1
|
Munn RGK, Wolff A, Speers LJ, Bilkey DK. Disrupted hippocampal synchrony following maternal immune activation in a rat model. Hippocampus 2023; 33:995-1008. [PMID: 37129454 DOI: 10.1002/hipo.23545] [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: 11/07/2022] [Revised: 03/14/2023] [Accepted: 04/16/2023] [Indexed: 05/03/2023]
Abstract
Maternal immune activation (MIA) is a risk factor for schizophrenia and other neurodevelopmental disorders. MIA in rats models a number of the brain and behavioral changes that are observed in schizophrenia, including impaired memory. Recent studies in the MIA model have shown that the firing of the hippocampal place cells that are involved in memory processes appear relatively normal, but with abnormalities in the temporal ordering of firing. In this study, we re-analyzed data from prior hippocampal electrophysiological recordings of MIA and control animals to determine whether temporal dysfunction was evident. We find that there is a decreased ratio of slow to fast gamma power, resulting from an increase in fast gamma power and a tendency toward reduced slow gamma power in MIA rats. Moreover, we observe a robust reduction in spectral coherence between hippocampal theta and both fast and slow gamma rhythms, as well as changes in the phase of theta at which fast gamma occurs. We also find the phasic organization of place cell phase precession on the theta wave to be abnormal in MIA rats. Lastly, we observe that the local field potential of MIA rats contains more frequent sharp-wave ripple events, and that place cells were more likely to fire spikes during ripples in these animals than control. These findings provide further evidence of desynchrony in MIA animals and may point to circuit-level changes that underlie failures to integrate and encode information in schizophrenia.
Collapse
Affiliation(s)
- Robert G K Munn
- Department of Anatomy, University of Otago, Dunedin, New Zealand
| | - Amy Wolff
- Department of Neuroscience and Medical Discovery Team on Addiction, University of Minnesota, Minneapolis, Minnesota, USA
| | - Lucinda J Speers
- Department of Psychology, University of Otago, Dunedin, New Zealand
| | - David K Bilkey
- Department of Psychology, University of Otago, Dunedin, New Zealand
| |
Collapse
|
2
|
Woods R, Lorusso J, Fletcher J, ElTaher H, McEwan F, Harris I, Kowash H, D'Souza SW, Harte M, Hager R, Glazier JD. Maternal immune activation and role of placenta in the prenatal programming of neurodevelopmental disorders. Neuronal Signal 2023; 7:NS20220064. [PMID: 37332846 PMCID: PMC10273029 DOI: 10.1042/ns20220064] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/16/2023] [Accepted: 05/17/2023] [Indexed: 06/20/2023] Open
Abstract
Maternal infection during pregnancy, leading to maternal immune activation (mIA) and cytokine release, increases the offspring risk of developing a variety of neurodevelopmental disorders (NDDs), including schizophrenia. Animal models have provided evidence to support these mechanistic links, with placental inflammatory responses and dysregulation of placental function implicated. This leads to changes in fetal brain cytokine balance and altered epigenetic regulation of key neurodevelopmental pathways. The prenatal timing of such mIA-evoked changes, and the accompanying fetal developmental responses to an altered in utero environment, will determine the scope of the impacts on neurodevelopmental processes. Such dysregulation can impart enduring neuropathological changes, which manifest subsequently in the postnatal period as altered neurodevelopmental behaviours in the offspring. Hence, elucidation of the functional changes that occur at the molecular level in the placenta is vital in improving our understanding of the mechanisms that underlie the pathogenesis of NDDs. This has notable relevance to the recent COVID-19 pandemic, where inflammatory responses in the placenta to SARS-CoV-2 infection during pregnancy and NDDs in early childhood have been reported. This review presents an integrated overview of these collective topics and describes the possible contribution of prenatal programming through placental effects as an underlying mechanism that links to NDD risk, underpinned by altered epigenetic regulation of neurodevelopmental pathways.
Collapse
Affiliation(s)
- Rebecca M. Woods
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| | - Jarred M. Lorusso
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| | - Jennifer Fletcher
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Medicine, Biology and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| | - Heidi ElTaher
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Medicine, Biology and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
- Department of Physiology, Faculty of Medicine, Alexandria University, Egypt
| | - Francesca McEwan
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| | - Isabella Harris
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| | - Hager M. Kowash
- Division of Developmental Biology and Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9WL, U.K
| | - Stephen W. D'Souza
- Division of Developmental Biology and Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9WL, U.K
| | - Michael Harte
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Medicine, Biology and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| | - Reinmar Hager
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| | - Jocelyn D. Glazier
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| |
Collapse
|
3
|
Abstract
PURPOSE OF REVIEW Schizophrenia is a psychiatric disorder that has a significant socioeconomic impact worldwide. Antipsychotic drugs targeting dopamine transmission alleviate psychotic symptoms but with limited efficacy and tolerability. Animal models have long proven useful for drug discovery. The continued need for new treatment highlights the importance of animal models to study schizophrenia. The lack of new therapeutic compounds combined with the shortcomings of clinical design studies potentially decreased the enthusiasm for animal model use. RECENT FINDINGS In the current review, we discuss the central role of animal models for schizophrenia in providing new insights into neurobiological features and therapeutic development. The US National Institute of Mental Health released the Research Domain Criteria to guide preclinical model studies. Here, we point out the advances of this approach and debate its potential limitations when using animal models to study schizophrenia from the drug discovery perspective. SUMMARY Cross-validated animal models for schizophrenia are crucial to comprehend the cause, pathophysiology, and behavioral and biological features of the disease, to advance prevention and treatment, and the need to carefully evaluate and select appropriate paradigms when investigating novel therapeutic targets.
Collapse
Affiliation(s)
- Daniela L Uliana
- Departments of Neuroscience, Psychiatry and Psychology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Felipe V Gomes
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Anthony A Grace
- Departments of Neuroscience, Psychiatry and Psychology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
4
|
Romero-Miguel D, Casquero-Veiga M, Fernández J, Lamanna-Rama N, Gómez-Rangel V, Gálvez-Robleño C, Santa-Marta C, Villar CJ, Lombó F, Abalo R, Desco M, Soto-Montenegro ML. Maternal Supplementation with N-Acetylcysteine Modulates the Microbiota-Gut-Brain Axis in Offspring of the Poly I:C Rat Model of Schizophrenia. Antioxidants (Basel) 2023; 12:970. [PMID: 37107344 PMCID: PMC10136134 DOI: 10.3390/antiox12040970] [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: 02/01/2023] [Revised: 04/03/2023] [Accepted: 04/04/2023] [Indexed: 04/29/2023] Open
Abstract
The microbiota-gut-brain axis is a complex interconnected system altered in schizophrenia. The antioxidant N-acetylcysteine (NAC) has been proposed as an adjunctive therapy to antipsychotics in clinical trials, but its role in the microbiota-gut-brain axis has not been sufficiently explored. We aimed to describe the effect of NAC administration during pregnancy on the gut-brain axis in the offspring from the maternal immune stimulation (MIS) animal model of schizophrenia. Pregnant Wistar rats were treated with PolyI:C/Saline. Six groups of animals were studied according to the study factors: phenotype (Saline, MIS) and treatment (no NAC, NAC 7 days, NAC 21 days). Offspring were subjected to the novel object recognition test and were scanned using MRI. Caecum contents were used for metagenomics 16S rRNA sequencing. NAC treatment prevented hippocampal volume reduction and long-term memory deficits in MIS-offspring. In addition, MIS-animals showed lower bacterial richness, which was prevented by NAC. Moreover, NAC7/NAC21 treatments resulted in a reduction of proinflammatory taxons in MIS-animals and an increase in taxa known to produce anti-inflammatory metabolites. Early approaches, like this one, with anti-inflammatory/anti-oxidative compounds, especially in neurodevelopmental disorders with an inflammatory/oxidative basis, may be useful in modulating bacterial microbiota, hippocampal size, as well as hippocampal-based memory impairments.
Collapse
Grants
- project number PI17/01766, and grant number BA21/00030 Ministerio de Ciencia e Innovación, Instituto de Salud Carlos III, co-financed by the European Regional Development Fund (ERDF), "A way to make Europe"
- project PID2021-128862OB-I00 MCIN /AEI /10.13039/501100011033 / FEDER, UE
- project number CB07/09/0031 CIBER de Salud Mental - Instituto de Salud Carlos III
- project numbers 2017/085, 2022/008917 Delegación del Gobierno para el Plan Nacional sobre Drogas
- 2016/01 Fundación Alicia Koplowitz
- grant, PEJD-2018-PRE/BMD-7899 Consejería de Educación e investigación, Comunidad de Madrid, co-funded by the European Social Fund "Investing in your future"
- "Programa Intramural de Impulso a la I+D+I 2019" Instituto de investigación Sanitaria Gregorio Marañón
- PT20/00044 Ministerio de Ciencia e Innovación, Instituto de Salud Carlos III
- x The CNIC is supported by the Instituto de Salud Carlos III (ISCIII), the Ministerio de Ciencia e Innovación (MCIN) and the Pro CNIC Foundation, and is a Severo Ochoa Center of Excellence (SEV-2015-0505)
- Contrato Intramural Postdoctoral FINBA
- SV-PA-21-AYUD/2021/51347 Ayudas para grupos de investigación de organismos del Principado de Asturias
Collapse
Affiliation(s)
| | | | - Javier Fernández
- Grupo de Investigación “Biotechnology in Nutraceuticals and Bioactive Compounds-BIONUC”, Departamento de Biología Funcional, Área de Microbiología, Universidad de Oviedo, 33006 Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), 33006 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Nicolás Lamanna-Rama
- Instituto de Investigación Sanitaria Gregorio Marañón, 28007 Madrid, Spain
- Departamento de Bioingeniería, Universidad Carlos III de Madrid, 28911 Madrid, Spain
| | | | - Carlos Gálvez-Robleño
- Grupo de Investigación de Alto Rendimiento en Fisiopatología y Farmacología del Sistema Digestivo (NeuGut-URJC), Universidad Rey Juan Carlos, 28922 Alcorcón, Spain
- Departamento de Ciencias Básicas de la Salud, Universidad Rey Juan Carlos (URJC), 28922 Alcorcón, Spain
| | - Cristina Santa-Marta
- Departamento de Física Matemática y de Fluidos, Universidad Nacional de Educación a Distancia (UNED), 28040 Madrid, Spain
| | - Claudio J. Villar
- Grupo de Investigación “Biotechnology in Nutraceuticals and Bioactive Compounds-BIONUC”, Departamento de Biología Funcional, Área de Microbiología, Universidad de Oviedo, 33006 Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), 33006 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Felipe Lombó
- Grupo de Investigación “Biotechnology in Nutraceuticals and Bioactive Compounds-BIONUC”, Departamento de Biología Funcional, Área de Microbiología, Universidad de Oviedo, 33006 Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), 33006 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Raquel Abalo
- Grupo de Investigación de Alto Rendimiento en Fisiopatología y Farmacología del Sistema Digestivo (NeuGut-URJC), Universidad Rey Juan Carlos, 28922 Alcorcón, Spain
- Departamento de Ciencias Básicas de la Salud, Universidad Rey Juan Carlos (URJC), 28922 Alcorcón, Spain
- Grupo de Trabajo de Ciencias Básicas en Dolor y Analgesia, Sociedad Española del Dolor (SED), 28046 Madrid, Spain
- Grupo de Trabajo de Cannabinoides, Sociedad Española del Dolor (SED), 28046 Madrid, Spain
- Unidad Asociada I+D+i del Instituto de Química Medica (IQM), Consejo Superior de Investigaciones Científicas (CSIC), 28006 Madrid, Spain
| | - Manuel Desco
- Instituto de Investigación Sanitaria Gregorio Marañón, 28007 Madrid, Spain
- Departamento de Bioingeniería, Universidad Carlos III de Madrid, 28911 Madrid, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), 28029 Madrid, Spain
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
| | - María Luisa Soto-Montenegro
- Instituto de Investigación Sanitaria Gregorio Marañón, 28007 Madrid, Spain
- Grupo de Investigación de Alto Rendimiento en Fisiopatología y Farmacología del Sistema Digestivo (NeuGut-URJC), Universidad Rey Juan Carlos, 28922 Alcorcón, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), 28029 Madrid, Spain
| |
Collapse
|
5
|
North HF, Weissleder C, Fullerton JM, Webster MJ, Weickert CS. Increased immune cell and altered microglia and neurogenesis transcripts in an Australian schizophrenia subgroup with elevated inflammation. Schizophr Res 2022; 248:208-218. [PMID: 36108465 DOI: 10.1016/j.schres.2022.08.025] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 07/18/2022] [Accepted: 08/27/2022] [Indexed: 11/16/2022]
Abstract
We previously identified a subgroup of schizophrenia cases (~40 %) with heightened inflammation in the neurogenic subependymal zone (SEZ) (North et al., 2021b). This schizophrenia subgroup had changes indicating reduced microglial activity, increased peripheral immune cells, increased stem cell dormancy/quiescence and reduced neuronal precursor cells. The present follow-up study aimed to replicate and extend those novel findings in an independent post-mortem cohort of schizophrenia cases and controls from Australia. RNA was extracted from SEZ tissue from 20 controls and 22 schizophrenia cases from the New South Wales Brain Tissue Resource Centre, and gene expression analysis was performed. Cluster analysis of inflammation markers (IL1B, IL1R1, SERPINA3 and CXCL8) revealed a high-inflammation schizophrenia subgroup comprising 52 % of cases, which was a significantly greater proportion than the 17 % of high-inflammation controls. Consistent with our previous report (North et al., 2021b), those with high-inflammation and schizophrenia had unchanged mRNA expression of markers for steady-state and activated microglia (IBA1, HEXB, CD68), decreased expression of phagocytic microglia markers (P2RY12, P2RY13), but increased expression of markers for macrophages (CD163), monocytes (CD14), natural killer cells (FCGR3A), and the adhesion molecule ICAM1. Similarly, the high-inflammation schizophrenia subgroup emulated increased quiescent stem cell marker (GFAPD) and decreased neuronal progenitor (DLX6-AS1) and immature neuron marker (DCX) mRNA expression; but also revealed a novel increase in a marker of immature astrocytes (VIM). Replicating primary results in an independent cohort demonstrates that inflammatory subgroups in the SEZ in schizophrenia are reliable, robust and enhance understanding of neuropathological heterogeneity when studying schizophrenia.
Collapse
Affiliation(s)
- Hayley F North
- Neuroscience Research Australia, Sydney, NSW, Australia; School of Psychiatry, Faculty of Medicine & Health, University of New South Wales, Sydney, NSW, Australia
| | - Christin Weissleder
- Neuroscience Research Australia, Sydney, NSW, Australia; German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany; Department of Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Janice M Fullerton
- Neuroscience Research Australia, Sydney, NSW, Australia; School of Medical Sciences, Faculty of Medicine & Health, University of New South Wales, Sydney, NSW, Australia
| | - Maree J Webster
- Laboratory of Brain Research, Stanley Medical Research Institute, 9800 Medical Center Drive, Rockville, MD, USA
| | - Cynthia Shannon Weickert
- Neuroscience Research Australia, Sydney, NSW, Australia; School of Psychiatry, Faculty of Medicine & Health, University of New South Wales, Sydney, NSW, Australia; Department of Neuroscience and Physiology, Upstate Medical University, Syracuse, NY, USA.
| |
Collapse
|
6
|
Infection of the murine placenta by Listeria monocytogenes induces sex-specific responses in the fetal brain. Pediatr Res 2022; 93:1566-1573. [PMID: 36127406 DOI: 10.1038/s41390-022-02307-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 08/06/2022] [Accepted: 08/30/2022] [Indexed: 11/09/2022]
Abstract
BACKGROUND Epidemiological data indicate that prenatal infection is associated with an increased risk of several neurodevelopmental disorders in the progeny. These disorders display sex differences in presentation. The role of the placenta in the sex-specificity of infection-induced neurodevelopmental abnormalities is not well-defined. We used an imaging-based animal model of the bacterial pathogen Listeria monocytogenes to identify sex-specific effects of placental infection on neurodevelopment of the fetus. METHODS Pregnant CD1 mice were infected with a bioluminescent strain of Listeria on embryonic day 14.5 (E14.5). Excised fetuses were imaged on E18.5 to identify the infected placentas. The associated fetal brains were analyzed for gene expression and altered brain structure due to infection. The behavior of adult offspring affected by prenatal Listeria infection was analyzed. RESULTS Placental infection induced sex-specific alteration of gene expression patterns in the fetal brain and resulted in abnormal cortical development correlated with placental infection levels. Furthermore, male offspring exhibited abnormal social interaction, whereas females exhibited elevated anxiety. CONCLUSION Placental infection by Listeria induced sex-specific abnormalities in neurodevelopment of the fetus. Prenatal infection also affected the behavior of the offspring in a sex-specific manner. IMPACT Placental infection with Listeria monocytogenes induces sexually dichotomous gene expression patterns in the fetal brains of mice. Abnormal cortical lamination is correlated with placental infection levels. Placental infection results in autism-related behavior in male offspring and heightened anxiety levels in female offspring.
Collapse
|
7
|
Uliana DL, Zhu X, Gomes FV, Grace AA. Using animal models for the studies of schizophrenia and depression: The value of translational models for treatment and prevention. Front Behav Neurosci 2022; 16:935320. [PMID: 36090659 PMCID: PMC9449416 DOI: 10.3389/fnbeh.2022.935320] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 08/04/2022] [Indexed: 11/29/2022] Open
Abstract
Animal models of psychiatric disorders have been highly effective in advancing the field, identifying circuits related to pathophysiology, and identifying novel therapeutic targets. In this review, we show how animal models, particularly those based on development, have provided essential information regarding circuits involved in disorders, disease progression, and novel targets for intervention and potentially prevention. Nonetheless, in recent years there has been a pushback, largely driven by the US National Institute of Mental Health (NIMH), to shift away from animal models and instead focus on circuits in normal subjects. This has been driven primarily from a lack of discovery of new effective therapeutic targets, and the failure of targets based on preclinical research to show efficacy. We discuss why animal models of complex disorders, when strongly cross-validated by clinical research, are essential to understand disease etiology as well as pathophysiology, and direct new drug discovery. Issues related to shortcomings in clinical trial design that confound translation from animal models as well as the failure to take patient pharmacological history into account are proposed to be a source of the failure of what are likely effective compounds from showing promise in clinical trials.
Collapse
Affiliation(s)
- Daniela L. Uliana
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Xiyu Zhu
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, United States
| | - Felipe V. Gomes
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Anthony A. Grace
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
8
|
Maternal immune activation in rats induces dysfunction of placental leucine transport and alters fetal brain growth. Clin Sci (Lond) 2022; 136:1117-1137. [PMID: 35852150 PMCID: PMC9366863 DOI: 10.1042/cs20220245] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 07/15/2022] [Accepted: 07/19/2022] [Indexed: 11/17/2022]
Abstract
Maternal infection during pregnancy increases the offspring risk of developing a variety of neurodevelopmental disorders (NDDs), including schizophrenia. While the mechanisms remain unclear, dysregulation of placental function is implicated. We hypothesised that maternal infection, leading to maternal immune activation and stimulated cytokine production, alters placental and yolk sac amino acid transport, affecting fetal brain development and thus NDD risk. Using a rat model of maternal immune activation induced by the viral mimetic polyinosinic:polycytidylic acid (poly(I:C)), we investigated placental and yolk sac expression of system L amino acid transporter subtypes which transport several essential amino acids including branched-chain amino acids (BCAA), maternal and fetal BCAA concentration, placental 14C-leucine transport activity and associated impacts on fetal growth and development. Poly(I:C) treatment increased acutely maternal IL-6 and TNFα concentration, contrasting with IL-1β. Transcriptional responses for these pro-inflammatory cytokines were found in placenta and yolk sac following poly(I:C) treatment. Placental and yolk sac weights were reduced by poly(I:C) treatment, yet fetal body weight was unaffected, while fetal brain weight was increased. Maternal plasma BCAA concentration was reduced 24 h post-poly(I:C) treatment, yet placental, but not yolk sac, BCAA concentration was increased. Placental and yolk sac gene expression of Slc7a5, Slc7a8 and Slc43a2 encoding LAT1, LAT2 and LAT4 transporter subtypes respectively, was altered by poly(I:C) treatment. Placental 14C-leucine transport was significantly reduced 24 h post-treatment, contrasting with a significant increase six days following poly(I:C) treatment. Maternal immune activation induces dysregulated placental transport of amino acids affecting fetal brain development, and NDD risk potential in offspring.
Collapse
|
9
|
Speers LJ, Schmidt R, Bilkey DK. Aberrant Phase Precession of Lateral Septal Cells in a Maternal Immune Activation Model of Schizophrenia Risk May Disrupt the Integration of Location with Reward. J Neurosci 2022; 42:4187-4201. [PMID: 35396329 PMCID: PMC9121831 DOI: 10.1523/jneurosci.0039-22.2022] [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: 12/22/2021] [Revised: 03/16/2022] [Accepted: 04/03/2022] [Indexed: 11/21/2022] Open
Abstract
Spatial memory and reward processing are known to be disrupted in schizophrenia. Since the lateral septum (LS) may play an important role in the integration of location and reward, we examined the effect of maternal immune activation (MIA), a known schizophrenia risk factor, on spatial representation in the rat LS. In support of a previous study, we found that spatial location is represented as a phase code in the rostral LS of adult male rats, so that LS cell spiking shifts systematically against the phase of the hippocampal, theta-frequency, local field potential as an animal moves along a track toward a reward (phase precession). Whereas shallow precession slopes were observed in control group cells, they were steeper in the MIA animals, such that firing frequently precessed across several theta cycles as the animal moved along the length of the apparatus, with subsequent ambiguity in the phase representation of location. Furthermore, an analysis of the phase trajectories of the control group cells revealed that the population tended to converge toward a common firing phase as the animal approached the reward location. This suggested that phase coding in these cells might signal both reward location and the distance to reward. By comparison, the degree of phase convergence in the MIA-group cells was weak, and the region of peak convergence was distal to the reward location. These findings suggest that a schizophrenia risk factor disrupts the phase-based encoding of location-reward relationships in the LS, potentially smearing reward representations across space.SIGNIFICANCE STATEMENT It is unclear how spatial or contextual information generated by hippocampal cells is converted to a code that can be used to signal reward location in regions, such as the VTA. Here we provide evidence that the firing phase of cells in the lateral septum, a region that links the two areas, may code reward location in the firing phase of cells. This phase coding is disrupted in a maternal immune activation model of schizophrenia risk such that representations of reward may be smeared across space in maternal immune activation animals. This could potentially underlie erroneous reward processing and misattribution of salience in schizophrenia.
Collapse
Affiliation(s)
- Lucinda J Speers
- Psychology Department, Otago University, Dunedin 9054, New Zealand
| | - Robert Schmidt
- Psychology Department, University of Sheffield, Sheffield, United Kingdom
| | - David K Bilkey
- Psychology Department, Otago University, Dunedin 9054, New Zealand
| |
Collapse
|
10
|
Liu R, Tang W, Wang W, Xu F, Fan W, Zhang Y, Zhang C. NLRP3 Influences Cognitive Function in Schizophrenia in Han Chinese. Front Genet 2021; 12:781625. [PMID: 34956329 PMCID: PMC8702823 DOI: 10.3389/fgene.2021.781625] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 11/19/2021] [Indexed: 12/14/2022] Open
Abstract
It has been proposed that immune abnormalities may be implicated with pathophysiology of schizophrenia. The nod-like receptor pyrin domain-contraining protein 3 (NLRP3) can trigger immune-inflammatory cascade reactions. In this study, we intended to identify the role of gene encoding NLRP3 (NLRP3) in susceptibility to schizophrenia and its clinical features. For the NLRP3 mRNA expression analysis, 53 drug-naïve patients with first-episode schizophrenia and 56 healthy controls were enrolled. For the genetic study, a total of 823 schizophrenia patients and 859 controls were recruited. Among them, 239 drug-naïve patients with first-episode schizophrenia were enrolled for clinical evaluation. There is no significant difference in NLRP3 mRNA levels between patients with schizophrenia and healthy controls (p = 0.07). We did not observe any significant differences in allele and genotype frequencies of rs10754558 polymorphism between the schizophrenia and control groups. We noticed significant differences in the scores of RBANS attention and total scores between the patients with different genotypes of rs10754558 polymorphism (p = 0.001 and p < 0.01, respectively). Further eQTL analysis presented a significant association between the rs10754558 polymorphism and NLRP3 in frontal cortex (p = 0.0028, p = 0.028 after Bonferroni correction). Although our findings did not support NLRP3 confer susceptibility to schizophrenia, NLRP3 may be a risk factor for cognitive impairment, especially attention deficit in this disorder.
Collapse
Affiliation(s)
- Ruimei Liu
- Schizophrenia Program, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Tang
- Department of Psychiatry, The Affiliated Kangning Hospital of Wenzhou Medical University, Wenzhou, China
| | - Weiping Wang
- Department of Psychiatry, Jinhua Second Hospital, Jinhua, China
| | - Feikang Xu
- Schizophrenia Program, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weixing Fan
- Department of Psychiatry, Jinhua Second Hospital, Jinhua, China
| | - Yi Zhang
- Schizophrenia Program, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen Zhang
- Schizophrenia Program, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
11
|
Speers LJ, Bilkey DK. Disorganization of Oscillatory Activity in Animal Models of Schizophrenia. Front Neural Circuits 2021; 15:741767. [PMID: 34675780 PMCID: PMC8523827 DOI: 10.3389/fncir.2021.741767] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 09/16/2021] [Indexed: 01/02/2023] Open
Abstract
Schizophrenia is a chronic, debilitating disorder with diverse symptomatology, including disorganized cognition and behavior. Despite considerable research effort, we have only a limited understanding of the underlying brain dysfunction. In this article, we review the potential role of oscillatory circuits in the disorder with a particular focus on the hippocampus, a region that encodes sequential information across time and space, as well as the frontal cortex. Several mechanistic explanations of schizophrenia propose that a loss of oscillatory synchrony between and within these brain regions may underlie some of the symptoms of the disorder. We describe how these oscillations are affected in several animal models of schizophrenia, including models of genetic risk, maternal immune activation (MIA) models, and models of NMDA receptor hypofunction. We then critically discuss the evidence for disorganized oscillatory activity in these models, with a focus on gamma, sharp wave ripple, and theta activity, including the role of cross-frequency coupling as a synchronizing mechanism. Finally, we focus on phase precession, which is an oscillatory phenomenon whereby individual hippocampal place cells systematically advance their firing phase against the background theta oscillation. Phase precession is important because it allows sequential experience to be compressed into a single 120 ms theta cycle (known as a 'theta sequence'). This time window is appropriate for the induction of synaptic plasticity. We describe how disruption of phase precession could disorganize sequential processing, and thereby disrupt the ordered storage of information. A similar dysfunction in schizophrenia may contribute to cognitive symptoms, including deficits in episodic memory, working memory, and future planning.
Collapse
Affiliation(s)
| | - David K. Bilkey
- Department of Psychology, Otago University, Dunedin, New Zealand
| |
Collapse
|
12
|
Speers LJ, Cheyne KR, Cavani E, Hayward T, Schmidt R, Bilkey DK. Hippocampal Sequencing Mechanisms Are Disrupted in a Maternal Immune Activation Model of Schizophrenia Risk. J Neurosci 2021; 41:6954-6965. [PMID: 34253630 PMCID: PMC8360689 DOI: 10.1523/jneurosci.0730-21.2021] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 06/07/2021] [Accepted: 07/04/2021] [Indexed: 01/02/2023] Open
Abstract
Episodic memory requires information to be stored and recalled in sequential order, and these processes are disrupted in schizophrenia. Hippocampal phase precession and theta sequences are thought to provide a biological mechanism for sequential ordering of experience at timescales suitable for plasticity. These phenomena have not previously been examined in any models of schizophrenia risk. Here, we examine these phenomena in a maternal immune activation (MIA) rodent model. We show that while individual pyramidal cells in the CA1 region continue to precess normally in MIA animals, the starting phase of precession as an animal enters a new place field is considerably more variable in MIA animals than in controls. A critical consequence of this change is a disorganization of the ordered representation of experience via theta sequences. These results provide the first evidence of a biological-level mechanism that, if it occurs in schizophrenia, may explain aspects of disorganized sequential processing that contribute to the cognitive symptoms of the disorder.SIGNIFICANCE STATEMENT Hippocampal phase precession and theta sequences have been proposed as biophysical mechanisms by which the sequential structure of cognition might be ordered. Disturbances of sequential processing have frequently been observed in schizophrenia. Here, we show for the first time that phase precession and theta sequences are disrupted in a maternal immune activation (MIA) model of schizophrenia risk. This is a result of greater variability in the starting phase of precession, indicating that the mechanisms that coordinate precession at the assembly level are disrupted. We propose that this disturbance in phase precession underlies some of the disorganized cognitive symptoms that occur in schizophrenia. These findings could have important preclinical significance for the identification and treatment of schizophrenia risk factors.
Collapse
Affiliation(s)
- Lucinda J Speers
- Psychology Department, Otago University, Dunedin 9016, New Zealand
| | - Kirsten R Cheyne
- Psychology Department, Otago University, Dunedin 9016, New Zealand
| | - Elena Cavani
- Psychology Department, Otago University, Dunedin 9016, New Zealand
- University of Tübingen, Tübingen 72076, Germany
| | - Tara Hayward
- Psychology Department, Otago University, Dunedin 9016, New Zealand
| | - Robert Schmidt
- Psychology Department, University of Sheffield, Sheffield, S10 2TN, United Kingdom
| | - David K Bilkey
- Psychology Department, Otago University, Dunedin 9016, New Zealand
| |
Collapse
|
13
|
Quagliato LA, de Matos U, Nardi AE. Maternal immune activation generates anxiety in offspring: A translational meta-analysis. Transl Psychiatry 2021; 11:245. [PMID: 33903587 PMCID: PMC8076195 DOI: 10.1038/s41398-021-01361-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 04/12/2021] [Indexed: 02/07/2023] Open
Abstract
Maternal immune activation (MIA) during pregnancy is recognized as an etiological risk factor for various psychiatric disorders, such as schizophrenia, major depressive disorder, and autism. Prenatal immune challenge may serve as a "disease primer" for alteration of the trajectory of fetal brain development that, in combination with other genetic and environmental factors, may ultimately result in the emergence of different psychiatric conditions. However, the association between MIA and an offspring's chance of developing anxiety disorders is less clear. To evaluate the effect of MIA on offspring anxiety, a systematic review and meta-analysis of the preclinical literature was conducted. We performed a systematic search of the PubMed, Web of Science, PsycINFO, and Cochrane Library electronic databases using the PRISMA and World Health Organization (WHO) methodologies for systematic reviews. Studies that investigated whether MIA during pregnancy could cause anxiety symptoms in rodent offspring were included. Overall, the meta-analysis showed that MIA induced anxiety behavior in offspring. The studies provide strong evidence that prenatal immune activation impacts specific molecular targets and synapse formation and function and induces an imbalance in neurotransmission that could be related to the generation of anxiety in offspring. Future research should further explore the role of MIA in anxiety endophenotypes. According to this meta-analysis, MIA plays an important role in the pathophysiological mechanisms of anxiety disorders and is a promising therapeutic target.
Collapse
Affiliation(s)
- Laiana A Quagliato
- Laboratory of Panic & Respiration, Institute of Psychiatry, Federal University of Rio de Janeiro, 22270-010, Rio de Janeiro, Brazil.
| | - Ursula de Matos
- Laboratory of Panic & Respiration, Institute of Psychiatry, Federal University of Rio de Janeiro, 22270-010, Rio de Janeiro, Brazil
| | - Antonio E Nardi
- Laboratory of Panic & Respiration, Institute of Psychiatry, Federal University of Rio de Janeiro, 22270-010, Rio de Janeiro, Brazil
| |
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
|
Maternal Immune Activation Causes Schizophrenia-like Behaviors in the Offspring through Activation of Immune-Inflammatory, Oxidative and Apoptotic Pathways, and Lowered Antioxidant Defenses and Neuroprotection. Mol Neurobiol 2020; 57:4345-4361. [PMID: 32720073 DOI: 10.1007/s12035-020-02028-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 07/14/2020] [Indexed: 12/13/2022]
Abstract
Schizophrenia is a complex neuropsychiatric disorder, influenced by a combined action of genes and environmental factors. The neurodevelopmental origin is one of the most widely recognized etiological models of this heterogeneous disorder. Environmental factors, especially infections during gestation, appear to be a major risk determinant of neurodevelopmental basis of schizophrenia. Prenatal infection may cause maternal immune activation (MIA) and enhance risk of schizophrenia in the offspring. However, the precise mechanistic basis through which MIA causes long-lasting schizophrenia-like behavioral deficits in offspring remains inadequately understood. Herein, we aimed to delineate whether prenatal infection-induced MIA causes schizophrenia-like behaviors through its long-lasting effects on immune-inflammatory and apoptotic pathways, oxidative stress toxicity, and antioxidant defenses in the brain of offspring. Sprague-Dawley rats were divided into three groups (n = 15/group) and were injected with poly (I:C), LPS, and saline at gestational day (GD)-12. Except IL-1β, plasma levels of IL-6, TNF-α, and IL-17A assessed after 24 h were significantly elevated in both the poly (I:C)- and LPS-treated pregnant rats, indicating MIA. The rats born to dams treated with poly (I:C) and LPS displayed increased anxiety-like behaviors and significant deficits in social behaviors. Furthermore, the hippocampus of the offspring rats of both the poly (I:C)- and LPS-treated groups showed increased signs of lipid peroxidation, diminished total antioxidant content, and differentially upregulated expression of inflammatory (TNFα, IL6, and IL1β), and apoptotic (Bax, Cas3, and Cas9) genes but decreased expression of neuroprotective (BDNF and Bcl2) genes. The results suggest long-standing effects of prenatal infections on schizophrenia-like behavioral deficits, which are mediated by immune-inflammatory and apoptotic pathways, increased oxidative stress toxicity, and lowered antioxidant and neuroprotective defenses. The findings suggest that prenatal infections may underpin neurodevelopmental aberrations and neuroprogression and subsequently schizophrenia-like symptoms.
Collapse
|
16
|
Haddad FL, Patel SV, Schmid S. Maternal Immune Activation by Poly I:C as a preclinical Model for Neurodevelopmental Disorders: A focus on Autism and Schizophrenia. Neurosci Biobehav Rev 2020; 113:546-567. [PMID: 32320814 DOI: 10.1016/j.neubiorev.2020.04.012] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 01/28/2020] [Accepted: 04/09/2020] [Indexed: 12/18/2022]
Abstract
Maternal immune activation (MIA) in response to a viral infection during early and mid-gestation has been linked through various epidemiological studies to a higher risk for the child to develop autism or schizophrenia-related symptoms.. This has led to the establishment of the pathogen-free poly I:C-induced MIA animal model for neurodevelopmental disorders, which shows relatively high construct and face validity. Depending on the experimental variables, particularly the timing of poly I:C administration, different behavioural and molecular phenotypes have been described that relate to specific symptoms of neurodevelopmental disorders such as autism spectrum disorder and/or schizophrenia. We here review and summarize epidemiological evidence for the effects of maternal infection and immune activation, as well as major findings in different poly I:C MIA models with a focus on poly I:C exposure timing, behavioural and molecular changes in the offspring, and characteristics of the model that relate it to autism spectrum disorder and schizophrenia.
Collapse
Affiliation(s)
- Faraj L Haddad
- Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON, Canada.
| | - Salonee V Patel
- Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON, Canada.
| | - Susanne Schmid
- Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON, Canada.
| |
Collapse
|
17
|
Xu R, Wu B, Liang J, He F, Gu W, Li K, Luo Y, Chen J, Gao Y, Wu Z, Wang Y, Zhou W, Wang M. Altered gut microbiota and mucosal immunity in patients with schizophrenia. Brain Behav Immun 2020; 85:120-127. [PMID: 31255682 DOI: 10.1016/j.bbi.2019.06.039] [Citation(s) in RCA: 103] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 06/26/2019] [Accepted: 06/26/2019] [Indexed: 10/26/2022] Open
Abstract
Evidence shows that gut microbiota may play important roles in schizophrenia pathogenesis via the "gut-brain" axis, but the mechanisms remain unclear. Here, eighty-four patients with schizophrenia and 84 sex- and age-matched healthy controls were enrolled. Shotgun metagenomic sequencing and 16S rRNA sequencing were performed, and the gut microbiota-associated epitopes (MEs) were predicted, which, together with IgA content, were used to determine the gut microbiota composition associated with gut immune status. Patients with schizophrenia had significantly reduced gut microbiota richnesses compared with those of the healthy controls, and the gut microbiota compositions clearly distinguished the patients with schizophrenia from the healthy controls. Based on two-stage metagenomic-wide association studies, nineteen gut microbiota taxonomies were associated with schizophrenia, and the microbial dysbiosis (MD) index was calculated based on the abundance of differential taxonomies. We found that MD index was positively correlated with MEs diversity and gut IgA levels, and negatively correlated with gut microbiota richness. Glutamate synthase (GOGAT) was more active in the guts of patients with schizophrenia than in those of healthy controls, and high GOGAT activity was associated with altered gut microbiota taxonomies associated with gut IgA levels. Our results may imply a role of the microbiome in the etiology of schizophrenia and contribute to the development of microbiome targeted interventions for schizophrenia.
Collapse
Affiliation(s)
- Ruihuan Xu
- Clinical Laboratory, Longgang Distric Central Hospital, Affliated to Guangzhou University of Traditional Chinese Medicine, Shenzhen, Guangdong 518116, China.
| | - Bingbing Wu
- Shanghai Key Laboratory of Birth Defects, The Molecular Medical Center, Pediatrics Research Institute, Children's Hospital of Fudan University, National Center for Children's Health, Shanghai 201102, China
| | - Jingwen Liang
- Clinical Laboratory, Longgang Distric Central Hospital, Affliated to Guangzhou University of Traditional Chinese Medicine, Shenzhen, Guangdong 518116, China
| | - Fusheng He
- Imunobio, Shenzhen, Guangdong 518001, China
| | - Wen Gu
- Shenzhen Key Laboratory for Psychological Healthcare, Shenzhen Institute of Mental Health, Shenzhen Kangning Hospital, Shenzhen Mental Health Center, Shenzhen 518020, China
| | - Kang Li
- Clinical Laboratory, Longgang Distric Central Hospital, Affliated to Guangzhou University of Traditional Chinese Medicine, Shenzhen, Guangdong 518116, China
| | - Yi Luo
- Clinical Laboratory, Longgang Distric Central Hospital, Affliated to Guangzhou University of Traditional Chinese Medicine, Shenzhen, Guangdong 518116, China
| | - Jianxia Chen
- Clinical Laboratory, Longgang Distric Central Hospital, Affliated to Guangzhou University of Traditional Chinese Medicine, Shenzhen, Guangdong 518116, China
| | - Yongbo Gao
- Clinical Laboratory, Longgang Distric Central Hospital, Affliated to Guangzhou University of Traditional Chinese Medicine, Shenzhen, Guangdong 518116, China
| | - Ze Wu
- Clinical Laboratory, Longgang Hand Surgery Hospital of Shenzhen, 518116 Guangdong, China
| | - Yongqiang Wang
- Shenzhen Following Precision Medical Research Institute, Luohu Hospital Group, Shenzhen 518000, China
| | - Wenhao Zhou
- Shanghai Key Laboratory of Birth Defects, Division of Neonatology, Children's Hospital of Fudan University, National Center for Children's Health, Shanghai 201102, China.
| | - Mingbang Wang
- Shanghai Key Laboratory of Birth Defects, Division of Neonatology, Children's Hospital of Fudan University, National Center for Children's Health, Shanghai 201102, China.
| |
Collapse
|
18
|
Discrimination difficulty, cognitive burden, and reversal impairments in a maternal immune activation model of schizophrenia risk. Behav Processes 2019; 167:103936. [DOI: 10.1016/j.beproc.2019.103936] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 07/22/2019] [Accepted: 08/01/2019] [Indexed: 12/14/2022]
|
19
|
Maternal Immune Activation during Pregnancy Alters the Behavior Profile of Female Offspring of Sprague Dawley Rats. eNeuro 2019; 6:eN-NWR-0437-18. [PMID: 31016229 PMCID: PMC6477592 DOI: 10.1523/eneuro.0437-18.2019] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 03/25/2019] [Accepted: 03/26/2019] [Indexed: 12/30/2022] Open
Abstract
Sex differences are documented in psychiatric and neurological disorders, yet most preclinical animal research has been conducted in males only. There is a need to better understand of the nature of sex differences in brain disease in order to meet the needs of psychiatric patients. We present the behavior profile of adult female offspring produced using a maternal immune activation (MIA) model where pregnant rats receive an immune stimulant and the offspring typically show various abnormalities consistent with psychiatric illnesses such as schizophrenia and autism. The results in female offspring were compared to a previously published cohort of their male siblings (Lins et al., 2018). We examined prepulse inhibition (PPI), sociability, MK-801-induced locomotor activity, crossmodal object recognition (CMOR), and oddity discrimination; behaviors relevant to the positive, negative, and cognitive symptoms of schizophrenia. No between-treatment differences in PPI or locomotor activity were noted. Tactile memory was observed in the control and treated female offspring, visual recognition memory was deficient in the polyinosinic:polycytidylic acid (polyI:C) offspring only, and both groups lacked crossmodal recognition. PolyI:C offspring were impaired in oddity preference and had reduced preference for a stranger conspecific in a sociability assay. Systemic maternal CXCL1, IL-6, and TNF-a levels 3 h after polyI:C treatment were determined, but no relationship was found between these cytokines and the behavior seen in the adult female offspring. Overall, female offspring of polyI:C-treated dams display an array of behavior abnormalities relevant to psychiatric illnesses such as schizophrenia similar to those previously reported in male rats.
Collapse
|
20
|
McColl ER, Piquette-Miller M. Poly(I:C) alters placental and fetal brain amino acid transport in a rat model of maternal immune activation. Am J Reprod Immunol 2019; 81:e13115. [PMID: 30924965 DOI: 10.1111/aji.13115] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 03/18/2019] [Accepted: 03/19/2019] [Indexed: 12/13/2022] Open
Abstract
PROBLEM Maternal immune activation (MIA) during pregnancy is associated with increased chances of neurodevelopmental disorders including schizophrenia and autism spectrum disorder (ASD). However, the exact mechanism through which MIA contributes to altered neurodevelopment is unknown. Due to the important role that amino acids play in neurodevelopment, altered amino acid transport could play a role in neurodevelopmental disorders. Indeed, altered plasma concentrations of multiple amino acids have been reported in individuals with ASD or schizophrenia. Therefore, our objective was to determine whether virally mediated MIA induces changes in amino acid transporters in the placenta and fetal brain. METHOD OF STUDY Pregnant rats were administered poly(I:C) on gestational day 14, and placental and fetal tissues were collected 6, 24, and 48 hours later. Amino acid transporter expression was measured in the placenta and fetal brain using qPCR, Western blotting, and Simple Western. Free amino acid concentrations in the fetal brain were quantified using HPLC. RESULTS Poly(I:C) increased mRNA expression of several amino acid transporters in the placenta and fetal brain over these timepoints. Conversely, poly(I:C) imposed significant decreases in the protein expression of ASCT1 and EAAT2 in placenta and expression of SNAT5, EAAT1, and GLYT1 in fetal brain. Functional consequences of altered transporter expression were demonstrated through widespread changes in the concentrations of free amino acids in the fetal brains. CONCLUSION Together, these results represent novel findings with the poly(I:C) MIA model and contribute to the understanding of how MIA during pregnancy potentially leads to neurodevelopmental disorders.
Collapse
Affiliation(s)
- Eliza R McColl
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Micheline Piquette-Miller
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
21
|
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]
|
22
|
Glass R, Norton S, Fox N, Kusnecov AW. Maternal immune activation with staphylococcal enterotoxin A produces unique behavioral changes in C57BL/6 mouse offspring. Brain Behav Immun 2019; 75:12-25. [PMID: 29772261 DOI: 10.1016/j.bbi.2018.05.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 04/27/2018] [Accepted: 05/04/2018] [Indexed: 12/25/2022] Open
Abstract
Stimulation of the immune system during pregnancy, known as maternal immune activation (MIA), can cause long-lasting neurobiological and behavioral changes in the offspring. This phenomenon has been implicated in the etiology of developmental psychiatric disorders, such as autism and schizophrenia. Much of this evidence is predicated on animal models using bacterial agents such as LPS and/or viral mimics such as Poly I:C, both of which act through toll-like receptors. However, fewer studies have examined the role of direct activation of maternal T-cells during pregnancy using microbial agents. Bacterial superantigens, such as Staphylococcal Enterotoxin A and B (SEA; SEB), are microbial proteins that activate CD4+ T-cells and cause prominent T-cell proliferation and cytokine production. We injected pregnant and non-pregnant adult female C57BL/6 mice with 200 μg/Kg of SEA, SEB, or 0.9% saline, and measured splenic T-cell-derived cytokine concentrations (viz., IL-2, IFN-γ, IL-6, and IL-4) 2 h later; animals injected with SEA were also measured for splenic concentrations of TNF-α and IL-17A. Half of the injected pregnant animals were brought to term, and their offspring were tested on a series of behavioral tasks starting at six weeks of age (postnatal day 42 [P42]). These tasks included social interaction, the elevated plus maze (EPM), an open field and object recognition (OR) task, prepulse inhibition (PPI) of sensorimotor gating, and the Morris water maze (MWM). Results showed that SEA and SEB induced significant concentrations of all measured cytokines, and in particular IFN-γ, although cytokine responses were greater following SEA exposure. In addition, pregnancy induced an inhibitory effect on cytokine production. Behavioral results showed distinct phenotypes among offspring from SEA- or SEB-injected mothers, very likely due to differences in the magnitude of cytokines generated in response to each toxin. Offspring from SEA-injected mothers displayed modest decreases in social behavior, but increased anxiety, locomotion, interest in a novel object, and short-term spatial memory, while offspring of SEB-injected mothers only exhibited increased anxiety and locomotion. There were no deficits in PPI, which was actually pronounced in SEA and SEB offspring. Overall, the novel use of SEA and SEB as prenatal immune challenges elicited distinct behavioral profiles in the offspring that both mirrors and diverges from previous models of maternal immune activation in important ways. We conclude that superantigen-induced T-cell-mediated maternal immune activation is a valid and valuable model for studying and expanding our understanding of the effects of prenatal immune challenge on neurodevelopmental and behavioral alterations in offspring.
Collapse
Affiliation(s)
- Ruthy Glass
- Rutgers University, 152 Frelinghuysen Rd, Piscataway, NJ 08854, USA.
| | - Sara Norton
- Rutgers University, 152 Frelinghuysen Rd, Piscataway, NJ 08854, USA
| | - Nicholas Fox
- Rutgers University, 152 Frelinghuysen Rd, Piscataway, NJ 08854, USA
| | | |
Collapse
|
23
|
Careaga M, Taylor SL, Chang C, Chiang A, Ku KM, Berman RF, Van de Water JA, Bauman MD. Variability in PolyIC induced immune response: Implications for preclinical maternal immune activation models. J Neuroimmunol 2018; 323:87-93. [PMID: 30196839 DOI: 10.1016/j.jneuroim.2018.06.014] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 06/19/2018] [Accepted: 06/25/2018] [Indexed: 12/18/2022]
Abstract
Maternal infection during pregnancy may increase the risk of offspring neurodevelopmental disorders. The preclinical Polyinosinic-polycytidylic acid (PolyIC) model has become one of the most widely used approaches in maternal immune activation (MIA) research. However, variability in molecular weight may impact the immune activating potential of PolyIC. Nulliparous rats injected with high molecular weight PolyIC exhibit pronounced cytokine response and sickness behavior that was not observed in rats injected low molecular weight PolyIC. Although an essential next step is to extend these studies to pregnant animals, the preliminary results suggest that PolyIC molecular weight is an important experimental design consideration.
Collapse
Affiliation(s)
- Milo Careaga
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, USA; The MIND Institute, University of California, Davis, USA
| | - Sandra L Taylor
- Department of Public Health Sciences, University of California, Davis, USA
| | - Carolyn Chang
- The MIND Institute, University of California, Davis, USA
| | - Alex Chiang
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, USA; The MIND Institute, University of California, Davis, USA
| | - Katherine M Ku
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, USA; The MIND Institute, University of California, Davis, USA
| | - Robert F Berman
- The MIND Institute, University of California, Davis, USA; Department of Neurological Surgery, University of California, Davis, USA
| | - Judy A Van de Water
- The MIND Institute, University of California, Davis, USA; Internal Medicine, Division of Rheumatology, University of California, Davis, USA
| | - Melissa D Bauman
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, USA; The MIND Institute, University of California, Davis, USA; California National Primate Research Center, University of California, Davis, USA.
| |
Collapse
|
24
|
Zhang J, Jing Y, Zhang H, Bilkey DK, Liu P. Effects of maternal immune activation on brain arginine metabolism of postnatal day 2 rat offspring. Schizophr Res 2018; 192:431-441. [PMID: 28526281 DOI: 10.1016/j.schres.2017.05.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 05/10/2017] [Accepted: 05/13/2017] [Indexed: 12/19/2022]
Abstract
l-Arginine is a versatile semi-essential amino acid with a number of bioactive metabolites, and altered arginine metabolism has been implicated in the pathogenesis of schizophrenia. Earlier research has demonstrated that maternal immune activation (MIA; a risk factor for schizophrenia) alters arginine metabolism in the prefrontal cortex and hippocampus of the adult offspring. The present study investigated how MIA affected the levels of l-arginine and its downstream metabolites in the whole forebrain, frontal cortex, hippocampus and cerebellum of male and female rat offspring at the age of postnatal day 2. While no effects were evident in the forebrain, MIA significantly increased l-arginine, glutamate, putrescine, spermidine and spermine levels and the glutamate/GABA ratio, but decreased the glutamine/glutamate ratio, in the frontal cortex, hippocampus and/or cerebellum with no marked sex differences. Cluster analyses revealed that l-arginine and its main metabolites formed distinct groups, which changed as a function of MIA or sex in all four brain regions examined. These results demonstrate, for the first time, that MIA alters brain arginine metabolism in the rat offspring during early neonatal development, and further support the involvement of arginine metabolism in the pathogenesis of schizophrenia.
Collapse
Affiliation(s)
- Jiaxian Zhang
- Department of Anatomy, University of Otago, Dunedin, New Zealand; Brain Health and Research Centre, University of Otago, Dunedin, New Zealand
| | - Yu Jing
- Department of Anatomy, University of Otago, Dunedin, New Zealand; Brain Health and Research Centre, University of Otago, Dunedin, New Zealand
| | - Hu Zhang
- School of Pharmacy, University of Otago, Dunedin, New Zealand; Brain Health and Research Centre, University of Otago, Dunedin, New Zealand
| | - David K Bilkey
- Department of Psychology, University of Otago, Dunedin, New Zealand; Brain Health and Research Centre, University of Otago, Dunedin, New Zealand
| | - Ping Liu
- Department of Anatomy, University of Otago, Dunedin, New Zealand; Brain Health and Research Centre, University of Otago, Dunedin, New Zealand.
| |
Collapse
|
25
|
Solek CM, Farooqi N, Verly M, Lim TK, Ruthazer ES. Maternal immune activation in neurodevelopmental disorders. Dev Dyn 2017; 247:588-619. [PMID: 29226543 DOI: 10.1002/dvdy.24612] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 11/30/2017] [Accepted: 12/01/2017] [Indexed: 12/12/2022] Open
Abstract
Converging lines of evidence from basic science and clinical studies suggest a relationship between maternal immune activation (MIA) and neurodevelopmental disorders such as autism spectrum disorder (ASD) and schizophrenia. The mechanisms through which MIA increases the risk of neurodevelopmental disorders have become a subject of intensive research. This review aims to describe how dysregulation of microglial function and immune mechanisms may link MIA and neurodevelopmental pathologies. We also summarize the current evidence in animal models of MIA. Developmental Dynamics 247:588-619, 2018. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Cynthia M Solek
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Nasr Farooqi
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Myriam Verly
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Tony K Lim
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Edward S Ruthazer
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
26
|
Maternal immune activation in rats produces temporal perception impairments in adult offspring analogous to those observed in schizophrenia. PLoS One 2017; 12:e0187719. [PMID: 29108010 PMCID: PMC5673217 DOI: 10.1371/journal.pone.0187719] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 10/24/2017] [Indexed: 02/07/2023] Open
Abstract
The neurophysiology underlying temporal perception significantly overlaps with areas of dysfunction identified in schizophrenia. Patients commonly exhibit distorted temporal perception, which likely contributes to functional impairments. Thus, study of temporal perception in animal models of the disease may help to understand both cognitive and neurobiological factors involved in functional impairments in patients. As maternal immune activation (MIA) has been shown to be a significant etiological risk factor in development of schizophrenia and other developmental psychiatric diseases, we tested interval timing in a rat model of MIA that has previously been shown to recapitulate several behavioural and neurophysiological impairments observed in the disease. Rats were tested on a temporal-bisection task, in which temporal duration stimuli were categorized as either “short” or “long” by responding to a corresponding lever. Data from this task were modeled to provide estimates of accuracy and sensitivity of temporal perception. Parameter estimates derived from the model fitting showed that MIA rats significantly overestimated the passage of time compared to controls. These results indicate that the MIA rat paradigm recapitulates timing distortions that are phenotypical of schizophrenia. These findings lend further support to the epidemiological validity of this MIA rat model, supporting its relevance for future research into the role of maternal immune activation in producing neurobiological and behavioural impairments in schizophrenia.
Collapse
|
27
|
Zhang J, Jing Y, Zhang H, Bilkey DK, Liu P. Maternal immune activation leads to increased nNOS immunoreactivity in the brain of postnatal day 2 rat offspring. Synapse 2017; 72. [PMID: 28921679 DOI: 10.1002/syn.22011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 08/29/2017] [Accepted: 09/11/2017] [Indexed: 01/10/2023]
Abstract
Neuronal nitric oxide synthase (nNOS) is a key arginine metabolising enzyme in the brain, and nNOS-derived nitric oxide (NO) plays an important role in regulating glutamatergic neurotransmission. NO and its related molecules are involved in the pathogenesis of schizophrenia, and human genetic studies have identified schizophrenia risk genes encoding nNOS. This study systematically investigated how maternal immune activation (MIA; a risk factor for schizophrenia) induced by polyinosinic:polycytidylic acid affected nNOS-immunoreactivity in the brain of the resulting male and female offspring at the age of postnatal day (PND) 2. Immunohistochemistry revealed a markedly increased intensity of nNOS-positive cells in the CA3 and dentate gyrus subregions of the hippocampus, the somatosensory cortex, and the striatum, but not the frontal cortex and hippocampal CA1 region, in the MIA offspring when compared to control group animals. There were no sex differences in the effect. Given the role of nNOS in glutamatergic neurotransmission and its functional relationship with glutamate NMDA receptors, increased nNOS immunoreactivity may indicate the up-regulation of NMDA receptor function in MIA rat offspring at an early postnatal age. Future research is required to determine whether these changes contribute to the neuronal and behavioral dysfunction observed in both juvenile and adult MIA rat offspring.
Collapse
Affiliation(s)
- Jiaxian Zhang
- Department of Anatomy, University of Otago, Dunedin, New Zealand.,Brain Health Research Centre, University of Otago, Dunedin, New Zealand
| | - Yu Jing
- Department of Anatomy, University of Otago, Dunedin, New Zealand.,Brain Health Research Centre, University of Otago, Dunedin, New Zealand
| | - Hu Zhang
- Brain Health Research Centre, University of Otago, Dunedin, New Zealand.,School of Pharmacy, University of Otago, Dunedin, New Zealand
| | - David K Bilkey
- Brain Health Research Centre, University of Otago, Dunedin, New Zealand.,Department of Psychology, University of Otago, Dunedin, New Zealand
| | - Ping Liu
- Department of Anatomy, University of Otago, Dunedin, New Zealand.,Brain Health Research Centre, University of Otago, Dunedin, New Zealand
| |
Collapse
|
28
|
Perinatal inflammation and adult psychopathology: From preclinical models to humans. Semin Cell Dev Biol 2017; 77:104-114. [PMID: 28890420 DOI: 10.1016/j.semcdb.2017.09.010] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Revised: 08/22/2017] [Accepted: 09/06/2017] [Indexed: 02/05/2023]
Abstract
Perinatal environment plays a crucial role in brain development and determines its function through life. Epidemiological studies and clinical reports link perinatal exposure to infection and/or immune activation to various psychiatric disorders. In addition, accumulating evidence from animal models shows that perinatal inflammation can affect various behaviors relevant to psychiatric disorders such as schizophrenia, autism, anxiety and depression. Remarkably, the effects on behavior and brain function do not always depend on the type of inflammatory stimulus or the perinatal age targeted, so diverse inflammatory events can have similar consequences on the brain. Moreover, other perinatal environmental factors that affect behavior (e.g. diet and stress) also elicit inflammatory responses. Understanding the interplay between perinatal environment and inflammation on brain development is required to identify the mechanisms through which perinatal inflammation affect brain function in the adult animal. Evidence for the role of the peripheral immune system and glia on perinatal programming of behavior is discussed in this review, along with recent evidence for the role of epigenetic mechanisms affecting gene expression in the brain.
Collapse
|
29
|
Meehan C, Harms L, Frost JD, Barreto R, Todd J, Schall U, Shannon Weickert C, Zavitsanou K, Michie PT, Hodgson DM. Effects of immune activation during early or late gestation on schizophrenia-related behaviour in adult rat offspring. Brain Behav Immun 2017; 63:8-20. [PMID: 27423491 DOI: 10.1016/j.bbi.2016.07.144] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 06/22/2016] [Accepted: 07/12/2016] [Indexed: 12/16/2022] Open
Abstract
Maternal exposure to infectious agents during gestation has been identified as a significant risk factor for schizophrenia. Using a mouse model, past work has demonstrated that the gestational timing of the immune-activating event can impact the behavioural phenotype and expression of dopaminergic and glutamatergic neurotransmission markers in the offspring. In order to determine the inter-species generality of this effect to rats, another commonly used model species, the current study investigated the impact of a viral mimetic Poly (I:C) at either an early (gestational day 10) or late (gestational day 19) time-point on schizophrenia-related behaviour and neurotransmitter receptor expression in rat offspring. Exposure to Poly (I:C) in late, but not early, gestation resulted in transient impairments in working memory. In addition, male rats exposed to maternal immune activation (MIA) in either early or late gestation exhibited sensorimotor gating deficits. Conversely, neither early nor late MIA exposure altered locomotor responses to MK-801 or amphetamine. In addition, increased dopamine 1 receptor mRNA levels were found in the nucleus accumbens of male rats exposed to early gestational MIA. The findings from this study diverge somewhat from previous findings in mice with MIA exposure, which were often found to exhibit a more comprehensive spectrum of schizophrenia-like phenotypes in both males and females, indicating potential differences in the neurodevelopmental vulnerability to MIA exposure in the rat with regards to schizophrenia related changes.
Collapse
Affiliation(s)
- Crystal Meehan
- School of Psychology, University of Newcastle, Callaghan, NSW, Australia; Priority Research Centre for Brain and Mental Health Research, University of Newcastle, Callaghan, NSW, Australia; Schizophrenia Research Institute, Randwick, NSW, Australia; Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Lauren Harms
- School of Psychology, University of Newcastle, Callaghan, NSW, Australia; Priority Research Centre for Brain and Mental Health Research, University of Newcastle, Callaghan, NSW, Australia; Schizophrenia Research Institute, Randwick, NSW, Australia; Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Jade D Frost
- School of Psychology, University of Newcastle, Callaghan, NSW, Australia; Priority Research Centre for Brain and Mental Health Research, University of Newcastle, Callaghan, NSW, Australia; Schizophrenia Research Institute, Randwick, NSW, Australia; Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Rafael Barreto
- School of Psychology, University of Newcastle, Callaghan, NSW, Australia
| | - Juanita Todd
- School of Psychology, University of Newcastle, Callaghan, NSW, Australia; Priority Research Centre for Brain and Mental Health Research, University of Newcastle, Callaghan, NSW, Australia; Schizophrenia Research Institute, Randwick, NSW, Australia; Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Ulrich Schall
- Priority Research Centre for Brain and Mental Health Research, University of Newcastle, Callaghan, NSW, Australia; Schizophrenia Research Institute, Randwick, NSW, Australia; Hunter Medical Research Institute, Newcastle, NSW, Australia; School of Medicine and Public Health, University of Newcastle, Callaghan, NSW, Australia
| | - Cynthia Shannon Weickert
- Schizophrenia Research Institute, Randwick, NSW, Australia; School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia; Neuroscience Research Australia, Randwick, NSW, Australia
| | | | - Patricia T Michie
- School of Psychology, University of Newcastle, Callaghan, NSW, Australia; Priority Research Centre for Brain and Mental Health Research, University of Newcastle, Callaghan, NSW, Australia; Schizophrenia Research Institute, Randwick, NSW, Australia; Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Deborah M Hodgson
- School of Psychology, University of Newcastle, Callaghan, NSW, Australia; Priority Research Centre for Brain and Mental Health Research, University of Newcastle, Callaghan, NSW, Australia; Schizophrenia Research Institute, Randwick, NSW, Australia; Hunter Medical Research Institute, Newcastle, NSW, Australia.
| |
Collapse
|
30
|
Millar J, Bilkey DK, Ward RD. Maternal immune activation alters sensitivity to action-outcome contingency in adult rat offspring. Brain Behav Immun 2017; 63:81-87. [PMID: 27592563 DOI: 10.1016/j.bbi.2016.08.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 08/16/2016] [Accepted: 08/31/2016] [Indexed: 01/01/2023] Open
Abstract
Epidemiological studies have provided convincing evidence for a role of maternal immune activation in the pathogenesis of neurodevelopmental disorders such as autism and schizophrenia. In recent years, several research groups have capitalised on this discovery and developed animal models such as the maternal immune activation (MIA) model that emulates many phenotypes characteristic of disorders such as schizophrenia. In the present series of experiments we used the MIA model to examine motivation, a core component of the negative symptomatology in schizophrenia. Contrary to what we expected, in the progressive ratio task, which assesses an animals' willingness to work for a reward under increasing effort requirements, we found that MIA rats appeared more motivated than controls. Subsequent tests showed that this seemingly enhanced motivation was not due to an overall increase in responding, nor due to enhanced attribution of incentive salience to reward associated responses. Instead, we found that the increased willingness to work exhibited by MIA animals was due to an inability to detect changes in the contingency between their behaviour and the resulting rewarding outcome. With regard to motivation, the experiments reported here are the first to subject the MIA model to a rigorous experimental analysis of behaviour by parsing underlying processes that give rise to the overt symptoms in psychiatric disease.
Collapse
Affiliation(s)
- Jessica Millar
- University of Otago, PO Box 56, Dunedin 9054, New Zealand
| | - David K Bilkey
- University of Otago, PO Box 56, Dunedin 9054, New Zealand
| | - Ryan D Ward
- University of Otago, PO Box 56, Dunedin 9054, New Zealand.
| |
Collapse
|
31
|
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
|
32
|
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
|
33
|
Rahman T, Zavitsanou K, Purves-Tyson T, Harms LR, Meehan C, Schall U, Todd J, Hodgson DM, Michie PT, Weickert CS. Effects of Immune Activation during Early or Late Gestation on N-Methyl-d-Aspartate Receptor Measures in Adult Rat Offspring. Front Psychiatry 2017; 8:77. [PMID: 28928676 PMCID: PMC5591421 DOI: 10.3389/fpsyt.2017.00077] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 04/21/2017] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Glutamatergic receptor [N-methyl-d-aspartate receptor (NMDAR)] alterations within cortex, hippocampus, and striatum are linked to schizophrenia pathology. Maternal immune activation (MIA) is an environmental risk factor for the development of schizophrenia in offspring. In rodents, gestational timing of MIA may result in distinct behavioral outcomes in adulthood, but how timing of MIA may impact the nature and extent of NMDAR-related changes in brain is not known. We hypothesize that NMDAR-related molecular changes in rat cortex, striatum, and hippocampus are induced by MIA and are dependent on the timing of gestational inflammation and sex of the offspring. METHODS Wistar dams were treated the with viral mimic, polyriboinosinic:polyribocytidylic acid (polyI:C), or vehicle on either gestational day 10 or 19. Fresh-frozen coronal brain sections were collected from offspring between postnatal day 63-91. Autoradiographic binding was used to infer levels of the NMDAR channel, and NR2A and NR2B subunits in cortex [cingulate (Cg), motor, auditory], hippocampus (dentate gyrus, cornu ammonis area 3, cornu ammonis area 1), and striatum [dorsal striatum, nucleus accumbens core, and nucleus accumbens shell (AS)]. NR1 and NR2A mRNA levels were measured by in situ hybridization in cortex, hippocampus, and striatum in male offspring only. RESULTS In the total sample, NMDAR channel binding was elevated in the Cg of polyI:C offspring. NR2A binding was elevated, while NR2B binding was unchanged, in all brain regions of polyI:C offspring overall. Male, but not female, polyI:C offspring exhibited increased NMDAR channel and NR2A binding in the striatum overall, and increased NR2A binding in the cortex overall. Male polyI:C offspring exhibited increased NR1 mRNA in the AS, and increased NR2A mRNA in cortex and subregions of the hippocampus. CONCLUSION MIA may alter glutamatergic signaling in cortical and hippocampal regions via alterations in NMDAR indices; however, this was independent of gestational timing. Male MIA offspring have exaggerated changes in NMDAR compared to females in both the cortex and striatum. The MIA-induced increase in NR2A may decrease brain plasticity and contribute to the exacerbated behavioral changes reported in males and indicate that the brains of male offspring are more susceptible to long-lasting changes in glutamate neurotransmission induced by developmental inflammation.
Collapse
Affiliation(s)
- Tasnim Rahman
- Faculty of Medicine, School of Psychiatry, University of New South Wales, Sydney, NSW, Australia.,Neuroscience Research Australia, Randwick, NSW, Australia.,Schizophrenia Research Institute, at Neuroscience Research Australia, Randwick, NSW, Australia
| | - Katerina Zavitsanou
- Faculty of Medicine, School of Psychiatry, University of New South Wales, Sydney, NSW, Australia.,Neuroscience Research Australia, Randwick, NSW, Australia.,Schizophrenia Research Institute, at Neuroscience Research Australia, Randwick, NSW, Australia
| | - Tertia Purves-Tyson
- Faculty of Medicine, School of Psychiatry, University of New South Wales, Sydney, NSW, Australia.,Neuroscience Research Australia, Randwick, NSW, Australia.,Schizophrenia Research Institute, at Neuroscience Research Australia, Randwick, NSW, Australia
| | - Lauren R Harms
- Schizophrenia Research Institute, at Neuroscience Research Australia, Randwick, NSW, Australia.,School of Psychology, University of Newcastle, Callaghan, NSW, Australia.,Priority Centre for Brain and Mental Health Research, University of Newcastle, Newcastle, NSW, Australia.,Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Crystal Meehan
- Schizophrenia Research Institute, at Neuroscience Research Australia, Randwick, NSW, Australia.,School of Psychology, University of Newcastle, Callaghan, NSW, Australia.,Priority Centre for Brain and Mental Health Research, University of Newcastle, Newcastle, NSW, Australia.,Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Ulrich Schall
- Priority Centre for Brain and Mental Health Research, University of Newcastle, Newcastle, NSW, Australia.,Hunter Medical Research Institute, Newcastle, NSW, Australia.,School of Medicine and Public Health, University of Newcastle, Callaghan, NSW, Australia
| | - Juanita Todd
- Schizophrenia Research Institute, at Neuroscience Research Australia, Randwick, NSW, Australia.,School of Psychology, University of Newcastle, Callaghan, NSW, Australia.,Priority Centre for Brain and Mental Health Research, University of Newcastle, Newcastle, NSW, Australia.,Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Deborah M Hodgson
- School of Psychology, University of Newcastle, Callaghan, NSW, Australia.,Priority Centre for Brain and Mental Health Research, University of Newcastle, Newcastle, NSW, Australia.,Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Patricia T Michie
- Schizophrenia Research Institute, at Neuroscience Research Australia, Randwick, NSW, Australia.,School of Psychology, University of Newcastle, Callaghan, NSW, Australia.,Priority Centre for Brain and Mental Health Research, University of Newcastle, Newcastle, NSW, Australia.,Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Cyndi Shannon Weickert
- Faculty of Medicine, School of Psychiatry, University of New South Wales, Sydney, NSW, Australia.,Neuroscience Research Australia, Randwick, NSW, Australia.,Schizophrenia Research Institute, at Neuroscience Research Australia, Randwick, NSW, Australia
| |
Collapse
|
34
|
Paylor JW, Lins BR, Greba Q, Moen N, de Moraes RS, Howland JG, Winship IR. Developmental disruption of perineuronal nets in the medial prefrontal cortex after maternal immune activation. Sci Rep 2016; 6:37580. [PMID: 27876866 PMCID: PMC5120325 DOI: 10.1038/srep37580] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 11/01/2016] [Indexed: 02/06/2023] Open
Abstract
Maternal infection during pregnancy increases the risk of offspring developing schizophrenia later in life. Similarly, animal models of maternal immune activation (MIA) induce behavioural and anatomical disturbances consistent with a schizophrenia-like phenotype in offspring. Notably, cognitive impairments in tasks dependent on the prefrontal cortex (PFC) are observed in humans with schizophrenia and in offspring after MIA during pregnancy. Recent studies of post-mortem tissue from individuals with schizophrenia revealed deficits in extracellular matrix structures called perineuronal nets (PNNs), particularly in PFC. Given these findings, we examined PNNs over the course of development in a well-characterized rat model of MIA using polyinosinic-polycytidylic acid (polyI:C). We found selective reductions of PNNs in the PFC of polyI:C offspring which did not manifest until early adulthood. These deficits were not associated with changes in parvalbumin cell density, but a decrease in the percentage of parvalbumin cells surrounded by a PNN. Developmental expression of PNNs was also significantly altered in the amygdala of polyI:C offspring. Our results indicate MIA causes region specific developmental abnormalities in PNNs in the PFC of offspring. These findings confirm the polyI:C model replicates neuropathological alterations associated with schizophrenia and may identify novel mechanisms for cognitive and emotional dysfunction in the disorder.
Collapse
Affiliation(s)
- John W Paylor
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, T6G 2E1, AB, Canada.,Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Edmonton, T6G 2B7, AB, Canada
| | - Brittney R Lins
- Department of Physiology, University of Saskatchewan, Saskatoon, S7N 5E5, SK, Canada
| | - Quentin Greba
- Department of Physiology, University of Saskatchewan, Saskatoon, S7N 5E5, SK, Canada
| | - Nicholas Moen
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, T6G 2E1, AB, Canada
| | | | - John G Howland
- Department of Physiology, University of Saskatchewan, Saskatoon, S7N 5E5, SK, Canada
| | - Ian R Winship
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, T6G 2E1, AB, Canada.,Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Edmonton, T6G 2B7, AB, Canada
| |
Collapse
|
35
|
Vernon AC, So PW, Lythgoe DJ, Chege W, Cooper JD, Williams SCR, Kapur S. Longitudinal in vivo maturational changes of metabolites in the prefrontal cortex of rats exposed to polyinosinic-polycytidylic acid in utero. Eur Neuropsychopharmacol 2015; 25:2210-20. [PMID: 26475576 DOI: 10.1016/j.euroneuro.2015.09.022] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Revised: 08/28/2015] [Accepted: 09/29/2015] [Indexed: 01/10/2023]
Abstract
Proton magnetic resonance spectroscopy ((1)H MRS) studies in schizophrenia patients generally report decreased levels of N-acetyl-aspartate (NAA), glutamate and glutathione, particularly in frontal cortex. However, these data are inconsistent in part due to confounds associated with clinical samples. The lack of validated diagnostic biomarkers also hampers analysis of the neurodevelopmental trajectory of neurochemical abnormalities. Rodent models are powerful tools to address these issues, particularly when combined with (1)H MRS (clinically comparable technology). We investigated the trajectory of metabolic changes in the prefrontal cortex during brain maturation from adolescence to adulthood in vivo using (1)H MRS in rats exposed prenatally to polyinosinic-polycytidylic acid (POL), a rodent model of maternal immune activation (MIA), an epidemiological risk factor for several psychiatric disorders with a neurodevelopmental origin. Longitudinal in vivo (1)H MRS revealed a significant decrease in PFC levels of GSH and taurine in adult, but not adolescent rats. Significant age×MIA interactions for PFC levels of NAA were also observed. These data replicate some deficits observed in the PFC of patients with schizophrenia. There were no significant changes in the levels of glutamate or any other metabolite. These data suggest prenatal exposure to POL leads to subtle metabolic perturbations of the normal maturing PFC, which may be related to subsequent behavioural abnormalities. Further work is however required to examine any potential confound of shipping stress on the presumed imbalances in PFC metabolites in POL-exposed offspring. Testing the interactions between MIA with stress or genetic risk variants will also be an important advance.
Collapse
Affiliation(s)
- Anthony C Vernon
- King's College London, Institute of Psychiatry Psychology and Neuroscience, Department of Psychosis Studies, De Crespigny Park, London SE5 8AF, UK.
| | - Po-Wah So
- King's College London, Institute of Psychiatry, Department of Neuroimaging, Centre for Neuroimaging Sciences, De Crespigny Park, London SE5 8AF, UK
| | - David J Lythgoe
- King's College London, Institute of Psychiatry, Department of Neuroimaging, Centre for Neuroimaging Sciences, De Crespigny Park, London SE5 8AF, UK
| | - Winfred Chege
- King's College London, Institute of Psychiatry Psychology and Neuroscience, Department of Psychosis Studies, De Crespigny Park, London SE5 8AF, UK
| | - Jonathan D Cooper
- King's College London, Institute of Psychiatry Psychology and Neuroscience, Department of Basic and Clinical Neuroscience, The James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Steven C R Williams
- King's College London, Institute of Psychiatry, Department of Neuroimaging, Centre for Neuroimaging Sciences, De Crespigny Park, London SE5 8AF, UK
| | - Shitij Kapur
- King's College London, Institute of Psychiatry Psychology and Neuroscience, Department of Psychosis Studies, De Crespigny Park, London SE5 8AF, UK
| |
Collapse
|
36
|
Wolff AR, Bilkey DK. Prenatal immune activation alters hippocampal place cell firing characteristics in adult animals. Brain Behav Immun 2015; 48:232-43. [PMID: 25843370 DOI: 10.1016/j.bbi.2015.03.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Revised: 03/12/2015] [Accepted: 03/24/2015] [Indexed: 12/27/2022] Open
Abstract
Prenatal maternal immune activation (MIA) is a risk factor for several developmental neuropsychiatric disorders, including autism, bipolar disorder and schizophrenia. Adults with these disorders display alterations in memory function that may result from changes in the structure and function of the hippocampus. In the present study we use an animal model to investigate the effect that a transient prenatal maternal immune activation episode has on the spatially-modulated firing activity of hippocampal neurons in adult animals. MIA was induced in pregnant rat dams with a single injection of the synthetic cytokine inducer polyinosinic:polycytidylic acid (poly I:C) on gestational day 15. Control dams were given a saline equivalent. Firing activity and local field potentials (LFPs) were recorded from the CA1 region of the adult male offspring of these dams as they moved freely in an open arena. Most neurons displayed characteristic spatially-modulated 'place cell' firing activity and while there was no between-group difference in mean firing rate between groups, place cells had smaller place fields in MIA-exposed animals when compared to control-group cells. Cells recorded in MIA-group animals also displayed an altered firing-phase synchrony relationship to simultaneously recorded LFPs. When the floor of the arena was rotated, the place fields of MIA-group cells were more likely to shift in the same direction as the floor rotation, suggesting that local cues may have been more salient for these animals. In contrast, place fields in control group cells were more likely to shift firing position to novel spatial locations suggesting an altered response to contextual cues. These findings show that a single MIA intervention is sufficient to change several important characteristics of hippocampal place cell activity in adult offspring. These changes could contribute to the memory dysfunction that is associated with MIA, by altering the encoding of spatial context and by disrupting plasticity mechanisms that are dependent on spike timing synchrony.
Collapse
Affiliation(s)
- Amy R Wolff
- Department of Psychology, Brain Health Research Centre, University of Otago, Dunedin, New Zealand
| | - David K Bilkey
- Department of Psychology, Brain Health Research Centre, University of Otago, Dunedin, New Zealand.
| |
Collapse
|
37
|
Möller M, Swanepoel T, Harvey BH. Neurodevelopmental Animal Models Reveal the Convergent Role of Neurotransmitter Systems, Inflammation, and Oxidative Stress as Biomarkers of Schizophrenia: Implications for Novel Drug Development. ACS Chem Neurosci 2015; 6:987-1016. [PMID: 25794269 DOI: 10.1021/cn5003368] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Schizophrenia is a life altering disease with a complex etiology and pathophysiology, and although antipsychotics are valuable in treating the disorder, certain symptoms and/or sufferers remain resistant to treatment. Our poor understanding of the underlying neuropathological mechanisms of schizophrenia hinders the discovery and development of improved pharmacological treatment, so that filling these gaps is of utmost importance for an improved outcome. A vast amount of clinical data has strongly implicated the role of inflammation and oxidative insults in the pathophysiology of schizophrenia. Preclinical studies using animal models are fundamental in our understanding of disease development and pathology as well as the discovery and development of novel treatment options. In particular, social isolation rearing (SIR) and pre- or postnatal inflammation (PPNI) have shown great promise in mimicking the biobehavioral manifestations of schizophrenia. Furthermore, the "dual-hit" hypothesis of schizophrenia states that a first adverse event such as genetic predisposition or a prenatal insult renders an individual susceptible to develop the disease, while a second insult (e.g., postnatal inflammation, environmental adversity, or drug abuse) may be necessary to precipitate the full-blown syndrome. Animal models that emphasize the "dual-hit" hypothesis therefore provide valuable insight into understanding disease progression. In this Review, we will discuss SIR, PPNI, as well as possible "dual-hit" animal models within the context of the redox-immune-inflammatory hypothesis of schizophrenia, correlating such changes with the recognized monoamine and behavioral alterations of schizophrenia. Finally, based on these models, we will review new therapeutic options, especially those targeting immune-inflammatory and redox pathways.
Collapse
Affiliation(s)
- M. Möller
- Department of Pharmacology and ‡Center of Excellence for Pharmaceutical Sciences,
School of Pharmacy, North-West University, Potchefstroom 2531, South Africa
| | - T. Swanepoel
- Department of Pharmacology and ‡Center of Excellence for Pharmaceutical Sciences,
School of Pharmacy, North-West University, Potchefstroom 2531, South Africa
| | - B. H. Harvey
- Department of Pharmacology and ‡Center of Excellence for Pharmaceutical Sciences,
School of Pharmacy, North-West University, Potchefstroom 2531, South Africa
| |
Collapse
|
38
|
Leza JC, García-Bueno B, Bioque M, Arango C, Parellada M, Do K, O'Donnell P, Bernardo M. Inflammation in schizophrenia: A question of balance. Neurosci Biobehav Rev 2015; 55:612-26. [PMID: 26092265 DOI: 10.1016/j.neubiorev.2015.05.014] [Citation(s) in RCA: 144] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 04/22/2015] [Accepted: 05/18/2015] [Indexed: 02/08/2023]
Abstract
In the past decade, there has been renewed interest in immune/inflammatory changes and their associated oxidative/nitrosative consequences as key pathophysiological mechanisms in schizophrenia and related disorders. Both brain cell components (microglia, astrocytes, and neurons) and peripheral immune cells have been implicated in inflammation and the resulting oxidative/nitrosative stress (O&NS) in schizophrenia. Furthermore, down-regulation of endogenous antioxidant and anti-inflammatory mechanisms has been identified in biological samples from patients, although the degree and progression of the inflammatory process and the nature of its self-regulatory mechanisms vary from early onset to full-blown disease. This review focuses on the interactions between inflammation and O&NS, their damaging consequences for brain cells in schizophrenia, the possible origins of inflammation and increased O&NS in the disorder, and current pharmacological strategies to deal with these processes (mainly treatments with anti-inflammatory or antioxidant drugs as add-ons to antipsychotics).
Collapse
Affiliation(s)
- Juan C Leza
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Complutense University, Madrid, Spain; Department of Pharmacology, Faculty of Medicine, Complutense University, Madrid, Spain; Instituto de Investigación Sanitaria (IIS) Hospital 12 de Octubre (i+12), Madrid, Spain.
| | - Borja García-Bueno
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Complutense University, Madrid, Spain; Department of Pharmacology, Faculty of Medicine, Complutense University, Madrid, Spain; Instituto de Investigación Sanitaria (IIS) Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Miquel Bioque
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Complutense University, Madrid, Spain; Barcelona Clínic Schizophrenia Unit, Hospital Clínic Barcelona, University of Barcelona, IDIBAPS, Barcelona, Spain
| | - Celso Arango
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Complutense University, Madrid, Spain; Department of Psychiatry, Faculty of Medicine, Complutense University, Madrid, Spain; Child and Adolescent Psychiatry Department, IIS Hospital Gregorio Marañón (IISGM), Madrid, Spain
| | - Mara Parellada
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Complutense University, Madrid, Spain; Department of Psychiatry, Faculty of Medicine, Complutense University, Madrid, Spain; Child and Adolescent Psychiatry Department, IIS Hospital Gregorio Marañón (IISGM), Madrid, Spain
| | - Kim Do
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
| | | | - Miguel Bernardo
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Complutense University, Madrid, Spain; Barcelona Clínic Schizophrenia Unit, Hospital Clínic Barcelona, University of Barcelona, IDIBAPS, Barcelona, Spain
| |
Collapse
|
39
|
Vorhees CV, Graham DL, Braun AA, Schaefer TL, Skelton MR, Richtand NM, Williams MT. Prenatal immune challenge in rats: effects of polyinosinic-polycytidylic acid on spatial learning, prepulse inhibition, conditioned fear, and responses to MK-801 and amphetamine. Neurotoxicol Teratol 2014; 47:54-65. [PMID: 25450663 DOI: 10.1016/j.ntt.2014.10.007] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2014] [Revised: 10/23/2014] [Accepted: 10/31/2014] [Indexed: 01/05/2023]
Abstract
Prenatal maternal immune activation increases risk for schizophrenia and/or autism. Previous data suggest that maternal weight change in response to the immune activator polyinosinic-polycytidylic (Poly IC) in rats influences the severity of effect in the offspring as does the exposure period. We treated gravid Sprague-Dawley rats from E14 to 18 with 8mg/kg/day Poly IC or saline. The Poly IC group was divided into those that gained the least weight or lost (Poly IC (L)) and those that gained the most (Poly IC (H)) weight. There were no effects of Poly IC on anxiety (elevated zero-maze, open-field, object burying), or Morris water maze cued learning or working memory or Cincinnati water maze egocentric learning. The Poly IC (H) group males had decreased acoustic startle whereas Poly IC (L) females had reduced startle and increased PPI. Poly IC offspring showed exaggerated hyperactivity in response to amphetamine (primarily in the Poly IC (H) group) and attenuated hyperactivity in response to MK-801 challenge (primarily in the Poly IC (L) group). Poly IC (L) males showed reduced cued conditioned freezing; both sexes showed less time in the dark in a light-dark test, and the Poly IC groups showed impaired Morris water maze hidden platform acquisition and probe performance. The data demonstrate that offspring from the most affected dams were more affected than those from less reactive dams indicating that degree of maternal immune activation predicts severity of effects on offspring behavior.
Collapse
Affiliation(s)
- Charles V Vorhees
- Division of Child Neurology, Cincinnati Children's Research Foundation, 3333 Burnet Ave., Cincinnati, OH 45229, United States; Department of Pediatrics, College of Medicine, University of Cincinnati, United States.
| | - Devon L Graham
- Department of Pharmacology and Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37232, United States
| | - Amanda A Braun
- Division of Child Neurology, Cincinnati Children's Research Foundation, 3333 Burnet Ave., Cincinnati, OH 45229, United States; Department of Pediatrics, College of Medicine, University of Cincinnati, United States
| | - Tori L Schaefer
- Division of Child Neurology, Cincinnati Children's Research Foundation, 3333 Burnet Ave., Cincinnati, OH 45229, United States; Department of Pediatrics, College of Medicine, University of Cincinnati, United States
| | - Matthew R Skelton
- Division of Child Neurology, Cincinnati Children's Research Foundation, 3333 Burnet Ave., Cincinnati, OH 45229, United States; Department of Pediatrics, College of Medicine, University of Cincinnati, United States
| | - Neil M Richtand
- Psychiatry Service, V-116A, VA San Diego Healthcare System, 3350 La Jolla Village Dr., San Diego, CA 92161, United States
| | - Michael T Williams
- Division of Child Neurology, Cincinnati Children's Research Foundation, 3333 Burnet Ave., Cincinnati, OH 45229, United States; Department of Pediatrics, College of Medicine, University of Cincinnati, United States
| |
Collapse
|
40
|
Richetto J, Riva MA. Prenatal maternal factors in the development of cognitive impairments in the offspring. J Reprod Immunol 2014; 104-105:20-5. [PMID: 24794049 DOI: 10.1016/j.jri.2014.03.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2013] [Revised: 03/11/2014] [Accepted: 03/17/2014] [Indexed: 01/16/2023]
Abstract
Different environmental factors acting during sensitive prenatal periods can have a negative impact on neurodevelopment and predispose the individual to the development of various psychiatric conditions that often share cognitive impairments as a common component. As cognitive symptoms remain one of the most challenging and resistant aspects of mental illness to be treated pharmacologically, it is important to investigate the mechanisms underlying such cognitive deficits, with particular focus on the impact of early life adverse events that predispose the individual to mental disorders. Multiple clinical studies have, in fact, repeatedly confirmed that prenatal maternal factors, such as infection, stress or malnutrition, are pivotal in shaping behavioral and cognitive functions of the offspring, and in the past decade many preclinical studies have investigated this hypothesis. The purpose of this review is to describe recent preclinical studies aimed at dissecting the relative impact of various prenatal maternal factors on the development of cognitive impairments in offspring, focusing on animal models of prenatal stress and prenatal infection. These recent studies point to the pivotal role of prenatal stressful experiences in shaping memory and learning functions associated with specific brain structures, such as the hippocampus and the prefrontal cortex. More importantly, such experimental evidence suggests that different insults converge on similar downstream functional targets, such as cognition, which may therefore represent an endophenotype for several pathological conditions. Future studies should thus focus on investigating the mechanisms contributing to the convergent action of different prenatal insults in order to identify targets for novel therapeutic intervention.
Collapse
Affiliation(s)
- Juliet Richetto
- Center of Neuropharmacology, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Marco A Riva
- Center of Neuropharmacology, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
41
|
Lin CW, Hsueh YP. Sarm1, a neuronal inflammatory regulator, controls social interaction, associative memory and cognitive flexibility in mice. Brain Behav Immun 2014; 37:142-51. [PMID: 24321214 DOI: 10.1016/j.bbi.2013.12.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Revised: 11/16/2013] [Accepted: 12/01/2013] [Indexed: 12/31/2022] Open
Abstract
Impaired neurodevelopment leads to several psychiatric disorders, including autism, schizophrenia and attention deficiency hyperactivity disorder. Our prior study showed that sterile alpha and TIR motif-containing 1 protein (Sarm1) regulates neuronal morphogenesis through at least two pathways. Sarm1 controls neuronal morphogenesis, including dendritic arborization, axonal outgrowth and establishment of neuronal polarity, through the MKK-JNK pathway. Neuronally expressed Sarm1 also regulates the expression of inflammatory cytokines in the brain, which have also been shown to impact brain development and function. Because the reduction of Sarm1 expression negatively influences neuronal development, here we investigated whether Sarm1 controls mouse behaviors. We analyzed two independent Sarm1 transgenic mouse lines using a series of behavioral assays, and found that the reduction of Sarm1 protein levels had a limited effect on locomotion and anxiety. However, Sarm1 knockdown mice exhibited impairments in cued and contextual fear conditioning as well as cognitive flexibility. Moreover, the three-chambered social test, reciprocal social interaction and social transmission of food preference further illustrated deficiencies in Sarm1 knockdown mice in social interaction. These findings suggest that Sarm1, a molecule that regulates innate immunity and neuronal morphogenesis, regulates social behaviors and cognition. We conclude that Sarm1 is involved in immune response, neural development and psychiatric disorders.
Collapse
Affiliation(s)
- Chia-Wen Lin
- Institute of Molecular Biology, Academia Sinica, Taipei 11529, Taiwan
| | - Yi-Ping Hsueh
- Institute of Molecular Biology, Academia Sinica, Taipei 11529, Taiwan.
| |
Collapse
|
42
|
Desbonnet L, O’Tuathaigh CMP, Waddington JL. Modeling schizophrenia: uncovering novel therapeutic targets. Expert Rev Clin Pharmacol 2014; 5:667-76. [DOI: 10.1586/ecp.12.57] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
43
|
Dickerson DD, Bilkey DK. Aberrant neural synchrony in the maternal immune activation model: using translatable measures to explore targeted interventions. Front Behav Neurosci 2013; 7:217. [PMID: 24409130 PMCID: PMC3873515 DOI: 10.3389/fnbeh.2013.00217] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Accepted: 12/16/2013] [Indexed: 01/01/2023] Open
Abstract
Maternal exposure to infection occurring mid-gestation produces a three-fold increase in the risk of schizophrenia in the offspring. The critical initiating factor appears to be the maternal immune activation (MIA) that follows infection. This process can be induced in rodents by exposure of pregnant dams to the viral mimic Poly I:C, which triggers an immune response that results in structural, functional, behavioral, and electrophysiological phenotypes in the adult offspring that model those seen in schizophrenia. We used this model to explore the role of synchronization in brain neural networks, a process thought to be dysfunctional in schizophrenia and previously associated with positive, negative, and cognitive symptoms of schizophrenia. Exposure of pregnant dams to Poly I:C on GD15 produced an impairment in long-range neural synchrony in adult offspring between two regions implicated in schizophrenia pathology; the hippocampus and the medial prefrontal cortex (mPFC). This reduction in synchrony was ameliorated by acute doses of the antipsychotic clozapine. MIA animals have previously been shown to have impaired pre-pulse inhibition (PPI), a gold-standard measure of schizophrenia-like deficits in animal models. Our data showed that deficits in synchrony were positively correlated with the impairments in PPI. Subsequent analysis of LFP activity during the PPI response also showed that reduced coupling between the mPFC and the hippocampus following processing of the pre-pulse was associated with reduced PPI. The ability of the MIA intervention to model neurodevelopmental aspects of schizophrenia pathology provides a useful platform from which to investigate the ontogeny of aberrant synchronous processes. Further, the way in which the model expresses translatable deficits such as aberrant synchrony and reduced PPI will allow researchers to explore novel intervention strategies targeted to these changes.
Collapse
Affiliation(s)
| | - David K Bilkey
- Department of Psychology, University of Otago Dunedin, New Zealand
| |
Collapse
|
44
|
Savanthrapadian S, Wolff AR, Logan BJ, Eckert MJ, Bilkey DK, Abraham WC. Enhanced hippocampal neuronal excitability and LTP persistence associated with reduced behavioral flexibility in the maternal immune activation model of schizophrenia. Hippocampus 2013; 23:1395-409. [PMID: 23966340 DOI: 10.1002/hipo.22193] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/16/2013] [Indexed: 12/23/2022]
Abstract
Individuals with schizophrenia display a number of structural and cytoarchitectural alterations in the hippocampus, suggesting that other functions such as synaptic plasticity may also be modified. Altered hippocampal plasticity is likely to affect memory processing, and therefore any such pathology may contribute to the cognitive symptoms of schizophrenia, which includes prominent memory impairment. The current study tested whether prenatal exposure to infection, an environmental risk factor that has previously been associated with schizophrenia produced changes in hippocampal synaptic transmission or plasticity, using the maternal immune activation (MIA) animal model. We also assessed performance in hippocampus-dependent memory tasks to determine whether altered plasticity is associated with memory dysfunction. MIA did not alter basal synaptic transmission in either the dentate gyrus or CA1 of freely moving adult rats. It did, however, result in increased paired-pulse facilitation of the dentate gyrus population spike and an enhanced persistence of dentate long-term potentiation. MIA animals displayed slower learning of a reversed platform location in the water maze, and a similarly slowed learning during reversal in a spatial plus maze task. Together these findings are indicative of reduced behavioral flexibility in response to changes in task requirements. The results are consistent with the hypothesis that hippocampal plasticity is altered in schizophrenia, and that this change in plasticity mechanisms may underlie some aspects of cognitive dysfunction in this disorder.
Collapse
Affiliation(s)
- Shakuntala Savanthrapadian
- Department of Psychology and the Brain Health Research Center, University of Otago, Dunedin, New Zealand
| | | | | | | | | | | |
Collapse
|
45
|
Venkatasubramanian G, Debnath M. The TRIPS (Toll-like receptors in immuno-inflammatory pathogenesis) Hypothesis: a novel postulate to understand schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry 2013; 44:301-11. [PMID: 23587629 DOI: 10.1016/j.pnpbp.2013.04.001] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Revised: 03/08/2013] [Accepted: 04/05/2013] [Indexed: 02/09/2023]
Abstract
Mounting evidence indicates that immune activation and/or immuno-inflammatory reactions during neurodevelopment apparently contribute to the pathogenesis and progression of schizophrenia. One of the important environmental factors that is known to trigger immune activation/inflammatory responses during early pregnancy is prenatal infection. Recent understanding from animal studies suggests that prenatal infection induced maternal immune activation (MIA)/inflammation in congruence with oxidative/nitrosative stress can lead to neurodevelopmental damage and behavioral abnormalities in the offspring. Although the underlying precise mechanistic processes of MIA/inflammation are yet to be completely elucidated, it is being increasingly recognized that Toll-like receptors (TLRs) that form the first line of defense against invading microorganisms could participate in the prenatal infection induced immune insults. Interestingly, some of the TLRs, especially TLR3 and TLR4 that modulate neurodevelopment, neuronal survival and neuronal plasticity by regulating the neuro-immune cross-talk in the developing and adult brain could also be affected by prenatal infection. Importantly, sustained activation of TLR3/TLR4 due to environmental factors including infection and stress has been found to generate excessive reactive oxygen species (ROS)/reactive nitrogen species (RNS) as well as pro-inflammatory mediators during embryogenesis, which result into neuronal damage by necrosis/apoptosis. In recent times, ROS/RNS and immuno-inflammatory mediators are being increasingly linked to progressive brain changes in schizophrenia. Although a significant role of TLR3/TLR4 in neurodegeneration is gaining certainty, their importance in establishing a causal link between prenatal infection and immuno-inflammatory, oxidative and nitrosative stress (IO&NS) responses and influence on adult presentation of schizophrenia is yet to be ascertained. We review here the current knowledge generated from the animal and human studies on the role of TLRs in schizophrenia and finally propose the "TRIPS Hypothesis" (Toll-like receptors in immuno-inflammatory pathogenesis) to elucidate the underlying mechanism(s) of TLR-mediated risk of schizophrenia. Considering the established role of TLR3 and TLR4 in antiviral and antibacterial responses respectively, we believe that in some cases of schizophrenia where IO&NS responses are evident, prenatal infection might lead to neuroprogressive changes in a TLR3/TLR4-dependent way.
Collapse
Affiliation(s)
- Ganesan Venkatasubramanian
- The Schizophrenia Clinic, Department of Psychiatry, National Institute of Mental Health and Neurosciences, Hosur Road, Bangalore 560029, India
| | | |
Collapse
|
46
|
Gibney SM, Drexhage HA. Evidence for a dysregulated immune system in the etiology of psychiatric disorders. J Neuroimmune Pharmacol 2013; 8:900-20. [PMID: 23645137 DOI: 10.1007/s11481-013-9462-8] [Citation(s) in RCA: 144] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Accepted: 04/17/2013] [Indexed: 02/06/2023]
Abstract
There is extensive bi-directional communication between the brain and the immune system in both health and disease. In recent years, the role of an altered immune system in the etiology of major psychiatric disorders has become more apparent. Studies have demonstrated that some patients with major psychiatric disorders exhibit characteristic signs of immune dysregulation and that this may be a common pathophysiological mechanism that underlies the development and progression of these disorders. Furthermore, many psychiatric disorders are also often accompanied by chronic medical conditions related to immune dysfunction such as autoimmune diseases, diabetes and atherosclerosis. One of the major psychiatric disorders that has been associated with an altered immune system is schizophrenia, with approximately one third of patients with this disorder showing immunological abnormalities such as an altered cytokine profile in serum and cerebrospinal fluid. An altered cytokine profile is also found in a proportion of patients with major depressive disorder and is thought to be potentially related to the pathophysiology of this disorder. Emerging evidence suggests that altered immune parameters may also be implicated in the neurobiological etiology of autism spectrum disorders. Further support for a role of immune dysregulation in the pathophysiology of these psychiatric disorders comes from studies showing the immunomodulating effects of antipsychotics and antidepressants, and the mood altering effects of anti-inflammatory therapies. This review will not attempt to discuss all of the psychiatric disorders that have been associated with an augmented immune system, but will instead focus on several key disorders where dysregulation of this system has been implicated in their pathophysiology including depression, schizophrenia and autism spectrum disorder.
Collapse
Affiliation(s)
- Sinead M Gibney
- Department of Immunology, Na1101, Erasmus MC, Dr. Molewaterplein 50, Erasmus MC, 3015 GE Rotterdam, The Netherlands.
| | | |
Collapse
|
47
|
Anderson G, Maes M. Schizophrenia: linking prenatal infection to cytokines, the tryptophan catabolite (TRYCAT) pathway, NMDA receptor hypofunction, neurodevelopment and neuroprogression. Prog Neuropsychopharmacol Biol Psychiatry 2013; 42:5-19. [PMID: 22800757 DOI: 10.1016/j.pnpbp.2012.06.014] [Citation(s) in RCA: 101] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2012] [Revised: 06/06/2012] [Accepted: 06/18/2012] [Indexed: 02/07/2023]
Abstract
In 1995, the macrophage-T lymphocyte theory of schizophrenia (Smith and Maes, 1995) considered that activated immuno-inflammatory pathways may account for the higher neurodevelopmental pathology linked with gestational infections through the detrimental effects of activated microglia, oxidative and nitrosative stress (O&NS), cytokine-induced activation of the tryptophan catabolite (TRYCAT) pathway and consequent modulation of the N-methyl d-aspartate receptor (NMDAr) and glutamate production. The aim of the present paper is to review the current state-of-the art regarding the role of the above pathways in schizophrenia. Accumulating data suggest a powerful role for prenatal infection, both viral and microbial, in driving an early developmental etiology to schizophrenia. Models of prenatal rodent infection show maintained activation of immuno-inflammatory pathways coupled to increased microglia activation. The ensuing activation of immuno-inflammatory pathways in schizophrenia may activate the TRYCAT pathway, including increased kynurenic acid (KA) and neurotoxic TRYCATs. Increased KA, via the inhibition of the α7 nicotinic acetylcholine receptor, lowers gamma-amino-butyric-acid (GABA)ergic post-synaptic current, contributing to dysregulated glutamatergic activity. Hypofunctioning of the NMDAr on GABAergic interneurons will contribute to glutamatergic dysregulation. Many susceptibility genes for schizophrenia are predominantly expressed in early development and will interact with these early developmental driven changes in the immuno-inflammatory and TRYCAT pathways. Maternal infection and subsequent immuno-inflammatory responses are additionally associated with O&NS, including lowered antioxidants such as glutathione. This will contribute to alterations in neurogenesis and myelination. In such a scenario a) a genetic or epigenetic potentiation of immuno-inflammatory pathways may constitute a double hit on their own, stimulating wider immuno-inflammatory responses and thus potentiating the TRYCAT pathway and subsequent NMDAr dysfunction and neuroprogression; and b) antipsychotic-induced changes in immuno-inflammatory, TRYCAT and O&NS pathways would modulate the CNS glia-neuronal interactions that determine synaptic plasticity as well as myelin generation and maintenance.
Collapse
|
48
|
Developmental neuroinflammation and schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry 2013; 42:20-34. [PMID: 22122877 DOI: 10.1016/j.pnpbp.2011.11.003] [Citation(s) in RCA: 226] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2011] [Revised: 10/18/2011] [Accepted: 11/09/2011] [Indexed: 12/27/2022]
Abstract
There is increasing interest in and evidence for altered immune factors in the etiology and pathophysiology of schizophrenia. Stimulated by various epidemiological findings reporting elevated risk of schizophrenia following prenatal exposure to infection, one line of current research aims to explore the potential contribution of immune-mediated disruption of early brain development in the precipitation of long-term psychotic disease. Since the initial formulation of the "prenatal cytokine hypothesis" more than a decade ago, extensive epidemiological research and remarkable advances in modeling prenatal immune activation effects in animal models have provided strong support for this hypothesis by underscoring the critical role of cytokine-associated inflammatory events, together with downstream pathophysiological processes such as oxidative stress, hypoferremia and zinc deficiency, in mediating the short- and long-term neurodevelopmental effects of prenatal infection. Longitudinal studies in animal models further indicate that infection-induced developmental neuroinflammation may be pathologically relevant beyond the antenatal and neonatal periods, and may contribute to disease progression associated with the gradual development of full-blown schizophrenic disease. According to this scenario, exposure to prenatal immune challenge primes early pre- and postnatal alterations in peripheral and central inflammatory response systems, which in turn may disrupt the normal development and maturation of neuronal systems from juvenile to adult stages of life. Such developmental neuroinflammation may adversely affect processes that are pivotal for normal brain maturation, including myelination, synaptic pruning, and neuronal remodeling, all of which occur to a great extent during postnatal brain maturation. Undoubtedly, our understanding of the role of developmental neuroinflammation in progressive brain changes relevant to schizophrenia is still in infancy. Identification of these mechanisms would be highly warranted because they may represent a valuable target to attenuate or even prevent the emergence of full-blown brain and behavioral pathology, especially in individuals with a history of prenatal complications such as in-utero exposure to infection and/or inflammation.
Collapse
|
49
|
Novak G, Fan T, O'Dowd BF, George SR. Postnatal maternal deprivation and pubertal stress have additive effects on dopamine D2 receptor and CaMKII beta expression in the striatum. Int J Dev Neurosci 2013; 31:189-95. [DOI: 10.1016/j.ijdevneu.2013.01.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Revised: 12/31/2012] [Accepted: 01/01/2013] [Indexed: 01/05/2023] Open
Affiliation(s)
- Gabriela Novak
- Centre for Addiction and Mental HealthTorontoOntarioCanada
- Department of PharmacologyUniversity of TorontoTorontoOntarioCanada
| | - Theresa Fan
- Centre for Addiction and Mental HealthTorontoOntarioCanada
| | - Brian F. O'Dowd
- Centre for Addiction and Mental HealthTorontoOntarioCanada
- Department of PharmacologyUniversity of TorontoTorontoOntarioCanada
| | - Susan R. George
- Centre for Addiction and Mental HealthTorontoOntarioCanada
- Department of PharmacologyUniversity of TorontoTorontoOntarioCanada
- Department of MedicineUniversity of TorontoTorontoOntarioCanada
| |
Collapse
|
50
|
Yee N, Schwarting RKW, Fuchs E, Wöhr M. Increased affective ultrasonic communication during fear learning in adult male rats exposed to maternal immune activation. J Psychiatr Res 2012; 46:1199-205. [PMID: 22687817 DOI: 10.1016/j.jpsychires.2012.05.010] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2012] [Revised: 04/27/2012] [Accepted: 05/21/2012] [Indexed: 02/05/2023]
Abstract
Maternal exposure to infection during pregnancy greatly increases the risk of psychopathology in the offspring. In support of clinical findings, rodent models of maternal immune activation (MIA) show that prenatal exposure to pathogens can induce phenotypic changes in the offspring associated with schizophrenia, autism, depression and anxiety. In the current study, we investigated the effects of MIA via polyinosinic:polycytidylic acid (poly I:C) on emotional behavior and communication in rats. Pregnant rats were administered poly I:C or saline on gestation day 15 and male offspring were tested in an auditory fear conditioning paradigm in early adulthood. We found that prenatal poly I:C exposure significantly altered affective signaling, namely, the production of aversive 22-kHz ultrasonic vocalizations (USVs), in terms of call number, structure and temporal patterning. MIA led to an increase in aversive 22-kHz USVs to 300% of saline controls. Offspring exposed to MIA not only emitted more 22-kHz USVs, but also emitted calls that were shorter in duration and occurred in bouts containing more calls. The production of appetitive 50-kHz USVs and audible calls was not affected. Intriguingly, alterations in aversive 22-kHz USV emission were observed despite no obvious changes in overt defensive behavior, which highlights the importance of assessing USVs as an additional measure of fear. Aversive 22-kHz USVs are a prominent part of the rat's defensive behavioral repertoire and serve important communicative functions, most notably as alarm calls. The observed changes in aversive 22-kHz USVs show that MIA has long-term effects on emotional behavior and communication in exposed rat offspring.
Collapse
Affiliation(s)
- Nicole Yee
- Clinical Neurobiology Laboratory, German Primate Center, Leibniz Institute for Primate Research, Kellnerweg 4, Göttingen 37077, Germany.
| | | | | | | |
Collapse
|