1
|
Wu R, Chou S, Li M. Continuous oral olanzapine or clozapine treatment initiated in adolescence has differential short- and long-term impacts on antipsychotic sensitivity than those initiated in adulthood. Eur J Pharmacol 2024; 972:176567. [PMID: 38582275 PMCID: PMC11128075 DOI: 10.1016/j.ejphar.2024.176567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 04/03/2024] [Accepted: 04/03/2024] [Indexed: 04/08/2024]
Abstract
One of the major discoveries in recent research on antipsychotic drugs is that antipsychotic treatment in adolescence could induce robust long-term alterations in antipsychotic sensitivity that persist into adulthood. These long-term impacts are likely influenced by various factors, including the "diseased" state of animals, sex, type of drugs, mode of drug administration, and age of treatment onset. In this study we compared the short- and long-term behavioral effects of 21-day continuous oral olanzapine (7.5 mg/kg/day) or clozapine (30.0 mg/kg/day) administration in heathy or maternal immune activated adolescent (33-53 days old) or adult (80-100 days old) rats of both sexes. We used a conditioned avoidance response model to assess the drug-induced alterations in antipsychotic sensitivity. Here, we report that while under the chronic drug treatment period, olanzapine progressively increased its suppression of avoidance responding over time, especially when treatment was initiated in adulthood. Clozapine's suppression depended on the age of drug exposure, with treatment initiated in adulthood showing a suppression while that initiated in adolescent did not. After a 17-day drug-free interval, in a drug challenge test, olanzapine treatment initiated in adolescence caused a decrease in drug sensitivity, as reflected by less avoidance suppression (a tolerance effect); whereas that initiated in adulthood appeared to cause an increase (more avoidance suppression, a sensitization effect). Clozapine treatments initiated in both adolescence and adulthood caused a similar tolerance effect. Our findings indicate that the same chronic antipsychotic treatment regimen initiated in adolescence or adulthood can have differential short- and long-term impacts on drug sensitivity.
Collapse
Affiliation(s)
- Ruiyong Wu
- College of Bioscience and Biotechnology, Yangzhou University, Yangzhou, China
| | - Shinnyi Chou
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ming Li
- Department of Psychology, Nanjing University, Nanjing, China.
| |
Collapse
|
2
|
Gogos A, Sbisa A, van den Buuse M. Disruption of NMDA receptor-mediated regulation of PPI in the maternal immune activation model of schizophrenia is restored by 17β-estradiol and raloxifene. Schizophr Res 2024; 267:432-440. [PMID: 38642484 DOI: 10.1016/j.schres.2024.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 02/01/2024] [Accepted: 04/03/2024] [Indexed: 04/22/2024]
Abstract
Maternal immune activation (MIA) during pregnancy is known to increase the risk of development of schizophrenia in the offspring. Sex steroid hormone analogues have been proposed as potential antipsychotic treatments but the mechanisms of action involved remain unclear. Estrogen has been shown to alter N-methyl-d-aspartate (NMDA) receptor binding in the brain. We therefore studied the effect of chronic treatment with 17β-estradiol, its isomer, 17α-estradiol, and the selective estrogen receptor modulator, raloxifene, on MIA-induced psychosis-like behaviour and the effect of the NMDA receptor antagonist, MK-801. Pregnant rats were treated with saline or the viral mimetic, poly(I:C), on gestational day 15. Adult female offspring were tested for changes in baseline prepulse inhibition (PPI) and the effects of acute treatment with MK-801 on PPI and locomotor activity. Poly(I:C) offspring had significantly lower baseline PPI compared to control offspring, and this effect was prevented by 17β-estradiol and raloxifene, but not 17α-estradiol. MK-801 reduced PPI in control offspring but had no effect in poly(I:C) offspring treated with vehicle. Chronic treatment with 17β-estradiol and raloxifene restored the effect of MK-801 on PPI. There were no effects of MIA or estrogenic treatment on MK-801 induced locomotor hyperactivity. These results show that MIA affects baseline PPI as well as NMDA receptor-mediated regulation of PPI in female rats, and strengthen the view that estrogenic treatment may have antipsychotic effects.
Collapse
Affiliation(s)
- Andrea Gogos
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Alyssa Sbisa
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia; School of Psychology and Public Health, La Trobe University, Bundoora, VIC, Australia
| | - Maarten van den Buuse
- School of Psychology and Public Health, La Trobe University, Bundoora, VIC, Australia.
| |
Collapse
|
3
|
da Rosa ALST, Bezerra OS, Rohde LA, Graeff-Martins AS. Exploring clozapine use in severe psychiatric symptoms associated with autism spectrum disorder: A scoping review. J Psychopharmacol 2024; 38:324-343. [PMID: 38576151 DOI: 10.1177/02698811241241384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/06/2024]
Abstract
BACKGROUND Patients with autism spectrum disorder (ASD) may experience severe psychiatric symptoms, often unresponsive to conventional pharmacological therapies, highlighting the need for more effective alternatives. AIMS This study aims to map and synthesize evidence on the use of clozapine as a therapeutic option for managing severe psychiatric symptomatology co-occurring with ASD. METHODS We conducted a scoping review on multiple sources following the JBI guidelines. The search strategy was inclusive, targeting both peer-reviewed publications and gray literature presenting empirical data on the use of clozapine therapy for patients with ASD accompanied by comorbid psychiatric symptoms. Two independent evaluators performed the selection of studies, data extraction, and critical appraisal. RESULTS The review included 46 studies, encompassing 122 ASD individuals who received clozapine therapy. The sources of evidence comprise 31 case reports, 8 case series, 6 retrospective observational studies, and 1 quasi-experimental prospective study. The tables present the findings along with a narrative summary. Clozapine treatment demonstrated benefits in four groups of severe and treatment-resistant psychiatric symptoms in ASD patients: disruptive behaviors, psychotic symptoms, catatonia, and mood symptoms. Although side effects were common, tolerability was generally satisfactory. However, severe adverse events, such as seizures, moderate neutropenia, and myocarditis, underscore the need for intensive clinical monitoring. CONCLUSIONS While clozapine shows promise as a pharmacological intervention for severe psychopathologies in ASD, more rigorous clinical studies are required to elucidate its efficacy and safety in this population. The limited robustness of the evidence calls for caution, signaling an early research stage into this topic.
Collapse
Affiliation(s)
- André Luiz Schuh Teixeira da Rosa
- Graduate Program of Psychiatry and Behavioral Sciences, Department of Psychiatry, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
- Division of Child and Adolescent Psychiatry, Department of Psychiatry, Hospital de Clínicas de Porto Alegre, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Olivia Sorato Bezerra
- Child Neurology Unit, Department of Pediatrics, Hospital de Clínicas de Porto Alegre, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Luis Augusto Rohde
- Graduate Program of Psychiatry and Behavioral Sciences, Department of Psychiatry, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
- Division of Child and Adolescent Psychiatry, Department of Psychiatry, Hospital de Clínicas de Porto Alegre, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Ana Soledade Graeff-Martins
- Graduate Program of Psychiatry and Behavioral Sciences, Department of Psychiatry, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
- Division of Child and Adolescent Psychiatry, Department of Psychiatry, Hospital de Clínicas de Porto Alegre, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
| |
Collapse
|
4
|
Chou S, Wu R, Li M. Long-term impacts of prenatal maternal immune activation and postnatal maternal separation on maternal behavior in adult female rats: Relevance to postpartum mental disorders. Behav Brain Res 2024; 461:114831. [PMID: 38142861 PMCID: PMC10872411 DOI: 10.1016/j.bbr.2023.114831] [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: 11/12/2023] [Revised: 12/19/2023] [Accepted: 12/19/2023] [Indexed: 12/26/2023]
Abstract
Early life adversities are known to exert long-term negative impacts on psychological and brain functions in adulthood. The present work examined how a prenatal brain insult and a postnatal stressor independently or interactively influence the quality of maternal care of postpartum female rats and their cognitive and emotional functions, as a way to identify the behavioral dysfunctions underlying childhood trauma-induced postpartum mental disorders (as indexed by impaired maternal care). Sprague-Dawley female offspring born from mother rats exposed to polyinosinic:polycytidylic acid (PolyI:C, 4.0-6.0 mg/kg) intended to cause gestational maternal immune activation (MIA) or saline were subjected to a repeated maternal separation stress (RMS, 3 h/day) or no separation for 9 days in the first two weeks of life (a 2 × 2 design). When these offspring became mothers, their attentional filtering ability (as measured in the prepulse inhibition of acoustic startle reflex test), positive hedonic response (as measured in the sucrose preference test), and negative emotional response (as measured in the startle reflex and fear-potentiated startle test) were examined, along with their home-cage maternal behavior. Virgin littermates served as controls in all the behavioral tests except in maternal behavior. Results showed that mother rats who experienced RMS displayed impaired nest building and crouching/nursing activities. RMS also interacted with MIA to alter pup retrieval latency and startle reactivity, such that MIA-RMS dams demonstrated significantly slower pup retrieval latency and higher startle magnitude compared to either RMS-only and MIA-only mothers. MIA also disrupted attentional filtering ability, with significantly lower prepulse inhibition. However, neither prenatal MIA nor postnatal RMS impaired sucrose preference or the acquisition of fear-potentiated startle. These results indicate that prenatal stress and postnatal adversity could impair maternal behavior individually, and interact with each other, causing impairments in attention, emotion and maternal motivation.
Collapse
Affiliation(s)
- Shinnyi Chou
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ruiyong Wu
- College of Bioscience and Biotechnology, Yangzhou University, Yangzhou, China
| | - Ming Li
- Department of Psychology, Nanjing University, Nanjing, China.
| |
Collapse
|
5
|
Speers LJ, Chin P, Bilkey DK. No evidence that acute clozapine administration alters CA1 phase precession in rats. Brain Res 2023; 1814:148446. [PMID: 37301424 DOI: 10.1016/j.brainres.2023.148446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 05/25/2023] [Accepted: 06/04/2023] [Indexed: 06/12/2023]
Abstract
Hippocampal phase precession, wherein there is a systematic shift in the phase of neural firing against the underlying theta activity, is proposed to play an important role in the sequencing of information in memory. Previous research shows that the starting phase of precession is more variable in rats following maternal immune activation (MIA), a known risk factor for schizophrenia. Since starting phase variability has the potential to disorganize the construction of sequences of information, we tested whether the atypical antipsychotic clozapine, which ameliorates some cognitive deficits in schizophrenia, alters this aspect of phase precession. Either saline or clozapine (5 mg/kg) was administered to rats and then CA1 place cell activity was recorded from the CA1 region of the hippocampus as the animals ran around a rectangular track for food reward. When compared to saline trials, acute administration of clozapine did not affect any place cell properties, including those related to phase precession, in either control or MIA animals. Clozapine did, however, produce a reduction in locomotion speed, indicating that its presence had some effect on behaviour. These results help to constrain explanations of phase precession mechanisms and their potential role in sequence learning deficits.
Collapse
Affiliation(s)
| | - Phoebe Chin
- Psychology Dept., Otago Univ., Dunedin, New Zealand
| | | |
Collapse
|
6
|
Murlanova K, Pletnikov MV. Modeling psychotic disorders: Environment x environment interaction. Neurosci Biobehav Rev 2023; 152:105310. [PMID: 37437753 DOI: 10.1016/j.neubiorev.2023.105310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 06/26/2023] [Accepted: 07/05/2023] [Indexed: 07/14/2023]
Abstract
Schizophrenia is a major psychotic disorder with multifactorial etiology that includes interactions between genetic vulnerability and environmental risk factors. In addition, interplay of multiple environmental adversities affects neurodevelopment and may increase the individual risk of developing schizophrenia. Consistent with the two-hit hypothesis of schizophrenia, we review rodent models that combine maternal immune activation as the first hit with other adverse environmental exposures as the second hit. We discuss the strengths and pitfalls of the current animal models of environment x environment interplay and propose some future directions to advance the field.
Collapse
Affiliation(s)
- Kateryna Murlanova
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14203, USA
| | - Mikhail V Pletnikov
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14203, USA; Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
7
|
Edemann-Callesen H, Bernhardt N, Hlusicka EB, Hintz F, Habelt B, Winter R, Neubert I, Pelz M, Filla A, Soto-Montenegro ML, Winter C, Hadar R. Supplement Treatment with NAC and Omega-3 Polyunsaturated Fatty Acids during Pregnancy Partially Prevents Schizophrenia-Related Outcomes in the Poly I:C Rat Model. Antioxidants (Basel) 2023; 12:antiox12051068. [PMID: 37237933 DOI: 10.3390/antiox12051068] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/02/2023] [Accepted: 05/04/2023] [Indexed: 05/28/2023] Open
Abstract
BACKGROUND Heightened levels of inflammation and oxidative stress are thought to be involved in the pathophysiology of schizophrenia. We aimed to assess whether intake of anti-inflammatory and anti-oxidant drugs during pregnancy prevents later schizophrenia-related outcomes in a neurodevelopmental rat model of this disorder. METHODS Pregnant Wistar rats were injected with polyriboinosinic-polyribocytidilic acid (Poly I:C) or saline and subsequently treated with either N-acetyl cysteine (NAC) or omega-3 polyunsaturated fatty acids (PUFAs) until delivery. Controls rats received no treatment. In the offspring, neuroinflammation and anti-oxidant enzyme activity were assessed on postnatal day (PND) 21, 33, 48, and 90. Behavioral testing was performed at PND 90, followed by post-mortem neurochemical assessment and ex vivo MRI. RESULTS The supplement treatment led to a quicker restoration of the wellbeing of dams. In the adolescent Poly I:C offspring, the supplement treatment prevented an increase in microglial activity and partially prevented a deregulation in the anti-oxidant defense system. In the adult Poly I:C offspring, supplement treatment partially prevented dopamine deficits, which was paralleled by some changes in behavior. Exposure to omega-3 PUFAs prevented the enlargement of lateral ventricles. CONCLUSION Intake of over-the-counter supplements may assist in especially targeting the inflammatory response related to schizophrenia pathophysiology, aiding in diminishing later disease severity in the offspring.
Collapse
Affiliation(s)
- Henriette Edemann-Callesen
- Department of Psychiatry and Neuroscience, Campus Mitte, Charité University Medicine Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Nadine Bernhardt
- Department of Psychiatry and Psychotherapy, Medical Faculty Carl Gustav Carus, Technische Universität, 01307 Dresden, Germany
| | - Elizabeth Barroeta Hlusicka
- Department of Psychiatry and Psychotherapy, Medical Faculty Carl Gustav Carus, Technische Universität, 01307 Dresden, Germany
| | - Franziska Hintz
- Department of Psychiatry and Neuroscience, Campus Mitte, Charité University Medicine Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Bettina Habelt
- Department of Psychiatry and Psychotherapy, Medical Faculty Carl Gustav Carus, Technische Universität, 01307 Dresden, Germany
- Leibniz Institute of Polymer Research Dresden, 01069 Dresden, Germany
| | - Rebecca Winter
- Department of Psychiatry and Psychotherapy, Medical Faculty Carl Gustav Carus, Technische Universität, 01307 Dresden, Germany
| | - Isabell Neubert
- Department of Psychiatry and Psychotherapy, Medical Faculty Carl Gustav Carus, Technische Universität, 01307 Dresden, Germany
| | - Meike Pelz
- Department of Psychiatry and Psychotherapy, Medical Faculty Carl Gustav Carus, Technische Universität, 01307 Dresden, Germany
| | - Alexandra Filla
- Department of Psychiatry and Psychotherapy, Medical Faculty Carl Gustav Carus, Technische Universität, 01307 Dresden, Germany
| | - Maria Luisa Soto-Montenegro
- Instituto de Investigación Sanitaria Gregorio Marañón, 28007 Madrid, Spain
- CIBER de Salud Mental, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Grupo de Investigación de Alto Rendimiento en Fisiopatología y Farmacología del Sistema Digestivo (NeuGut-URJC), Universidad Rey Juan Carlos, 28922 Alcorcón, Spain
| | - Christine Winter
- Department of Psychiatry and Neuroscience, Campus Mitte, Charité University Medicine Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Ravit Hadar
- Department of Psychiatry and Neuroscience, Campus Mitte, Charité University Medicine Berlin, Charitéplatz 1, 10117 Berlin, Germany
| |
Collapse
|
8
|
Petty A, Howes O, Eyles D. Animal Models of Relevance to the Schizophrenia Prodrome. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2023; 3:22-32. [PMID: 36712558 PMCID: PMC9874082 DOI: 10.1016/j.bpsgos.2021.12.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 11/29/2021] [Accepted: 12/01/2021] [Indexed: 02/01/2023] Open
Abstract
Patients with schizophrenia often undergo a prodromal phase prior to diagnosis. Given the absence of significant therapeutic improvements, attention has recently shifted to the possibility of intervention during this early stage to delay or diminish symptom severity or even prevent onset. Unfortunately, the 20 or so trials of intervention to date have not been successful in either preventing onset or improving long-term outcomes in subjects who are at risk of developing schizophrenia. One reason may be that the biological pathways an effective intervention must target are not static. The prodromal phase typically occurs during late adolescence, a period during which a number of brain circuits and structures are still maturing. We propose that developing a deeper understanding of which circuits/processes and brain structures are still maturing at this time and which processes drive the transition to schizophrenia will take us a step closer to developing better prophylactic interventions. Fortunately, such knowledge is now emerging from clinical studies, complemented by work in animal models. Our task here is to describe what would constitute an appropriate animal model to study and to potentially intervene in such processes. Such a model would allow invasive analysis of the cellular and molecular substrates of the progressive neurobiology that defines the schizophrenia prodrome and hopefully offer valuable insights into potential prophylactic targets.
Collapse
Affiliation(s)
- Alice Petty
- Neuroscience Research Australia, Sydney, New South Wales, Australia.,Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia
| | | | - Darryl Eyles
- Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia.,Queensland Centre for Mental Health Research, Wacol, Queensland, Australia
| |
Collapse
|
9
|
Garcia-Partida JA, Torres-Sanchez S, MacDowell K, Fernández-Ponce MT, Casas L, Mantell C, Soto-Montenegro ML, Romero-Miguel D, Lamanna-Rama N, Leza JC, Desco M, Berrocoso E. The effects of mango leaf extract during adolescence and adulthood in a rat model of schizophrenia. Front Pharmacol 2022; 13:886514. [PMID: 35959428 PMCID: PMC9360613 DOI: 10.3389/fphar.2022.886514] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 06/30/2022] [Indexed: 11/13/2022] Open
Abstract
There is evidence that in schizophrenia, imbalances in inflammatory and oxidative processes occur during pregnancy and in the early postnatal period, generating interest in the potential therapeutic efficacy of anti-inflammatory and antioxidant compounds. Mangiferin is a polyphenolic compound abundant in the leaves of Mangifera indica L. that has robust antioxidant and anti-inflammatory properties, making it a potential candidate for preventive or co-adjuvant therapy in schizophrenia. Hence, this study set-out to evaluate the effect of mango leaf extract (MLE) in a model of schizophrenia based on maternal immune activation, in which Poly I:C (4 mg/kg) is administered intravenously to pregnant rats. Young adult (postnatal day 60-70) or adolescent (postnatal day 35-49) male offspring received MLE (50 mg/kg of mangiferin) daily, and the effects of MLE in adolescence were compared to those of risperidone, assessing behavior, brain magnetic resonance imaging (MRI), and oxidative/inflammatory and antioxidant mediators in the adult offspring. MLE treatment in adulthood reversed the deficit in prepulse inhibition (PPI) but it failed to attenuate the sensitivity to amphetamine and the deficit in novel object recognition (NOR) induced. By contrast, adolescent MLE treatment prevented the sensorimotor gating deficit in the PPI test, producing an effect similar to that of risperidone. This MLE treatment also produced a reduction in grooming behavior, but it had no effect on anxiety or novel object recognition memory. MRI studies revealed that adolescent MLE administration partially counteracted the cortical shrinkage, and cerebellum and ventricle enlargement. In addition, MLE administration in adolescence reduced iNOS mediated inflammatory activation and it promoted the expression of biomarkers of compensatory antioxidant activity in the prefrontal cortex and hippocampus, as witnessed through the reduction of Keap1 and the accumulation of NRF2 and HO1. Together, these findings suggest that MLE might be an alternative therapeutic or preventive add-on strategy to improve the clinical expression of schizophrenia in adulthood, while also modifying the time course of this disease at earlier stages in populations at high-risk.
Collapse
Affiliation(s)
- Jose Antonio Garcia-Partida
- Neuropsychopharmacology and Psychobiology Research Group, Department of Neuroscience, University of Cádiz, Cádiz, Spain
- Instituto de Investigación e Innovación en Ciencias Biomédicas de Cádiz, INiBICA, Hospital Universitario Puerta del Mar, Cádiz, Spain
| | - Sonia Torres-Sanchez
- Instituto de Investigación e Innovación en Ciencias Biomédicas de Cádiz, INiBICA, Hospital Universitario Puerta del Mar, Cádiz, Spain
- Neuropsychopharmacology and Psychobiology Research Group, Psychobiology Area, Department of Psychology, University of Cádiz, Cádiz, Spain
- Ciber of Mental Health (CIBERSAM), ISCIII, Madrid, Spain
| | - Karina MacDowell
- Ciber of Mental Health (CIBERSAM), ISCIII, Madrid, Spain
- Department of Pharmacology and Toxicology, Faculty of Medicine, Universidad Complutense de Madrid (UCM), Health Research Institute Hospital 12 de Octubre (imas12), Institute of Research in Neurochemistry IUIN-UCM, Madrid, Spain
| | | | - Lourdes Casas
- Department of Chemical Engineering and Food Technology, Science Faculty, University of Cádiz, Cádiz, Spain
| | - Casimiro Mantell
- Department of Chemical Engineering and Food Technology, Science Faculty, University of Cádiz, Cádiz, Spain
| | - María Luisa Soto-Montenegro
- Ciber of Mental Health (CIBERSAM), ISCIII, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- High Performance Research Group in Physiopathology and Pharmacology of the Digestive System (NeuGut), Universidad Rey Juan Carlos, Madrid, Spain
| | - Diego Romero-Miguel
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Departamento de Bioingeniería e Ingeniería Aeroespacial, Universidad Carlos III de Madrid, Madrid, Spain
| | - Nicolás Lamanna-Rama
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Departamento de Bioingeniería e Ingeniería Aeroespacial, Universidad Carlos III de Madrid, Madrid, Spain
| | - Juan Carlos Leza
- Ciber of Mental Health (CIBERSAM), ISCIII, Madrid, Spain
- Department of Pharmacology and Toxicology, Faculty of Medicine, Universidad Complutense de Madrid (UCM), Health Research Institute Hospital 12 de Octubre (imas12), Institute of Research in Neurochemistry IUIN-UCM, Madrid, Spain
| | - Manuel Desco
- Ciber of Mental Health (CIBERSAM), ISCIII, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Departamento de Bioingeniería e Ingeniería Aeroespacial, Universidad Carlos III de Madrid, Madrid, Spain
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Esther Berrocoso
- Instituto de Investigación e Innovación en Ciencias Biomédicas de Cádiz, INiBICA, Hospital Universitario Puerta del Mar, Cádiz, Spain
- Neuropsychopharmacology and Psychobiology Research Group, Psychobiology Area, Department of Psychology, University of Cádiz, Cádiz, Spain
- Ciber of Mental Health (CIBERSAM), ISCIII, Madrid, Spain
| |
Collapse
|
10
|
Maternal immune activation with high molecular weight poly(I:C) in Wistar rats leads to elevated immune cell chemoattractants. J Neuroimmunol 2022; 364:577813. [DOI: 10.1016/j.jneuroim.2022.577813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 12/19/2021] [Accepted: 01/10/2022] [Indexed: 11/20/2022]
|
11
|
Büki A, Bohár Z, Kekesi G, Vécsei L, Horvath G. Wisket rat model of schizophrenia: Impaired motivation and, altered brain structure, but no anhedonia. Physiol Behav 2021; 244:113651. [PMID: 34800492 DOI: 10.1016/j.physbeh.2021.113651] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 11/12/2021] [Accepted: 11/13/2021] [Indexed: 01/17/2023]
Abstract
It is well-known that the poor cognition in schizophrenia is strongly linked to negative symptoms, including motivational deficit, which due to, at least partially, anhedonia. The goal of this study was to explore whether the schizophrenia-like Wisket animals with impaired motivation (obtained in the reward-based hole-board test), also show decreased hedonic behavior (investigated with the sucrose preference test). While neurochemical alterations of different neurotransmitter systems have been detected in the Wisket rats, no research has been performed on structural changes. Therefore, our additional aim was to reveal potential neuroanatomical and structural alterations in different brain regions in these rats. The rats showed decreased general motor activity (locomotion, rearing and exploration) and impaired task performance in the hole-board test compared to the controls, whereas no significant difference was observed in the sucrose preference test between the groups. The Wisket rats exhibited a significant decrease in the frontal cortical thickness and the hippocampal area, and moderate increases in the lateral ventricles and cell disarray in the CA3 subfield of hippocampus. To our knowledge, this is the first study to investigate the hedonic behavior and neuroanatomical alterations in a multi-hit animal model of schizophrenia. The results obtained in the sucrose preference test suggest that anhedonic behavior might not be involved in the impaired motivation obtained in the hole-board test. The neuropathological changes agree with findings obtained in patients with schizophrenia, which refine the high face validity of the Wisket model.
Collapse
Affiliation(s)
- Alexandra Büki
- Department of Physiology, Albert Szent-Györgyi Medical School, University of Szeged, Dóm tér 10., Szeged, H-6720, Hungary.
| | - Zsuzsanna Bohár
- MTA-SZTE Neuroscience Research Group, Semmelweis u. 6, Szeged, H-6725, Hungary
| | - Gabriella Kekesi
- Department of Physiology, Albert Szent-Györgyi Medical School, University of Szeged, Dóm tér 10., Szeged, H-6720, Hungary
| | - László Vécsei
- MTA-SZTE Neuroscience Research Group, Semmelweis u. 6, Szeged, H-6725, Hungary; Department of Neurology, Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis u. 6., Szeged, H-6725, Hungary; Interdisciplinary Excellence Center, Department of Neurology, Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis u. 6., Szeged, H-6725 Hungary
| | - Gyongyi Horvath
- Department of Physiology, Albert Szent-Györgyi Medical School, University of Szeged, Dóm tér 10., Szeged, H-6720, Hungary
| |
Collapse
|
12
|
Guma E, Bordignon PDC, Devenyi GA, Gallino D, Anastassiadis C, Cvetkovska V, Barry AD, Snook E, Germann J, Greenwood CMT, Misic B, Bagot RC, Chakravarty MM. Early or Late Gestational Exposure to Maternal Immune Activation Alters Neurodevelopmental Trajectories in Mice: An Integrated Neuroimaging, Behavioral, and Transcriptional Study. Biol Psychiatry 2021; 90:328-341. [PMID: 34053674 DOI: 10.1016/j.biopsych.2021.03.017] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/23/2021] [Accepted: 03/15/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND Exposure to maternal immune activation (MIA) in utero is a risk factor for neurodevelopmental disorders later in life. The impact of the gestational timing of MIA exposure on downstream development remains unclear. METHODS We characterized neurodevelopmental trajectories of mice exposed to the viral mimetic poly I:C (polyinosinic:polycytidylic acid) either on gestational day 9 (early) or on day 17 (late) using longitudinal structural magnetic resonance imaging from weaning to adulthood. Using multivariate methods, we related neuroimaging and behavioral variables for the time of greatest alteration (adolescence/early adulthood) and identified regions for further investigation using RNA sequencing. RESULTS Early MIA exposure was associated with accelerated brain volume increases in adolescence/early adulthood that normalized in later adulthood in the striatum, hippocampus, and cingulate cortex. Similarly, alterations in anxiety-like, stereotypic, and sensorimotor gating behaviors observed in adolescence normalized in adulthood. MIA exposure in late gestation had less impact on anatomical and behavioral profiles. Multivariate maps associated anxiety-like, social, and sensorimotor gating deficits with volume of the dorsal and ventral hippocampus and anterior cingulate cortex, among others. The most transcriptional changes were observed in the dorsal hippocampus, with genes enriched for fibroblast growth factor regulation, autistic behaviors, inflammatory pathways, and microRNA regulation. CONCLUSIONS Leveraging an integrated hypothesis- and data-driven approach linking brain-behavior alterations to the transcriptome, we found that MIA timing differentially affects offspring development. Exposure in late gestation leads to subthreshold deficits, whereas exposure in early gestation perturbs brain development mechanisms implicated in neurodevelopmental disorders.
Collapse
Affiliation(s)
- Elisa Guma
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada; Computational Brain Imaging Lab, Cerebral Imaging Center, Douglas Mental Health University Institute, Montreal, Quebec, Canada.
| | - Pedro do Couto Bordignon
- Department of Psychology, McGill University, Montreal, Quebec, Canada; Ludmer Center for Neuroinformatics and Mental Health, Montreal, Quebec, Canada
| | - Gabriel A Devenyi
- Department of Psychiatry, McGill University, Montreal, Quebec, Canada; Computational Brain Imaging Lab, Cerebral Imaging Center, Douglas Mental Health University Institute, Montreal, Quebec, Canada
| | - Daniel Gallino
- Computational Brain Imaging Lab, Cerebral Imaging Center, Douglas Mental Health University Institute, Montreal, Quebec, Canada
| | - Chloe Anastassiadis
- Computational Brain Imaging Lab, Cerebral Imaging Center, Douglas Mental Health University Institute, Montreal, Quebec, Canada; Institute of Medical Science & Collaborative Program in Neuroscience, University of Toronto, Toronto, Ontario, Canada
| | | | - Amadou D Barry
- Departments of Human Genetics and Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Quebec, Canada; Ludmer Center for Neuroinformatics and Mental Health, Montreal, Quebec, Canada
| | - Emily Snook
- Computational Brain Imaging Lab, Cerebral Imaging Center, Douglas Mental Health University Institute, Montreal, Quebec, Canada; Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Jurgen Germann
- Computational Brain Imaging Lab, Cerebral Imaging Center, Douglas Mental Health University Institute, Montreal, Quebec, Canada; University Health Network, Toronto, Ontario, Canada
| | - Celia M T Greenwood
- Gerald Bronfman Department of Oncology, McGill University, Montreal, Quebec, Canada; Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University, Montreal, Quebec, Canada; Departments of Human Genetics and Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Quebec, Canada; Ludmer Center for Neuroinformatics and Mental Health, Montreal, Quebec, Canada
| | - Bratislav Misic
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Rosemary C Bagot
- Department of Psychology, McGill University, Montreal, Quebec, Canada; Ludmer Center for Neuroinformatics and Mental Health, Montreal, Quebec, Canada
| | - M Mallar Chakravarty
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada; Department of Psychiatry, McGill University, Montreal, Quebec, Canada; Department of Biological and Biomedical Engineering, McGill University, Montreal, Quebec, Canada; Computational Brain Imaging Lab, Cerebral Imaging Center, Douglas Mental Health University Institute, Montreal, Quebec, Canada.
| |
Collapse
|
13
|
Page NF, Gandal MJ, Estes ML, Cameron S, Buth J, Parhami S, Ramaswami G, Murray K, Amaral DG, Van de Water JA, Schumann CM, Carter CS, Bauman MD, McAllister AK, Geschwind DH. Alterations in Retrotransposition, Synaptic Connectivity, and Myelination Implicated by Transcriptomic Changes Following Maternal Immune Activation in Nonhuman Primates. Biol Psychiatry 2021; 89:896-910. [PMID: 33386132 PMCID: PMC8052273 DOI: 10.1016/j.biopsych.2020.10.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 10/07/2020] [Accepted: 10/09/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND Maternal immune activation (MIA) is a proposed risk factor for multiple neuropsychiatric disorders, including schizophrenia. However, the molecular mechanisms through which MIA imparts risk remain poorly understood. A recently developed nonhuman primate model of exposure to the viral mimic poly:ICLC during pregnancy shows abnormal social and repetitive behaviors and elevated striatal dopamine, a molecular hallmark of human psychosis, providing an unprecedented opportunity for studying underlying molecular correlates. METHODS We performed RNA sequencing across psychiatrically relevant brain regions (prefrontal cortex, anterior cingulate, hippocampus) and primary visual cortex for comparison from 3.5- to 4-year-old male MIA-exposed and control offspring-an age comparable to mid adolescence in humans. RESULTS We identify 266 unique genes differentially expressed in at least one brain region, with the greatest number observed in hippocampus. Co-expression networks identified region-specific alterations in synaptic signaling and oligodendrocytes. Although we observed temporal and regional differences, transcriptomic changes were shared across first- and second-trimester exposures, including for the top differentially expressed genes-PIWIL2 and MGARP. In addition to PIWIL2, several other regulators of retrotransposition and endogenous transposable elements were dysregulated following MIA, potentially connecting MIA to retrotransposition. CONCLUSIONS Together, these results begin to elucidate the brain-level molecular processes through which MIA may impart risk for psychiatric disease.
Collapse
Affiliation(s)
- Nicholas F Page
- Department of Psychiatry, Center for Autism Research and Treatment, Los Angeles, California; Department of Cell Biology and Neuroscience, Rutgers University-New Brunswick, Piscataway, New Jersey
| | - Michael J Gandal
- Department of Psychiatry, Center for Autism Research and Treatment, Los Angeles, California
| | - Myka L Estes
- Center for Neuroscience, School of Medicine, University of California, Davis, Davis, California
| | - Scott Cameron
- Center for Neuroscience, School of Medicine, University of California, Davis, Davis, California
| | - Jessie Buth
- Department of Psychiatry, Center for Autism Research and Treatment, Los Angeles, California; Program in Neurobehavioral Genetics, Center for Autism Research and Treatment, Los Angeles, California
| | - Sepideh Parhami
- Department of Psychiatry, Center for Autism Research and Treatment, Los Angeles, California; Program in Neurobehavioral Genetics, Center for Autism Research and Treatment, Los Angeles, California
| | - Gokul Ramaswami
- Department of Psychiatry, Center for Autism Research and Treatment, Los Angeles, California; Program in Neurobehavioral Genetics, Center for Autism Research and Treatment, Los Angeles, California
| | - Karl Murray
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California, Davis, Davis, California
| | - David G Amaral
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California, Davis, Davis, California
| | - Judy A Van de Water
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California, Davis, Davis, California
| | - Cynthia M Schumann
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California, Davis, Davis, California
| | - Cameron S Carter
- Center for Neuroscience, School of Medicine, University of California, Davis, Davis, California; Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California, Davis, Davis, California
| | - Melissa D Bauman
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California, Davis, Davis, California
| | - A Kimberley McAllister
- Center for Neuroscience, School of Medicine, University of California, Davis, Davis, California; Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California, Davis, Davis, California
| | - Daniel H Geschwind
- Department of Psychiatry, Center for Autism Research and Treatment, Los Angeles, California; Program in Neurobehavioral Genetics, Center for Autism Research and Treatment, Los Angeles, California; Department of Neurology, Center for Autism Research and Treatment, Los Angeles, California; Department of Human Genetics, Semel Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California.
| |
Collapse
|
14
|
Rittweger N, Ishorst T, Barmashenko G, Aliane V, Winter C, Funke K. Effects of iTBS-rTMS on the Behavioral Phenotype of a Rat Model of Maternal Immune Activation. Front Behav Neurosci 2021; 15:670699. [PMID: 33967716 PMCID: PMC8098712 DOI: 10.3389/fnbeh.2021.670699] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 03/30/2021] [Indexed: 12/23/2022] Open
Abstract
Repetitive transcranial magnetic stimulation (rTMS) is considered a promising therapeutic tool for treating neuropsychiatric diseases. Previously, we found intermittent theta-burst stimulation (iTBS) rTMS to be most effective in modulating cortical excitation-inhibition balance in rats, accompanied by improved cortical sensory processing and sensory learning performance. Using an animal schizophrenia model based on maternal immune activation (MIA) we tested if iTBS applied to either adult or juvenile rats can affect the behavioral phenotype in a therapeutic or preventive manner, respectively. In a sham-controlled fashion, iTBS effects in MIA rats were compared with rats receiving vehicle NaCl injection instead of the synthetic viral strand. Prior to iTBS, adult MIA rats showed deficits in sensory gating, as tested with prepulse inhibition (PPI) of the acoustic startle reflex, and deficits in novel object recognition (NOR). No differences between MIA and control rats were evident with regard to signs of anxiety, anhedonia and depression but MIA rats were somewhat superior to controls during the training phase of Morris Water Maze (MWM) test. MIA but not control rats significantly improved in PPI following iTBS at adulthood but without significant differences between verum and sham application. If applied during adolescence, verum but not sham-iTBS improved NOR at adulthood but no difference in PPI was evident in rats treated either with sham or verum-iTBS. MIA and control rat responses to sham-iTBS applied at adulthood differed remarkably, indicating a different physiological reaction to the experimental experiences. Although verum-iTBS was not superior to sham-iTBS, MIA rats seemed to benefit from the treatment procedure in general, since differences-in relation to control rats declined or disappeared. Even if classical placebo effects can be excluded, motor or cognitive challenges or the entire handling procedure during the experiments appear to alleviate the behavioral impairments of MIA rats.
Collapse
Affiliation(s)
- Nadine Rittweger
- Department of Neurophysiology, Medical Faculty, Ruhr-University, Bochum, Germany
| | - Tanja Ishorst
- Department of Neurophysiology, Medical Faculty, Ruhr-University, Bochum, Germany
| | - Gleb Barmashenko
- Department of Neurophysiology, Medical Faculty, Ruhr-University, Bochum, Germany.,AIO-Studien-gGmbH, Berlin, Germany
| | - Verena Aliane
- Department of Neurophysiology, Medical Faculty, Ruhr-University, Bochum, Germany
| | - Christine Winter
- Department of Psychiatry and Psychotherapy, Charité University Medicine Berlin, Berlin, Germany.,Department of Psychiatry and Psychotherapy, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Klaus Funke
- Department of Neurophysiology, Medical Faculty, Ruhr-University, Bochum, Germany
| |
Collapse
|
15
|
Edemann-Callesen H, Winter C, Hadar R. Using cortical non-invasive neuromodulation as a potential preventive treatment in schizophrenia - A review. Brain Stimul 2021; 14:643-651. [PMID: 33819680 DOI: 10.1016/j.brs.2021.03.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 02/11/2021] [Accepted: 03/23/2021] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Evidence suggests that schizophrenia constitutes a neurodevelopmental disorder, characterized by a gradual emergence of behavioral and neurobiological abnormalities over time. Therefore, applying early interventions to prevent later manifestation of symptoms is appealing. OBJECTIVE This review focuses on the use of cortical neuromodulation in schizophrenia and its potential as a preventive treatment approach. We present clinical and preclinical findings investigating the use of neuromodulation in schizophrenia, including the current research focusing on cortical non-invasive stimulation and its possibility as a future preventive treatment. METHODS We performed a search in Medline (PubMed) in September 2020 using a combination of relevant medical subject headings (MeSH) and text words. The search included human and preclinical trials as well as existing systematic reviews and meta-analysis. There were no restrictions on language or the date of publication. RESULTS Neurodevelopmental animal models may be used to investigate how the disease progresses and thus which brain areas ideally should be targeted at a given time point. Here, abnormalities of the prefrontal cortex have been often identified as an early and persistent impairment in schizophrenia. Currently there is insufficient evidence to either support or refute the use of neuromodulation to the cortex in adult patients with already manifested symptoms. However, preclinical results show that early non-invasive neuromodulation to the prefrontal cortex of adolescent animals, sufficiently prevents later psychosis-relevant abnormalities in adulthood. This points to the promising potential of cortical non-invasive neuromodulation as a preventive treatment when applied early in the course of the disease. CONCLUSION Preclinical translational-oriented findings indicate, that neuromodulation to cortical areas offers the possibility of targeting early neuropathology and through this diminish the progression of a later schizophrenic profile. Further studies are needed to investigate whether such early cortical stimulation may serve as a future preventive treatment in schizophrenia.
Collapse
Affiliation(s)
- Henriette Edemann-Callesen
- Department of Psychiatry and Psychotherapy, Charité Campus Mitte, Charité -Universitätsmedizin, Berlin, Germany
| | - Christine Winter
- Department of Psychiatry and Psychotherapy, Charité Campus Mitte, Charité -Universitätsmedizin, Berlin, Germany
| | - Ravit Hadar
- Department of Psychiatry and Psychotherapy, Charité Campus Mitte, Charité -Universitätsmedizin, Berlin, Germany.
| |
Collapse
|
16
|
Sbisa A, Kusljic S, Zethoven D, van den Buuse M, Gogos A. The effect of 17β-estradiol on maternal immune activation-induced changes in prepulse inhibition and dopamine receptor and transporter binding in female rats. Schizophr Res 2020; 223:249-257. [PMID: 32878698 DOI: 10.1016/j.schres.2020.08.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 08/09/2020] [Accepted: 08/17/2020] [Indexed: 12/20/2022]
Abstract
Maternal immune activation (MIA) during pregnancy is associated with an increased risk of development of schizophrenia in later life. 17β-estradiol treatment may improve schizophrenia symptoms, but little is known about its efficacy on MIA-induced psychosis-like behavioural deficits in animals. Therefore, in this study we used the poly(I:C) neurodevelopmental model of schizophrenia to examine whether MIA-induced psychosis-like behavioural and neurochemical changes can be attenuated by chronic treatment (2-6 weeks) with 17β-estradiol. Pregnant rats were treated with saline or the viral mimetic, poly(I:C), on gestational day 15 and adult female offspring were tested for changes in prepulse inhibition (PPI) and density of dopamine D1 and D2 receptors and dopamine transporters in the forebrain compared to control offspring. Poly(I:C)-treated offspring exhibited significantly disrupted PPI, an effect which was reversed by chronic treatment with 17β-estradiol. In control offspring, but not poly(I:C) offspring, PPI was significantly reduced by acute treatment with either the dopamine D1/D2 receptor agonist, apomorphine, or dopamine releaser, methamphetamine. 17β-estradiol restored the effect of apomorphine, but not methamphetamine, on PPI in poly(I:C) offspring. There was a strong trend for a dopamine D2 receptor binding density increase in the nucleus accumbens core region in poly(I:C) offspring, and this was reversed by chronic 17β-estradiol treatment. No changes were found in the nucleus accumbens shell, caudate putamen or frontal cortex or in the density of dopamine D1 receptors or transporters. These findings suggest that 17β-estradiol may improve some symptoms of schizophrenia, an effect that may be mediated by selective changes in dopamine D2 receptor density.
Collapse
Affiliation(s)
- Alyssa Sbisa
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia; School of Psychology and Public Health, La Trobe University, Bundoora, VIC, Australia
| | - Snezana Kusljic
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia; Department of Nursing, University of Melbourne, Parkville, VIC, Australia
| | - Damon Zethoven
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Maarten van den Buuse
- School of Psychology and Public Health, La Trobe University, Bundoora, VIC, Australia; Department of Pharmacology, University of Melbourne, Parkville, VIC, Australia; The College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, QLD, Australia
| | - Andrea Gogos
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
17
|
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
|
18
|
Donegan JJ, Boley AM, Glenn JP, Carless MA, Lodge DJ. Developmental alterations in the transcriptome of three distinct rodent models of schizophrenia. PLoS One 2020; 15:e0232200. [PMID: 32497066 PMCID: PMC7272013 DOI: 10.1371/journal.pone.0232200] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 04/09/2020] [Indexed: 11/25/2022] Open
Abstract
Schizophrenia is a debilitating disorder affecting just under 1% of the population. While the symptoms of this disorder do not appear until late adolescence, pathological alterations likely occur earlier, during development in utero. While there is an increasing literature examining transcriptome alterations in patients, it is not possible to examine the changes in gene expression that occur during development in humans that will develop schizophrenia. Here we utilize three distinct rodent developmental disruption models of schizophrenia to examine potential overlapping alterations in the transcriptome, with a specific focus on markers of interneuron development. Specifically, we administered either methylazoxymethanol acetate (MAM), Polyinosinic:polycytidylic acid (Poly I:C), or chronic protein malnutrition, on GD 17 and examined mRNA expression in the developing hippocampus of the offspring 18 hours later. Here, we report alterations in gene expression that may contribute to the pathophysiology of schizophrenia, including significant alterations in interneuron development and ribosome function.
Collapse
Affiliation(s)
- Jennifer J. Donegan
- Department of Pharmacology and Center for Biomedical Neuroscience, University of Texas Health Science Center, San Antonio, TX, United States of America
| | - Angela M. Boley
- Department of Pharmacology and Center for Biomedical Neuroscience, University of Texas Health Science Center, San Antonio, TX, United States of America
| | - Jeremy P. Glenn
- Population Health Program, Texas Biomedical Research Institute, San Antonio, TX, United States of America
| | - Melanie A. Carless
- Population Health Program, Texas Biomedical Research Institute, San Antonio, TX, United States of America
| | - Daniel J. Lodge
- Department of Pharmacology and Center for Biomedical Neuroscience, University of Texas Health Science Center, San Antonio, TX, United States of America
| |
Collapse
|
19
|
Stark T, Di Bartolomeo M, Di Marco R, Drazanova E, Platania CBM, Iannotti FA, Ruda-Kucerova J, D'Addario C, Kratka L, Pekarik V, Piscitelli F, Babinska Z, Fedotova J, Giurdanella G, Salomone S, Sulcova A, Bucolo C, Wotjak CT, Starcuk Z, Drago F, Mechoulam R, Di Marzo V, Micale V. Altered dopamine D3 receptor gene expression in MAM model of schizophrenia is reversed by peripubertal cannabidiol treatment. Biochem Pharmacol 2020; 177:114004. [PMID: 32360362 DOI: 10.1016/j.bcp.2020.114004] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 04/24/2020] [Indexed: 12/16/2022]
Abstract
Gestational methylazoxymethanol acetate (MAM) treatment produces offspring with adult phenotype relevant to schizophrenia, including positive- and negative-like symptoms, cognitive deficits, dopaminergic dysfunction, structural and functional abnormalities. Here we show that adult rats prenatally treated with MAM at gestational day 17 display significant increase in dopamine D3 receptor (D3) mRNA expression in prefrontal cortex (PFC), hippocampus and nucleus accumbens, accompanied by increased expression of dopamine D2 receptor (D2) mRNA exclusively in the PFC. Furthermore, a significant change in the blood perfusion at the level of the circle of Willis and hippocampus, paralleled by the enlargement of lateral ventricles, was also detected by magnetic resonance imaging (MRI) techniques. Peripubertal treatment with the non-euphoric phytocannabinoid cannabidiol (30 mg/kg) from postnatal day (PND) 19 to PND 39 was able to reverse in MAM exposed rats: i) the up-regulation of the dopamine D3 receptor mRNA (only partially prevented by haloperidol 0.6 mg/kg/day); and ii) the regional blood flow changes in MAM exposed rats. Molecular modelling predicted that cannabidiol could bind preferentially to dopamine D3 receptor, where it may act as a partial agonist according to conformation of ionic-lock, which is highly conserved in GPCRs. In summary, our results demonstrate that the mRNA expression of both dopamine D2 and D3 receptors is altered in the MAM model; however only the transcript levels of D3 are affected by cannabidiol treatment, likely suggesting that this gene might not only contribute to the schizophrenia symptoms but also represent an unexplored target for the antipsychotic activity of cannabidiol.
Collapse
Affiliation(s)
- Tibor Stark
- Department of Pharmacology, Faculty of Medicine, Masaryk University, Brno, Czech Republic; RG "Neuronal Plasticity", Max Planck Institute of Psychiatry, Munich, Germany
| | - Martina Di Bartolomeo
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Roberta Di Marco
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Eva Drazanova
- Department of Pharmacology, Faculty of Medicine, Masaryk University, Brno, Czech Republic; Institute of Scientific Instruments of the Czech Academy of Sciences, Brno, Czech Republic
| | | | - Fabio Arturo Iannotti
- Institute of Biomolecular Chemistry, Consiglio Nazionale delle Ricerche, Endocannabinoid Research Group, Naples, Italy
| | - Jana Ruda-Kucerova
- Department of Pharmacology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Claudio D'Addario
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Lucie Kratka
- Institute of Scientific Instruments of the Czech Academy of Sciences, Brno, Czech Republic
| | - Vladimir Pekarik
- Department of Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Fabiana Piscitelli
- Institute of Biomolecular Chemistry, Consiglio Nazionale delle Ricerche, Endocannabinoid Research Group, Naples, Italy
| | - Zuzana Babinska
- Department of Pharmacology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Julia Fedotova
- International Research Centre "Biotechnologies of the Third Millennium", ITMO University, St. Petersburg, Russian Federation; Laboratory of Neuroendocrinology, I.P. Pavlov Institute of Physiology RASci., St. Petersburg, Russian Federation; Lobachevsky State University of Nizhny Novgorod, Institute of Biology and Biomedicine, Nizhny Novgorod, Russian Federation
| | - Giovanni Giurdanella
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Salvatore Salomone
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Alexandra Sulcova
- ICCI - International Cannabis and Cannabinoid Institute, Praha, Czech Republic
| | - Claudio Bucolo
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Carsten T Wotjak
- RG "Neuronal Plasticity", Max Planck Institute of Psychiatry, Munich, Germany; Boehringer Ingelheim Pharma GmbH & KO KG, Germany
| | - Zenon Starcuk
- Institute of Scientific Instruments of the Czech Academy of Sciences, Brno, Czech Republic
| | - Filippo Drago
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Raphael Mechoulam
- Institute for Drug Research, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Vincenzo Di Marzo
- Institute of Biomolecular Chemistry, Consiglio Nazionale delle Ricerche, Endocannabinoid Research Group, Naples, Italy; Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, Université Laval, Quebec City, Canada; Joint International Unit on Chemical and Biomolecular Research on the Microbiome and its Impact on Metabolic Health and Nutrition (UMI-MicroMeNu), Université Laval and Institute of Biomolecular Chemistry, CNR, Pozzuoli, Italy
| | - Vincenzo Micale
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy; National Institute of Mental Health, Klecany, Czech Republic.
| |
Collapse
|
20
|
Bauman MD, Van de Water J. Translational opportunities in the prenatal immune environment: Promises and limitations of the maternal immune activation model. Neurobiol Dis 2020; 141:104864. [PMID: 32278881 DOI: 10.1016/j.nbd.2020.104864] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 03/03/2020] [Accepted: 04/02/2020] [Indexed: 12/15/2022] Open
Abstract
The prenatal environment, and in particular, the maternal-fetal immune environment, has emerged as a targeted area of research for central nervous system (CNS) diseases with neurodevelopmental origins. Converging evidence from both clinical and preclinical research indicates that changes in the maternal gestational immune environment can alter fetal brain development and increase the risk for certain neurodevelopmental disorders. Here we focus on the translational potential of one prenatal animal model - the maternal immune activation (MIA) model. This model stems from the observation that a subset of pregnant women who are exposed to infection during pregnancy have an increased risk of giving birth to a child who will later be diagnosed with a neurodevelopmental disorder, such as autism spectrum disorder (ASD) or schizophrenia (SZ). The preclinical MIA model provides a system in which to explore causal relationships, identify underlying neurobiological mechanisms, and, ultimately, develop novel therapeutic interventions and preventative strategies. In this review, we will highlight converging evidence from clinical and preclinical research that links changes in the maternal-fetal immune environment with lasting changes in offspring brain and behavioral development. We will then explore the promises and limitations of the MIA model as a translational tool to develop novel therapeutic interventions. As the translational potential of the MIA model has been the focus of several excellent review articles, here we will focus on what is perhaps the least well developed area of MIA model research - novel preventative strategies and therapeutic interventions.
Collapse
Affiliation(s)
- Melissa D Bauman
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, United States of America; California National Primate Research Center, University of California, Davis, United States of America; The MIND Institute, University of California, Davis, United States of America.
| | - Judy Van de Water
- The MIND Institute, University of California, Davis, United States of America; Rheumatology/Allergy and Clinical Immunology, University of California, Davis, United States of America
| |
Collapse
|
21
|
Gogos A, Sbisa A, Witkamp D, van den Buuse M. Sex differences in the effect of maternal immune activation on cognitive and psychosis-like behaviour in Long Evans rats. Eur J Neurosci 2020; 52:2614-2626. [PMID: 31901174 DOI: 10.1111/ejn.14671] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Revised: 12/16/2019] [Accepted: 12/23/2019] [Indexed: 02/06/2023]
Abstract
Maternal immune activation during pregnancy is associated with increased risk of development of schizophrenia in later life. There are sex differences in schizophrenia, particularly in terms of age of onset, course of illness and severity of symptoms. However, there is limited and inconsistent literature on sex differences in the effects of maternal immune activation on behaviour with relevance to schizophrenia. The aim of this study was therefore to investigate sex differences in the effects of maternal immune activation by treating Long Evans rats with poly(I:C) on gestational day 15. We compared adult male and female offspring on spatial working memory in the touchscreen trial-unique nonmatching-to-location task, pairwise discrimination and reversal learning, as well as on prepulse inhibition and psychotropic drug-induced locomotor hyperactivity. Male, but not female poly(I:C) offspring displayed a deficit in spatial working memory, particularly at the longer delay. Neither pairwise discrimination nor reversal learning showed an effect of poly(I:C), but female controls outperformed male controls in the reversal learning task. Significant reduction of prepulse inhibition and enhancement of acute methamphetamine-induced locomotor hyperactivity was found similarly in male and female poly(I:C) offspring. These results show that maternal immune activation induces a range of behavioural effects in the offspring, with sex specificity in the effects of maternal immune activation on some aspects of cognition, but not psychosis-like behaviour.
Collapse
Affiliation(s)
- Andrea Gogos
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Vic., Australia
| | - Alyssa Sbisa
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Vic., Australia.,School of Psychology and Public Health, La Trobe University, Melbourne, Vic., Australia
| | - Diede Witkamp
- School of Psychology and Public Health, La Trobe University, Melbourne, Vic., Australia
| | - Maarten van den Buuse
- School of Psychology and Public Health, La Trobe University, Melbourne, Vic., Australia.,Department of Pharmacology, University of Melbourne, Melbourne, Vic., Australia.,The College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, QLD, Australia
| |
Collapse
|
22
|
Di Biase MA, Katabi G, Piontkewitz Y, Cetin-Karayumak S, Weiner I, Pasternak O. Increased extracellular free-water in adult male rats following in utero exposure to maternal immune activation. Brain Behav Immun 2020; 83:283-287. [PMID: 31521731 DOI: 10.1016/j.bbi.2019.09.010] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 08/27/2019] [Accepted: 09/12/2019] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND In previous work, we applied novel in vivo imaging methods to reveal that white matter pathology in patients with first-episode psychosis (FEP) is mainly characterized by excessive extracellular free-water, and to a lesser extent by cellular processes, such as demyelination. Here, we apply a back-translational approach to evaluate whether or not a rodent model of maternal immune activation (MIA) induces patterns of white matter pathology that we observed in patients with FEP. To this end, we examined free-water and tissue-specific white matter alterations in rats born to mothers exposed to the viral mimic polyriboinosinic-polyribocytidylic acid (Poly-I:C) in pregnancy, which is widely used to produce alterations relevant to schizophrenia and is characterized by a robust neuroinflammatory response. METHOD Pregnant dams were injected on gestational day 15 with the viral mimic Poly-I:C (4 mg/kg) or saline. Diffusion-weighted magnetic resonance images were acquired from 17 male offspring (9 Poly-I:C and 8 saline) on postnatal day 90, after the emergence of brain structural and behavioral abnormalities. The free-water fraction (FW) and tissue-specific fractional anisotropy (FAT), as well as conventional fractional anisotropy (FA) were computed across voxels traversing a white matter skeleton. Voxel-wise and whole-brain averaged white matter were tested for significant microstructural alterations in immune-challenged, relative to saline-exposed offspring. RESULTS Compared to saline-exposed offspring, those exposed to maternal Poly-I:C displayed increased extracellular FW averaged across voxels comprising a white matter skeleton (t(15) = 2.74; p = 0.01). Voxel-wise analysis ascribed these changes to white matter within the corpus callosum, external capsule and the striatum. In contrast, no significant between-group differences emerged for FAT or for conventional FA, measured across average and voxel-wise white matter. CONCLUSION We identified excess FW across frontal white matter fibers of rats exposed to prenatal immune activation, analogous to our "bedside" observation in FEP patients. Findings from this initial experiment promote use of the MIA model to examine pathological pathways underlying FW alterations observed in patients with schizophrenia. Establishing these mechanisms has important implications for clinical studies, as free-water imaging reflects a feasible biomarker that has so far yielded consistent findings in the early stages of schizophrenia.
Collapse
Affiliation(s)
- Maria A Di Biase
- Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Gili Katabi
- School of Psychological Sciences and Sagol School of Neuroscience, Tel-Aviv University, Tel-Aviv, Israel
| | - Yael Piontkewitz
- Straus Center for Computational Neuroimaging, George S. Wise Faculty of Life Sciences, Tel-Aviv University, Israel
| | - Suheyla Cetin-Karayumak
- Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ina Weiner
- School of Psychological Sciences and Sagol School of Neuroscience, Tel-Aviv University, Tel-Aviv, Israel
| | - Ofer Pasternak
- Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
23
|
Duchatel RJ, Harms LR, Meehan CL, Michie PT, Bigland MJ, Smith DW, Jobling P, Hodgson DM, Tooney PA. Reduced cortical somatostatin gene expression in a rat model of maternal immune activation. Psychiatry Res 2019; 282:112621. [PMID: 31648143 DOI: 10.1016/j.psychres.2019.112621] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 10/14/2019] [Accepted: 10/14/2019] [Indexed: 12/28/2022]
Abstract
Alterations in GABAergic interneurons and glutamic acid decarboxylase (GAD) are observed in the brains of people with schizophrenia. Studies also show increased density of interstitial white matter neurons (IWMN), including those containing GAD and somatostatin (SST) in the brain in schizophrenia. Maternal immune activation can be modelled in rodents to investigate the relationship between prenatal exposure to infections and increased risk of developing schizophrenia. We reported that maternal immune activation induced an increase in density of somatostatin-positive IWMN in the adult rat offspring. Here we hypothesised that maternal immune activation induced in pregnant rats by polyinosinic:polycytidylic acid would alter SST and GAD gene expression as well as increase the density of GAD-positive IWMNs in the adult offspring. SST gene expression was significantly reduced in the cingulate cortex of adult offspring exposed to late gestation maternal immune activation. There was no change in cortical GAD gene expression nor GAD-positive IWMN density in adults rats exposed to maternal immune activation at either early or late gestation. This suggests that our model of maternal immune activation induced by prenatal exposure of rats to polyinosinic:polycytidylic acid during late gestation is able to recapitulate changes in SST but not other GABAergic neuropathologies observed in schizophrenia.
Collapse
Affiliation(s)
- Ryan J Duchatel
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, 2308 Australia; Priority Centre for Brain and Mental Health Research, University of Newcastle, Callaghan, NSW, 2308 Australia; Hunter Medical Research Institute, New Lambton Heights, NSW, 2305 Australia.
| | - Lauren R Harms
- School of Psychology, Faculty of Science, University of Newcastle, Callaghan, NSW, 2308 Australia; Priority Centre for Brain and Mental Health Research, University of Newcastle, Callaghan, NSW, 2308 Australia; Hunter Medical Research Institute, New Lambton Heights, NSW, 2305 Australia.
| | - Crystal L Meehan
- School of Psychology, Faculty of Science, University of Newcastle, Callaghan, NSW, 2308 Australia; Priority Centre for Brain and Mental Health Research, University of Newcastle, Callaghan, NSW, 2308 Australia; Hunter Medical Research Institute, New Lambton Heights, NSW, 2305 Australia.
| | - Patricia T Michie
- School of Psychology, Faculty of Science, University of Newcastle, Callaghan, NSW, 2308 Australia; Priority Centre for Brain and Mental Health Research, University of Newcastle, Callaghan, NSW, 2308 Australia; Hunter Medical Research Institute, New Lambton Heights, NSW, 2305 Australia.
| | - Mark J Bigland
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, 2308 Australia; Priority Centre for Brain and Mental Health Research, University of Newcastle, Callaghan, NSW, 2308 Australia; Hunter Medical Research Institute, New Lambton Heights, NSW, 2305 Australia.
| | - Doug W Smith
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, 2308 Australia; Priority Centre for Brain and Mental Health Research, University of Newcastle, Callaghan, NSW, 2308 Australia; Hunter Medical Research Institute, New Lambton Heights, NSW, 2305 Australia.
| | - Phillip Jobling
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, 2308 Australia; Priority Centre for Brain and Mental Health Research, University of Newcastle, Callaghan, NSW, 2308 Australia; Hunter Medical Research Institute, New Lambton Heights, NSW, 2305 Australia.
| | - Deborah M Hodgson
- School of Psychology, Faculty of Science, University of Newcastle, Callaghan, NSW, 2308 Australia; Priority Centre for Brain and Mental Health Research, University of Newcastle, Callaghan, NSW, 2308 Australia; Hunter Medical Research Institute, New Lambton Heights, NSW, 2305 Australia.
| | - Paul A Tooney
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, 2308 Australia; Priority Centre for Brain and Mental Health Research, University of Newcastle, Callaghan, NSW, 2308 Australia; Hunter Medical Research Institute, New Lambton Heights, NSW, 2305 Australia.
| |
Collapse
|
24
|
The role of maternal immune activation in altering the neurodevelopmental trajectories of offspring: A translational review of neuroimaging studies with implications for autism spectrum disorder and schizophrenia. Neurosci Biobehav Rev 2019; 104:141-157. [DOI: 10.1016/j.neubiorev.2019.06.020] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 05/24/2019] [Accepted: 06/13/2019] [Indexed: 02/01/2023]
|
25
|
Casquero-Veiga M, García-García D, MacDowell KS, Pérez-Caballero L, Torres-Sánchez S, Fraguas D, Berrocoso E, Leza JC, Arango C, Desco M, Soto-Montenegro ML. Risperidone administered during adolescence induced metabolic, anatomical and inflammatory/oxidative changes in adult brain: A PET and MRI study in the maternal immune stimulation animal model. Eur Neuropsychopharmacol 2019; 29:880-896. [PMID: 31229322 DOI: 10.1016/j.euroneuro.2019.05.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 04/30/2019] [Accepted: 05/29/2019] [Indexed: 12/22/2022]
Abstract
Inflammation and oxidative stress (IOS) are considered key pathophysiological elements in the development of mental disorders. Recent studies demonstrated that the antipsychotic risperidone elicits an antiinflammatory effect in the brain. We administered risperidone for 2-weeks at adolescence to assess its role in preventing brain-related IOS changes in the maternal immune stimulation (MIS) model at adulthood. We also investigated the development of volumetric and neurotrophic abnormalities in areas related to the HPA-axis. Poly I:C (MIS) or saline (Sal) were injected into pregnant Wistar rats on GD15. Male offspring received risperidone or vehicle daily from PND35-PND49. We studied 4 groups (8-15 animals/group): Sal-vehicle, MIS-vehicle, Sal-risperidone and MIS-risperidone. [18F]FDG-PET and MRI studies were performed at adulthood and analyzed using SPM12 software. IOS and neurotrophic markers were measured using WB and ELISA assays in brain tissue. Risperidone elicited a protective function of schizophrenia-related IOS deficits. In particular, risperidone elicited the following effects: reduced volume in the ventricles and the pituitary gland; reduced glucose metabolism in the cerebellum, periaqueductal gray matter, and parietal cortex; higher FDG uptake in the cingulate cortex, hippocampus, thalamus, and brainstem; reduced NFκB activity and iNOS expression; and increased enzymatic activity of CAT and SOD in some brain areas. Our study suggests that some schizophrenia-related IOS changes can be prevented in the MIS model. It also stresses the need to search for novel strategies based on anti-inflammatory compounds in risk populations at early stages in order to alter the course of the disease.
Collapse
Affiliation(s)
- Marta Casquero-Veiga
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain; CIBER de Salud Mental (CIBERSAM), Madrid, Spain
| | - David García-García
- Department of Bioengineering and Aerospace Engineering, Universidad Carlos III de Madrid, Leganés, Spain; Facultad de Ciencia y Tecnología, Universidad Isabel I, Burgos, Spain
| | - Karina S MacDowell
- CIBER de Salud Mental (CIBERSAM), Madrid, Spain; Department of Pharmacology & Toxicology, School of Medicine, Universidad Complutense (UCM), IIS Imas12, IUIN, Madrid, Spain
| | - Laura Pérez-Caballero
- CIBER de Salud Mental (CIBERSAM), Madrid, Spain; Neuropsychopharmacology & 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
| | - Sonia Torres-Sánchez
- CIBER de Salud Mental (CIBERSAM), Madrid, Spain; Instituto de Investigación e Innovación en Ciencias Biomédicas de Cádiz, INiBICA, Hospital Universitario Puerta del Mar, Cádiz, Spain; Neuropsychopharmacology & Psychobiology Research Group, Universidad de Cádiz, Cádiz, Spain
| | - David Fraguas
- CIBER de Salud Mental (CIBERSAM), Madrid, Spain; Department of Child and Adolescent Psychiatry, Hospital General Universitario Gregorio Marañón, School of Medicine, Universidad Complutense (UCM), Madrid, Spain
| | - Esther Berrocoso
- CIBER de Salud Mental (CIBERSAM), Madrid, Spain; Neuropsychopharmacology & 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 & Toxicology, School of Medicine, Universidad Complutense (UCM), IIS Imas12, IUIN, Madrid, Spain
| | - Celso Arango
- CIBER de Salud Mental (CIBERSAM), Madrid, Spain; Department of Child and Adolescent Psychiatry, Hospital General Universitario Gregorio Marañón, School of Medicine, Universidad Complutense (UCM), Madrid, Spain
| | - Manuel Desco
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain; Department of Bioengineering and Aerospace Engineering, Universidad Carlos III de Madrid, Leganés, Spain; CIBER de Salud Mental (CIBERSAM), Madrid, Spain; Centro Nacional de Investigaciones Cardiovasculares, CNIC, Madrid, Spain.
| | - María Luisa Soto-Montenegro
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain; CIBER de Salud Mental (CIBERSAM), Madrid, Spain
| |
Collapse
|
26
|
Murray KN, Edye ME, Manca M, Vernon AC, Oladipo JM, Fasolino V, Harte MK, Mason V, Grayson B, McHugh PC, Knuesel I, Prinssen EP, Hager R, Neill JC. Evolution of a maternal immune activation (mIA) model in rats: Early developmental effects. Brain Behav Immun 2019; 75:48-59. [PMID: 30218784 DOI: 10.1016/j.bbi.2018.09.005] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 08/05/2018] [Accepted: 09/04/2018] [Indexed: 01/17/2023] Open
Abstract
Maternal immune activation (mIA) in rodents is rapidly emerging as a key model for neurodevelopmental disorders such as autism spectrum disorder (ASD) and schizophrenia. Here, we optimise a mIA model in rats, aiming to address certain limitations of current work in this field. Specifically, the lack of clear evidence for methodology chosen, identification of successful induction of mIA in the dams and investigation of male offspring only. We focus on gestational and early juvenile changes in offspring following mIA, as detailed information on these critical early developmental time points is sparse. Following strain (Wistar, Lister Hooded, Sprague Dawley) comparison and selection, and polyriboinosinic-polyribocytidylic acid (poly I:C) dose selection (2.5-15 mg/kg single or once daily for 5 days), mIA was induced in pregnant Wistar rats with 10 mg/kg poly I:C i.p. on gestational day (GD) 15. Early morphometric analysis was conducted in male and female offspring at GD21 and postnatal day (PD) 21, eight dams for each treatment at each time point were used, 32 in total. Subsequent microglia analysis was conducted at PD21 in a small group of offspring. Poly I:C at 10 mg/kg i.p. induced a robust, but variable, plasma IL-6 response 3 h post-injection and reduced body weight at 6 h and 24 h post-injection in two separate cohorts of Wistar rats at GD15. Plasma IL-6 was not elevated at PD21 in offspring or dams. Poly I:C-induced mIA did not affect litter numbers, but resulted in PD21 pup, and GD21 placenta growth restriction. Poly I:C significantly increased microglial activation at PD21 in male hippocampi. We have identified 10 mg/kg poly I:C i.p on GD15 as a robust experimental approach for inducing mIA in Wistar rats and used this to identify early neurodevelopmental changes. This work provides a framework to study the developmental trajectory of disease-relevant, sex-specific phenotypic changes in rats.
Collapse
Affiliation(s)
- Katie N Murray
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester M13 9PT, United Kingdom
| | - Michelle E Edye
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester M13 9PT, United Kingdom
| | - Maurizio Manca
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester M13 9PT, United Kingdom
| | - Anthony C Vernon
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, 5 Cutcombe Road, London SE5 9RT, United Kingdom; King's College London, MRC Centre for Neurodevelopmental Disorders, New Hunt's House, Guy's Hospital Campus, London SE1 1UL, United Kingdom
| | - Joanna M Oladipo
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester M13 9PT, United Kingdom
| | - Victoria Fasolino
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester M13 9PT, United Kingdom
| | - Michael K Harte
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester M13 9PT, United Kingdom
| | - Varsha Mason
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester M13 9PT, United Kingdom
| | - Ben Grayson
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester M13 9PT, United Kingdom
| | - Patrick C McHugh
- Centre for Biomarker Research and Department of Pharmacy, School of Applied Sciences, University of Huddersfield, HD1 3DH, United Kingdom
| | - Irene Knuesel
- Roche Innovation Center Basel, 124 Grenzacherstrasse, Basel, CH 4070, Switzerland
| | - Eric P Prinssen
- Roche Innovation Center Basel, 124 Grenzacherstrasse, Basel, CH 4070, Switzerland
| | - Reinmar Hager
- Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester M13 9PT, United Kingdom.
| | - Joanna C Neill
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester M13 9PT, United Kingdom.
| |
Collapse
|
27
|
Gray A, Tattoli R, Dunn A, Hodgson D, Michie P, Harms L. Maternal immune activation in mid-late gestation alters amphetamine sensitivity and object recognition, but not other schizophrenia-related behaviours in adult rats. Behav Brain Res 2019; 356:358-364. [DOI: 10.1016/j.bbr.2018.08.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 08/17/2018] [Accepted: 08/17/2018] [Indexed: 12/17/2022]
|
28
|
Peres FF, Diana MC, Levin R, Suiama MA, Almeida V, Vendramini AM, Santos CM, Zuardi AW, Hallak JEC, Crippa JA, Abílio VC. Cannabidiol Administered During Peri-Adolescence Prevents Behavioral Abnormalities in an Animal Model of Schizophrenia. Front Pharmacol 2018; 9:901. [PMID: 30186164 PMCID: PMC6113576 DOI: 10.3389/fphar.2018.00901] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 07/23/2018] [Indexed: 01/08/2023] Open
Abstract
Schizophrenia is considered a debilitating neurodevelopmental psychiatric disorder and its pharmacotherapy remains problematic without recent major advances. The development of interventions able to prevent the emergence of schizophrenia would therefore represent an enormous progress. Here, we investigated whether treatment with cannabidiol (CBD - a compound of Cannabis sativa that presents an antipsychotic profile in animals and humans) during peri-adolescence would prevent schizophrenia-like behavioral abnormalities in an animal model of schizophrenia: the spontaneously hypertensive rat (SHR) strain. Wistar rats and SHRs were treated with vehicle or CBD from 30 to 60 post-natal days. In experiment 1, schizophrenia-like behaviors (locomotor activity, social interaction, prepulse inhibition of startle and contextual fear conditioning) were assessed on post-natal day 90. Side effects commonly associated with antipsychotic treatment were also evaluated: body weight gain and catalepsy throughout the treatment, and oral dyskinesia 48 h after treatment interruption and on post-natal day 90. In experiment 2, serum levels of triglycerides and glycemia were assessed on post-natal day 61. In experiment 3, levels of BDNF, monoamines, and their metabolites were evaluated on post-natal days 61 and 90 in the prefrontal cortex and striatum. Treatment with CBD prevented the emergence of SHRs' hyperlocomotor activity (a model for the positive symptoms of schizophrenia) and deficits in prepulse inhibition of startle and contextual fear conditioning (cognitive impairments). CBD did not induce any of the potential motor or metabolic side effects evaluated. Treatment with CBD increased the prefrontal cortex 5-HIAA/serotonin ratio and the levels of 5-HIAA on post-natal days 61 and 90, respectively. Our data provide pre-clinical evidence for a safe and beneficial effect of peripubertal and treatment with CBD on preventing positive and cognitive symptoms of schizophrenia, and suggest the involvement of the serotoninergic system on this effect.
Collapse
Affiliation(s)
- Fernanda F Peres
- Department of Pharmacology, Federal University of São Paulo, São Paulo, Brazil.,National Institute for Translational Medicine, National Council for Scientific and Technological Development, Ribeirão Preto, Brazil.,Interdisciplinary Laboratory of Clinical Neurosciences, Department of Psychiatry, Federal University of São Paulo, São Paulo, Brazil
| | - Mariana C Diana
- Department of Pharmacology, Federal University of São Paulo, São Paulo, Brazil.,Interdisciplinary Laboratory of Clinical Neurosciences, Department of Psychiatry, Federal University of São Paulo, São Paulo, Brazil
| | - Raquel Levin
- Department of Pharmacology, Federal University of São Paulo, São Paulo, Brazil.,Interdisciplinary Laboratory of Clinical Neurosciences, Department of Psychiatry, Federal University of São Paulo, São Paulo, Brazil
| | - Mayra A Suiama
- Department of Pharmacology, Federal University of São Paulo, São Paulo, Brazil.,Interdisciplinary Laboratory of Clinical Neurosciences, Department of Psychiatry, Federal University of São Paulo, São Paulo, Brazil
| | - Valéria Almeida
- Department of Pharmacology, Federal University of São Paulo, São Paulo, Brazil.,Interdisciplinary Laboratory of Clinical Neurosciences, Department of Psychiatry, Federal University of São Paulo, São Paulo, Brazil
| | - Ana M Vendramini
- Department of Pharmacology, Federal University of São Paulo, São Paulo, Brazil.,Interdisciplinary Laboratory of Clinical Neurosciences, Department of Psychiatry, Federal University of São Paulo, São Paulo, Brazil
| | - Camila M Santos
- Department of Pharmacology, Federal University of São Paulo, São Paulo, Brazil.,Interdisciplinary Laboratory of Clinical Neurosciences, Department of Psychiatry, Federal University of São Paulo, São Paulo, Brazil
| | - Antônio W Zuardi
- National Institute for Translational Medicine, National Council for Scientific and Technological Development, Ribeirão Preto, Brazil.,Department of Neuroscience and Behavior, University of São Paulo, Ribeirão Preto, Brazil
| | - Jaime E C Hallak
- National Institute for Translational Medicine, National Council for Scientific and Technological Development, Ribeirão Preto, Brazil.,Department of Neuroscience and Behavior, University of São Paulo, Ribeirão Preto, Brazil
| | - José A Crippa
- National Institute for Translational Medicine, National Council for Scientific and Technological Development, Ribeirão Preto, Brazil.,Department of Neuroscience and Behavior, University of São Paulo, Ribeirão Preto, Brazil
| | - Vanessa C Abílio
- Department of Pharmacology, Federal University of São Paulo, São Paulo, Brazil.,National Institute for Translational Medicine, National Council for Scientific and Technological Development, Ribeirão Preto, Brazil.,Department of Neuroscience and Behavior, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
29
|
Drazanova E, Ruda-Kucerova J, Kratka L, Horska K, Demlova R, Starcuk Z, Kasparek T. Poly(I:C) model of schizophrenia in rats induces sex-dependent functional brain changes detected by MRI that are not reversed by aripiprazole treatment. Brain Res Bull 2017; 137:146-155. [PMID: 29155259 DOI: 10.1016/j.brainresbull.2017.11.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 11/06/2017] [Accepted: 11/08/2017] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND PURPOSE One of the hallmarks of schizophrenia is altered brain structure, potentially due to antipsychotic treatment, the disorder itself or both. It was proposed that functional changes may precede the structural ones. In order to understand and potentially prevent this unwanted process, brain function assessment should be validated as a diagnostic tool. METHODS We used Arterial Spin Labelling MRI technique for the evaluation of brain perfusion in several brain regions in a neurodevelopmental poly(I:C) model of schizophrenia (8mg/kg on a gestational day 15) in rats taking into account sex-dependent effects and chronic treatment with aripiprazole (30days), an atypical antipsychotic acting as a partial agonist on dopaminergic receptors. RESULTS We found the sex of the animal to have a highly significant effect in all regions of interest, with females showing lower blood perfusion than males. However, both males and females treated prenatally with poly(I:C) showed enlargement of the lateral ventricles. Furthermore, we detected increased perfusion in the circle of Willis, hippocampus, and sensorimotor cortex, which was not influenced by chronic atypical antipsychotic aripiprazole treatment in male poly(I:C) rats. CONCLUSION We hypothesize that perfusion alterations may be caused by the hyperdopaminergic activity in the poly(I:C) model, and the absence of aripiprazole effect on perfusion in brain regions related to schizophrenia may be due to its partial agonistic mechanism.
Collapse
Affiliation(s)
- Eva Drazanova
- Department of Pharmacology, Faculty of Medicine, Masaryk University, Brno, Czech Republic; Institute of Scientific Instruments, The Czech Academy of Sciences, Brno, Czech Republic
| | - Jana Ruda-Kucerova
- Department of Pharmacology, Faculty of Medicine, Masaryk University, Brno, Czech Republic.
| | - Lucie Kratka
- Institute of Scientific Instruments, The Czech Academy of Sciences, Brno, Czech Republic
| | - Katerina Horska
- Department of Human Pharmacology and Toxicology, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences, Brno, Czech Republic
| | - Regina Demlova
- Department of Pharmacology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Zenon Starcuk
- Institute of Scientific Instruments, The Czech Academy of Sciences, Brno, Czech Republic
| | - Tomas Kasparek
- Department of Psychiatry, University Hospital and Masaryk University, Brno, Czech Republic
| |
Collapse
|
30
|
Hadar R, Dong L, Del-Valle-Anton L, Guneykaya D, Voget M, Edemann-Callesen H, Schweibold R, Djodari-Irani A, Goetz T, Ewing S, Kettenmann H, Wolf SA, Winter C. Deep brain stimulation during early adolescence prevents microglial alterations in a model of maternal immune activation. Brain Behav Immun 2017; 63:71-80. [PMID: 27939248 DOI: 10.1016/j.bbi.2016.12.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 11/23/2016] [Accepted: 12/05/2016] [Indexed: 11/25/2022] Open
Abstract
In recent years schizophrenia has been recognized as a neurodevelopmental disorder likely involving a perinatal insult progressively affecting brain development. The poly I:C maternal immune activation (MIA) rodent model is considered as a neurodevelopmental model of schizophrenia. Using this model we and others demonstrated the association between neuroinflammation in the form of altered microglia and a schizophrenia-like endophenotype. Therapeutic intervention using the anti-inflammatory drug minocycline affected altered microglia activation and was successful in the adult offspring. However, less is known about the effect of preventive therapeutic strategies on microglia properties. Previously we found that deep brain stimulation of the medial prefrontal cortex applied pre-symptomatically to adolescence MIA rats prevented the manifestation of behavioral and structural deficits in adult rats. We here studied the effects of deep brain stimulation during adolescence on microglia properties in adulthood. We found that in the hippocampus and nucleus accumbens, but not in the medial prefrontal cortex, microglial density and soma size were increased in MIA rats. Pro-inflammatory cytokine mRNA was unchanged in all brain areas before and after implantation and stimulation. Stimulation of either the medial prefrontal cortex or the nucleus accumbens normalized microglia density and soma size in main projection areas including the hippocampus and in the area around the electrode implantation. We conclude that in parallel to an alleviation of the symptoms in the rat MIA model, deep brain stimulation has the potential to prevent the neuroinflammatory component in this disease.
Collapse
Affiliation(s)
- Ravit Hadar
- Department of Psychiatry and Psychotherapy, Medical Faculty Carl Gustav Carus, Technische Universitaet Dresden, Germany
| | - Le Dong
- Cellular Neuroscience, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Lucia Del-Valle-Anton
- Cellular Neuroscience, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Dilansu Guneykaya
- Cellular Neuroscience, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Mareike Voget
- Department of Psychiatry and Psychotherapy, Medical Faculty Carl Gustav Carus, Technische Universitaet Dresden, Germany; International Graduate Program Medical Neurosciences, Charité - Universitaetsmedizin Berlin, Germany
| | - Henriette Edemann-Callesen
- Department of Psychiatry and Psychotherapy, Medical Faculty Carl Gustav Carus, Technische Universitaet Dresden, Germany; International Graduate Program Medical Neurosciences, Charité - Universitaetsmedizin Berlin, Germany
| | - Regina Schweibold
- Cellular Neuroscience, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Anais Djodari-Irani
- Cellular Neuroscience, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Thomas Goetz
- Department of Psychiatry and Psychotherapy, Medical Faculty Carl Gustav Carus, Technische Universitaet Dresden, Germany
| | - Samuel Ewing
- Department of Psychiatry and Psychotherapy, Medical Faculty Carl Gustav Carus, Technische Universitaet Dresden, Germany
| | - Helmut Kettenmann
- Cellular Neuroscience, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Susanne A Wolf
- Cellular Neuroscience, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.
| | - Christine Winter
- Department of Psychiatry and Psychotherapy, Medical Faculty Carl Gustav Carus, Technische Universitaet Dresden, Germany
| |
Collapse
|
31
|
Crum WR, Sawiak SJ, Chege W, Cooper JD, Williams SC, Vernon AC. Evolution of structural abnormalities in the rat brain following in utero exposure to maternal immune activation: A longitudinal in vivo MRI study. Brain Behav Immun 2017; 63:50-59. [PMID: 27940258 PMCID: PMC5441572 DOI: 10.1016/j.bbi.2016.12.008] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 11/07/2016] [Accepted: 12/07/2016] [Indexed: 02/08/2023] Open
Abstract
Genetic and environmental risk factors for psychiatric disorders are suggested to disrupt the trajectory of brain maturation during adolescence, leading to the development of psychopathology in adulthood. Rodent models are powerful tools to dissect the specific effects of such risk factors on brain maturational profiles, particularly when combined with Magnetic Resonance Imaging (MRI; clinically comparable technology). We therefore investigated the effect of maternal immune activation (MIA), an epidemiological risk factor for adult-onset psychiatric disorders, on rat brain maturation using atlas and tensor-based morphometry analysis of longitudinal in vivo MR images. Exposure to MIA resulted in decreases in the volume of several cortical regions, the hippocampus, amygdala, striatum, nucleus accumbens and unexpectedly, the lateral ventricles, relative to controls. In contrast, the volumes of the thalamus, ventral mesencephalon, brain stem and major white matter tracts were larger, relative to controls. These volumetric changes were maximal between post-natal day 50 and 100 with no differences between the groups thereafter. These data are consistent with and extend prior studies of brain structure in MIA-exposed rodents. Apart from the ventricular findings, these data have robust face validity to clinical imaging findings reported in studies of individuals at high clinical risk for a psychiatric disorder. Further work is now required to address the relationship of these MRI changes to behavioral dysfunction and to establish thier cellular correlates.
Collapse
Affiliation(s)
- William R. Crum
- Department of Neuroimaging Institute of Psychiatry, Psychology and Neuroscience, King’s College London, De Crespigny Park, London SE5 8AF, UK
| | - Stephen J. Sawiak
- Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Addenbrooke’s Hospital, Hills Road, Cambridge, UK
| | - Winfred Chege
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, De Crespigny Park, London SE5 8AF, UK
| | - Jonathan D. Cooper
- 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, UK
| | - Steven C.R. Williams
- Department of Neuroimaging Institute of Psychiatry, Psychology and Neuroscience, King’s College London, De Crespigny Park, London SE5 8AF, UK,MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK
| | - Anthony C. Vernon
- 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, UK,MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK,Corresponding author at: 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, UK.Department of Basic and Clinical NeuroscienceInstitute of PsychiatryPsychology and NeuroscienceKing’s College LondonMaurice Wohl Clinical Neuroscience Institute5 Cutcombe RoadLondonSE5 9RTUK
| |
Collapse
|
32
|
Positive modulation of α5 GABA A receptors in preadolescence prevents reduced locomotor response to amphetamine in adult female but not male rats prenatally exposed to lipopolysaccharide. Int J Dev Neurosci 2017; 61:31-39. [PMID: 28610974 DOI: 10.1016/j.ijdevneu.2017.06.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 06/02/2017] [Accepted: 06/03/2017] [Indexed: 12/16/2022] Open
Abstract
We previously demonstrated that lipopolysaccharide (LPS) administered intraperitoneally (i.p.) to pregnant Wistar rat dams, at embryonic days 15 and 16 (E15/16), induced a decrease of baseline locomotor activity and diminished reactivity to amphetamine in adult female offspring. In the present study we aimed to assess the duration of LPS-induced maternal immune activation (MIA) and investigate possible changes in levels of main neurotransmitters in fetal brain during MIA. We hypothesized that the observed behavioral changes may be linked with MIA-induced disturbance of prenatal GABAergic system development, especially with α5 GABAA receptors (α5GABAARs), expression of which takes place between E14 and E17. Thereafter, we set to investigate if later potentiation of α5GABAARs in offspring's preadolescence (from postnatal day 22-28) could prevent the deficit in locomotor reactivity to amphetamine observed in adulthood, at postnatal day P60. The elevation of IL-6 in amniotic fluid 6h after LPS treatment (100μg/kg, i.p.) at E15 was concurrent with a significant increase of GABA and decrease of glutamate concentration in fetal brain. Moreover, repeated administration of MP-III-022, a selective positive allosteric modulator of α5GABAARs, at a dose (2mg/kg daily, i.p.) derived from a separate pharmacokinetic study, prevented the LPS-induced decrease in locomotor reactivity to amphetamine (0.5mg/kg, i.p.) in adult females. These results were not mirrored in the parallel set of experiments with male offspring from LPS-treated rats. The results suggest that pharmacological potentiation of α5GABAARs activity in preadolescence may ameliorate at least some of adverse consequences of exposure to MIA in utero.
Collapse
|
33
|
Improved Social Interaction, Recognition and Working Memory with Cannabidiol Treatment in a Prenatal Infection (poly I:C) Rat Model. Neuropsychopharmacology 2017; 42:1447-1457. [PMID: 28230072 PMCID: PMC5436124 DOI: 10.1038/npp.2017.40] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 02/10/2017] [Accepted: 02/19/2017] [Indexed: 12/16/2022]
Abstract
Neuropsychiatric disorders such as schizophrenia are associated with cognitive impairment, including learning, memory and attention deficits. Antipsychotic drugs are limited in their efficacy to improve cognition; therefore, new therapeutic agents are required. Cannabidiol (CBD), the non-intoxicating component of cannabis, has anti-inflammatory, neuroprotective and antipsychotic-like properties; however, its ability to improve the cognitive deficits of schizophrenia remains unclear. Using a prenatal infection model, we examined the effect of chronic CBD treatment on cognition and social interaction. Time-mated pregnant Sprague-Dawley rats (n=16) were administered polyinosinic-polycytidilic acid (poly I:C) (POLY; 4 mg/kg) or saline (CONT) at gestation day 15. Male offspring (PN56) were injected twice daily with 10 mg/kg CBD (CONT+CBD, POLY+CBD; n=12 per group) or vehicle (VEH; CONT+VEH, POLY+VEH; n=12 per group) for 3 weeks. Body weight, food and water intake was measured weekly. The Novel Object Recognition and rewarded T-maze alternation tests assessed recognition and working memory, respectively, and the social interaction test assessed sociability. POLY+VEH offspring exhibited impaired recognition and working memory, and reduced social interaction compared to CONT+VEH offspring (p<0.01). CBD treatment significantly improved recognition, working memory and social interaction deficits in the poly I:C model (p<0.01 vs POLY+VEH), did not affect total body weight gain, food or water intake, and had no effect in control animals (all p>0.05). In conclusion, chronic CBD administration can attenuate the social interaction and cognitive deficits induced by prenatal poly I:C infection. These novel findings present interesting implications for potential use of CBD in treating the cognitive deficits and social withdrawal of schizophrenia.
Collapse
|
34
|
Early neuromodulation prevents the development of brain and behavioral abnormalities in a rodent model of schizophrenia. Mol Psychiatry 2017; 23:943-951. [PMID: 28373685 PMCID: PMC5552352 DOI: 10.1038/mp.2017.52] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 02/07/2017] [Accepted: 02/10/2017] [Indexed: 12/18/2022]
Abstract
The notion that schizophrenia is a neurodevelopmental disorder in which neuropathologies evolve gradually over the developmental course indicates a potential therapeutic window during which pathophysiological processes may be modified to halt disease progression or reduce its severity. Here we used a neurodevelopmental maternal immune stimulation (MIS) rat model of schizophrenia to test whether early targeted modulatory intervention would affect schizophrenia's neurodevelopmental course. We applied deep brain stimulation (DBS) or sham stimulation to the medial prefrontal cortex (mPFC) of adolescent MIS rats and respective controls, and investigated its behavioral, biochemical, brain-structural and -metabolic effects in adulthood. We found that mPFC-DBS successfully prevented the emergence of deficits in sensorimotor gating, attentional selectivity and executive function in adulthood, as well as the enlargement of lateral ventricle volumes and mal-development of dopaminergic and serotonergic transmission. These data suggest that the mPFC may be a valuable target for effective preventive treatments. This may have significant translational value, suggesting that targeting the mPFC before the onset of psychosis via less invasive neuromodulation approaches may be a viable preventive strategy.
Collapse
|
35
|
Careaga M, Murai T, Bauman MD. Maternal Immune Activation and Autism Spectrum Disorder: From Rodents to Nonhuman and Human Primates. Biol Psychiatry 2017; 81:391-401. [PMID: 28137374 PMCID: PMC5513502 DOI: 10.1016/j.biopsych.2016.10.020] [Citation(s) in RCA: 238] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 10/20/2016] [Accepted: 10/20/2016] [Indexed: 12/18/2022]
Abstract
A subset of women who are exposed to infection during pregnancy have an increased risk of giving birth to a child who will later be diagnosed with a neurodevelopmental or neuropsychiatric disorder. Although epidemiology studies have primarily focused on the association between maternal infection and an increased risk of offspring schizophrenia, mounting evidence indicates that maternal infection may also increase the risk of autism spectrum disorder. A number of factors, including genetic susceptibility, the intensity and timing of the infection, and exposure to additional aversive postnatal events, may influence the extent to which maternal infection alters fetal brain development and which disease phenotype (autism spectrum disorder, schizophrenia, other neurodevelopmental disorders) is expressed. Preclinical animal models provide a test bed to systematically evaluate the effects of maternal infection on fetal brain development, determine the relevance to human central nervous system disorders, and to evaluate novel preventive and therapeutic strategies. Maternal immune activation models in mice, rats, and nonhuman primates suggest that the maternal immune response is the critical link between exposure to infection during pregnancy and subsequent changes in brain and behavioral development of offspring. However, differences in the type, severity, and timing of prenatal immune challenge paired with inconsistencies in behavioral phenotyping approaches have hindered the translation of preclinical results to human studies. Here we highlight the promises and limitations of the maternal immune activation model as a preclinical tool to study prenatal risk factors for autism spectrum disorder, and suggest specific changes to improve reproducibility and maximize translational potential.
Collapse
Affiliation(s)
- Milo Careaga
- UC Davis MIND Institute, University of California, Davis, California; Department of Psychiatry and Behavioral Sciences, University of California, Davis, California
| | - Takeshi Murai
- UC Davis MIND Institute, University of California, Davis, California; California National Primate Research Center, University of California, Davis, California; Biomarker Group, Drug Development Research Laboratories, Sumitomo Dainippon Pharma Co., Ltd., Osaka, Japan
| | - Melissa D Bauman
- UC Davis MIND Institute, University of California, Davis, California; Department of Psychiatry and Behavioral Sciences, University of California, Davis, California; California National Primate Research Center, University of California, Davis, California.
| |
Collapse
|
36
|
Luoni A, Richetto J, Longo L, Riva MA. Chronic lurasidone treatment normalizes GABAergic marker alterations in the dorsal hippocampus of mice exposed to prenatal immune activation. Eur Neuropsychopharmacol 2017; 27:170-179. [PMID: 27939135 DOI: 10.1016/j.euroneuro.2016.12.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 11/21/2016] [Accepted: 12/01/2016] [Indexed: 12/31/2022]
Abstract
Prenatal maternal infection represents a risk factor for the development of psychopathologic conditions later in life. Clinical evidence is also supported by animal models in which the vulnerability to develop a schizophrenic-like phenotype likely originates from inflammatory processes as early as in the womb. Prenatal immune challenge, for example, induces a variety of long-term behavioral alterations in mice, such as deficits in recognition and spatial working memory, perseverative behaviors and social impairments, which are relevant to different symptom clusters of schizophrenia. Here, we investigated the modulation of GABAergic markers in the dorsal and ventral hippocampus of adult mice exposed to late prenatal immune challenge with the viral mimetic Poly(I:C) (polyriboinosinic-polyribocytidilic-acid) at gestational day 17, and we evaluated the ability of chronic treatment with the multi-receptor antipsychotic lurasidone to modulate the alterations produced by maternal infection. Poly(I:C) mice show a significant reduction of key GABAergic markers, such as GAD67 and parvalbumin, specifically in the dorsal hippocampus, which were normalized by chronic lurasidone administration. Moreover, chronic drug administration increases the expression of the pool of brain derived neurotrophic factor (BDNF) transcripts with the long 3'-UTR as well as the levels of mature BDNF protein in the synaptosomal compartment, selectively in dorsal hippocampus. All in all, our findings demonstrate that lurasidone is effective in ameliorating molecular abnormalities observed in Poly(I:C) mice, providing further support to the neuroplastic properties of this multi-receptor antipsychotic drug.
Collapse
Affiliation(s)
- A Luoni
- Department of Pharmacological and Biomolecular Sciences, University of Milan, via Balzaretti 9, 20133 Milan, Italy
| | - J Richetto
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland
| | - L Longo
- Department of Pharmacological and Biomolecular Sciences, University of Milan, via Balzaretti 9, 20133 Milan, Italy
| | - M A Riva
- Department of Pharmacological and Biomolecular Sciences, University of Milan, via Balzaretti 9, 20133 Milan, Italy.
| |
Collapse
|
37
|
da Silveira VT, Medeiros DDC, Ropke J, Guidine PA, Rezende GH, Moraes MFD, Mendes EMAM, Macedo D, Moreira FA, de Oliveira ACP. Effects of early or late prenatal immune activation in mice on behavioral and neuroanatomical abnormalities relevant to schizophrenia in the adulthood. Int J Dev Neurosci 2017; 58:1-8. [PMID: 28122258 DOI: 10.1016/j.ijdevneu.2017.01.009] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 01/19/2017] [Accepted: 01/19/2017] [Indexed: 12/20/2022] Open
Abstract
Maternal immune activation (MIA) during pregnancy in rodents increases the risk of the offspring to develop schizophrenia-related behaviors, suggesting a relationship between the immune system and the brain development. Here we tested the hypothesis that MIA induced by the viral mimetic polyinosinic-polycytidylic acid (poly I:C) in early or late gestation of mice leads to behavioral and neuroanatomical disorders in the adulthood. On gestational days (GDs) 9 or 17 pregnant dams were treated with poly I:C or saline via intravenous route and the offspring behaviors were measured during adulthood. Considering the progressive structural neuroanatomical alterations in the brain of individuals with schizophrenia, we used magnetic resonance imaging (MRI) to perform brain morphometric analysis of the offspring aged one year. MIA on GD9 or GD17 led to increased basal locomotor activity, enhanced motor responses to ketamine, a psychotomimetic drug, and reduced time spent in the center of the arena, suggesting an increased anxiety-like behavior. In addition, MIA on GD17 reduced glucose preference in the offspring. None of the treatments altered the relative volume of the lateral ventricles. However, a decrease in brain volume, especially for posterior structures, was observed for one-year-old animals treated with poly I:C compared with control groups. Thus, activation of the maternal immune system at different GDs lead to neuroanatomical and behavioral alterations possibly related to the positive and negative symptoms of schizophrenia. These results provide insights on neuroimmunonological and neurodevelopmental aspects of certain psychopathologies, such as schizophrenia.
Collapse
Affiliation(s)
- Vivian T da Silveira
- Department of Pharmacology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Brazil
| | - Daniel de Castro Medeiros
- Center for Technology and Research in Magneto-Resonance (CTPMAG), Graduate Program in Electrical Engineering - Universidade Federal de Minas Gerais, Brazil
| | - Jivago Ropke
- Department of Pharmacology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Brazil
| | - Patricia A Guidine
- Center for Technology and Research in Magneto-Resonance (CTPMAG), Graduate Program in Electrical Engineering - Universidade Federal de Minas Gerais, Brazil
| | - Gustavo H Rezende
- Center for Technology and Research in Magneto-Resonance (CTPMAG), Graduate Program in Electrical Engineering - Universidade Federal de Minas Gerais, Brazil
| | - Marcio Flavio D Moraes
- Center for Technology and Research in Magneto-Resonance (CTPMAG), Graduate Program in Electrical Engineering - Universidade Federal de Minas Gerais, Brazil
| | - Eduardo Mazoni A M Mendes
- Center for Technology and Research in Magneto-Resonance (CTPMAG), Graduate Program in Electrical Engineering - Universidade Federal de Minas Gerais, Brazil
| | - Danielle Macedo
- Drug Research and Development Center, Department of Physiology and Pharmacology, Medical School, Universidade Federal do Ceará, Brazil
| | - Fabricio A Moreira
- Department of Pharmacology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Brazil
| | | |
Collapse
|
38
|
Eßlinger M, Wachholz S, Manitz MP, Plümper J, Sommer R, Juckel G, Friebe A. Schizophrenia associated sensory gating deficits develop after adolescent microglia activation. Brain Behav Immun 2016; 58:99-106. [PMID: 27235930 DOI: 10.1016/j.bbi.2016.05.018] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 05/05/2016] [Accepted: 05/24/2016] [Indexed: 10/21/2022] Open
Abstract
Maternal infection during pregnancy is a well-established risk factor for schizophrenia in the adult offspring. Consistently, prenatal Poly(I:C) treatment in mice has been validated to model behavioral and neurodevelopmental abnormalities associated with schizophrenia. By using the Poly(I:C) BALB/c mouse model, we investigated the functional profile of microglia by flow cytometry in relation to progressive behavioral changes from adolescence to adulthood. Prenatal Poly(I:C) treatment induced the expected sensory gating deficits (pre-pulse inhibition (PPI) of the acoustic startle response) in 100day-old adult offspring, but only in female not in male descendants. No PPI-deficits were present in 30day-old adolescent mice. Sensory gating deficits in adult females were preceded by a strong M1-type microglia polarization pattern during puberty as determined by flow cytometric analysis of multiple pro- and anti-inflammatory surface markers. Microglia activation in females did not persist until adulthood and was absent in behaviorally unaffected male descendants. Further, the specific activation pattern of microglia was not mirrored by a similar activation of peripheral immune cells. We conclude that prenatal Poly(I:C) treatment induces post pubertal deficits in sensory gating which are specifically preceded by a pro-inflammatory activation pattern of microglia during puberty.
Collapse
Affiliation(s)
- Manuela Eßlinger
- Laboratory of Psychoneuroimmunology, Department of Psychiatry, Center of Clinical Research 1 (ZKF1 2/052), Universitätsstraße 150, Ruhr University Bochum, D-44801 Bochum, Germany; Department of Psychiatry, LWL University Hospital, Ruhr-University Bochum, Alexandrinenstr. 1, D-44791 Bochum, Germany.
| | - Simone Wachholz
- Laboratory of Psychoneuroimmunology, Department of Psychiatry, Center of Clinical Research 1 (ZKF1 2/052), Universitätsstraße 150, Ruhr University Bochum, D-44801 Bochum, Germany; Department of Psychiatry, LWL University Hospital, Ruhr-University Bochum, Alexandrinenstr. 1, D-44791 Bochum, Germany.
| | - Marie-Pierre Manitz
- Laboratory of Psychoneuroimmunology, Department of Psychiatry, Center of Clinical Research 1 (ZKF1 2/052), Universitätsstraße 150, Ruhr University Bochum, D-44801 Bochum, Germany; Department of Psychiatry, LWL University Hospital, Ruhr-University Bochum, Alexandrinenstr. 1, D-44791 Bochum, Germany.
| | - Jennifer Plümper
- Laboratory of Psychoneuroimmunology, Department of Psychiatry, Center of Clinical Research 1 (ZKF1 2/052), Universitätsstraße 150, Ruhr University Bochum, D-44801 Bochum, Germany; Department of Psychiatry, LWL University Hospital, Ruhr-University Bochum, Alexandrinenstr. 1, D-44791 Bochum, Germany.
| | - Rainer Sommer
- Laboratory of Psychoneuroimmunology, Department of Psychiatry, Center of Clinical Research 1 (ZKF1 2/052), Universitätsstraße 150, Ruhr University Bochum, D-44801 Bochum, Germany.
| | - Georg Juckel
- Laboratory of Psychoneuroimmunology, Department of Psychiatry, Center of Clinical Research 1 (ZKF1 2/052), Universitätsstraße 150, Ruhr University Bochum, D-44801 Bochum, Germany; Department of Psychiatry, LWL University Hospital, Ruhr-University Bochum, Alexandrinenstr. 1, D-44791 Bochum, Germany.
| | - Astrid Friebe
- Laboratory of Psychoneuroimmunology, Department of Psychiatry, Center of Clinical Research 1 (ZKF1 2/052), Universitätsstraße 150, Ruhr University Bochum, D-44801 Bochum, Germany; Department of Psychiatry, LWL University Hospital, Ruhr-University Bochum, Alexandrinenstr. 1, D-44791 Bochum, Germany.
| |
Collapse
|
39
|
Gomes FV, Rincón-Cortés M, Grace AA. Adolescence as a period of vulnerability and intervention in schizophrenia: Insights from the MAM model. Neurosci Biobehav Rev 2016; 70:260-270. [PMID: 27235082 PMCID: PMC5074867 DOI: 10.1016/j.neubiorev.2016.05.030] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 05/24/2016] [Accepted: 05/24/2016] [Indexed: 11/30/2022]
Abstract
Adolescence is a time of extensive neuroanatomical, functional and chemical reorganization of the brain, which parallels substantial maturational changes in behavior and cognition. Environmental factors that impinge on the timing of these developmental factors, including stress and drug exposure, increase the risk for psychiatric disorders. Indeed, antecedents to affective and psychotic disorders, which have clinical and pathophysiological overlap, are commonly associated with risk factors during adolescence that predispose to these disorders. In the context of schizophrenia, psychosis typically begins in late adolescence/early adulthood, which has been replicated by animal models. Rats exposed during gestational day (GD) 17 to the mitotoxin methylazoxymethanol acetate (MAM) exhibit behavioral, pharmacological, and anatomical characteristics consistent with an animal model of schizophrenia. Here we provide an overview of adolescent changes within the dopamine system and the PFC and review recent findings regarding the effects of stress and cannabis exposure during the peripubertal period as risk factors for the emergence of schizophrenia-like deficits. Finally, we discuss peripubertal interventions appearing to circumvent the emergence of adult schizophrenia-like deficits.
Collapse
Affiliation(s)
- Felipe V Gomes
- Departments of Neuroscience, Psychiatry and Psychology, United States
| | | | - Anthony A Grace
- Departments of Neuroscience, Psychiatry and Psychology, United States.
| |
Collapse
|
40
|
Santos CM, Peres FF, Diana MC, Justi V, Suiama MA, Santana MG, Abílio VC. Peripubertal exposure to environmental enrichment prevents schizophrenia-like behaviors in the SHR strain animal model. Schizophr Res 2016; 176:552-559. [PMID: 27338757 DOI: 10.1016/j.schres.2016.06.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 06/04/2016] [Accepted: 06/07/2016] [Indexed: 11/28/2022]
Abstract
Schizophrenia is a highly disabling mental disorder, in which genetics and environmental factors interact culminating in the disease. The treatment of negative symptoms and cognitive deficits with antipsychotics is currently inefficient and is an important field of research. Environmental enrichment (EE) has been suggested to improve some cognitive deficits in animal models of various psychiatric disorders. In this study, we aimed to evaluate a possible beneficial effect of early and long-term exposure to EE on an animal model of schizophrenia, the SHR strain. Young male Wistar rats (control strain) and SHRs (21 post-natal days) were housed for 6weeks in two different conditions: in large cages (10 animals per cage) containing objects of different textures, forms, colors and materials that were changed 3 times/week (EE condition) or in standard cages (5 animals per cage - Control condition). Behavioral evaluations - social interaction (SI), locomotion, prepulse inhibition of startle (PPI) and spontaneous alternation (SA) - were performed 6weeks after the end of EE. SHRs presented deficits in PPI (a sensorimotor impairment), SI (mimicking the negative symptoms) and SA (a working memory deficit), and also hyperlocomotion (modeling the positive symptoms). EE was able to reduce locomotion and increase PPI in both strains, and to prevent the working memory deficit in SHRs. EE also increased the number of neurons in the CA1 and CA3 of the hippocampus. In conclusion, EE can be a potential nonpharmacological strategy to prevent some behavioral deficits associated with schizophrenia.
Collapse
Affiliation(s)
- Camila Mauricio Santos
- LiNC - Interdisciplinary Laboratory of Clinical Neurosciences, Department of Psychiatry, Universidade Federal de São Paulo (UNIFESP), Rua Pedro de Toledo, 669, 3° andar, Ed. de Pesquisas II, CEP 04039-032 São Paulo, SP, Brazil
| | - Fernanda Fiel Peres
- LiNC - Interdisciplinary Laboratory of Clinical Neurosciences, Department of Psychiatry, Universidade Federal de São Paulo (UNIFESP), Rua Pedro de Toledo, 669, 3° andar, Ed. de Pesquisas II, CEP 04039-032 São Paulo, SP, Brazil; Department of Pharmacology, Universidade Federal de São Paulo (UNIFESP), Rua Pedro de Toledo, 669, 5° andar, Ed. de Pesquisas II, CEP 04039-032 São Paulo, SP, Brazil
| | - Mariana Cepollaro Diana
- LiNC - Interdisciplinary Laboratory of Clinical Neurosciences, Department of Psychiatry, Universidade Federal de São Paulo (UNIFESP), Rua Pedro de Toledo, 669, 3° andar, Ed. de Pesquisas II, CEP 04039-032 São Paulo, SP, Brazil
| | - Veronica Justi
- LiNC - Interdisciplinary Laboratory of Clinical Neurosciences, Department of Psychiatry, Universidade Federal de São Paulo (UNIFESP), Rua Pedro de Toledo, 669, 3° andar, Ed. de Pesquisas II, CEP 04039-032 São Paulo, SP, Brazil
| | - Mayra Akimi Suiama
- LiNC - Interdisciplinary Laboratory of Clinical Neurosciences, Department of Psychiatry, Universidade Federal de São Paulo (UNIFESP), Rua Pedro de Toledo, 669, 3° andar, Ed. de Pesquisas II, CEP 04039-032 São Paulo, SP, Brazil; Department of Pharmacology, Universidade Federal de São Paulo (UNIFESP), Rua Pedro de Toledo, 669, 5° andar, Ed. de Pesquisas II, CEP 04039-032 São Paulo, SP, Brazil
| | - Marcela Gonçalves Santana
- LiNC - Interdisciplinary Laboratory of Clinical Neurosciences, Department of Psychiatry, Universidade Federal de São Paulo (UNIFESP), Rua Pedro de Toledo, 669, 3° andar, Ed. de Pesquisas II, CEP 04039-032 São Paulo, SP, Brazil; Department of Pharmacology, Universidade Federal de São Paulo (UNIFESP), Rua Pedro de Toledo, 669, 5° andar, Ed. de Pesquisas II, CEP 04039-032 São Paulo, SP, Brazil
| | - Vanessa Costhek Abílio
- LiNC - Interdisciplinary Laboratory of Clinical Neurosciences, Department of Psychiatry, Universidade Federal de São Paulo (UNIFESP), Rua Pedro de Toledo, 669, 3° andar, Ed. de Pesquisas II, CEP 04039-032 São Paulo, SP, Brazil; Department of Pharmacology, Universidade Federal de São Paulo (UNIFESP), Rua Pedro de Toledo, 669, 5° andar, Ed. de Pesquisas II, CEP 04039-032 São Paulo, SP, Brazil
| |
Collapse
|
41
|
Amygdala Hyperactivity in MAM Model of Schizophrenia is Normalized by Peripubertal Diazepam Administration. Neuropsychopharmacology 2016; 41:2455-62. [PMID: 27000940 PMCID: PMC4987842 DOI: 10.1038/npp.2016.42] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 03/16/2016] [Accepted: 03/17/2016] [Indexed: 01/12/2023]
Abstract
In addition to prefrontal cortex (PFC) and hippocampus, amygdala may have a role in the pathophysiology of schizophrenia, given its pivotal role in emotion and extensive connectivity with the PFC and hippocampus. Moreover, abnormal activities of amygdala may be related to the anxiety observed in schizophrenia patients and at-risk adolescents. These at-risk subjects demonstrated heightened levels of anxiety, which are correlated with the onset of psychosis later in life. Similarly, rats that received methyl azoxymethanol acetate (MAM) gestationally exhibited higher levels of anxiety peripubertally. In the current study, the heightened anxiety was also observed in adult MAM animals, as well as higher firing rates of BLA neurons in both peripubertal and adult MAM rats. In addition, the power of BLA theta oscillations of adult MAM rats showed a larger increase in response to conditioned stimuli (CS). We showed previously that administration of the antianxiety drug diazepam during the peripubertal period prevents the hyperdopaminergic state in adult MAM rats. In this study, we found that peripubertal diazepam treatment reduced heightened anxiety, decreased BLA neuron firing rates and attenuated the CS-induced increase in BLA theta power in adult MAM rats, supporting a persistent normalization by this treatment. This study provides a link between BLA hyperactivity and anxiety in schizophrenia model rats and that circumvention of stress may prevent the emergence of pathology in the adult.
Collapse
|
42
|
Li M. Antipsychotic-induced sensitization and tolerance: Behavioral characteristics, developmental impacts, and neurobiological mechanisms. J Psychopharmacol 2016; 30:749-70. [PMID: 27371498 PMCID: PMC4944179 DOI: 10.1177/0269881116654697] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Antipsychotic sensitization and tolerance refer to the increased and decreased drug effects due to past drug use, respectively. Both effects reflect the long-term impacts of antipsychotic treatment on the brain and result from the brain's adaptive response to the foreign property of the drug. In this review, clinical evidence of the behavioral aspect of antipsychotic sensitization and tolerance is selectively reviewed, followed by an overview of preclinical literature that examines these behavioral characteristics and the related pharmacological and nonpharmacological factors. Next, recent work on the developmental impacts of adolescent antipsychotic sensitization and tolerance is presented and recent research that delineates the neurobiological mechanisms of antipsychotic sensitization and tolerance is summarized. A theoretical framework based on "drug learning and memory" principles is proposed to account for the phenomena of antipsychotic sensitization and tolerance. It is maintained that antipsychotic sensitization and tolerance follow basic principles of learning or acquisition ("induction") and memory ("expression"). The induction and expression of both effects reflect the consequences of associative and nonassociative processing and are strongly influenced by various pharmacological, environmental, and behavioral factors. Drug-induced neuroplasticity, such as functional changes of striatal dopamine D2 and prefrontal serotonin (5-HT)2A receptors and their mediated signaling pathways, in principle, is responsible for antipsychotic sensitization and tolerance. Understanding the behavioral characteristics and neurobiological underpinnings of antipsychotic sensitization and tolerance has greatly enhanced our understanding of mechanisms of antipsychotic action, and may have important implications for future drug discovery and clinical practice.
Collapse
Affiliation(s)
- Ming Li
- Department of Psychology, University of Nebraska-Lincoln, Lincoln, NE, USA
| |
Collapse
|
43
|
Moe AAK, Scott JG, Burne TH, Eyles DW. Neural changes induced by antipsychotic administration in adolescence: A review of studies in laboratory rodents. J Psychopharmacol 2016; 30:771-94. [PMID: 27413140 DOI: 10.1177/0269881116654776] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Adolescence is characterized by major remodelling processes in the brain. Use of antipsychotic drugs (APDs) in adolescents has increased dramatically in the last 20 years; however, our understanding of the neurobiological consequences of APD treatment on the adolescent brain has not kept the same pace and significant concerns have been raised. In this review, we examined currently available preclinical studies of the effects of APDs on the adolescent brain. In animal models of neuropsychiatric disorders, adolescent APD treatment appears to be protective against selected structural, behavioural and neurochemical phenotypes. In "neurodevelopmentally normal" adolescent animals, a range of short- and long-term alterations in behaviour and neurochemistry have been reported. In particular, the adolescent brain appears to be sensitive to long-term locomotor/reward effects of chronic atypical APDs in contrast with the outcomes in adults. Long-lasting changes in dopaminergic, glutamatergic and gamma-amino butyric acid-ergic systems induced by adolescent APD administration have been observed in the nucleus accumbens. A detailed examination of other potential target regions such as striatum, prefrontal cortex and ventral tegmental area is still required. Through identification of specific neural pathways targeted by adolescent APD treatment, future studies will expand the current knowledge on long-term neural outcomes which are of translational value.
Collapse
Affiliation(s)
- Aung Aung Kywe Moe
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD, Australia
| | - James G Scott
- Queensland Centre for Mental Health Research, Wacol, QLD, Australia Discipline of Psychiatry, School of Medicine, The University of Queensland Centre for Clinical Research, Herston, QLD, Australia Metro North Mental Health Service, Royal Brisbane and Women's Hospital, Herston, QLD, Australia
| | - Thomas Hj Burne
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD, Australia Queensland Centre for Mental Health Research, Wacol, QLD, Australia
| | - Darryl W Eyles
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD, Australia Queensland Centre for Mental Health Research, Wacol, QLD, Australia
| |
Collapse
|
44
|
Bikovsky L, Hadar R, Soto-Montenegro ML, Klein J, Weiner I, Desco M, Pascau J, Winter C, Hamani C. Deep brain stimulation improves behavior and modulates neural circuits in a rodent model of schizophrenia. Exp Neurol 2016; 283:142-50. [PMID: 27302677 DOI: 10.1016/j.expneurol.2016.06.012] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 03/07/2016] [Accepted: 06/10/2016] [Indexed: 12/11/2022]
Abstract
Schizophrenia is a debilitating psychiatric disorder with a significant number of patients not adequately responding to treatment. Deep brain stimulation (DBS) is a surgical technique currently investigated for medically-refractory psychiatric disorders. Here, we use the poly I:C rat model of schizophrenia to study the effects of medial prefrontal cortex (mPFC) and nucleus accumbens (Nacc) DBS on two behavioral schizophrenia-like deficits, i.e. sensorimotor gating, as reflected by disrupted prepulse inhibition (PPI), and attentional selectivity, as reflected by disrupted latent inhibition (LI). In addition, the neurocircuitry influenced by DBS was studied using FDG PET. We found that mPFC- and Nacc-DBS alleviated PPI and LI abnormalities in poly I:C offspring, whereas Nacc- but not mPFC-DBS disrupted PPI and LI in saline offspring. In saline offspring, mPFC-DBS increased metabolism in the parietal cortex, striatum, ventral hippocampus and Nacc, while reducing it in the brainstem, cerebellum, hypothalamus and periaqueductal gray. Nacc-DBS, on the other hand, increased activity in the ventral hippocampus and olfactory bulb and reduced it in the septal area, brainstem, periaqueductal gray and hypothalamus. In poly I:C offspring changes in metabolism following mPFC-DBS were similar to those recorded in saline offspring, except for a reduced activity in the brainstem and hypothalamus. In contrast, Nacc-DBS did not induce any statistical changes in brain metabolism in poly I:C offspring. Our study shows that mPFC- or Nacc-DBS delivered to the adult progeny of poly I:C treated dams improves deficits in PPI and LI. Despite common behavioral responses, stimulation in the two targets induced different metabolic effects.
Collapse
Affiliation(s)
- Lior Bikovsky
- School of Psychological Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Ravit Hadar
- Department of Psychiatry and Psychotherapy, University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany
| | | | - Julia Klein
- Department of Psychiatry and Psychotherapy, Charité University Medicine Berlin, Campus Charité Mitte, Berlin, Germany
| | - Ina Weiner
- School of Psychological Sciences, Tel Aviv University, Tel Aviv, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Manuel Desco
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain; CIBERSAM, Madrid, Spain; Departamento de Bioingeniería e Ingeniería Aeroespacial, Universidad Carlos III de Madrid, Spain
| | - Javier Pascau
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain; CIBERSAM, Madrid, Spain; Departamento de Bioingeniería e Ingeniería Aeroespacial, Universidad Carlos III de Madrid, Spain
| | - Christine Winter
- Department of Psychiatry and Psychotherapy, University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany.
| | - Clement Hamani
- Behavioural Neurobiology Laboratory, Centre for Addiction and Mental Health, 250 College Street, Toronto, ON, M5T 1R8, Canada; Campbell Family Mental Health Research Institute, CAMH, Canada; Division of Neurosurgery, Toronto Western Hospital, 399 Bathurst Street, Toronto, ON, M5T 2S8, Canada
| |
Collapse
|
45
|
Serati M, Redaelli M, Buoli M, Altamura AC. Perinatal Major Depression Biomarkers: A systematic review. J Affect Disord 2016; 193:391-404. [PMID: 26802316 DOI: 10.1016/j.jad.2016.01.027] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Revised: 12/04/2015] [Accepted: 01/12/2016] [Indexed: 12/20/2022]
Abstract
Postpartum depression, now termed perinatal depression by the DSM-5, is a clinically relevant disorder reaching 15% of incidence. Although it is quite frequent and associated with high social dysfunction, only recently its underpinning biological pathways have been explored, while multiple and concomitant risk factors have been identified (e.g. psychosocial stress). Peripartum depression usually has its onset during the third trimester of pregnancy or in the postpartum, being one of the most common medical complications in new mothers. Purpose of the present review is to summarize the state of art of biological biomarkers involved in the pathogenesis of perinatal depression, in view of the fact that suboptimal prenatal milieu can induce permanent damage in subsequent offspring life and have a negative impact on mother-child relationship. Furthermore, parents' biological changes due to medical/psychiatric disorders or stress exposure could influence offspring life: a concept known as 'intergenerational transmission', acting by variations into gametes and the gestational uterine environment. Given the evidence that perinatal mental disorders involve risks for the mother and offspring, the search for reliable biomarkers in high-risk mothers actually represents a medical priority to prevent perinatal depression.
Collapse
Affiliation(s)
- M Serati
- Department of Psychiatry, University of Milan, Fondazione IRCCS Ca'Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122 Milan, Italy.
| | - M Redaelli
- Department of Psychiatry, University of Milan, Fondazione IRCCS Ca'Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122 Milan, Italy
| | - M Buoli
- Department of Psychiatry, University of Milan, Fondazione IRCCS Ca'Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122 Milan, Italy
| | - A C Altamura
- Department of Psychiatry, University of Milan, Fondazione IRCCS Ca'Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122 Milan, Italy
| |
Collapse
|
46
|
Abstract
Despite a lack of recent progress in the treatment of schizophrenia, our understanding of its genetic and environmental causes has considerably improved, and their relationship to aberrant patterns of neurodevelopment has become clearer. This raises the possibility that 'disease-modifying' strategies could alter the course to - and of - this debilitating disorder, rather than simply alleviating symptoms. A promising window for course-altering intervention is around the time of the first episode of psychosis, especially in young people at risk of transition to schizophrenia. Indeed, studies performed in both individuals at risk of developing schizophrenia and rodent models for schizophrenia suggest that pre-diagnostic pharmacotherapy and psychosocial or cognitive-behavioural interventions can delay or moderate the emergence of psychosis. Of particular interest are 'hybrid' strategies that both relieve presenting symptoms and reduce the risk of transition to schizophrenia or another psychiatric disorder. This Review aims to provide a broad-based consideration of the challenges and opportunities inherent in efforts to alter the course of schizophrenia.
Collapse
|
47
|
Duchatel RJ, Jobling P, Graham BA, Harms LR, Michie PT, Hodgson DM, Tooney PA. Increased white matter neuron density in a rat model of maternal immune activation - Implications for schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry 2016; 65:118-26. [PMID: 26385575 DOI: 10.1016/j.pnpbp.2015.09.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 09/04/2015] [Accepted: 09/11/2015] [Indexed: 10/23/2022]
Abstract
Interstitial neurons are located among white matter tracts of the human and rodent brain. Post-mortem studies have identified increased interstitial white matter neuron (IWMN) density in the fibre tracts below the cortex in people with schizophrenia. The current study assesses IWMN pathology in a model of maternal immune activation (MIA); a risk factor for schizophrenia. Experimental MIA was produced by an injection of polyinosinic:polycytidylic acid (PolyI:C) into pregnant rats on gestational day (GD) 10 or GD19. A separate control group received saline injections. The density of neuronal nuclear antigen (NeuN(+)) and somatostatin (SST(+)) IWMNs was determined in the white matter of the corpus callosum in two rostrocaudally adjacent areas in the 12week old offspring of GD10 (n=10) or GD19 polyI:C dams (n=18) compared to controls (n=20). NeuN(+) IWMN density trended to be higher in offspring from dams exposed to polyI:C at GD19, but not GD10. A subpopulation of these NeuN(+) IWMNs was shown to express SST. PolyI:C treatment of dams induced a significant increase in the density of SST(+) IWMNs in the offspring when delivered at both gestational stages with more regionally widespread effects observed at GD19. A positive correlation was observed between NeuN(+) and SST(+) IWMN density in animals exposed to polyI:C at GD19, but not controls. This is the first study to show that MIA increases IWMN density in adult offspring in a similar manner to that seen in the brain in schizophrenia. This suggests the MIA model will be useful in future studies aimed at probing the relationship between IWMNs and schizophrenia.
Collapse
Affiliation(s)
- Ryan J Duchatel
- Preclinical Neurobiology Research Group, School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW 2308, Australia; Centre for Translational Neuroscience and Mental Health, The University of Newcastle, Callaghan, NSW 2308, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia; Schizophrenia Research Institute, Sydney, NSW, Australia.
| | - Phillip Jobling
- Preclinical Neurobiology Research Group, School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW 2308, Australia; Centre for Translational Neuroscience and Mental Health, The University of Newcastle, Callaghan, NSW 2308, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia.
| | - Brett A Graham
- Preclinical Neurobiology Research Group, School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW 2308, Australia; Centre for Translational Neuroscience and Mental Health, The University of Newcastle, Callaghan, NSW 2308, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia.
| | - Lauren R Harms
- School of Psychology, Faculty of Science and IT, University of Newcastle, Callaghan, NSW 2308, Australia; Schizophrenia Research Institute, Sydney, NSW, Australia.
| | - Patricia T Michie
- School of Psychology, Faculty of Science and IT, University of Newcastle, Callaghan, NSW 2308, Australia; Schizophrenia Research Institute, Sydney, NSW, Australia.
| | - Deborah M Hodgson
- School of Psychology, Faculty of Science and IT, University of Newcastle, Callaghan, NSW 2308, Australia; Schizophrenia Research Institute, Sydney, NSW, Australia.
| | - Paul A Tooney
- Preclinical Neurobiology Research Group, School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW 2308, Australia; Centre for Translational Neuroscience and Mental Health, The University of Newcastle, Callaghan, NSW 2308, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia; Schizophrenia Research Institute, Sydney, NSW, Australia.
| |
Collapse
|
48
|
Maternal immune activation produces neonatal excitability defects in offspring hippocampal neurons from pregnant rats treated with poly I:C. Sci Rep 2016; 6:19106. [PMID: 26742695 PMCID: PMC4705483 DOI: 10.1038/srep19106] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 12/02/2015] [Indexed: 12/26/2022] Open
Abstract
Maternal immune activation (MIA) resulting from prenatal exposure to infectious pathogens or inflammatory stimuli is increasingly recognized to play an important etiological role in neuropsychiatric disorders with neurodevelopmental features. MIA in pregnant rodents induced by injection of the synthetic double-stranded RNA, Poly I:C, a mimic of viral infection, leads to a wide spectrum of behavioral abnormalities as well as structural and functional defects in the brain. Previous MIA studies using poly I:C prenatal treatment suggested that neurophysiological alterations occur in the hippocampus. However, these investigations used only juvenile or adult animals. We postulated that MIA-induced alterations could occur earlier at neonatal/early postnatal stages. Here we examined the neurophysiological properties of cultured pyramidal-like hippocampal neurons prepared from neonatal (P0-P2) offspring of pregnant rats injected with poly I:C. Offspring neurons from poly I:C-treated mothers exhibited significantly lower intrinsic excitability and stronger spike frequency adaptation, compared to saline. A similar lower intrinsic excitability was observed in CA1 pyramidal neurons from hippocampal slices of two weeks-old poly I:C offspring. Cultured hippocampal neurons also displayed lower frequency of spontaneous firing, higher charge transfer of IPSCs and larger amplitude of miniature IPSCs. Thus, maternal immune activation leads to strikingly early neurophysiological abnormalities in hippocampal neurons.
Collapse
|
49
|
Patrich E, Piontkewitz Y, Peretz A, Weiner I, Attali B. Maturation- and sex-sensitive depression of hippocampal excitatory transmission in a rat schizophrenia model. Brain Behav Immun 2016; 51:240-251. [PMID: 26327125 DOI: 10.1016/j.bbi.2015.08.021] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 08/20/2015] [Accepted: 08/27/2015] [Indexed: 11/28/2022] Open
Abstract
Schizophrenia is associated with behavioral and brain structural abnormalities, of which the hippocampus appears to be one of the most consistent region affected. Previous studies performed on the poly I:C model of schizophrenia suggest that alterations in hippocampal synaptic transmission and plasticity take place in the offspring. However, these investigations yielded conflicting results and the neurophysiological alterations responsible for these deficits are still unclear. Here we performed for the first time a longitudinal study examining the impact of prenatal poly I:C treatment and of gender on hippocampal excitatory neurotransmission. In addition, we examined the potential preventive/curative effects of risperidone (RIS) treatment during the peri-adolescence period. Excitatory synaptic transmission was determined by stimulating Schaffer collaterals and monitoring fiber volley amplitude and slope of field-EPSP (fEPSP) in CA1 pyramidal neurons in male and female offspring hippocampal slices from postnatal days (PNDs) 18-20, 34, 70 and 90. Depression of hippocampal excitatory transmission appeared at juvenile age in male offspring of the poly I:C group, while it expressed with a delay in female, manifesting at adulthood. In addition, a reduced hippocampal size was found in both adult male and female offspring of poly I:C treated dams. Treatment with RIS at the peri-adolescence period fully restored in males but partly repaired in females these deficiencies. A maturation- and sex-dependent decrease in hippocampal excitatory transmission occurs in the offspring of poly I:C treated pregnant mothers. Pharmacological intervention with RIS during peri-adolescence can cure in a gender-sensitive fashion early occurring hippocampal synaptic deficits.
Collapse
Affiliation(s)
- Eti Patrich
- Department of Physiology & Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel; Department of Psychology, Gordon Faculty of Social Sciences, Tel Aviv University, Tel Aviv 69978, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel
| | - Yael Piontkewitz
- Strauss Center for Computational Neuroimaging, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Asher Peretz
- Department of Physiology & Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Ina Weiner
- Department of Psychology, Gordon Faculty of Social Sciences, Tel Aviv University, Tel Aviv 69978, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel
| | - Bernard Attali
- Department of Physiology & Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel.
| |
Collapse
|
50
|
Hemmerle AM, Ahlbrand R, Bronson SL, Lundgren KH, Richtand NM, Seroogy KB. Modulation of schizophrenia-related genes in the forebrain of adolescent and adult rats exposed to maternal immune activation. Schizophr Res 2015; 168. [PMID: 26206493 PMCID: PMC4591187 DOI: 10.1016/j.schres.2015.07.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Maternal immune activation (MIA) is an environmental risk factor for schizophrenia, and may contribute to other developmental disorders including autism and epilepsy. Activation of pro-inflammatory cytokine systems by injection of the synthetic double-stranded RNA polyriboinosinic-polyribocytidilic acid (Poly I:C) mediates important neurochemical and behavioral corollaries of MIA, which have relevance to deficits observed in schizophrenia. We examined the consequences of MIA on forebrain expression of neuregulin-1 (NRG-1), brain-derived neurotrophic factor (BDNF) and their receptors, ErbB4 and trkB, respectively, genes associated with schizophrenia. On gestational day 14, pregnant rats were injected with Poly I:C or vehicle. Utilizing in situ hybridization, expression of NRG-1, ErbB4, BDNF, and trkB was examined in male rat offspring at postnatal day (P) 14, P30 and P60. ErbB4 mRNA expression was significantly increased at P30 in the anterior cingulate (AC Ctx), frontal, and parietal cortices, with increases in AC Ctx expression continuing through P60. ErbB4 expression was also elevated in the prefrontal cortex (PFC) at P14. In contrast, NRG-1 mRNA was decreased in the PFC at P60. Expression of BDNF mRNA was significantly upregulated in the PFC at P60 and decreased in the AC Ctx at P14. Expression of trkB was increased in two regions, the piriform cortex at P14 and the striatum at P60. These findings demonstrate developmentally and regionally selective alterations in the expression of schizophrenia-related genes as a consequence of MIA. Further study is needed to determine contributions of these effects to the development of alterations of relevance to neuropsychiatric diseases.
Collapse
Affiliation(s)
- Ann M. Hemmerle
- Department of Neurology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA,Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Rebecca Ahlbrand
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Stefanie L. Bronson
- Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH 45267, USA,Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Kerstin H. Lundgren
- Department of Neurology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Neil M. Richtand
- Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH 45267, USA,Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA,San Diego Veterans Affairs Healthcare System, San Diego, CA 92161USA,Department of Psychiatry, University of California, San Diego School of Medicine, La Jolla, CA 92093, USA
| | - Kim B. Seroogy
- Department of Neurology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA,Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH 45267, USA,Corresponding Author: Kim B. Seroogy, PhD, The Selma Schottenstein Harris Laboratory for Research in Parkinson’s, Gardner Family Center for Parkinson’s Disease and Movement Disorders, Department of Neurology, University of Cincinnati College of Medicine, Medical Sciences Building, ML0536, 231 Albert Sabin Way, Cincinnati, OH 45267-0536, USA. Telephone: 513-558-7086; Fax: 513-558-7009;
| |
Collapse
|