1
|
Munarriz-Cuezva E, Meana JJ. Poly (I:C)-induced maternal immune activation generates impairment of reversal learning performance in offspring. J Neurochem 2025; 169:e16212. [PMID: 39183542 DOI: 10.1111/jnc.16212] [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: 01/29/2024] [Revised: 08/07/2024] [Accepted: 08/09/2024] [Indexed: 08/27/2024]
Abstract
Maternal immune activation (MIA) induces a variety of behavioral and brain abnormalities in offspring of rodent models, compatible with neurodevelopmental disorders, such as schizophrenia or autism. However, it remains controversial whether MIA impairs reversal learning, a basic expression of cognitive flexibility that seems to be altered in schizophrenia. In the present study, MIA was induced by administration of a single dose of polyriboinosinic-polyribocytidylic acid (Poly (I:C) (5 mg/kg i.p.)) or saline to mouse pregnant dams in gestational day (GD) 9.5. Immune activation was monitored through changes in weight and temperature. The offspring were evaluated when they reached adulthood (8 weeks) using a touchscreen-based system to investigate the effects of Poly (I:C) on discrimination and reversal learning performance. After an initial pre-training, mice were trained to discriminate between two different stimuli, of which only one was rewarded (acquisition phase). When the correct response reached above 80% values for two consecutive days, the images were reversed (reversal phase) to assess the adaptation capacity to a changing environment. Maternal Poly (I:C) treatment did not interfere with the learning process but induced deficits in reversal learning compared to control saline animals. Thus, the accuracy in the reversal phase was lower, and Poly (I:C) animals required more sessions to complete it, suggesting impairments in cognitive flexibility. This study advances the knowledge of how MIA affects behavior, especially cognitive domains that are impaired in schizophrenia. The findings support the validity of the Poly (I:C)-based MIA model as a tool to develop pharmacological treatments targeting cognitive deficits associated with neurodevelopmental disorders.
Collapse
Affiliation(s)
- Eva Munarriz-Cuezva
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country, Leioa, Bizkaia, Spain
- Centro de Investigación Biomédica en Red de Salud Mental, Leioa, Bizkaia, Spain
| | - Jose Javier Meana
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country, Leioa, Bizkaia, Spain
- Centro de Investigación Biomédica en Red de Salud Mental, Leioa, Bizkaia, Spain
- Biobizkaia Health Research Institute, Barakaldo, Bizkaia, Spain
| |
Collapse
|
2
|
Drehmer I, Santos-Terra J, Gottfried C, Deckmann I. mTOR signaling pathway as a pathophysiologic mechanism in preclinical models of autism spectrum disorder. Neuroscience 2024; 563:33-42. [PMID: 39481829 DOI: 10.1016/j.neuroscience.2024.10.050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 10/23/2024] [Accepted: 10/28/2024] [Indexed: 11/03/2024]
Abstract
Autism spectrum disorder (ASD) is a highly prevalent multifactorial disorder characterized by social deficits and stereotypies. Despite extensive research efforts, the etiology of ASD remains poorly understood. However, studies using preclinical models have identified the mechanistic target of rapamycin kinase (mTOR) signaling pathway as a key player in ASD-related features. This review examines genetic and environmental models of ASD, focusing on their association with the mTOR pathway. We organize findings on alterations within this pathway, providing insights about the potential mechanisms involved in the onset and maintenance of ASD symptoms. Our analysis highlights the central role of mTOR hyperactivation in disrupting autophagic processes, neural organization, and neurotransmitter pathways, which collectively contribute to ASD phenotypes. The review also discusses the therapeutic potential of mTOR pathway inhibitors, such as rapamycin, in mitigating ASD characteristics. These insights underscore the importance of the mTOR pathway as a target for future research and therapeutic intervention in ASD. This review innovates by bringing the convergence of disrupted mTOR signaling in preclinical models and clinical data associated with ASD.
Collapse
Affiliation(s)
- Isabela Drehmer
- Translational Research Group on Autism Spectrum Disorder - GETTEA, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; National Institute of Science and Technology in Neuroimmunomodulation - INCT-NIM, Brazil; Autism Wellbeing and Research Development - AWARD - Initiative BR-UK-CA, Brazil; Psychiatry Molecular Laboratory, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil; Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Júlio Santos-Terra
- Translational Research Group on Autism Spectrum Disorder - GETTEA, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; National Institute of Science and Technology in Neuroimmunomodulation - INCT-NIM, Brazil; Autism Wellbeing and Research Development - AWARD - Initiative BR-UK-CA, Brazil; Psychiatry Molecular Laboratory, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Carmem Gottfried
- Translational Research Group on Autism Spectrum Disorder - GETTEA, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; National Institute of Science and Technology in Neuroimmunomodulation - INCT-NIM, Brazil; Autism Wellbeing and Research Development - AWARD - Initiative BR-UK-CA, Brazil; Psychiatry Molecular Laboratory, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Iohanna Deckmann
- Translational Research Group on Autism Spectrum Disorder - GETTEA, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; National Institute of Science and Technology in Neuroimmunomodulation - INCT-NIM, Brazil; Autism Wellbeing and Research Development - AWARD - Initiative BR-UK-CA, Brazil; Psychiatry Molecular Laboratory, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.
| |
Collapse
|
3
|
Chen T, Meng H, Fang N, Shi P, Chen M, Liu Q, Lv L, Li W. Age-related changes in behavior profile in male offspring of rats treated with poly I:C-induced maternal immune activation in early gestation. Animal Model Exp Med 2024; 7:914-925. [PMID: 38741390 DOI: 10.1002/ame2.12417] [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: 06/27/2023] [Accepted: 03/21/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND Autism and schizophrenia are environmental risk factors associated with prenatal viral infection during pregnancy. It is still unclear whether behavior phenotypes change at different developmental stages in offspring following the activation of the maternal immune system. METHODS Sprague-Dawley rats received a single caudal vein injection of 10 mg/kg polyinosinic:polycytidylic acid (poly I:C) on gestational day 9 and the offspring were comprehensively tested for behaviors in adolescence and adulthood. RESULTS Maternal serum levels of interleukin (IL)-6, IL-1β and tumor necrosis factor-α were elevated in poly I:C-treated dams. The offspring of maternal poly I:C-induced rats showed increased anxiety, impaired social approach, and progressive impaired cognitive and sensorimotor gating function. CONCLUSION Maternal immune activation led to developmental specificity behavioral impairment in offspring.
Collapse
Affiliation(s)
- Tengfei Chen
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Lab of Biological Psychiatry of Xinxiang Medical University, Xinxiang, China
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, China
| | - Huadan Meng
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Lab of Biological Psychiatry of Xinxiang Medical University, Xinxiang, China
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, China
| | - Ni Fang
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Lab of Biological Psychiatry of Xinxiang Medical University, Xinxiang, China
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, China
| | - Peiling Shi
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Lab of Biological Psychiatry of Xinxiang Medical University, Xinxiang, China
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, China
| | - Mengxue Chen
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Lab of Biological Psychiatry of Xinxiang Medical University, Xinxiang, China
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, China
| | - Qing Liu
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Lab of Biological Psychiatry of Xinxiang Medical University, Xinxiang, China
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, China
| | - Luxian Lv
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Lab of Biological Psychiatry of Xinxiang Medical University, Xinxiang, China
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, China
- Henan Province People's Hospital, Zhengzhou, Henan, China
| | - Wenqiang Li
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Key Lab of Biological Psychiatry of Xinxiang Medical University, Xinxiang, China
- International Joint Research Laboratory for Psychiatry and Neuroscience of Henan, Xinxiang, China
- Henan Collaborative Innovation Center of Prevention and Treatment of Mental Disorder, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
4
|
Yotova AY, Li LL, O'Leary A, Tegeder I, Reif A, Courtney MJ, Slattery DA, Freudenberg F. Synaptic proteome perturbations after maternal immune activation: Identification of embryonic and adult hippocampal changes. Brain Behav Immun 2024; 121:351-364. [PMID: 39089536 DOI: 10.1016/j.bbi.2024.07.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 07/10/2024] [Accepted: 07/28/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND Maternal immune activation (MIA) triggers neurobiological changes in offspring, potentially reshaping the molecular synaptic landscape, with the hippocampus being particularly vulnerable. However, critical details regarding developmental timing of these changes and whether they differ between males and females remain unclear. METHODS We induced MIA in C57BL/6J mice on gestational day nine using the viral mimetic poly(I:C) and performed mass spectrometry-based proteomic analyses on hippocampal synaptoneurosomes of embryonic (E18) and adult (20 ± 1 weeks) MIA offspring. RESULTS In the embryonic synaptoneurosomes, MIA led to lipid, polysaccharide, and glycoprotein metabolism pathway disruptions. In the adult synaptic proteome, we observed a dynamic shift toward transmembrane trafficking, intracellular signalling cascades, including cell death and growth, and cytoskeletal organisation. In adults, many associated pathways overlapped between males and females. However, we found distinct sex-specific enrichment of dopaminergic and glutamatergic pathways. We identified 50 proteins altered by MIA in both embryonic and adult samples (28 with the same directionality), mainly involved in presynaptic structure and synaptic vesicle function. We probed human phenome-wide association study data in the cognitive and psychiatric domains, and 49 of the 50 genes encoding these proteins were significantly associated with the investigated phenotypes. CONCLUSIONS Our data emphasise the dynamic effects of viral-like MIA on developing and mature hippocampi and provide novel targets for study following prenatal immune challenges. The 22 proteins that changed directionality from the embryonic to adult hippocampus, suggestive of compensatory over-adaptions, are particularly attractive for future investigations.
Collapse
Affiliation(s)
- Anna Y Yotova
- Goethe University Frankfurt, University Hospital, Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, Frankfurt, Germany; Goethe University Frankfurt, Faculty of Biological Sciences, Institute of Cell Biology and Neuroscience, Frankfurt, Germany
| | - Li-Li Li
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland; Turku Brain and Mind Center, University of Turku and Åbo Akademi University, 20014 Turku, Finland
| | - Aet O'Leary
- Goethe University Frankfurt, University Hospital, Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, Frankfurt, Germany; Department of Neuropsychopharmacology, Institute of Chemistry, University of Tartu, Tartu, Estonia
| | - Irmgard Tegeder
- Goethe University Frankfurt, Faculty of Medicine, Institute of Clinical Pharmacology, Frankfurt, Germany
| | - Andreas Reif
- Goethe University Frankfurt, University Hospital, Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, Frankfurt, Germany; Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| | - Michael J Courtney
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland; Turku Brain and Mind Center, University of Turku and Åbo Akademi University, 20014 Turku, Finland
| | - David A Slattery
- Goethe University Frankfurt, University Hospital, Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, Frankfurt, Germany
| | - Florian Freudenberg
- Goethe University Frankfurt, University Hospital, Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, Frankfurt, Germany; Goethe University Frankfurt, Faculty of Biological Sciences, Institute of Cell Biology and Neuroscience, Frankfurt, Germany.
| |
Collapse
|
5
|
King C, Plakke B. Maternal choline supplementation modulates cognition and induces anti-inflammatory signaling in the prefrontal cortices of adolescent rats exposed to maternal immune activation. Brain Behav Immun Health 2024; 40:100836. [PMID: 39206430 PMCID: PMC11350509 DOI: 10.1016/j.bbih.2024.100836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 07/22/2024] [Accepted: 08/01/2024] [Indexed: 09/04/2024] Open
Abstract
Maternal infection has long been described as a risk factor for neurodevelopmental disorders, especially autism spectrum disorders (ASD) and schizophrenia. Although many pathogens do not cross the placenta and infect the developing fetus directly, the maternal immune response to them is sufficient to alter fetal neurodevelopment, a phenomenon termed maternal immune activation (MIA). Low maternal choline is also a risk factor for neurodevelopmental disorders, and most pregnant people do not receive enough of it. In addition to its role in neurodevelopment, choline is capable of inducing anti-inflammatory signaling through a nicotinic pathway. Therefore, it was hypothesized that maternal choline supplementation would blunt the neurodevelopmental impact of MIA in offspring through long-term instigation of cholinergic anti-inflammatory signaling. To model MIA in rats, the viral mimetic polyinosinic:polycytidylic acid (poly(I:C)) was used to elicit a maternal antiviral innate immune response in dams both with and without choline supplementation. Offspring were reared to both early and late adolescent stages (postnatal days 28 and 50, respectively), where anxiety-related behaviors and cognition were examined. After behavioral testing, animals were euthanized, and their prefrontal cortices (PFCs) were collected for analysis. MIA offspring demonstrated sex-specific patterns of altered cognition and repetitive behaviors, which were modulated by maternal choline supplementation. Choline supplementation also bolstered anti-inflammatory signaling in the PFCs of MIA animals at both early and late adolescent stages. These findings suggest that maternal choline supplementation may be sufficient to blunt some of the behavioral and neurobiological impacts of inflammatory exposures in utero, indicating that it may be a cheap, safe, and effective intervention for neurodevelopmental disorders.
Collapse
Affiliation(s)
- Cole King
- Department of Psychological Sciences, Kansas State University, 1114 Mid-Campus Drive, Manhattan, KS, 66502, USA
| | - Bethany Plakke
- Department of Psychological Sciences, Kansas State University, 1114 Mid-Campus Drive, Manhattan, KS, 66502, USA
| |
Collapse
|
6
|
Sal-Sarria S, Conejo NM, González-Pardo H. Maternal immune activation and its multifaceted effects on learning and memory in rodent offspring: A systematic review. Neurosci Biobehav Rev 2024; 164:105844. [PMID: 39106940 DOI: 10.1016/j.neubiorev.2024.105844] [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: 05/27/2024] [Revised: 07/25/2024] [Accepted: 08/02/2024] [Indexed: 08/09/2024]
Abstract
This systematic review explored the impact of maternal immune activation (MIA) on learning and memory behavior in offspring, with a particular focus on sexual dimorphism. We analyzed 20 experimental studies involving rodent models (rats and mice) exposed to either lipopolysaccharide (LPS) or POLY I:C during gestation following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. Our findings reveal that most studies report a detrimental impact of MIA on the learning and memory performance of offspring, highlighting the significant role of prenatal environmental factors in neurodevelopment. Furthermore, this review underscores the complex effects of sex, with males often exhibiting more pronounced cognitive impairment compared to females. Notably, a small subset of studies report enhanced cognitive function following MIA, suggesting complex, context-dependent outcomes of prenatal immune challenges. This review also highlights sex differences caused by the effects of MIA in terms of cytokine responses, alterations in gene expression, and differences in microglial responses as factors that contribute to the cognitive outcomes observed.
Collapse
Affiliation(s)
- Saúl Sal-Sarria
- Laboratory of Neuroscience, Department of Psychology, University of Oviedo, Oviedo, Spain; Institute of Neurosciences of the Principality of Asturias (INEUROPA), Oviedo, Spain; Health Research Institute of the Principality of Asturias (ISPA), Oviedo, Spain.
| | - Nélida M Conejo
- Laboratory of Neuroscience, Department of Psychology, University of Oviedo, Oviedo, Spain; Institute of Neurosciences of the Principality of Asturias (INEUROPA), Oviedo, Spain; Health Research Institute of the Principality of Asturias (ISPA), Oviedo, Spain.
| | - Héctor González-Pardo
- Laboratory of Neuroscience, Department of Psychology, University of Oviedo, Oviedo, Spain; Institute of Neurosciences of the Principality of Asturias (INEUROPA), Oviedo, Spain; Health Research Institute of the Principality of Asturias (ISPA), Oviedo, Spain.
| |
Collapse
|
7
|
Godavarthi SK, Li HQ, Pratelli M, Spitzer NC. Embryonic exposure to environmental factors drives transmitter switching in the neonatal mouse cortex causing autistic-like adult behavior. Proc Natl Acad Sci U S A 2024; 121:e2406928121. [PMID: 39178233 PMCID: PMC11363343 DOI: 10.1073/pnas.2406928121] [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: 04/06/2024] [Accepted: 07/23/2024] [Indexed: 08/25/2024] Open
Abstract
Autism spectrum disorders (ASD) can be caused by environmental factors. These factors act early in the development of the nervous system and induce stereotyped repetitive behaviors and diminished social interactions, among other outcomes. Little is known about how these behaviors are produced. In pregnant women, delivery of valproic acid (VPA) (to control seizure activity or stabilize mood) or immune activation by a virus increases the incidence of ASD in offspring. We found that either VPA or Poly Inosine:Cytosine (which mimics a viral infection), administered at mouse embryonic day 12.5, induced a neurotransmitter switch from GABA to glutamate in PV- and CCK-expressing interneurons in the medial prefrontal cortex by postnatal day 10. The switch was present for only a brief period during early postnatal development, observed in male and female mice at postnatal day 21 and reversed in both males and females by postnatal day 30. At postnatal day 90, male mice exhibited stereotyped repetitive behaviors and diminished social interaction while female mice exhibited only stereotyped repetitive behavior. Transfecting GAD1 in PV- and CCK-expressing interneurons at postnatal day 10, to reintroduce GABA expression, overrode the switch and prevented expression of autistic-like behavior. These findings point to an important role of neurotransmitter switching in mediating the environmental causes of autism.
Collapse
Affiliation(s)
- Swetha K. Godavarthi
- Neurobiology Department, University of California San Diego, La Jolla, CA92093
- Kavli Institute for Brain & Mind, University of California San Diego, La Jolla, CA92093
| | - Hui-quan Li
- Neurobiology Department, University of California San Diego, La Jolla, CA92093
- Kavli Institute for Brain & Mind, University of California San Diego, La Jolla, CA92093
| | - Marta Pratelli
- Neurobiology Department, University of California San Diego, La Jolla, CA92093
- Kavli Institute for Brain & Mind, University of California San Diego, La Jolla, CA92093
| | - Nicholas C. Spitzer
- Neurobiology Department, University of California San Diego, La Jolla, CA92093
- Kavli Institute for Brain & Mind, University of California San Diego, La Jolla, CA92093
| |
Collapse
|
8
|
Maggioni E, Pigoni A, Fontana E, Delvecchio G, Bonivento C, Bianchi V, Mauri M, Bellina M, Girometti R, Agarwal N, Nobile M, Brambilla P. Right frontal cingulate cortex mediates the effect of prenatal complications on youth internalizing behaviors. Mol Psychiatry 2024; 29:2074-2083. [PMID: 38378927 PMCID: PMC11408263 DOI: 10.1038/s41380-024-02475-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 01/26/2024] [Accepted: 01/31/2024] [Indexed: 02/22/2024]
Abstract
Prenatal and perinatal complications represent well-known risk factors for the future development of psychiatric disorders. Such influence might become manifested during childhood and adolescence, as key periods for brain and behavioral changes. Internalizing and externalizing behaviors in adolescence have been associated with the risk of psychiatric onset later in life. Both brain morphology and behavior seem to be affected by obstetric complications, but a clear link among these three aspects is missing. Here, we aimed at analyzing the association between prenatal and perinatal complications, behavioral issues, and brain volumes in a group of children and adolescents. Eighty-two children and adolescents with emotional-behavioral problems underwent clinical and 3 T brain magnetic resonance imaging (MRI) assessments. The former included information on behavior, through the Child Behavior Checklist/6-18 (CBCL/6-18), and on the occurrence of obstetric complications. The relationships between clinical and gray matter volume (GMV) measures were investigated through multiple generalized linear models and mediation models. We found a mutual link between prenatal complications, GMV alterations in the frontal gyrus, and withdrawn problems. Specifically, complications during pregnancy were associated with higher CBCL/6-18 withdrawn scores and GMV reductions in the right superior frontal gyrus and anterior cingulate cortex. Finally, a mediation effect of these GMV measures on the association between prenatal complications and the withdrawn dimension was identified. Our findings suggest a key role of obstetric complications in affecting brain structure and behavior. For the first time, a mediator role of frontal GMV in the relationship between prenatal complications and internalizing symptoms was suggested. Once replicated on independent cohorts, this evidence will have relevant implications for planning preventive interventions.
Collapse
Affiliation(s)
- Eleonora Maggioni
- Department of Electronics Information and Bioengineering, Politecnico di Milano, Milano, Italy
| | - Alessandro Pigoni
- Department of Neurosciences and Mental Health, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Social and Affective Neuroscience Group, MoMiLab, IMT School for Advanced Studies Lucca, Lucca, Italy
| | - Elisa Fontana
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Giuseppe Delvecchio
- Department of Neurosciences and Mental Health, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | | | - Valentina Bianchi
- Child and Adolescent Psychiatry Unit, Scientific Institute IRCCS "Eugenio Medea", Bosisio Parini (Lc), Italy
| | - Maddalena Mauri
- Child and Adolescent Psychiatry Unit, Scientific Institute IRCCS "Eugenio Medea", Bosisio Parini (Lc), Italy
| | - Monica Bellina
- Child and Adolescent Psychiatry Unit, Scientific Institute IRCCS "Eugenio Medea", Bosisio Parini (Lc), Italy
| | - Rossano Girometti
- Institute of Radiology, Department of Medicine (DMED), University of Udine, Udine, Italy
- University Hospital S. Maria Della Misericordia, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC), Udine, Italy
| | - Nivedita Agarwal
- Neuroimaging Unit, Scientific Institute IRCCS "Eugenio Medea", Bosisio Parini (Lc), Italy
| | - Maria Nobile
- Child and Adolescent Psychiatry Unit, Scientific Institute IRCCS "Eugenio Medea", Bosisio Parini (Lc), Italy
| | - Paolo Brambilla
- Department of Neurosciences and Mental Health, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy.
| |
Collapse
|
9
|
Ünal D, Varol AB, Köse TB, Koçak EE. Morphological Correlates of Behavioral Variation in Autism Spectrum Disorder Groups in A Maternal Immune Activation Model. Noro Psikiyatr Ars 2024; 67:195-201. [PMID: 39258126 PMCID: PMC11382561 DOI: 10.29399/npa.28637] [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: 10/09/2023] [Accepted: 11/16/2023] [Indexed: 09/12/2024] Open
Abstract
Introduction Clinical heterogeneity is one of the biggest challenges for researchers studying underlying neurobiological mechanisms in Autism Spectrum Disorder (ASD). We aimed to use polyinosinic-polycytidylic acid [Poly (I:C)] induced maternal immune activation mice model to investigate the behavioral variation and the role of brain circuits related to symptom clusters in ASD. For this purpose, behavioral tests were applied to offsprings and regional thickness was measured from histological brain sections in medial prefrontal cortex, hippocampus and striatum. Methods Pups of intraperitoneal Poly (I:C)-applied mothers (n: 14) and phosphate buffered saline-applied mothers (n: 6) were used for this study. We used three chamber socialization test and social memory test to evaluate social behavior deficit in mice. Marble burying test was used for assessing stereotypic behavior and new object recognition test for learning and cognitive flexibility. Three subgroups (n: 4 for each) were determined according to behavioral test parameters. Regional thickness was measured in medial prefrontal cortex, hippocampus and striatum and compared between subgroups. Results We detected that the behavioral differences were distributed in a spectrum as expected in the clinic and also detected increased hippocampus thickness in the stereotypic behavior dominant Poly (I:C) subgroup. Conclusion Poly (I:C) induced maternal immune activation model creates the behavioral variation and cortical development differences that are seen in relation with symptom groups in ASD.
Collapse
Affiliation(s)
- Dilek Ünal
- Hacettepe University School of Medicine, Department of Child and Adolescent Psychiatry, Ankara, Turkey
| | - Aslıhan Bahadır Varol
- Hacettepe University School of Medicine, Neurological and Psychiatric Sciences Institute, Ankara, Turkey
| | - Tansu Bilge Köse
- Hacettepe University School of Medicine, Neurological and Psychiatric Sciences Institute, Ankara, Turkey
| | - Emine Eren Koçak
- Hacettepe University School of Medicine, Neurological and Psychiatric Sciences Institute, Ankara, Turkey
| |
Collapse
|
10
|
Vacharasin JM, Ward JA, McCord MM, Cox K, Imitola J, Lizarraga SB. Neuroimmune mechanisms in autism etiology - untangling a complex problem using human cellular models. OXFORD OPEN NEUROSCIENCE 2024; 3:kvae003. [PMID: 38665176 PMCID: PMC11044813 DOI: 10.1093/oons/kvae003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 01/13/2024] [Accepted: 01/31/2024] [Indexed: 04/28/2024]
Abstract
Autism spectrum disorder (ASD) affects 1 in 36 people and is more often diagnosed in males than in females. Core features of ASD are impaired social interactions, repetitive behaviors and deficits in verbal communication. ASD is a highly heterogeneous and heritable disorder, yet its underlying genetic causes account only for up to 80% of the cases. Hence, a subset of ASD cases could be influenced by environmental risk factors. Maternal immune activation (MIA) is a response to inflammation during pregnancy, which can lead to increased inflammatory signals to the fetus. Inflammatory signals can cross the placenta and blood brain barriers affecting fetal brain development. Epidemiological and animal studies suggest that MIA could contribute to ASD etiology. However, human mechanistic studies have been hindered by a lack of experimental systems that could replicate the impact of MIA during fetal development. Therefore, mechanisms altered by inflammation during human pre-natal brain development, and that could underlie ASD pathogenesis have been largely understudied. The advent of human cellular models with induced pluripotent stem cell (iPSC) and organoid technology is closing this gap in knowledge by providing both access to molecular manipulations and culturing capability of tissue that would be otherwise inaccessible. We present an overview of multiple levels of evidence from clinical, epidemiological, and cellular studies that provide a potential link between higher ASD risk and inflammation. More importantly, we discuss how stem cell-derived models may constitute an ideal experimental system to mechanistically interrogate the effect of inflammation during the early stages of brain development.
Collapse
Affiliation(s)
- Janay M Vacharasin
- Department of Biological Sciences, and Center for Childhood Neurotherapeutics, Univ. of South Carolina, 715 Sumter Street, Columbia, SC 29208, USA
- Department of Biological Sciences, Francis Marion University, 4822 East Palmetto Street, Florence, S.C. 29506, USA
| | - Joseph A Ward
- Department of Molecular Biology, Cell Biology, & Biochemistry, Brown University, 185 Meeting Street, Providence, RI 02912, USA
- Center for Translational Neuroscience, Carney Institute of Brain Science, Brown University, 70 Ship Street, Providence, RI 02903, USA
| | - Mikayla M McCord
- Department of Biological Sciences, and Center for Childhood Neurotherapeutics, Univ. of South Carolina, 715 Sumter Street, Columbia, SC 29208, USA
| | - Kaitlin Cox
- Department of Biological Sciences, and Center for Childhood Neurotherapeutics, Univ. of South Carolina, 715 Sumter Street, Columbia, SC 29208, USA
| | - Jaime Imitola
- Laboratory of Neural Stem Cells and Functional Neurogenetics, UConn Health, Departments of Neuroscience, Neurology, Genetics and Genome Sciences, UConn Health, 263 Farmington Avenue, Farmington, CT 06030-5357, USA
| | - Sofia B Lizarraga
- Department of Molecular Biology, Cell Biology, & Biochemistry, Brown University, 185 Meeting Street, Providence, RI 02912, USA
- Center for Translational Neuroscience, Carney Institute of Brain Science, Brown University, 70 Ship Street, Providence, RI 02903, USA
| |
Collapse
|
11
|
Zhang L, Lin C, Zhu J, He Y, Zhan M, Xia X, Yang N, Yang K, Wang B, Zhong Z, Wang Y, Ding W, Yang Y. Restoring prefrontal cortical excitation-inhibition balance with cannabidiol ameliorates neurobehavioral abnormalities in a mouse model of neurodevelopmental disorders. Neuropharmacology 2023; 240:109715. [PMID: 37716533 DOI: 10.1016/j.neuropharm.2023.109715] [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: 06/08/2023] [Revised: 09/07/2023] [Accepted: 09/08/2023] [Indexed: 09/18/2023]
Abstract
Maternal immune activation (MIA) resulting from viral infections during pregnancy is linked to increased rates of neurodevelopmental disorders in offspring. However, the mechanisms underlying MIA-induced neurobehavioral abnormalities remain unclear. Here, we used a poly (I:C)-induced MIA mouse model to demonstrate the presence of multiple behavioral deficits in male offspring. Through RNA sequencing (RNA-seq), we identified significant upregulation of genes involved in axonogenesis, synaptogenesis, and glutamatergic synaptic neurotransmission in the mPFC of MIA mice. Electrophysiological analyses further revealed an excitatory-inhibitory (E/I) synaptic imbalance in mPFC pyramidal neurons, leading to hyperactivity in this brain region. Cannabidiol (CBD) effectively alleviated the behavioral abnormalities observed in MIA offspring by reducing glutamatergic transmission and enhancing GABAergic neurotransmission of mPFC pyramidal neurons. Activation of GPR55 by lipid lysophosphatidylinositol (LPI), an endogenous GPR55 agonist, specifically in the mPFC of healthy animals led to MIA-associated behavioral phenotypes, which CBD could effectively reverse. Moreover, we found that a GPR55 antagonist can mimic CBD's beneficial effects, indicating that CBD's therapeutic effects are mediated via the LPI-GPR55 signaling pathway. Therefore, we identified mPFC as a primary node of a neural network that mediates MIA-induced behavioral abnormalities in offspring. Our work provides insights into the mechanisms underlying the developmental consequences of MIA and identifies CBD as a promising therapeutic approach to alleviate these effects.
Collapse
Affiliation(s)
- Lu Zhang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Chunqiao Lin
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Jiushuang Zhu
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yan He
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Meng Zhan
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Xiuwen Xia
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Ni Yang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Kun Yang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Baojia Wang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Zhanqion Zhong
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yili Wang
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Weijun Ding
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Youjun Yang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| |
Collapse
|
12
|
Devaraju M, Li A, Ha S, Li M, Shivakumar M, Li H, Nishiguchi EP, Gérardin P, Waldorf KA, Al-Haddad BJS. Beyond TORCH: A narrative review of the impact of antenatal and perinatal infections on the risk of disability. Neurosci Biobehav Rev 2023; 153:105390. [PMID: 37708918 PMCID: PMC10617835 DOI: 10.1016/j.neubiorev.2023.105390] [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: 06/13/2023] [Revised: 09/07/2023] [Accepted: 09/10/2023] [Indexed: 09/16/2023]
Abstract
Infections and inflammation during pregnancy or early life can alter child neurodevelopment and increase the risk for structural brain abnormalities and mental health disorders. There is strong evidence that TORCH infections (i.e., Treponema pallidum, Toxoplasma gondii, rubella virus, cytomegalovirus, herpes virus) alter fetal neurodevelopment across multiple developmental domains and contribute to motor and cognitive disabilities. However, the impact of a broader range of viral and bacterial infections on fetal development and disability is less well understood. We performed a literature review of human studies to identify gaps in the link between maternal infections, inflammation, and several neurodevelopmental domains. We found strong and moderate evidence respectively for a higher risk of motor and cognitive delays and disabilities in offspring exposed to a range of non-TORCH pathogens during fetal life. In contrast, there is little evidence for an increased risk of language and sensory disabilities. While guidelines for TORCH infection prevention during pregnancy are common, further consideration for prevention of non-TORCH infections during pregnancy for fetal neuroprotection may be warranted.
Collapse
Affiliation(s)
- Monica Devaraju
- University of Washington, School of Medicine, 1959 NE Pacific St, Seattle, WA 98195, USA; University of Washington, Department of Obstetrics, 1959 NE Pacific St, Seattle, WA 98195, USA
| | - Amanda Li
- University of Washington, Department of Obstetrics, 1959 NE Pacific St, Seattle, WA 98195, USA; Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH, USA
| | - Sandy Ha
- University of Washington, Department of Obstetrics, 1959 NE Pacific St, Seattle, WA 98195, USA
| | - Miranda Li
- University of Washington, School of Medicine, 1959 NE Pacific St, Seattle, WA 98195, USA; University of Washington, Department of Obstetrics, 1959 NE Pacific St, Seattle, WA 98195, USA
| | - Megana Shivakumar
- University of Washington, Department of Obstetrics, 1959 NE Pacific St, Seattle, WA 98195, USA
| | - Hanning Li
- University of Washington, Department of Obstetrics, 1959 NE Pacific St, Seattle, WA 98195, USA
| | - Erika Phelps Nishiguchi
- University of Hawaii, Department of Pediatrics, Division of Community Pediatrics, 1319 Punahou St, Honolulu, HI, USA
| | - Patrick Gérardin
- INSERM CIC1410, Centre Hospitalier Universitaire de la Réunion, Saint Pierre, Réunion, France; Platform for Clinical and Translational Research, Centre Hospitalier Universitaire, Saint Pierre, Réunion, France
| | - Kristina Adams Waldorf
- University of Washington, Department of Obstetrics, 1959 NE Pacific St, Seattle, WA 98195, USA.
| | - Benjamin J S Al-Haddad
- University of Minnesota, Department of Pediatrics, Division of Neonatology, Academic Office Building, 2450 Riverside Ave S AO-401, Minneapolis, MN 55454, USA; Masonic Institute for the Developing Brain, 2025 E River Pkwy, Minneapolis, MN 55414, USA.
| |
Collapse
|
13
|
Powell SB, Swerdlow NR. The Relevance of Animal Models of Social Isolation and Social Motivation for Understanding Schizophrenia: Review and Future Directions. Schizophr Bull 2023; 49:1112-1126. [PMID: 37527471 PMCID: PMC10483472 DOI: 10.1093/schbul/sbad098] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/03/2023]
Abstract
BACKGROUND AND HYPOTHESES Social dysfunction in schizophrenia includes symptoms of withdrawal and deficits in social skills, social cognition, and social motivation. Based on the course of illness, with social withdrawal occurring prior to psychosis onset, it is likely that the severity of social withdrawal/isolation contributes to schizophrenia neuropathology. STUDY DESIGN We review the current literature on social isolation in rodent models and provide a conceptual framework for its relationship to social withdrawal and neural circuit dysfunction in schizophrenia. We next review preclinical tasks of social behavior used in schizophrenia-relevant models and discuss strengths and limitations of existing approaches. Lastly, we consider new effort-based tasks of social motivation and their potential for translational studies in schizophrenia. STUDY RESULTS Social isolation rearing in rats produces profound differences in behavior, pharmacologic sensitivity, and neurochemistry compared to socially reared rats. Rodent models relevant to schizophrenia exhibit deficits in social behavior as measured by social interaction and social preference tests. Newer tasks of effort-based social motivation are being developed in rodents to better model social motivation deficits in neuropsychiatric disorders. CONCLUSIONS While experimenter-imposed social isolation provides a viable experimental model for understanding some biological mechanisms linking social dysfunction to clinical and neural pathology in schizophrenia, it bypasses critical antecedents to social isolation in schizophrenia, notably deficits in social reward and social motivation. Recent efforts at modeling social motivation using effort-based tasks in rodents have the potential to quantify these antecedents, identify models (eg, developmental, genetic) that produce deficits, and advance pharmacological treatments for social motivation.
Collapse
Affiliation(s)
- Susan B Powell
- Research Service, VA San Diego Healthcare System, La Jolla, CA, USA
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
- Veterans Affairs VISN22 Mental Illness Research, Education and Clinical Center, La Jolla, CA, USA
| | - Neal R Swerdlow
- Research Service, VA San Diego Healthcare System, La Jolla, CA, USA
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
- Veterans Affairs VISN22 Mental Illness Research, Education and Clinical Center, La Jolla, CA, USA
| |
Collapse
|
14
|
Tranter MM, Aggarwal S, Young JW, Dillon DG, Barnes SA. Reinforcement learning deficits exhibited by postnatal PCP-treated rats enable deep neural network classification. Neuropsychopharmacology 2023; 48:1377-1385. [PMID: 36509858 PMCID: PMC10354061 DOI: 10.1038/s41386-022-01514-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 11/21/2022] [Accepted: 11/26/2022] [Indexed: 12/14/2022]
Abstract
The ability to appropriately update the value of a given action is a critical component of flexible decision making. Several psychiatric disorders, including schizophrenia, are associated with impairments in flexible decision making that can be evaluated using the probabilistic reversal learning (PRL) task. The PRL task has been reverse-translated for use in rodents. Disrupting glutamate neurotransmission during early postnatal neurodevelopment in rodents has induced behavioral, cognitive, and neuropathophysiological abnormalities relevant to schizophrenia. Here, we tested the hypothesis that using the NMDA receptor antagonist phencyclidine (PCP) to disrupt postnatal glutamatergic transmission in rats would lead to impaired decision making in the PRL. Consistent with this hypothesis, compared to controls the postnatal PCP-treated rats completed fewer reversals and exhibited disruptions in reward and punishment sensitivity (i.e., win-stay and lose-shift responding, respectively). Moreover, computational analysis of behavior revealed that postnatal PCP-treatment resulted in a pronounced impairment in the learning rate throughout PRL testing. Finally, a deep neural network (DNN) trained on the rodent behavior could accurately predict the treatment group of subjects. These data demonstrate that disrupting early postnatal glutamatergic neurotransmission impairs flexible decision making and provides evidence that DNNs can be trained on behavioral datasets to accurately predict the treatment group of new subjects, highlighting the potential for DNNs to aid in the diagnosis of schizophrenia.
Collapse
Affiliation(s)
- Michael M Tranter
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093, USA
- Department of Mental Health, VA San Diego Healthcare System, La Jolla, CA, 92093, USA
| | - Samarth Aggarwal
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093, USA
| | - Jared W Young
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093, USA
- Department of Mental Health, VA San Diego Healthcare System, La Jolla, CA, 92093, USA
| | - Daniel G Dillon
- Center for Depression, Anxiety and Stress Research, McLean Hospital, Belmont, MA, 02478, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Samuel A Barnes
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093, USA.
- Department of Mental Health, VA San Diego Healthcare System, La Jolla, CA, 92093, USA.
| |
Collapse
|
15
|
Yotova AY, Li LL, O’Leary A, Tegeder I, Reif A, Courtney MJ, Slattery DA, Freudenberg F. Embryonic and adult synaptic proteome perturbations after maternal immune activation: Identification of persistent changes relevant for early intervention. RESEARCH SQUARE 2023:rs.3.rs-3100753. [PMID: 37461513 PMCID: PMC10350178 DOI: 10.21203/rs.3.rs-3100753/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/28/2023]
Abstract
Maternal infections during pregnancy pose an increased risk for neurodevelopmental psychiatric disorders (NPDs) in the offspring. Here, we examined age- and sex-dependent dynamic changes of the hippocampal synaptic proteome after maternal immune activation (MIA) in embryonic and adult mice. Adult male and female MIA offspring exhibited social deficits and sex-specific depression-like behaviours, among others, validating the model. Furthermore, we observed dose-, age-, and sex-dependent synaptic proteome differences. Analysis of the embryonic synaptic proteome implicates sphingolipid and ketoacid metabolism pathway disruptions during neurodevelopment for NPD-pertinent sequelae. In the embryonic hippocampus, prenatal immune activation also led to changes in neuronal guidance, glycosphingolipid metabolism important for signalling and myelination, and post-translational modification of proteins that regulate intercellular interaction and developmental timing. In adulthood, the observed changes in synaptoneurosomes revealed a dynamic shift toward transmembrane trafficking, intracellular signalling cascades, and hormone-mediated metabolism. Importantly, 68 of the proteins with differential abundance in the embryonic brains of MIA offspring were also altered in adulthood, 75% of which retained their directionality. These proteins are involved in synaptic organisation, neurotransmitter receptor regulation, and the vesicle cycle. A cluster of persistently upregulated proteins, including AKT3, PAK1/3, PPP3CA, formed a functional network enriched in the embryonic brain that is involved in cellular responses to environmental stimuli. To infer a link between the overlapping protein alterations and cognitive and psychiatric traits, we probed human phenome-wise association study data for cognitive and psychiatric phenotypes and all, but PORCN were significantly associated with the investigated domains. Our data provide insights into the dynamic effects of an early prenatal immune activation on developing and mature hippocampi and highlights targets for early intervention in individuals exposed to such immune challenges.
Collapse
Affiliation(s)
- Anna Y. Yotova
- Goethe University Frankfurt, University Hospital, Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, Frankfurt, Germany
- Goethe University Frankfurt, Faculty of Biological Sciences, Institute of Cell Biology and Neuroscience, Frankfurt, Germany
| | - Li-Li Li
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland; Turku Brain and Mind Center, University of Turku and Åbo Akademi University, 20014, Turku, Finland
| | - Aet O’Leary
- Goethe University Frankfurt, University Hospital, Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, Frankfurt, Germany
- Department of Neuropsychopharmacology, Institute of Chemistry, University of Tartu, Tartu, Estonia
| | - Irmgard Tegeder
- Goethe University Frankfurt, Faculty of Medicine, Institute of Clinical Pharmacology, Frankfurt, Germany
| | - Andreas Reif
- Goethe University Frankfurt, University Hospital, Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, Frankfurt, Germany
| | - Michael J Courtney
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland; Turku Brain and Mind Center, University of Turku and Åbo Akademi University, 20014, Turku, Finland
| | - David A. Slattery
- Goethe University Frankfurt, University Hospital, Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, Frankfurt, Germany
| | - Florian Freudenberg
- Goethe University Frankfurt, University Hospital, Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, Frankfurt, Germany
- Goethe University Frankfurt, Faculty of Biological Sciences, Institute of Cell Biology and Neuroscience, Frankfurt, Germany
| |
Collapse
|
16
|
Taskiran SY, Taskiran M, Unal G, Golgeli A. Group I mGluRs positive allosteric modulators improved schizophrenia-related behavioral and molecular deficits in the Poly I:C rat model. Pharmacol Biochem Behav 2023:173593. [PMID: 37390974 DOI: 10.1016/j.pbb.2023.173593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/16/2023] [Accepted: 06/26/2023] [Indexed: 07/02/2023]
Abstract
RATIONALE Maternal polyinosinic-polycytidylic acid (Poly I:C) exposure leads to an increase in various proinflammatory cytokines and causes schizophrenia-like symptoms in offspring. In recent years, group I metabotropic glutamate receptors (mGluRs) have emerged as a potential target in the pathophysiology of schizophrenia. OBJECTIVES The aim of our study was to investigate the behavioral and molecular changes by using the mGlu1 receptor positive allosteric modulator (PAM) agent RO 67-7476, and the negative allosteric modulator (NAM) agent JNJ 16259685 and the mGlu5 receptor PAM agent VU-29, and NAM agent fenobam in the Poly I:C-induced schizophrenia model in rats. METHODS Female Wistar albino rats were treated with Poly I:C on day 14 of gestation after mating. On the postnatal day (PND) 35, 56 and 84, behavioral tests were performed in the male offspring. On the PND84, brain tissue was collected and the level of proinflammatory cytokines was determined by ELISA method. RESULTS Poly I:C caused impairments in all behavioral tests and increased the levels of proinflammatory cytokines. While PAM agents caused significant improvements in prepulse inhibition (PPI), novel object recognition (NOR), spontaneous alternation and reference memory tests, they brought the levels of proinflammatory cytokines closer to the control group. NAM agents were ineffective on behavioral tests. It was observed that PAM agents significantly improved Poly I:C-induced disruption in behavioral and molecular analyses. CONCLUSIONS These results suggest that PAM agents, particularly the mGlu5 receptor VU-29, are also promising and could be a potential target in schizophrenia.
Collapse
Affiliation(s)
| | - Mehmet Taskiran
- Department of Biology, Faculty of Science, Erciyes University, Kayseri, Turkey.
| | - Gokhan Unal
- Department of Pharmacology, Faculty of Pharmacy, Erciyes University, Kayseri, Turkey.
| | - Asuman Golgeli
- Department of Physiology, Faculty of Medicine, Erciyes University, Kayseri, Turkey.
| |
Collapse
|
17
|
Chamera K, Curzytek K, Kamińska K, Trojan E, Leśkiewicz M, Tylek K, Regulska M, Basta-Kaim A. Insights into the Potential Impact of Quetiapine on the Microglial Trajectory and Inflammatory Response in Organotypic Cortical Cultures Derived from Rat Offspring. Biomedicines 2023; 11:biomedicines11051405. [PMID: 37239076 DOI: 10.3390/biomedicines11051405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/27/2023] [Accepted: 05/05/2023] [Indexed: 05/28/2023] Open
Abstract
Atypical antipsychotics currently constitute the first-line medication for schizophrenia, with quetiapine being one of the most commonly prescribed representatives of the group. Along with its specific affinity for multiple receptors, this compound exerts other biological characteristics, among which anti-inflammatory effects are strongly suggested. Simultaneously, published data indicated that inflammation and microglial activation could be diminished by stimulation of the CD200 receptor (CD200R), which takes place by binding to its ligand (CD200) or soluble CD200 fusion protein (CD200Fc). Therefore, in the present study, we sought to evaluate whether quetiapine could affect certain aspects of microglial activity, including the CD200-CD200R and CX3CL1-CX3CR1 axes, which are involved in the regulation of neuron-microglia interactions, as well as the expression of selected markers of the pro- and anti-inflammatory profile of microglia (Cd40, Il-1β, Il-6, Cebpb, Cd206, Arg1, Il-10 and Tgf-β). Concurrently, we examined the impact of quetiapine and CD200Fc on the IL-6 and IL-10 protein levels. The abovementioned aspects were investigated in organotypic cortical cultures (OCCs) prepared from the offspring of control rats (control OCCs) or those subjected to maternal immune activation (MIA OCCs), which is a widely implemented approach to explore schizophrenia-like disturbances in animals. The experiments were performed under basal conditions and after additional exposure to the bacterial endotoxin lipopolysaccharide (LPS), according to the "two-hit" hypothesis of schizophrenia. The results of our research revealed differences between control and MIA OCCs under basal conditions and in response to treatment with LPS in terms of lactate dehydrogenase and nitric oxide release as well as Cd200r, Il-1β, Il-6 and Cd206 expression. The additional stimulation with the bacterial endotoxin resulted in a notable change in the mRNA levels of pro- and anti-inflammatory microglial markers in both types of OCCs. Quetiapine diminished the influence of LPS on Il-1β, Il-6, Cebpb and Arg1 expression in control OCCs as well as on IL-6 and IL-10 levels in MIA OCCs. Moreover, CD200Fc reduced the impact of the bacterial endotoxin on IL-6 production in MIA OCCs. Thus, our results demonstrated that quetiapine, as well as the stimulation of CD200R by CD200Fc, beneficially affected LPS-induced neuroimmunological changes, including microglia-related activation.
Collapse
Affiliation(s)
- Katarzyna Chamera
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St., 31-343 Kraków, Poland
| | - Katarzyna Curzytek
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St., 31-343 Kraków, Poland
| | - Kinga Kamińska
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St., 31-343 Kraków, Poland
| | - Ewa Trojan
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St., 31-343 Kraków, Poland
| | - Monika Leśkiewicz
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St., 31-343 Kraków, Poland
| | - Kinga Tylek
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St., 31-343 Kraków, Poland
| | - Magdalena Regulska
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St., 31-343 Kraków, Poland
| | - Agnieszka Basta-Kaim
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St., 31-343 Kraków, Poland
| |
Collapse
|
18
|
Dong Y, Weng J, Zhu Y, Sun D, He W, Chen Q, Cheng J, Zhu Y, Jiang Y. Transcriptomic profiling of the developing brain revealed cell-type and brain-region specificity in a mouse model of prenatal stress. BMC Genomics 2023; 24:86. [PMID: 36829105 PMCID: PMC9951484 DOI: 10.1186/s12864-023-09186-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 02/14/2023] [Indexed: 02/26/2023] Open
Abstract
BACKGROUND Prenatal stress (PS) is considered as a risk factor for many mental disorders. PS-induced transcriptomic alterations may contribute to the functional dysregulation during brain development. Here, we used RNA-seq to explore changes of gene expression in the mouse fetal brain after prenatal exposure to chronic unpredictable mild stress (CUMS). RESULTS We compared the stressed brains to the controls and identified groups of significantly differentially expressed genes (DEGs). GO analysis on up-regulated DEGs revealed enrichment for the cell cycle pathways, while down-regulated DEGs were mostly enriched in the neuronal pathways related to synaptic transmission. We further performed cell-type enrichment analysis using published scRNA-seq data from the fetal mouse brain and revealed cell-type-specificity for up- and down-regulated DEGs, respectively. The up-regulated DEGs were highly enriched in the radial glia, while down-regulated DEGs were enriched in different types of neurons. Cell deconvolution analysis further showed altered cell fractions in the stressed brain, indicating accumulation of neuroblast and impaired neurogenesis. Moreover, we also observed distinct brain-region expression pattern when mapping DEGs onto the developing Allen brain atlas. The up-regulated DEGs were primarily enriched in the dorsal forebrain regions including the cortical plate and hippocampal formation. Surprisingly, down-regulated DEGs were found excluded from the cortical region, but highly expressed on various regions in the ventral forebrain, midbrain and hindbrain. CONCLUSION Taken together, we provided an unbiased data source for transcriptomic alterations of the whole fetal brain after chronic PS, and reported differential cell-type and brain-region vulnerability of the developing brain in response to environmental insults during the pregnancy.
Collapse
Affiliation(s)
- Yuhao Dong
- grid.8547.e0000 0001 0125 2443Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, 200032 Shanghai, China
| | - Jie Weng
- grid.8547.e0000 0001 0125 2443Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, 200032 Shanghai, China
| | - Yueyan Zhu
- grid.8547.e0000 0001 0125 2443Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, 200032 Shanghai, China
| | - Daijing Sun
- grid.8547.e0000 0001 0125 2443Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, 200032 Shanghai, China
| | - Wei He
- grid.8547.e0000 0001 0125 2443Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, 200032 Shanghai, China
| | - Qi Chen
- grid.8547.e0000 0001 0125 2443Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, 200032 Shanghai, China
| | - Jin Cheng
- grid.8547.e0000 0001 0125 2443Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, 200032 Shanghai, China
| | - Ying Zhu
- grid.8547.e0000 0001 0125 2443Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, 200032 Shanghai, China
| | - Yan Jiang
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, 200032, Shanghai, China.
| |
Collapse
|
19
|
Wang Y, Xu L, Fang H, Wang F, Gao T, Zhu Q, Jiao G, Ke X. Social Brain Network of Children with Autism Spectrum Disorder: Characterization of Functional Connectivity and Potential Association with Stereotyped Behavior. Brain Sci 2023; 13:brainsci13020280. [PMID: 36831823 PMCID: PMC9953760 DOI: 10.3390/brainsci13020280] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 01/21/2023] [Accepted: 01/26/2023] [Indexed: 02/11/2023] Open
Abstract
Objective: To identify patterns of social dysfunction in adolescents with autism spectrum disorder (ASD), study the potential linkage between social brain networks and stereotyped behavior, and further explore potential targets of non-invasive nerve stimulation to improve social disorders. Methods: Voxel-wise and ROI-wise analysis methods were adopted to explore abnormalities in the functional activity of social-related regions of the brain. Then, we analyzed the relationships between clinical variables and the statistical indicators of social-related brain regions. Results: Compared with the typically developing group, the functional connectivity strength of social-related brain regions with the precentral gyrus, postcentral gyrus, supplementary motor area, paracentral lobule, median cingulum, and paracingulum gyri was significantly weakened in the ASD group (all p < 0. 01). The functional connectivity was negatively correlated with communication, social interaction, communication + social interaction, and the total score of the ADOS scale (r = -0.38, -0.39, -0.40, and -0.3, respectively; all p < 0.01), with social awareness, social cognition, social communication, social motivation, autistic mannerisms, and the total score of the SRS scale (r = -0.32, -0.32, -0.40, -0.30, -0.28, and -0.27, respectively; all p < 0.01), and with the total score of SCQ (r = -0.27, p < 0.01). In addition, significant intergroup differences in clustering coefficients and betweenness centrality were seen across multiple brain regions in the ASD group. Conclusions: The functional connectivity between social-related brain regions and many other brain regions was significantly weakened compared to the typically developing group, and it was negatively correlated with social disorders. Social network dysfunction seems to be related to stereotyped behavior. Therefore, these social-related brain regions may be taken as potential stimulation targets of non-invasive nerve stimulation to improve social dysfunction in children with ASD in the future.
Collapse
|
20
|
Boktor JC, Adame MD, Rose DR, Schumann CM, Murray KD, Bauman MD, Careaga M, Mazmanian SK, Ashwood P, Needham BD. Global metabolic profiles in a non-human primate model of maternal immune activation: implications for neurodevelopmental disorders. Mol Psychiatry 2022; 27:4959-4973. [PMID: 36028571 PMCID: PMC9772216 DOI: 10.1038/s41380-022-01752-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 08/02/2022] [Accepted: 08/12/2022] [Indexed: 01/14/2023]
Abstract
Epidemiological evidence implicates severe maternal infections as risk factors for neurodevelopmental disorders, such as ASD and schizophrenia. Accordingly, animal models mimicking infection during pregnancy, including the maternal immune activation (MIA) model, result in offspring with neurobiological, behavioral, and metabolic phenotypes relevant to human neurodevelopmental disorders. Most of these studies have been performed in rodents. We sought to better understand the molecular signatures characterizing the MIA model in an organism more closely related to humans, rhesus monkeys (Macaca mulatta), by evaluating changes in global metabolic profiles in MIA-exposed offspring. Herein, we present the global metabolome in six peripheral tissues (plasma, cerebrospinal fluid, three regions of intestinal mucosa scrapings, and feces) from 13 MIA and 10 control offspring that were confirmed to display atypical neurodevelopment, elevated immune profiles, and neuropathology. Differences in lipid, amino acid, and nucleotide metabolism discriminated these MIA and control samples, with correlations of specific metabolites to behavior scores as well as to cytokine levels in plasma, intestinal, and brain tissues. We also observed modest changes in fecal and intestinal microbial profiles, and identify differential metabolomic profiles within males and females. These findings support a connection between maternal immune activation and the metabolism, microbiota, and behavioral traits of offspring, and may further the translational applications of the MIA model and the advancement of biomarkers for neurodevelopmental disorders such as ASD or schizophrenia.
Collapse
Affiliation(s)
- Joseph C Boktor
- Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Mark D Adame
- Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Destanie R Rose
- Department of Medical Microbiology and Immunology, University of California Davis, Davis, CA, 95616, USA
- The M.I.N.D. Institute, University of California, Davis, Sacramento, CA, 95817, USA
| | - Cynthia M Schumann
- The M.I.N.D. Institute, University of California, Davis, Sacramento, CA, 95817, USA
| | - Karl D Murray
- The M.I.N.D. Institute, University of California, Davis, Sacramento, CA, 95817, USA
| | - Melissa D Bauman
- The M.I.N.D. Institute, University of California, Davis, Sacramento, CA, 95817, USA
| | - Milo Careaga
- Department of Medical Microbiology and Immunology, University of California Davis, Davis, CA, 95616, USA
- The M.I.N.D. Institute, University of California, Davis, Sacramento, CA, 95817, USA
| | - Sarkis K Mazmanian
- Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Paul Ashwood
- Department of Medical Microbiology and Immunology, University of California Davis, Davis, CA, 95616, USA.
- The M.I.N.D. Institute, University of California, Davis, Sacramento, CA, 95817, USA.
| | - Brittany D Needham
- Department of Anatomy, Cell Biology & Physiology, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| |
Collapse
|
21
|
Snihirova Y, Linden DEJ, van Amelsvoort T, van der Meer D. Environmental Influences on the Relation between the 22q11.2 Deletion Syndrome and Mental Health: A Literature Review. Genes (Basel) 2022; 13:2003. [PMID: 36360240 PMCID: PMC9690390 DOI: 10.3390/genes13112003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 10/21/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022] Open
Abstract
22q11.2 deletion syndrome (22q11DS) is a clinically heterogeneous genetic syndrome, associated with a wide array of neuropsychiatric symptoms. The clinical presentation is likely to be influenced by environmental factors, yet little is known about this. Here, we review the available research literature on the role of the environment in 22q11DS. We find that within-patient design studies have mainly investigated the role of parental factors, stress, and substance use, reporting significant effects of these factors on the clinical profile. Case-control studies have been less successful, with almost no reports of significant moderating effects of the environment. We go on to hypothesize which specific environmental measures are most likely to interact with the 22q11 deletion, based on the genes in this region and their involvement in molecular pathways. We end by discussing potential reasons for the limited findings so far, including modest sample sizes and limited availability of environmental measures, and make recommendations how to move forward.
Collapse
Affiliation(s)
- Yelyzaveta Snihirova
- School for Mental Health and Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6211 LK Maastricht, The Netherlands
| | - David E. J. Linden
- School for Mental Health and Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6211 LK Maastricht, The Netherlands
| | - Therese van Amelsvoort
- School for Mental Health and Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6211 LK Maastricht, The Netherlands
| | - Dennis van der Meer
- School for Mental Health and Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6211 LK Maastricht, The Netherlands
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, 0315 Oslo, Norway
| |
Collapse
|
22
|
Griego E, Segura-Villalobos D, Lamas M, Galván EJ. Maternal immune activation increases excitability via downregulation of A-type potassium channels and reduces dendritic complexity of hippocampal neurons of the offspring. Brain Behav Immun 2022; 105:67-81. [PMID: 35803480 DOI: 10.1016/j.bbi.2022.07.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 06/29/2022] [Accepted: 07/03/2022] [Indexed: 11/29/2022] Open
Abstract
The epidemiological association between bacterial or viral maternal infections during pregnancy and increased risk for developing psychiatric disorders in offspring is well documented. Numerous rodent and non-human primate studies of viral- or, to a lesser extent, bacterial-induced maternal immune activation (MIA) have documented a series of neurological alterations that may contribute to understanding the pathophysiology of schizophrenia and autism spectrum disorders. Long-term neuronal and behavioral alterations are now ascribed to the effect of maternal proinflammatory cytokines rather than the infection itself. However, detailed electrophysiological alterations in brain areas relevant to psychiatric disorders, such as the dorsal hippocampus, are lacking in response to bacterial-induced MIA. This study determined if electrophysiological and morphological alterations converge in CA1 pyramidal cells (CA1 PC) from the dorsal hippocampus in bacterial-induced MIA offspring. A series of changes in the functional expression of K+ and Na+ ion channels altered the passive and active membrane properties and triggered hyperexcitability of CA1 PC. Contributing to the hyperexcitability, the somatic A-type potassium current (IA) was decreased in MIA CA1 PC. Likewise, the spontaneous glutamatergic and GABAergic inputs were dysregulated and biased toward increased excitation, thereby reshaping the excitation-inhibition balance. Consistent with these findings, the dendritic branching complexity of MIA CA1 PC was reduced. Together, these morphophysiological alterations modify CA1 PC computational capabilities and contribute to explaining cellular alterations that may underlie the cognitive symptoms of MIA-associated psychiatric disorders.
Collapse
Affiliation(s)
- Ernesto Griego
- Departamento de Farmacobiología, CINVESTAV Unidad Sur, Ciudad de México, Mexico
| | | | - Mónica Lamas
- Departamento de Farmacobiología, CINVESTAV Unidad Sur, Ciudad de México, Mexico
| | - Emilio J Galván
- Departamento de Farmacobiología, CINVESTAV Unidad Sur, Ciudad de México, Mexico.
| |
Collapse
|
23
|
Hanson KL, Grant SE, Funk LH, Schumann CM, Bauman MD. Impact of Maternal Immune Activation on Nonhuman Primate Prefrontal Cortex Development: Insights for Schizophrenia. Biol Psychiatry 2022; 92:460-469. [PMID: 35773097 PMCID: PMC9888668 DOI: 10.1016/j.biopsych.2022.04.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 03/30/2022] [Accepted: 04/13/2022] [Indexed: 02/02/2023]
Abstract
Late adolescence is a period of dynamic change in the brain as humans learn to navigate increasingly complex environments. In particular, prefrontal cortical (PFC) regions undergo extensive remodeling as the brain is fine-tuned to orchestrate cognitive control over attention, reasoning, and emotions. Late adolescence also presents a uniquely vulnerable period as neurodevelopmental illnesses, such as schizophrenia, become evident and worsen into young adulthood. Challenges in early development, including prenatal exposure to infection, may set the stage for a cascade of maladaptive events that ultimately result in aberrant PFC connectivity and function before symptoms emerge. A growing body of research suggests that activation of the mother's immune system during pregnancy may act as a disease primer, in combination with other environmental and genetic factors, contributing to an increased risk of neurodevelopmental disorders, including schizophrenia. Animal models provide an invaluable opportunity to examine the course of brain and behavioral changes in offspring exposed to maternal immune activation (MIA). Although the vast majority of MIA research has been carried out in rodents, here we highlight the translational utility of the nonhuman primate (NHP) as a model species more closely related to humans in PFC structure and function. In this review, we consider the protracted period of brain and behavioral maturation in the NHP, describe emerging findings from MIA NHP offspring in the context of rodent preclinical models, and lastly explore the translational relevance of the NHP MIA model to expand understanding of the etiology and developmental course of PFC pathology in schizophrenia.
Collapse
Affiliation(s)
- Kari L Hanson
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, Davis, California; MIND Institute, University of California, Davis, Davis, California
| | - Simone E Grant
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, Davis, California
| | - Lucy H Funk
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, Davis, California
| | - Cynthia M Schumann
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, Davis, California; MIND Institute, University of California, Davis, Davis, California.
| | - Melissa D Bauman
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, Davis, California; MIND Institute, University of California, Davis, Davis, California; California National Primate Research Center, University of California, Davis, Davis, California.
| |
Collapse
|
24
|
Garcia-Partida JA, Torres-Sanchez S, MacDowell K, Fernández-Ponce MT, Casas L, Mantell C, Soto-Montenegro ML, Romero-Miguel D, Lamanna-Rama N, Leza JC, Desco M, Berrocoso E. The effects of mango leaf extract during adolescence and adulthood in a rat model of schizophrenia. Front Pharmacol 2022; 13:886514. [PMID: 35959428 PMCID: PMC9360613 DOI: 10.3389/fphar.2022.886514] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 06/30/2022] [Indexed: 11/13/2022] Open
Abstract
There is evidence that in schizophrenia, imbalances in inflammatory and oxidative processes occur during pregnancy and in the early postnatal period, generating interest in the potential therapeutic efficacy of anti-inflammatory and antioxidant compounds. Mangiferin is a polyphenolic compound abundant in the leaves of Mangifera indica L. that has robust antioxidant and anti-inflammatory properties, making it a potential candidate for preventive or co-adjuvant therapy in schizophrenia. Hence, this study set-out to evaluate the effect of mango leaf extract (MLE) in a model of schizophrenia based on maternal immune activation, in which Poly I:C (4 mg/kg) is administered intravenously to pregnant rats. Young adult (postnatal day 60-70) or adolescent (postnatal day 35-49) male offspring received MLE (50 mg/kg of mangiferin) daily, and the effects of MLE in adolescence were compared to those of risperidone, assessing behavior, brain magnetic resonance imaging (MRI), and oxidative/inflammatory and antioxidant mediators in the adult offspring. MLE treatment in adulthood reversed the deficit in prepulse inhibition (PPI) but it failed to attenuate the sensitivity to amphetamine and the deficit in novel object recognition (NOR) induced. By contrast, adolescent MLE treatment prevented the sensorimotor gating deficit in the PPI test, producing an effect similar to that of risperidone. This MLE treatment also produced a reduction in grooming behavior, but it had no effect on anxiety or novel object recognition memory. MRI studies revealed that adolescent MLE administration partially counteracted the cortical shrinkage, and cerebellum and ventricle enlargement. In addition, MLE administration in adolescence reduced iNOS mediated inflammatory activation and it promoted the expression of biomarkers of compensatory antioxidant activity in the prefrontal cortex and hippocampus, as witnessed through the reduction of Keap1 and the accumulation of NRF2 and HO1. Together, these findings suggest that MLE might be an alternative therapeutic or preventive add-on strategy to improve the clinical expression of schizophrenia in adulthood, while also modifying the time course of this disease at earlier stages in populations at high-risk.
Collapse
Affiliation(s)
- Jose Antonio Garcia-Partida
- Neuropsychopharmacology and Psychobiology Research Group, Department of Neuroscience, University of Cádiz, Cádiz, Spain
- Instituto de Investigación e Innovación en Ciencias Biomédicas de Cádiz, INiBICA, Hospital Universitario Puerta del Mar, Cádiz, Spain
| | - Sonia Torres-Sanchez
- Instituto de Investigación e Innovación en Ciencias Biomédicas de Cádiz, INiBICA, Hospital Universitario Puerta del Mar, Cádiz, Spain
- Neuropsychopharmacology and Psychobiology Research Group, Psychobiology Area, Department of Psychology, University of Cádiz, Cádiz, Spain
- Ciber of Mental Health (CIBERSAM), ISCIII, Madrid, Spain
| | - Karina MacDowell
- Ciber of Mental Health (CIBERSAM), ISCIII, Madrid, Spain
- Department of Pharmacology and Toxicology, Faculty of Medicine, Universidad Complutense de Madrid (UCM), Health Research Institute Hospital 12 de Octubre (imas12), Institute of Research in Neurochemistry IUIN-UCM, Madrid, Spain
| | | | - Lourdes Casas
- Department of Chemical Engineering and Food Technology, Science Faculty, University of Cádiz, Cádiz, Spain
| | - Casimiro Mantell
- Department of Chemical Engineering and Food Technology, Science Faculty, University of Cádiz, Cádiz, Spain
| | - María Luisa Soto-Montenegro
- Ciber of Mental Health (CIBERSAM), ISCIII, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- High Performance Research Group in Physiopathology and Pharmacology of the Digestive System (NeuGut), Universidad Rey Juan Carlos, Madrid, Spain
| | - Diego Romero-Miguel
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Departamento de Bioingeniería e Ingeniería Aeroespacial, Universidad Carlos III de Madrid, Madrid, Spain
| | - Nicolás Lamanna-Rama
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Departamento de Bioingeniería e Ingeniería Aeroespacial, Universidad Carlos III de Madrid, Madrid, Spain
| | - Juan Carlos Leza
- Ciber of Mental Health (CIBERSAM), ISCIII, Madrid, Spain
- Department of Pharmacology and Toxicology, Faculty of Medicine, Universidad Complutense de Madrid (UCM), Health Research Institute Hospital 12 de Octubre (imas12), Institute of Research in Neurochemistry IUIN-UCM, Madrid, Spain
| | - Manuel Desco
- Ciber of Mental Health (CIBERSAM), ISCIII, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Departamento de Bioingeniería e Ingeniería Aeroespacial, Universidad Carlos III de Madrid, Madrid, Spain
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Esther Berrocoso
- Instituto de Investigación e Innovación en Ciencias Biomédicas de Cádiz, INiBICA, Hospital Universitario Puerta del Mar, Cádiz, Spain
- Neuropsychopharmacology and Psychobiology Research Group, Psychobiology Area, Department of Psychology, University of Cádiz, Cádiz, Spain
- Ciber of Mental Health (CIBERSAM), ISCIII, Madrid, Spain
| |
Collapse
|
25
|
Eng ME, Imperio GE, Bloise E, Matthews SG. ATP-binding cassette (ABC) drug transporters in the developing blood-brain barrier: role in fetal brain protection. Cell Mol Life Sci 2022; 79:415. [PMID: 35821142 PMCID: PMC11071850 DOI: 10.1007/s00018-022-04432-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 05/27/2022] [Accepted: 06/15/2022] [Indexed: 12/19/2022]
Abstract
The blood-brain barrier (BBB) provides essential neuroprotection from environmental toxins and xenobiotics, through high expression of drug efflux transporters in endothelial cells of the cerebral capillaries. However, xenobiotic exposure, stress, and inflammatory stimuli have the potential to disrupt BBB permeability in fetal and post-natal life. Understanding the role and ability of the BBB in protecting the developing brain, particularly with respect to drug/toxin transport, is key to promoting long-term brain health. Drug transporters, particularly P-gp and BCRP are expressed in early gestation at the developing BBB and have a crucial role in developmental homeostasis and fetal brain protection. We have highlighted several factors that modulate drug transporters at the developing BBB, including synthetic glucocorticoid (sGC), cytokines, maternal infection, and growth factors. Some factors have the potential to increase expression and function of drug transporters and increase brain protection (e.g., sGC, transforming growth factor [TGF]-β). However, others inhibit drug transporters expression and function at the BBB, increasing brain exposure to xenobiotics (e.g., tumor necrosis factor [TNF], interleukin [IL]-6), negatively impacting brain development. This has implications for pregnant women and neonates, who represent a vulnerable population and may be exposed to drugs and environmental toxins, many of which are P-gp and BCRP substrates. Thus, alterations in regulated transport across the developing BBB may induce long-term changes in brain health and compromise pregnancy outcome. Furthermore, a large portion of neonatal adverse drug reactions are attributed to agents that target or access the nervous system, such as stimulants (e.g., caffeine), anesthetics (e.g., midazolam), analgesics (e.g., morphine) and antiretrovirals (e.g., Zidovudine); thus, understanding brain protection is key for the development of strategies to protect the fetal and neonatal brain.
Collapse
Affiliation(s)
- Margaret E Eng
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Medical Sciences Bldg. Rm. 3207. 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
| | | | - Enrrico Bloise
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Medical Sciences Bldg. Rm. 3207. 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Canada
- Department of Morphology, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Stephen G Matthews
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Medical Sciences Bldg. Rm. 3207. 1 King's College Circle, Toronto, ON, M5S 1A8, Canada.
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Canada.
- Department of Obstetrics and Gynecology, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada.
- Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada.
| |
Collapse
|
26
|
Desbonnet L, Konkoth A, Laighneach A, McKernan D, Holleran L, McDonald C, Morris DW, Donohoe G, Kelly J. Dual hit mouse model to examine the long-term effects of maternal immune activation and post-weaning social isolation on schizophrenia endophenotypes. Behav Brain Res 2022; 430:113930. [DOI: 10.1016/j.bbr.2022.113930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 04/27/2022] [Accepted: 05/14/2022] [Indexed: 11/02/2022]
|
27
|
Nakamura JP, Schroeder A, Gibbons A, Sundram S, Hill RA. Timing of maternal immune activation and sex influence schizophrenia-relevant cognitive constructs and neuregulin and GABAergic pathways. Brain Behav Immun 2022; 100:70-82. [PMID: 34808289 DOI: 10.1016/j.bbi.2021.11.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 10/17/2021] [Accepted: 11/13/2021] [Indexed: 12/24/2022] Open
Abstract
Maternal immune activation (MIA) during pregnancy is an established environmental risk factor for schizophrenia. Timing of immune activation exposure as well as sex of the exposed offspring are critical factors in defining the effects of MIA. However, the specificity of MIA on the component structure of schizophrenia, especially cognition, has been difficult to assess due to a lack of translational validity of maze-like testing paradigms. We aimed to assess cognitive domains relevant to schizophrenia using highly translational touchscreen-based tasks in male and female mice exposed to the viral mimetic, poly(I:C) (5 mg/k, i.p.), during early (gestational day (GD) 9-11) and late (GD13-15) gestational time points. Gene expression of schizophrenia candidate pathways were assessed in fetal brain immediately following poly(I:C) exposure and in adulthood to identify its influence on neurodevelopmental processes. Sex and window specific alterations in cognitive performance were found with the early window of MIA exposure causing female-specific disruptions to working memory and reduced perseverative behaviour, while late MIA exposure caused male-specific changes to working memory and deficits in reversal learning. GABAergic specification marker, Nkx2.1 gene expression was reduced in fetal brains and reelin expression was reduced in adult hippocampus of both early and late poly(I:C) exposed mice. Neuregulin and EGF signalling were initially upregulated in the fetal brain, but were reduced in the adult hippocampus, with male mice exposed in the late window showing reduced Nrg3 expression. Serine racemase was reduced in both fetal and adult brain, but again, adult reductions were specific to male mice exposed at the late time point. Overall, we show that cognitive constructs relevant to schizophrenia are altered by in utero exposure to maternal immune activation, but are highly dependent on the timing of infection and the sex of the offspring. Glutamatergic and epidermal growth factor pathways were similarly altered by MIA in a timing and sex dependent manner, while MIA-induced GABAergic deficits were independent of timing or sex.
Collapse
Affiliation(s)
- J P Nakamura
- Department of Psychiatry, School of Clinical Sciences, Monash University, Clayton, VIC 3168, Australia
| | - A Schroeder
- Department of Psychiatry, School of Clinical Sciences, Monash University, Clayton, VIC 3168, Australia
| | - A Gibbons
- Department of Psychiatry, School of Clinical Sciences, Monash University, Clayton, VIC 3168, Australia
| | - S Sundram
- Department of Psychiatry, School of Clinical Sciences, Monash University, Clayton, VIC 3168, Australia; Mental Health Program, Monash Health, Clayton, VIC 3168, Australia
| | - R A Hill
- Department of Psychiatry, School of Clinical Sciences, Monash University, Clayton, VIC 3168, Australia.
| |
Collapse
|
28
|
Sato A, Kotajima-Murakami H, Tanaka M, Katoh Y, Ikeda K. Influence of Prenatal Drug Exposure, Maternal Inflammation, and Parental Aging on the Development of Autism Spectrum Disorder. Front Psychiatry 2022; 13:821455. [PMID: 35222122 PMCID: PMC8863673 DOI: 10.3389/fpsyt.2022.821455] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 01/12/2022] [Indexed: 12/17/2022] Open
Abstract
Autism spectrum disorder (ASD) affects reciprocal social interaction and produces abnormal repetitive, restrictive behaviors and interests. The diverse causes of ASD are divided into genetic alterations and environmental risks. The prevalence of ASD has been rising for several decades, which might be related to environmental risks as it is difficult to consider that the prevalence of genetic disorders related to ASD would increase suddenly. The latter includes (1) exposure to medications, such as valproic acid (VPA) and selective serotonin reuptake inhibitors (SSRIs) (2), maternal complications during pregnancy, including infection and hypertensive disorders of pregnancy, and (3) high parental age. Epidemiological studies have indicated a pathogenetic role of prenatal exposure to VPA and maternal inflammation in the development of ASD. VPA is considered to exert its deleterious effects on the fetal brain through several distinct mechanisms, such as alterations of γ-aminobutyric acid signaling, the inhibition of histone deacetylase, the disruption of folic acid metabolism, and the activation of mammalian target of rapamycin. Maternal inflammation that is caused by different stimuli converges on a higher load of proinflammatory cytokines in the fetal brain. Rodent models of maternal exposure to SSRIs generate ASD-like behavior in offspring, but clinical correlations with these preclinical findings are inconclusive. Hypertensive disorders of pregnancy and advanced parental age increase the risk of ASD in humans, but the mechanisms have been poorly investigated in animal models. Evidence of the mechanisms by which environmental factors are related to ASD is discussed, which may contribute to the development of preventive and therapeutic interventions for ASD.
Collapse
Affiliation(s)
- Atsushi Sato
- Department of Pediatrics, The University of Tokyo Hospital, Tokyo, Japan.,Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | | | - Miho Tanaka
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.,Department of Psychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yoshihisa Katoh
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.,Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kazutaka Ikeda
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| |
Collapse
|
29
|
Bao M, Hofsink N, Plösch T. LPS vs. Poly I:C Model: Comparison of Long-Term Effects of Bacterial and Viral Maternal Immune Activation (MIA) on the Offspring. Am J Physiol Regul Integr Comp Physiol 2021; 322:R99-R111. [PMID: 34874190 PMCID: PMC8782664 DOI: 10.1152/ajpregu.00087.2021] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
A prominent health issue nowadays is the COVID-19 pandemic, which poses acute risks to human health. However, the long-term health consequences are largely unknown and cannot be neglected. An especially vulnerable period for infection is pregnancy, when infections could have long-term health effect on the child. Evidence suggests that maternal immune activation (MIA) induced by either bacteria or viruses presents various effects on the offspring, leading to adverse phenotypes in many organ systems. This review compares the mechanisms of bacterial and viral MIA and the possible long-term outcomes for the offspring by summarizing the outcome in animal LPS and Poly I:C models. Both models are activated immune responses mediated by Toll-like receptors. The outcomes for MIA offspring include neurodevelopment, immune response, circulation, metabolism, and reproduction. Some of these changes continue to exist until later life. Besides different doses and batches of LPS and Poly I:C, the injection day, administration route, and also different animal species influence the outcomes. Here, we specifically aim to support colleagues when choosing their animal models for future studies.
Collapse
Affiliation(s)
- Mian Bao
- Department of Obstetrics and Gynaecology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Naomi Hofsink
- Department of Obstetrics and Gynaecology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Torsten Plösch
- Department of Obstetrics and Gynaecology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.,Perinatal Neurobiology, Department of Human Medicine, School of Medicine and Health Sciences Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| |
Collapse
|
30
|
Su Y, Lian J, Hodgson J, Zhang W, Deng C. Prenatal Poly I:C Challenge Affects Behaviors and Neurotransmission via Elevated Neuroinflammation Responses in Female Juvenile Rats. Int J Neuropsychopharmacol 2021; 25:160-171. [PMID: 34893855 PMCID: PMC8832231 DOI: 10.1093/ijnp/pyab087] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 11/12/2021] [Accepted: 12/04/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Exposure to polyriboinosinic-polyribocytidylic acid (Poly I:C) in pregnant rats has been reported to cause schizophrenia-like behaviors and abnormal neurotransmissions in adult, particularly male, offspring. However, what is less well understood are the effects of maternal Poly I:C exposure on adolescent behaviors and neurotransmission in female juvenile rats. METHODS Female adolescent Poly I:C offspring were constructed by treating with 5 mg/kg Poly I:C on timed pregnant rats (gestation day 15). A battery of behavioral tests was conducted during postnatal day 35-60. Neurotransmitter receptors and inflammation markers in brain regions were evaluated by RT-qPCR on postnatal day 60. RESULTS Open field, elevated plus maze, and forced swimming tests revealed that prenatal Poly I:C exposure led to elevated anxiety-like and depression-like behaviors in female adolescent offspring. Deficits in pre-pulse inhibition and social interaction were also observed. However, the Poly I:C rats had better performance than the controls in the novel object recognition memory test, which demonstrated a behavioral phenotype with improved cognitive function. Prenatal Poly I:C exposure caused brain region-specific elevation of the P2X7 receptor- and NF-κB-NLRP3-IL-1β inflammatory signaling in female juvenile rats. Prenatal Poly I:C exposure decreased expression of GABAA receptor subunits Gabrb3 in the prefrontal cortex and Gabrb1 and dopamine D2 receptor in the hippocampus, but increased NMDA receptor subunit Grin2a in the prefrontal cortex, 5-HT2A in the hippocampus, and Gabrb3 and D2 receptor in the nucleus accumben. CONCLUSIONS Prenatal Poly I:C challenge causes behavioral deficits and brain-specific neurotransmission changes via elevated neuroinflammation responses in female adolescent offspring rats.
Collapse
Affiliation(s)
- Yueqing Su
- The School of Public Health, Fujian Medical University, Fuzhou, China,Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China,Antipsychotic Research Laboratory, Illawarra Health and Medical Research Institute, Wollongong, NSW, Australia,School of Medicine, and Molecular Horizons, University of Wollongong, Wollongong, NSW, Australia
| | - Jiamei Lian
- School of Medicine, and Molecular Horizons, University of Wollongong, Wollongong, NSW, Australia
| | - James Hodgson
- Antipsychotic Research Laboratory, Illawarra Health and Medical Research Institute, Wollongong, NSW, Australia,School of Medicine, and Molecular Horizons, University of Wollongong, Wollongong, NSW, Australia
| | - Wenchang Zhang
- The School of Public Health, Fujian Medical University, Fuzhou, China
| | - Chao Deng
- Antipsychotic Research Laboratory, Illawarra Health and Medical Research Institute, Wollongong, NSW, Australia,School of Medicine, and Molecular Horizons, University of Wollongong, Wollongong, NSW, Australia,Correspondence: Chao Deng, PhD, Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, 2522, Australia ()
| |
Collapse
|
31
|
Disruption of Alternative Splicing in the Amygdala of Pigs Exposed to Maternal Immune Activation. IMMUNO 2021. [DOI: 10.3390/immuno1040035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The inflammatory response of gestating females to infection or stress can disrupt gene expression in the offspring’s amygdala, resulting in lasting neurodevelopmental, physiological, and behavioral disorders. The effects of maternal immune activation (MIA) can be impacted by the offspring’s sex and exposure to additional stressors later in life. The objectives of this study were to investigate the disruption of alternative splicing patterns associated with MIA in the offspring’s amygdala and characterize this disruption in the context of the second stress of weaning and sex. Differential alternative splicing was tested on the RNA-seq profiles of a pig model of viral-induced MIA. Compared to controls, MIA was associated with the differential alternative splicing (FDR-adjusted p-value < 0.1) of 292 and 240 genes in weaned females and males, respectively, whereas 132 and 176 genes were differentially spliced in control nursed female and male, respectively. The majority of the differentially spliced (FDR-adjusted p-value < 0.001) genes (e.g., SHANK1, ZNF672, KCNA6) and many associated enriched pathways (e.g., Fc gamma R-mediated phagocytosis, non-alcoholic fatty liver disease, and cGMP-PKG signaling) have been reported in MIA-related disorders including autism and schizophrenia in humans. Differential alternative splicing associated with MIA was detected in the gene MAG across all sex-stress groups except for unstressed males and SLC2A11 across all groups except unstressed females. Precise understanding of the effect of MIA across second stressors and sexes necessitates the consideration of splicing isoform profiles.
Collapse
|
32
|
Serra I, Manusama OR, Kaiser FMP, Floriano II, Wahl L, van der Zalm C, IJspeert H, van Hagen PM, van Beveren NJM, Arend SM, Okkenhaug K, Pel JJM, Dalm VASH, Badura A. Activated PI3Kδ syndrome, an immunodeficiency disorder, leads to sensorimotor deficits recapitulated in a murine model. Brain Behav Immun Health 2021; 18:100377. [PMID: 34786564 PMCID: PMC8579111 DOI: 10.1016/j.bbih.2021.100377] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 09/24/2021] [Accepted: 10/18/2021] [Indexed: 02/08/2023] Open
Abstract
The phosphoinositide-3-kinase (PI3K) family plays a major role in cell signaling and is predominant in leukocytes. Gain-of-function (GOF) mutations in the PIK3CD gene lead to the development of activated PI3Kδ syndrome (APDS), a rare primary immunodeficiency disorder. A subset of APDS patients also displays neurodevelopmental delay symptoms, suggesting a potential role of PIK3CD in cognitive and behavioural function. However, the extent and nature of the neurodevelopmental deficits has not been previously quantified. Here, we assessed the cognitive functions of two APDS patients, and investigated the causal role of the PIK3CD GOF mutation in neurological deficits using a murine model of this disease. We used p110δE1020K knock-in mice, harbouring the most common APDS mutation in patients. We found that APDS patients present with visuomotor deficits, exacerbated by autism spectrum disorder comorbidity, whereas p110δE1020K mice exhibited impairments in motor behaviour, learning and repetitive behaviour patterning. Our data indicate that PIK3CD GOF mutations increase the risk for neurodevelopmental deficits, supporting previous findings on the interplay between the nervous and the immune system. Further, our results validate the knock-in mouse model, and offer an objective assessment tool for patients that could be incorporated in diagnosis and in the evaluation of treatments.
Collapse
Affiliation(s)
- Ines Serra
- Department of Neuroscience, Erasmus MC, Rotterdam, the Netherlands
| | | | - Fabian M P Kaiser
- Department of Immunology, Erasmus MC, Rotterdam, the Netherlands.,Department of Pediatrics, Erasmus MC, Rotterdam, the Netherlands
| | | | - Lucas Wahl
- Department of Neuroscience, Erasmus MC, Rotterdam, the Netherlands
| | | | - Hanna IJspeert
- Department of Immunology, Erasmus MC, Rotterdam, the Netherlands
| | - P Martin van Hagen
- Department of Immunology, Erasmus MC, Rotterdam, the Netherlands.,Division of Clinical Immunology, Department of Internal Medicine, Erasmus MC, Rotterdam, the Netherlands
| | | | - Sandra M Arend
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, the Netherlands
| | - Klaus Okkenhaug
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Johan J M Pel
- Department of Neuroscience, Erasmus MC, Rotterdam, the Netherlands
| | - Virgil A S H Dalm
- Department of Immunology, Erasmus MC, Rotterdam, the Netherlands.,Division of Clinical Immunology, Department of Internal Medicine, Erasmus MC, Rotterdam, the Netherlands.,Academic Center for Rare Immunological Diseases (RIDC), Erasmus MC, Rotterdam, the Netherlands
| | | |
Collapse
|
33
|
Factors Regulating the Activity of LINE1 Retrotransposons. Genes (Basel) 2021; 12:genes12101562. [PMID: 34680956 PMCID: PMC8535693 DOI: 10.3390/genes12101562] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/21/2021] [Accepted: 09/22/2021] [Indexed: 12/15/2022] Open
Abstract
LINE-1 (L1) is a class of autonomous mobile genetic elements that form somatic mosaicisms in various tissues of the organism. The activity of L1 retrotransposons is strictly controlled by many factors in somatic and germ cells at all stages of ontogenesis. Alteration of L1 activity was noted in a number of diseases: in neuropsychiatric and autoimmune diseases, as well as in various forms of cancer. Altered activity of L1 retrotransposons for some pathologies is associated with epigenetic changes and defects in the genes involved in their repression. This review discusses the molecular genetic mechanisms of the retrotransposition and regulation of the activity of L1 elements. The contribution of various factors controlling the expression and distribution of L1 elements in the genome occurs at all stages of the retrotransposition. The regulation of L1 elements at the transcriptional, post-transcriptional and integration into the genome stages is described in detail. Finally, this review also focuses on the evolutionary aspects of L1 accumulation and their interplay with the host regulation system.
Collapse
|
34
|
Laighneach A, Desbonnet L, Kelly JP, Donohoe G, Morris DW. Meta-Analysis of Brain Gene Expression Data from Mouse Model Studies of Maternal Immune Activation Using Poly(I:C). Genes (Basel) 2021; 12:genes12091363. [PMID: 34573345 PMCID: PMC8471627 DOI: 10.3390/genes12091363] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 08/20/2021] [Accepted: 08/30/2021] [Indexed: 02/07/2023] Open
Abstract
Maternal immune activation (MIA) is a known risk factor for schizophrenia (SCZ) and autism spectrum disorder (ASD) and is often modelled in animal studies in order to study the effect of prenatal infection on brain function including behaviour and gene expression. Although the effect of MIA on gene expression are highly heterogeneous, combining data from multiple gene expression studies in a robust method may shed light on the true underlying biological effects caused by MIA and this could inform studies of SCZ and ASD. This study combined four RNA-seq and microarray datasets in an overlap analysis and ranked meta-analysis in order to investigate genes, pathways and cell types dysregulated in the MIA mouse models. Genes linked to SCZ and ASD and crucial in neurodevelopmental processes including neural tube folding, regulation of cellular stress and neuronal/glial cell differentiation were among the most consistently dysregulated in these ranked analyses. Gene ontologies including K+ ion channel function, neuron and glial cell differentiation, synaptic structure, axonal outgrowth, cilia function and lipid metabolism were also strongly implicated. Single-cell analysis identified excitatory and inhibitory cell types in the cortex, hippocampus and striatum that may be affected by MIA and are also enriched for genes associated with SCZ, ASD and cognitive phenotypes. This points to the cellular location of molecular mechanisms that may be consistent between the MIA model and neurodevelopmental disease, improving our understanding of its utility to study prenatal infection as an environmental stressor.
Collapse
Affiliation(s)
- Aodán Laighneach
- Centre for Neuroimaging, Cognition and Genomics, Discipline of Biochemistry and School of Psychology, National University of Ireland Galway, H91 TK33 Galway, Ireland; (A.L.); (G.D.)
| | - Lieve Desbonnet
- Discipline of Pharmacology and Therapeutics, National University of Ireland Galway, H91 TK33 Galway, Ireland; (L.D.); (J.P.K.)
| | - John P. Kelly
- Discipline of Pharmacology and Therapeutics, National University of Ireland Galway, H91 TK33 Galway, Ireland; (L.D.); (J.P.K.)
| | - Gary Donohoe
- Centre for Neuroimaging, Cognition and Genomics, Discipline of Biochemistry and School of Psychology, National University of Ireland Galway, H91 TK33 Galway, Ireland; (A.L.); (G.D.)
| | - Derek W. Morris
- Centre for Neuroimaging, Cognition and Genomics, Discipline of Biochemistry and School of Psychology, National University of Ireland Galway, H91 TK33 Galway, Ireland; (A.L.); (G.D.)
- Correspondence:
| |
Collapse
|
35
|
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: 8.8] [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
|
36
|
Zhao X, Mohammed R, Tran H, Erickson M, Kentner AC. Poly (I:C)-induced maternal immune activation modifies ventral hippocampal regulation of stress reactivity: prevention by environmental enrichment. Brain Behav Immun 2021; 95:203-215. [PMID: 33766701 PMCID: PMC8187276 DOI: 10.1016/j.bbi.2021.03.018] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 02/28/2021] [Accepted: 03/17/2021] [Indexed: 12/14/2022] Open
Abstract
Environmental enrichment (EE) has been successfully implemented in human rehabilitation settings. However, the mechanisms underlying its success are not understood. Incorporating components of EE protocols into our animal models allows for the exploration of these mechanisms and their role in mitigation. Using a mouse model of maternal immune activation (MIA), the present study explored disruptions in social behavior and associated hypothalamic pituitary adrenal (HPA) axis functioning, and whether a supportive environment could prevent these effects. We show that prenatal immune activation of toll-like receptor 3, by the viral mimetic polyinosinic-polycytidylic acid (poly(I:C)), led to disrupted maternal care in that dams built poorer quality nests, an effect corrected by EE housing. Standard housed male and female MIA mice engaged in higher rates of repetitive rearing and had lower levels of social interaction, alongside sex-specific expression of several ventral hippocampal neural stress markers. Moreover, MIA males had delayed recovery of plasma corticosterone in response to a novel social encounter. Enrichment housing, likely mediated by improved maternal care, protected against these MIA-induced effects. We also evaluated c-Fos immunoreactivity associated with the novel social experience and found MIA to decrease neural activation in the dentate gyrus. Activation in the hypothalamus was blunted in EE housed animals, suggesting that the putative circuits modulating social behaviors may be different between standard and complex housing environments. These data demonstrate that augmentation of the environment supports parental care and offspring safety/security, which can offset effects of early health adversity by buffering HPA axis dysregulation. Our findings provide further evidence for the viability of EE interventions in maternal and pediatric settings.
Collapse
Affiliation(s)
| | | | | | | | - Amanda C. Kentner
- Corresponding author: Amanda Kentner, , Office #617-274-3360, Fax # 617-732-2959
| |
Collapse
|
37
|
Barcik W, Chiacchierini G, Bimpisidis Z, Papaleo F. Immunology and microbiology: how do they affect social cognition and emotion recognition? Curr Opin Immunol 2021; 71:46-54. [PMID: 34058687 DOI: 10.1016/j.coi.2021.05.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 05/01/2021] [Indexed: 12/25/2022]
Abstract
Social interactions profoundly influence animals' life. The quality of social interactions and many everyday life decisions are determined by a proper perception, processing and reaction to others' emotions. Notably, alterations in these social processes characterize a number of neurodevelopmental disorders, including autism spectrum disorders and schizophrenia. Increasing evidences support an implication of immune system vulnerability and inflammatory processes in disparate behavioral functions and the aforementioned neurodevelopmental disorders. In this review, we show a possible unifying view on how immune responses, within and outside the brain, and the communication between the immune system and brain responses might influence emotion recognition and related social responses. In particular, we highlight the importance of combining genetics, immunology and microbiology factors in understanding social behaviors. We underline the importance of better disentangling the whole machinery between brain-immune system interactions to better address the complexity of social processes.
Collapse
Affiliation(s)
- Weronika Barcik
- Genetics of Cognition Laboratory, Neuroscience Area, Istituto Italiano di Tecnologia, Genova, Italy
| | - Giulia Chiacchierini
- Genetics of Cognition Laboratory, Neuroscience Area, Istituto Italiano di Tecnologia, Genova, Italy
| | - Zisis Bimpisidis
- Genetics of Cognition Laboratory, Neuroscience Area, Istituto Italiano di Tecnologia, Genova, Italy
| | - Francesco Papaleo
- Genetics of Cognition Laboratory, Neuroscience Area, Istituto Italiano di Tecnologia, Genova, Italy; Fondazione IRCCS Ca'Granda Ospedale Maggiore Policlinico, Milano, Italy.
| |
Collapse
|
38
|
Page NF, Gandal MJ, Estes ML, Cameron S, Buth J, Parhami S, Ramaswami G, Murray K, Amaral DG, Van de Water JA, Schumann CM, Carter CS, Bauman MD, McAllister AK, Geschwind DH. Alterations in Retrotransposition, Synaptic Connectivity, and Myelination Implicated by Transcriptomic Changes Following Maternal Immune Activation in Nonhuman Primates. Biol Psychiatry 2021; 89:896-910. [PMID: 33386132 PMCID: PMC8052273 DOI: 10.1016/j.biopsych.2020.10.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 10/07/2020] [Accepted: 10/09/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND Maternal immune activation (MIA) is a proposed risk factor for multiple neuropsychiatric disorders, including schizophrenia. However, the molecular mechanisms through which MIA imparts risk remain poorly understood. A recently developed nonhuman primate model of exposure to the viral mimic poly:ICLC during pregnancy shows abnormal social and repetitive behaviors and elevated striatal dopamine, a molecular hallmark of human psychosis, providing an unprecedented opportunity for studying underlying molecular correlates. METHODS We performed RNA sequencing across psychiatrically relevant brain regions (prefrontal cortex, anterior cingulate, hippocampus) and primary visual cortex for comparison from 3.5- to 4-year-old male MIA-exposed and control offspring-an age comparable to mid adolescence in humans. RESULTS We identify 266 unique genes differentially expressed in at least one brain region, with the greatest number observed in hippocampus. Co-expression networks identified region-specific alterations in synaptic signaling and oligodendrocytes. Although we observed temporal and regional differences, transcriptomic changes were shared across first- and second-trimester exposures, including for the top differentially expressed genes-PIWIL2 and MGARP. In addition to PIWIL2, several other regulators of retrotransposition and endogenous transposable elements were dysregulated following MIA, potentially connecting MIA to retrotransposition. CONCLUSIONS Together, these results begin to elucidate the brain-level molecular processes through which MIA may impart risk for psychiatric disease.
Collapse
Affiliation(s)
- Nicholas F Page
- Department of Psychiatry, Center for Autism Research and Treatment, Los Angeles, California; Department of Cell Biology and Neuroscience, Rutgers University-New Brunswick, Piscataway, New Jersey
| | - Michael J Gandal
- Department of Psychiatry, Center for Autism Research and Treatment, Los Angeles, California
| | - Myka L Estes
- Center for Neuroscience, School of Medicine, University of California, Davis, Davis, California
| | - Scott Cameron
- Center for Neuroscience, School of Medicine, University of California, Davis, Davis, California
| | - Jessie Buth
- Department of Psychiatry, Center for Autism Research and Treatment, Los Angeles, California; Program in Neurobehavioral Genetics, Center for Autism Research and Treatment, Los Angeles, California
| | - Sepideh Parhami
- Department of Psychiatry, Center for Autism Research and Treatment, Los Angeles, California; Program in Neurobehavioral Genetics, Center for Autism Research and Treatment, Los Angeles, California
| | - Gokul Ramaswami
- Department of Psychiatry, Center for Autism Research and Treatment, Los Angeles, California; Program in Neurobehavioral Genetics, Center for Autism Research and Treatment, Los Angeles, California
| | - Karl Murray
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California, Davis, Davis, California
| | - David G Amaral
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California, Davis, Davis, California
| | - Judy A Van de Water
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California, Davis, Davis, California
| | - Cynthia M Schumann
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California, Davis, Davis, California
| | - Cameron S Carter
- Center for Neuroscience, School of Medicine, University of California, Davis, Davis, California; Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California, Davis, Davis, California
| | - Melissa D Bauman
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California, Davis, Davis, California
| | - A Kimberley McAllister
- Center for Neuroscience, School of Medicine, University of California, Davis, Davis, California; Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California, Davis, Davis, California
| | - Daniel H Geschwind
- Department of Psychiatry, Center for Autism Research and Treatment, Los Angeles, California; Program in Neurobehavioral Genetics, Center for Autism Research and Treatment, Los Angeles, California; Department of Neurology, Center for Autism Research and Treatment, Los Angeles, California; Department of Human Genetics, Semel Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California.
| |
Collapse
|
39
|
Chamera K, Trojan E, Kotarska K, Szuster-Głuszczak M, Bryniarska N, Tylek K, Basta-Kaim A. Role of Polyinosinic:Polycytidylic Acid-Induced Maternal Immune Activation and Subsequent Immune Challenge in the Behaviour and Microglial Cell Trajectory in Adult Offspring: A Study of the Neurodevelopmental Model of Schizophrenia. Int J Mol Sci 2021; 22:ijms22041558. [PMID: 33557113 PMCID: PMC7913889 DOI: 10.3390/ijms22041558] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/22/2021] [Accepted: 02/01/2021] [Indexed: 12/21/2022] Open
Abstract
Multiple lines of evidence support the pathogenic role of maternal immune activation (MIA) in the occurrence of the schizophrenia-like disturbances in offspring. While in the brain the homeostatic role of neuron-microglia protein systems is well documented, the participation of the CX3CL1-CX3CR1 and CD200-CD200R dyads in the adverse impact of MIA often goes under-recognized. Therefore, in the present study, we examined the effect of MIA induced by polyinosinic:polycytidylic acid (Poly I:C) on the CX3CL1-CX3CR1 and CD200-CD200R axes, microglial trajectory (MhcII, Cd40, iNos, Il-1β, Tnf-α, Il-6, Arg1, Igf-1, Tgf-β and Il-4), and schizophrenia-like behaviour in adult male offspring of Sprague-Dawley rats. Additionally, according to the “two-hit” hypothesis of schizophrenia, we evaluated the influence of acute challenge with Poly I:C in adult prenatally MIA-exposed animals on the above parameters. In the present study, MIA evoked by Poly I:C injection in the late period of gestation led to the appearance of schizophrenia-like disturbances in adult offspring. Our results revealed the deficits manifested as a diminished number of aggressive interactions, presence of depressive-like episodes, and increase of exploratory activity, as well as a dichotomy in the sensorimotor gating in the prepulse inhibition (PPI) test expressed as two behavioural phenotypes (MIAPPI-low and MIAPPI-high). Furthermore, in the offspring rats subjected to a prenatal challenge (i.e., MIA) we noticed the lack of modulation of behavioural changes after the additional acute immune stimulus (Poly I:C) in adulthood. The important finding reported in this article is that MIA affects the expression and levels of the neuron-microglia proteins in the frontal cortex and hippocampus of adult offspring. We found that the changes in the CX3CL1-CX3CR1 axis could affect microglial trajectory, including decreased hippocampal mRNA level of MhcII and elevated cortical expression of Igf-1 in the MIAPPI-high animals and/or could cause the up-regulation of an inflammatory response (Il-6, Tnf-α, iNos) after the “second hit” in both examined brain regions and, at least in part, might differentiate behavioural disturbances in adult offspring. Consequently, the future effort to identify the biological background of these interactions in the Poly I:C-induced MIA model in Sprague-Dawley rats is desirable to unequivocally clarify this issue.
Collapse
|
40
|
Santana-Coelho D, Layne-Colon D, Valdespino R, Ross CC, Tardif SD, O'Connor JC. Advancing Autism Research From Mice to Marmosets: Behavioral Development of Offspring Following Prenatal Maternal Immune Activation. Front Psychiatry 2021; 12:705554. [PMID: 34421684 PMCID: PMC8377364 DOI: 10.3389/fpsyt.2021.705554] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 07/07/2021] [Indexed: 12/31/2022] Open
Abstract
Understanding the mechanism(s) by which maternal immune activation (MIA) during gestation may disrupt neurodevelopment and increase the susceptibility for disorders such as autism spectrum disorder (ASD) or schizophrenia is a critical step in the development of better treatments and preventive measures. A large body of literature has investigated the pathophysiology of MIA in rodents. However, a translatability gap plagues pre-clinical research of complex behavioral/developmental diseases and those diseases requiring clinical diagnosis, such as ASD. While ideal for their genetic flexibility, vast reagent toolkit, and practicality, rodent models often lack important elements of ethological validity. Hence, our study aimed to develop and characterize the prenatal MIA model in marmosets. Here, we adapted the well-characterized murine maternal immune activation model. Pregnant dams were administered 5 mg/kg poly-L-lysine stabilized polyinosinic-polycytidylic acid (Poly ICLC) subcutaneously three times during gestation (gestational day 63, 65, and 67). Dams were allowed to deliver naturally with no further experimental treatments. After parturition, offspring were screened for general health and vigor, and individual assessment of communication development and social behavior was measured during neonatal or adolescent periods. Similar to rodent models, offspring subjected to MIA exhibited a disruption in patterns of communication during early development. Assessment of social behavior in a marmoset-modified 3-chamber test at 3 and 9 months of age revealed alterations in social behavior that, in some instances, was sex-dependent. Together, our data indicate that marmosets are an excellent non-human primate model for investigating the neurodevelopmental and behavioral consequences of exposure to prenatal challenges, like MIA. Additional studies are necessary to more completely characterize the effect of prenatal inflammation on marmoset development and explore therapeutic intervention strategies that may be applicable in a clinical setting.
Collapse
Affiliation(s)
- Danielle Santana-Coelho
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Donna Layne-Colon
- Southwest National Primate Center, Texas Biomedical Research Institute, San Antonio, TX, United States
| | - Roslyn Valdespino
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Corinna C Ross
- Southwest National Primate Center, Texas Biomedical Research Institute, San Antonio, TX, United States
| | - Suzette D Tardif
- Southwest National Primate Center, Texas Biomedical Research Institute, San Antonio, TX, United States
| | - Jason C O'Connor
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States.,Audie L. Murphy Veterans Affairs, South Texas Veterans Health System, San Antonio, TX, United States
| |
Collapse
|
41
|
Increased RNA editing in maternal immune activation model of neurodevelopmental disease. Nat Commun 2020; 11:5236. [PMID: 33067431 PMCID: PMC7567798 DOI: 10.1038/s41467-020-19048-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 09/23/2020] [Indexed: 12/19/2022] Open
Abstract
The etiology of major neurodevelopmental disorders such as schizophrenia and autism is unclear, with evidence supporting a combination of genetic factors and environmental insults, including viral infection during pregnancy. Here we utilized a mouse model of maternal immune activation (MIA) with the viral mimic PolyI:C infection during early gestation. We investigated the transcriptional changes in the brains of mouse fetuses following MIA during the prenatal period, and evaluated the behavioral and biochemical changes in the adult brain. The results reveal an increase in RNA editing levels and dysregulation in brain development-related gene pathways in the fetal brains of MIA mice. These MIA-induced brain editing changes are not observed in adulthood, although MIA-induced behavioral deficits are observed. Taken together, our findings suggest that MIA induces transient dysregulation of RNA editing at a critical time in brain development.
Collapse
|
42
|
Goh JY, O'Sullivan SE, Shortall SE, Zordan N, Piccinini AM, Potter HG, Fone KCF, King MV. Gestational poly(I:C) attenuates, not exacerbates, the behavioral, cytokine and mTOR changes caused by isolation rearing in a rat 'dual-hit' model for neurodevelopmental disorders. Brain Behav Immun 2020; 89:100-117. [PMID: 32485291 DOI: 10.1016/j.bbi.2020.05.076] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 05/28/2020] [Accepted: 05/28/2020] [Indexed: 12/12/2022] Open
Abstract
Many psychiatric illnesses have a multifactorial etiology involving genetic and environmental risk factors that trigger persistent neurodevelopmental impairments. Several risk factors have been individually replicated in rodents, to understand disease mechanisms and evaluate novel treatments, particularly for poorly-managed negative and cognitive symptoms. However, the complex interplay between various factors remains unclear. Rodent dual-hit neurodevelopmental models offer vital opportunities to examine this and explore new strategies for early therapeutic intervention. This study combined gestational administration of polyinosinic:polycytidylic acid (poly(I:C); PIC, to mimic viral infection during pregnancy) with post-weaning isolation of resulting offspring (to mirror adolescent social adversity). After in vitro and in vivo studies required for laboratory-specific PIC characterization and optimization, we administered 10 mg/kg i.p. PIC potassium salt to time-mated Lister hooded dams on gestational day 15. This induced transient hypothermia, sickness behavior and weight loss in the dams, and led to locomotor hyperactivity, elevated striatal cytokine levels, and increased frontal cortical JNK phosphorylation in the offspring at adulthood. Remarkably, instead of exacerbating the well-characterized isolation syndrome, gestational PIC exposure actually protected against a spectrum of isolation-induced behavioral and brain regional changes. Thus isolation reared rats exhibited locomotor hyperactivity, impaired associative memory and reversal learning, elevated hippocampal and frontal cortical cytokine levels, and increased mammalian target of rapamycin (mTOR) activation in the frontal cortex - which were not evident in isolates previously exposed to gestational PIC. Brains from adolescent littermates suggest little contribution of cytokines, mTOR or JNK to early development of the isolation syndrome, or resilience conferred by PIC. But notably hippocampal oxytocin, which can protect against stress, was higher in adolescent PIC-exposed isolates so might contribute to a more favorable outcome. These findings have implications for identifying individuals at risk for disorders like schizophrenia who may benefit from early therapeutic intervention, and justify preclinical assessment of whether adolescent oxytocin manipulations can modulate disease onset or progression.
Collapse
Affiliation(s)
- Jen-Yin Goh
- School of Life Sciences, University of Nottingham, Medical School, Queen's Medical Centre, Nottingham NG7 2UH, UK
| | - Saoirse E O'Sullivan
- School of Medicine, University of Nottingham, Royal Derby Hospital, Derby DE22 3DT, UK
| | - Sinead E Shortall
- School of Life Sciences, University of Nottingham, Medical School, Queen's Medical Centre, Nottingham NG7 2UH, UK
| | - Nicole Zordan
- School of Pharmacy, University of Nottingham, University Park, Nottingham NG7 2RD, UK
| | - Anna M Piccinini
- School of Pharmacy, University of Nottingham, University Park, Nottingham NG7 2RD, UK
| | - Harry G Potter
- Division of Evolution and Genomic Sciences, School of Biological Sciences, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, UK
| | - Kevin C F Fone
- School of Life Sciences, University of Nottingham, Medical School, Queen's Medical Centre, Nottingham NG7 2UH, UK
| | - Madeleine V King
- School of Life Sciences, University of Nottingham, Medical School, Queen's Medical Centre, Nottingham NG7 2UH, UK.
| |
Collapse
|
43
|
Maternal Immunity in Autism Spectrum Disorders: Questions of Causality, Validity, and Specificity. J Clin Med 2020; 9:jcm9082590. [PMID: 32785127 PMCID: PMC7464885 DOI: 10.3390/jcm9082590] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/05/2020] [Accepted: 08/06/2020] [Indexed: 02/07/2023] Open
Abstract
Autism spectrum disorders (ASD) are complex neurodevelopmental disorders with unknown heterogeneous aetiologies. Epidemiological studies have found an association between maternal infection and development of ASD in the offspring, and clinical findings reveal a state of immune dysregulation in the pre- and postnatal period of affected subjects. Maternal immune activation (MIA) has been proposed to mediate this association by altering fetal neurodevelopment and leading to autism. Although animal models have supported a causal link between MIA and development of ASD, their validity needs to be explored. Moreover, considering that only a small proportion of affected offspring develop autism, and that MIA has been implicated in related diseases such as schizophrenia, a key unsolved question is how disease specificity and phenotypic outcome are determined. Here, we have integrated preclinical and clinical evidence, including the use of animal models for establishing causality, to explore the role of maternal infections in ASD. A proposed priming/multi-hit model may offer insights into the clinical heterogeneity of ASD, its convergence with related disorders, and therapeutic strategies.
Collapse
|
44
|
Saxena R, Babadi M, Namvarhaghighi H, Roullet FI. Role of environmental factors and epigenetics in autism spectrum disorders. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020; 173:35-60. [PMID: 32711816 DOI: 10.1016/bs.pmbts.2020.05.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Autism Spectrum Disorder (ASD) is a neurodevelopmental disorder thought to be caused by predisposing high-risk genes that may be altered during the early development by environmental factors. The impact of maternal challenges during pregnancy on the prevalence of ASD has been widely studied in clinical and animal studies. Here, we review some clinical and pre-clinical evidence that links environmental factors (i.e., infection, air pollution, pesticides, valproic acid and folic acid) and the risk of ASD. Additionally, certain prenatal environmental challenges such as the valproate and folate prenatal exposures allow us to study mechanisms possibly linked to the etiology of ASD, for instance the epigenetic processes. These mechanistic pathways are also presented and discussed in this chapter.
Collapse
Affiliation(s)
- Roheeni Saxena
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, United States
| | - Melika Babadi
- School of Interdisciplinary Science, McMaster University, Hamilton, ON, Canada
| | | | - Florence I Roullet
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
45
|
Discrimination difficulty, cognitive burden, and reversal impairments in a maternal immune activation model of schizophrenia risk. Behav Processes 2019; 167:103936. [DOI: 10.1016/j.beproc.2019.103936] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 07/22/2019] [Accepted: 08/01/2019] [Indexed: 12/14/2022]
|
46
|
[Research advances in the role of mTOR signaling pathway in autism spectrum disorder]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2019; 21. [PMID: 31315775 PMCID: PMC7389111 DOI: 10.7499/j.issn.1008-8830.2019.07.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Mammalian target of rapamycin (mTOR) is an intracellular signaling pathway molecule which regulates various fundamental physiological processes. The mTOR signaling pathway plays an important role in synaptic plasticity, information transmission and processing, and neuroregulation. Dysregulation of the mTOR signaling pathway is generally considered to be related to the pathogenesis of autism spectrum disorder (ASD); meanwhile, the mTOR inhibitor can ameliorate the symptoms of ASD. The role of mTOR in the pathogenesis of ASD is summarized in this article to provide a theoretical basis for targeted therapy of ASD.
Collapse
|