1
|
Otero AM, Connolly MG, Gonzalez-Ricon RJ, Wang SS, Allen JM, Antonson AM. Influenza A virus during pregnancy disrupts maternal intestinal immunity and fetal cortical development in a dose- and time-dependent manner. Mol Psychiatry 2025; 30:13-28. [PMID: 38961232 DOI: 10.1038/s41380-024-02648-9] [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: 12/19/2023] [Revised: 06/19/2024] [Accepted: 06/21/2024] [Indexed: 07/05/2024]
Abstract
Epidemiological studies link exposure to viral infection during pregnancy, including influenza A virus (IAV) infection, with increased incidence of neurodevelopmental disorders (NDDs) in offspring. Models of maternal immune activation (MIA) using viral mimetics demonstrate that activation of maternal intestinal T helper 17 (TH17) cells, which produce effector cytokine interleukin (IL)-17, leads to aberrant fetal brain development, such as neocortical malformations. Fetal microglia and border-associated macrophages (BAMs) also serve as potential cellular mediators of MIA-induced cortical abnormalities. However, neither the inflammation-induced TH17 cell pathway nor fetal brain-resident macrophages have been thoroughly examined in models of live viral infection during pregnancy. Here, we inoculated pregnant mice with two infectious doses of IAV and evaluated peak innate and adaptive immune responses in the dam and fetus. While respiratory IAV infection led to dose-dependent maternal colonic shortening and microbial dysregulation, there was no elevation in intestinal TH17 cells nor IL-17. Systemically, IAV resulted in consistent dose- and time-dependent increases in IL-6 and IFN-γ. Fetal cortical abnormalities and global changes in fetal brain transcripts were observable in the high-but not the moderate-dose IAV group. Profiling of fetal microglia and BAMs revealed dose- and time-dependent differences in the numbers of meningeal but not choroid plexus BAMs, while microglial numbers and proliferative capacity of Iba1+ cells remained constant. Fetal brain-resident macrophages increased phagocytic CD68 expression, also in a dose- and time-dependent fashion. Taken together, our findings indicate that certain features of MIA are conserved between mimetic and live virus models, while others are not. Overall, we provide consistent evidence of an infection severity threshold for downstream maternal inflammation and fetal cortical abnormalities, which recapitulates a key feature of the epidemiological data and further underscores the importance of using live pathogens in NDD modeling to better evaluate the complete immune response and to improve translation to the clinic.
Collapse
Affiliation(s)
- Ashley M Otero
- Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Meghan G Connolly
- Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | | | - Selena S Wang
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jacob M Allen
- Department of Kinesiology and Community Health, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Adrienne M Antonson
- Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL, USA.
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
2
|
Buonfiglioli A, Kübler R, Missall R, De Jong R, Chan S, Haage V, Wendt S, Lin AJ, Mattei D, Graziani M, Latour B, Gigase F, Chiu R, Zhang Y, Nygaard HB, De Jager PL, De Witte LD. A microglia-containing cerebral organoid model to study early life immune challenges. Brain Behav Immun 2025; 123:1127-1146. [PMID: 39500415 DOI: 10.1016/j.bbi.2024.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 10/10/2024] [Accepted: 11/02/2024] [Indexed: 11/13/2024] Open
Abstract
Prenatal infections and activation of the maternal immune system have been proposed to contribute to causing neurodevelopmental disorders (NDDs), chronic conditions often linked to brain abnormalities. Microglia are the resident immune cells of the brain and play a key role in neurodevelopment. Disruption of microglial functions can lead to brain abnormalities and increase the risk of developing NDDs. How the maternal as well as the fetal immune system affect human neurodevelopment and contribute to NDDs remains unclear. An important reason for this knowledge gap is the fact that the impact of exposure to prenatal risk factors has been challenging to study in the human context. Here, we characterized a model of cerebral organoids (CO) with integrated microglia (COiMg). These organoids express typical microglial markers and respond to inflammatory stimuli. The presence of microglia influences cerebral organoid development, including cell density and neural differentiation, and regulates the expression of several ciliated and mesenchymal cell markers. Moreover, COiMg and organoids without microglia show similar but also distinct responses to inflammatory stimuli. Additionally, IFN-γ induced significant transcriptional and structural changes in the cerebral organoids, that appear to be regulated by the presence of microglia. Specifically, interferon-gamma (IFN-γ) was found to alter the expression of genes linked to autism. This model provides a valuable tool to study how inflammatory perturbations and microglial presence affect neurodevelopmental processes.
Collapse
Affiliation(s)
- Alice Buonfiglioli
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Raphael Kübler
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Human Genetics, Radboud UMC, Nijmegen, Netherlands (the)
| | - Roy Missall
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Renske De Jong
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Stephanie Chan
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Verena Haage
- Center for Translational & Computational Neuroimmunology, Department of Neurology and the Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Stefan Wendt
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver V6T 1Z3, Canada
| | - Ada J Lin
- Division of Neurology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver V6T 1Z3, Canada
| | - Daniele Mattei
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mara Graziani
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Human Genetics, Radboud UMC, Nijmegen, Netherlands (the); Donders Institute for Brain, Cognition and Behaviour, 6500 HB, Nijmegen, Netherlands (the)
| | - Brooke Latour
- Department of Human Genetics, Radboud UMC, Nijmegen, Netherlands (the); Donders Institute for Brain, Cognition and Behaviour, 6500 HB, Nijmegen, Netherlands (the)
| | - Frederieke Gigase
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Rebecca Chiu
- Center for Translational & Computational Neuroimmunology, Department of Neurology and the Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Ya Zhang
- Center for Translational & Computational Neuroimmunology, Department of Neurology and the Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Haakon B Nygaard
- Division of Neurology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver V6T 1Z3, Canada
| | - Philip L De Jager
- Center for Translational & Computational Neuroimmunology, Department of Neurology and the Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Lot D De Witte
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Human Genetics, Radboud UMC, Nijmegen, Netherlands (the); Donders Institute for Brain, Cognition and Behaviour, 6500 HB, Nijmegen, Netherlands (the); Department of Psychiatry, Radboud UMC, Nijmegen, Netherlands (the)
| |
Collapse
|
3
|
Griego E, Cerna C, Sollozo-Dupont I, Fuenzalida M, Galván EJ. Maternal immune activation alters temporal Precision of spike generation of CA1 pyramidal neurons by Unbalancing GABAergic inhibition intheOffspring. Brain Behav Immun 2025; 123:211-228. [PMID: 39293693 DOI: 10.1016/j.bbi.2024.09.012] [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: 02/17/2024] [Revised: 09/05/2024] [Accepted: 09/13/2024] [Indexed: 09/20/2024] Open
Abstract
Infection during pregnancy represents a risk factor for neuropsychiatric disorders associated with neurodevelopmental alterations. A growing body of evidence from rodents and non-human primates shows that maternal inflammation induced by viral or bacterial infections results in several neurobiological alterations in the offspring. These changes may play an important role in the pathophysiology of psychiatric disorders like schizophrenia and autism spectrum disorders, whose clinical features include impairments in cognitive processing and social performance. Such alterations are causally associated with the maternal inflammatory response to infection rather than with the infection itself. Previously, we reported that CA1 pyramidal neurons of mice exposed to MIA exhibit increased excitability accompanied by a reduction in dendritic complexity. However, potential alterations in cellular and synaptic rules that shape the neuronal computational properties of the offspring remain to be determined. In this study, using mice as subjects, we identified a series of cellular and synaptic alterations endured by CA1 pyramidal neurons of the dorsal hippocampus in a lipopolysaccharide-induced maternal immune activation (MIA) model. Our data indicate that MIA reshapes the excitation-inhibition balance by decreasing the perisomatic GABAergic inhibition predominantly mediated by cholecystokinin-expressing Interneurons but not parvalbumin-expressing interneurons impinging on CA1 pyramidal neurons. These alterations yield a dysregulated amplification of the temporal and spatial synaptic integration. In addition, MIA-exposed offspring displayed social and anxiety-like abnormalities. These findings collectively contribute to understanding the cellular and synaptic alterations underlying the behavioral symptoms present in neurodevelopmental disorders associated with MIA.
Collapse
Affiliation(s)
- Ernesto Griego
- Departamento de Farmacobiología, Cinvestav, Ciudad de México, México; Current Address: Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, United States
| | - Camila Cerna
- Centro de Neurobiología y Fisiopatología Integrativa, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile; Millennium Nucleus of Neuroepigenetics and Plasticity (EpiNeuro), Santiago, Chile
| | - Isabel Sollozo-Dupont
- Instituto Nacional de Perinatología, Isidro Espinosa de los Reyes. Ciudad de México, México
| | - Marco Fuenzalida
- Centro de Neurobiología y Fisiopatología Integrativa, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile; Millennium Nucleus of Neuroepigenetics and Plasticity (EpiNeuro), Santiago, Chile
| | - Emilio J Galván
- Departamento de Farmacobiología, Cinvestav, Ciudad de México, México; Centro de Investigaciones sobre el Envejecimiento, CIE-Cinvestav, Ciudad de México, México.
| |
Collapse
|
4
|
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
|
5
|
Niskanen A, Barron A, Azaryah H, Kerkelä M, Pulli E, Tuulari JJ, Lukkarinen M, Karlsson L, Muetzel RL, Campoy C, Catena A, Tiemeier H, Khandaker GM, Karlsson H, Veijola J, Björnholm L. Sex-specific associations between maternal prenatal inflammation and offspring cortical morphology in youth: A harmonised study across four birth cohorts. Brain Behav Immun 2025; 123:1081-1090. [PMID: 39505051 DOI: 10.1016/j.bbi.2024.11.010] [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: 04/23/2024] [Revised: 09/29/2024] [Accepted: 11/03/2024] [Indexed: 11/08/2024] Open
Abstract
Maternal immune activation (MIA) during pregnancy is implicated in offspring psychiatric disorders. However, it is unknown to what extent MIA affects neurodevelopment, particularly cerebrocortical anatomy, in the general population, and whether effects differ by sex. The current study used vertex-wise statistics to examine the association between maternal prenatal CRP, an archetypal systemic inflammatory marker, and offspring cortical thickness, surface area, and volume, in 2635 mother-child dyads (5.4-26.5 years) from three population-based cohorts, and one clinical cohort enriched for presence of inflammation markers. Maternal CRP within a normal physiological range (<10 mg/L) exhibited sex-specific quadratic associations with cortical morphological measures in 2 regions in males and 1 region in females at childhood. Elevated (>10 mg/L) CRP was associated with regional cortical morphology in females and in a pooled sample of sexes. Overall, MIA is associated with cortical development in a regional and sex-specific manner in studies spanning childhood to adulthood.
Collapse
Affiliation(s)
- Anni Niskanen
- Research Unit of Clinical Medicine, Department of Psychiatry, University of Oulu, Oulu, Finland; Department of Psychiatry, Oulu University Hospital, Oulu, Finland; Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Aaron Barron
- FinnBrain Birth Cohort Study, Turku Brain and Mind Centre, Department of Clinical Medicine, University of Turku, Turku, Finland; Department of Clinical Medicine, Psychiatry, University of Turku and Turku University Hospital, Turku, Finland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Hatim Azaryah
- Department of Pediatrics, University of Granada, Granada, Spain; EURISTIKOS Excellence Centre for Pediatric Research, School of Medicine, University of Granada, Granada, Spain; Instituto Biosanitario de Granada (Ibs-Granada), Granada, Spain
| | - Martta Kerkelä
- Research Unit of Clinical Medicine, Department of Psychiatry, University of Oulu, Oulu, Finland; Department of Psychiatry, Oulu University Hospital, Oulu, Finland
| | - Elmo Pulli
- FinnBrain Birth Cohort Study, Turku Brain and Mind Centre, Department of Clinical Medicine, University of Turku, Turku, Finland; Centre for Population Health Research, Turku University Hospital and University of Turku, Turku, Finland
| | - Jetro J Tuulari
- FinnBrain Birth Cohort Study, Turku Brain and Mind Centre, Department of Clinical Medicine, University of Turku, Turku, Finland; Department of Clinical Medicine, Psychiatry, University of Turku and Turku University Hospital, Turku, Finland; Centre for Population Health Research, Turku University Hospital and University of Turku, Turku, Finland; Turku Collegium for Science, Medicine and Technology (TCSMT), University of Turku, Turku, Finland
| | - Minna Lukkarinen
- FinnBrain Birth Cohort Study, Turku Brain and Mind Centre, Department of Clinical Medicine, University of Turku, Turku, Finland; Centre for Population Health Research, Turku University Hospital and University of Turku, Turku, Finland; The Department of Pediatrics and Adolescent Medicine, University of Turku, and Turku University Hospital, Turku, Finland
| | - Linnea Karlsson
- FinnBrain Birth Cohort Study, Turku Brain and Mind Centre, Department of Clinical Medicine, University of Turku, Turku, Finland; Centre for Population Health Research, Turku University Hospital and University of Turku, Turku, Finland; Department of Clinical Medicine, Unit of Public Health, University of Turku, Finland; Department of Child Psychiatry, Turku University Hospital, Finland
| | - Ryan L Muetzel
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus University Medical Center, Rotterdam, Netherlands (the); Department of Radiology and Nuclear Medicine, Erasmus University Medical Center,Netherlands (the)
| | - Cristina Campoy
- Department of Pediatrics, University of Granada, Granada, Spain; EURISTIKOS Excellence Centre for Pediatric Research, School of Medicine, University of Granada, Granada, Spain; Instituto Biosanitario de Granada (Ibs-Granada), Granada, Spain; CIBERESP, Spanish Research Network on Epidemiology and Public Health, ISCIII, Madrid, Spain
| | - Andrés Catena
- Department of Experimental Psychology, School of Psychology, University of Granada, Granada, Spain; Mind, Brain & Behaviour Centre (CIMCYC), University of Granada, Spain
| | - Henning Tiemeier
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus University Medical Center, Rotterdam, Netherlands (the); Department of Social and Behavioral Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Golam M Khandaker
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK; Centre for Academic Mental Health, Bristol Medical School, University of Bristol, Bristol, UK; National Institute of Health and Care Research Bristol Biomedical Research Centre, University Hospitals Bristol and Weston NHS Foundation Trust and University of Bristol, Bristol, UK; Avon and Wiltshire Mental Health Partnership NHS Trust, Bristol, UK
| | - Hasse Karlsson
- FinnBrain Birth Cohort Study, Turku Brain and Mind Centre, Department of Clinical Medicine, University of Turku, Turku, Finland; Department of Clinical Medicine, Psychiatry, University of Turku and Turku University Hospital, Turku, Finland; Centre for Population Health Research, Turku University Hospital and University of Turku, Turku, Finland
| | - Juha Veijola
- Research Unit of Clinical Medicine, Department of Psychiatry, University of Oulu, Oulu, Finland; Department of Psychiatry, Oulu University Hospital, Oulu, Finland; Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Lassi Björnholm
- Research Unit of Clinical Medicine, Department of Psychiatry, University of Oulu, Oulu, Finland; Department of Psychiatry, Oulu University Hospital, Oulu, Finland; Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland.
| |
Collapse
|
6
|
Guma E, Chakravarty MM. Immune Alterations in the Intrauterine Environment Shape Offspring Brain Development in a Sex-Specific Manner. Biol Psychiatry 2025; 97:12-27. [PMID: 38679357 PMCID: PMC11511788 DOI: 10.1016/j.biopsych.2024.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 03/20/2024] [Accepted: 04/17/2024] [Indexed: 05/01/2024]
Abstract
Exposure to immune dysregulation in utero or in early life has been shown to increase risk for neuropsychiatric illness. The sources of inflammation can be varied, including acute exposures due to maternal infection or acute stress, or persistent exposures due to chronic stress, obesity, malnutrition, or autoimmune diseases. These exposures may cause subtle alteration in brain development, structure, and function that can become progressively magnified across the lifespan, potentially increasing the likelihood of developing a neuropsychiatric conditions. There is some evidence that males are more susceptible to early-life inflammatory challenges than females. In this review, we discuss the various sources of in utero or early-life immune alteration and the known effects on fetal development with a sex-specific lens. To do so, we leveraged neuroimaging, behavioral, cellular, and neurochemical findings. Gaining clarity about how the intrauterine environment affects offspring development is critically important for informing preventive and early intervention measures that may buffer against the effects of these early-life risk factors.
Collapse
Affiliation(s)
- Elisa Guma
- Section on Developmental Neurogenomics, Human Genetics Branch, National Institute of Mental Health, Bethesda, Maryland; Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts.
| | - M Mallar Chakravarty
- Computational Brain Anatomy Laboratory, Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec, Canada; Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec, Canada; Department of Psychiatry, McGill University, Montreal, Quebec, Canada; Department of Biomedical Engineering, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
7
|
Taskiran M, Yildiz Taskiran S, Unal G, Bozkurt NM, Golgeli A. Vortioxetine improved schizophrenia-like behavioral deficits in a Poly I:C-induced maternal immune activation model of schizophrenia in rats. Fundam Clin Pharmacol 2024; 38:1069-1079. [PMID: 38962906 DOI: 10.1111/fcp.13028] [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: 04/07/2023] [Revised: 04/21/2024] [Accepted: 06/26/2024] [Indexed: 07/05/2024]
Abstract
BACKGROUND Several studies provide clear evidence that exposure to various infections during pregnancy are linked with an increased risk for schizophrenia. In preclinical studies, administration of polyinosinic-polycytidylic acid (Poly I:C) in pregnant rodents can induce maternal immune activation leading to impairments in brain function in the offspring. OBJECTIVES The aim of this study was to investigate the effect of vortioxetine, a multimodal selective serotonin reuptake inhibitor (SSRI), in the pathophysiology of Poly I:C-induced schizophrenia-like model in rats. METHODS For this purpose, Poly I:C (8 mg/kg, ip) was injected into pregnant animals 14 days after mating, and tail blood was taken for determination of IL-6 levels after 2 h. At postnatal days 83-86, behavioral tests were performed. RESULTS Our results revealed that Poly I:C caused impairments in prepulse inhibition, novel object recognition, social interaction, and open-field tests. Chronic administration of vortioxetine (2.5, 5, and 10 mg/kg, ip, postnatal days 69-83) caused significant improvements in these deficits. CONCLUSION Overall, our findings indicate that vortioxetine may provide new therapeutic approaches for the treatment of schizophrenia. We think that increased serotonergic activity in frontal brain regions may provide the ameliorative effect of vortioxetine, especially on negative and cognitive symptoms. Therefore, it will be useful to determine the efficacy of vortioxetine with combined drugs with further studies.
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
| | - Nuh Mehmet Bozkurt
- Department of Pharmacology, Faculty of Pharmacy, Erciyes University, Kayseri, Turkey
| | - Asuman Golgeli
- Department of Physiology, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| |
Collapse
|
8
|
Restellini R, Golay P, Jenni R, Baumann PS, Alameda L, Allgäuer L, Steullet P, Abrahamyan Empson L, Mebdouhi N, Do KQ, Conus P, Dwir D, Klauser P. Winter birth: A factor of poor functional outcome in a Swiss early psychosis cohort. Schizophr Res 2024; 274:206-211. [PMID: 39341100 DOI: 10.1016/j.schres.2024.09.022] [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: 05/13/2024] [Revised: 09/12/2024] [Accepted: 09/20/2024] [Indexed: 09/30/2024]
Abstract
OBJECTIVE Winter birth has consistently been identified as a risk factor for schizophrenia. This study aimed to determine whether individuals born during this season are also at higher risk for early psychosis and whether this is associated with distinct functional and clinical outcomes. METHODS We conducted a prospective study on 222 patients during their early phase of psychosis in Switzerland, nested in the Treatment and Early Intervention in Psychosis (TIPP) cohort. We compared the birth trimesters of these patients with those of the general Swiss population. Additionally, we evaluated the Global Assessment of Functioning scale (GAF) and the Positive and Negative Syndrome Scale (PANSS) scores among patients born in winter (January to March) versus those born during the rest of the year during a three-year follow-up period. RESULTS A significantly higher proportion of patients experiencing early psychosis were born in winter compared to the general Swiss population. Patients born in winter had significantly lower GAF scores at 6 months, 24 months, and 36 months of follow-up, compared to patients born during the rest of the year. They also manifested fewer positive symptoms, as indicated by the PANSS positive subscale. CONCLUSION Birth in winter appears to be associated with a lower functional outcome and potentially distinct symptomatology in the early phase of psychosis.
Collapse
Affiliation(s)
- Romeo Restellini
- Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland; Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland; Service of Emergency Medicine, Department of Acute Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Philippe Golay
- Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland; La Source School of Nursing, HES-SO University of Applied Sciences and Arts Western Switzerland, Lausanne, Switzerland
| | - Raoul Jenni
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Philipp S Baumann
- Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Luis Alameda
- Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland; Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College of London, London, UK; Centro Investigacion Biomedica en Red de Salud Mental (CIBERSAM), Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocio, Departamento de Psiquiatria, Universidad de Sevilla, Sevilla, Spain
| | - Larissa Allgäuer
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Pascal Steullet
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Lilith Abrahamyan Empson
- Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Nadir Mebdouhi
- Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Kim Quang Do
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Philippe Conus
- Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Daniella Dwir
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Paul Klauser
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland; Service of Child and Adolescent Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
9
|
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
|
10
|
Zeng X, Fan L, Qin Q, Zheng D, Wang H, Li M, Jiang Y, Wang H, Liu H, Liang S, Wu L, Liang S. Exogenous PD-L1 binds to PD-1 to alleviate and prevent autism-like behaviors in maternal immune activation-induced male offspring mice. Brain Behav Immun 2024; 122:527-546. [PMID: 39182588 DOI: 10.1016/j.bbi.2024.08.042] [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: 04/04/2024] [Revised: 08/15/2024] [Accepted: 08/22/2024] [Indexed: 08/27/2024] Open
Abstract
Autism Spectrum Disorder (ASD) is a neurodevelopmental disorder caused by the interaction of multiple pathogenic factors. Epidemiological studies and animal experiments indicate that maternal immune activation (MIA) is closely related to the development of ASD in offspring. A large number of pro-inflammatory cytokines are transferred from the placenta to the fetal brain during MIA, which impedes fetal neurodevelopment and is accompanied by activation of immune cells and microglia. Programmed cell death protein 1 (PD-1) can be highly expressed on the surface of various activated immune cells, when combined with programmed cell death-ligand 1 (PD-L1), it can activate the PD-1/PD-L1 pathway and exert powerful immunosuppressive effects, suggesting that this immune checkpoint may have the potential to treat MIA-induced ASD. This study combined bioinformatics analysis and experimental validation to explore the efficacy of Fc-fused PD-L1 (PD-L1-Fc) in treating MIA-induced ASD. Bioinformatics analysis results showed that in human placental inflammation, IL-6 was upregulated, T cells proliferated significantly, and the PD-1/PD-L1 pathway was significantly enriched. The experimental results showed that intraperitoneal injection of poly(I:C) induced MIA in pregnant mice resulted in significant expression of IL-6 in their serum, placenta, and fetal brain. At the same time, the expression of PD-1 and PD-L1 in the placenta and fetal brain increased, CD4+ T cells in the spleen were significantly activated, and PD-1 expression increased. Their offspring mice exhibited typical ASD-like behaviors. In vitro experiments on primary microglia of offspring mice have confirmed that the expression of IL-6, PD-1, and PD-L1 is significantly increased, and PD-L1-Fc effectively reduced their expression levels. In the prefrontal cortex of MIA offspring mice, there was an increase in the expression of IL-6, PD-1, and PD-L1; activation of microglial cells, and colocalization with PD-1. Then we administered brain stereotaxic injections of PD-L1-Fc to MIA offspring mice and intraperitoneal injections to MIA pregnant mice. The results indicated that PD-L1-Fc effectively suppressed neuroinflammation in the frontal cortex of offspring mice and partially ameliorated ASD-like behaviors; MIA in pregnant mice was significantly alleviated, and the offspring mice they produced did not exhibit neuroinflammation or ASD-like behaviors. In summary, we have demonstrated the therapeutic ability of PD-L1-Fc for MIA-induced ASD, aiming to provide new strategies and insights for the treatment of ASD.
Collapse
Affiliation(s)
- Xin Zeng
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, Harbin 150081, China
| | - Linlin Fan
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, Harbin 150081, China
| | - Qian Qin
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, Harbin 150081, China
| | - Danyang Zheng
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, Harbin 150081, China
| | - Han Wang
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, Harbin 150081, China
| | - Mengyue Li
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, Harbin 150081, China
| | - Yutong Jiang
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, Harbin 150081, China
| | - Hui Wang
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, Harbin 150081, China
| | - Hao Liu
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, Harbin 150081, China
| | - Shengjun Liang
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, Harbin 150081, China
| | - Lijie Wu
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, Harbin 150081, China.
| | - Shuang Liang
- Department of Child and Adolescent Health, Public Health College, Harbin Medical University, Harbin 150081, China.
| |
Collapse
|
11
|
Serrano J, Womack S, Yount C, Chowdhury SF, Arnold M, Brunner J, Duberstein Z, Barrett ES, Scheible K, Miller RK, O'Connor TG. Prenatal maternal immune activation predicts observed fearfulness in infancy. Dev Psychol 2024; 60:2052-2061. [PMID: 38546567 PMCID: PMC11436485 DOI: 10.1037/dev0001718] [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] [Indexed: 05/12/2024]
Abstract
Fear reactivity is an early emerging temperament trait that predicts longer term behavioral and health outcomes. The current analysis tests the hypothesis, an extension of prior research on maternal immune activation (MIA), that the prenatal maternal immune system is a reliable predictor of observed fear reactivity in infancy. The analysis is based on a prospective longitudinal cohort study that collected data from the first trimester and conducted observational assessments of temperament at approximately 12 months of age (n = 281 infants). MIA was assessed from immune biomarkers measured in maternal blood at each trimester; infant temperament was assessed using the Laboratory Temperament Assessment Battery assessment at 12 months; covariates included family and sociodemographic factors. Patterns of inflammatory markers across gestation reliably predicted observed temperament: elevated prenatal MIA was associated with high fear reactivity to novel stimuli. The findings provide novel evidence of prenatal origins of fear reactivity and suggest developmental mechanisms that may underlie early emerging individual differences in child temperament. (PsycInfo Database Record (c) 2024 APA, all rights reserved).
Collapse
Affiliation(s)
| | - Sean Womack
- Department of Psychiatry and Human Behavior, Alpert Medical School, Brown University
| | - Catherine Yount
- Department of Public Health Sciences, University of Rochester School of Medicine and Dentistry
| | | | - Molly Arnold
- Department of Psychology, University of Rochester
| | - Jessica Brunner
- Department of Obstetrics and Gynecology, University of Rochester School of Medicine and Dentistry
| | | | - Emily S Barrett
- Department of Obstetrics and Gynecology, University of Rochester School of Medicine and Dentistry
| | - Kristin Scheible
- Department of Pediatrics, University of Rochester School of Medicine and Dentistry
| | - Richard K Miller
- Department of Obstetrics and Gynecology, University of Rochester School of Medicine and Dentistry
| | - Thomas G O'Connor
- Department of Psychiatry, University of Rochester School of Medicine and Dentistry
| |
Collapse
|
12
|
Haker R, Helft C, Natali Shamir E, Shahar M, Solomon H, Omer N, Blumenfeld-Katzir T, Zlotzover S, Piontkewitz Y, Weiner I, Ben-Eliezer N. Characterization of Brain Abnormalities in Lactational Neurodevelopmental Poly I:C Rat Model of Schizophrenia and Depression Using Machine-Learning and Quantitative MRI. J Magn Reson Imaging 2024. [PMID: 39466009 DOI: 10.1002/jmri.29634] [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: 01/28/2024] [Revised: 10/03/2024] [Accepted: 10/05/2024] [Indexed: 10/29/2024] Open
Abstract
BACKGROUND A recent neurodevelopmental rat model, utilizing lactational exposure to polyriboinosinic-polyribocytidilic acid (Poly I:C) leads to mimics of behavioral phenotypes resembling schizophrenia-like symptoms in male offspring and depression-like symptoms in female offspring. PURPOSE To identify mechanisms of neuronal abnormalities in lactational Poly I:C offspring using quantitative MRI (qMRI) tools. STUDY TYPE Prospective. ANIMAL MODEL Twenty Poly I:C rats and 20 healthy control rats, age 130 postnatal day. FIELD STRENGTH/SEQUENCE 7 T. Multiflip-angle FLASH protocol for T1 mapping; multi-echo spin-echo T2-mapping protocol; echo planar imaging protocol for diffusion tensor imaging. ASSESSMENT Nursing dams were injected with the viral mimic Poly I:C or saline (control group). In adulthood, quantitative maps of T1, T2, proton density, and five diffusion metrics were generated for the offsprings. Seven regions of interest (ROIs) were segmented, followed by extracting 10 quantitative features for each ROI. STATISTICAL TESTS Random forest machine learning (ML) tool was employed to identify MRI markers of disease and classify Poly I:C rats from healthy controls based on quantitative features. RESULTS Poly I:C rats were identified from controls with an accuracy of 82.5 ± 25.9% for females and 85.0 ± 24.0% for males. Poly I:C females exhibited differences mainly in diffusion-derived parameters in the thalamus and the medial prefrontal cortex (MPFC), while males displayed changes primarily in diffusion-derived parameters in the corpus callosum and MPFC. DATA CONCLUSION qMRI shows potential for identifying sex-specific brain abnormalities in the Poly I:C model of neurodevelopmental disorders. LEVEL OF EVIDENCE NA TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Rona Haker
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Coral Helft
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | | | - Moni Shahar
- The AI and Data Science Center, Tel Aviv University, Tel Aviv, Israel
| | - Hadas Solomon
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Noam Omer
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv, Israel
| | | | - Sharon Zlotzover
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv, Israel
| | - Yael Piontkewitz
- School of Psychological Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Ina Weiner
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- School of Psychological Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Noam Ben-Eliezer
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv, Israel
- Center for Advanced Imaging Innovation and Research (CAI2R), New York University School of Medicine, New York, New York, USA
| |
Collapse
|
13
|
Sarnyai Z, Ben-Shachar D. Schizophrenia, a disease of impaired dynamic metabolic flexibility: A new mechanistic framework. Psychiatry Res 2024; 342:116220. [PMID: 39369460 DOI: 10.1016/j.psychres.2024.116220] [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: 03/22/2024] [Revised: 09/21/2024] [Accepted: 09/30/2024] [Indexed: 10/08/2024]
Abstract
Schizophrenia is a chronic, neurodevelopmental disorder with unknown aetiology and pathophysiology that emphasises the role of neurotransmitter imbalance and abnormalities in synaptic plasticity. The currently used pharmacological approach, the antipsychotic drugs, which have limited efficacy and an array of side-effects, have been developed based on the neurotransmitter hypothesis. Recent research has uncovered systemic and brain abnormalities in glucose and energy metabolism, focusing on altered glycolysis and mitochondrial oxidative phosphorylation. These findings call for a re-conceptualisation of schizophrenia pathophysiology as a progressing bioenergetics failure. In this review, we provide an overview of the fundamentals of brain bioenergetics and the changes identified in schizophrenia. We then propose a new explanatory framework positing that schizophrenia is a disease of impaired dynamic metabolic flexibility, which also reconciles findings of abnormal glucose and energy metabolism in the periphery and in the brain along the course of the disease. This evidence-based framework and testable hypothesis has the potential to transform the way we conceptualise this debilitating condition and to develop novel treatment approaches.
Collapse
Affiliation(s)
- Zoltán Sarnyai
- Laboratory of Psychobiology, Department of Neuroscience, The Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Department of Psychiatry, Rambam Health Campus, Haifa, Israel; Laboratory of Psychiatric Neuroscience, Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, QLD, Australia.
| | - Dorit Ben-Shachar
- Laboratory of Psychobiology, Department of Neuroscience, The Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Department of Psychiatry, Rambam Health Campus, Haifa, Israel.
| |
Collapse
|
14
|
ElGrawani W, Mueller FS, Schalbetter SM, Brown SA, Weber-Stadlbauer U, Tarokh L. Maternal immune activation exerts long-term effects on activity and sleep in male offspring mice. Eur J Neurosci 2024; 60:5505-5521. [PMID: 39210746 DOI: 10.1111/ejn.16506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 07/16/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024]
Abstract
Exposure to infectious or non-infectious immune activation during early development is a serious risk factor for long-term behavioural dysfunctions. Mouse models of maternal immune activation (MIA) have increasingly been used to address neuronal and behavioural dysfunctions in response to prenatal infections. One commonly employed MIA model involves administering poly(I:C) (polyriboinosinic-polyribocytdilic acid), a synthetic analogue of double-stranded RNA, during gestation, which robustly induces an acute viral-like inflammatory response. Using electroencephalography (EEG) and infrared (IR) activity recordings, we explored alterations in sleep/wake, circadian and locomotor activity patterns on the adult male offspring of poly(I:C)-treated mothers. Our findings demonstrate that these offspring displayed reduced home cage activity during the (subjective) night under both light/dark or constant darkness conditions. In line with this finding, these mice exhibited an increase in non-rapid eye movement (NREM) sleep duration as well as an increase in sleep spindles density. Following sleep deprivation, poly(I:C)-exposed offspring extended NREM sleep duration and prolonged NREM sleep bouts during the dark phase as compared with non-exposed mice. Additionally, these mice exhibited a significant alteration in NREM sleep EEG spectral power under heightened sleep pressure. Together, our study highlights the lasting effects of infection and/or immune activation during pregnancy on circadian activity and sleep/wake patterns in the offspring.
Collapse
Affiliation(s)
- Waleed ElGrawani
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Flavia S Mueller
- Institute of Pharmacology and Toxicology, University of Zurich - Vetsuisse, Zurich, Switzerland
| | - Sina M Schalbetter
- Institute of Pharmacology and Toxicology, University of Zurich - Vetsuisse, Zurich, Switzerland
| | - Steven A Brown
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Ulrike Weber-Stadlbauer
- Institute of Pharmacology and Toxicology, University of Zurich - Vetsuisse, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Leila Tarokh
- Translational Research Center, University Hospital of Psychiatry and Psychotherapy, University of Bern, Bern, Switzerland
- University Hospital of Child and Adolescent Psychiatry and Psychotherapy, University of Bern, Bern, Switzerland
| |
Collapse
|
15
|
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
|
16
|
Pando P, Langen TJ, Kentner AC. Neighborly Influence: Intrauterine position accounts for individual variability in a mouse model of maternal immune activation. Brain Behav Immun 2024; 121:72-73. [PMID: 39043343 PMCID: PMC11380580 DOI: 10.1016/j.bbi.2024.07.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 07/20/2024] [Indexed: 07/25/2024] Open
Affiliation(s)
- Penelope Pando
- School of Arts & Sciences, Massachusetts College of Pharmacy and Health Sciences, 179 Longwood Ave, Boston, MA 02115, USA
| | - Tristen J Langen
- School of Arts & Sciences, Massachusetts College of Pharmacy and Health Sciences, 179 Longwood Ave, Boston, MA 02115, USA
| | - Amanda C Kentner
- School of Arts & Sciences, Massachusetts College of Pharmacy and Health Sciences, 179 Longwood Ave, Boston, MA 02115, USA.
| |
Collapse
|
17
|
Maddock RJ, Vlasova RM, Chen S, Iosif AM, Bennett J, Tanase C, Ryan AM, Murai T, Hogrefe CE, Schumann CD, Geschwind DH, Van de Water J, Amaral DG, Lesh TA, Styner MA, Kimberley McAllister A, Carter CS, Bauman MD. Altered brain metabolites in male nonhuman primate offspring exposed to maternal immune activation. Brain Behav Immun 2024; 121:280-290. [PMID: 39032543 DOI: 10.1016/j.bbi.2024.07.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 07/04/2024] [Accepted: 07/15/2024] [Indexed: 07/23/2024] Open
Abstract
Converging data show that exposure to maternal immune activation (MIA) in utero alters brain development in animals and increases the risk of neurodevelopmental disorders in humans. A recently developed non-human primate MIA model affords opportunities for studies with uniquely strong translational relevance to human neurodevelopment. The current longitudinal study used 1H-MRS to investigate the developmental trajectory of prefrontal cortex metabolites in male rhesus monkey offspring of dams (n = 14) exposed to a modified form of the inflammatory viral mimic, polyinosinic:polycytidylic acid (Poly IC), in the late first trimester. Brain metabolites in these animals were compared to offspring of dams that received saline (n = 10) or no injection (n = 4). N-acetylaspartate (NAA), glutamate, creatine, choline, myo-inositol, taurine, and glutathione were estimated from PRESS and MEGA-PRESS acquisitions obtained at 6, 12, 24, 36, and 45 months of age. Prior investigations of this cohort reported reduced frontal cortical gray and white matter and subtle cognitive impairments in MIA offspring. We hypothesized that the MIA-induced neurodevelopmental changes would extend to abnormal brain metabolite levels, which would be associated with the observed cognitive impairments. Prefrontal NAA was significantly higher in the MIA offspring across all ages (p < 0.001) and was associated with better performance on the two cognitive measures most sensitive to impairment in the MIA animals (both p < 0.05). Myo-inositol was significantly lower across all ages in MIA offspring but was not associated with cognitive performance. Taurine was elevated in MIA offspring at 36 and 45 months. Glutathione did not differ between groups. MIA exposure in male non-human primates is associated with altered prefrontal cortex metabolites during childhood and adolescence. A positive association between elevated NAA and cognitive performance suggests the hypothesis that elevated NAA throughout these developmental stages reflects a protective or resilience-related process in MIA-exposed offspring. The potential relevance of these findings to human neurodevelopmental disorders is discussed.
Collapse
Affiliation(s)
- Richard J Maddock
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California Davis, Sacramento, CA, USA.
| | - Roza M Vlasova
- Department of Psychiatry, University of North Carolina, Chapel Hill, NC, USA
| | - Shuai Chen
- Division of Biostatistics, Department of Public Health Sciences, School of Medicine, University of California Davis, Sacramento, CA, USA
| | - Ana-Maria Iosif
- Division of Biostatistics, Department of Public Health Sciences, School of Medicine, University of California Davis, Sacramento, CA, USA
| | - Jeffrey Bennett
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California Davis, Sacramento, CA, USA
| | - Costin Tanase
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California Davis, Sacramento, CA, USA
| | - Amy M Ryan
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California Davis, Sacramento, CA, USA
| | - Takeshi Murai
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California Davis, Sacramento, CA, USA
| | - Casey E Hogrefe
- California National Primate Research Center, University of California Davis, Davis, CA, USA
| | - Cynthia D Schumann
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California Davis, Sacramento, CA, USA
| | - Daniel H Geschwind
- Neurogenetics Program, Department of Neurology, University of California Los Angeles, Los Angeles, CA, USA
| | - Judy Van de Water
- Rheumatology/Allergy and Clinical Immunology, School of Medicine, University of California Davis, Sacramento, CA, USA; MIND Institute, School of Medicine, University of California Davis, Sacramento, CA, USA
| | - David G Amaral
- MIND Institute, School of Medicine, University of California Davis, Sacramento, CA, USA
| | - Tyler A Lesh
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California Davis, Sacramento, CA, USA
| | - Martin A Styner
- Department of Psychiatry, University of North Carolina, Chapel Hill, NC, USA; Department of Computer Science, University of North Carolina, Chapel Hill, NC, USA
| | | | - Cameron S Carter
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California Davis, Sacramento, CA, USA.
| | - Melissa D Bauman
- California National Primate Research Center, University of California Davis, Davis, CA, USA; MIND Institute, School of Medicine, University of California Davis, Sacramento, CA, USA; Physiology and Membrane Biology, School of Medicine, University of California Davis, Sacramento, CA, USA.
| |
Collapse
|
18
|
Armario A, Nadal R, Fuentes S, Visa J, Belda X, Serrano S, Labad J. Prenatal immune activation in rats and adult exposure to inescapable shocks reveal sex-dependent effects on fear conditioning that might be relevant for schizophrenia. Psychiatry Res 2024; 342:116219. [PMID: 39388806 DOI: 10.1016/j.psychres.2024.116219] [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: 05/13/2024] [Revised: 09/18/2024] [Accepted: 09/29/2024] [Indexed: 10/12/2024]
Abstract
Prenatal infection is considered a relevant factor for neurodevelopmental alterations and psychiatric diseases. Administration of bacterial and viral components during pregnancy in rodents results in maternal immune activation (MIA), leading to schizophrenia-like neurochemical and behavioral changes. Despite some evidence for abnormal fear conditioning in schizophrenia, only a few animal studies have focused on this issue. Therefore, we addressed the impact of the administration of the viral mimetic polyI:C to pregnant Long-Evans rats on the adult offspring response to inescapable shocks (IS) and contextual fear conditioning. In males, polyI:C induced a greater endocrine (plasma ACTH) response to IS and both polyI:C and IS enhanced fear conditioning and generalization to a completely different novel environment (hole-board), with no additive effects, probably due to a ceiling effect. In contrast, a modest impact of polyI:C and a lower impact of IS on contextual fear conditioning and generalization was observed in females. Thus, the present results demonstrate that polyI:C dramatically affected fear response to IS in adult males and support the hypothesis that males are more sensitive than females to this treatment. This model might allow to explore neurobiological mechanisms underlying abnormal responsiveness to fear conditioning and stressors in schizophrenia.
Collapse
Affiliation(s)
- Antonio Armario
- Institut de Neurociències, Universitat Autònoma de Barcelona, Spain; Animal Physiology Unit, Faculty of Biosciences, Department of Cellular Biology, Physiology and Imunology, Universitat Autònoma de Barcelona, Spain; Unitat de Neurociència Traslacional, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí I3PT, Universitat Autònoma de Barcelona, Spain; CIBERSAM, Instituto de Salud Carlos III, Spain.
| | - Roser Nadal
- Institut de Neurociències, Universitat Autònoma de Barcelona, Spain; Deparment of Psychobiology and Methodology of Health Sciences, Faculty of Psychology, Universitat Autònoma de Barcelona, Spain; Unitat de Neurociència Traslacional, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí I3PT, Universitat Autònoma de Barcelona, Spain; CIBERSAM, Instituto de Salud Carlos III, Spain
| | - Silvia Fuentes
- Institut de Neurociències, Universitat Autònoma de Barcelona, Spain; Deparment of Psychobiology and Methodology of Health Sciences, Faculty of Psychology, Universitat Autònoma de Barcelona, Spain
| | - Joan Visa
- Institut de Neurociències, Universitat Autònoma de Barcelona, Spain
| | - Xavier Belda
- Institut de Neurociències, Universitat Autònoma de Barcelona, Spain; Animal Physiology Unit, Faculty of Biosciences, Department of Cellular Biology, Physiology and Imunology, Universitat Autònoma de Barcelona, Spain
| | - Sara Serrano
- Institut de Neurociències, Universitat Autònoma de Barcelona, Spain; Animal Physiology Unit, Faculty of Biosciences, Department of Cellular Biology, Physiology and Imunology, Universitat Autònoma de Barcelona, Spain
| | - Javier Labad
- Department of Mental Health and Addictions, Consorci Sanitari del Maresme, Mataró, Spain; Unitat de Neurociència Traslacional, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí I3PT, Universitat Autònoma de Barcelona, Spain; CIBERSAM, Instituto de Salud Carlos III, Spain
| |
Collapse
|
19
|
Schaer R, Mueller FS, Notter T, Weber-Stadlbauer U, Meyer U. Intrauterine position effects in a mouse model of maternal immune activation. Brain Behav Immun 2024; 120:391-402. [PMID: 38897330 DOI: 10.1016/j.bbi.2024.06.015] [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: 01/28/2024] [Revised: 05/27/2024] [Accepted: 06/16/2024] [Indexed: 06/21/2024] Open
Abstract
Rodent models of maternal immune activation (MIA) are increasingly used as experimental tools in preclinical research of immune-mediated neurodevelopmental disorders and mental illnesses. Using a viral-like MIA model that is based on prenatal poly(I:C) exposure in mice, we have recently identified the existence of subgroups of MIA-exposed offspring that show dissociable behavioral, transcriptional, brain network and inflammatory profiles even under conditions of genetic homogeneity and identical MIA. Here, we tested the hypothesis that the intrauterine positions of fetuses, which are known to shape individual variability in litter-bearing mammals through variations in fetal hormone exposure, may contribute to the variable outcomes of MIA in mice. MIA was induced by maternal administration of poly(I:C) on gestation day 12 in C57BL/6N mice. Determining intrauterine positions using delivery by Cesarean section (C-section), we found that MIA-exposed offspring developing between female fetuses only (0M-MIA offspring) displayed significant deficits in sociability and sensorimotor gating at adult age, whereas MIA-exposed offspring developing between one or two males in utero (1/2M-MIA offspring) did not show the same deficits. These intrauterine position effects similarly emerged in male and female offspring. Furthermore, while MIA elevated fetal brain levels of pro- and anti-inflammatory cytokines independently of the precise intrauterine position and sex of adjacent fetuses during the acute phase, fetal brain levels of TNF-α remained elevated in 0M-MIA but not 1/2M-MIA offspring until the post-acute phase in late gestation. As expected, 1/2M offspring generally showed higher testosterone levels in the fetal brain during late gestation as compared to 0M offspring, confirming the transfer of testosterone from male fetuses to adjacent male or female fetuses. Taken together, our findings identify a novel source of within-litter variability contributing to heterogeneous outcomes of short- and long-term effects in a mouse model of MIA. In broader context, our findings highlight that individual differences in fetal exposure to hormonal and inflammatory signals may be a perinatal factor that shapes risk and resilience to MIA.
Collapse
Affiliation(s)
- Ron Schaer
- Institute of Veterinary Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Flavia S Mueller
- Institute of Veterinary Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Tina Notter
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland; Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Ulrike Weber-Stadlbauer
- Institute of Veterinary Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland; Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Urs Meyer
- Institute of Veterinary Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland; Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
20
|
Mattei D, Ivanov A, Hammer J, Ugursu B, Schalbetter S, Richetto J, Weber-Stadlbauer U, Mueller F, Scarborough J, Wolf SA, Kettenmann H, Wollscheid B, Beule D, Meyer U. Microglia undergo molecular and functional adaptations to dark and light phases in male laboratory mice. Brain Behav Immun 2024; 120:571-583. [PMID: 38986723 DOI: 10.1016/j.bbi.2024.07.007] [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: 04/25/2024] [Revised: 06/20/2024] [Accepted: 07/06/2024] [Indexed: 07/12/2024] Open
Abstract
Microglia are increasingly recognized to contribute to brain health and disease. Preclinical studies using laboratory rodents are essential to advance our understanding of the physiological and pathophysiological roles of these cells in the central nervous system. Rodents are nocturnal animals, and they are mostly maintained in a defined light-dark cycle within animal facilities, with many laboratories investigating the molecular and functional profiles of microglia exclusively during the animals' light (sleep) phase. However, only a few studies have considered possible differences in microglial functions between the active and sleep phases. Based on initial evidence suggesting that microglial intrinsic clock genes can affect their phenotypes, we sought to investigate differences in transcriptional, proteotype and functional profiles of microglia between light (sleep) and dark (active) phases, and how these changes are affected in pathological models. We found marked transcriptional and proteotype differences between microglia harvested from male mice during the light or dark phase. Amongst others, these differences related to genes and proteins associated with immune responses, motility, and phagocytosis, which were reflected by functional alterations in microglial synaptic pruning and response to bacterial stimuli. Possibly accounting for such changes, we found RNA and protein regulation in SWI/SNF and NuRD chromatin remodeling complexes between light and dark phases. Importantly, we also show that the time of microglial sample collection influences the nature of microglial transcriptomic changes in a model of immune-mediated neurodevelopmental disorders. Our findings emphasize the importance of considering diurnal factors in studying microglial cells and indicate that implementing a circadian perspective is pivotal for advancing our understanding of their physiological and pathophysiological roles in brain health and disease.
Collapse
Affiliation(s)
- Daniele Mattei
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, 8057 Zurich, Switzerland; Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America.
| | - Andranik Ivanov
- Core Unit Bioinformatics, Berlin Institute of Health, Charité-Universitätsmedizin, Berlin, Germany
| | - Jacqueline Hammer
- Institute of Molecular Systems Biology and Department for Health Sciences and Technology, ETH Zürich, Switzerland
| | - Bilge Ugursu
- Cellular Neuroscience, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany; Department of Ophthalmology, Charité - Universitätsmedizin Berlin, Germany; Psychoneuroimmunology, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Sina Schalbetter
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, 8057 Zurich, Switzerland
| | - Juliet Richetto
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, 8057 Zurich, Switzerland
| | - Ulrike Weber-Stadlbauer
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, 8057 Zurich, Switzerland
| | - Flavia Mueller
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, 8057 Zurich, Switzerland
| | - Joseph Scarborough
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, 8057 Zurich, Switzerland
| | - Susanne A Wolf
- Cellular Neuroscience, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany; Department of Ophthalmology, Charité - Universitätsmedizin Berlin, Germany; Psychoneuroimmunology, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Helmut Kettenmann
- Cellular Neuroscience, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany; Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Bernd Wollscheid
- Institute of Molecular Systems Biology and Department for Health Sciences and Technology, ETH Zürich, Switzerland
| | - Dieter Beule
- Core Unit Bioinformatics, Berlin Institute of Health, Charité-Universitätsmedizin, Berlin, Germany
| | - Urs Meyer
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, 8057 Zurich, Switzerland; Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
21
|
Vasileva S, Yap CX, Whitehouse AJ, Gratten J, Eyles D. Absence of association between maternal adverse events and long-term gut microbiome outcomes in the Australian autism biobank. Brain Behav Immun Health 2024; 39:100814. [PMID: 39027090 PMCID: PMC11254947 DOI: 10.1016/j.bbih.2024.100814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 06/16/2024] [Indexed: 07/20/2024] Open
Abstract
Introduction Maternal immune activation (MIA) and prenatal maternal stress (MatS) are well-studied risk factors for psychiatric conditions such as autism and schizophrenia. Animal studies have proposed the gut microbiome as a mechanism underlying this association and have found that risk factor-related gut microbiome alterations persist in the adult offspring. In this cross-sectional study, we assessed whether maternal immune activation and prenatal maternal stress were associated with long-term gut microbiome alterations in children using shotgun metagenomics. Methods This cross-sectional study included children diagnosed with autism (N = 92), siblings without a diagnosis (N = 42), and unrelated children (N = 40) without a diagnosis who were recruited into the Australian Autism Biobank and provided a faecal sample. MIA exposure was inferred from self-reported data and included asthma/allergies, complications during pregnancy triggering an immune response, auto-immune conditions, and acute inflammation. Maternal stress included any of up to 9 stressful life events during pregnancy, such as divorce, job loss, and money problems. Data were analysed for a total of 174 children, of whom 63 (36%) were born to mothers with MIA and 84 (48%) were born to mothers who experienced maternal stress during pregnancy (where 33 [19%] experienced both). Gut microbiome data was assessed using shotgun metagenomic sequencing of the children's faecal samples. Results In our cohort, MIA, but not MatS, was associated with ASD. Variance component analysis revealed no associations between any of the gut microbiome datasets and neither MIA nor MatS. After adjusting for age, sex, diet and autism diagnosis, there was no significant difference between groups for bacterial richness, α-diversity or β-diversity. We found no significant differences in species abundance in the main analyses. However, when stratifying the cohort by age, we found that Faecalibacterium prausnitzii E was significantly decreased in MIA children aged 11-17. Discussion Consistent with previous findings, we found that children who were born to mothers with MIA were more likely to be diagnosed with autism. Unlike within animal studies, we found negligible microbiome differences associated with MIA and maternal stress. Given the current interest in the microbiome-gut-brain axis, researchers should exercise caution in translating microbiome findings from animal models to human contexts and the clinical setting.
Collapse
Affiliation(s)
- Svetlina Vasileva
- Queensland Brain Institute, University of Queensland, Brisbane, Australia
| | - Chloe X. Yap
- Mater Research Institute, The University of Queensland, Translational Research Institute, Brisbane, Australia
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
- Cooperative Research Centre for Living with Autism (Autism CRC), Long Pocket, Brisbane, Australia
| | | | - Jacob Gratten
- Mater Research Institute, The University of Queensland, Translational Research Institute, Brisbane, Australia
- Cooperative Research Centre for Living with Autism (Autism CRC), Long Pocket, Brisbane, Australia
| | - Darryl Eyles
- Queensland Brain Institute, University of Queensland, Brisbane, Australia
- Queensland Centre for Mental Health Research, Wacol, Australia
| |
Collapse
|
22
|
Liong S, Choy KHC, De Luca SN, Liong F, Coward-Smith M, Oseghale O, Miles MA, Vlahos R, Valant C, Nithianantharajah J, Pantelis C, Christopoulos A, Selemidis S. Brain region-specific alterations in gene expression trajectories in the offspring born from influenza A virus infected mice. Brain Behav Immun 2024; 120:488-498. [PMID: 38925418 DOI: 10.1016/j.bbi.2024.06.025] [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: 03/13/2024] [Revised: 06/04/2024] [Accepted: 06/23/2024] [Indexed: 06/28/2024] Open
Abstract
Influenza A virus (IAV) infection during pregnancy can increase the risk for neurodevelopmental disorders in the offspring, however, the underlying neurobiological mechanisms are largely unknown. To recapitulate viral infection, preclinical studies have traditionally focused on using synthetic viral mimetics, rather than live IAV, to examine consequences of maternal immune activation (MIA)-dependent processes on offspring. In contrast, few studies have used live IAV to assess effects on global gene expression, and none to date have addressed whether moderate IAV, mimicking seasonal influenza disease, alters normal gene expression trajectories in different brain regions across different stages of development. Herein, we show that moderate IAV infection during pregnancy, which causes mild maternal disease and no overt foetal complications in utero, induces lasting effects on the offspring into adulthood. We observed behavioural changes in adult offspring, including disrupted prepulse inhibition, dopaminergic hyper-responsiveness, and spatial recognition memory deficits. Gene profiling in the offspring brain from neonate to adolescence revealed persistent alterations to normal gene expression trajectories in the prefronal cortex, hippocampus, hypothalamus and cerebellum. Alterations were found in genes involved in inflammation and neurogenesis, which were predominately dysregulated in neonatal and early adolescent offspring. Notably, late adolescent offspring born from IAV infected mice displayed altered microglial morphology in the hippocampus. In conclusion, we show that moderate IAV during pregnancy perturbs neurodevelopmental trajectories in the offspring, including alterations in the neuroinflammatory gene expression profile and microglial number and morphology in the hippocampus, resulting in behavioural changes in adult offspring. Such early perturbations may underlie the vulnerability in human offspring for the later development of neurodevelopmental disorders, including schizophrenia. Our work highlights the importance of using live IAV in developing novel preclinical models that better recapitulate the real-world impact of inflammatory insults during pregnancy on offspring neurodevelopmental trajectories and disease susceptibility later in life.
Collapse
Affiliation(s)
- Stella Liong
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - K H Christopher Choy
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia
| | - Simone N De Luca
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - Felicia Liong
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - Madison Coward-Smith
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - Osezua Oseghale
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - Mark A Miles
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - Ross Vlahos
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - Celine Valant
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia
| | - Jess Nithianantharajah
- The Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia; Department of Florey Neuroscience, University of Melbourne, Melbourne, VIC, Australia.
| | - Christos Pantelis
- The Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia; Melbourne Neuropsychiatry Centre, Department of Psychiatry, University of Melbourne, Melbourne, VIC, Australia; Neuromedicines Discovery Centre, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia.
| | - Arthur Christopoulos
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia; ARC Centre for Cryo-electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia; Neuromedicines Discovery Centre, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia.
| | - Stavros Selemidis
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia.
| |
Collapse
|
23
|
Tillmann KE, Schaer R, Mueller FS, Mueller K, Voelkl B, Weber-Stadlbauer U, Pollak DD. Differential effects of purified low molecular weight Poly(I:C) in the maternal immune activation model depend on the laboratory environment. Transl Psychiatry 2024; 14:300. [PMID: 39033141 PMCID: PMC11271296 DOI: 10.1038/s41398-024-03014-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 06/27/2024] [Accepted: 07/04/2024] [Indexed: 07/23/2024] Open
Abstract
The Poly (I:C) (polyriboinosinic-polyribocytidilic acid) paradigm of maternal immune activation (MIA) is most widely used as experimental model for the evaluation of the effects of gestational infection on the brain and behavior of the progeny. We have previously reported significant batch-to-batch variability in the effects of Poly (I:C), purchased from the same supplier (Sigma-Aldrich), on maternal and fetal immune responses and found these differences to be dependent on the relative amount of synthetic double-stranded RNA fragments in the high versus low molecular weight (LMW) range contained in the compound. We here resorted to Poly (I:C) purified for LMW dsRNA fragments to establish a MIA paradigm with increased reproducibility and enhanced standardization in an effort to refine the MIA paradigm and characterize its effect on offspring behavior. We found that the parallel application of LMW Poly (I:C) in two different MIA-experienced laboratories (Vienna and Zurich) yielded differential outcomes in terms of maternal immune responses and behavioral phenotypes in the offspring generation. In both experimental sites, administration of LMW Poly (I:C) induced a significant sickness response and cytokine induction in the pregnant dam and fetal brains, while the expected deficit in sociability as one main behavioral outcome parameter in the MIA progeny, was only present in the Zurich, but not the Vienna cohort. We conclude that although using Poly (I:C) purified for a defined molecular weight range reduces batch-to-batch variability, it does not make the MIA model more reliable and robust. The differential response in behavioral phenotypes of the MIA offspring between the two laboratories illustrates the highly complex interaction between prenatal and postnatal milieus - including the laboratory environment - that determine offspring phenotypic outcomes after MIA. Consequently, establishing a new MIA protocol or implementing the MIA model firstly under new or changed environmental conditions must include the assessment of offspring behavior to ensure solid and reproducible experimental outcomes.
Collapse
Affiliation(s)
- Katharina E Tillmann
- Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Vienna, Austria
| | - Ron Schaer
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland
| | - Flavia S Mueller
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland
| | - Karin Mueller
- Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Vienna, Austria
| | - Bernhard Voelkl
- Animal Welfare Division, Veterinary Public Health Institute University of Bern, Bern, Switzerland
| | - Ulrike Weber-Stadlbauer
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland.
- Neuroscience Center Zurich, University of Zurich and ETH, Winterthurerstrasse 190, 8057, Zurich, Switzerland.
| | - Daniela D Pollak
- Department of Neurophysiology and Neuropharmacology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
24
|
Schneider Gasser EM, Schaer R, Mueller FS, Bernhardt AC, Lin HY, Arias-Reyes C, Weber-Stadlbauer U. Prenatal immune activation in mice induces long-term alterations in brain mitochondrial function. Transl Psychiatry 2024; 14:289. [PMID: 39009558 PMCID: PMC11251165 DOI: 10.1038/s41398-024-03010-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 06/23/2024] [Accepted: 07/03/2024] [Indexed: 07/17/2024] Open
Abstract
Prenatal exposure to infections is a risk factor for neurodevelopmental disorders in offspring, and alterations in mitochondrial function are discussed as a potential underlying factor. Here, using a mouse model of viral-like maternal immune activation (MIA) based on poly(I:C) (POL) treatment at gestational day (GD) 12, we show that adult offspring exhibit behavioral deficits, such as reduced levels of social interaction. In addition, we found increased nicotinamidadenindinucleotid (NADH)- and succinate-linked mitochondrial respiration and maximal electron transfer capacity in the prefrontal cortex (PFC) and in the amygdala (AMY) of males and females. The increase in respiratory capacity resulted from an increase in mitochondrial mass in neurons (as measured by complex IV activity and transcript expression), presumably to compensate for a reduction in mitochondrion-specific respiration. Moreover, in the PFC of control (CON) male offspring a higher excess capacity compared to females was observed, which was significantly reduced in the POL-exposed male offspring, and, along with a higher leak respiration, resulted in a lower mitochondrial coupling efficiency. Transcript expression of the uncoupling proteins (UCP4 and UCP5) showed a reduction in the PFC of POL male mice, suggesting mitochondrial dysfunction. In addition, in the PFC of CON females, a higher expression of the antioxidant enzyme superoxide dismutase (SOD1) was observed, suggesting a higher antioxidant capacity as compared to males. Finally, transcripts analysis of genes involved in mitochondrial biogenesis and dynamics showed reduced expression of fission/fusion transcripts in PFC of POL offspring of both sexes. In conclusion, we show that MIA causes alterations in neuronal mitochondrial function and mass in the PFC and AMY of adult offspring with some effects differing between males and females.
Collapse
Affiliation(s)
- Edith M Schneider Gasser
- Institute of Veterinary Pharmacology and Toxicology, Vetsuisse Faculty, University of Zurich, Zurich, 8057, Switzerland.
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zurich, 8057, Switzerland.
- Department of Pediatrics, Faculty of Medicine, Université Laval, Québec, QC, Canada.
- Neuroscience Center Zurich, University of Zurich, and ETH, Zurich, 8057, Switzerland.
| | - Ron Schaer
- Institute of Veterinary Pharmacology and Toxicology, Vetsuisse Faculty, University of Zurich, Zurich, 8057, Switzerland
| | - Flavia S Mueller
- Institute of Veterinary Pharmacology and Toxicology, Vetsuisse Faculty, University of Zurich, Zurich, 8057, Switzerland
| | - Alexandra C Bernhardt
- Institute of Veterinary Pharmacology and Toxicology, Vetsuisse Faculty, University of Zurich, Zurich, 8057, Switzerland
| | - Han-Yu Lin
- Institute of Veterinary Pharmacology and Toxicology, Vetsuisse Faculty, University of Zurich, Zurich, 8057, Switzerland
| | | | - Ulrike Weber-Stadlbauer
- Institute of Veterinary Pharmacology and Toxicology, Vetsuisse Faculty, University of Zurich, Zurich, 8057, Switzerland
- Neuroscience Center Zurich, University of Zurich, and ETH, Zurich, 8057, Switzerland
| |
Collapse
|
25
|
Liao H, Lu D, Reisinger SN, Kleeman EA, van de Garde N, Gubert C, Hannan AJ. Mimicking bacterial infection in male mice changes sperm small RNA profiles and multigenerationally alters offspring behavior and physiology. Brain Behav Immun 2024; 119:520-538. [PMID: 38636562 DOI: 10.1016/j.bbi.2024.04.017] [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: 12/21/2023] [Revised: 03/22/2024] [Accepted: 04/16/2024] [Indexed: 04/20/2024] Open
Abstract
Paternal pre-conceptual exposures, including stress, diet, substance abuse, parasite infection, and viral immune activation via Poly I:C, have been reported to influence the brains and behavior of offspring through sperm epigenetic changes. However, the effects of paternal (F0) pre-conceptual exposure to bacterial-induced immune activation on the behavior and physiology of F1 and F2 generations remain unexplored. We examined this using C57BL/6J mice. Eight-week-old males (F0) received a single intraperitoneal injection of the bacterial mimetic lipopolysaccharide (LPS: 5 mg/kg) or 0.9 % saline (vehicle control) before mating with naïve females at four weeks post-injection. Comprehensive behavioral assessments were conducted to investigate anxiety, social behaviors, depressive-like behaviors and cognition in both the F1 and F2 generations within the age range of 8 to 14 weeks. Results demonstrated that only female offspring of LPS-exposed fathers exhibited reduced anxiety levels in the light/dark box, large open field, and novelty-suppressed feeding test. These F1 female offspring also exhibited heightened sociability in the 3-chambered social interaction test and a reduced preference for saccharin in the saccharin preference test. Additionally, the F1 male offspring of LPS-challenged males demonstrated an increased total distance traveled in the light/dark box and a longer distance covered in the light zone. They also exhibited diminished preference for social novelty in the 3-chambered social interaction test and an elevated novel arm preference index in the Y-maze. In the F2 generation, male descendants of LPS-treated fathers showed reduced latency to feed in the novelty-suppressed feeding test. Additionally, the F2 generation of LPS-challenged fathers, but not the F1 generation, displayed enhanced immune response in both sexes after an acute LPS immune challenge (5 mg/kg). Analysis of sperm small noncoding RNA profiles from LPS-treated F0 mice revealed significant changes at 4 weeks after administration of LPS. These changes included three microRNAs, eight PIWI-interacting RNAs, and two transfer RNAs, exhibiting significant upregulation (mmu-miR-146a-5p, mmu-piR-27082 and mmu-piR-29102) or downregulation (mmu-miR-5110, mmu-miR-467e-3p, mmu-piR-22583, mmu-piR-23548, mmu-piR-36341, mmu-piR-50293, mmu-piR-16583, mmu-piR-36507, Mus_musculus_tRNA-Ile-AAT-2-1 and Mus_musculus_tRNA-Tyr-GTA-1-1). Additionally, we detected 52 upregulated small noncoding RNAs (including 9 miRNAs, 41 piRNAs, and 2 tRNAs) and 7 downregulated small noncoding RNAs (3 miRNAs, 3 piRNAs, and 1 tRNA) in the sperm of F1 offspring from LPS-treated males. These findings provide compelling evidence for the involvement of epigenetic mechanisms in the modulation of brain function and immunity, and associated behavioral and immunological traits, across generations, in response to bacterial infection.
Collapse
Affiliation(s)
- Huan Liao
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia.
| | - Da Lu
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Sonali N Reisinger
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Elizabeth A Kleeman
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Nicholas van de Garde
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Carolina Gubert
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Anthony J Hannan
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia; Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia.
| |
Collapse
|
26
|
Hope H, Pierce M, Gabr H, Radojčić MR, Swift E, Taxiarchi VP, Abel KM. The causal association between maternal depression, anxiety, and infection in pregnancy and neurodevelopmental disorders among 410 461 children: a population study using quasi-negative control cohorts and sibling analysis. Psychol Med 2024; 54:1693-1701. [PMID: 38205522 DOI: 10.1017/s0033291723003604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2024]
Abstract
BACKGROUND To address if the long-standing association between maternal infection, depression/anxiety in pregnancy, and offspring neurodevelopmental disorder (NDD) is causal, we conducted two negative-control studies. METHODS Four primary care cohorts of UK children (pregnancy, 1 and 2 years prior to pregnancy, and siblings) born between 1 January 1990 and 31 December 2017 were constructed. NDD included autism/autism spectrum disorder, attention-deficit/hyperactivity disorder, intellectual disability, cerebral palsy, and epilepsy. Maternal exposures included depression/anxiety and/or infection. Maternal (age, smoking status, comorbidities, body mass index, NDD); child (gender, ethnicity, birth year); and area-level (region and level of deprivation) confounders were captured. The NDD incidence rate among (1) children exposed during or outside of pregnancy and (2) siblings discordant for exposure in pregnancy was compared using Cox-regression models, unadjusted and adjusted for confounders. RESULTS The analysis included 410 461 children of 297 426 mothers and 2 793 018 person-years of follow-up with 8900 NDD cases (incidence rate = 3.2/1000 person years). After adjustments, depression and anxiety consistently associated with NDD (pregnancy-adjusted HR = 1.58, 95% CI 1.46-1.72; 1-year adj. HR = 1.49, 95% CI 1.39-1.60; 2-year adj. HR = 1.62, 95% CI 1.50-1.74); and to a lesser extent, of infection (pregnancy adj. HR = 1.16, 95% CI 1.10-1.22; 1-year adj. HR = 1.20, 95% CI 1.14-1.27; 2-year adj. HR = 1.19, 95% CI 1.12-1.25). NDD risk did not differ among siblings discordant for pregnancy exposure to mental illness HR = 0.97, 95% CI 0.77-1.21 or infection HR = 0.99, 95% CI 0.90-1.08. CONCLUSIONS Maternal risk appears to be unspecific to pregnancy: our study provided no evidence of a specific, and therefore causal, link between in-utero exposure to infection, common mental illness, and later development of NDD.
Collapse
Affiliation(s)
- Holly Hope
- Centre for Women's Mental Health, Division of Psychology and Mental Health, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Matthias Pierce
- Centre for Women's Mental Health, Division of Psychology and Mental Health, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Hend Gabr
- Centre for Women's Mental Health, Division of Psychology and Mental Health, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Department of Mathematics, Insurance, and Statistics, Faculty of Commerce, Menoufia University, Shebeen El-Kom, Menoufia, Egypt
| | - Maja R Radojčić
- Centre for Women's Mental Health, Division of Psychology and Mental Health, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Eleanor Swift
- Centre for Women's Mental Health, Division of Psychology and Mental Health, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Greater Manchester Mental Health NHS Foundation Trust, Manchester, UK
| | - Vicky P Taxiarchi
- Centre for Women's Mental Health, Division of Psychology and Mental Health, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Kathryn M Abel
- Centre for Women's Mental Health, Division of Psychology and Mental Health, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Greater Manchester Mental Health NHS Foundation Trust, Manchester, UK
| |
Collapse
|
27
|
Buonfiglioli A, Kübler R, Missall R, De Jong R, Chan S, Haage V, Wendt S, Lin AJ, Mattei D, Graziani M, Latour B, Gigase F, Nygaard HB, De Jager PL, De Witte LD. A microglia-containing cerebral organoid model to study early life immune challenges. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.24.595814. [PMID: 38826204 PMCID: PMC11142229 DOI: 10.1101/2024.05.24.595814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Prenatal infections and activation of the maternal immune system have been proposed to contribute to causing neurodevelopmental disorders (NDDs), chronic conditions often linked to brain abnormalities. Microglia are the resident immune cells of the brain and play a key role in neurodevelopment. Disruption of microglial functions can lead to brain abnormalities and increase the risk of developing NDDs. How the maternal as well as the fetal immune system affect human neurodevelopment and contribute to NDDs remains unclear. An important reason for this knowledge gap is the fact that the impact of exposure to prenatal risk factors has been challenging to study in the human context. Here, we characterized a model of cerebral organoids (CO) with integrated microglia (COiMg). These organoids express typical microglial markers and respond to inflammatory stimuli. The presence of microglia influences cerebral organoid development, including cell density and neural differentiation, and regulates the expression of several ciliated mesenchymal cell markers. Moreover, COiMg and organoids without microglia show similar but also distinct responses to inflammatory stimuli. Additionally, IFN-γ induced significant transcriptional and structural changes in the cerebral organoids, that appear to be regulated by the presence of microglia. Specifically, interferon-gamma (IFN-γ) was found to alter the expression of genes linked to autism. This model provides a valuable tool to study how inflammatory perturbations and microglial presence affect neurodevelopmental processes.
Collapse
Affiliation(s)
- Alice Buonfiglioli
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Raphael Kübler
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Human Genetics, Radboud UMC, Nijmegen, The Netherlands
| | - Roy Missall
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Renske De Jong
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Stephanie Chan
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Verena Haage
- Center for Translational & Computational Neuroimmunology, Department of Neurology and the Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Stefan Wendt
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6T 1Z3, Canada
| | - Ada J. Lin
- Division of Neurology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6T 1Z3, Canada
| | - Daniele Mattei
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mara Graziani
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Human Genetics, Radboud UMC, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, 6500 HB, Nijmegen, The Netherlands
| | - Brooke Latour
- Department of Human Genetics, Radboud UMC, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, 6500 HB, Nijmegen, The Netherlands
| | - Frederieke Gigase
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Haakon B. Nygaard
- Division of Neurology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6T 1Z3, Canada
| | - Philip L. De Jager
- Center for Translational & Computational Neuroimmunology, Department of Neurology and the Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Lot D. De Witte
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Human Genetics, Radboud UMC, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, 6500 HB, Nijmegen, The Netherlands
- Department of Psychiatry, Radboud UMC, Nijmegen, The Netherlands
| |
Collapse
|
28
|
Lipner E, Mac Giollabhui N, Breen EC, Cohn BA, Krigbaum NY, Cirillo PM, Olino TM, Alloy LB, Ellman LM. Sex-Specific Pathways From Prenatal Maternal Inflammation to Adolescent Depressive Symptoms. JAMA Psychiatry 2024; 81:498-505. [PMID: 38324324 PMCID: PMC10851141 DOI: 10.1001/jamapsychiatry.2023.5458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 10/30/2023] [Indexed: 02/08/2024]
Abstract
Importance Prenatal maternal inflammation has been associated with major depressive disorder in offspring in adulthood as well as with internalizing and externalizing symptoms in childhood; however, the association between prenatal inflammation and offspring depression in adolescence has yet to be examined. Objective To determine whether maternal levels of inflammatory biomarkers during pregnancy are associated with depressive symptomatology in adolescent-aged offspring and to examine how gestational timing, offspring sex, and childhood psychiatric symptoms impact these associations. Design, Setting, and Participants This was an observational study of a population-based birth cohort from the Child Health and Development Studies (CHDS), which recruited almost all mothers receiving obstetric care from the Kaiser Foundation Health Plan (KFHP) in Alameda County, California, between June 1959 and September 1966. Pregnancy data and blood sera were collected from mothers, and offspring psychiatric symptom data were collected in childhood (ages 9-11 years) and adolescence (ages 15-17 years). Mother-offspring dyads with available maternal prenatal inflammatory biomarkers during first and/or second trimesters and offspring depressive symptom data at adolescent follow-up were included. Data analyses took place between March 2020 and June 2023. Exposures Levels of inflammatory biomarkers (interleukin 6 [IL-6], IL-8, IL-1 receptor antagonist [IL-1RA], and soluble tumor necrosis factor receptor-II) assayed from maternal sera in the first and second trimesters of pregnancy. Main Outcomes and Measures Self-reported depressive symptoms at adolescent follow-up. Results A total of 674 mothers (mean [SD] age, 28.1 [5.9] years) and their offspring (350 male and 325 female) were included in this study. Higher second trimester IL-6 was significantly associated with greater depressive symptoms in offspring during adolescence (b, 0.57; SE, 0.26); P = .03). Moderated mediation analyses showed that childhood externalizing symptoms significantly mediated the association between first trimester IL-6 and adolescent depressive symptoms in male offspring (b, 0.18; 95% CI, 0.02-0.47), while childhood internalizing symptoms mediated the association between second trimester IL-1RA and adolescent depressive symptoms in female offspring (b, 0.80; 95% CI, 0.19-1.75). Conclusions and Relevance In this study, prenatal maternal inflammation was associated with depressive symptoms in adolescent-aged offspring. The findings of the study suggest that pathways to adolescent depressive symptomatology from prenatal risk factors may differ based on both the timing of exposure to prenatal inflammation and offspring sex.
Collapse
Affiliation(s)
- Emily Lipner
- Department of Psychology and Neuroscience, Temple University, Philadelphia, Pennsylvania
| | - Naoise Mac Giollabhui
- Department of Psychology and Neuroscience, Temple University, Philadelphia, Pennsylvania
- Department of Psychiatry, Massachusetts General Hospital, Boston
| | - Elizabeth C. Breen
- Cousins Center for Psychoneuroimmunology, Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles
| | - Barbara A. Cohn
- Child Health and Development Studies, Public Health Institute, Berkeley, California
| | - Nickilou Y. Krigbaum
- Child Health and Development Studies, Public Health Institute, Berkeley, California
| | - Piera M. Cirillo
- Child Health and Development Studies, Public Health Institute, Berkeley, California
| | - Thomas M. Olino
- Department of Psychology and Neuroscience, Temple University, Philadelphia, Pennsylvania
| | - Lauren B. Alloy
- Department of Psychology and Neuroscience, Temple University, Philadelphia, Pennsylvania
| | - Lauren M. Ellman
- Department of Psychology and Neuroscience, Temple University, Philadelphia, Pennsylvania
| |
Collapse
|
29
|
Duko B, Gebremedhin AT, Tessema GA, Dunne J, Alati R, Pereira G. The effects of pre-eclampsia on social and emotional developmental vulnerability in children at age five in Western Australia: A population data linkage study. J Affect Disord 2024; 352:349-356. [PMID: 38360367 DOI: 10.1016/j.jad.2024.02.042] [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: 03/24/2023] [Revised: 02/07/2024] [Accepted: 02/12/2024] [Indexed: 02/17/2024]
Abstract
BACKGROUND It is important to explore factors that may hinder early childhood development in AEDC Emotional Maturity and Social Competence domains as these underpin the foundation for health, well-being, and productivity over the life course. No previous study has examined whether, or to what extent, preeclampsia increases the risk of developmental vulnerability in social and emotional domains in early childhood. METHODS We conducted a retrospective population-based cohort study on the association between preeclampsia and childhood developmental vulnerability in emotional maturity and social competence domains in children born in Western Australia in 2009, 2012 and 2015. We obtained records of births, developmental anomalies, midwives notifications and hospitalisations. These data were linked to the Australian Early Development Census (AEDC), from which developmental vulnerability in emotional maturity and social competence domains at a median age of 5 years was ascertained. Causal relative risks (RR) were estimated with doubly robust estimation. RESULTS A total of 64,391 mother-offspring pairs were included in the final analysis. For the whole cohort, approximately 25 % and 23 % of children were classified as developmentally vulnerable or at-risk on AEDC emotional maturity and social competence domains, respectively. Approximately 2.8 % of children were exposed in utero to preeclampsia. Children exposed to preeclampsia were more likely to be classified as developmentally vulnerable or at-risk on the emotional maturity (RR = 1.19, 95%CI:1.11-1.28) and social competence domains (RR = 1.22, 95 % CI:1.13-1.31). CONCLUSION Children exposed to pre-eclampsia in utero were more likely to be developmentally vulnerable in emotional maturity and social competence domains in this cohort. Our findings provide new insights into the harmful effect of preeclampsia on childhood developmental vulnerability.
Collapse
Affiliation(s)
- Bereket Duko
- Australian Centre for Precision Health, UniSA Clinical & Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; Curtin School of Population Health, Curtin University, Kent St, Bentley, WA 6102, Australia; South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia.
| | | | - Gizachew Assefa Tessema
- Curtin School of Population Health, Curtin University, Kent St, Bentley, WA 6102, Australia; enAble Institute, Curtin University, Kent Street, Bentley, Western Australia 6102, Australia
| | - Jennifer Dunne
- Curtin School of Population Health, Curtin University, Kent St, Bentley, WA 6102, Australia
| | - Rosa Alati
- Curtin School of Population Health, Curtin University, Kent St, Bentley, WA 6102, Australia; enAble Institute, Curtin University, Kent Street, Bentley, Western Australia 6102, Australia; Institute for Social Sciences Research, The University of Queensland, 80 Meier's Rd, Indooroopilly, Queensland 4068, Australia
| | - Gavin Pereira
- Curtin School of Population Health, Curtin University, Kent St, Bentley, WA 6102, Australia; enAble Institute, Curtin University, Kent Street, Bentley, Western Australia 6102, Australia
| |
Collapse
|
30
|
Lu X, Sun Q, Wu L, Liao M, Yao J, Xiu M. The neutrophil-lymphocyte ratio in first-episode medication-naïve patients with schizophrenia: A 12-week longitudinal follow-up study. Prog Neuropsychopharmacol Biol Psychiatry 2024; 131:110959. [PMID: 38311095 DOI: 10.1016/j.pnpbp.2024.110959] [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: 11/25/2023] [Revised: 01/30/2024] [Accepted: 02/01/2024] [Indexed: 02/06/2024]
Abstract
Inflammation has been related to schizophrenia (SZ). The neutrophil-to-lymphocyte ratio (NLR) is an inexpensive inflammatory marker, however, its potential predictive value in patients with SZ has not been extensively investigated. This study aimed to examine whether NLR could predict the clinical response to antipsychotics in this population. One hundred and ninety-five medication-naïve first-episode schizophrenia (MNFES) patients were recruited and received treatment with risperidone for 12 weeks in the present study. Clinical symptoms were evaluated at week 0 and the end of 12 weeks of treatment using the PANSS scales. Complete blood counts were determined at baseline. We found that baseline NEU counts and NLR were positively associated with improvements in clinical symptoms in patients. In addition, MNFES patients with higher baseline NLR values showed a better treatment response to antipsychotics. Linear regression analysis revealed a predictive role of baseline NLR for the improvements of clinical symptoms in SZ patients. Our findings demonstrate that higher NLR was related to better improvements in symptoms after 12 weeks of treatment with antipsychotics, which renders it a promising biomarker of the response to antipsychotics in clinical practice.
Collapse
Affiliation(s)
- Xiaobing Lu
- Department of Nutritional and Metabolic Psychiatry, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, China
| | | | - Ling Wu
- Qingdao Mental Health Center, Qingdao, China
| | - Meisi Liao
- North University of China, Taiyuan, China
| | - Jing Yao
- Peking University HuiLongGuan Clinical Medical School, Beijing HuiLongGuan Hospital, Beijing, China
| | - Meihong Xiu
- Peking University HuiLongGuan Clinical Medical School, Beijing HuiLongGuan Hospital, Beijing, China.
| |
Collapse
|
31
|
Liu Y, Hang X, Zhang Y, Fang Y, Yuan S, Zhang Y, Wu B, Kong Y, Kuang Z, Sun W. Maternal immune activation induces sex-dependent behavioral differences in a rat model of schizophrenia. Front Psychiatry 2024; 15:1375999. [PMID: 38659461 PMCID: PMC11040086 DOI: 10.3389/fpsyt.2024.1375999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 03/19/2024] [Indexed: 04/26/2024] Open
Abstract
Background Maternal immune activation (MIA) is a mature means to construct a schizophrenia model. However, some preclinical studies have reported that a MIA-induced schizophrenia model seemed to have gender heterogeneity in behavioral phenotype. On the other hand, the MIA's paradigms were diverse in different studies, and many details could affect the effect of MIA. To some extent, it is not credible and scientific to directly compare the gender differences of different MIA programs. Therefore, it is necessary to study whether the sex of the exposed offspring leads to behavioral differences on the premise of maintaining a consistent MIA mode. Methods An animal model of schizophrenia was established by the administration of 10 mg/kg Poly (I: C) when dams were on day 9 of gestation. Then, a number of female and male offspring completed a series of behavioral tests during postnatal days 61-75. Results Compared with the female control group (n = 14), female MIA offspring (n = 12) showed a longer movement distance (d = 1.07, p < 0.05) and higher average speed (d = 1.08, p < 0.05) in the open field test (OFT). In the Y maze test, the percentage of entering the novel arm of female MIA offspring was lower (d = 0.92, p < 0.05). Compared with the male control group (n = 14), male MIA offspring (n = 13) displayed less movement distance (d = 0.93, p < 0.05) and a lower average speed (d = 0.94, p < 0.05) in the OFT. In the Y maze test, the proportion of exploration time in the novel arm of male MIA offspring was lower (d = 0.96, p < 0.05). In the EPM, male MIA offspring showed less time (d = 0.85, p < 0.05) and a lower percentage of time spent in the open arms (d = 0.85, p < 0.05). Male MIA offspring also had a lower PPI index (76 dB + 120 dB, d = 0.81, p < 0.05; 80 dB + 120 dB, d = 1.45, p < 0.01). Conclusions Our results showed that the behavioral phenotypes induced by prenatal immune activation were highly dependent on the sex of the offspring.
Collapse
Affiliation(s)
- Yunxia Liu
- The Third Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaoyi Hang
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yijie Zhang
- The Third Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Yilin Fang
- The Third Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Shanfang Yuan
- The Third Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Yi Zhang
- Department of Encephalopathy, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China
| | - Bin Wu
- The Third Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Yan Kong
- The Third Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Zihe Kuang
- The Third Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Wenjun Sun
- Department of Encephalopathy, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China
| |
Collapse
|
32
|
Du J, Nakachi Y, Murata Y, Kiyota E, Kato T, Bundo M, Iwamoto K. Exploration of cell type-specific somatic mutations in schizophrenia and the impact of maternal immune activation on the somatic mutation profile in the brain. Psychiatry Clin Neurosci 2024; 78:237-247. [PMID: 38334156 DOI: 10.1111/pcn.13640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 12/10/2023] [Accepted: 12/20/2023] [Indexed: 02/10/2024]
Abstract
AIM Schizophrenia (SZ) is a severe psychiatric disorder caused by the interaction of genetic and environmental factors. Although somatic mutations that occur in the brain after fertilization may play an important role in the cause of SZ, their frequencies and patterns in the brains of patients and related animal models have not been well studied. This study aimed to find somatic mutations related to the pathophysiology of SZ. METHODS We performed whole-exome sequencing (WES) of neuronal and nonneuronal nuclei isolated from the postmortem prefrontal cortex of patients with SZ (n = 10) and controls (n = 10). After detecting somatic mutations, we explored the similarities and differences in shared common mutations between two cell types and cell type-specific mutations. We also performed WES of prefrontal cortex samples from an animal model of SZ based on maternal immune activation (MIA) and explored the possible impact of MIA on the patterns of somatic mutations. RESULTS We did not find quantitative differences in somatic mutations but found higher variant allele fractions of neuron-specific mutations in patients with SZ. In the mouse model, we found a larger variation in the number of somatic mutations in the offspring of MIA mice, with the occurrence of somatic mutations in neurodevelopment-related genes. CONCLUSION Somatic mutations occurring at an earlier stage of brain cell differentiation toward neurons may be important for the cause of SZ. MIA may affect somatic mutation profiles in the brain.
Collapse
Affiliation(s)
- Jianbin Du
- Department of Molecular Brain Science, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yutaka Nakachi
- Department of Molecular Brain Science, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yui Murata
- Department of Molecular Brain Science, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
- Laboratory for Molecular Dynamics of Mental Disorders, RIKEN Brain Science Institute, Wako, Japan
| | - Emi Kiyota
- Department of Molecular Brain Science, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Tadafumi Kato
- Laboratory for Molecular Dynamics of Mental Disorders, RIKEN Brain Science Institute, Wako, Japan
- Department of Psychiatry and Behavioral Science, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Miki Bundo
- Department of Molecular Brain Science, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
- Laboratory for Molecular Dynamics of Mental Disorders, RIKEN Brain Science Institute, Wako, Japan
| | - Kazuya Iwamoto
- Department of Molecular Brain Science, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
- Laboratory for Molecular Dynamics of Mental Disorders, RIKEN Brain Science Institute, Wako, Japan
| |
Collapse
|
33
|
Nielsen TC, Nassar N, Shand AW, Jones HF, Han VX, Patel S, Guastella AJ, Dale RC, Lain SJ. Association between cumulative maternal exposures related to inflammation and child attention-deficit/hyperactivity disorder: A cohort study. Paediatr Perinat Epidemiol 2024; 38:241-250. [PMID: 38009577 DOI: 10.1111/ppe.13022] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 11/10/2023] [Accepted: 11/11/2023] [Indexed: 11/29/2023]
Abstract
BACKGROUND Preclinical studies suggest synergistic effects of maternal inflammatory exposures on offspring neurodevelopment, but human studies have been limited. OBJECTIVES To examine the cumulative association and potential interactions between seven maternal exposures related to inflammation and child attention-deficit/hyperactivity disorder (ADHD). METHODS We conducted a population-based cohort study of children born from July 2001 to December 2011 in New South Wales, Australia, and followed up until December 2014. Seven maternal exposures were identified from birth data and hospital admissions during pregnancy: autoimmune disease, asthma, hospitalization for infection, mood or anxiety disorder, smoking, hypertension, and diabetes. Child ADHD was identified from stimulant prescription records. Multivariable Cox regression assessed the association between individual and cumulative exposures and ADHD and potential interaction between exposures, controlling for potential confounders. RESULTS The cohort included 908,770 children, one-third (281,724) with one or more maternal exposures. ADHD was identified in 16,297 children (incidence 3.5 per 1000 person-years) with median age of 7 (interquartile range 2) years at first treatment. Each exposure was independently associated with ADHD, and risk increased with additional exposures: one exposure (hazard ratio (HR) 1.59, 95% confidence interval (CI) 1.54, 1.65), two exposures (HR 2.25, 95% CI 2.13, 2.37), and three or more exposures (HR 3.28, 95% CI 2.95, 3.64). Positive interaction was found between smoking and infection. The largest effect size was found for cumulative exposure of asthma, infection, mood or anxiety disorder, and smoking (HR 6.12, 95% CI 3.47, 10.70). CONCLUSIONS This study identifies cumulative effects of multiple maternal exposures related to inflammation on ADHD, most potentially preventable or modifiable. Future studies should incorporate biomarkers of maternal inflammation and consider gene-environment interactions.
Collapse
Affiliation(s)
- Timothy C Nielsen
- Children's Hospital Westmead Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Natasha Nassar
- Children's Hospital Westmead Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Antonia W Shand
- Children's Hospital Westmead Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Royal Hospital for Women, Randwick, New South Wales, Australia
| | - Hannah F Jones
- Children's Hospital Westmead Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Starship Children's Hospital, Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Velda X Han
- Children's Hospital Westmead Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Khoo Teck Puat-National University Children's Medical Institute, National University Health System, Singapore City, Singapore
| | - Shrujna Patel
- Children's Hospital Westmead Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Adam J Guastella
- Children's Hospital Westmead Clinical School, Brain and Mind Centre, Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - Russell C Dale
- Children's Hospital Westmead Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Samantha J Lain
- Children's Hospital Westmead Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
34
|
Moreno-Fernández M, Ucha M, Reis-de-Paiva R, Marcos A, Ambrosio E, Higuera-Matas A. Lack of interactions between prenatal immune activation and Δ 9-tetrahydrocannabinol exposure during adolescence in behaviours relevant to symptom dimensions of schizophrenia in rats. Prog Neuropsychopharmacol Biol Psychiatry 2024; 129:110889. [PMID: 37918558 DOI: 10.1016/j.pnpbp.2023.110889] [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: 01/20/2023] [Revised: 07/27/2023] [Accepted: 10/29/2023] [Indexed: 11/04/2023]
Abstract
The causality in the association between cannabis use and the risk of developing schizophrenia has been the subject of intense debate in the last few years. The development of animal models recapitulating several aspects of the disease is crucial for shedding light on this issue. Given that maternal infections are a known risk for schizophrenia, here, we used the maternal immune activation (MIA) model combined with THC exposure during adolescence to examine several behaviours in rats (working memory in the Y maze, sociability in the three-chamber test, sucrose preference as a measure, prepulse inhibition and formation of incidental associations) that are similar to the different symptom clusters of the disease. To this end, we administered LPS to pregnant dams and when the offspring reached adolescence, we exposed them to a mild dose of THC to examine their behaviour in adulthood. We also studied several parameters in the dams, including locomotor activity in the open field, elevated plus maze performance and their response to LPS, that could predict symptom severity of the offspring, but found no evidence of any predictive value of these variables. In the adult offspring, MIA was associated with impaired working memory and sensorimotor gating, but surprisingly, it increased sociability, social novelty and sucrose preference. THC, on its own, impaired sociability and social memory, but there were no interactions between MIA and THC exposure. These results suggest that, in this model, THC during adolescence does not trigger or aggravate symptoms related to schizophrenia in rats.
Collapse
Affiliation(s)
- Mario Moreno-Fernández
- Department of Psychobiology, School of Psychology, National University for Distance Learning (UNED), Madrid, Spain; UNED International Graduate School (EIDUNED), Madrid, Spain
| | - Marcos Ucha
- Department of Psychobiology, School of Psychology, National University for Distance Learning (UNED), Madrid, Spain.
| | - Raquel Reis-de-Paiva
- Department of Psychobiology, School of Psychology, National University for Distance Learning (UNED), Madrid, Spain
| | - Alberto Marcos
- Department of Psychobiology, School of Psychology, National University for Distance Learning (UNED), Madrid, Spain
| | - Emilio Ambrosio
- Department of Psychobiology, School of Psychology, National University for Distance Learning (UNED), Madrid, Spain
| | - Alejandro Higuera-Matas
- Department of Psychobiology, School of Psychology, National University for Distance Learning (UNED), Madrid, Spain.
| |
Collapse
|
35
|
Santos CAD, Paula AP, Filho GGF, Alves MM, Nery AF, Pontes MGA, Macedo EYL, Oliveira RM, Freitas SM, Lima S, Varela FVC, Viana ALS, Silva ALP, Silva ÉGC, D'Souza-Li L. Developmental impairment in children exposed during pregnancy to maternal SARS-COV2: A Brazilian cohort study. Int J Infect Dis 2024; 139:146-152. [PMID: 38061413 DOI: 10.1016/j.ijid.2023.12.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 11/29/2023] [Accepted: 12/02/2023] [Indexed: 12/31/2023] Open
Abstract
OBJECTIVES To determine the effects of in-utero exposure to maternal SARS-CoV-2 infection on offspring's neurodevelopment during the first year of life. METHODS We performed a prospective cohort of babies exposed to SARS-CoV-2 during pregnancy, and a control group (CG) of unexposed babies in a low-income area in Brazil. Children's neurodevelopment was assessed using the guide for Monitoring Child Development in the Integrated Management of Childhood Illness context for both groups (at 1,2,3,4,5,6, 9, and 12 months), and the Ages & Stages Questionnaire (ASQ-3) for the exposed group (EG) (at 4, 6 and 12 months). RESULTS We followed 137 children for 1 year, 69 in the COVID-19-EG, and 68 in the CG. All mothers were unvaccinated at the time of cohort inclusion, and maternal demographics were similar in the two groups. 20.3% of EG children and 5.9% of the CG received a diagnosis of neurodevelopmental delay within 12 months of life (P = 0.013, relative risk = 3.44; 95% confidence interval, 1.19- 9.95). For the EG, the prevalence of neurodevelopment impairment using Ages & Stages Questionnaire was 35.7% at 4 months, 7% at 6 months, and 32.1% at 12 months. CONCLUSION SARS-CoV-2 exposure was associated with neurodevelopmental impairment, and specific guidelines are needed for the follow-up of these high-risk children to mitigate the long-term effects on children's health.
Collapse
Affiliation(s)
- Carolina A D Santos
- University of Campinas, UNICAMP, Faculty of Medical Science, Campinas, Brazil; Santos Dumont Institute, Anita Garibaldi Center for Education and Research in Health, Macaíba, Brazil.
| | - Artemis P Paula
- Santos Dumont Institute, Anita Garibaldi Center for Education and Research in Health, Macaíba, Brazil
| | - Gentil G Fonseca Filho
- Federal University of Rio Grande do Norte, Natal, Brazil; Federal University of Rio Grande do Norte, Santa Cruz, Brazil
| | - Manoella M Alves
- Santos Dumont Institute, Anita Garibaldi Center for Education and Research in Health, Macaíba, Brazil; Federal University of Rio Grande do Norte, Natal, Brazil
| | - Andréia F Nery
- Federal University of Rio Grande do Norte, Natal, Brazil
| | - Monise G A Pontes
- Santos Dumont Institute, Anita Garibaldi Center for Education and Research in Health, Macaíba, Brazil
| | - Erianna Y L Macedo
- Santos Dumont Institute, Anita Garibaldi Center for Education and Research in Health, Macaíba, Brazil
| | - Ruy M Oliveira
- Santos Dumont Institute, Anita Garibaldi Center for Education and Research in Health, Macaíba, Brazil
| | - Sabrinna M Freitas
- Santos Dumont Institute, Anita Garibaldi Center for Education and Research in Health, Macaíba, Brazil
| | - Sarah Lima
- Santos Dumont Institute, Anita Garibaldi Center for Education and Research in Health, Macaíba, Brazil
| | - Fernanda V C Varela
- Santos Dumont Institute, Anita Garibaldi Center for Education and Research in Health, Macaíba, Brazil
| | - Andrezza L S Viana
- Santos Dumont Institute, Anita Garibaldi Center for Education and Research in Health, Macaíba, Brazil
| | - Aline L P Silva
- Santos Dumont Institute, Anita Garibaldi Center for Education and Research in Health, Macaíba, Brazil
| | - Érika G C Silva
- Santos Dumont Institute, Anita Garibaldi Center for Education and Research in Health, Macaíba, Brazil
| | - Lília D'Souza-Li
- University of Campinas, UNICAMP, Faculty of Medical Science, Department of Pediatrics, Campinas, Brazil
| |
Collapse
|
36
|
Gillespie B, Panthi S, Sundram S, Hill RA. The impact of maternal immune activation on GABAergic interneuron development: A systematic review of rodent studies and their translational implications. Neurosci Biobehav Rev 2024; 156:105488. [PMID: 38042358 DOI: 10.1016/j.neubiorev.2023.105488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 11/09/2023] [Accepted: 11/27/2023] [Indexed: 12/04/2023]
Abstract
Mothers exposed to infections during pregnancy disproportionally birth children who develop autism and schizophrenia, disorders associated with altered GABAergic function. The maternal immune activation (MIA) model recapitulates this risk factor, with many studies also reporting disruptions to GABAergic interneuron expression, protein, cellular density and function. However, it is unclear if there are species, sex, age, region, or GABAergic subtype specific vulnerabilities to MIA. Furthermore, to fully comprehend the impact of MIA on the GABAergic system a synthesised account of molecular, cellular, electrophysiological and behavioural findings was required. To this end we conducted a systematic review of GABAergic interneuron changes in the MIA model, focusing on the prefrontal cortex and hippocampus. We reviewed 102 articles that revealed robust changes in a number of GABAergic markers that present as gestationally-specific, region-specific and sometimes sex-specific. Disruptions to GABAergic markers coincided with distinct behavioural phenotypes, including memory, sensorimotor gating, anxiety, and sociability. Findings suggest the MIA model is a valid tool for testing novel therapeutics designed to recover GABAergic function and associated behaviour.
Collapse
Affiliation(s)
- Brendan Gillespie
- Department of Psychiatry, School of Clinical Sciences, Monash University, Clayton, VIC 3168, Australia
| | - Sandesh Panthi
- Department of Psychiatry, School of Clinical Sciences, Monash University, Clayton, VIC 3168, Australia
| | - Suresh Sundram
- Department of Psychiatry, School of Clinical Sciences, Monash University, Clayton, VIC 3168, Australia
| | - Rachel A Hill
- Department of Psychiatry, School of Clinical Sciences, Monash University, Clayton, VIC 3168, Australia.
| |
Collapse
|
37
|
Jain S, Oltman S, Rogers E, Ryckman K, Petersen M, Baer RJ, Rand L, Piao X, Jelliffe-Pawlowski L. Assessing for prenatal risk factors associated with infant neurologic morbidity using a multivariate analysis. J Perinatol 2023; 43:1486-1493. [PMID: 37950045 PMCID: PMC10716040 DOI: 10.1038/s41372-023-01820-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 10/19/2023] [Accepted: 10/30/2023] [Indexed: 11/12/2023]
Abstract
OBJECTIVE To characterize the biochemical and demographic profiles of pregnant people with maternal immune activation (MIA) and identify the prenatal characteristics associated with neurologic morbidity in offspring. STUDY DESIGN This was a retrospective cohort study of 602 mother-infant dyads with births between 2009 and 2010 in California. Multivariable logistic regression was used to build a MIA vulnerability profile including mid-pregnancy biochemical markers and maternal demographic characteristics, and its relationship with infant neurologic morbidity was examined. RESULTS Of the 602 mother-infant dyads, 80 mothers and 61 infants had diagnoses suggestive of MIA and neurologic morbidity, respectively. Our model, including two demographic and seven biochemical characteristics, identified mothers with MIA with good performance (AUC:0.814; 95% CI:0.7-0.8). Three demographic and five inflammatory markers together identified 80% of infants with neurological morbidity (AUC:0.802, 95% CI:0.7-0.8). CONCLUSION Inflammatory environment in mothers with pre-existing risk factors like obesity, poverty, and prematurity renders offspring more susceptible to neurologic morbidities.
Collapse
Affiliation(s)
- Samhita Jain
- Division of Neonatology, Department of Pediatrics, University of California, San Francisco, CA, USA.
| | - Scott Oltman
- California Preterm Birth Initiative, University of California San Francisco, San Francisco, CA, USA
- Department of Epidemiology & Biostatistics, University of California, San Francisco, CA, USA
| | - Elizabeth Rogers
- Division of Neonatology, Department of Pediatrics, University of California, San Francisco, CA, USA
| | - Kelli Ryckman
- Department of Epidemiology and Biostatistics, Indiana University Bloomington, Bloomington, IN, USA
| | - Mark Petersen
- Division of Neonatology, Department of Pediatrics, University of California, San Francisco, CA, USA
| | - Rebecca J Baer
- California Preterm Birth Initiative, University of California San Francisco, San Francisco, CA, USA
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| | - Larry Rand
- California Preterm Birth Initiative, University of California San Francisco, San Francisco, CA, USA
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco School of Medicine, San Francisco, CA, USA
| | - Xianhua Piao
- Division of Neonatology, Department of Pediatrics, University of California, San Francisco, CA, USA
- Newborn Brain Research Institute, University of California, San Francisco, CA, USA
- Weill Institute for Neuroscience, University of California, San Francisco, CA, USA
| | - Laura Jelliffe-Pawlowski
- California Preterm Birth Initiative, University of California San Francisco, San Francisco, CA, USA
- Department of Epidemiology & Biostatistics, University of California, San Francisco, CA, USA
| |
Collapse
|
38
|
Guerrin CG, Prasad K, Vazquez-Matias DA, Zheng J, Franquesa-Mullerat M, Barazzuol L, Doorduin J, de Vries EF. Prenatal infection and adolescent social adversity affect microglia, synaptic density, and behavior in male rats. Neurobiol Stress 2023; 27:100580. [PMID: 37920548 PMCID: PMC10618826 DOI: 10.1016/j.ynstr.2023.100580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/27/2023] [Accepted: 10/12/2023] [Indexed: 11/04/2023] Open
Abstract
Maternal infection during pregnancy and childhood social trauma have been associated with neurodevelopmental and affective disorders, such as schizophrenia, autism spectrum disorders, bipolar disorder and depression. These disorders are characterized by changes in microglial cells, which play a notable role in synaptic pruning, and synaptic deficits. Here, we investigated the effect of prenatal infection and social adversity during adolescence - either alone or in combination - on behavior, microglia, and synaptic density. Male offspring of pregnant rats injected with poly I:C, mimicking prenatal infection, were exposed to repeated social defeat during adolescence. We found that maternal infection during pregnancy prevented the reduction in social behavior and increase in anxiety induced by social adversity during adolescence. Furthermore, maternal infection and social adversity, alone or in combination, induced hyperlocomotion in adulthood. Longitudinal in vivo imaging with [11C]PBR28 positron emission tomography revealed that prenatal infection alone and social adversity during adolescence alone induced a transient increase in translocator protein TSPO density, an indicator of glial reactivity, whereas their combination induced a long-lasting increase that remained until adulthood. Furthermore, only the combination of prenatal infection and social adversity during adolescence induced an increase in microglial cell density in the frontal cortex. Prenatal infection increased proinflammatory cytokine IL-1β protein levels in hippocampus and social adversity reduced anti-inflammatory cytokine IL-10 protein levels in hippocampus during adulthood. This reduction in IL-10 was prevented if rats were previously exposed to prenatal infection. Adult offspring exposed to prenatal infection or adolescent social adversity had a higher synaptic density in the frontal cortex, but not hippocampus, as evaluated by synaptophysin density. Interestingly, such an increase in synaptic density was not observed in rats exposed to the combination of prenatal infection and social adversity, perhaps due to the long-lasting increase in microglial density, which may lead to an increase in microglial synaptic pruning. These findings suggest that changes in microglia activity and cytokine release induced by prenatal infection and social adversity during adolescence may be related to a reduced synaptic pruning, resulting in a higher synaptic density and behavioral changes in adulthood.
Collapse
Affiliation(s)
- Cyprien G.J. Guerrin
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713, GZ, Groningen, the Netherlands
| | - Kavya Prasad
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713, GZ, Groningen, the Netherlands
| | - Daniel A. Vazquez-Matias
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713, GZ, Groningen, the Netherlands
| | - Jing Zheng
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713, GZ, Groningen, the Netherlands
| | - Maria Franquesa-Mullerat
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713, GZ, Groningen, the Netherlands
| | - Lara Barazzuol
- Department of Radiation Oncology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, the Netherlands
- Department of Biomedical Sciences of Cells and Systems, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, the Netherlands
| | - Janine Doorduin
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713, GZ, Groningen, the Netherlands
| | - Erik F.J. de Vries
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713, GZ, Groningen, the Netherlands
| |
Collapse
|
39
|
de Oliveira EG, de Lima DA, da Silva Júnior JC, de Souza Barbosa MV, de Andrade Silva SC, de Santana JH, Dos Santos Junior OH, Lira EC, Lagranha CJ, Duarte FS, Gomes DA. (R)-ketamine attenuates neurodevelopmental disease-related phenotypes in a mouse model of maternal immune activation. Eur Arch Psychiatry Clin Neurosci 2023; 273:1501-1512. [PMID: 37249625 DOI: 10.1007/s00406-023-01629-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 05/22/2023] [Indexed: 05/31/2023]
Abstract
Infections during pregnancy are associated with an increased risk of neuropsychiatric disorders with developmental etiologies, such as schizophrenia and autism spectrum disorders (ASD). Studies have shown that the animal model of maternal immune activation (MIA) reproduces a wide range of phenotypes relevant to the study of neurodevelopmental disorders. Emerging evidence shows that (R)-ketamine attenuates behavioral, cellular, and molecular changes observed in animal models of neuropsychiatric disorders. Here, we investigate whether (R)-ketamine administration during adolescence attenuates some of the phenotypes related to neurodevelopmental disorders in an animal model of MIA. For MIA, pregnant Swiss mice received intraperitoneally (i.p.) lipopolysaccharide (LPS; 100 µg/kg/day) or saline on gestational days 15 and 16. The two MIA-based groups of male offspring received (R)-ketamine (20 mg/kg/day; i.p.) or saline from postnatal day (PND) 36 to 50. At PND 62, the animals were examined for anxiety-like behavior and locomotor activity in the open-field test (OFT), as well as in the social interaction test (SIT). At PND 63, the prefrontal cortex (PFC) was collected for analysis of oxidative balance and gene expression of the cytokines IL-1β, IL-6, and TGF-β1. We show that (R)-ketamine abolishes anxiety-related behavior and social interaction deficits induced by MIA. Additionally, (R)-ketamine attenuated the increase in lipid peroxidation and the cytokines in the PFC of the offspring exposed to MIA. The present work suggests that (R)-ketamine administration may have a long-lasting attenuation in deficits in emotional behavior induced by MIA, and that these effects may be attributed to its antioxidant and anti-inflammatory activity in the PFC.
Collapse
Affiliation(s)
- Elifrances Galdino de Oliveira
- Laboratory of Neuroendocrinology and Metabolism, Department of Physiology and Pharmacology, Bioscience Center, Federal University of Pernambuco, Av. Prof. Moraes Rego, 1235 Cidade Universitária, Recife, PE, 50670-901, Brazil.
- Graduate Program of Neuropsychiatry and Behavioral Sciences, Federal University of Pernambuco, Recife, PE, Brazil.
| | - Diógenes Afonso de Lima
- Laboratory of Neuroendocrinology and Metabolism, Department of Physiology and Pharmacology, Bioscience Center, Federal University of Pernambuco, Av. Prof. Moraes Rego, 1235 Cidade Universitária, Recife, PE, 50670-901, Brazil
| | - José Carlos da Silva Júnior
- Laboratory of Neuroendocrinology and Metabolism, Department of Physiology and Pharmacology, Bioscience Center, Federal University of Pernambuco, Av. Prof. Moraes Rego, 1235 Cidade Universitária, Recife, PE, 50670-901, Brazil
| | - Mayara Victória de Souza Barbosa
- Laboratory of Neuroendocrinology and Metabolism, Department of Physiology and Pharmacology, Bioscience Center, Federal University of Pernambuco, Av. Prof. Moraes Rego, 1235 Cidade Universitária, Recife, PE, 50670-901, Brazil
| | - Severina Cassia de Andrade Silva
- Graduate Program of Neuropsychiatry and Behavioral Sciences, Federal University of Pernambuco, Recife, PE, Brazil
- Laboratory of Biochemistry and Exercise Biochemistry, Department of Physical Education and Sports Science, Federal University of Pernambuco, Vitória de Santo Antão, PE, Brazil
| | - Jonata Henrique de Santana
- Graduate Program of Neuropsychiatry and Behavioral Sciences, Federal University of Pernambuco, Recife, PE, Brazil
- Laboratory of Biochemistry and Exercise Biochemistry, Department of Physical Education and Sports Science, Federal University of Pernambuco, Vitória de Santo Antão, PE, Brazil
| | - Osmar Henrique Dos Santos Junior
- Graduate Program of Neuropsychiatry and Behavioral Sciences, Federal University of Pernambuco, Recife, PE, Brazil
- Laboratory of Biochemistry and Exercise Biochemistry, Department of Physical Education and Sports Science, Federal University of Pernambuco, Vitória de Santo Antão, PE, Brazil
| | - Eduardo Carvalho Lira
- Laboratory of Neuroendocrinology and Metabolism, Department of Physiology and Pharmacology, Bioscience Center, Federal University of Pernambuco, Av. Prof. Moraes Rego, 1235 Cidade Universitária, Recife, PE, 50670-901, Brazil
| | - Claudia Jacques Lagranha
- Graduate Program of Neuropsychiatry and Behavioral Sciences, Federal University of Pernambuco, Recife, PE, Brazil
- Laboratory of Biochemistry and Exercise Biochemistry, Department of Physical Education and Sports Science, Federal University of Pernambuco, Vitória de Santo Antão, PE, Brazil
| | - Filipe Silveira Duarte
- Laboratory of Neuroendocrinology and Metabolism, Department of Physiology and Pharmacology, Bioscience Center, Federal University of Pernambuco, Av. Prof. Moraes Rego, 1235 Cidade Universitária, Recife, PE, 50670-901, Brazil
| | - Dayane Aparecida Gomes
- Laboratory of Neuroendocrinology and Metabolism, Department of Physiology and Pharmacology, Bioscience Center, Federal University of Pernambuco, Av. Prof. Moraes Rego, 1235 Cidade Universitária, Recife, PE, 50670-901, Brazil
- Graduate Program of Neuropsychiatry and Behavioral Sciences, Federal University of Pernambuco, Recife, PE, Brazil
| |
Collapse
|
40
|
Hashimoto K. Arketamine for cognitive impairment in psychiatric disorders. Eur Arch Psychiatry Clin Neurosci 2023; 273:1513-1525. [PMID: 36786865 PMCID: PMC9925943 DOI: 10.1007/s00406-023-01570-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 02/01/2023] [Indexed: 02/15/2023]
Abstract
Cognitive impairment has been observed in patients with various psychiatric disorders, including schizophrenia, major depressive disorder (MDD), and bipolar disorder (BD). Although modern therapeutic drugs can improve certain symptoms (i.e., psychosis, depression) in these patients, these drugs have not been found to improve cognitive impairment. The N-methyl-D-aspartate receptor antagonist (R,S)-ketamine has attracted attention as a rapidly acting antidepressant. In addition to its robust antidepressant effects, (R,S)-ketamine has been suggested to improve cognitive impairment in patients with MDD and BD, despite causing cognitive impairment in healthy control subjects. (R,S)-ketamine is a racemic mixture of equal amounts of (R)-ketamine (or arketamine) and (S)-ketamine (or esketamine). Arketamine has been found to have more potent antidepressant-like actions than esketamine in rodents. Interestingly, arketamine, but not esketamine, has been suggested to improve phencyclidine-induced cognitive deficits in mice. Furthermore, arketamine has been suggested to ameliorate cognitive deficits in rodent offspring after maternal immune activation. In the current article, it is proposed that arketamine has therapeutic potential for treating cognitive impairment in patients with psychiatric disorders. Additionally, the potential role of the gut-microbiome-brain axis in cognitive impairment in psychiatric disorders is discussed.
Collapse
Affiliation(s)
- Kenji Hashimoto
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, 1-8-1 Inohana, Chiba, 260-8670, Japan.
| |
Collapse
|
41
|
Lesh TA, Iosif AM, Tanase C, Vlasova RM, Ryan AM, Bennett J, Hogrefe CE, Maddock RJ, Geschwind DH, Van de Water J, McAllister AK, Styner MA, Bauman MD, Carter CS. Extracellular free water elevations are associated with brain volume and maternal cytokine response in a longitudinal nonhuman primate maternal immune activation model. Mol Psychiatry 2023; 28:4185-4194. [PMID: 37582858 PMCID: PMC10867284 DOI: 10.1038/s41380-023-02213-w] [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: 08/31/2022] [Revised: 07/21/2023] [Accepted: 08/02/2023] [Indexed: 08/17/2023]
Abstract
Maternal infection has emerged as an important environmental risk factor for neurodevelopmental disorders, including schizophrenia and autism spectrum disorders. Animal model systems of maternal immune activation (MIA) suggest that the maternal immune response plays a significant role in the offspring's neurodevelopment and behavioral outcomes. Extracellular free water is a measure of freely diffusing water in the brain that may be associated with neuroinflammation and impacted by MIA. The present study evaluates the brain diffusion characteristics of male rhesus monkeys (Macaca mulatta) born to MIA-exposed dams (n = 14) treated with a modified form of the viral mimic polyinosinic:polycytidylic acid at the end of the first trimester. Control dams received saline injections at the end of the first trimester (n = 10) or were untreated (n = 4). Offspring underwent diffusion MRI scans at 6, 12, 24, 36, and 45 months. Offspring born to MIA-exposed dams showed significantly increased extracellular free water in cingulate cortex gray matter starting as early as 6 months of age and persisting through 45 months. In addition, offspring gray matter free water in this region was significantly correlated with the magnitude of the maternal IL-6 response in the MIA-exposed dams. Significant correlations between brain volume and extracellular free water in the MIA-exposed offspring also indicate converging, multimodal evidence of the impact of MIA on brain development. These findings provide strong evidence for the construct validity of the nonhuman primate MIA model as a system of relevance for investigating the pathophysiology of human neurodevelopmental psychiatric disorders. Elevated free water in individuals exposed to immune activation in utero could represent an early marker of a perturbed or vulnerable neurodevelopmental trajectory.
Collapse
Affiliation(s)
- Tyler A Lesh
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, CA, USA
| | - Ana-Maria Iosif
- Division of Biostatistics, Department of Public Health Sciences, University of California, Davis, CA, USA
| | - Costin Tanase
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, CA, USA
| | - Roza M Vlasova
- Department of Psychiatry, University of North Carolina, Chapel Hill, NC, USA
| | - Amy M Ryan
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, CA, USA
- MIND Institute, University of California, Davis, CA, USA
- California National Primate Research Center, Davis, CA, USA
| | - Jeffrey Bennett
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, CA, USA
| | | | - Richard J Maddock
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, CA, USA
| | - Daniel H Geschwind
- Neurogenetics Program, Department of Neurology, University of California, Los Angeles, CA, USA
| | - Judy Van de Water
- MIND Institute, University of California, Davis, CA, USA
- Rheumatology/Allergy and Clinical Immunology, University of California, Davis, CA, USA
| | - A Kimberley McAllister
- MIND Institute, University of California, Davis, CA, USA
- Center for Neuroscience, University of California, Davis, CA, USA
| | - Martin A Styner
- Department of Psychiatry, University of North Carolina, Chapel Hill, NC, USA
| | - Melissa D Bauman
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, CA, USA
- MIND Institute, University of California, Davis, CA, USA
- California National Primate Research Center, Davis, CA, USA
| | - Cameron S Carter
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, CA, USA.
| |
Collapse
|
42
|
Fujii S, Murata Y, Imamura Y, Nakachi Y, Bundo M, Kubota-Sakashita M, Kato T, Iwamoto K. Sex-dependent behavioral alterations in a poly(I:C)-induced maternal immune activation mouse model without segment filamentous bacteria. Neurosci Lett 2023; 814:137467. [PMID: 37652351 DOI: 10.1016/j.neulet.2023.137467] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 08/19/2023] [Accepted: 08/28/2023] [Indexed: 09/02/2023]
Abstract
Maternal immune activation is one of the environmental risk factors for offspring to develop psychiatric disorders. A synthetic viral mimetic immunogen, polyinosinic-polycytidylic acid (poly(I:C)), is used to induce maternal immune activation in animal models of psychiatric disorders. In the mouse poly(I:C) model, the existence of segment filamentous bacteria (SFB) in the maternal intestine has been reported to be important for the induction of ASD-related behavioral alterations as well as atypical cortical development called cortical patches. This study aimed to elucidate the effect of a single poly(I:C) injection during embryonic day (E) 9 to E16 on offspring's behavior in the ensured absence of maternal SFB by vancomycin drinking in C57BL/6N mice. The cortical patches were not found at either injection timings with poly(I:C) or PBS vehicle, tested in male or female offspring at postnatal day 0 or 1. Prepulse inhibition was decreased in male adult offspring most strongly at poly(I:C) injection timings later than E11, whereas a modest but significant decrease was observed in female offspring with an injection during E12 to E15. The decrease in social interaction was observed in female offspring most conspicuously at injection timings later than E11, whereas a significant decrease was observed in male offspring with an injection during E12 to E15. In conclusion, this study indicated that behavioral alterations could be induced without maternal SFB. The effect on behavior was substantially different between males and females.
Collapse
Affiliation(s)
- Shinya Fujii
- Department of Molecular Brain Science, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto-shi, Kumamoto 860-8556, Japan
| | - Yui Murata
- Department of Molecular Brain Science, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto-shi, Kumamoto 860-8556, Japan
| | - Yuko Imamura
- Department of Molecular Brain Science, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto-shi, Kumamoto 860-8556, Japan
| | - Yutaka Nakachi
- Department of Molecular Brain Science, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto-shi, Kumamoto 860-8556, Japan
| | - Miki Bundo
- Department of Molecular Brain Science, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto-shi, Kumamoto 860-8556, Japan
| | - Mie Kubota-Sakashita
- Department of Psychiatry and Behavioral Science, Juntendo University Graduate School of Medicine, Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan; Department of Molecular Pathology of Mood Disorders, Juntendo University Graduate School of Medicine, Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan
| | - Tadafumi Kato
- Department of Psychiatry and Behavioral Science, Juntendo University Graduate School of Medicine, Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan; Department of Molecular Pathology of Mood Disorders, Juntendo University Graduate School of Medicine, Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan
| | - Kazuya Iwamoto
- Department of Molecular Brain Science, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto-shi, Kumamoto 860-8556, Japan.
| |
Collapse
|
43
|
Steen NE, Rahman Z, Szabo A, Hindley GFL, Parker N, Cheng W, Lin A, O’Connell KS, Sheikh MA, Shadrin A, Bahrami S, Karthikeyan S, Hoseth EZ, Dale AM, Aukrust P, Smeland OB, Ueland T, Frei O, Djurovic S, Andreassen OA. Shared Genetic Loci Between Schizophrenia and White Blood Cell Counts Suggest Genetically Determined Systemic Immune Abnormalities. Schizophr Bull 2023; 49:1345-1354. [PMID: 37319439 PMCID: PMC10483470 DOI: 10.1093/schbul/sbad082] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
BACKGROUND Immune mechanisms are indicated in schizophrenia (SCZ). Recent genome-wide association studies (GWAS) have identified genetic variants associated with SCZ and immune-related phenotypes. Here, we use cutting edge statistical tools to identify shared genetic variants between SCZ and white blood cell (WBC) counts and further understand the role of the immune system in SCZ. STUDY DESIGN GWAS results from SCZ (patients, n = 53 386; controls, n = 77 258) and WBC counts (n = 56 3085) were analyzed. We applied linkage disequilibrium score regression, the conditional false discovery rate method and the bivariate causal mixture model for analyses of genetic associations and overlap, and 2 sample Mendelian randomization to estimate causal effects. STUDY RESULTS The polygenicity for SCZ was 7.5 times higher than for WBC count and constituted 32%-59% of WBC count genetic loci. While there was a significant but weak positive genetic correlation between SCZ and lymphocytes (rg = 0.05), the conditional false discovery rate method identified 383 shared genetic loci (53% concordant effect directions), with shared variants encompassing all investigated WBC subtypes: lymphocytes, n = 215 (56% concordant); neutrophils, n = 158 (49% concordant); monocytes, n = 146 (47% concordant); eosinophils, n = 135 (56% concordant); and basophils, n = 64 (53% concordant). A few causal effects were suggested, but consensus was lacking across different Mendelian randomization methods. Functional analyses indicated cellular functioning and regulation of translation as overlapping mechanisms. CONCLUSIONS Our results suggest that genetic factors involved in WBC counts are associated with the risk of SCZ, indicating a role of immune mechanisms in subgroups of SCZ with potential for stratification of patients for immune targeted treatment.
Collapse
Affiliation(s)
- Nils Eiel Steen
- NORMENT Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Zillur Rahman
- NORMENT Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Attila Szabo
- NORMENT Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- K.G. Jebsen Centre for Neurodevelopmental Disorders, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Guy F L Hindley
- NORMENT Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| | - Nadine Parker
- NORMENT Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Weiqiu Cheng
- NORMENT Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Aihua Lin
- NORMENT Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Kevin S O’Connell
- NORMENT Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Mashhood A Sheikh
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Alexey Shadrin
- NORMENT Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Shahram Bahrami
- NORMENT Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Sandeep Karthikeyan
- NORMENT Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Eva Z Hoseth
- NORMENT Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Division of Mental Health, Helse Møre Romsdal HF, Kristiansund, Norway
| | - Anders M Dale
- Department of Radiology, University of California, San Diego, La Jolla, CA, USA
- Department of Cognitive Sciences, University of California, San Diego, La Jolla, CA, USA
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Faculty of Medicine, University of Oslo, Oslo, Norway
- Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Olav B Smeland
- NORMENT Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Thor Ueland
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Faculty of Medicine, University of Oslo, Oslo, Norway
- K.G. Jebsen—Thrombosis Research and Expertise Center (TREC), University of Tromsø, Tromsø, Norway
| | - Oleksandr Frei
- NORMENT Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Center for Bioinformatics, Department of Informatics, University of Oslo, Oslo, Norway
| | - Srdjan Djurovic
- NORMENT Centre, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Ole A Andreassen
- NORMENT Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- K.G. Jebsen Centre for Neurodevelopmental Disorders, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
44
|
Munn RGK, Wolff A, Speers LJ, Bilkey DK. Disrupted hippocampal synchrony following maternal immune activation in a rat model. Hippocampus 2023; 33:995-1008. [PMID: 37129454 DOI: 10.1002/hipo.23545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 03/14/2023] [Accepted: 04/16/2023] [Indexed: 05/03/2023]
Abstract
Maternal immune activation (MIA) is a risk factor for schizophrenia and other neurodevelopmental disorders. MIA in rats models a number of the brain and behavioral changes that are observed in schizophrenia, including impaired memory. Recent studies in the MIA model have shown that the firing of the hippocampal place cells that are involved in memory processes appear relatively normal, but with abnormalities in the temporal ordering of firing. In this study, we re-analyzed data from prior hippocampal electrophysiological recordings of MIA and control animals to determine whether temporal dysfunction was evident. We find that there is a decreased ratio of slow to fast gamma power, resulting from an increase in fast gamma power and a tendency toward reduced slow gamma power in MIA rats. Moreover, we observe a robust reduction in spectral coherence between hippocampal theta and both fast and slow gamma rhythms, as well as changes in the phase of theta at which fast gamma occurs. We also find the phasic organization of place cell phase precession on the theta wave to be abnormal in MIA rats. Lastly, we observe that the local field potential of MIA rats contains more frequent sharp-wave ripple events, and that place cells were more likely to fire spikes during ripples in these animals than control. These findings provide further evidence of desynchrony in MIA animals and may point to circuit-level changes that underlie failures to integrate and encode information in schizophrenia.
Collapse
Affiliation(s)
- Robert G K Munn
- Department of Anatomy, University of Otago, Dunedin, New Zealand
| | - Amy Wolff
- Department of Neuroscience and Medical Discovery Team on Addiction, University of Minnesota, Minneapolis, Minnesota, USA
| | - Lucinda J Speers
- Department of Psychology, University of Otago, Dunedin, New Zealand
| | - David K Bilkey
- Department of Psychology, University of Otago, Dunedin, New Zealand
| |
Collapse
|
45
|
Perez-Palomar B, Erdozain AM, Erkizia-Santamaría I, Ortega JE, Meana JJ. Maternal Immune Activation Induces Cortical Catecholaminergic Hypofunction and Cognitive Impairments in Offspring. J Neuroimmune Pharmacol 2023; 18:348-365. [PMID: 37208550 PMCID: PMC10577104 DOI: 10.1007/s11481-023-10070-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 05/12/2023] [Indexed: 05/21/2023]
Abstract
BACKGROUND Impairment of specific cognitive domains in schizophrenia has been associated with prefrontal cortex (PFC) catecholaminergic deficits. Among other factors, prenatal exposure to infections represents an environmental risk factor for schizophrenia development in adulthood. However, it remains largely unknown whether the prenatal infection-induced changes in the brain may be associated with concrete switches in a particular neurochemical circuit, and therefore, if they could alter behavioral functions. METHODS In vitro and in vivo neurochemical evaluation of the PFC catecholaminergic systems was performed in offspring from mice undergoing maternal immune activation (MIA). The cognitive status was also evaluated. Prenatal viral infection was mimicked by polyriboinosinic-polyribocytidylic acid (poly(I:C)) administration to pregnant dams (7.5 mg/kg i.p., gestational day 9.5) and consequences were evaluated in adult offspring. RESULTS MIA-treated offspring showed disrupted recognition memory in the novel object recognition task (t = 2.30, p = 0.031). This poly(I:C)-based group displayed decreased extracellular dopamine (DA) concentrations compared to controls (t = 3.17, p = 0.0068). Potassium-evoked release of DA and noradrenaline (NA) were impaired in the poly(I:C) group (DA: Ft[10,90] = 43.33, p < 0.0001; Ftr[1,90] = 1.224, p = 0.2972; Fi[10,90] = 5.916, p < 0.0001; n = 11); (NA: Ft[10,90] = 36.27, p < 0.0001; Ftr[1,90] = 1.841, p = 0.208; Fi[10,90] = 8.686, p < 0.0001; n = 11). In the same way, amphetamine-evoked release of DA and NA were also impaired in the poly(I:C) group (DA: Ft[8,328] = 22.01, p < 0.0001; Ftr[1,328] = 4.507, p = 0.040; Fi[8,328] = 2.319, p = 0.020; n = 43); (NA: Ft[8,328] = 52.07; p < 0.0001; Ftr[1,328] = 4.322; p = 0.044; Fi[8,398] = 5.727; p < 0.0001; n = 43). This catecholamine imbalance was accompanied by increased dopamine D1 and D2 receptor expression (t = 2.64, p = 0.011 and t = 3.55, p = 0.0009; respectively), whereas tyrosine hydroxylase, DA and NA tissue content, DA and NA transporter (DAT/NET) expression and function were unaltered. CONCLUSIONS MIA induces in offspring a presynaptic catecholaminergic hypofunction in PFC with cognitive impairment. This poly(I:C)-based model reproduces catecholamine phenotypes reported in schizophrenia and represents an opportunity for the study of cognitive impairment associated to this disorder.
Collapse
Affiliation(s)
- Blanca Perez-Palomar
- Department of Pharmacology, University of the Basque Country UPV/EHU, Leioa, Bizkaia, E-48940, Spain
- Centro de Investigación Biomédica en Red de Salud Mental CIBERSAM, ISCIII, Leioa, Spain
- Biocruces Bizkaia Health Research Institute, Bizkaia, Spain
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, 63110, USA
- Department of Pharmaceutical and Administrative Sciences, University of Health Sciences and Pharmacy in St. Louis, St. Louis, MO, 63110, USA
| | - Amaia M Erdozain
- Department of Pharmacology, University of the Basque Country UPV/EHU, Leioa, Bizkaia, E-48940, Spain
- Centro de Investigación Biomédica en Red de Salud Mental CIBERSAM, ISCIII, Leioa, Spain
| | - Ines Erkizia-Santamaría
- Department of Pharmacology, University of the Basque Country UPV/EHU, Leioa, Bizkaia, E-48940, Spain
| | - Jorge E Ortega
- Department of Pharmacology, University of the Basque Country UPV/EHU, Leioa, Bizkaia, E-48940, Spain.
- Centro de Investigación Biomédica en Red de Salud Mental CIBERSAM, ISCIII, Leioa, Spain.
- Biocruces Bizkaia Health Research Institute, Bizkaia, Spain.
| | - J Javier Meana
- Department of Pharmacology, University of the Basque Country UPV/EHU, Leioa, Bizkaia, E-48940, Spain
- Centro de Investigación Biomédica en Red de Salud Mental CIBERSAM, ISCIII, Leioa, Spain
- Biocruces Bizkaia Health Research Institute, Bizkaia, Spain
| |
Collapse
|
46
|
Speers LJ, Chin P, Bilkey DK. No evidence that acute clozapine administration alters CA1 phase precession in rats. Brain Res 2023; 1814:148446. [PMID: 37301424 DOI: 10.1016/j.brainres.2023.148446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 05/25/2023] [Accepted: 06/04/2023] [Indexed: 06/12/2023]
Abstract
Hippocampal phase precession, wherein there is a systematic shift in the phase of neural firing against the underlying theta activity, is proposed to play an important role in the sequencing of information in memory. Previous research shows that the starting phase of precession is more variable in rats following maternal immune activation (MIA), a known risk factor for schizophrenia. Since starting phase variability has the potential to disorganize the construction of sequences of information, we tested whether the atypical antipsychotic clozapine, which ameliorates some cognitive deficits in schizophrenia, alters this aspect of phase precession. Either saline or clozapine (5 mg/kg) was administered to rats and then CA1 place cell activity was recorded from the CA1 region of the hippocampus as the animals ran around a rectangular track for food reward. When compared to saline trials, acute administration of clozapine did not affect any place cell properties, including those related to phase precession, in either control or MIA animals. Clozapine did, however, produce a reduction in locomotion speed, indicating that its presence had some effect on behaviour. These results help to constrain explanations of phase precession mechanisms and their potential role in sequence learning deficits.
Collapse
Affiliation(s)
| | - Phoebe Chin
- Psychology Dept., Otago Univ., Dunedin, New Zealand
| | | |
Collapse
|
47
|
Murlanova K, Pletnikov MV. Modeling psychotic disorders: Environment x environment interaction. Neurosci Biobehav Rev 2023; 152:105310. [PMID: 37437753 DOI: 10.1016/j.neubiorev.2023.105310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 06/26/2023] [Accepted: 07/05/2023] [Indexed: 07/14/2023]
Abstract
Schizophrenia is a major psychotic disorder with multifactorial etiology that includes interactions between genetic vulnerability and environmental risk factors. In addition, interplay of multiple environmental adversities affects neurodevelopment and may increase the individual risk of developing schizophrenia. Consistent with the two-hit hypothesis of schizophrenia, we review rodent models that combine maternal immune activation as the first hit with other adverse environmental exposures as the second hit. We discuss the strengths and pitfalls of the current animal models of environment x environment interplay and propose some future directions to advance the field.
Collapse
Affiliation(s)
- Kateryna Murlanova
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14203, USA
| | - Mikhail V Pletnikov
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14203, USA; Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
48
|
Hashimoto K. Emerging role of the host microbiome in neuropsychiatric disorders: overview and future directions. Mol Psychiatry 2023; 28:3625-3637. [PMID: 37845499 PMCID: PMC10730413 DOI: 10.1038/s41380-023-02287-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 09/23/2023] [Accepted: 09/29/2023] [Indexed: 10/18/2023]
Abstract
The human body harbors a diverse ecosystem of microorganisms, including bacteria, viruses, and fungi, collectively known as the microbiota. Current research is increasingly focusing on the potential association between the microbiota and various neuropsychiatric disorders. The microbiota resides in various parts of the body, such as the oral cavity, nasal passages, lungs, gut, skin, bladder, and vagina. The gut microbiota in the gastrointestinal tract has received particular attention due to its high abundance and its potential role in psychiatric and neurodegenerative disorders. However, the microbiota presents in other body tissues, though less abundant, also plays crucial role in immune system and human homeostasis, thus influencing the development and progression of neuropsychiatric disorders. For example, oral microbiota imbalance and associated periodontitis might increase the risk for neuropsychiatric disorders. Additionally, studies using the postmortem brain samples have detected the widespread presence of oral bacteria in the brains of patients with Alzheimer's disease. This article provides an overview of the emerging role of the host microbiota in neuropsychiatric disorders and discusses future directions, such as underlying biological mechanisms, reliable biomarkers associated with the host microbiota, and microbiota-targeted interventions, for research in this field.
Collapse
Affiliation(s)
- Kenji Hashimoto
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, 260-8670, Japan.
| |
Collapse
|
49
|
Yildiz Taskiran S, Taskiran M, Unal G, Bozkurt NM, Golgeli A. The long-lasting effects of aceclofenac, a COX-2 inhibitor, in a Poly I:C-Induced maternal immune activation model of schizophrenia in rats. Behav Brain Res 2023; 452:114565. [PMID: 37414224 DOI: 10.1016/j.bbr.2023.114565] [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: 03/13/2023] [Revised: 07/01/2023] [Accepted: 07/02/2023] [Indexed: 07/08/2023]
Abstract
It is well established that rats exposed to inflammation during pregnancy or the perinatal period have an increased chance of developing schizophrenia-like symptoms and behaviors, and people with schizophrenia also have raised levels of inflammatory markers. Therefore, there is evidence supporting the idea that anti-inflammatory drugs may have therapeutic benefits. Aceclofenac is a nonsteroidal anti-inflammatory drug that has anti-inflammatory properties and is used clinically to treat inflammatory and painful processes such as osteoarthritis and rheumatoid arthritis, making it a potential candidate for preventive or adjunctive therapy in schizophrenia. This study therefore examined the effect of aceclofenac in a maternal immune activation model of schizophrenia, in which polyinosinic-polycytidylic acid (Poly I:C) (8 mg/kg, i.p.) was administered to pregnant rat dams. Young female rat pups received daily aceclofenac (5, 10, and 20 mg/kg, i.p., n = 10) between postnatal day 56 and 76. The effects of aceclofenac were compared with assessment of behavioral tests and ELISA results. During the postnatal days (PNDs) 73-76, behavioral tests were conducted in rats, and on PND 76, ELISA tests were performed to examine the changes in Tumor necrosis factor alpha (TNF-α), Interleukin-1β (IL-1β), Brain-derived neurotrophic factor (BDNF), and nestin levels. Aceclofenac treatment reversed deficits in prepulse inhibition, novel object recognition, social interaction, and locomotor activity tests. In addition, aceclofenac administration decreased TNF-α and IL-1β expression in the prefrontal cortex and hippocampus. In contrast, BDNF and nestin levels did not change significantly during treatment with aceclofenac. Taken together, these results suggest that aceclofenac may be an alternative therapeutic adjunctive strategy to improve the clinical expression of schizophrenia in the further studies.
Collapse
Affiliation(s)
| | - Mehmet Taskiran
- Department of Biology, Faculty of Science, Erciyes University, Kayseri, Türkiye
| | - Gokhan Unal
- Department of Pharmacology, Faculty of Pharmacy, Erciyes University, Kayseri, Türkiye
| | - Nuh Mehmet Bozkurt
- Department of Pharmacology, Faculty of Pharmacy, Erciyes University, Kayseri, Türkiye
| | - Asuman Golgeli
- Department of Physiology, Faculty of Medicine, Erciyes University, Kayseri, Türkiye
| |
Collapse
|
50
|
Guerrin CGJ, de Vries EFJ, Prasad K, Vazquez-Matias DA, Manusiwa LE, Barazzuol L, Doorduin J. Maternal infection during pregnancy aggravates the behavioral response to an immune challenge during adolescence in female rats. Behav Brain Res 2023; 452:114566. [PMID: 37419332 DOI: 10.1016/j.bbr.2023.114566] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 05/23/2023] [Accepted: 07/03/2023] [Indexed: 07/09/2023]
Abstract
Prenatal and early postnatal infection have been associated with changes in microglial activity and the development of psychiatric disorders. Here, we investigated the effect of prenatal immune activation and postnatal immune challenge, alone and combined, on behavior and microglial cell density in female Wistar rats. Pregnant rats were injected with poly I:C to induce a maternal immune activation (MIA). Their female offspring were subsequently exposed to a lipopolysaccharide (LPS) immune challenge during adolescence. Anhedonia, social behavior, anxiety, locomotion, and working memory were measured with the sucrose preference, social interaction, open field, elevated-plus maze, and Y-maze test, respectively. Microglia cell density was quantified by counting the number of Iba-1 positive cells in the brain cortex. Female MIA offspring were more susceptible to the LPS immune challenge during adolescence than control offspring as demonstrated by a more pronounced reduction in sucrose preference and body weight on the days following the LPS immune challenge. Furthermore, only the rats exposed to both MIA and LPS showed long-lasting changes in social behavior and locomotion. Conversely, the combination MIA and LPS prevented the anxiety induced by MIA alone during adulthood. MIA, LPS, or their combination did not change microglial cell density in the parietal and frontal cortex of adult rats. The results of our study suggest that the maternal immune activation during pregnancy aggravates the response to an immune challenge during adolescence in female rats.
Collapse
Affiliation(s)
- Cyprien G J Guerrin
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, Groningen 9713 GZ, the Netherlands
| | - Erik F J de Vries
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, Groningen 9713 GZ, the Netherlands
| | - Kavya Prasad
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, Groningen 9713 GZ, the Netherlands
| | - Daniel A Vazquez-Matias
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, Groningen 9713 GZ, the Netherlands
| | - Lesley E Manusiwa
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, Groningen 9713 GZ, the Netherlands
| | - Lara Barazzuol
- Department of Radiation Oncology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, Groningen 9713 GZ, the Netherlands; Department of Biomedical Sciences of Cells and Systems, University of Groningen, University Medical Center Groningen, Hanzeplein 1, Groningen 9713 GZ, the Netherlands
| | - Janine Doorduin
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, Groningen 9713 GZ, the Netherlands.
| |
Collapse
|