1
|
Topchiy I, Mohbat J, Folorunso OO, Wang ZZ, Lazcano-Etchebarne C, Engin E. GABA system as the cause and effect in early development. Neurosci Biobehav Rev 2024; 161:105651. [PMID: 38579901 PMCID: PMC11081854 DOI: 10.1016/j.neubiorev.2024.105651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 03/05/2024] [Accepted: 04/01/2024] [Indexed: 04/07/2024]
Abstract
GABA is the primary inhibitory neurotransmitter in the adult brain and through its actions on GABAARs, it protects against excitotoxicity and seizure activity, ensures temporal fidelity of neurotransmission, and regulates concerted rhythmic activity of neuronal populations. In the developing brain, the development of GABAergic neurons precedes that of glutamatergic neurons and the GABA system serves as a guide and framework for the development of other brain systems. Despite this early start, the maturation of the GABA system also continues well into the early postnatal period. In this review, we organize evidence around two scenarios based on the essential and protracted nature of GABA system development: 1) disruptions in the development of the GABA system can lead to large scale disruptions in other developmental processes (i.e., GABA as the cause), 2) protracted maturation of this system makes it vulnerable to the effects of developmental insults (i.e., GABA as the effect). While ample evidence supports the importance of GABA/GABAAR system in both scenarios, large gaps in existing knowledge prevent strong mechanistic conclusions.
Collapse
Affiliation(s)
- Irina Topchiy
- Division of Basic Neuroscience, McLean Hospital, Belmont, MA 02478, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02215, USA
| | - Julie Mohbat
- Division of Basic Neuroscience, McLean Hospital, Belmont, MA 02478, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02215, USA; School of Life Sciences, Ecole Polytechnique Federale de Lausanne, Lausanne CH-1015, Switzerland
| | - Oluwarotimi O Folorunso
- Division of Basic Neuroscience, McLean Hospital, Belmont, MA 02478, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02215, USA
| | - Ziyi Zephyr Wang
- Division of Basic Neuroscience, McLean Hospital, Belmont, MA 02478, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02215, USA
| | | | - Elif Engin
- Division of Basic Neuroscience, McLean Hospital, Belmont, MA 02478, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
2
|
Gillespie B, Panthi S, Sundram S, Hill RA. The impact of maternal immune activation on GABAergic interneuron development: A systematic review of rodent studies and their translational implications. Neurosci Biobehav Rev 2024; 156:105488. [PMID: 38042358 DOI: 10.1016/j.neubiorev.2023.105488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 11/09/2023] [Accepted: 11/27/2023] [Indexed: 12/04/2023]
Abstract
Mothers exposed to infections during pregnancy disproportionally birth children who develop autism and schizophrenia, disorders associated with altered GABAergic function. The maternal immune activation (MIA) model recapitulates this risk factor, with many studies also reporting disruptions to GABAergic interneuron expression, protein, cellular density and function. However, it is unclear if there are species, sex, age, region, or GABAergic subtype specific vulnerabilities to MIA. Furthermore, to fully comprehend the impact of MIA on the GABAergic system a synthesised account of molecular, cellular, electrophysiological and behavioural findings was required. To this end we conducted a systematic review of GABAergic interneuron changes in the MIA model, focusing on the prefrontal cortex and hippocampus. We reviewed 102 articles that revealed robust changes in a number of GABAergic markers that present as gestationally-specific, region-specific and sometimes sex-specific. Disruptions to GABAergic markers coincided with distinct behavioural phenotypes, including memory, sensorimotor gating, anxiety, and sociability. Findings suggest the MIA model is a valid tool for testing novel therapeutics designed to recover GABAergic function and associated behaviour.
Collapse
Affiliation(s)
- Brendan Gillespie
- Department of Psychiatry, School of Clinical Sciences, Monash University, Clayton, VIC 3168, Australia
| | - Sandesh Panthi
- Department of Psychiatry, School of Clinical Sciences, Monash University, Clayton, VIC 3168, Australia
| | - Suresh Sundram
- Department of Psychiatry, School of Clinical Sciences, Monash University, Clayton, VIC 3168, Australia
| | - Rachel A Hill
- Department of Psychiatry, School of Clinical Sciences, Monash University, Clayton, VIC 3168, Australia.
| |
Collapse
|
3
|
Raymann S, Schalbetter SM, Schaer R, Bernhardt AC, Mueller FS, Meyer U, Weber-Stadlbauer U. Late prenatal immune activation in mice induces transgenerational effects via the maternal and paternal lineages. Cereb Cortex 2023; 33:2273-2286. [PMID: 36857721 DOI: 10.1093/cercor/bhac207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 04/24/2022] [Accepted: 04/25/2022] [Indexed: 11/14/2022] Open
Abstract
Prenatal exposure to infectious or noninfectious immune activation is an environmental risk factor for neurodevelopmental disorders and mental illnesses. Recent research using animal models suggests that maternal immune activation (MIA) during early to middle stages of pregnancy can induce transgenerational effects on brain and behavior, likely via inducing stable epigenetic modifications across generations. Using a mouse model of viral-like MIA, which is based on gestational treatment with poly(I:C), the present study explored whether transgenerational effects can also emerge when MIA occurs in late pregnancy. Our findings demonstrate that the direct descendants born to poly(I:C)-treated mothers display deficits in temporal order memory, which are similarly present in second- and third-generation offspring. These transgenerational effects were mediated via both the maternal and paternal lineages and were accompanied by transient changes in maternal care. In addition to the cognitive effects, late prenatal immune activation induced generation-spanning effects on the prefrontal expression of gamma-aminobutyric acid (GABA)ergic genes, including parvalbumin and distinct alpha-subunits of the GABAA receptor. Together, our results suggest that MIA in late pregnancy has the potential to affect cognitive functions and prefrontal gene expression patterns in multiple generations, highlighting its role in shaping disease risk across generations.
Collapse
Affiliation(s)
- Stephanie Raymann
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Winterthurerstrasse 260, 8057 Zurich, Switzerland
| | - Sina M Schalbetter
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Winterthurerstrasse 260, 8057 Zurich, Switzerland
| | - Ron Schaer
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Winterthurerstrasse 260, 8057 Zurich, Switzerland
| | - Alexandra C Bernhardt
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Winterthurerstrasse 260, 8057 Zurich, Switzerland
| | - Flavia S Mueller
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Winterthurerstrasse 260, 8057 Zurich, Switzerland
| | - Urs Meyer
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Winterthurerstrasse 260, 8057 Zurich, Switzerland.,Neuroscience Center Zurich, University of Zurich and ETH, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Ulrike Weber-Stadlbauer
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Winterthurerstrasse 260, 8057 Zurich, Switzerland.,Neuroscience Center Zurich, University of Zurich and ETH, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| |
Collapse
|
4
|
Rong J, Yang Y, Liang M, Zhong H, Li Y, Zhu Y, Sha S, Chen L, Zhou R. Neonatal inflammation increases hippocampal KCC2 expression through methylation-mediated TGF-β1 downregulation leading to impaired hippocampal cognitive function and synaptic plasticity in adult mice. J Neuroinflammation 2023; 20:15. [PMID: 36691035 PMCID: PMC9872321 DOI: 10.1186/s12974-023-02697-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 01/12/2023] [Indexed: 01/25/2023] Open
Abstract
The mechanisms by which neonatal inflammation leads to cognitive deficits in adulthood remain poorly understood. Inhibitory GABAergic synaptic transmission plays a vital role in controlling learning, memory and synaptic plasticity. Since early-life inflammation has been reported to adversely affect the GABAergic synaptic transmission, the aim of this study was to investigate whether and how neonatal inflammation affects GABAergic synaptic transmission resulting in cognitive impairment. Neonatal mice received a daily subcutaneous injection of lipopolysaccharide (LPS, 50 μg/kg) or saline on postnatal days 3-5. It was found that blocking GABAergic synaptic transmission reversed the deficit in hippocampus-dependent memory or the induction failure of long-term potentiation in the dorsal CA1 in adult LPS mice. An increase of mIPSCs amplitude was further detected in adult LPS mice indicative of postsynaptic potentiation of GABAergic transmission. Additionally, neonatal LPS resulted in the increased expression and function of K+-Cl--cotransporter 2 (KCC2) and the decreased expression of transforming growth factor-beta 1 (TGF-β1) in the dorsal CA1 during adulthood. The local TGF-β1 overexpression improved KCC2 expression and function, synaptic plasticity and memory of adult LPS mice. Adult LPS mice show hypermethylation of TGFb1 promoter and negatively correlate with reduced TGF-β1 transcripts. 5-Aza-deoxycytidine restored the changes in TGFb1 promoter methylation and TGF-β1 expression. Altogether, the results suggest that hypermethylation-induced reduction of TGF-β1 leads to enhanced GABAergic synaptic inhibition through increased KCC2 expression, which is a underlying mechanism of neonatal inflammation-induced hippocampus-dependent memory impairment in adult mice.
Collapse
Affiliation(s)
- Jing Rong
- grid.89957.3a0000 0000 9255 8984Department of Physiology, Nanjing Medical University, Longmian Avenue 101, Jiangning District, Nanjing, 211166 Jiangsu China
| | - Yang Yang
- grid.89957.3a0000 0000 9255 8984Department of Physiology, Nanjing Medical University, Longmian Avenue 101, Jiangning District, Nanjing, 211166 Jiangsu China
| | - Min Liang
- grid.89957.3a0000 0000 9255 8984Department of Physiology, Nanjing Medical University, Longmian Avenue 101, Jiangning District, Nanjing, 211166 Jiangsu China
| | - Haiquan Zhong
- grid.89957.3a0000 0000 9255 8984Department of Physiology, Nanjing Medical University, Longmian Avenue 101, Jiangning District, Nanjing, 211166 Jiangsu China
| | - Yingchun Li
- grid.89957.3a0000 0000 9255 8984Department of Physiology, Nanjing Medical University, Longmian Avenue 101, Jiangning District, Nanjing, 211166 Jiangsu China
| | - Yichao Zhu
- grid.89957.3a0000 0000 9255 8984Department of Physiology, Nanjing Medical University, Longmian Avenue 101, Jiangning District, Nanjing, 211166 Jiangsu China
| | - Sha Sha
- grid.89957.3a0000 0000 9255 8984Department of Physiology, Nanjing Medical University, Longmian Avenue 101, Jiangning District, Nanjing, 211166 Jiangsu China
| | - Lei Chen
- grid.89957.3a0000 0000 9255 8984Department of Physiology, Nanjing Medical University, Longmian Avenue 101, Jiangning District, Nanjing, 211166 Jiangsu China
| | - Rong Zhou
- grid.89957.3a0000 0000 9255 8984Department of Physiology, Nanjing Medical University, Longmian Avenue 101, Jiangning District, Nanjing, 211166 Jiangsu China
| |
Collapse
|
5
|
Zhong H, Rong J, Yang Y, Liang M, Li Y, Zhou R. Neonatal inflammation via persistent TGF-β1 downregulation decreases GABA AR expression in basolateral amygdala leading to the imbalance of the local excitation-inhibition circuits and anxiety-like phenotype in adult mice. Neurobiol Dis 2022; 169:105745. [PMID: 35513229 DOI: 10.1016/j.nbd.2022.105745] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 04/26/2022] [Accepted: 04/27/2022] [Indexed: 11/29/2022] Open
Abstract
Neonatal inflammation can increase the risk of anxiety disorder in adulthood. The balance between glutamatergic excitatory and GABAergic inhibitory transmissions in the basolateral amygdala (BLA) plays a vital role in controlling anxiety state. Based on the reports that early-life inflammation had adverse effects on GABAergic system, the aim of this study was to investigate whether and how neonatal inflammation affects excitatory-inhibitory circuits in the BLA resulting in anxiety disorder. Neonatal mice received a daily subcutaneous injection of lipopolysaccharide (LPS, 50 μg/kg) or saline on postnatal days 3-5. LPS-treated mice developed anxiety behaviors accompanied by the hyperactivity of adrenal axis in adulthood. Electrophysiological study revealed the increase of postsynaptic neuronal excitability in the cortical-BLA excitatory synapses of LPS mice which could be recovered by bath-application of GABAAR agonist suggesting the impairment of GABAergic system in LPS mice. Compared with controls, GABAARα2 subunit expression and density of GABA-evoked current in BLA principal neurons were reduced in LPS mice. Additionally, neonatal LPS treatment resulted in the down-regulation of transforming growth factor-beta 1 (TGF-β1) expression and PKC signaling pathway in the adult BLA. The local TGF-β1 overexpression in the BLA improved GABAARα2 expression via up-regulating the activity of PKC signaling, which corrected GABAAR-mediated inhibition leading to the abolishment of anxiety-like change in adrenal axis regulation and behaviors in LPS mice. These data suggest the persistent TGF-β1deficit induces the down-regulation of GABAARα2 expression and subsequent disruption of the excitation-inhibition balance in the BLA circuits, which is the important mechanisms of neonatal inflammation-induced anxiety disorder.
Collapse
Affiliation(s)
- Haiquan Zhong
- Department of Physiology, Nanjing Medical University, Nanjing 211166, China
| | - Jing Rong
- Department of Physiology, Nanjing Medical University, Nanjing 211166, China
| | - Yang Yang
- Department of Physiology, Nanjing Medical University, Nanjing 211166, China
| | - Min Liang
- Department of Physiology, Nanjing Medical University, Nanjing 211166, China
| | - Yingchun Li
- Department of Physiology, Nanjing Medical University, Nanjing 211166, China
| | - Rong Zhou
- Department of Physiology, Nanjing Medical University, Nanjing 211166, China.
| |
Collapse
|
6
|
Stress induced microglial activation contributes to depression. Pharmacol Res 2022; 179:106145. [DOI: 10.1016/j.phrs.2022.106145] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 02/08/2022] [Accepted: 02/22/2022] [Indexed: 02/06/2023]
|
7
|
Blaylock RL, Faria M. New concepts in the development of schizophrenia, autism spectrum disorders, and degenerative brain diseases based on chronic inflammation: A working hypothesis from continued advances in neuroscience research. Surg Neurol Int 2021; 12:556. [PMID: 34877042 PMCID: PMC8645502 DOI: 10.25259/sni_1007_2021] [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: 10/04/2021] [Accepted: 10/05/2021] [Indexed: 12/14/2022] Open
Abstract
This paper was written prompted by a poignant film about adolescent girl with schizophrenia who babysits for a younger girl in an isolated cabin. Schizophrenia is an illness that both authors are fascinated with and that they continue to study and investigate. There is now compelling evidence that schizophrenia is a very complex syndrome that involves numerous neural pathways in the brain, far more than just dopaminergic and serotonergic systems. One of the more popular theories in recent literature is that it represents a hypo glutaminergic deficiency of certain pathways, including thalamic ones. After much review of research and study in this area, we have concluded that most such theories contain a number of shortcomings. Most are based on clinical responses to certain drugs, particularly antipsychotic drugs affecting the dopaminergic neurotransmitters; thus, assuming dopamine release was the central cause of the psychotic symptoms of schizophrenia. The theory was limited in that dopamine excess could only explain the positive symptoms of the disorder. Antipsychotic medications have minimal effectiveness for the negative and cognitive symptoms associated with schizophrenia. It has been estimated that 20–30% of patients show either a partial or no response to antipsychotic medications. In addition, the dopamine hypothesis does not explain the neuroanatomic findings in schizophrenia.
Collapse
Affiliation(s)
| | - Miguel Faria
- Clinical Professor of Surgery (Neurosurgery, ret.) and Adjunct Professor of Medical History (ret.), Mercer University School of Medicine, United States
| |
Collapse
|
8
|
Kassotaki I, Valsamakis G, Mastorakos G, Grammatopoulos DK. Placental CRH as a Signal of Pregnancy Adversity and Impact on Fetal Neurodevelopment. Front Endocrinol (Lausanne) 2021; 12:714214. [PMID: 34408727 PMCID: PMC8366286 DOI: 10.3389/fendo.2021.714214] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 07/09/2021] [Indexed: 11/13/2022] Open
Abstract
Early life is a period of considerable plasticity and vulnerability and insults during that period can disrupt the homeostatic equilibrium of the developing organism, resulting in adverse developmental programming and enhanced susceptibility to disease. Fetal exposure to prenatal stress can impede optimum brain development and deranged mother's hypothalamic-pituitary-adrenal axis (HPA axis) stress responses can alter the neurodevelopmental trajectories of the offspring. Corticotropin-releasing hormone (CRH) and glucocorticoids, regulate fetal neurogenesis and while CRH exerts neuroprotective actions, increased levels of stress hormones have been associated with fetal brain structural alterations such as reduced cortical volume, impoverishment of neuronal density in the limbic brain areas and alterations in neuronal circuitry, synaptic plasticity, neurotransmission and G-protein coupled receptor (GPCR) signalling. Emerging evidence highlight the role of epigenetic changes in fetal brain programming, as stress-induced methylation of genes encoding molecules that are implicated in HPA axis and major neurodevelopmental processes. These serve as molecular memories and have been associated with long term modifications of the offspring's stress regulatory system and increased susceptibility to psychosomatic disorders later in life. This review summarises our current understanding on the roles of CRH and other mediators of stress responses on fetal neurodevelopment.
Collapse
Affiliation(s)
- Ifigeneia Kassotaki
- Department of Internal Medicine, 2nd Internal Medicine Clinic, Venizeleio Pananeio General Hospital, Heraklion, Greece
| | - Georgios Valsamakis
- Second University Department of Obs and Gynae, Aretaieion Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- Translational Medicine, Warwick Medical School, Coventry, United Kingdom
| | - George Mastorakos
- Unit of Endocrinology, Diabetes Mellitus and Metabolism, Aretaieion Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitris K. Grammatopoulos
- Translational Medicine, Warwick Medical School, Coventry, United Kingdom
- Institute of Precision Diagnostics and Translational Medicine, Pathology, University Hospitals Coventry and Warwickshire (UHCW) NHS Trust, Coventry, United Kingdom
| |
Collapse
|
9
|
Woods RM, Lorusso JM, Potter HG, Neill JC, Glazier JD, Hager R. Maternal immune activation in rodent models: A systematic review of neurodevelopmental changes in gene expression and epigenetic modulation in the offspring brain. Neurosci Biobehav Rev 2021; 129:389-421. [PMID: 34280428 DOI: 10.1016/j.neubiorev.2021.07.015] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 05/11/2021] [Accepted: 07/11/2021] [Indexed: 01/06/2023]
Abstract
Maternal immune activation (mIA) during pregnancy is hypothesised to disrupt offspring neurodevelopment and predispose offspring to neurodevelopmental disorders such as schizophrenia. Rodent models of mIA have explored possible mechanisms underlying this paradigm and provide a vital tool for preclinical research. However, a comprehensive analysis of the molecular changes that occur in mIA-models is lacking, hindering identification of robust clinical targets. This systematic review assesses mIA-driven transcriptomic and epigenomic alterations in specific offspring brain regions. Across 118 studies, we focus on 88 candidate genes and show replicated changes in expression in critical functional areas, including elevated inflammatory markers, and reduced myelin and GABAergic signalling proteins. Further, disturbed epigenetic markers at nine of these genes support mIA-driven epigenetic modulation of transcription. Overall, our results demonstrate that current outcome measures have direct relevance for the hypothesised pathology of schizophrenia and emphasise the importance of mIA-models in contributing to the understanding of biological pathways impacted by mIA and the discovery of new drug targets.
Collapse
Affiliation(s)
- Rebecca M Woods
- Division of Evolution & Genomic Sciences, School of Biological Sciences, Manchester Academic Health Science Center, Faculty of Biology, Medicine & Health, University of Manchester, Manchester, M13 9PT, United Kingdom.
| | - Jarred M Lorusso
- Division of Evolution & Genomic Sciences, School of Biological Sciences, Manchester Academic Health Science Center, Faculty of Biology, Medicine & Health, University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Harry G Potter
- Division of Evolution & Genomic Sciences, School of Biological Sciences, Manchester Academic Health Science Center, Faculty of Biology, Medicine & Health, University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Joanna C Neill
- Division of Pharmacy & Optometry, School of Health Sciences, Manchester Academic Health Science Center, Faculty of Biology, Medicine & Health, University of Manchester, Manchester, M13 9PL, United Kingdom
| | - Jocelyn D Glazier
- Division of Evolution & Genomic Sciences, School of Biological Sciences, Manchester Academic Health Science Center, Faculty of Biology, Medicine & Health, University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Reinmar Hager
- Division of Evolution & Genomic Sciences, School of Biological Sciences, Manchester Academic Health Science Center, Faculty of Biology, Medicine & Health, University of Manchester, Manchester, M13 9PT, United Kingdom
| |
Collapse
|
10
|
Nakagawa K, Yoshino H, Ogawa Y, Yamamuro K, Kimoto S, Noriyama Y, Makinodan M, Yamashita M, Saito Y, Kishimoto T. Maternal Immune Activation Affects Hippocampal Excitatory and Inhibitory Synaptic Transmission in Offspring From an Early Developmental Period to Adulthood. Front Cell Neurosci 2020; 14:241. [PMID: 32903758 PMCID: PMC7438877 DOI: 10.3389/fncel.2020.00241] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 07/09/2020] [Indexed: 12/31/2022] Open
Abstract
One of the risk factors for schizophrenia is maternal infection. We have previously shown that Polyriboinosinic-polyribocytidylic acid (poly I:C) induced maternal immune activation in mice caused histological changes in the hippocampal CA1 area of offspring during the developmental period and impaired sensorimotor gating in offspring during adulthood, resulting in behavioral changes. However, it remains unclear how maternal immune activation functionally impacts the hippocampal neuronal activity of offspring. We studied the effect of prenatal poly I:C treatment on synaptic transmission of hippocampal CA1 pyramidal cells in postnatal and adult offspring. Treatment with poly I:C diminished excitatory and enhanced inhibitory (GABAergic) synaptic transmission on pyramidal cells in adult offspring. During the early developmental period, we still observed that treatment with poly I:C decreased excitatory synaptic transmission and potentially increased GABAergic synaptic transmission, which was uncovered under a condition of high extracellular potassium-activated neurons. In conclusion, we demonstrate that maternal immune activation decreased excitatory and increased inhibitory synaptic transmission on hippocampal pyramidal cells from an early developmental period to adulthood, which could result in net inhibition in conjunction with poor functional organization and integration of hippocampal circuits.
Collapse
Affiliation(s)
- Keiju Nakagawa
- Department of Psychiatry, Nara Medical University, Kashihara, Japan
| | - Hiroki Yoshino
- Department of Psychiatry, Nara Medical University, Kashihara, Japan
| | - Yoichi Ogawa
- Department of Neurophysiology, Nara Medical University, Kashihara, Japan
| | | | - Sohei Kimoto
- Department of Psychiatry, Nara Medical University, Kashihara, Japan
| | | | - Manabu Makinodan
- Department of Psychiatry, Nara Medical University, Kashihara, Japan
| | - Masayuki Yamashita
- Center for Medical Science, International University of Health and Welfare, Otawara, Japan
| | - Yasuhiko Saito
- Department of Neurophysiology, Nara Medical University, Kashihara, Japan
| | | |
Collapse
|
11
|
Chronic maternal interleukin-17 and autism-related cortical gene expression, neurobiology, and behavior. Neuropsychopharmacology 2020; 45:1008-1017. [PMID: 32074626 PMCID: PMC7162858 DOI: 10.1038/s41386-020-0640-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 02/06/2020] [Accepted: 02/11/2020] [Indexed: 12/18/2022]
Abstract
Chronic inflammation during pregnancy (e.g., preeclampsia, diabetes) is linked to increased risk for offspring neurodevelopmental disorders such as autism spectrum disorder (ASD). However, mediators of such exposures that could be targeted with maternal intervention are unclear, as few chronic gestational inflammation models have been tested. One potential mediator is interleukin-17 (IL-17), a pro-inflammatory cytokine implicated in neurodevelopmental disorders and gestational disease. To test chronic maternal IL-17 impacts on offspring, C57BL/6J dams were administered IL-17A continuously throughout pregnancy. Offspring were assessed for body weight; cortical volume, gene expression, and cellular composition; and adult behavior. IL-17A-condition offspring exhibited decreased somatic and cortical size at embryonic day 18 (E18) and as adults. mRNA sequencing of E18 cortex revealed 320 differentially expressed genes in males, but none in females. These were significantly enriched for ASD (Simons Foundation Autism Research Initiative), synaptic, and cell cycle genes. By adulthood, neocortical glial cell density and gene expression were decreased, while GABAergic synaptic gene expression was increased in males. Furthermore, IL-17A-condition male but not female offspring exhibited reduced anxiety-like behavior. Social approach deficits in males were negatively correlated with neocortical GABAergic synaptic gene expression. Chronic gestational IL-17A was sufficient to cause ASD-like phenotypes early and persistently in male offspring. This echoes the male bias, altered cortical development, and behavioral findings in ASD, suggesting that chronic maternal IL-17 contributes to offspring ASD pathogenesis. Furthermore, the trajectory from embryonically dysregulated synaptic and cell cycle genes to disrupted adult glia, inhibitory synapses, and behavior suggests a mechanism for chronic maternal IL-17 effects on offspring.
Collapse
|
12
|
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
|
13
|
Baratta AM, Kanyuch NR, Cole CA, Valafar H, Deslauriers J, Pocivavsek A. Acute sleep deprivation during pregnancy in rats: Rapid elevation of placental and fetal inflammation and kynurenic acid. Neurobiol Stress 2019; 12:100204. [PMID: 32258253 PMCID: PMC7109515 DOI: 10.1016/j.ynstr.2019.100204] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Revised: 11/27/2019] [Accepted: 12/11/2019] [Indexed: 01/19/2023] Open
Abstract
The kynurenine pathway (KP) is the dominant pathway for tryptophan degradation in the mammalian body and emerging evidence suggests that acute episodes of sleep deprivation (SD) disrupt tryptophan metabolism via the KP. Increases in the neuroactive KP metabolite kynurenic acid (KYNA) during pregnancy may lead to a higher risk for disrupted neurodevelopment in the offspring. As pregnancy is a critical period during which several factors, including sleep disruptions, could disrupt the fetal environment, we presently explored the relationship between maternal SD and KP metabolism and immune pathways in maternal, placenta, and fetal tissues. Pregnant Wistar rat dams were sleep deprived by gentle handling for 5 h from zeitgeber time (ZT) 0 to ZT 5. Experimental cohorts included: i) controls, ii) one session of SD on embryonic day (ED) 18 or iii) three sessions of SD occurring daily on ED 16, ED 17 and ED 18. Maternal (plasma, brain), placental and fetal (plasma, brain) tissues were collected immediately after the last session of SD or after 24 h of recovery from SD. Respective controls were euthanized at ZT 5 on ED 18 or ED 19. Maternal plasma corticosterone and fetal brain KYNA were significantly elevated only after one session of SD on ED 18. Importantly, maternal plasma corticosterone levels correlated significantly with fetal brain KYNA levels. In addition, placental levels of the proinflammatory cytokines interleukin-1β (IL-1β) and interleukin-6 (IL-6) were increased following maternal SD, suggesting a relationship between placental immune response to SD and fetal brain KYNA accumulation. Collectively, our results demonstrate that sleep loss during the last week of gestation can adversely impact maternal stress, placental immune function, and fetal brain KYNA levels. We introduce KYNA as a novel molecular target influenced by sleep loss during pregnancy. Prenatal sleep deprivation influences kynurenine pathway metabolism in utero. Fetal brain kynurenic acid (KYNA) is elevated after maternal sleep deprivation. Maternal plasma corticosterone is increased after sleep deprivation. Prenatal sleep deprivation induces placental and fetal brain cytokines. These data support an interplay with stress, in utero inflammation, and KYNA.
Collapse
Affiliation(s)
- Annalisa M Baratta
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Nickole R Kanyuch
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Casey A Cole
- College of Engineering and Computing, University of South Carolina, Columba, South Carolina, USA
| | - Homayoun Valafar
- College of Engineering and Computing, University of South Carolina, Columba, South Carolina, USA
| | - Jessica Deslauriers
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA.,Center of Excellence for Stress and Mental Health, Veterans Affairs Hospital, La Jolla, CA, USA
| | - Ana Pocivavsek
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA.,Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| |
Collapse
|
14
|
Liang M, Zhong H, Rong J, Li Y, Zhu C, Zhou L, Zhou R. Postnatal Lipopolysaccharide Exposure Impairs Adult Neurogenesis and Causes Depression-like Behaviors Through Astrocytes Activation Triggering GABAA Receptor Downregulation. Neuroscience 2019; 422:21-31. [DOI: 10.1016/j.neuroscience.2019.10.025] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 09/30/2019] [Accepted: 10/14/2019] [Indexed: 01/20/2023]
|
15
|
Osborne AL, Solowij N, Babic I, Lum JS, Huang XF, Newell KA, Weston-Green K. Cannabidiol improves behavioural and neurochemical deficits in adult female offspring of the maternal immune activation (poly I:C) model of neurodevelopmental disorders. Brain Behav Immun 2019; 81:574-587. [PMID: 31326506 DOI: 10.1016/j.bbi.2019.07.018] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 07/03/2019] [Accepted: 07/15/2019] [Indexed: 11/18/2022] Open
Abstract
Cognitive impairment is a major source of disability in schizophrenia and current antipsychotic drugs (APDs) have minimal efficacy for this symptom domain. Cannabidiol (CBD), the major non-intoxicating component of Cannabis sativa L., exhibits antipsychotic and neuroprotective properties. We recently reported the effects of CBD on cognition in male offspring of a maternal immune activation (polyinosinic-polycytidilic acid (poly I:C)) model relevant to the aetiology of schizophrenia; however, the effects of CBD treatment in females are unknown. Sex differences are observed in the onset of schizophrenia symptoms and response to APD treatment. Furthermore, the endogenous cannabinoid system, a direct target of CBD, is sexually dimorphic in humans and rodents. Therefore, the present work aimed to assess the therapeutic impact of CBD treatment on behaviour and neurochemical signalling markers in female poly I:C offspring. Time-mated pregnant Sprague-Dawley rats (n = 16) were administered poly I:C (4 mg/kg; i.v.) or saline (control) on gestational day 15. From postnatal day 56, female offspring received CBD (10 mg/kg, i.p.) or vehicle treatment for approximately 3 weeks. Following 2 weeks of CBD treatment, offspring underwent behavioural testing, including the novel object recognition, rewarded alternation T-maze and social interaction tests to assess recognition memory, working memory and sociability, respectively. After 3 weeks of CBD treatment, the prefrontal cortex (PFC) and hippocampus (HPC) were collected to assess effects on endocannabinoid, glutamatergic and gamma-aminobutyric acid (GABA) signalling markers. CBD attenuated poly I:C-induced deficits in recognition memory, social interaction and glutamatergic N-methyl-d-aspartate receptor (NMDAR) binding in the PFC of poly I:C offspring. Working memory performance was similar between treatment groups. CBD also increased glutamate decarboxylase 67, the rate-limiting enzyme that converts glutamate to GABA, and parvalbumin protein levels in the HPC. In contrast to the CBD treatment effects observed in poly I:C offspring, CBD administration to control rats reduced social interaction, cannabinoid CB1 receptor and NMDAR binding density in the PFC, suggesting that CBD administration to healthy rats may have negative consequences on social behaviour and brain maturation in adulthood. Overall, the findings of this study support the therapeutic benefits of CBD on recognition memory and sociability in female poly I:C offspring, and provide insight into the neurochemical changes that may underlie the therapeutic benefits of CBD in the poly I:C model.
Collapse
Affiliation(s)
- Ashleigh L Osborne
- Neuropharmacology and Molecular Psychiatry Laboratory, School of Medicine, University of Wollongong, Wollongong, NSW 2522, Australia; Centre for Translational Neuroscience, Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia; Molecular Horizons, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Nadia Solowij
- School of Psychology, Faculty of Social Sciences, University of Wollongong, and Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia; Australian Centre for Cannabinoid Clinical and Research Excellence, New Lambton Heights, NSW 2305, Australia
| | - Ilijana Babic
- Neuropharmacology and Molecular Psychiatry Laboratory, School of Medicine, University of Wollongong, Wollongong, NSW 2522, Australia; Centre for Translational Neuroscience, Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia; Molecular Horizons, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW 2522, Australia; Illawarra and Shoalhaven Local Health District, Wollongong, NSW 2500, Australia
| | - Jeremy S Lum
- Neuropharmacology and Molecular Psychiatry Laboratory, School of Medicine, University of Wollongong, Wollongong, NSW 2522, Australia; Centre for Translational Neuroscience, Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia; Molecular Horizons, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Xu-Feng Huang
- Centre for Translational Neuroscience, Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia; Molecular Horizons, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW 2522, Australia; Australian Centre for Cannabinoid Clinical and Research Excellence, New Lambton Heights, NSW 2305, Australia
| | - Kelly A Newell
- Neuropharmacology and Molecular Psychiatry Laboratory, School of Medicine, University of Wollongong, Wollongong, NSW 2522, Australia; Centre for Translational Neuroscience, Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia; Molecular Horizons, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Katrina Weston-Green
- Neuropharmacology and Molecular Psychiatry Laboratory, School of Medicine, University of Wollongong, Wollongong, NSW 2522, Australia; Centre for Translational Neuroscience, Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia; Molecular Horizons, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW 2522, Australia; Australian Centre for Cannabinoid Clinical and Research Excellence, New Lambton Heights, NSW 2305, Australia.
| |
Collapse
|
16
|
Dabbah-Assadi F, Alon D, Golani I, Doron R, Kremer I, Beloosesky R, Shamir A. The influence of immune activation at early vs late gestation on fetal NRG1-ErbB4 expression and behavior in juvenile and adult mice offspring. Brain Behav Immun 2019; 79:207-215. [PMID: 30738182 DOI: 10.1016/j.bbi.2019.02.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 01/28/2019] [Accepted: 02/05/2019] [Indexed: 01/01/2023] Open
Abstract
Maternal inflammation during pregnancy is associated with a higher incidence of mental disorders (e.g. schizophrenia and autism) in the offspring. In our study, we investigate the involvement of the NRG-ErbB signaling pathway in rodent fetal brains four hours following maternal immune activation (MIA) insult at two different gestational days (i.e. early vs late). Furthermore, we test the long-term behavioral alteration of the exposed MIA mice at juvenile and adulthood. We demonstrate that MIA at late, but not at early gestation day, altered the expression of NRG1, its receptor ErbB4, and the dopamine D2 receptor four hours post injection of viral or bacterial mimic material in fetal brain. At the behavioral levels, adult late-MIA-exposed female offspring, but not juvenile, display lack preference to a novel object. While working memory alteration observed only in adult male MIA-exposed offspring at late gestation day. In addition, we found that adult females MIA-exposed mice spent more time in the center of the open field than female-saline groups. On the other hand, juvenile male offspring exposed to MIA at early, but not late, gestation day displayed a significant alteration in social interaction. Our results suggest that MIA during late gestation immediately influences the expression levels of the NRG1 and ErbB4 genes, and affects long-term behavioral changes at adulthood. These behavioral changes are time related and sex-specific. Thus, immune activation at late stages of the embryonic brain development initiates the activation of the NRG1-ErbB4 pathway and this disturbance might result in cognitive dysfunction in adulthood.
Collapse
Affiliation(s)
- F Dabbah-Assadi
- Psychobiology Research Laboratory, Mazor Mental Health Center, Akko, Israel; The Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - D Alon
- Psychobiology Research Laboratory, Mazor Mental Health Center, Akko, Israel
| | - I Golani
- Department of Biotechnology Engineering, ORT Braude College, Karmiel, Israel
| | - R Doron
- Psychobiology Laboratory, School of Behavioral Sciences, The Academic College of Tel Aviv-Yaffo, Israel; Department of Education and Psychology, The Open University, Raanana, Israel
| | - I Kremer
- Psychobiology Research Laboratory, Mazor Mental Health Center, Akko, Israel; The Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - R Beloosesky
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel; Department of Obstetrics and Gynecology, Rambam Medical Center, Haifa, Israel
| | - A Shamir
- Psychobiology Research Laboratory, Mazor Mental Health Center, Akko, Israel; The Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
17
|
Dunn GA, Nigg JT, Sullivan EL. Neuroinflammation as a risk factor for attention deficit hyperactivity disorder. Pharmacol Biochem Behav 2019; 182:22-34. [PMID: 31103523 PMCID: PMC6855401 DOI: 10.1016/j.pbb.2019.05.005] [Citation(s) in RCA: 138] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Revised: 05/08/2019] [Accepted: 05/14/2019] [Indexed: 01/08/2023]
Abstract
Attention Deficit Hyperactivity Disorder (ADHD) is a persistent, and impairing pediatric-onset neurodevelopmental condition. Its high prevalence, and recurrent controversy over its widespread identification and treatment, drive strong interest in its etiology and mechanisms. Emerging evidence for a role for neuroinflammation in ADHD pathophysiology is of great interest. This evidence includes 1) the above-chance comorbidity of ADHD with inflammatory and autoimmune disorders, 2) initial studies indicating an association with ADHD and increased serum cytokines, 3) preliminary evidence from genetic studies demonstrating associations between polymorphisms in genes associated with inflammatory pathways and ADHD, 4) emerging evidence that early life exposure to environmental factors may increase risk for ADHD via an inflammatory mechanism, and 5) mechanistic evidence from animal models of maternal immune activation documenting behavioral and neural outcomes consistent with ADHD. Prenatal exposure to inflammation is associated with changes in offspring brain development including reductions in cortical gray matter volume and the volume of certain cortical areas -parallel to observations associated with ADHD. Alterations in neurotransmitter systems, including the dopaminergic, serotonergic and glutamatergic systems, are observed in ADHD populations. Animal models provide strong evidence that development and function of these neurotransmitters systems are sensitive to exposure to in utero inflammation. In summary, accumulating evidence from human studies and animal models, while still incomplete, support a potential role for neuroinflammation in the pathophysiology of ADHD. Confirmation of this association and the underlying mechanisms have become valuable targets for research. If confirmed, such a picture may be important in opening new intervention routes.
Collapse
Affiliation(s)
| | - Joel T Nigg
- Oregon Health and Science University, United States of America
| | - Elinor L Sullivan
- University of Oregon, United States of America; Oregon Health and Science University, United States of America; Oregon National Primate Research Center, United States of America.
| |
Collapse
|
18
|
Okojie AK, Rauf K, Iyare E. The Impact of Plasmodium Berghei Exposure In-utero on Neurobehavioral Profile in Mice. Basic Clin Neurosci 2019; 10:99-107. [PMID: 31031897 PMCID: PMC6484196 DOI: 10.32598/bcn.9.10.95] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 02/27/2018] [Accepted: 05/26/2018] [Indexed: 12/25/2022] Open
Abstract
Introduction: The World Health Organization estimates that about 25 million pregnant mothers are currently at risk for malaria, and that malaria accounts for over 10,000 maternal and 200,000 neonatal deaths per year. The current hypothesis of early life programming supports the premise that many developmental delay and disorders may have their origin In-utero. Therefore, the current study aimed at evaluating the possible impact of experimental malaria exposure In-utero on neurobehavioral profile in mice offspring. Methods: Pregnant mice were intraperitoneally infected on gestational day 13 with 1.02×105 infected red blood cells. Pregnant mice (both infected and uninfected) were allowed to deliver and the offspring were monitored up to postnatal day 42 when anxiety-like, Obsessive-Compulsive Disorder (OCD), and locomotor activity were evaluated using elevated plus maze, marble burying, and Open Field Test, respectively. Results: The current study showed that maternal infection with Plasmodium berghei resulted in an interesting behavior in offspring characterized by increased anxiety-like and OCD behaviors. Locomotor activity was however not affected. Conclusion: It may be concluded that In-utero exposure to experimental malaria in mice causes behavioral changes.
Collapse
Affiliation(s)
- Akhabue Keneth Okojie
- Reproductive and Developmental Programming Research Group, Department of Physiology, Enugu Campus, University of Nigeria, Enugu, Nigeria.,Department of Pharmacy, COMSATS Institute of Information Technology, Abbottabad, Pakistan.,Department of Physiology, Edo University Iyamho, Uzairue, Edo State, Nigeria
| | - Khalid Rauf
- Department of Pharmacy, COMSATS Institute of Information Technology, Abbottabad, Pakistan
| | - Eghosa Iyare
- Reproductive and Developmental Programming Research Group, Department of Physiology, Enugu Campus, University of Nigeria, Enugu, Nigeria
| |
Collapse
|
19
|
Missault S, Anckaerts C, Ahmadoun S, Blockx I, Barbier M, Bielen K, Shah D, Kumar-Singh S, De Vos WH, Van der Linden A, Dedeurwaerdere S, Verhoye M. Hypersynchronicity in the default mode-like network in a neurodevelopmental animal model with relevance for schizophrenia. Behav Brain Res 2019; 364:303-316. [PMID: 30807809 DOI: 10.1016/j.bbr.2019.02.040] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 02/22/2019] [Accepted: 02/22/2019] [Indexed: 12/31/2022]
Abstract
BACKGROUND Immune activation during pregnancy is an important risk factor for schizophrenia. Brain dysconnectivity and NMDA receptor (NMDAR) hypofunction have been postulated to be central to schizophrenia pathophysiology. The aim of this study was to investigate resting-state functional connectivity (resting-state functional MRI-rsfMRI), microstructure (diffusion tension imaging-DTI) and response to NMDAR antagonist (pharmacological fMRI-phMRI) using multimodal MRI in offspring of pregnant dams exposed to immune challenge (maternal immune activation-MIA model), and determine whether these neuroimaging readouts correlate with schizophrenia-related behaviour. METHODS Pregnant rats were injected with Poly I:C or saline on gestational day 15. The maternal weight response was assessed. Since previous research has shown behavioural deficits can differ between MIA offspring dependent on the maternal response to immune stimulus, offspring were divided into three groups: controls (saline, n = 11), offspring of dams that gained weight (Poly I:C WG, n = 12) and offspring of dams that lost weight post-MIA (Poly I:C WL, n = 16). Male adult offspring were subjected to rsfMRI, DTI, phMRI with NMDAR antagonist, behavioural testing and histological assessment. RESULTS Poly I:C WL offspring exhibited increased functional connectivity in default mode-like network (DMN). Poly I:C WG offspring showed the most pronounced attenuation in NMDAR antagonist response versus controls. DTI revealed no differences in Poly I:C offspring versus controls. Poly I:C offspring exhibited anxiety. CONCLUSIONS MIA offspring displayed a differential pathophysiology depending on the maternal response to immune challenge. While Poly I:C WL offspring displayed hypersynchronicity in the DMN, altered NMDAR antagonist response was most pronounced in Poly I:C WG offspring.
Collapse
Affiliation(s)
- Stephan Missault
- Experimental Laboratory of Translational Neuroscience and Otolaryngology, Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium; Bio-Imaging Lab, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium.
| | - Cynthia Anckaerts
- Bio-Imaging Lab, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Soumaya Ahmadoun
- Experimental Laboratory of Translational Neuroscience and Otolaryngology, Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Ines Blockx
- Bio-Imaging Lab, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Michaël Barbier
- Laboratory of Cell Biology and Histology, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Kenny Bielen
- Molecular Pathology Group, Laboratory of Cell Biology and Histology, Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Disha Shah
- Bio-Imaging Lab, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Samir Kumar-Singh
- Molecular Pathology Group, Laboratory of Cell Biology and Histology, Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Winnok H De Vos
- Laboratory of Cell Biology and Histology, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium; Cell Systems & Imaging, Faculty of Bioscience Engineering, University of Ghent, Coupure Links 653, 9000 Gent, Belgium
| | - Annemie Van der Linden
- Bio-Imaging Lab, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Stefanie Dedeurwaerdere
- Experimental Laboratory of Hematology, Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Marleen Verhoye
- Bio-Imaging Lab, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| |
Collapse
|
20
|
Abstract
Hippocampal abnormalities have been heavily implicated in the pathophysiology of schizophrenia. The dentate gyrus of the hippocampus was shown to manifest an immature molecular profile in schizophrenia subjects, as well as in various animal models of the disorder. In this position paper, we advance a hypothesis that this immature molecular profile is accompanied by an identifiable immature morphology of the dentate gyrus granule cell layer. We adduce evidence for arrested maturation of the dentate gyrus in the human schizophrenia-affected brain, as well as multiple rodent models of the disease. Implications of this neurohistopathological signature for current theory regarding the development of schizophrenia are discussed.
Collapse
Affiliation(s)
- Ayda Tavitian
- Department of Neurology & Neurosurgery, Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
| | - Wei Song
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
| | - Hyman M. Schipper
- Department of Neurology & Neurosurgery, Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
- Department of Medicine, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
21
|
Duchatel RJ, Meehan CL, Harms LR, Michie PT, Bigland MJ, Smith DW, Walker FR, Jobling P, Hodgson DM, Tooney PA. Late gestation immune activation increases IBA1-positive immunoreactivity levels in the corpus callosum of adult rat offspring. Psychiatry Res 2018; 266:175-185. [PMID: 29864618 DOI: 10.1016/j.psychres.2018.05.063] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 03/20/2018] [Accepted: 05/23/2018] [Indexed: 11/17/2022]
Abstract
Animal models of maternal immune activation study the effects of infection, an environmental risk factor for schizophrenia, on brain development. Microglia activation and cytokine upregulation may have key roles in schizophrenia neuropathology. We hypothesised that maternal immune activation induces changes in microglia and cytokines in the brains of the adult offspring. Maternal immune activation was induced by injecting polyriboinosinic:polyribocytidylic acid into pregnant rats on gestational day (GD) 10 or GD19, with brain tissue collected from the offspring at adulthood. We observed no change in Iba1, Gfap, IL1-β and TNF-α mRNA levels in the cingulate cortex (CC) in adult offspring exposed to maternal immune activation. Prenatal exposure to immune activation had a significant main effect on microglial IBA1-positive immunoreactive material (IBA1+IRM) in the corpus callosum; post-hoc analyses identified a significant increase in GD19 offspring, but not GD10. No change in was observed in the CC. In contrast, maternal immune activation had a significant main effect on GFAP+IRM in the CC at GD19 (not GD10); post-hoc analyses only identified a strong trend towards increased GFAP+IRM in the GD19 offspring, with no white matter changes. This suggests late gestation maternal immune activation causes subtle alterations to microglia and astrocytes in the adult offspring.
Collapse
Affiliation(s)
- Ryan J Duchatel
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW 2308, Australia; Priority Centre for Brain and Mental Health Research, University of Newcastle, Callaghan, NSW 2308, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia.
| | - Crystal L Meehan
- School of Psychology, Faculty of Science, University of Newcastle, Callaghan, NSW 2308, Australia; Priority Centre for Brain and Mental Health Research, University of Newcastle, Callaghan, NSW 2308, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia.
| | - Lauren R Harms
- School of Psychology, Faculty of Science, University of Newcastle, Callaghan, NSW 2308, Australia; Priority Centre for Brain and Mental Health Research, University of Newcastle, Callaghan, NSW 2308, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia.
| | - Patricia T Michie
- School of Psychology, Faculty of Science, University of Newcastle, Callaghan, NSW 2308, Australia; Priority Centre for Brain and Mental Health Research, University of Newcastle, Callaghan, NSW 2308, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia.
| | - Mark J Bigland
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW 2308, Australia; Priority Centre for Brain and Mental Health Research, University of Newcastle, Callaghan, NSW 2308, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia.
| | - Doug W Smith
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW 2308, Australia; Priority Centre for Brain and Mental Health Research, University of Newcastle, Callaghan, NSW 2308, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia.
| | - Frederick R Walker
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW 2308, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia.
| | - Phillip Jobling
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW 2308, Australia; Priority Centre for Brain and Mental Health Research, University of Newcastle, Callaghan, NSW 2308, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia.
| | - Deborah M Hodgson
- School of Psychology, Faculty of Science, University of Newcastle, Callaghan, NSW 2308, Australia; Priority Centre for Brain and Mental Health Research, University of Newcastle, Callaghan, NSW 2308, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia.
| | - Paul A Tooney
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW 2308, Australia; Priority Centre for Brain and Mental Health Research, University of Newcastle, Callaghan, NSW 2308, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia.
| |
Collapse
|
22
|
Hsueh PT, Lin HH, Wang HH, Liu CL, Ni WF, Liu JK, Chang HH, Sun DS, Chen YS, Chen YL. Immune imbalance of global gene expression, and cytokine, chemokine and selectin levels in the brains of offspring with social deficits via maternal immune activation. GENES BRAIN AND BEHAVIOR 2018; 17:e12479. [PMID: 29656594 DOI: 10.1111/gbb.12479] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 02/27/2018] [Accepted: 04/05/2018] [Indexed: 12/13/2022]
Abstract
The murine maternal immune activation (MIA) offspring model enables longitudinal studies to explore aberrant social behaviors similar to those observed in humans. High levels of cytokines, chemokines and cell adhesion molecules (CAM) have been found in the plasma and/or brains of psychiatric patients. We hypothesized that upregulation of the systemic or brain immune response has an augmenting effect by potentially increasing the interplay between the neuronal and immune systems during the growth of the MIA offspring. In this study, a C57BL/6j MIA female offspring model exhibiting social deficits was established. The expression of fetal interferon (IFN)-stimulated (gbp3, irgm1, ifi44), adolescent immunodevelopmental transcription factor (eg, r2, tfap2b), hormone (pomc, hcrt), adult selectin (sell, selp) and neuroligin (nlgn2) genes was altered. Systemic upregulation of endogenous IL-10 occurred at the adult stage, while both IL-1β and IL-6 were increased and persisted in the sera throughout the growth of the MIA offspring. The cerebral IL-6 levels were endogenously upregulated, but both MCP-1 (macrophage inflammatory protein-1) and L-selectin levels were downregulated at the adolescent and/or adult stages. However, the MIA offspring were susceptible to lipopolysaccharide (LPS) stimulation. After reinjecting the MIA offspring with LPS in adulthood, a variety of sera and cerebral cytokines, chemokines and CAMs were increased. Particularly, both MCP-1 and L-selectin showed relatively high expression in the brain compared with the expression levels in phosphate-buffered saline (PBS)-treated offspring injected with LPS. Potentially, MCP-1 was attracted to the L-selectin-mediated immune cells due to augmentation of the immune response following stimulation in MIA female offspring.
Collapse
Affiliation(s)
- P-T Hsueh
- Department of Internal Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - H-H Lin
- Department of Internal Medicine, National Yang-Ming University, Taipei, Taiwan.,Section of Infectious Disease, Department of Medicine, E-Da Hospital, Kaohsiung, Taiwan
| | - H-H Wang
- Department of Biotechnology, National Kaohsiung Normal University, Kaohsiung, Taiwan
| | - C-L Liu
- Department of Biotechnology, National Kaohsiung Normal University, Kaohsiung, Taiwan
| | - W-F Ni
- Department of Biotechnology, National Kaohsiung Normal University, Kaohsiung, Taiwan
| | - J-K Liu
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - H-H Chang
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien, Taiwan
| | - D-S Sun
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien, Taiwan
| | - Y-S Chen
- Department of Internal Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Y-L Chen
- Department of Biotechnology, National Kaohsiung Normal University, Kaohsiung, Taiwan
| |
Collapse
|
23
|
Graham AM, Rasmussen JM, Rudolph MD, Heim CM, Gilmore JH, Styner M, Potkin SG, Entringer S, Wadhwa PD, Fair DA, Buss C. Maternal Systemic Interleukin-6 During Pregnancy Is Associated With Newborn Amygdala Phenotypes and Subsequent Behavior at 2 Years of Age. Biol Psychiatry 2018; 83:109-119. [PMID: 28754515 PMCID: PMC5723539 DOI: 10.1016/j.biopsych.2017.05.027] [Citation(s) in RCA: 200] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 05/09/2017] [Accepted: 05/17/2017] [Indexed: 12/11/2022]
Abstract
BACKGROUND Maternal inflammation during pregnancy increases the risk for offspring psychiatric disorders and other adverse long-term health outcomes. The influence of inflammation on the developing fetal brain is hypothesized as one potential mechanism but has not been examined in humans. METHODS Participants were adult women (N = 86) who were recruited during early pregnancy and whose offspring were born after 34 weeks' gestation. A biological indicator of maternal inflammation (interleukin-6) that has been shown to influence fetal brain development in animal models was quantified serially in early, mid-, and late pregnancy. Structural and functional brain magnetic resonance imaging scans were acquired in neonates shortly after birth. Infants' amygdalae were individually segmented for measures of volume and as seeds for resting state functional connectivity. At 24 months of age, children completed a snack delay task to assess impulse control. RESULTS Higher average maternal interleukin-6 concentration during pregnancy was prospectively associated with larger right amygdala volume and stronger bilateral amygdala connectivity to brain regions involved in sensory processing and integration (fusiform, somatosensory cortex, and thalamus), salience detection (anterior insula), and learning and memory (caudate and parahippocampal gyrus). Larger newborn right amygdala volume and stronger left amygdala connectivity were in turn associated with lower impulse control at 24 months of age, and mediated the association between higher maternal interleukin-6 concentrations and lower impulse control. CONCLUSIONS These findings provide new evidence in humans linking maternal inflammation during pregnancy with newborn brain and emerging behavioral phenotypes relevant for psychiatric disorders. A better understanding of intrauterine conditions that influence offspring disease susceptibility is warranted to inform targeted early intervention and prevention efforts.
Collapse
Affiliation(s)
- Alice M Graham
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon
| | - Jerod M Rasmussen
- Development, Health and Disease Research Program, University of California, Irvine, Irvine, California
| | - Marc D Rudolph
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon
| | - Christine M Heim
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Medical Psychology, Berlin, Germany; Department of Biobehavioral Health, Pennsylvania State University, University Park, Pennsylvania
| | - John H Gilmore
- Department of Psychiatry, University of North Carolina, Chapel Hill, North Carolina
| | - Martin Styner
- Department of Psychiatry, University of North Carolina, Chapel Hill, North Carolina
| | - Steven G Potkin
- Department of Psychiatry and Human Behavior, University of California, Irvine, Irvine, California
| | - Sonja Entringer
- Development, Health and Disease Research Program, University of California, Irvine, Irvine, California; Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Medical Psychology, Berlin, Germany
| | - Pathik D Wadhwa
- Development, Health and Disease Research Program, University of California, Irvine, Irvine, California
| | - Damien A Fair
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon; Advanced Imaging Research Center, Oregon Health & Science University, Portland, Oregon
| | - Claudia Buss
- Development, Health and Disease Research Program, University of California, Irvine, Irvine, California; Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Medical Psychology, Berlin, Germany.
| |
Collapse
|
24
|
Konefal SC, Stellwagen D. Tumour necrosis factor-mediated homeostatic synaptic plasticity in behavioural models: testing a role in maternal immune activation. Philos Trans R Soc Lond B Biol Sci 2017; 372:rstb.2016.0160. [PMID: 28093554 DOI: 10.1098/rstb.2016.0160] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/09/2016] [Indexed: 12/25/2022] Open
Abstract
The proinflammatory cytokine tumour necrosis factor-alpha (TNFα) has long been characterized for its role in the innate immune system, but more recently has been found to have a distinct role in the nervous system that does not overlap with other proinflammatory cytokines. Through regulation of neuronal glutamate and GABA receptor trafficking, TNF mediates a homeostatic form of synaptic plasticity, but plays no direct role in Hebbian forms of plasticity. As yet, there is no evidence to suggest that this adaptive plasticity plays a significant role in normal development, but it does maintain neuronal circuit function in the face of several types of disruption. This includes developmental plasticity in primary sensory cortices, as well as modulating the response to antidepressants, chronic antipsychotics and drugs of abuse. TNF is also a prominent component of the neuroinflammation occurring in most neuropathologies, but the role of TNF-mediated synaptic plasticity in this context remains to be determined. We tested this in a maternal immune activation (MIA) model of neurodevelopmental disorders. Using TNF-/- mice, we observed that TNF is not required for the expression of abnormal social or anxious behaviour in this model. This indicates that TNF does not uniquely contribute to the development of neuronal dysfunction in this model, and suggests that during neuroinflammatory events, compensation between the various proinflammatory cytokines is the norm.This article is part of the themed issue 'Integrating Hebbian and homeostatic plasticity'.
Collapse
Affiliation(s)
- Sarah C Konefal
- Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, The Research Institute of the McGill University Health Center, Montreal, Quebec, Canada H3G 1A4
| | - David Stellwagen
- Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, The Research Institute of the McGill University Health Center, Montreal, Quebec, Canada H3G 1A4
| |
Collapse
|
25
|
Crum WR, Sawiak SJ, Chege W, Cooper JD, Williams SC, Vernon AC. Evolution of structural abnormalities in the rat brain following in utero exposure to maternal immune activation: A longitudinal in vivo MRI study. Brain Behav Immun 2017; 63:50-59. [PMID: 27940258 PMCID: PMC5441572 DOI: 10.1016/j.bbi.2016.12.008] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 11/07/2016] [Accepted: 12/07/2016] [Indexed: 02/08/2023] Open
Abstract
Genetic and environmental risk factors for psychiatric disorders are suggested to disrupt the trajectory of brain maturation during adolescence, leading to the development of psychopathology in adulthood. Rodent models are powerful tools to dissect the specific effects of such risk factors on brain maturational profiles, particularly when combined with Magnetic Resonance Imaging (MRI; clinically comparable technology). We therefore investigated the effect of maternal immune activation (MIA), an epidemiological risk factor for adult-onset psychiatric disorders, on rat brain maturation using atlas and tensor-based morphometry analysis of longitudinal in vivo MR images. Exposure to MIA resulted in decreases in the volume of several cortical regions, the hippocampus, amygdala, striatum, nucleus accumbens and unexpectedly, the lateral ventricles, relative to controls. In contrast, the volumes of the thalamus, ventral mesencephalon, brain stem and major white matter tracts were larger, relative to controls. These volumetric changes were maximal between post-natal day 50 and 100 with no differences between the groups thereafter. These data are consistent with and extend prior studies of brain structure in MIA-exposed rodents. Apart from the ventricular findings, these data have robust face validity to clinical imaging findings reported in studies of individuals at high clinical risk for a psychiatric disorder. Further work is now required to address the relationship of these MRI changes to behavioral dysfunction and to establish thier cellular correlates.
Collapse
Affiliation(s)
- William R. Crum
- Department of Neuroimaging Institute of Psychiatry, Psychology and Neuroscience, King’s College London, De Crespigny Park, London SE5 8AF, UK
| | - Stephen J. Sawiak
- Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Addenbrooke’s Hospital, Hills Road, Cambridge, UK
| | - Winfred Chege
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, De Crespigny Park, London SE5 8AF, UK
| | - Jonathan D. Cooper
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, Maurice Wohl Clinical Neuroscience Institute, 5 Cutcombe Road, London SE5 9RT, UK
| | - Steven C.R. Williams
- Department of Neuroimaging Institute of Psychiatry, Psychology and Neuroscience, King’s College London, De Crespigny Park, London SE5 8AF, UK,MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK
| | - Anthony C. Vernon
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, Maurice Wohl Clinical Neuroscience Institute, 5 Cutcombe Road, London SE5 9RT, UK,MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK,Corresponding author at: Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, Maurice Wohl Clinical Neuroscience Institute, 5 Cutcombe Road, London SE5 9RT, UK.Department of Basic and Clinical NeuroscienceInstitute of PsychiatryPsychology and NeuroscienceKing’s College LondonMaurice Wohl Clinical Neuroscience Institute5 Cutcombe RoadLondonSE5 9RTUK
| |
Collapse
|
26
|
Ronovsky M, Berger S, Molz B, Berger A, Pollak DD. Animal Models of Maternal Immune Activation in Depression Research. Curr Neuropharmacol 2017; 14:688-704. [PMID: 26666733 PMCID: PMC5050397 DOI: 10.2174/1570159x14666151215095359] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 10/24/2015] [Accepted: 11/09/2015] [Indexed: 01/17/2023] Open
Abstract
Abstract: Background Depression and schizophrenia are debilitating mental illnesses with significant socio-economic impact. The high degree of comorbidity between the two disorders, and shared symptoms and risk factors, suggest partly common pathogenic mechanisms. Supported by human and animal studies, maternal immune activation (MIA) has been intimately associated with the development of schizophrenia. However, the link between MIA and depression has remained less clear, in part due to the lack of appropriate animal models. Objective Here we aim to summarize findings obtained from studies using MIA animal models and discuss their relevance for preclinical depression research. Methods Results on molecular, cellular and behavioral phenotypes in MIA animal models were collected by literature search (PubMed) and evaluated for their significance for depression. Results Several reports on offspring depression-related behavioral alterations indicate an involvement of MIA in the development of depression later in life. Depression-related behavioral phenotypes were frequently paralleled by neurogenic and neurotrophic deficits and modulated by several genetic and environmental factors. Conclusion Literature evidence analyzed in this review supports a relevance of MIA as animal model for a specific early life adversity, which may prime an individual for the development of distinct psychopathologies later life. MIA animal models may present a unique tool for the identification of additional exogenous and endogenous factors, which are required for the manifestation of a specific neuropsychiatric disorder, such as depression, later in life. Hereby, novel insights into the molecular mechanisms involved in the pathophysiology of depression may be obtained, supporting the identification of alternative therapeutic strategies.
Collapse
Affiliation(s)
| | | | | | | | - Daniela D Pollak
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstrasse 17, A-1090 Vienna, Austria
| |
Collapse
|
27
|
Transgenerational transmission and modification of pathological traits induced by prenatal immune activation. Mol Psychiatry 2017; 22:102-112. [PMID: 27021823 DOI: 10.1038/mp.2016.41] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 02/05/2016] [Accepted: 02/17/2016] [Indexed: 12/16/2022]
Abstract
Prenatal exposure to infectious or inflammatory insults is increasingly recognized to contribute to the etiology of psychiatric disorders with neurodevelopmental components, including schizophrenia, autism and bipolar disorder. It remains unknown, however, if such immune-mediated brain anomalies can be transmitted to subsequent generations. Using an established mouse model of prenatal immune activation by the viral mimetic poly(I:C), we show that reduced sociability and increased cued fear expression are similarly present in the first- and second-generation offspring of immune-challenged ancestors. We further demonstrate that sensorimotor gating impairments are confined to the direct descendants of infected mothers, whereas increased behavioral despair emerges as a novel phenotype in the second generation. These transgenerational effects are mediated via the paternal lineage and are stable until the third generation, demonstrating transgenerational non-genetic inheritance of pathological traits following in-utero immune activation. Next-generation sequencing further demonstrated unique and overlapping genome-wide transcriptional changes in first- and second-generation offspring of immune-challenged ancestors. These transcriptional effects mirror the transgenerational effects on behavior, showing that prenatal immune activation leads to a transgenerational transmission (presence of similar phenotypes across generations) and modification (presence of distinct phenotypes across generations) of pathological traits. Together, our study demonstrates for, we believe, the first time that prenatal immune activation can negatively affect brain and behavioral functions in multiple generations. These findings thus highlight a novel pathological aspect of this early-life adversity in shaping disease risk across generations.
Collapse
|
28
|
Giovanoli S, Weber-Stadlbauer U, Schedlowski M, Meyer U, Engler H. Prenatal immune activation causes hippocampal synaptic deficits in the absence of overt microglia anomalies. Brain Behav Immun 2016; 55:25-38. [PMID: 26408796 DOI: 10.1016/j.bbi.2015.09.015] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 09/22/2015] [Accepted: 09/23/2015] [Indexed: 12/11/2022] Open
Abstract
Prenatal exposure to infectious or inflammatory insults can increase the risk of developing neuropsychiatric disorder in later life, including schizophrenia, bipolar disorder, and autism. These brain disorders are also characterized by pre- and postsynaptic deficits. Using a well-established mouse model of maternal exposure to the viral mimetic polyriboinosinic-polyribocytidilic acid [poly(I:C)], we examined whether prenatal immune activation might cause synaptic deficits in the hippocampal formation of pubescent and adult offspring. Based on the widely appreciated role of microglia in synaptic pruning, we further explored possible associations between synaptic deficits and microglia anomalies in offspring of poly(I:C)-exposed and control mothers. We found that prenatal immune activation induced an adult onset of presynaptic hippocampal deficits (as evaluated by synaptophysin and bassoon density). The early-life insult further caused postsynaptic hippocampal deficits in pubescence (as evaluated by PSD95 and SynGAP density), some of which persisted into adulthood. In contrast, prenatal immune activation did not change microglia (or astrocyte) density, nor did it alter their activation phenotypes. The prenatal manipulation did also not cause signs of persistent systemic inflammation. Despite the absence of overt glial anomalies or systemic inflammation, adult offspring exposed to prenatal immune activation displayed increased hippocampal IL-1β levels. Taken together, our findings demonstrate that age-dependent synaptic deficits and abnormal pro-inflammatory cytokine expression can occur during postnatal brain maturation in the absence of microglial anomalies or systemic inflammation.
Collapse
Affiliation(s)
- Sandra Giovanoli
- Physiology and Behavior Laboratory, ETH Zurich, Schwerzenbach, Switzerland
| | - Ulrike Weber-Stadlbauer
- Physiology and Behavior Laboratory, ETH Zurich, Schwerzenbach, Switzerland; Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland
| | - Manfred Schedlowski
- Institute of Medical Psychology and Behavioral Immunobiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Urs Meyer
- Physiology and Behavior Laboratory, ETH Zurich, Schwerzenbach, Switzerland; Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland.
| | - Harald Engler
- Institute of Medical Psychology and Behavioral Immunobiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
29
|
Meyer U, Yee BK, Feldon J. The Neurodevelopmental Impact of Prenatal Infections at Different Times of Pregnancy: The Earlier the Worse? Neuroscientist 2016; 13:241-56. [PMID: 17519367 DOI: 10.1177/1073858406296401] [Citation(s) in RCA: 188] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Environmental insults taking place in early brain development may have long-lasting consequences for adult brain functioning. There is a large body of epidemiological data linking maternal infections during pregnancy to a higher incidence of psychiatric disorders with a presumed neurodevelopmental origin in the offspring, including schizophrenia and autism. Although specific gestational windows may be associated with a differing vulnerability to infection-mediated disturbances in normal brain development, it still remains debatable whether and/or why certain gestation periods may confer maximal risk for neurodevelopmental disturbances following the prenatal exposure to infectious events. In this review, the authors integrate both epidemiological and experimental findings supporting the hypothesis that infection-associated immunological events in early fetal life may have a stronger neurodevelopmental impact compared to late pregnancy infections. This is because infections in early gestation may not only interfere with fundamental neurodevelopmental events such as cell proliferation and differentiation, but it may also predispose the developing nervous system to additional failures in subsequent cell migration, target selection, and synapse maturation, eventually leading to multiple brain and behavioral abnormalities in the adult offspring. The temporal dependency of the epidemiological link between maternal infections during pregnancy and a higher risk for brain disorders in the offspring may thus be explained by specific spatiotemporal events in the course of fetal brain development. NEUROSCIENTIST 13(3):241—256, 2007.
Collapse
Affiliation(s)
- Urs Meyer
- Laboratory of Behavioral Neurobiology, ETH Zurich, Switzerland
| | | | | |
Collapse
|
30
|
Crowley T, Cryan JF, Downer EJ, O'Leary OF. Inhibiting neuroinflammation: The role and therapeutic potential of GABA in neuro-immune interactions. Brain Behav Immun 2016; 54:260-277. [PMID: 26851553 DOI: 10.1016/j.bbi.2016.02.001] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 01/22/2016] [Accepted: 02/02/2016] [Indexed: 12/25/2022] Open
Abstract
The central nervous system, once thought to be a site of immunological privilege, has since been found to harbour immunocompetent cells and to communicate with the peripheral nervous system. In the central nervous system (CNS), glial cells display immunological responses to pathological and physiological stimuli through pro- and anti-inflammatory cytokine and chemokine signalling, antigen presentation and the clearing of cellular debris through phagocytosis. While this neuroinflammatory signalling can act to reduce neuronal damage and comprises a key facet of CNS homeostasis, persistent inflammation or auto-antigen-mediated immunoreactivity can induce a positive feedback cycle of neuroinflammation that ultimately results in necrosis of glia and neurons. Persistent neuroinflammation has been recognised as a major pathological component of virtually all neurodegenerative diseases and has also been a focus of research into the pathology underlying psychiatric disorders. Thus, pharmacological strategies to curb the pathological effects of persistent neuroinflammation are of interest for many disorders of the CNS. Accumulating evidence suggests that GABAergic activities are closely bound to immune processes and signals, and thus the GABAergic neurotransmitter system might represent an important therapeutic target in modulating neuroinflammation. Here, we review evidence that inflammation induces changes in the GABA neurotransmitter system in the CNS and that GABAergic signalling exerts a reciprocal influence over neuroinflammatory processes. Together, the data support the hypothesis that the GABA system is a potential therapeutic target in the modulation of central inflammation.
Collapse
Affiliation(s)
- Tadhg Crowley
- Department of Anatomy and Neuroscience, University College Cork, Ireland
| | - John F Cryan
- Department of Anatomy and Neuroscience, University College Cork, Ireland; APC Microbiome Institute, University College Cork, Ireland
| | - Eric J Downer
- School of Medicine, Discipline of Physiology, Trinity Biomedical Sciences Institute, Trinity College, Dublin 2, Ireland.
| | - Olivia F O'Leary
- Department of Anatomy and Neuroscience, University College Cork, Ireland; APC Microbiome Institute, University College Cork, Ireland.
| |
Collapse
|
31
|
Cassella SN, Hemmerle AM, Lundgren KH, Kyser TL, Ahlbrand R, Bronson SL, Richtand NM, Seroogy KB. Maternal immune activation alters glutamic acid decarboxylase-67 expression in the brains of adult rat offspring. Schizophr Res 2016; 171:195-9. [PMID: 26830319 PMCID: PMC4803111 DOI: 10.1016/j.schres.2016.01.041] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Revised: 01/15/2016] [Accepted: 01/19/2016] [Indexed: 12/21/2022]
Abstract
Activation of the maternal innate immune system, termed "maternal immune activation" (MIA), represents a common environmental risk factor for schizophrenia. Whereas evidence suggests dysregulation of GABA systems may underlie the pathophysiology of schizophrenia, a role for MIA in alteration of GABAergic systems is less clear. Here, pregnant rats received either the viral mimetic polyriboinosinic-polyribocytidilic acid or vehicle injection on gestational day 14. Glutamic acid decarboxylase-67 (GAD67) mRNA expression was examined in male offspring at postnatal day (P)14, P30 and P60. At P60, GAD67 mRNA was elevated in hippocampus and thalamus and decreased in prefrontal cortex of MIA offspring. MIA-induced alterations in GAD expression could contribute to the pathophysiology of schizophrenia.
Collapse
Affiliation(s)
- Sarah N Cassella
- Department of Neurology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Ann M Hemmerle
- Department of Neurology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Kerstin H Lundgren
- Department of Neurology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Tara L Kyser
- Department of Neurology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Rebecca Ahlbrand
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Stefanie L Bronson
- Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH 45267, USA; Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Neil M Richtand
- Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH 45267, USA; Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; San Diego Veterans Affairs Healthcare System, San Diego, CA 92161, USA; Department of Psychiatry, University of California, San Diego School of Medicine, La Jolla, CA 92093, USA
| | - Kim B Seroogy
- Department of Neurology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH 45267, USA.
| |
Collapse
|
32
|
Maternal immune activation produces neonatal excitability defects in offspring hippocampal neurons from pregnant rats treated with poly I:C. Sci Rep 2016; 6:19106. [PMID: 26742695 PMCID: PMC4705483 DOI: 10.1038/srep19106] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 12/02/2015] [Indexed: 12/26/2022] Open
Abstract
Maternal immune activation (MIA) resulting from prenatal exposure to infectious pathogens or inflammatory stimuli is increasingly recognized to play an important etiological role in neuropsychiatric disorders with neurodevelopmental features. MIA in pregnant rodents induced by injection of the synthetic double-stranded RNA, Poly I:C, a mimic of viral infection, leads to a wide spectrum of behavioral abnormalities as well as structural and functional defects in the brain. Previous MIA studies using poly I:C prenatal treatment suggested that neurophysiological alterations occur in the hippocampus. However, these investigations used only juvenile or adult animals. We postulated that MIA-induced alterations could occur earlier at neonatal/early postnatal stages. Here we examined the neurophysiological properties of cultured pyramidal-like hippocampal neurons prepared from neonatal (P0-P2) offspring of pregnant rats injected with poly I:C. Offspring neurons from poly I:C-treated mothers exhibited significantly lower intrinsic excitability and stronger spike frequency adaptation, compared to saline. A similar lower intrinsic excitability was observed in CA1 pyramidal neurons from hippocampal slices of two weeks-old poly I:C offspring. Cultured hippocampal neurons also displayed lower frequency of spontaneous firing, higher charge transfer of IPSCs and larger amplitude of miniature IPSCs. Thus, maternal immune activation leads to strikingly early neurophysiological abnormalities in hippocampal neurons.
Collapse
|
33
|
Turkheimer FE, Leech R, Expert P, Lord LD, Vernon AC. The brain's code and its canonical computational motifs. From sensory cortex to the default mode network: A multi-scale model of brain function in health and disease. Neurosci Biobehav Rev 2015; 55:211-22. [PMID: 25956253 DOI: 10.1016/j.neubiorev.2015.04.014] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 04/01/2015] [Accepted: 04/25/2015] [Indexed: 12/21/2022]
Affiliation(s)
| | - Robert Leech
- Division of Brain Sciences, Imperial College London, London, UK
| | - Paul Expert
- Institute of Psychiatry, King's College London, London, UK
| | | | | |
Collapse
|
34
|
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
|
35
|
Sahila MM, Babitha PP, Bandaru S, Nayarisseri A, Doss VA. Molecular docking based screening of GABA (A) receptor inhibitors from plant derivatives. Bioinformation 2015; 11:280-9. [PMID: 26229288 PMCID: PMC4512002 DOI: 10.6026/97320630011280] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2015] [Revised: 06/25/2015] [Accepted: 06/04/2015] [Indexed: 12/05/2022] Open
Abstract
UNLABELLED The present antipsychotic drugs have known to show serious concerns like extra pyramidal side effects therefore, pursuit for novel antipsychotic GABAnergic drugs has lately focused on the folkloric medicine from plant derivatives as better treatment option of schizophrenia. The present study centers to identify potential inhibitors of plant origin for GABA receptor through in silico approaches. Three compound datasets were undertaken in the study. The first set consisted of seven compounds which included Magnolol, Honokiol and other plant derivatives. The second set consisted of 16 derivatives of N-diarylalkenyl-piperidinecarboxylic acid synthesized by Zheng et al., 2006. The third dataset had thirty two compounds which were Magnolol and Honokiol analogues synthesized by Fuchs et al., 2014. All the compounds were docked at the allosteric site of the GABA (A) receptor. The compounds were further tested for ADMET and biological activity. We observed Honokiol and its derivatives demonstrated superior druglike properties than any compound undertaken in the study. Further, compound 61 [2-(4-methoxyphenyl)-4-propylphenol] of dataset three - a synthetic derivative of honokiol had better profile than its parent compound. In a possible attempt to identify compound with even better efficacious compound than 61, virtual screening was performed, 135 compounds akin to compound 61 were retrieved. Interestingly none of the 135 compounds showed better druggable properties than compound 61. Our in silico pharmacological profiling of compounds is in coherence and is complemented by the findings of Fuchs et al, which also revealed compound 61 to be the good potentiator of GABA receptor. ABBREVIATIONS GABA (A) R - Gamma Amino Butyric Acid Receptor, subtype A, GPCR - G Protein Coupled Receptor, OPLS - Optimized Potentials for Liquid Simulations, PDB - Protein Data Bank, PLP - Piece wise Linear Potential, T.E.S.T - Toxicity Estimation Software Tool, TCM - Traditional Chinese Medicine.
Collapse
Affiliation(s)
- Mohammed Marunnan Sahila
- Department of Bioinformatics, School of Life Sciences, Karpagam University, Coimbatore 641021, India
- Department of Bioinformatics, SIAS-Centre for Scientific, Research, Safi Institute of Advanced Study(SIAS) , Rasiya Nagar, Vazhayoor East, Malappuram-673633, Kerala, India
| | | | - Srinivas Bandaru
- Bioinformatics Research Laboratory, Eminent Biosciences, Vijaynagar, Indore - 452010, India
| | - Anuraj Nayarisseri
- Bioinformatics Research Laboratory, Eminent Biosciences, Vijaynagar, Indore - 452010, India
| | - Victor Arokia Doss
- Department of Biochemistry, PSG College of Arts and Science, Coimbatore - 641 014, India
| |
Collapse
|
36
|
Reisinger S, Khan D, Kong E, Berger A, Pollak A, Pollak DD. The poly(I:C)-induced maternal immune activation model in preclinical neuropsychiatric drug discovery. Pharmacol Ther 2015; 149:213-26. [PMID: 25562580 DOI: 10.1016/j.pharmthera.2015.01.001] [Citation(s) in RCA: 169] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 12/30/2014] [Indexed: 12/28/2022]
Abstract
Increasing epidemiological and experimental evidence implicates gestational infections as one important factor involved in the pathogenesis of several neuropsychiatric disorders. Corresponding preclinical model systems based upon maternal immune activation (MIA) by treatment of the pregnant female have been developed. These MIA animal model systems have been successfully used in basic and translational research approaches, contributing to the investigation of the underlying pathophysiological mechanisms at the molecular, cellular and behavioral levels. The present article focuses on the application of a specific MIA rodent paradigm, based upon treatment of the gestating dam with the viral mimic polyinosinic-polycytidilic acid (Poly(I:C)), a synthetic analog of double-stranded RNA (dsRNA) which activates the Toll-like receptor 3 (TLR3) pathway. Important advantages and constraints of this animal model will be discussed, specifically in light of gestational infection as one vulnerability factor contributing to the complex etiology of mood and psychotic disorders, which are likely the result of intricate multi-level gene×environment interactions. Improving our currently incomplete understanding of the molecular pathomechanistic principles underlying these disorders is a prerequisite for the development of alternative therapeutic approaches which are critically needed in light of the important drawbacks and limitations of currently available pharmacological treatment options regarding efficacy and side effects. The particular relevance of the Poly(I:C) MIA model for the discovery of novel drug targets for symptomatic and preventive therapeutic strategies in mood and psychotic disorders is highlighted in this review article.
Collapse
Affiliation(s)
- Sonali Reisinger
- Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Austria
| | - Deeba Khan
- Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Austria
| | - Eryan Kong
- Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Austria
| | - Angelika Berger
- Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Austria
| | - Arnold Pollak
- Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Austria
| | - Daniela D Pollak
- Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Austria.
| |
Collapse
|
37
|
Wischhof L, Irrsack E, Osorio C, Koch M. Prenatal LPS-exposure--a neurodevelopmental rat model of schizophrenia--differentially affects cognitive functions, myelination and parvalbumin expression in male and female offspring. Prog Neuropsychopharmacol Biol Psychiatry 2015; 57:17-30. [PMID: 25455585 DOI: 10.1016/j.pnpbp.2014.10.004] [Citation(s) in RCA: 119] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 10/08/2014] [Accepted: 10/10/2014] [Indexed: 01/15/2023]
Abstract
Maternal infection during pregnancy increases the risk for the offspring to develop schizophrenia. Gender differences can be seen in various features of the illness and sex steroid hormones (e.g. estrogen) have strongly been implicated in the disease pathology. In the present study, we evaluated sex differences in the effects of prenatal exposure to a bacterial endotoxin (lipopolysaccharide, LPS) in rats. Pregnant dams received LPS-injections (100 μg/kg) at gestational day 15 and 16. The offspring was then tested for prepulse inhibition (PPI), locomotor activity, anxiety-like behavior and object recognition memory at various developmental time points. At postnatal day (PD) 33 and 60, prenatally LPS-exposed rats showed locomotor hyperactivity which was similar in male and female offspring. Moreover, prenatal LPS-treatment caused PPI deficits in pubertal (PD45) and adult (PD90) males while PPI impairments were found only at PD45 in prenatally LPS-treated females. Following prenatal LPS-administration, recognition memory for objects was impaired in both sexes with males being more severely affected. Additionally, we assessed prenatal infection-induced alterations of parvalbumin (Parv) expression and myelin fiber density. Male offspring born to LPS-challenged mothers showed decreased myelination in cortical and limbic brain regions as well as reduced numbers of Parv-expressing cells in the medial prefrontal cortex (mPFC), hippocampus and entorhinal cortex. In contrast, LPS-exposed female rats showed only a modest decrease in myelination and Parv immunoreactivity. Collectively, our data indicate that some of the prenatal immune activation effects are sex dependent and further strengthen the importance of taking into account gender differences in animal models of schizophrenia.
Collapse
Affiliation(s)
- Lena Wischhof
- Brain Research Institute, Department of Neuropharmacology, University of Bremen, Hochschulring 18, 28359 Bremen, Germany.
| | - Ellen Irrsack
- Brain Research Institute, Department of Neuropharmacology, University of Bremen, Hochschulring 18, 28359 Bremen, Germany
| | - Carmen Osorio
- Brain Research Institute, Department of Neuropharmacology, University of Bremen, Hochschulring 18, 28359 Bremen, Germany
| | - Michael Koch
- Brain Research Institute, Department of Neuropharmacology, University of Bremen, Hochschulring 18, 28359 Bremen, Germany
| |
Collapse
|
38
|
Richetto J, Labouesse MA, Poe MM, Cook JM, Grace AA, Riva MA, Meyer U. Behavioral effects of the benzodiazepine-positive allosteric modulator SH-053-2'F-S-CH₃ in an immune-mediated neurodevelopmental disruption model. Int J Neuropsychopharmacol 2015; 18:pyu055. [PMID: 25636893 PMCID: PMC4360215 DOI: 10.1093/ijnp/pyu055] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Impaired γ-aminobutyric acid (GABA) signaling may contribute to the emergence of cognitive deficits and subcortical dopaminergic hyperactivity in patients with schizophrenia and related psychotic disorders. Against this background, it has been proposed that pharmacological interventions targeting GABAergic dysfunctions may prove useful in correcting such cognitive impairments and dopaminergic imbalances. METHODS Here, we explored possible beneficial effects of the benzodiazepine-positive allosteric modulator SH-053-2'F-S-CH₃, with partial selectivity at the α2, α3, and α5 subunits of the GABAA receptor in an immune-mediated neurodevelopmental disruption model. The model is based on prenatal administration of the viral mimetic polyriboinosinic-polyribocytidilic acid [poly(I:C)] in mice, which is known to capture various GABAergic, dopamine-related, and cognitive abnormalities implicated in schizophrenia and related disorders. RESULTS Real-time polymerase chain reaction analyses confirmed the expected alterations in GABAA receptor α subunit gene expression in the medial prefrontal cortices and ventral hippocampi of adult poly(I:C) offspring relative to control offspring. Systemic administration of SH-053-2'F-S-CH₃ failed to normalize the poly(I:C)-induced deficits in working memory and social interaction, but instead impaired performance in these cognitive and behavioral domains both in control and poly(I:C) offspring. In contrast, SH-053-2'F-S-CH₃ was highly effective in mitigating the poly(I:C)-induced amphetamine hypersensitivity phenotype without causing side effects in control offspring. CONCLUSIONS Our preclinical data suggest that benzodiazepine-like positive allosteric modulators with activity at the α2, α3, and α5 subunits of the GABAA receptor may be particularly useful in correcting pathological overactivity of the dopaminergic system, but they may be ineffective in targeting multiple pathological domains that involve the co-existence of psychotic, social, and cognitive dysfunctions.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Urs Meyer
- Center of Neuropharmacology, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy (Drs Richetto and Riva); Physiology and Behavior Laboratory, ETH Zurich, Schorenstrasse 16, 8603 Schwerzenbach, Switzerland (Drs Labouesse and Meyer); Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, Milwaukee, WI (Drs Poe and Cook); Departments of Neuroscience, Psychiatry and Psychology, University of Pittsburgh, Pittsburgh, PA (Dr Grace); Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Milan, Italy (Dr Riva).
| |
Collapse
|
39
|
Schmidt MJ, Mirnics K. Neurodevelopment, GABA system dysfunction, and schizophrenia. Neuropsychopharmacology 2015; 40:190-206. [PMID: 24759129 PMCID: PMC4262918 DOI: 10.1038/npp.2014.95] [Citation(s) in RCA: 147] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 04/03/2014] [Accepted: 04/11/2014] [Indexed: 02/07/2023]
Abstract
The origins of schizophrenia have eluded clinicians and researchers since Kraepelin and Bleuler began documenting their findings. However, large clinical research efforts in recent decades have identified numerous genetic and environmental risk factors for schizophrenia. The combined data strongly support the neurodevelopmental hypothesis of schizophrenia and underscore the importance of the common converging effects of diverse insults. In this review, we discuss the evidence that genetic and environmental risk factors that predispose to schizophrenia disrupt the development and normal functioning of the GABAergic system.
Collapse
Affiliation(s)
- Martin J Schmidt
- Department of Psychiatry, Vanderbilt University, Nashville, TN, USA
| | - Karoly Mirnics
- Department of Psychiatry, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, TN, USA
- Department of Psychiatry, University of Szeged, Szeged, Hungary
| |
Collapse
|
40
|
Mattei D, Schweibold R, Wolf SA. Brain in flames - animal models of psychosis: utility and limitations. Neuropsychiatr Dis Treat 2015; 11:1313-29. [PMID: 26064050 PMCID: PMC4455860 DOI: 10.2147/ndt.s65564] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The neurodevelopmental hypothesis of schizophrenia posits that schizophrenia is a psychopathological condition resulting from aberrations in neurodevelopmental processes caused by a combination of environmental and genetic factors which proceed long before the onset of clinical symptoms. Many studies discuss an immunological component in the onset and progression of schizophrenia. We here review studies utilizing animal models of schizophrenia with manipulations of genetic, pharmacologic, and immunological origin. We focus on the immunological component to bridge the studies in terms of evaluation and treatment options of negative, positive, and cognitive symptoms. Throughout the review we link certain aspects of each model to the situation in human schizophrenic patients. In conclusion we suggest a combination of existing models to better represent the human situation. Moreover, we emphasize that animal models represent defined single or multiple symptoms or hallmarks of a given disease.
Collapse
Affiliation(s)
- Daniele Mattei
- Department of Cellular Neuroscience, Max-Delbrueck-Center for Molecular Medicine, Berlin, Germany
| | - Regina Schweibold
- Department of Cellular Neuroscience, Max-Delbrueck-Center for Molecular Medicine, Berlin, Germany ; Department of Neurosurgery, Helios Clinics, Berlin, Germany
| | - Susanne A Wolf
- Department of Cellular Neuroscience, Max-Delbrueck-Center for Molecular Medicine, Berlin, Germany
| |
Collapse
|
41
|
Greenberg Z, Ramshaw H, Schwarz Q. Time Windows of Interneuron Development: Implications to Our Understanding of the Aetiology and Treatment of Schizophrenia. AIMS Neurosci 2015. [DOI: 10.3934/neuroscience.2015.4.294] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
42
|
Modinos G, Allen P, Grace AA, McGuire P. Translating the MAM model of psychosis to humans. Trends Neurosci 2014; 38:129-38. [PMID: 25554679 DOI: 10.1016/j.tins.2014.12.005] [Citation(s) in RCA: 121] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Revised: 11/12/2014] [Accepted: 12/02/2014] [Indexed: 12/13/2022]
Abstract
Elevated dopamine function and alterations in medial temporal lobe (MTL) structure and function are two of the most robust findings in schizophrenia, but how interactions between these abnormalities underlie the onset of psychosis is unclear. The methylazoxymethanol acetate (MAM) rodent model proposes that psychosis develops as a result of a perturbation of MTL function, leading to elevated striatal dopamine dysfunction. Here, we review several recent neuroimaging studies that examine components of the putative model in humans with an ultra high risk (UHR) of the psychosis. While data from these studies are broadly consistent with the MAM model, caution is required when comparing data across animal and human studies.
Collapse
Affiliation(s)
- Gemma Modinos
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
| | - Paul Allen
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Anthony A Grace
- Departments of Neuroscience, Psychiatry, and Psychology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Philip McGuire
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| |
Collapse
|
43
|
McDougle CJ, Landino SM, Vahabzadeh A, O'Rourke J, Zurcher NR, Finger BC, Palumbo ML, Helt J, Mullett JE, Hooker JM, Carlezon WA. Toward an immune-mediated subtype of autism spectrum disorder. Brain Res 2014; 1617:72-92. [PMID: 25445995 DOI: 10.1016/j.brainres.2014.09.048] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Revised: 09/15/2014] [Accepted: 09/20/2014] [Indexed: 11/25/2022]
Abstract
A role for immunological involvement in autism spectrum disorder (ASD) has long been hypothesized. This review includes four sections describing (1) evidence for a relationship between familial autoimmune disorders and ASD; (2) results from post-mortem and neuroimaging studies that investigated aspects of neuroinflammation in ASD; (3) findings from animal model work in ASD involving inflammatory processes; and (4) outcomes from trials of anti-inflammatory/immune-modulating drugs in ASD that have appeared in the literature. Following each section, ideas are provided for future research, suggesting paths forward in the continuing effort to define the role of immune factors and inflammation in the pathophysiology of a subtype of ASD. This article is part of a Special Issue entitled SI: Neuroimmunology in Health And Disease.
Collapse
Affiliation(s)
- Christopher J McDougle
- Lurie Center for Autism, Lexington, MA, United States; Massachusetts General Hospital, Boston, MA, United States; Harvard Medical School, Boston, MA, United States.
| | - Samantha M Landino
- Behavioral Genetics Laboratory, Belmont, MA, United States; McLean Hospital, Belmont, MA, United States
| | - Arshya Vahabzadeh
- Massachusetts General Hospital, Boston, MA, United States; McLean Hospital, Belmont, MA, United States; Harvard Medical School, Boston, MA, United States
| | - Julia O'Rourke
- Lurie Center for Autism, Lexington, MA, United States; Massachusetts General Hospital, Boston, MA, United States; Harvard Medical School, Boston, MA, United States
| | - Nicole R Zurcher
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, United States; Massachusetts General Hospital, Boston, MA, United States; Harvard Medical School, Boston, MA, United States
| | - Beate C Finger
- Behavioral Genetics Laboratory, Belmont, MA, United States; McLean Hospital, Belmont, MA, United States; Harvard Medical School, Boston, MA, United States
| | - Michelle L Palumbo
- Lurie Center for Autism, Lexington, MA, United States; Massachusetts General Hospital, Boston, MA, United States; Harvard Medical School, Boston, MA, United States
| | - Jessica Helt
- Lurie Center for Autism, Lexington, MA, United States; Massachusetts General Hospital, Boston, MA, United States
| | - Jennifer E Mullett
- Lurie Center for Autism, Lexington, MA, United States; Massachusetts General Hospital, Boston, MA, United States
| | - Jacob M Hooker
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA, United States; Massachusetts General Hospital, Boston, MA, United States; Harvard Medical School, Boston, MA, United States
| | - William A Carlezon
- Behavioral Genetics Laboratory, Belmont, MA, United States; McLean Hospital, Belmont, MA, United States; Harvard Medical School, Boston, MA, United States
| |
Collapse
|
44
|
Solati J, Kleehaupt E, Kratz O, Moll GH, Golub Y. Inverse effects of lipopolysaccharides on anxiety in pregnant mice and their offspring. Physiol Behav 2014; 139:369-74. [PMID: 25447752 DOI: 10.1016/j.physbeh.2014.10.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 10/14/2014] [Indexed: 11/29/2022]
Abstract
This study aimed to evaluate the effects of the bacterial lipopolysaccharide (LPS) exposure during early pregnancy on anxiety-related behaviour of both pregnant female mice and their male offspring. Pregnant NMRI mice were treated with subcutaneous injections of LPS (30, 60, 120, 240 and 480 μg/kg) on the tenth gestational day of pregnancy. Pro-inflammatory cytokines, such as TNF-α, IL-1β, IL-6 and corticosterone levels, were measured in maternal serum 1.5h following the LPS injections. Baseline anxiety levels of pregnant mice (1.5h after LPS administration) and their male offspring (at postnatal days 60-70) were investigated with the elevated plus maze (EPM) test. In addition, anxiety levels in the offspring were measured after 2h restraint stress or TNF-α (10 μg/kg) administration. Our results demonstrate that LPS administration induces anxiety-like behaviour and a significant increase in cytokines and corticosterone levels in maternal serum. However, in male offspring, prenatal LPS administration has no significant effects on serum cytokines and corticosterone secretion with an exception of the lowest LPS dose that slightly reduced corticosterone levels. Interestingly, prenatal LPS treatment seemed to decrease the baseline anxiety levels, while pretreatment with restraint stress or TNF-α abolished this anxiolytic effects. In summary, our results suggest that prenatal exposure to LPS during early pregnancy may result in reduced baseline anxiety in adult male offspring.
Collapse
Affiliation(s)
- Jalal Solati
- Department of Child and Adolescent Mental Health, University of Erlangen-Nürnberg, Erlangen, Germany; Department of Biology, College of Science, Karaj branch, Islamic Azad University, Alborz, Iran
| | - Eva Kleehaupt
- Department of Child and Adolescent Mental Health, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Oliver Kratz
- Department of Child and Adolescent Mental Health, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Gunther H Moll
- Department of Child and Adolescent Mental Health, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Yulia Golub
- Department of Child and Adolescent Mental Health, University of Erlangen-Nürnberg, Erlangen, Germany.
| |
Collapse
|
45
|
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
|
46
|
Maternal immune activation and abnormal brain development across CNS disorders. Nat Rev Neurol 2014; 10:643-60. [PMID: 25311587 DOI: 10.1038/nrneurol.2014.187] [Citation(s) in RCA: 607] [Impact Index Per Article: 60.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Epidemiological studies have shown a clear association between maternal infection and schizophrenia or autism in the progeny. Animal models have revealed maternal immune activation (mIA) to be a profound risk factor for neurochemical and behavioural abnormalities in the offspring. Microglial priming has been proposed as a major consequence of mIA, and represents a critical link in a causal chain that leads to the wide spectrum of neuronal dysfunctions and behavioural phenotypes observed in the juvenile, adult or aged offspring. Such diversity of phenotypic outcomes in the mIA model are mirrored by recent clinical evidence suggesting that infectious exposure during pregnancy is also associated with epilepsy and, to a lesser extent, cerebral palsy in children. Preclinical research also suggests that mIA might precipitate the development of Alzheimer and Parkinson diseases. Here, we summarize and critically review the emerging evidence that mIA is a shared environmental risk factor across CNS disorders that varies as a function of interactions between genetic and additional environmental factors. We also review ongoing clinical trials targeting immune pathways affected by mIA that may play a part in disease manifestation. In addition, future directions and outstanding questions are discussed, including potential symptomatic, disease-modifying and preventive treatment strategies.
Collapse
|
47
|
Mattei D, Djodari-Irani A, Hadar R, Pelz A, de Cossío LF, Goetz T, Matyash M, Kettenmann H, Winter C, Wolf SA. Minocycline rescues decrease in neurogenesis, increase in microglia cytokines and deficits in sensorimotor gating in an animal model of schizophrenia. Brain Behav Immun 2014; 38:175-84. [PMID: 24509090 DOI: 10.1016/j.bbi.2014.01.019] [Citation(s) in RCA: 139] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Revised: 01/06/2014] [Accepted: 01/27/2014] [Indexed: 01/27/2023] Open
Abstract
Adult neurogenesis in the hippocampus is impaired in schizophrenic patients and in an animal model of schizophrenia. Amongst a plethora of regulators, the immune system has been shown repeatedly to strongly modulate neurogenesis under physiological and pathological conditions. It is well accepted, that schizophrenic patients have an aberrant peripheral immune status, which is also reflected in the animal model. The microglia as the intrinsic immune competent cells of the brain have recently come into focus as possible therapeutic targets in schizophrenia. We here used a maternal immune stimulation rodent model of schizophrenia in which polyinosinic-polycytidilic acid (Poly I:C) was injected into pregnant rats to mimic an anti-viral immune response. We identified microglia IL-1β and TNF-α increase constituting the factors correlating best with decreases in net-neurogenesis and impairment in pre-pulse inhibition of a startle response in the Poly I:C model. Treatment with the antibiotic minocycline (3mg/kg/day) normalized microglial cytokine production in the hippocampus and rescued neurogenesis and behavior. We could also show that enhanced microglial TNF-α and IL-1β production in the hippocampus was accompanied by a decrease in the pro-proliferative TNFR2 receptor expression on neuronal progenitor cells, which could be attenuated by minocycline. These findings strongly support the idea to use anti-inflammatory drugs to target microglia activation as an adjunctive therapy in schizophrenic patients.
Collapse
Affiliation(s)
- Daniele Mattei
- Max-Delbrück-Center of Molecular Medicine, Cellular Neuroscience, 13125 Berlin, Germany
| | - Anaïs Djodari-Irani
- Max-Delbrück-Center of Molecular Medicine, Cellular Neuroscience, 13125 Berlin, Germany; Department of Psychiatry and Psychotherapy, Charité - Universitätsmedizin Berlin, Charité Campus Mitte, 10117 Berlin, Germany
| | - Ravit Hadar
- University Hospital, Clinic for Psychiatry and Psychotherapy, Experimental Psychiatry, 01307 Dresden, Germany
| | - Andreas Pelz
- Max-Delbrück-Center of Molecular Medicine, Cellular Neuroscience, 13125 Berlin, Germany
| | | | - Thomas Goetz
- University Hospital, Clinic for Psychiatry and Psychotherapy, Experimental Psychiatry, 01307 Dresden, Germany
| | - Marina Matyash
- Max-Delbrück-Center of Molecular Medicine, Cellular Neuroscience, 13125 Berlin, Germany
| | - Helmut Kettenmann
- Max-Delbrück-Center of Molecular Medicine, Cellular Neuroscience, 13125 Berlin, Germany
| | - Christine Winter
- University Hospital, Clinic for Psychiatry and Psychotherapy, Experimental Psychiatry, 01307 Dresden, Germany
| | - Susanne A Wolf
- Max-Delbrück-Center of Molecular Medicine, Cellular Neuroscience, 13125 Berlin, Germany.
| |
Collapse
|
48
|
Bitanihirwe BKY, Woo TUW. Perineuronal nets and schizophrenia: the importance of neuronal coatings. Neurosci Biobehav Rev 2014; 45:85-99. [PMID: 24709070 DOI: 10.1016/j.neubiorev.2014.03.018] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Revised: 02/19/2014] [Accepted: 03/25/2014] [Indexed: 12/17/2022]
Abstract
Schizophrenia is a complex brain disorder associated with deficits in synaptic connectivity. The insidious onset of this illness during late adolescence and early adulthood has been reported to be dependent on several key processes of brain development including synaptic refinement, myelination and the physiological maturation of inhibitory neural networks. Interestingly, these events coincide with the appearance of perineuronal nets (PNNs), reticular structures composed of components of the extracellular matrix that coat a variety of cells in the mammalian brain. Until recently, the functions of the PNN had remained enigmatic, but are now considered to be important in development of the central nervous system, neuronal protection and synaptic plasticity, all elements which have been associated with schizophrenia. Here, we review the emerging evidence linking PNNs to schizophrenia. Future studies aimed at further elucidating the functions of PNNs will provide new insights into the pathophysiology of schizophrenia leading to the identification of novel therapeutic targets with the potential to restore normal synaptic integrity in the brain of patients afflicted by this illness.
Collapse
Affiliation(s)
| | - Tsung-Ung W Woo
- Program in Cellular Neuropathology, McLean Hospital, Belmont, MA, USA; Department of Psychiatry, Harvard Medical School, Boston, MA, USA; Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, MA, USA.
| |
Collapse
|
49
|
Richetto J, Calabrese F, Riva MA, Meyer U. Prenatal immune activation induces maturation-dependent alterations in the prefrontal GABAergic transcriptome. Schizophr Bull 2014; 40:351-61. [PMID: 23328159 PMCID: PMC3932076 DOI: 10.1093/schbul/sbs195] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Neuronal dysfunctions in the cortical GABAergic system have been widely documented in neuropsychiatric disorders with prenatal infectious etiologies, including schizophrenia. At least some of these abnormalities may stem from transcriptional impairments in the GABAergic transcriptome. However, the extent to which prenatal exposure to immune challenge can induce long-term alterations in GABAergic gene transcription remains largely elusive. Here, we use an established mouse model of prenatal immune activation induced by maternal gestational administration of the viral mimetic poly(I:C) (= polyriboinosinic-polyribocytidilic acid) to demonstrate that prenatal immune activation causes maturation-dependent alterations in prefrontal GABAergic gene expression. The spectrum of abnormalities included altered mRNA expression levels of enzymes regulating γ-aminobutyric acid (GABA) biosynthesis (glutamic acid decarboxylase 65-kDa [GAD65] and GAD67), vesicular GABA transporter (VGAT), alpha-subunits of the GABA(A) receptor (α2, α3, α4, and α5), and the chloride transporters sodium-potassium-chloride cotransporter 1 and potassium-chloride cotransporter 2. Additional western blot analyses confirmed the deficits in prefrontal GAD65/GAD67 and VGAT expression at the protein level. Intriguingly, the prefrontal GABAergic transcriptome was found to be more strongly affected in adult compared with peripubertal offspring born to immune-challenged mothers, and these age-dependent changes in GABAergic gene expression were paralleled by an adult onset of working memory deficiency. Collectively, our data emphasize a critical impact of prenatal immune-related insults on long-term GABAergic changes relevant to neuropsychiatric disorders with prenatal infectious etiologies, especially for those with delayed onset in early adulthood.
Collapse
Affiliation(s)
- Juliet Richetto
- *To whom correspondence should be addressed; tel: +41 44 655 7403, fax: +41 44 655 7203, e-mail:
| | - Francesca Calabrese
- 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;,Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Milan, Italy;,These authors contributed equally to the present study
| | - Urs Meyer
- Physiology and Behavior Laboratory, Swiss Federal Institute of Technology (ETH) Zurich, Schorenstrasse 16, 8603 Schwerzenbach, Switzerland,These authors contributed equally to the present study.,*To whom correspondence should be addressed; tel: +41 44 655 7403, fax: +41 44 655 7203, e-mail:
| |
Collapse
|
50
|
Na KS, Jung HY, Kim YK. The role of pro-inflammatory cytokines in the neuroinflammation and neurogenesis of schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry 2014; 48:277-86. [PMID: 23123365 DOI: 10.1016/j.pnpbp.2012.10.022] [Citation(s) in RCA: 294] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Revised: 10/11/2012] [Accepted: 10/26/2012] [Indexed: 12/22/2022]
Abstract
Schizophrenia is a serious mental illness with chronic symptoms and significant impairment in psychosocial functioning. Although novel antipsychotics have been developed, the negative and cognitive symptoms of schizophrenia are still unresponsive to pharmacotherapy. The high level of social impairment and a chronic deteriorating course suggest that schizophrenia likely has neurodegenerative characteristics. Inflammatory markers such as pro-inflammatory cytokines are well-known etiological factors for psychiatric disorders, including schizophrenia. Inflammation in the central nervous system is closely related to neurodegeneration. In addition to pro-inflammatory cytokines, microglia also play an important role in the inflammatory process in the CNS. Uncontrolled activity of pro-inflammatory cytokines and microglia can induce schizophrenia in tandem with genetic vulnerability and glutamatergic neurotransmitters. Several studies have investigated the possible effects of antipsychotics on inflammation and neurogenesis. Additionally, anti-inflammatory adjuvant therapy has been under investigation as a treatment option for schizophrenia. Further studies should consider the confounding effects of systemic factors such as metabolic syndrome and smoking. In addition, the unique mechanisms by which pro-inflammatory cytokines are involved in the etiopathology of schizophrenia should be investigated. In this article, we aimed to review (1) major findings regarding neuroinflammation and pro-inflammatory cytokine alterations in schizophrenia, (2) interactions between neuroinflammation and neurogenesis as possible neural substrates for schizophrenia, and (3) novel pharmacological approaches.
Collapse
Affiliation(s)
- Kyoung-Sae Na
- Department of Psychiatry, Soonchunhyang University Bucheon Hospital, Bucheon, Republic of Korea
| | | | | |
Collapse
|