Persons AL, Napier TC. Hippocampal blood-brain barrier of methamphetamine self-administering HIV-1 transgenic rats. Eur J Neurosci 2021; 53:416-429. [PMID: 32725911 PMCID: PMC9949894 DOI: 10.1111/ejn.14925] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 07/19/2020] [Accepted: 07/20/2020] [Indexed: 12/16/2022]
Abstract
Combined antiretroviral therapy for HIV infection reduces plasma viral load and prolongs life. However, the brain is a viral reservoir, and pathologies such as cognitive decline and blood-brain barrier (BBB) disruption persist. Methamphetamine abuse is prevalent among HIV-infected individuals. Methamphetamine and HIV toxic proteins can disrupt the BBB, but it is unclear if there exists a common pathway by which HIV proteins and methamphetamine induce BBB damage. Also unknown are the BBB effects imposed by chronic exposure to HIV proteins in the comorbid context of chronic methamphetamine abuse. To evaluate these scenarios, we trained HIV-1 transgenic (Tg) and non-Tg rats to self-administer methamphetamine using a 21-day paradigm that produced an equivalency dose range at the low end of the amounts self-titrated by humans. Markers of BBB integrity were measured for the hippocampus, a brain region involved in cognitive function. Outcomes revealed that tight junction proteins, claudin-5 and occludin, were reduced in Tg rats independent of methamphetamine, and this co-occurred with increased levels of lipopolysaccharide, albumin (indicating barrier breakdown) and matrix metalloproteinase-9 (MMP-9; indicating barrier matrix disruption); reductions in GFAP (indicating astrocytic dysfunction); and microglial activation (indicating inflammation). Evaluations of markers for two signaling pathways that regulate MMP-9 transcription, NF-κB and ERK/∆FosB revealed an overall genotype effect for NF-κB. Methamphetamine did not alter measurements from Tg rats, but in non-Tg rats, methamphetamine reduced occludin and GFAP, and increased MMP-9 and NF-κB. Study outcomes suggest that BBB dysregulation resulting from chronic exposure to HIV-1 proteins or methamphetamine both involve NF-κB/MMP-9.
Collapse
Affiliation(s)
- Michael Ohene-Nyako
- Department of Pharmacology, Rush University, Chicago, IL, USA,Department of Physician Assistant Studies, Rush University, Chicago, IL, USA
| | - Amanda L. Persons
- Department of Physician Assistant Studies, Rush University, Chicago, IL, USA,Department of Psychiatry and Behavioral Sciences, Rush University, Chicago, IL, USA,Center for Compulsive Behavior and Addiction, Rush University, Chicago, IL, USA
| | - T. Celeste Napier
- Department of Pharmacology, Rush University, Chicago, IL, USA,Department of Physician Assistant Studies, Rush University, Chicago, IL, USA,Center for Compulsive Behavior and Addiction, Rush University, Chicago, IL, USA
| |
Collapse
|
11
|
Ash NF, Massengill MT, Harmer L, Jafri A, Lewin AS. Automated segmentation and analysis of retinal microglia within ImageJ. Exp Eye Res 2020; 203:108416. [PMID: 33359513 DOI: 10.1016/j.exer.2020.108416] [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] [Received: 06/26/2020] [Revised: 11/18/2020] [Accepted: 12/16/2020] [Indexed: 01/12/2023]
Abstract
Microglia are immune cells of the central nervous system capable of distinct phenotypic changes and migration in response to injury. These changes most notably include the retraction of fine dendritic structures and adoption of a globular, phagocytic morphology. Due to their characteristic responses, microglia frequently act as histological indicators of injury progression. While algorithms seeking to automate microglia counts and morphological analysis are becoming increasingly popular, few exist that are adequate for use within the retina and manual analysis remains prevalent. To address this, we propose a novel segmentation routine, implemented within FIJI-ImageJ, to perform automated segmentation and cell counting of retinal microglia. We show that our routine could perform cell counts with accuracy similar to manual observers using the I307N Rho model. Tracking cell position relative to retinal vasculature, we observed population migration towards the photoreceptor layer beginning 12 h post light damage. Using feature selection with Chi2 and principal component analysis, we resolved cells along a morphological gradient, demonstrating that extracted features were sufficiently descriptive to capture subtle morphological changes within cell populations in I307N Rho and Balb/c TLR2-/- retinal degeneration models. Taken together, we introduce a novel automated routine capable of efficient image processing and segmentation. Using data retrieved following segmentation, we perform morphological analysis simultaneously on whole populations of cells, rather than individually. Our algorithm was built entirely with open-source software, for use on retinal microglia.
Collapse
Affiliation(s)
- Neil F Ash
- University of Florida Department of Molecular Genetics and Microbiology, Box 100266 Gainesville, FL, 32610, USA
| | - Michael T Massengill
- University of Florida Department of Molecular Genetics and Microbiology, Box 100266 Gainesville, FL, 32610, USA
| | - Lindsey Harmer
- University of Florida Department of Molecular Genetics and Microbiology, Box 100266 Gainesville, FL, 32610, USA
| | - Ahmed Jafri
- University of Florida Department of Molecular Genetics and Microbiology, Box 100266 Gainesville, FL, 32610, USA
| | - Alfred S Lewin
- University of Florida Department of Molecular Genetics and Microbiology, Box 100266 Gainesville, FL, 32610, USA.
| |
Collapse
|
12
|
Dinesh AA, Islam J, Khan J, Turkheimer F, Vernon AC. Effects of Antipsychotic Drugs: Cross Talk Between the Nervous and Innate Immune System. CNS Drugs 2020; 34:1229-1251. [PMID: 32975758 DOI: 10.1007/s40263-020-00765-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/29/2020] [Indexed: 12/11/2022]
Abstract
Converging lines of evidence suggest that activation of microglia (innate immune cells in the central nervous system [CNS]) is present in a subset of patients with schizophrenia. The extent to which antipsychotic drug treatment contributes to or combats this effect remains unclear. To address this question, we reviewed the literature for evidence that antipsychotic exposure influences brain microglia as indexed by in vivo neuroimaging and post-mortem studies in patients with schizophrenia and experimental animal models. We found no clear evidence from clinical studies for an effect of antipsychotics on either translocator protein (TSPO) radioligand binding (an in vivo neuroimaging measure of putative gliosis) or markers of brain microglia in post-mortem studies. In experimental animals, where drug and illness effects may be differentiated, we also found no clear evidence for consistent effects of antipsychotic drugs on TSPO radioligand binding. By contrast, we found evidence that chronic antipsychotic exposure may influence central microglia density and morphology. However, these effects were dependent on the dose and duration of drug exposure and whether an immune stimulus was present or not. In the latter case, antipsychotics were generally reported to suppress expression of inflammatory cytokines and inducible inflammatory enzymes such as cyclooxygenase and microglia activation. No clear conclusions could be drawn with regard to any effect of antipsychotics on brain microglia from current clinical data. There is evidence to suggest that antipsychotic drugs influence brain microglia in experimental animals, including possible anti-inflammatory actions. However, we lack detailed information on how these drugs influence brain microglia function at the molecular level. The clinical relevance of the animal data with regard to beneficial treatment effects and detrimental side effects of antipsychotic drugs also remains unknown, and further studies are warranted.
Collapse
Affiliation(s)
- Ayushi Anna Dinesh
- School of Medicine, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - Juned Islam
- School of Medicine, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - Javad Khan
- School of Medicine, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - Federico Turkheimer
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, Centre for Neuroimaging Sciences, De Crespigny Park, London, SE5 8AF, United Kingdom
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, SE1 1UL, United Kingdom
| | - Anthony C Vernon
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, SE1 1UL, United Kingdom.
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, Maurice Wohl Clinical Neuroscience Institute, 5 Cutcombe Road, London, SE5 9RT, United Kingdom.
| |
Collapse
|
13
|
Elmadany N, Logiacco F, Buonfiglioli A, Haage VC, Wright-Jin EC, Schattenberg A, Papawassiliou RM, Kettenmann H, Semtner M, Gutmann DH. Neurofibromatosis 1 - Mutant microglia exhibit sexually-dimorphic cyclic AMP-dependent purinergic defects. Neurobiol Dis 2020; 144:105030. [PMID: 32736084 DOI: 10.1016/j.nbd.2020.105030] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 07/09/2020] [Accepted: 07/23/2020] [Indexed: 12/30/2022] Open
Abstract
As critical regulators of brain homeostasis, microglia are influenced by numerous factors, including sex and genetic mutations. To study the impact of these factors on microglia biology, we employed genetically engineered mice that model Neurofibromatosis type 1 (NF1), a disorder characterized by clinically relevant sexually dimorphic differences. While microglia phagocytic activity was reduced in both male and female heterozygous Nf1 mutant (Nf1+/-) mice, purinergic control of phagocytosis was only affected in male Nf1+/- mice. ATP-induced P2Y-mediated membrane currents and P2RY12-dependent laser lesion-induced accumulation of microglial processes were also only impaired in male, but not female Nf1+/-, microglia. These defects resulted from Nf1+/- male-specific defects in cyclic AMP regulation, rather than from changes in purinergic receptor expression. Cyclic AMP elevation by phosphodiesterase blockade restored the male Nf1+/- microglia defects in P2Y-dependent membrane currents and process motility. Taken together, these data establish a sex-by-genotype interaction important to microglia function in the adult mouse brain.
Collapse
Affiliation(s)
- Nirmeen Elmadany
- Cellular Neurosciences, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany; Department of Biology, Chemistry, and Pharmacy, Freie Universität Berlin, 12169 Berlin, Germany
| | - Francesca Logiacco
- Cellular Neurosciences, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany; Department of Biology, Chemistry, and Pharmacy, Freie Universität Berlin, 12169 Berlin, Germany
| | - Alice Buonfiglioli
- Cellular Neurosciences, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany; Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany
| | - Verena C Haage
- Cellular Neurosciences, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany
| | - Elizabeth C Wright-Jin
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Alexander Schattenberg
- Cellular Neurosciences, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany
| | - Roxane M Papawassiliou
- Cellular Neurosciences, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany
| | - Helmut Kettenmann
- Cellular Neurosciences, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany.
| | - Marcus Semtner
- Cellular Neurosciences, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany.
| | - David H Gutmann
- Cellular Neurosciences, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany; Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
14
|
Hanger B, Couch A, Rajendran L, Srivastava DP, Vernon AC. Emerging Developments in Human Induced Pluripotent Stem Cell-Derived Microglia: Implications for Modelling Psychiatric Disorders With a Neurodevelopmental Origin. Front Psychiatry 2020; 11:789. [PMID: 32848951 PMCID: PMC7433763 DOI: 10.3389/fpsyt.2020.00789] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 07/23/2020] [Indexed: 12/15/2022] Open
Abstract
Microglia, the resident tissue macrophages of the brain, are increasingly implicated in the pathophysiology of psychiatric disorders with a neurodevelopmental origin, including schizophrenia. To date, however, our understanding of the potential role for these cells in schizophrenia has been informed by studies of aged post-mortem samples, low resolution in vivo neuroimaging and rodent models. Whilst these have provided important insights, including signs of the heterogeneous nature of microglia, we currently lack a validated human in vitro system to characterize microglia in the context of brain health and disease during neurodevelopment. Primarily, this reflects a lack of access to human primary tissue during developmental stages. In this review, we first describe microglia, including their ontogeny and heterogeneity and consider their role in brain development. We then provide an evaluation of the potential for differentiating microglia from human induced pluripotent stem cells (hiPSCs) as a robust in vitro human model system to study these cells. We find the majority of protocols for hiPSC-derived microglia generate cells characteristically similar to foetal stage microglia when exposed to neuronal environment-like cues. This may represent a robust and relevant model for the study of cellular and molecular mechanisms in schizophrenia. Each protocol however, provides unique benefits as well as shortcomings, highlighting the need for context-dependent protocol choice and cross-lab collaboration and communication to identify the most robust and translatable microglia model.
Collapse
Affiliation(s)
- Bjørn Hanger
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King’s College London, London, United Kingdom
| | - Amalie Couch
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King’s College London, London, United Kingdom
| | - Lawrence Rajendran
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King’s College London, London, United Kingdom
- UK Dementia Research Institute, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, United Kingdom
| | - Deepak P. Srivastava
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King’s College London, London, United Kingdom
- MRC Centre for Neurodevelopmental Disorders, King’s College London, London, United Kingdom
| | - Anthony C. Vernon
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King’s College London, London, United Kingdom
- MRC Centre for Neurodevelopmental Disorders, King’s College London, London, United Kingdom
| |
Collapse
|
15
|
Scarborough J, Mueller F, Arban R, Dorner-Ciossek C, Weber-Stadlbauer U, Rosenbrock H, Meyer U, Richetto J. Preclinical validation of the micropipette-guided drug administration (MDA) method in the maternal immune activation model of neurodevelopmental disorders. Brain Behav Immun 2020; 88:461-470. [PMID: 32278850 DOI: 10.1016/j.bbi.2020.04.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 04/06/2020] [Accepted: 04/06/2020] [Indexed: 12/31/2022] Open
Abstract
Pharmacological treatments in laboratory rodents remain a cornerstone of preclinical psychopharmacological research and drug development. There are numerous ways in which acute or chronic pharmacological treatments can be implemented, with each method having certain advantages and drawbacks. Here, we describe and validate a novel treatment method in mice, which we refer to as the micropipette-guided drug administration (MDA) procedure. This administration method is based on a sweetened condensed milk solution as a vehicle for pharmacological substances, which motivates the animals to consume vehicle and/or drug solutions voluntarily in the presence of the experimenter. In a proof-of-concept study, we show that the pharmacokinetic profiles of the atypical antipsychotic drug, risperidone, were similar whether administered via the MDA procedure or via the conventional oral gavage method. Unlike the latter, however, MDA did not induce the stress hormone, corticosterone. Furthermore, we assessed the suitability and validity of the MDA method in a mouse model of maternal immune activation, which is frequently used as a model of immune-mediated neurodevelopmental disorders. Using this model, we found that chronic treatment (>4 weeks, once per day) with risperidone via MDA led to a dose-dependent mitigation of MIA-induced social interaction deficits and amphetamine hypersensitivity. Taken together, the MDA procedure described herein represents a novel pharmacological administration method for per os treatments in mice that is easy to implement, cost effective, non-invasive, and less stressful for the animals than conventional oral gavage methods.
Collapse
Affiliation(s)
- Joseph Scarborough
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland
| | - Flavia Mueller
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland
| | - Roberto Arban
- Boehringer-Ingelheim Pharma GmbH & Co KG, Dept. of CNS Discovery Research, Biberach, Germany
| | - Cornelia Dorner-Ciossek
- Boehringer-Ingelheim Pharma GmbH & Co KG, Dept. of CNS Discovery Research, Biberach, Germany
| | - Ulrike Weber-Stadlbauer
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland
| | - Holger Rosenbrock
- Boehringer-Ingelheim Pharma GmbH & Co KG, Dept. of CNS Discovery Research, Biberach, Germany
| | - Urs Meyer
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland; Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Juliet Richetto
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland; Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
16
|
Turkheimer FE, Selvaggi P, Mehta MA, Veronese M, Zelaya F, Dazzan P, Vernon AC. Normalizing the Abnormal: Do Antipsychotic Drugs Push the Cortex Into an Unsustainable Metabolic Envelope? Schizophr Bull 2020; 46:484-495. [PMID: 31755955 PMCID: PMC7147598 DOI: 10.1093/schbul/sbz119] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The use of antipsychotic medication to manage psychosis, principally in those with a diagnosis of schizophrenia or bipolar disorder, is well established. Antipsychotics are effective in normalizing positive symptoms of psychosis in the short term (delusions, hallucinations and disordered thought). Their long-term use is, however, associated with side effects, including several types of movement (extrapyramidal syndrome, dyskinesia, akathisia), metabolic and cardiac disorders. Furthermore, higher lifetime antipsychotic dose-years may be associated with poorer cognitive performance and blunted affect, although the mechanisms driving the latter associations are not well understood. In this article, we propose a novel model of the long-term effects of antipsychotic administration focusing on the changes in brain metabolic homeostasis induced by the medication. We propose here that the brain metabolic normalization, that occurs in parallel to the normalization of psychotic symptoms following antipsychotic treatment, may not ultimately be sustainable by the cerebral tissue of some patients; these patients may be characterized by already reduced oxidative metabolic capacity and this may push the brain into an unsustainable metabolic envelope resulting in tissue remodeling. To support this perspective, we will review the existing data on the brain metabolic trajectories of patients with a diagnosis of schizophrenia as indexed using available neuroimaging tools before and after use of medication. We will also consider data from pre-clinical studies to provide mechanistic support for our model.
Collapse
Affiliation(s)
- Federico E Turkheimer
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
- MRC Centre for Neurodevelopmental Disorders, King’s College London, London, UK
| | - Pierluigi Selvaggi
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| | - Mitul A Mehta
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| | - Mattia Veronese
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| | - Fernando Zelaya
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| | - Paola Dazzan
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| | - Anthony C Vernon
- MRC Centre for Neurodevelopmental Disorders, King’s College London, London, UK
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| |
Collapse
|
17
|
Elevated serum chemokine CCL22 levels in first-episode psychosis: associations with symptoms, peripheral immune state and in vivo brain glial cell function. Transl Psychiatry 2020; 10:94. [PMID: 32179746 PMCID: PMC7075957 DOI: 10.1038/s41398-020-0776-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 02/18/2020] [Accepted: 03/03/2020] [Indexed: 02/08/2023] Open
Abstract
Several lines of research support immune system dysregulation in psychotic disorders. However, it remains unclear whether the immunological marker alterations are stable and how they associate with brain glial cell function. This longitudinal study aimed at investigating whether peripheral immune functions are altered in the early phases of psychotic disorders, whether the changes are associated with core symptoms, remission, brain glial cell function, and whether they persist in a one-year follow-up. Two independent cohorts comprising in total of 129 first-episode psychosis (FEP) patients and 130 controls were assessed at baseline and at the one-year follow-up. Serum cyto-/chemokines were measured using a 38-plex Luminex assay. The FEP patients showed a marked increase in chemokine CCL22 levels both at baseline (p < 0.0001; Cohen's d = 0.70) and at the 12-month follow-up (p = 0.0007) compared to controls. The group difference remained significant (p = 0.0019) after accounting for relevant covariates including BMI, smoking, and antipsychotic medication. Elevated serum CCL22 levels were significantly associated with hallucinations (ρ = 0.20) and disorganization (ρ = 0.23), and with worse verbal performance (ρ = -0.23). Brain glial cell activity was indexed with positron emission tomography and the translocator protein radiotracer [11C]PBR28 in subgroups of 15 healthy controls and 14 FEP patients with serum CCL22/CCL17 measurements. The distribution volume (VT) of [11C]PBR28 was lower in patients compared to controls (p = 0.026; Cohen's d = 0.94) without regionally specific effects, and was inversely associated with serum CCL22 and CCL17 levels (p = 0.036). Our results do not support the over-active microglia hypothesis of psychosis, but indicate altered CCR4 immune signaling in early psychosis with behavioral correlates possibly mediated through cross-talk between chemokine networks and dysfunctional or a decreased number of glial cells.
Collapse
|
18
|
Sneeboer MAM, Snijders GJLJ, Berdowski WM, Fernández-Andreu A, van Mierlo HC, Berdenis van Berlekom A, Litjens M, Kahn RS, Hol EM, de Witte LD. Microglia in post-mortem brain tissue of patients with bipolar disorder are not immune activated. Transl Psychiatry 2019; 9:153. [PMID: 31127084 PMCID: PMC6534632 DOI: 10.1038/s41398-019-0490-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 04/10/2019] [Accepted: 04/29/2019] [Indexed: 12/22/2022] Open
Abstract
Genetic, epidemiological, and biomarker studies suggest that the immune system is involved in the pathogenesis of bipolar disorder (BD). It has therefore been hypothesized that immune activation of microglia, the resident immune cells of the brain, is associated with the disease. Only a few studies have addressed the involvement of microglia in BD so far and a more detailed immune profiling of microglial activation is lacking. Here, we applied a multi-level approach to determine the activation state of microglia in BD post-mortem brain tissue. We did not find differences in microglial density, and mRNA expression of microglial markers in the medial frontal gyrus (MFG) of patients with BD. Furthermore, we performed in-depth characterization of human primary microglia isolated from fresh brain tissue of the MFG, superior temporal gyrus (STG), and thalamus (THA). Similarly, these ex vivo isolated microglia did not show elevated expression of inflammatory markers. Finally, challenging the isolated microglia with LPS did not result in an increased immune response in patients with BD compared to controls. In conclusion, our study shows that microglia in post-mortem brain tissue of patients with BD are not immune activated.
Collapse
Affiliation(s)
- Marjolein A M Sneeboer
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht University (BCRM-UMCU-UU), 3584 CG, Utrecht, The Netherlands.
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht University (BCRM-UMCU-UU), 3584 CG, Utrecht, The Netherlands.
| | - Gijsje J L J Snijders
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht University (BCRM-UMCU-UU), 3584 CG, Utrecht, The Netherlands
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht University (BCRM-UMCU-UU), 3584 CG, Utrecht, The Netherlands
| | - Woutje M Berdowski
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht University (BCRM-UMCU-UU), 3584 CG, Utrecht, The Netherlands
| | - Alba Fernández-Andreu
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht University (BCRM-UMCU-UU), 3584 CG, Utrecht, The Netherlands
| | - Hans C van Mierlo
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht University (BCRM-UMCU-UU), 3584 CG, Utrecht, The Netherlands
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht University (BCRM-UMCU-UU), 3584 CG, Utrecht, The Netherlands
| | - Amber Berdenis van Berlekom
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht University (BCRM-UMCU-UU), 3584 CG, Utrecht, The Netherlands
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht University (BCRM-UMCU-UU), 3584 CG, Utrecht, The Netherlands
| | - Manja Litjens
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht University (BCRM-UMCU-UU), 3584 CG, Utrecht, The Netherlands
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht University (BCRM-UMCU-UU), 3584 CG, Utrecht, The Netherlands
| | - René S Kahn
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht University (BCRM-UMCU-UU), 3584 CG, Utrecht, The Netherlands
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mental Illness Research, Education and Clinical Center (MIRECC), James J Peters VA Medical Center, Bronx, NY, USA
| | - Elly M Hol
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht University (BCRM-UMCU-UU), 3584 CG, Utrecht, The Netherlands
- Neuroimmunology, Netherlands Institute for Neuroscience, an Institute of The Royal Academy of Arts and Sciences, 1105 BA, Amsterdam, The Netherlands
| | - Lot D de Witte
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht University (BCRM-UMCU-UU), 3584 CG, Utrecht, The Netherlands
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mental Illness Research, Education and Clinical Center (MIRECC), James J Peters VA Medical Center, Bronx, NY, USA
| |
Collapse
|
19
|
Nishigaki A, Kawano T, Iwata H, Aoyama B, Yamanaka D, Tateiwa H, Shigematsu-Locatelli M, Eguchi S, Locatelli FM, Yokoyama M. Acute and long-term effects of haloperidol on surgery-induced neuroinflammation and cognitive deficits in aged rats. J Anesth 2019; 33:416-425. [PMID: 31049689 DOI: 10.1007/s00540-019-02646-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 04/23/2019] [Indexed: 12/19/2022]
Abstract
PURPOSE Neuroinflammation may contribute to the pathogenesis of the cognitive symptoms of postoperative delirium (POD) and its subsequent long-term cognitive impairment. Haloperidol (HAL), a dopamine receptor antagonist, is widely used to treat POD, whereas the effects of HAL on postoperative neuroinflammation and related cognitive deficits have been underdetermined. METHODS Aged rats underwent sham or abdominal surgery and were subcutaneously treated with either vehicle, low-dose (0.5 mg/kg bolus, then 0.5 mg/kg/day infusion), or high-dose (2.0 mg/kg bolus, then 2.0 mg/kg/day infusion) HAL. All treatments were initiated immediately after surgery and continued for 48 h. On either postoperative day 2 (early) or 7 (late), all rats were tested for trace and context fear memory retention after acquisition of trace fear conditioning. Following the cognitive testing, the levels of pro-inflammatory cytokines, as well as dopamine and its metabolite, in hippocampus and medial prefrontal cortex (mPFC) were measured. RESULTS In the early postoperative period, surgery induced acute neuroinflammation along with related trace and context memory dysfunction. Dopamine turnover was increased in both hippocampus and mPFC, whereas no relationship with memory functions was observed. However, HAL even at high-dose failed to restore the surgery-induced neuroinflammation and related cognitive deficits. In the late postoperative period, chronic neuroinflammation was detected only in hippocampus, which was associated with context, but not trace memory dysfunction. Neither low- nor high-dose HAL could prevent the development of these late-phase neurocognitive deficits. CONCLUSION Our findings indicate that perioperative administration with HAL may have no effects on postoperative neuroinflammation and related cognitive impairment.
Collapse
Affiliation(s)
- Atsushi Nishigaki
- Department of Anesthesiology and Intensive Care Medicine, Kochi Medical School, Kohasu, Oko-cho, Nankoku, Kochi, 783-8505, Japan
| | - Takashi Kawano
- Department of Anesthesiology and Intensive Care Medicine, Kochi Medical School, Kohasu, Oko-cho, Nankoku, Kochi, 783-8505, Japan.
| | - Hideki Iwata
- Department of Anesthesiology and Intensive Care Medicine, Kochi Medical School, Kohasu, Oko-cho, Nankoku, Kochi, 783-8505, Japan
| | - Bun Aoyama
- Department of Anesthesiology and Intensive Care Medicine, Kochi Medical School, Kohasu, Oko-cho, Nankoku, Kochi, 783-8505, Japan
| | - Daiki Yamanaka
- Department of Anesthesiology and Intensive Care Medicine, Kochi Medical School, Kohasu, Oko-cho, Nankoku, Kochi, 783-8505, Japan
| | - Hiroki Tateiwa
- Department of Anesthesiology and Intensive Care Medicine, Kochi Medical School, Kohasu, Oko-cho, Nankoku, Kochi, 783-8505, Japan
| | - Marie Shigematsu-Locatelli
- Department of Anesthesiology and Intensive Care Medicine, Kochi Medical School, Kohasu, Oko-cho, Nankoku, Kochi, 783-8505, Japan
| | - Satoru Eguchi
- Department of Dental Anesthesiology, Tokushima University School of Dentistry, Tokushima, Japan
| | - Fabricio M Locatelli
- Department of Anesthesiology and Intensive Care Medicine, Kochi Medical School, Kohasu, Oko-cho, Nankoku, Kochi, 783-8505, Japan
| | - Masataka Yokoyama
- Department of Anesthesiology and Intensive Care Medicine, Kochi Medical School, Kohasu, Oko-cho, Nankoku, Kochi, 783-8505, Japan
| |
Collapse
|
20
|
De Picker L, Ottoy J, Verhaeghe J, Deleye S, Wyffels L, Fransen E, Kosten L, Sabbe B, Coppens V, Timmers M, de Boer P, Van Nueten L, Op De Beeck K, Oberacher H, Vanhoenacker F, Ceyssens S, Stroobants S, Staelens S, Morrens M. State-associated changes in longitudinal [ 18F]-PBR111 TSPO PET imaging of psychosis patients: Evidence for the accelerated ageing hypothesis? Brain Behav Immun 2019; 77:46-54. [PMID: 30503836 DOI: 10.1016/j.bbi.2018.11.318] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 11/23/2018] [Accepted: 11/29/2018] [Indexed: 01/08/2023] Open
Abstract
OBJECTIVE To determine whether state-associated changes in microglial activity, measured with translocator-protein positron emission tomography (TSPO PET), can be identified in psychosis patients through longitudinal evaluation of their regional tracer uptake over the clinical course from acute psychosis to post-treatment follow-up, and comparison to healthy controls. We also evaluated the relation between tracer uptake, clinical symptoms and peripheral immunological markers. METHOD Second-generation radioligand [18F]-PBR111 TSPO PET-CT was used for longitudinal dynamic imaging in 14 male psychosis patients and 17 male age-matched healthy control subjects. Patients were first scanned during an acute psychotic episode followed by a second scan after treatment. Prior genotyping of subjects for the rs6917 polymorphism distinguished high- and mixed-affinity binders. The main outcome was regional volume of distribution (VT), representing TSPO binding. Plasma concentrations of CRP, cytokines and kynurenines were measured at each timepoint. RESULTS We found a significant three-way interaction between time of scan, age and cohort (cortical grey matter F6.50, p.020). Age-dependent differences in VT existed between cohorts during the psychotic state, but not at follow-up. Patients' relative change in VT over time correlated with age (cortical grey matter Pearson's r.574). PANSS positive subscale scores correlated with regional VT during psychosis (cortical grey matter r.767). Plasma CRP and quinolinic acid were independently associated with lower VT. CONCLUSIONS We identified a differential age-dependent pattern of TSPO binding from psychosis to follow-up in our cohort of male psychosis patients. We recommend future TSPO PET studies in psychosis patients to differentiate between clinical states and consider potential age-related effects.
Collapse
Affiliation(s)
- Livia De Picker
- Collaborative Antwerp Psychiatric Research Institute, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium; University Psychiatric Hospital Antwerp, Campus Duffel, Duffel, Belgium.
| | - Julie Ottoy
- Molecular Imaging Center Antwerp, University of Antwerp, Antwerp, Belgium
| | - Jeroen Verhaeghe
- Molecular Imaging Center Antwerp, University of Antwerp, Antwerp, Belgium
| | - Steven Deleye
- Molecular Imaging Center Antwerp, University of Antwerp, Antwerp, Belgium
| | - Leonie Wyffels
- Department of Nuclear Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Erik Fransen
- StatUa Center for Statistics, University of Antwerp, Belgium
| | - Lauren Kosten
- Molecular Imaging Center Antwerp, University of Antwerp, Antwerp, Belgium
| | - Bernard Sabbe
- Collaborative Antwerp Psychiatric Research Institute, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium; University Psychiatric Hospital Antwerp, Campus Duffel, Duffel, Belgium
| | - Violette Coppens
- Collaborative Antwerp Psychiatric Research Institute, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium; University Psychiatric Hospital Antwerp, Campus Duffel, Duffel, Belgium
| | - Maarten Timmers
- Janssen Research and Development, Janssen Pharmaceutica N.V., Beerse, Belgium; Reference Center for Biological Markers of Dementia (BIODEM), Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Peter de Boer
- Janssen Research and Development, Janssen Pharmaceutica N.V., Beerse, Belgium
| | - Luc Van Nueten
- Janssen Research and Development, Janssen Pharmaceutica N.V., Beerse, Belgium
| | - Ken Op De Beeck
- Medical Genetics Research Group, University of Antwerp, Antwerp, Belgium
| | - Herbert Oberacher
- Institute of Legal Medicine and Core Facility Metabolomics, Medical University of Innsbruck, Innsbruck, Austria
| | - Filip Vanhoenacker
- Department of Radiology, Sint-Maarten General Hospital, Mechelen, Belgium; Faculty of Medicine and Health Sciences, Universities of Antwerp and Ghent, Belgium
| | - Sarah Ceyssens
- Department of Nuclear Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Sigrid Stroobants
- Department of Nuclear Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Steven Staelens
- Molecular Imaging Center Antwerp, University of Antwerp, Antwerp, Belgium
| | - Manuel Morrens
- Collaborative Antwerp Psychiatric Research Institute, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium; University Psychiatric Hospital Antwerp, Campus Duffel, Duffel, Belgium
| |
Collapse
|