101
|
Sandoval KC, Thackray SE, Wong A, Niewinski N, Chipak C, Rehal S, Dyck RH. Lack of Vesicular Zinc Does Not Affect the Behavioral Phenotype of Polyinosinic:Polycytidylic Acid-Induced Maternal Immune Activation Mice. Front Behav Neurosci 2022; 16:769322. [PMID: 35273483 PMCID: PMC8902171 DOI: 10.3389/fnbeh.2022.769322] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 01/24/2022] [Indexed: 11/13/2022] Open
Abstract
Zinc is important in neural and synaptic development and neuronal transmission. Within the brain, zinc transporter 3 (ZnT3) is essential for zinc uptake into vesicles. Loss of vesicular zinc has been shown to produce neurodevelopmental disorder (NDD)-like behavior, such as decreased social interaction and increased anxiety- and repetitive-like behavior. Maternal immune activation (MIA) has been identified as an environmental factor for NDDs, such as autism spectrum disorders (ASDs) and schizophrenia (SZ), in offspring, which occurs during pregnancy when the mother’s immune system reacts to the exposure to viruses or infectious diseases. In this study, we investigated the interaction effect of a genetic factor [ZnT3 knockout (KO) mice] and an environmental factor (MIA). We induced MIA in pregnant female (dams) mice during mid-gestation, using polyinosinic:polycytidylic acid (polyI:C), which mimics a viral infection. Male and female ZnT3 KO and wild-type (WT) offspring were tested in five behavioral paradigms: Ultrasonic Vocalizations (USVs) at postnatal day 9 (P9), Open Field Test, Marble Burying Test, three-Chamber Social Test, and Pre-pulse Inhibition (PPI) in adulthood (P60–75). Our results indicate that loss of vesicular zinc does not result in enhanced ASD- and SZ-like phenotype compared to WT, nor does it show a more pronounced phenotype in male ZnT3 KO compared to female ZnT3 KO. Finally, MIA offspring demonstrated an ASD- and SZ-like phenotype only in specific behavioral tests: increased calls emitted in USVs and fewer marbles buried. Our results suggest that there is no interaction between the loss of vesicular zinc and MIA induction in the susceptibility to developing an ASD- and SZ-like phenotype.
Collapse
Affiliation(s)
- Katy Celina Sandoval
- Department of Psychology, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute (HBI), University of Calgary, Calgary, AB, Canada
- Alberta Children’s Hospital Research Institute (ACHRI), University of Calgary, Calgary, AB, Canada
| | - Sarah E. Thackray
- Department of Psychology, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute (HBI), University of Calgary, Calgary, AB, Canada
- Alberta Children’s Hospital Research Institute (ACHRI), University of Calgary, Calgary, AB, Canada
| | - Alison Wong
- Department of Psychology, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute (HBI), University of Calgary, Calgary, AB, Canada
- Alberta Children’s Hospital Research Institute (ACHRI), University of Calgary, Calgary, AB, Canada
| | - Nicole Niewinski
- Department of Psychology, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute (HBI), University of Calgary, Calgary, AB, Canada
- Alberta Children’s Hospital Research Institute (ACHRI), University of Calgary, Calgary, AB, Canada
| | - Colten Chipak
- Department of Psychology, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute (HBI), University of Calgary, Calgary, AB, Canada
- Alberta Children’s Hospital Research Institute (ACHRI), University of Calgary, Calgary, AB, Canada
| | - Suhkjinder Rehal
- Department of Psychology, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute (HBI), University of Calgary, Calgary, AB, Canada
- Alberta Children’s Hospital Research Institute (ACHRI), University of Calgary, Calgary, AB, Canada
| | - Richard H. Dyck
- Department of Psychology, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute (HBI), University of Calgary, Calgary, AB, Canada
- Alberta Children’s Hospital Research Institute (ACHRI), University of Calgary, Calgary, AB, Canada
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB, Canada
- *Correspondence: Richard H. Dyck,
| |
Collapse
|
102
|
李 正, 肖 贵, 何 春, 刘 霞, 樊 欣, 赵 妍, 王 念. Serum levels of insulin-like growth factor-1 and insulin-like growth factor binding protein-3 in children with autism spectrum disorder. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2022; 24:186-191. [PMID: 35209985 PMCID: PMC8884054 DOI: 10.7499/j.issn.1008-8830.2110020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 11/15/2021] [Indexed: 06/14/2023]
Abstract
OBJECTIVES To study the serum levels of insulin-like growth factor-1 (IGF-1) and insulin-like growth factor binding protein-3 (IGFBP-3) in children with autism spectrum disorder (ASD) and their association with the core symptoms of ASD. METHODS A total of 150 ASD children aged 2-7 years (ASD group) and 165 healthy children matched for age and sex (control group) who were recruited at the outpatient service of Chongqing Health Center for Women and Children were enrolled as subjects. Autism Behavior Checklist (ABC) and Childhood Autism Rating Scale (CARS) were used to evaluate the core symptoms of the ASD children. Chemiluminescence was used to measure the serum levels of IGF-1 and IGFBP-3 in both groups. RESULTS The ASD group had a significantly lower serum level of IGF-1 than the control group (P<0.05). The children with severe ASD had significantly lower serum levels of IGF-1 and IGFBP-3 than those with mild-to-moderate ASD (P<0.001). For the children aged 2-3 years, the ASD group had a significantly lower serum level of IGF-1 than the control group (P<0.05). Boys had a significantly lower serum level of IGF-1 than girls in both ASD and control groups (P<0.05). The serum levels of IGF-1 and IGFBP-3 were negatively correlated with the total score of CARS (r=-0.32 and -0.40 respectively, P<0.001). CONCLUSIONS The reduction in serum IGF-1 level in early childhood may be associated with the development of ASD, and the serum levels of IGF-1 and IGFBP-3 are associated with the core symptoms of ASD children.
Collapse
|
103
|
Increased Monocyte Production of IL-6 after Toll-like Receptor Activation in Children with Autism Spectrum Disorder (ASD) Is Associated with Repetitive and Restricted Behaviors. Brain Sci 2022; 12:brainsci12020220. [PMID: 35203983 PMCID: PMC8870658 DOI: 10.3390/brainsci12020220] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 01/31/2022] [Accepted: 02/02/2022] [Indexed: 01/27/2023] Open
Abstract
The prevalence of autism spectrum disorder (ASD) has starkly increased, instigating research into risk factors for ASD. This research has identified immune risk factors for ASD, along with evidence of immune dysfunction and excess inflammation frequently experienced by autistic individuals. Increased innate inflammatory cytokines, including interleukin (IL)-6, are seen repeatedly in ASD; however, the origin of excess IL-6 in ASD has not been identified. Here we explore specific responses of circulating monocytes from autistic children. We isolated CD14+ monocytes from whole blood and stimulated them for 24 h under three conditions: media alone, lipoteichoic acid to activate TLR2, and lipopolysaccharide to activate TLR4. We then measured secreted cytokine concentrations in cellular supernatant using a human multiplex bead immunoassay. We found that after TLR4 activation, CD14+ monocytes from autistic children produce increased IL-6 compared to monocytes from children with typical development. IL-6 concentration also correlated with worsening restrictive and repetitive behaviors. These findings suggest dysfunctional activation of myeloid cells, and may indicate that other cells of this lineage, including macrophages, and microglia in the brain, might have a similar dysfunction. Further research on myeloid cells in ASD is warranted.
Collapse
|
104
|
Vanderplow AM, Kermath BA, Bernhardt CR, Gums KT, Seablom EN, Radcliff AB, Ewald AC, Jones MV, Baker TL, Watters JJ, Cahill ME. A feature of maternal sleep apnea during gestation causes autism-relevant neuronal and behavioral phenotypes in offspring. PLoS Biol 2022; 20:e3001502. [PMID: 35113852 PMCID: PMC8812875 DOI: 10.1371/journal.pbio.3001502] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 11/29/2021] [Indexed: 12/26/2022] Open
Abstract
Mounting epidemiologic and scientific evidence indicates that many psychiatric disorders originate from a complex interplay between genetics and early life experiences, particularly in the womb. Despite decades of research, our understanding of the precise prenatal and perinatal experiences that increase susceptibility to neurodevelopmental disorders remains incomplete. Sleep apnea (SA) is increasingly common during pregnancy and is characterized by recurrent partial or complete cessations in breathing during sleep. SA causes pathological drops in blood oxygen levels (intermittent hypoxia, IH), often hundreds of times each night. Although SA is known to cause adverse pregnancy and neonatal outcomes, the long-term consequences of maternal SA during pregnancy on brain-based behavioral outcomes and associated neuronal functioning in the offspring remain unknown. We developed a rat model of maternal SA during pregnancy by exposing dams to IH, a hallmark feature of SA, during gestational days 10 to 21 and investigated the consequences on the offspring's forebrain synaptic structure, synaptic function, and behavioral phenotypes across multiples stages of development. Our findings represent a rare example of prenatal factors causing sexually dimorphic behavioral phenotypes associated with excessive (rather than reduced) synapse numbers and implicate hyperactivity of the mammalian target of rapamycin (mTOR) pathway in contributing to the behavioral aberrations. These findings have implications for neuropsychiatric disorders typified by superfluous synapse maintenance that are believed to result, at least in part, from largely unknown insults to the maternal environment.
Collapse
Affiliation(s)
- Amanda M. Vanderplow
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Bailey A. Kermath
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Cassandra R. Bernhardt
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Kimberly T. Gums
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Erin N. Seablom
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Abigail B. Radcliff
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Andrea C. Ewald
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Mathew V. Jones
- Department of Neuroscience, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Tracy L. Baker
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Jyoti J. Watters
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Michael E. Cahill
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| |
Collapse
|
105
|
Jash S, Sharma S. Pathogenic Infections during Pregnancy and the Consequences for Fetal Brain Development. Pathogens 2022; 11:pathogens11020193. [PMID: 35215136 PMCID: PMC8877441 DOI: 10.3390/pathogens11020193] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/21/2022] [Accepted: 01/27/2022] [Indexed: 12/10/2022] Open
Abstract
Pathogens comprised of viruses, bacteria, gut microbiome, and parasites are a leading cause of ever-emerging diseases in humans. Studying pathogens for their ability to cause diseases is a topic of critical discussion among scientists and pharmaceutical centers for effective drug development that diagnose, treat, and prevent infection-associated disorders. Pathogens impact health either directly by invading the host or by eliciting an acute inflammatory immune response. This paradigm of inflammatory immune responses is even more consequential in people who may be immunocompromised. In this regard, pregnancy offers an altered immunity scenario, which may allow the onset of severe diseases. Viruses, such as Influenza, HIV, and now SARS-CoV-2, associated with the COVID-19 pandemic, raise new concerns for maternal and fetal/neonatal health. Intrauterine bacterial and parasitic infections are also known to impact pregnancy outcomes and neonatal health. More importantly, viral and bacterial infections during pregnancy have been identified as a common contributor to fetal brain development defects. Infection-mediated inflammatory uterine immune milieu is thought to be the main trigger for causing poor fetal brain development, resulting in long-term cognitive impairments. The concept of in utero programming of childhood and adult disorders has revolutionized the field of neurodevelopment and its associated complications. Recent findings in mice and humans clearly support the idea that uterine immunity during pregnancy controls the health trajectory of the child and considerably influences the cognitive function and mental health. In this review, we focus on the in utero programming of autism spectrum disorders (ASD) and assess the effects of pathogens on the onset of ASD-like symptoms.
Collapse
|
106
|
Jantsch J, Tassinari ID, Giovenardi M, Bambini-Junior V, Guedes RP, de Fraga LS. Mood Disorders Induced by Maternal Overnutrition: The Role of the Gut-Brain Axis on the Development of Depression and Anxiety. Front Cell Dev Biol 2022; 10:795384. [PMID: 35155424 PMCID: PMC8826230 DOI: 10.3389/fcell.2022.795384] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 01/10/2022] [Indexed: 11/13/2022] Open
Abstract
Since the first evidence suggesting that maternal nutrition can impact the development of diseases in the offspring, much has been elucidated about its effects on the offspring’s nervous system. Animal studies demonstrated that maternal obesity can predispose the offspring to greater chances of metabolic and neurodevelopmental diseases. However, the mechanisms underlying these responses are not well established. In recent years, the role of the gut-brain axis in the development of anxiety and depression in people with obesity has emerged. Studies investigating changes in the maternal microbiota during pregnancy and also in the offspring demonstrate that conditions such as maternal obesity can modulate the microbiota, leading to long-term outcomes in the offspring. Considering that maternal obesity has also been linked to the development of psychiatric conditions (anxiety and depression), the gut-brain axis is a promising target to be further explored in these neuropsychiatric contexts. In the present study, we review the relationship between maternal obesity and anxious and depressive features, exploring the gut-brain axis as a potential mechanism underlying this relationship.
Collapse
Affiliation(s)
- Jeferson Jantsch
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil
| | - Isadora D’Ávila Tassinari
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Márcia Giovenardi
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil
| | - Victorio Bambini-Junior
- School of Pharmacy and Biomedical Sciences, University of Central Lancashire (UCLan), Preston, United Kingdom
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster, United Kingdom
| | - Renata Padilha Guedes
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil
| | - Luciano Stürmer de Fraga
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- *Correspondence: Luciano Stürmer de Fraga,
| |
Collapse
|
107
|
Nordahl CW, Andrews DS, Dwyer P, Waizbard-Bartov E, Restrepo B, Lee JK, Heath B, Saron C, Rivera SM, Solomon M, Ashwood P, Amaral DG. The Autism Phenome Project: Toward Identifying Clinically Meaningful Subgroups of Autism. Front Neurosci 2022; 15:786220. [PMID: 35110990 PMCID: PMC8801875 DOI: 10.3389/fnins.2021.786220] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 12/20/2021] [Indexed: 12/23/2022] Open
Abstract
One of the most universally accepted facts about autism is that it is heterogenous. Individuals diagnosed with autism spectrum disorder have a wide range of behavioral presentations and a variety of co-occurring medical and mental health conditions. The identification of more homogenous subgroups is likely to lead to a better understanding of etiologies as well as more targeted interventions and treatments. In 2006, we initiated the UC Davis MIND Institute Autism Phenome Project (APP) with the overarching goal of identifying clinically meaningful subtypes of autism. This ongoing longitudinal multidisciplinary study now includes over 400 children and involves comprehensive medical, behavioral, and neuroimaging assessments from early childhood through adolescence (2-19 years of age). We have employed several strategies to identify sub-populations within autistic individuals: subgrouping by neural, biological, behavioral or clinical characteristics as well as by developmental trajectories. In this Mini Review, we summarize findings to date from the APP cohort and describe progress made toward identifying meaningful subgroups of autism.
Collapse
Affiliation(s)
- Christine Wu Nordahl
- MIND Institute, University of California, Davis, Davis, CA, United States
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Derek Sayre Andrews
- MIND Institute, University of California, Davis, Davis, CA, United States
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Patrick Dwyer
- Center for Mind and Brain, University of California, Davis, Davis, CA, United States
- Department of Psychology, University of California, Davis, Davis, CA, United States
| | - Einat Waizbard-Bartov
- MIND Institute, University of California, Davis, Davis, CA, United States
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Bibiana Restrepo
- MIND Institute, University of California, Davis, Davis, CA, United States
- Department of Pediatrics, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Joshua K. Lee
- MIND Institute, University of California, Davis, Davis, CA, United States
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Brianna Heath
- MIND Institute, University of California, Davis, Davis, CA, United States
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Clifford Saron
- MIND Institute, University of California, Davis, Davis, CA, United States
- Center for Mind and Brain, University of California, Davis, Davis, CA, United States
| | - Susan M. Rivera
- MIND Institute, University of California, Davis, Davis, CA, United States
- Center for Mind and Brain, University of California, Davis, Davis, CA, United States
- Department of Psychology, University of California, Davis, Davis, CA, United States
| | - Marjorie Solomon
- MIND Institute, University of California, Davis, Davis, CA, United States
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Paul Ashwood
- MIND Institute, University of California, Davis, Davis, CA, United States
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, Davis, CA, United States
| | - David G. Amaral
- MIND Institute, University of California, Davis, Davis, CA, United States
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California, Davis, Davis, CA, United States
| |
Collapse
|
108
|
Carter M, Casey S, O'Keeffe GW, Gibson L, Gallagher L, Murray DM. Maternal Immune Activation and Interleukin 17A in the Pathogenesis of Autistic Spectrum Disorder and Why It Matters in the COVID-19 Era. Front Psychiatry 2022; 13:823096. [PMID: 35250672 PMCID: PMC8891512 DOI: 10.3389/fpsyt.2022.823096] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 01/21/2022] [Indexed: 11/30/2022] Open
Abstract
Autism spectrum disorder (ASD) is the commonest neurodevelopmental disability. It is a highly complex disorder with an increasing prevalence and an unclear etiology. Consensus indicates that ASD arises as a genetically modulated, and environmentally influenced condition. Although pathogenic rare genetic variants are detected in around 20% of cases of ASD, no single factor is responsible for the vast majority of ASD cases or that explains their characteristic clinical heterogeneity. However, a growing body of evidence suggests that ASD susceptibility involves an interplay between genetic factors and environmental exposures. One such environmental exposure which has received significant attention in this regard is maternal immune activation (MIA) resulting from bacterial or viral infection during pregnancy. Reproducible rodent models of ASD are well-established whereby induction of MIA in pregnant dams, leads to offspring displaying neuroanatomical, functional, and behavioral changes analogous to those seen in ASD. Blockade of specific inflammatory cytokines such as interleukin-17A during gestation remediates many of these observed behavioral effects, suggesting a causative or contributory role. Here, we review the growing body of animal and human-based evidence indicating that interleukin-17A may mediate the observed effects of MIA on neurodevelopmental outcomes in the offspring. This is particularly important given the current corona virus disease-2019 (COVID-19) pandemic as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection during pregnancy is a potent stimulator of the maternal immune response, however the long-term effects of maternal SARS-CoV-2 infection on neurodevelopmental outcomes is unclear. This underscores the importance of monitoring neurodevelopmental outcomes in children exposed to SARS-CoV-2-induced MIA during gestation.
Collapse
Affiliation(s)
- Michael Carter
- INFANT Research Centre, University College Cork, Cork, Ireland.,Department of Paediatrics and Child Health, University College Cork, Cork, Ireland.,National Children's Research Centre, Dublin, Ireland
| | - Sophie Casey
- INFANT Research Centre, University College Cork, Cork, Ireland.,Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Gerard W O'Keeffe
- INFANT Research Centre, University College Cork, Cork, Ireland.,Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Louise Gibson
- INFANT Research Centre, University College Cork, Cork, Ireland.,Department of Paediatrics and Child Health, University College Cork, Cork, Ireland
| | - Louise Gallagher
- Department of Psychiatry, School of Medicine, Trinity College Dublin, Dublin, Ireland.,Trinity Translational Medicine Institute, St. James's Hospital, Dublin, Ireland
| | - Deirdre M Murray
- INFANT Research Centre, University College Cork, Cork, Ireland.,Department of Paediatrics and Child Health, University College Cork, Cork, Ireland
| |
Collapse
|
109
|
Gardella B, Dominoni M, Scatigno AL, Cesari S, Fiandrino G, Orcesi S, Spinillo A. What is known about neuroplacentology in fetal growth restriction and in preterm infants: A narrative review of literature. Front Endocrinol (Lausanne) 2022; 13:936171. [PMID: 36060976 PMCID: PMC9437342 DOI: 10.3389/fendo.2022.936171] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 07/11/2022] [Indexed: 11/13/2022] Open
Abstract
The placenta plays a fundamental role during pregnancy for fetal growth and development. A suboptimal placental function may result in severe consequences during the infant's first years of life. In recent years, a new field known as neuroplacentology has emerged and it focuses on the role of the placenta in fetal and neonatal brain development. Because of the limited data, our aim was to provide a narrative review of the most recent knowledge about the relation between placental lesions and fetal and newborn neurological development. Papers published online from 2000 until February 2022 were taken into consideration and particular attention was given to articles in which placental lesions were related to neonatal morbidity and short-term and long-term neurological outcome. Most research regarding the role of placental lesions in neurodevelopment has been conducted on fetal growth restriction and preterm infants. Principal neurological outcomes investigated were periventricular leukomalacia, intraventricular hemorrhages, neonatal encephalopathy and autism spectrum disorder. No consequences in motor development were found. All the considered studies agree about the crucial role played by placenta in fetal and neonatal neurological development and outcome. However, the causal mechanisms remain largely unknown. Knowledge on the pathophysiological mechanisms and on placenta-related risks for neurological problems may provide clues for early interventions aiming to improve neurological outcomes, especially among pediatricians and child psychiatrists.
Collapse
Affiliation(s)
- Barbara Gardella
- Department of Obstetrics and Gynecology, Fondazione Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San Matteo, Pavia, Italy
- Department of Obstetrics and Gynecology, University of Pavia, Pavia, Italy
- *Correspondence: Barbara Gardella,
| | - Mattia Dominoni
- Department of Obstetrics and Gynecology, Fondazione Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San Matteo, Pavia, Italy
- Department of Obstetrics and Gynecology, University of Pavia, Pavia, Italy
| | - Annachiara Licia Scatigno
- Department of Obstetrics and Gynecology, Fondazione Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San Matteo, Pavia, Italy
- Department of Obstetrics and Gynecology, University of Pavia, Pavia, Italy
| | - Stefania Cesari
- Department of Pathology, Fondazione Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San Matteo, Pavia, Italy
| | - Giacomo Fiandrino
- Department of Pathology, Fondazione Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San Matteo, Pavia, Italy
| | - Simona Orcesi
- Department of Brain and Behavioural Sciences, University of Pavia, Pavia, Italy
- Child Neurology and Psychiatry Unit, Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS) Mondino Foundation, Pavia, Italy
| | - Arsenio Spinillo
- Department of Obstetrics and Gynecology, Fondazione Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San Matteo, Pavia, Italy
- Department of Obstetrics and Gynecology, University of Pavia, Pavia, Italy
| |
Collapse
|
110
|
Maternal immune activation with high molecular weight poly(I:C) in Wistar rats leads to elevated immune cell chemoattractants. J Neuroimmunol 2022; 364:577813. [DOI: 10.1016/j.jneuroim.2022.577813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 12/19/2021] [Accepted: 01/10/2022] [Indexed: 11/20/2022]
|
111
|
Fujitani M, Miyajima H, Otani Y, Liu X. Maternal and Adult Interleukin-17A Exposure and Autism Spectrum Disorder. Front Psychiatry 2022; 13:836181. [PMID: 35211045 PMCID: PMC8861354 DOI: 10.3389/fpsyt.2022.836181] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 01/14/2022] [Indexed: 11/26/2022] Open
Abstract
Epidemiological evidence in humans has suggested that maternal infections and maternal autoimmune diseases are involved in the pathogenesis of autism spectrum disorder. Animal studies supporting human results have shown that maternal immune activation causes brain and behavioral alterations in offspring. Several underlying mechanisms, including interleukin-17A imbalance, have been identified. Apart from the pro-inflammatory effects of interleukin-17A, there is also evidence to support the idea that it activates neuronal function and defines cognitive behavior. In this review, we examined the signaling pathways in both immunological and neurological contexts that may contribute to the improvement of autism spectrum disorder symptoms associated with maternal blocking of interleukin-17A and adult exposure to interleukin-17A. We first describe the epidemiology of maternal immune activation then focus on molecular signaling of the interleukin-17 family regarding its physiological and pathological roles in the embryonic and adult brain. In the future, it may be possible to use interleukin-17 antibodies to prevent autism spectrum disorder.
Collapse
Affiliation(s)
- Masashi Fujitani
- Department of Anatomy and Neuroscience, Faculty of Medicine, Shimane University, Shimane, Japan
| | - Hisao Miyajima
- Department of Anatomy and Neuroscience, Faculty of Medicine, Shimane University, Shimane, Japan
| | - Yoshinori Otani
- Department of Anatomy and Neuroscience, Faculty of Medicine, Shimane University, Shimane, Japan
| | - Xinlang Liu
- Department of Anatomy and Neuroscience, Faculty of Medicine, Shimane University, Shimane, Japan
| |
Collapse
|
112
|
|
113
|
Chernikova MA, Flores GD, Kilroy E, Labus JS, Mayer EA, Aziz-Zadeh L. The Brain-Gut-Microbiome System: Pathways and Implications for Autism Spectrum Disorder. Nutrients 2021; 13:nu13124497. [PMID: 34960049 PMCID: PMC8704412 DOI: 10.3390/nu13124497] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/08/2021] [Accepted: 12/09/2021] [Indexed: 12/13/2022] Open
Abstract
Gastrointestinal dysfunction is one of the most prevalent physiological symptoms of autism spectrum disorder (ASD). A growing body of largely preclinical research suggests that dysbiotic gut microbiota may modulate brain function and social behavior, yet little is known about the mechanisms that underlie these relationships and how they may influence the pathogenesis or severity of ASD. While various genetic and environmental risk factors have been implicated in ASD, this review aims to provide an overview of studies elucidating the mechanisms by which gut microbiota, associated metabolites, and the brain interact to influence behavior and ASD development, in at least a subgroup of individuals with gastrointestinal problems. Specifically, we review the brain-gut-microbiome system and discuss findings from current animal and human studies as they relate to social-behavioral and neurological impairments in ASD, microbiota-targeted therapies (i.e., probiotics, fecal microbiota transplantation) in ASD, and how microbiota may influence the brain at molecular, structural, and functional levels, with a particular interest in social and emotion-related brain networks. A deeper understanding of microbiome-brain-behavior interactions has the potential to inform new therapies aimed at modulating this system and alleviating both behavioral and physiological symptomatology in individuals with ASD.
Collapse
Affiliation(s)
- Michelle A. Chernikova
- USC Chan Division of Occupational Science and Occupational Therapy, University of Southern California, Los Angeles, CA 90033, USA; (M.A.C.); (G.D.F.); (E.K.)
- Brain and Creativity Institute, University of Southern California, Los Angeles, CA 90089, USA
- Psychology Department, Loyola Marymount University, Los Angeles, CA 90045, USA
| | - Genesis D. Flores
- USC Chan Division of Occupational Science and Occupational Therapy, University of Southern California, Los Angeles, CA 90033, USA; (M.A.C.); (G.D.F.); (E.K.)
- Brain and Creativity Institute, University of Southern California, Los Angeles, CA 90089, USA
- Psychology Department, California State Polytechnic University, Pomona, CA 91768, USA
| | - Emily Kilroy
- USC Chan Division of Occupational Science and Occupational Therapy, University of Southern California, Los Angeles, CA 90033, USA; (M.A.C.); (G.D.F.); (E.K.)
- Brain and Creativity Institute, University of Southern California, Los Angeles, CA 90089, USA
| | - Jennifer S. Labus
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, University of California Los Angeles, Los Angeles, CA 90095, USA;
- Vatche and Tamar Manoukian Division of Digestive Diseases, University of California Los Angeles, Los Angeles, CA 90095, USA
- David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Gonda (Goldschmied) Neuroscience and Genetics Research Center, Brain Research Institute UCLA, Los Angeles, CA 90095, USA
| | - Emeran A. Mayer
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, University of California Los Angeles, Los Angeles, CA 90095, USA;
- Vatche and Tamar Manoukian Division of Digestive Diseases, University of California Los Angeles, Los Angeles, CA 90095, USA
- David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Correspondence: (E.A.M.); (L.A.-Z.)
| | - Lisa Aziz-Zadeh
- USC Chan Division of Occupational Science and Occupational Therapy, University of Southern California, Los Angeles, CA 90033, USA; (M.A.C.); (G.D.F.); (E.K.)
- Brain and Creativity Institute, University of Southern California, Los Angeles, CA 90089, USA
- Correspondence: (E.A.M.); (L.A.-Z.)
| |
Collapse
|
114
|
Maly IV, Morales MJ, Pletnikov MV. Astrocyte Bioenergetics and Major Psychiatric Disorders. ADVANCES IN NEUROBIOLOGY 2021; 26:173-227. [PMID: 34888836 DOI: 10.1007/978-3-030-77375-5_9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Ongoing research continues to add new elements to the emerging picture of involvement of astrocyte energy metabolism in the pathophysiology of major psychiatric disorders, including schizophrenia, mood disorders, and addictions. This review outlines what is known about the energy metabolism in astrocytes, the most numerous cell type in the brain, and summarizes the recent work on how specific perturbations of astrocyte bioenergetics may contribute to the neuropsychiatric conditions. The role of astrocyte energy metabolism in mental health and disease is reviewed on the organism, organ, and cell level. Data arising from genomic, metabolomic, in vitro, and neurobehavioral studies is critically analyzed to suggest future directions in research and possible metabolism-focused therapeutic interventions.
Collapse
Affiliation(s)
- Ivan V Maly
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York, Buffalo, NY, USA
| | - Michael J Morales
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York, Buffalo, NY, USA
| | - Mikhail V Pletnikov
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York, Buffalo, NY, USA.
| |
Collapse
|
115
|
Vlasova RM, Iosif AM, Ryan AM, Funk LH, Murai T, Chen S, Lesh TA, Rowland DJ, Bennett J, Hogrefe CE, Maddock RJ, Gandal MJ, Geschwind DH, Schumann CM, Van de Water J, McAllister AK, Carter CS, Styner MA, Amaral DG, Bauman MD. Maternal Immune Activation during Pregnancy Alters Postnatal Brain Growth and Cognitive Development in Nonhuman Primate Offspring. J Neurosci 2021; 41:9971-9987. [PMID: 34607967 PMCID: PMC8638691 DOI: 10.1523/jneurosci.0378-21.2021] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 07/28/2021] [Accepted: 09/06/2021] [Indexed: 11/21/2022] Open
Abstract
Human epidemiological studies implicate exposure to infection during gestation in the etiology of neurodevelopmental disorders. Animal models of maternal immune activation (MIA) have identified the maternal immune response as the critical link between maternal infection and aberrant offspring brain and behavior development. Here we evaluate neurodevelopment of male rhesus monkeys (Macaca mulatta) born to MIA-treated dams (n = 14) injected 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 same gestational time points (n = 10) or were untreated (n = 4). MIA-treated dams exhibited a strong immune response as indexed by transient increases in sickness behavior, temperature, and inflammatory cytokines. Although offspring born to control or MIA-treated dams did not differ on measures of physical growth and early developmental milestones, the MIA-treated animals exhibited subtle changes in cognitive development and deviated from species-typical brain growth trajectories. Longitudinal MRI revealed significant gray matter volume reductions in the prefrontal and frontal cortices of MIA-treated offspring at 6 months that persisted through the final time point at 45 months along with smaller frontal white matter volumes in MIA-treated animals at 36 and 45 months. These findings provide the first evidence of early postnatal changes in brain development in MIA-exposed nonhuman primates and establish a translationally relevant model system to explore the neurodevelopmental trajectory of risk associated with prenatal immune challenge from birth through late adolescence.SIGNIFICANCE STATEMENT Women exposed to infection during pregnancy have an increased risk of giving birth to a child who will later be diagnosed with a neurodevelopmental disorder. Preclinical maternal immune activation (MIA) models have demonstrated that the effects of maternal infection on fetal brain development are mediated by maternal immune response. Since the majority of MIA models are conducted in rodents, the nonhuman primate provides a unique system to evaluate the MIA hypothesis in a species closely related to humans. Here we report the first longitudinal study conducted in a nonhuman primate MIA model. MIA-exposed offspring demonstrate subtle changes in cognitive development paired with marked reductions in frontal gray and white matter, further supporting the association between prenatal immune challenge and alterations in offspring neurodevelopment.
Collapse
Affiliation(s)
- Roza M Vlasova
- Department of Psychiatry, University of North Carolina, Chapel Hill, North Carolina, 27514
| | - Ana-Maria Iosif
- Division of Biostatistics, Department of Public Health Sciences, School of Medicine, University of California, Davis, Sacramento, California, 95817
| | - Amy M Ryan
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California, Davis, Sacramento, California, 95817
- The MIND Institute, School of Medicine, University of California, Davis, Sacramento, California, 95817
- California National Primate Research Center, University of California, Davis, California, 95616
| | - Lucy H Funk
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California, Davis, Sacramento, California, 95817
| | - Takeshi Murai
- California National Primate Research Center, University of California, Davis, California, 95616
| | - Shuai Chen
- Division of Biostatistics, Department of Public Health Sciences, School of Medicine, University of California, Davis, Sacramento, California, 95817
| | - Tyler A Lesh
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California, Davis, Sacramento, California, 95817
| | - Douglas J Rowland
- Center for Genomic and Molecular Imaging, University of California, Davis, California, 95616
| | - Jeffrey Bennett
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California, Davis, Sacramento, California, 95817
| | - Casey E Hogrefe
- California National Primate Research Center, University of California, Davis, California, 95616
| | - Richard J Maddock
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California, Davis, Sacramento, California, 95817
| | - Michael J Gandal
- Neurogenetics Program, Department of Neurology, University of California, Los Angeles, California, 90095
| | - Daniel H Geschwind
- Neurogenetics Program, Department of Neurology, University of California, Los Angeles, California, 90095
| | - Cynthia M Schumann
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California, Davis, Sacramento, California, 95817
- The MIND Institute, School of Medicine, University of California, Davis, Sacramento, California, 95817
| | - Judy Van de Water
- The MIND Institute, School of Medicine, University of California, Davis, Sacramento, California, 95817
- Rheumatology/Allergy and Clinical Immunology, School of Medicine, University of California, Davis, Sacramento, California, 95817
| | - A Kimberley McAllister
- The MIND Institute, School of Medicine, University of California, Davis, Sacramento, California, 95817
- Center for Neuroscience, University of California, Davis, California, 95618
| | - Cameron S Carter
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California, Davis, Sacramento, California, 95817
| | - Martin A Styner
- Department of Psychiatry, University of North Carolina, Chapel Hill, North Carolina, 27514
- Department of Computer Science, University of North Carolina, Chapel Hill, North Carolina, 27599
| | - David G Amaral
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California, Davis, Sacramento, California, 95817
- The MIND Institute, School of Medicine, University of California, Davis, Sacramento, California, 95817
- California National Primate Research Center, University of California, Davis, California, 95616
| | - Melissa D Bauman
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California, Davis, Sacramento, California, 95817
- The MIND Institute, School of Medicine, University of California, Davis, Sacramento, California, 95817
- California National Primate Research Center, University of California, Davis, California, 95616
| |
Collapse
|
116
|
Taylor M, Pillaye J, Horsnell WGC. Inherent maternal type 2 immunity: Consequences for maternal and offspring health. Semin Immunol 2021; 53:101527. [PMID: 34838445 DOI: 10.1016/j.smim.2021.101527] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 11/10/2021] [Accepted: 11/10/2021] [Indexed: 02/08/2023]
Abstract
An inherent elevation in type 2 immunity is a feature of maternal and offspring immune systems. This has diverse implications for maternal and offspring biology including influencing success of pregnancy, offspring immune development and maternal and offspring ability to control infection and diseases such as allergies. In this review we provide a broad insight into how this immunological feature of pregnancy and early life impacts both maternal and offspring biology. We also suggest how understanding of this axis of immune influence is and may be utilised to improve maternal and offspring health.
Collapse
Affiliation(s)
- Matthew Taylor
- Institute of Immunology and Infection Research, Ashworth Laboratories, The Kings Buildings, University of Edinburgh, West Mains Road, Edinburgh, EH9 3JT, UK.
| | - Jamie Pillaye
- Institute of Microbiology and Infection, University of Birmingham, Birmingham, UK
| | - William Gordon Charles Horsnell
- Institute of Microbiology and Infection, University of Birmingham, Birmingham, UK; Wellcome Centre for Infectious Diseases Research in Africa (CIDRI-Africa), Institute of Infectious Disease and Molecular Medicine (IDM), Department of Pathology, Division of Immunology, Faculty of Health Science, University of Cape Town, Cape Town, 7925, South Africa.
| |
Collapse
|
117
|
MCP-1 Signaling Disrupts Social Behavior by Modulating Brain Volumetric Changes and Microglia Morphology. Mol Neurobiol 2021; 59:932-949. [PMID: 34797523 DOI: 10.1007/s12035-021-02649-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 11/15/2021] [Indexed: 10/19/2022]
Abstract
Autism spectrum disorder (ASD) is a disease characterized by reduced social interaction and stereotypic behaviors and related to macroscopic volumetric changes in cerebellar and somatosensory cortices (SPP). Epidemiological and preclinical models have confirmed that a proinflammatory profile during fetal development increases ASD susceptibility after birth. Here, we aimed to globally identify the effect of maternal exposure to high-energy dense diets, which we refer to as cafeteria diet (CAF) on peripheral and central proinflammatory profiles, microglia reactivity, and volumetric brain changes related to assisting defective social interaction in the mice offspring. We found a sex-dependent effect of maternal exposure to CAF diet or inoculation of the dsARN mimetic Poly (I:C) on peripheral proinflammatory and social interaction in the offspring. Notably, maternal exposure to CAF diet impairs social interaction and favors an increase in anxiety in male but not female offspring. Also, CAF diet exposure or Poly (I:C) inoculation during fetal programming promote peripheral proinflammatory profile in the ASD-diagnosed male but not in females. Selectively, we found a robust accumulation of the monocyte chemoattractant protein-1 (MCP-1) in plasma of ASD-diagnosed males exposed to CAF during fetal development. Biological assessment of MCP-1 signaling in brain confirms that systemic injection of MCP-1-neutralizing antibody reestablished social interaction and blocked anxiety, accompanied by a reduction in cerebellar lobule X (CbX) volume and an increase volume of the primary somatosensory (SSP) cortex in male offspring. These data highlight the contribution of diet-dependent MCP-1 signaling on volumetric brain changes and microglia morphology promoting ASD-like behavior in male mice.
Collapse
|
118
|
Carter M, Casey S, O’Keeffe GW, Gibson L, Murray DM. Mid-gestation cytokine profiles in mothers of children affected by autism spectrum disorder: a case-control study. Sci Rep 2021; 11:22315. [PMID: 34785716 PMCID: PMC8595633 DOI: 10.1038/s41598-021-01662-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 10/15/2021] [Indexed: 12/28/2022] Open
Abstract
Autism Spectrum disorder is one of the commonest and most important neurodevelopmental conditions affecting children today. With an increasing prevalence and an unclear aetiology, it is imperative we find early markers of autism, which may facilitate early identification and intervention. Alterations of gestational cytokine profiles have been reported in mothers of autistic children. Increasing evidence suggests that the intrauterine environment is an important determinant of autism risk. This study aims to examine the mid-gestational serum cytokine profiles of the mothers of autistic children from a well-characterised birth cohort. A nested sub-cohort within a large mother-child birth cohort were identified based on a confirmed multi-disciplinary diagnosis of autism before the age 10 years and neuro-typical matched controls in a 2:1 ratio. IFN-γ, IL-1β, IL-4, IL-6, IL-8, IL-17A, GMCSF and TNFα were measured in archived maternal 20-week serum using MesoScale Diagnostics multiplex technology and validation of our IL-17A measurements was performed using an ultrasensitive assay. From a cohort of 2137 children, 25 had confirmed autism before 10 years and stored maternal serum from mid-gestation. We examined the sera of these 25 cases and 50 matched controls. The sex ratio was 4:1 males to females in each group, and the mean age at diagnosis was 5.09 years (SD 2.13). We found that concentrations of IL-4 were significantly altered between groups. The other analytes did not differ significantly using either multiplex or ultra-sensitive assays. In our well-characterised prospective cohort of autistic children, we confirmed mid-gestational alterations in maternal IL-4 concentrations in autism affected pregnancies versus matched controls. These findings add to promising evidence from animal models and retrospective screening programmes and adds to the knowledge in this field.
Collapse
Affiliation(s)
- Michael Carter
- The Irish Centre for Maternal and Child Health Research, University College Cork, Cork, Ireland. .,National Children's Research Centre, Crumlin, Dublin 12, Ireland. .,Department of Paediatrics and Child Health, University College Cork (UCC), Cork, Ireland.
| | - Sophie Casey
- grid.7872.a0000000123318773The Irish Centre for Maternal and Child Health Research, University College Cork, Cork, Ireland ,grid.7872.a0000000123318773Department of Anatomy and Neuroscience, University College Cork (UCC), Cork, Ireland
| | - Gerard W. O’Keeffe
- grid.7872.a0000000123318773The Irish Centre for Maternal and Child Health Research, University College Cork, Cork, Ireland ,grid.7872.a0000000123318773Department of Anatomy and Neuroscience, University College Cork (UCC), Cork, Ireland
| | - Louise Gibson
- grid.7872.a0000000123318773The Irish Centre for Maternal and Child Health Research, University College Cork, Cork, Ireland ,grid.7872.a0000000123318773Department of Paediatrics and Child Health, University College Cork (UCC), Cork, Ireland
| | - Deirdre M. Murray
- grid.7872.a0000000123318773The Irish Centre for Maternal and Child Health Research, University College Cork, Cork, Ireland ,grid.7872.a0000000123318773Department of Paediatrics and Child Health, University College Cork (UCC), Cork, Ireland
| |
Collapse
|
119
|
O'Connor TG, Ciesla AA. Maternal Immune Activation Hypotheses for Human Neurodevelopment: Some Outstanding Questions. BIOLOGICAL PSYCHIATRY: COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2021; 7:471-479. [PMID: 34688920 PMCID: PMC9021321 DOI: 10.1016/j.bpsc.2021.10.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 10/05/2021] [Accepted: 10/13/2021] [Indexed: 12/17/2022]
Abstract
The Maternal Immune Activation (MIA) hypothesis is a leading model for understanding prenatal influences on individual differences in, and clinical syndromes of, neurodevelopment. Experimental animal and human research has proliferated in recent years, and there is now a sizable research base. Several meta-analyses demonstrate general support for an association between prenatal immune activation and neurodevelopment in human research. However, questions remain about the nature of the immune activation, the network of underlying mechanisms involved, and the breadth of impact across behavioral phenotypes. Complementing recent reviews of results, the current review places particular emphasis on how advances in understanding mechanisms may be improved with greater attention to addressing the methodological variation and limitations of existing studies, and identifies areas for further clinical research.
Collapse
Affiliation(s)
- Thomas G O'Connor
- Department of Psychiatry, University of Rochester; Department of Psycholog, University of Rochestery; Department of Neuroscience, University of Rochester; Department of Obstetrics and Gynecology, University of Rochester; Wynne Center for Family Research, University of Rochester.
| | | |
Collapse
|
120
|
Zhu B, Deng F, Yan S, Huang K, Wu X, Tao X, Wang S, Tao F. Gestational diabetes mellitus, autistic traits and ADHD symptoms in toddlers: Placental inflammatory and oxidative stress cytokines do not play an intermediary role. Psychoneuroendocrinology 2021; 134:105435. [PMID: 34649104 DOI: 10.1016/j.psyneuen.2021.105435] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 08/24/2021] [Accepted: 09/30/2021] [Indexed: 11/29/2022]
Abstract
OBJECTIVE To evaluate whether gestational diabetes mellitus (GDM) is associated with increased risks of autistic traits and attention deficit/hyperactivity disorder (ADHD) among offspring and whether placental inflammatory and oxidative stress cytokines play an intermediary role. METHODS Based on a prospective cohort study from China, namely, the Ma'anshan Birth Cohort study (MABC), 3260 mother-child pairs were included. Autistic traits and ADHD symptoms among children were assessed at 18 months and 36 months, respectively. The mRNA expression levels of fourteen placental cytokines were determined using PCR. Logistic regression analysis was used to examine the associations between GDM and the risks of autistic traits or ADHD symptoms. Mediation analysis was used to assess the potential mediation effects of certain placental inflammatory factors. RESULTS Of the 3260 children, 419 (12.85%) were exposed to GDM. The prevalence rates of autistic traits and ADHD symptoms were 13.86% and 6.4%, respectively. A 48.6% increased risk of autistic traits was observed among offspring born to mothers with GDM [odds ratio (OR) = 1.49, 95% confidence interval (95%CI): 1.11-2.00)], while no significant association was found in terms of ADHD symptoms. There were significant positive associations between GDM and IL-10 expression and between HIF1-α and CRP mRNA expression and a significant negative association between GDM and CD206 mRNA expression. The expression of MCP-1 mRNA was negatively associated with the risk of autistic traits [adjusted OR = 0.73 (95%CI: 0.73-0.55)]. The levels of TNF-α were positively associated with the risk of ADHD symptoms [OR = 2.11 (95%CI: 1.39-3.21)], while GRP78 was inversely associated with it [OR = 0.64 (95%CI: 0.44-0.94)]. However, none of the 14 placental cytokines was involved as a key mediator. CONCLUSION Our findings suggest that GDM may act as a risk factor for autistic traits in offspring, while the biological mechanisms may not involve the 14 placental cytokines studied. No significant association between GDM and ADHD symptoms was observed.
Collapse
Affiliation(s)
- Beibei Zhu
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, No. 81 Meishan Road, Hefei 230032, Anhui, China; Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University; No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Fen Deng
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, No. 81 Meishan Road, Hefei 230032, Anhui, China; Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University; No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Shuangqin Yan
- Ma'anshan Maternal and Child Health Care Center, Ma'anshan, China
| | - Kun Huang
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, No. 81 Meishan Road, Hefei 230032, Anhui, China; Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University; No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Xiaoyan Wu
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, No. 81 Meishan Road, Hefei 230032, Anhui, China; Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University; No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Xingyong Tao
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, No. 81 Meishan Road, Hefei 230032, Anhui, China; Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University; No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Sufang Wang
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, No. 81 Meishan Road, Hefei 230032, Anhui, China; Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University; No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Fangbiao Tao
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, No. 81 Meishan Road, Hefei 230032, Anhui, China; Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University; No 81 Meishan Road, Hefei 230032, Anhui, China.
| |
Collapse
|
121
|
Haddad FL, Lu L, Baines KJ, Schmid S. Sensory filtering disruption caused by poly I:C - Timing of exposure and other experimental considerations. Brain Behav Immun Health 2021; 9:100156. [PMID: 34589898 PMCID: PMC8474281 DOI: 10.1016/j.bbih.2020.100156] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 10/06/2020] [Indexed: 02/08/2023] Open
Abstract
Maternal immune activation (MIA) in response to infection during pregnancy has been linked through various epidemiological and preclinical studies to an increased risk of neurodevelopmental disorders such as autism spectrum disorder (ASD) and schizophrenia in exposed offspring. Sensory filtering disruptions occur in both of these disorders and are typically measured using the acoustic startle response in both humans and rodents. Our study focuses on characterizing the baseline reactivity, habituation and prepulse inhibition (PPI) of the acoustic startle response following exposure to MIA. We induced MIA using polyinosinic: polycytidylic acid (poly I:C) at gestational day (GD) 9.5 or 14.5, and we tested sensory filtering phenotypes in adolescent and adult offspring. Our results show that startle reactivity was robustly increased in adult GD9.5 but not GD14.5 poly I:C offspring. In contrast to some previous studies, we found no consistent changes in short-term habituation, long-term habituation or prepulse inhibition of startle. Our study highlights the importance of MIA exposure timing and discusses sensory filtering phenotypes as they relate to ASD, schizophrenia and the poly I:C MIA model. Moreover, we analyze and discuss the potential impact of between- and within-litter variability on behavioural findings in poly I:C studies.
Collapse
Affiliation(s)
- Faraj L Haddad
- Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, The University of Western Ontario, Canada
| | - Lu Lu
- Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, The University of Western Ontario, Canada.,Department of Neurology, West China Hospital of Sichuan University, Chengdu, China
| | - Kelly J Baines
- Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, The University of Western Ontario, Canada
| | - Susanne Schmid
- Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, The University of Western Ontario, Canada
| |
Collapse
|
122
|
The urgent need for more basic research on SARS-Cov2 infection and vaccines in assessing potential psychoneurological effects using maternal immune activation (MIA) and other preclinical modeling. Brain Behav Immun 2021; 97:1-3. [PMID: 34217811 PMCID: PMC8247198 DOI: 10.1016/j.bbi.2021.06.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/09/2021] [Accepted: 06/12/2021] [Indexed: 11/20/2022] Open
Abstract
The rapid development and application of different SARS-Cov2 vaccines world-wide has resulted in impressive efficacy and protection from this deadly pandemic. However, the existence of different and continuously developing vaccine candidates coupled with the likelihood of continued application due to both waning immune responses and emergence of viral mutants, means that more basic research regarding their efficacy and continued application are needed. This is particularly true with use of preclinical models involving effects when given during pregnancy. The substantial body of data on the impact of maternal immune activation (MIA) on neurologic development and behavior in the progeny necessitates the need to have all vaccine candidates, particularly when inducing strong toll receptor (TLR) responses, involving these models. Use of other preclinical models involving autoimmunity and allergy coupled with incorporation of human modifying variables of aging and obesity should also be applied to better reflect the heterogeneity of the general population and potential off-target effects that may arise. Additionally, the use of human ACE2 receptor transgenic mouse models can shed insights given the differential tissues expression at different stages in development. However, to foster these types of basic research studies involving different vaccine products, initiatives must first be implemented and supported at the governmental level even while clinical data still accumulates.
Collapse
|
123
|
Maternal Mid-Gestation Cytokine Dysregulation in Mothers of Children with Autism Spectrum Disorder. J Autism Dev Disord 2021; 52:3919-3932. [PMID: 34505185 PMCID: PMC9349096 DOI: 10.1007/s10803-021-05271-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/01/2021] [Indexed: 12/25/2022]
Abstract
Autism spectrum disorder (ASD) is a developmental disorder characterised by deficits in social interactions and communication, with stereotypical and repetitive behaviours. Recent evidence suggests that maternal immune dysregulation may predispose offspring to ASD. Independent samples t-tests revealed downregulation of IL-17A concentrations in cases, when compared to controls, at both 15 weeks (p = 0.02), and 20 weeks (p = 0.02), which persisted at 20 weeks following adjustment for confounding variables. This adds to the growing body of evidence that maternal immune regulation may play a role in foetal neurodevelopment.
Collapse
|
124
|
Han VX, Patel S, Jones HF, Dale RC. Maternal immune activation and neuroinflammation in human neurodevelopmental disorders. Nat Rev Neurol 2021; 17:564-579. [PMID: 34341569 DOI: 10.1038/s41582-021-00530-8] [Citation(s) in RCA: 275] [Impact Index Per Article: 68.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/23/2021] [Indexed: 02/06/2023]
Abstract
Maternal health during pregnancy plays a major role in shaping health and disease risks in the offspring. The maternal immune activation hypothesis proposes that inflammatory perturbations in utero can affect fetal neurodevelopment, and evidence from human epidemiological studies supports an association between maternal inflammation during pregnancy and offspring neurodevelopmental disorders (NDDs). Diverse maternal inflammatory factors, including obesity, asthma, autoimmune disease, infection and psychosocial stress, are associated with an increased risk of NDDs in the offspring. In addition to inflammation, epigenetic factors are increasingly recognized to operate at the gene-environment interface during NDD pathogenesis. For example, integrated brain transcriptome and epigenetic analyses of individuals with NDDs demonstrate convergent dysregulated immune pathways. In this Review, we focus on the emerging human evidence for an association between maternal immune activation and childhood NDDs, including autism spectrum disorder, attention-deficit/hyperactivity disorder and Tourette syndrome. We refer to established pathophysiological concepts in animal models, including immune signalling across the placenta, epigenetic 'priming' of offspring microglia and postnatal immune-brain crosstalk. The increasing incidence of NDDs has created an urgent need to mitigate the risk and severity of these conditions through both preventive strategies in pregnancy and novel postnatal therapies targeting disease mechanisms.
Collapse
Affiliation(s)
- Velda X Han
- Kids Neuroscience Centre, The Children's Hospital at Westmead, 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, Singapore
| | - Shrujna Patel
- Kids Neuroscience Centre, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia.,The Children's Hospital at Westmead Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Hannah F Jones
- Kids Neuroscience Centre, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia.,Department of Neuroservices, Starship Children's Hospital, Auckland, New Zealand
| | - Russell C Dale
- Kids Neuroscience Centre, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia. .,The Children's Hospital at Westmead Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia. .,The Brain and Mind Centre, The University of Sydney, Sydney, New South Wales, Australia.
| |
Collapse
|
125
|
McColl ER, Piquette-Miller M. Viral model of maternal immune activation alters placental AMPK and mTORC1 signaling in rats. Placenta 2021; 112:36-44. [PMID: 34256323 DOI: 10.1016/j.placenta.2021.07.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 05/28/2021] [Accepted: 07/06/2021] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Maternal immune activation (MIA) is associated with neurodevelopmental disorders in offspring. We previously demonstrated that poly(I:C)-mediated MIA alters placental and fetal brain amino acid transporter expression in rats, which could potentially play a role in altered neurodevelopment; however, the mechanism(s) underlying these changes in amino acid transporter expression remain unknown. The objective of the current study was to investigate the mechanism(s) underlying poly(I:C)-mediated changes in the expression of the amino acid transporters in the placenta. METHODS Pregnant rats received poly(I:C) on gestational day 14 and placentas were collected 6 h later. Mass spectrometry-based proteomics of placentas was performed followed by pathway enrichment analysis. Activation of mTORC1 and its upstream regulator, AMPK, was investigated using immunoblotting. Finally, the role of mTORC1 and AMPK in regulating the expression and localization of the amino acid transporters EAAT2 and ASCT1 was investigated in the human choriocarcinoma cell line JAR. RESULTS The impact of poly(I:C) on the placental proteome was highly sexually dimorphic. While proteomics-based pathway enrichment analysis indicated enrichment of mTOR signaling in male placentas only, further investigation revealed inhibition of mTORC1 in both male and female placentas in addition to activation of AMPK. In vitro, activation of AMPK and inhibition of mTORC1 decreased membrane localization of EAAT2 and ASCT1. DISCUSSION Poly(I:C)-mediated MIA activates AMPK and inhibits mTORC1 in rat placenta, both of which decrease expression and membrane localization of EAAT2 and ASCT1 in vitro. Thus, AMPK/mTORC1 signaling could be a novel treatment target for alleviating MIA-mediated changes in placental amino acid transport.
Collapse
Affiliation(s)
- Eliza R McColl
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College St, Toronto, ON, M5S 3M2, Canada.
| | - Micheline Piquette-Miller
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College St, Toronto, ON, M5S 3M2, Canada.
| |
Collapse
|
126
|
Lee GA, Lin YK, Lai JH, Lo YC, Yang YCSH, Ye SY, Lee CJ, Wang CC, Chiang YH, Tseng SH. Maternal Immune Activation Causes Social Behavior Deficits and Hypomyelination in Male Rat Offspring with an Autism-Like Microbiota Profile. Brain Sci 2021; 11:1085. [PMID: 34439704 PMCID: PMC8391334 DOI: 10.3390/brainsci11081085] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 08/16/2021] [Accepted: 08/16/2021] [Indexed: 01/03/2023] Open
Abstract
Maternal immune activation (MIA) increases the risk of autism spectrum disorder (ASD) in offspring. Microbial dysbiosis is associated with ASD symptoms. However, the alterations in the brain-gut-microbiota axis in lipopolysaccharide (LPS)-induced MIA offspring remain unclear. Here, we examined the social behavior, anxiety-like and repetitive behavior, microbiota profile, and myelination levels in LPS-induced MIA rat offspring. Compared with control offspring, MIA male rat offspring spent less time in an active social interaction with stranger rats, displayed more anxiety-like and repetitive behavior, and had more hypomyelination in the prefrontal cortex and thalamic nucleus. A fecal microbiota analysis revealed that MIA offspring had a higher abundance of Alistipes, Fusobacterium, and Ruminococcus and a lower abundance of Coprococcus, Erysipelotrichaies, and Actinobacteria than control offspring, which is consistent with that of humans with ASD. The least absolute shrinkage and selection operator (LASSO) method was applied to determine the relative importance of the microbiota, which indicated that the abundance of Alistipes and Actinobacteria was the most relevant for the profile of defective social behavior, whereas Fusobacterium and Coprococcus was associated with anxiety-like and repetitive behavior. In summary, LPS-induced MIA offspring showed an abnormal brain-gut-microbiota axis with social behavior deficits, anxiety-like and repetitive behavior, hypomyelination, and an ASD-like microbiota profile.
Collapse
Affiliation(s)
- Gilbert Aaron Lee
- Department of Medical Research, Taipei Medical University Hospital, Taipei 110, Taiwan; (G.A.L.); (S.-Y.Y.)
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Yen-Kuang Lin
- Graduate Institute of Athletics and Coaching Science, National Taiwan Sport University, Taoyuan 333, Taiwan;
| | - Jing-Huei Lai
- Core Laboratory of Neuroscience, Office of R&D, Taipei Medical University, Taipei 110, Taiwan; (J.-H.L.); (Y.-H.C.)
- Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei 110, Taiwan
| | - Yu-Chun Lo
- PhD Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan;
| | - Yu-Chen S. H. Yang
- Joint Biobank, Office of Human Research, Taipei Medical University, Taipei 110, Taiwan;
| | - Syuan-You Ye
- Department of Medical Research, Taipei Medical University Hospital, Taipei 110, Taiwan; (G.A.L.); (S.-Y.Y.)
| | - Chia-Jung Lee
- PhD Program for Clinical Drug Discovery of Herbal Medicine, College of Pharmacy, Taipei Medical University, Taipei 110, Taiwan;
- Graduate Institute of Pharmacognosy Science, College of Pharmacy, Taipei Medical University, Taipei 110, Taiwan
| | - Ching-Chiung Wang
- Department of Pharmaceutical Sciences, Taipei Medical University, Taipei 110, Taiwan;
| | - Yung-Hsiao Chiang
- Core Laboratory of Neuroscience, Office of R&D, Taipei Medical University, Taipei 110, Taiwan; (J.-H.L.); (Y.-H.C.)
- Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei 110, Taiwan
- Department of Surgery, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- Graduate Institute of Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan
| | - Sung-Hui Tseng
- Department of Physical Medicine and Rehabilitation, School of Medicine, College of Medicine, Taipei Medical University, No. 250 Wu Hsing Street, Taipei 110, Taiwan
- Department of Physical Medicine and Rehabilitation, Taipei Medical University Hospital, Taipei 110, Taiwan
| |
Collapse
|
127
|
Alvares GA, Licari MK, Stevenson PG, Bebbington K, Cooper MN, Glasson EJ, Tan DW, Uljarević M, Varcin KJ, Wray J, Whitehouse AJO. Investigating associations between birth order and autism diagnostic phenotypes. J Child Psychol Psychiatry 2021; 62:961-970. [PMID: 33164221 DOI: 10.1111/jcpp.13349] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/05/2020] [Indexed: 12/29/2022]
Abstract
BACKGROUND Birth order effects have been linked to variability in intelligence, educational attainment and sexual orientation. First- and later-born children have been linked to an increased likelihood of an Autism Spectrum Disorder (ASD) diagnosis, with a smaller body of evidence implicating decreases in cognitive functioning with increased birth order. The present study investigated the potential association between birth order and ASD diagnostic phenotypes in a large and representative population sample. METHODS Data were obtained from an ongoing prospective diagnostic registry, collected between 1999 and 2017, including children (1-18 years of age, n = 5,404) diagnosed with ASD in the state of Western Australia. Children with ASD were ranked relative to sibling's birth to establish birth order within families at time of ASD diagnosis. Information reported to the registry by health professionals at the time of diagnostic evaluation included demographic and family characteristics, functional abilities and intellectual capacity. RESULTS Adaptive functioning and intelligence scores decreased with increasing birth order, with later-born children more likely to have an intellectual disability. Compared to first-born children with siblings, first-born children without siblings at the time of diagnosis also exhibited decreased cognitive functioning. CONCLUSIONS These findings demonstrate for the first time an association between increasing birth order and variability in ASD clinical phenotypes at diagnosis, with potential evidence of reproductive curtailment in children without siblings. Taken together, these findings have significant implications for advancing understanding about the potential mechanisms that contribute to heterogeneity in ASD clinical presentations as a function of birth order and family size.
Collapse
Affiliation(s)
- Gail A Alvares
- Telethon Kids Institute, The University of Western Australia, Perth, Western Australia, Australia
| | - Melissa K Licari
- Telethon Kids Institute, The University of Western Australia, Perth, Western Australia, Australia
| | - Paul G Stevenson
- Telethon Kids Institute, The University of Western Australia, Perth, Western Australia, Australia
| | - Keely Bebbington
- Telethon Kids Institute, The University of Western Australia, Perth, Western Australia, Australia
| | - Matthew N Cooper
- Telethon Kids Institute, The University of Western Australia, Perth, Western Australia, Australia
| | - Emma J Glasson
- Telethon Kids Institute, The University of Western Australia, Perth, Western Australia, Australia
| | - Diana W Tan
- School of Psychological Science, The University of Western Australia, Perth, Western Australia, Australia
| | - Mirko Uljarević
- Melbourne School of Psychological Sciences, Faculty of Medicine, Dentistry, and Health Sciences, University of Melbourne, Melbourne, Victoria, Australia
| | - Kandice J Varcin
- Telethon Kids Institute, The University of Western Australia, Perth, Western Australia, Australia
| | - John Wray
- Child and Adolescent Health Service, Western Australia Department of Health, Perth, WA, Australia
| | - Andrew J O Whitehouse
- Telethon Kids Institute, The University of Western Australia, Perth, Western Australia, Australia
| |
Collapse
|
128
|
Yang Y, Wang B, Zhong Z, Chen H, Ding W, Hoi MPM. Clonazepam attenuates neurobehavioral abnormalities in offspring exposed to maternal immune activation by enhancing GABAergic neurotransmission. Biochem Pharmacol 2021; 192:114711. [PMID: 34324871 DOI: 10.1016/j.bcp.2021.114711] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/23/2021] [Accepted: 07/23/2021] [Indexed: 10/20/2022]
Abstract
Ample evidence indicates that maternal immune activation (MIA) during gestation is linked to an increased risk for neurodevelopmental and psychiatric disorders, such as autism spectrum disorder (ASD), anxiety and depression, in offspring. However, the underlying mechanism for such a link remains largely elusive. Here, we performed RNA sequencing (RNA-seq) to examine the transcriptional profiles changes in mice in response to MIA and identified that the expression of Scn1a gene, encoding the pore-forming α-subunit of the brain voltage-gated sodium channel type-1 (NaV1.1) primarily in fast-spiking inhibitory interneurons, was significantly decreased in the medial prefrontal cortex (mPFC) of juvenile offspring after MIA. Moreover, diminished excitatory drive onto interneurons causes reduction of spontaneous gamma-aminobutyric acid (GABA)ergic neurotransmission in the mPFC of MIA offspring, leading to hyperactivity in this brain region. Remarkably, treatment with low-dose benzodiazepines clonazepam, an agonist of GABAA receptors, completely prevented the behavioral abnormalities, including stereotypies, social deficits, anxiety- and depression-like behavior, via increasing inhibitory neurotransmission as well as decreasing neural activity in the mPFC of MIA offspring. Our results demonstrate that decreased expression of NaV1.1 in the mPFC leads to abnormalities in maternal inflammation-related behaviors and provides a potential therapeutic strategy for the abnormal behavioral phenotypes observed in the offspring exposed to MIA.
Collapse
Affiliation(s)
- Youjun Yang
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China; School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Baojia Wang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Zhanqion Zhong
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Hanbin Chen
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Weijun Ding
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Maggie Pui Man Hoi
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China.
| |
Collapse
|
129
|
Maternal effects in mammals: Broadening our understanding of offspring programming. Front Neuroendocrinol 2021; 62:100924. [PMID: 33992652 DOI: 10.1016/j.yfrne.2021.100924] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 04/18/2021] [Accepted: 05/11/2021] [Indexed: 12/17/2022]
Abstract
The perinatal period is a sensitive time in mammalian development that can have long-lasting consequences on offspring phenotype via maternal effects. Maternal effects have been most intensively studied with respect to two major conditions: maternal diet and maternal stress. In this review, we shift the focus by discussing five major additional maternal cues and their influence on offspring phenotype: maternal androgen levels, photoperiod (melatonin), microbiome, immune regulation, and milk composition. We present the key findings for each of these topics in mammals, their mechanisms of action, and how they interact with each other and with the maternal influences of diet and stress. We explore their impacts in the contexts of both predictive adaptive responses and the developmental origins of disease, identify knowledge gaps and research opportunities in the field, and place a particular emphasis on the application and consideration of these effects in non-model species and natural ecological systems.
Collapse
|
130
|
Novel treatments for autism spectrum disorder based on genomics and systems biology. Pharmacol Ther 2021; 230:107939. [PMID: 34174273 DOI: 10.1016/j.pharmthera.2021.107939] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 06/03/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND Autism spectrum disorder (ASD) is a highly heterogeneous neurodevelopmental disorder with a complex underlying genetic architecture. There are currently no known pharmacologic treatments for the core ASD symptoms of social deficits and restricted/ repetitive behavior. However, there are dozens of clinical trials currently underway that are testing the impact of novel and existing agents on core and associated symptoms in ASD. METHODS We present a narrative synthesis of the historical and contemporary challenges to drug discovery in ASD. We then provide an overview of novel treatments currently under investigation from a genomics and systems biology perspective. RESULTS Data driven network and cluster analyses suggest alterations in transcriptional regulation, chromatin remodelling, synaptic transmission, neuropeptide signalling, and/or immunological mechanisms may contribute to or underlie the development of ASD. Agents and upcoming trials targeting each of the above listed systems are reviewed. CONCLUSION Identifying effective pharmacologic treatments for the core and associated symptom domains in ASD will require further collaboration and innovation in the areas of outcome measurement, biomarker research, and genomics, as well as systematic efforts to identify and treat subgroups of individuals with ASD who may be differentially responsive to specific treatments.
Collapse
|
131
|
Rescue of maternal immune activation-induced behavioral abnormalities in adult mouse offspring by pathogen-activated maternal T reg cells. Nat Neurosci 2021; 24:818-830. [PMID: 33859437 DOI: 10.1038/s41593-021-00837-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 03/10/2021] [Indexed: 02/01/2023]
Abstract
Maternal immune activation (MIA) induced by lipopolysaccharides or polyinosinic:polycytidylic acid injections can induce behavioral abnormalities in adult mouse offspring. Here, we used the soluble tachyzoite antigen from Toxoplasma gondii, a parasite that infects approximately two billion people, to induce MIA in mice. The adult male offspring showed autism-relevant behaviors and abnormal brain microstructure, along with a pro-inflammatory T-cell immune profile in the periphery and upregulation of interleukin-6 in brain astrocytes. We show that adoptive transfer of regulatory T (Treg) cells largely reversed these MIA-induced phenotypes. Notably, pathogen-activated maternal Treg cells showed greater rescue efficacy than those from control donors. Single-cell RNA sequencing identified and characterized a unique group of pathogen-activated Treg cells that constitute 32.6% of the pathogen-activated maternal Treg population. Our study establishes a new preclinical parasite-mimicking MIA model and suggests therapeutic potential of adoptive Treg cell transfer in neuropsychiatric disorders associated with immune alterations.
Collapse
|
132
|
Feng S, Huang H, Wang N, Wei Y, Liu Y, Qin D. Sleep Disorders in Children With Autism Spectrum Disorder: Insights From Animal Models, Especially Non-human Primate Model. Front Behav Neurosci 2021; 15:673372. [PMID: 34093147 PMCID: PMC8173056 DOI: 10.3389/fnbeh.2021.673372] [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: 02/27/2021] [Accepted: 04/16/2021] [Indexed: 02/05/2023] Open
Abstract
Autism Spectrum Disorder (ASD) is a heterogeneous neurodevelopmental disorder with deficient social skills, communication deficits and repetitive behaviors. The prevalence of ASD has increased among children in recent years. Children with ASD experience more sleep problems, and sleep appears to be essential for the survival and integrity of most living organisms, especially for typical synaptic development and brain plasticity. Many methods have been used to assess sleep problems over past decades such as sleep diaries and parent-reported questionnaires, electroencephalography, actigraphy and videosomnography. A substantial number of rodent and non-human primate models of ASD have been generated. Many of these animal models exhibited sleep disorders at an early age. The aim of this review is to examine and discuss sleep disorders in children with ASD. Toward this aim, we evaluated the prevalence, clinical characteristics, phenotypic analyses, and pathophysiological brain mechanisms of ASD. We highlight the current state of animal models for ASD and explore their implications and prospects for investigating sleep disorders associated with ASD.
Collapse
Affiliation(s)
- Shufei Feng
- Department of Pediatric Rehabilitation Medicine, Kunming Children’s Hospital, Kunming, China
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, China
| | - Haoyu Huang
- Department of Pediatric Rehabilitation Medicine, Kunming Children’s Hospital, Kunming, China
| | - Na Wang
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, China
| | - Yuanyuan Wei
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, China
| | - Yun Liu
- Department of Pediatric Rehabilitation Medicine, Kunming Children’s Hospital, Kunming, China
| | - Dongdong Qin
- Department of Pediatric Rehabilitation Medicine, Kunming Children’s Hospital, Kunming, China
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, China
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, China
| |
Collapse
|
133
|
Adjimann TS, Argañaraz CV, Soiza-Reilly M. Serotonin-related rodent models of early-life exposure relevant for neurodevelopmental vulnerability to psychiatric disorders. Transl Psychiatry 2021; 11:280. [PMID: 33976122 PMCID: PMC8113523 DOI: 10.1038/s41398-021-01388-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 04/16/2021] [Accepted: 04/21/2021] [Indexed: 01/22/2023] Open
Abstract
Mental disorders including depression and anxiety are continuously rising their prevalence across the globe. Early-life experience of individuals emerges as a main risk factor contributing to the developmental vulnerability to psychiatric disorders. That is, perturbing environmental conditions during neurodevelopmental stages can have detrimental effects on adult mood and emotional responses. However, the possible maladaptive neural mechanisms contributing to such psychopathological phenomenon still remain poorly understood. In this review, we explore preclinical rodent models of developmental vulnerability to psychiatric disorders, focusing on the impact of early-life environmental perturbations on behavioral aspects relevant to stress-related and psychiatric disorders. We limit our analysis to well-established models in which alterations in the serotonin (5-HT) system appear to have a crucial role in the pathophysiological mechanisms. We analyze long-term behavioral outcomes produced by early-life exposures to stress and psychotropic drugs such as the selective 5-HT reuptake inhibitor (SSRI) antidepressants or the anticonvulsant valproic acid (VPA). We perform a comparative analysis, identifying differences and commonalities in the behavioral effects produced in these models. Furthermore, this review discusses recent advances on neurodevelopmental substrates engaged in these behavioral effects, emphasizing the possible existence of maladaptive mechanisms that could be shared by the different models.
Collapse
Affiliation(s)
- Tamara S. Adjimann
- grid.7345.50000 0001 0056 1981Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Carla V. Argañaraz
- grid.7345.50000 0001 0056 1981Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Mariano Soiza-Reilly
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina.
| |
Collapse
|
134
|
Page NF, Gandal MJ, Estes ML, Cameron S, Buth J, Parhami S, Ramaswami G, Murray K, Amaral DG, Van de Water JA, Schumann CM, Carter CS, Bauman MD, McAllister AK, Geschwind DH. Alterations in Retrotransposition, Synaptic Connectivity, and Myelination Implicated by Transcriptomic Changes Following Maternal Immune Activation in Nonhuman Primates. Biol Psychiatry 2021; 89:896-910. [PMID: 33386132 PMCID: PMC8052273 DOI: 10.1016/j.biopsych.2020.10.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 10/07/2020] [Accepted: 10/09/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND Maternal immune activation (MIA) is a proposed risk factor for multiple neuropsychiatric disorders, including schizophrenia. However, the molecular mechanisms through which MIA imparts risk remain poorly understood. A recently developed nonhuman primate model of exposure to the viral mimic poly:ICLC during pregnancy shows abnormal social and repetitive behaviors and elevated striatal dopamine, a molecular hallmark of human psychosis, providing an unprecedented opportunity for studying underlying molecular correlates. METHODS We performed RNA sequencing across psychiatrically relevant brain regions (prefrontal cortex, anterior cingulate, hippocampus) and primary visual cortex for comparison from 3.5- to 4-year-old male MIA-exposed and control offspring-an age comparable to mid adolescence in humans. RESULTS We identify 266 unique genes differentially expressed in at least one brain region, with the greatest number observed in hippocampus. Co-expression networks identified region-specific alterations in synaptic signaling and oligodendrocytes. Although we observed temporal and regional differences, transcriptomic changes were shared across first- and second-trimester exposures, including for the top differentially expressed genes-PIWIL2 and MGARP. In addition to PIWIL2, several other regulators of retrotransposition and endogenous transposable elements were dysregulated following MIA, potentially connecting MIA to retrotransposition. CONCLUSIONS Together, these results begin to elucidate the brain-level molecular processes through which MIA may impart risk for psychiatric disease.
Collapse
Affiliation(s)
- Nicholas F Page
- Department of Psychiatry, Center for Autism Research and Treatment, Los Angeles, California; Department of Cell Biology and Neuroscience, Rutgers University-New Brunswick, Piscataway, New Jersey
| | - Michael J Gandal
- Department of Psychiatry, Center for Autism Research and Treatment, Los Angeles, California
| | - Myka L Estes
- Center for Neuroscience, School of Medicine, University of California, Davis, Davis, California
| | - Scott Cameron
- Center for Neuroscience, School of Medicine, University of California, Davis, Davis, California
| | - Jessie Buth
- Department of Psychiatry, Center for Autism Research and Treatment, Los Angeles, California; Program in Neurobehavioral Genetics, Center for Autism Research and Treatment, Los Angeles, California
| | - Sepideh Parhami
- Department of Psychiatry, Center for Autism Research and Treatment, Los Angeles, California; Program in Neurobehavioral Genetics, Center for Autism Research and Treatment, Los Angeles, California
| | - Gokul Ramaswami
- Department of Psychiatry, Center for Autism Research and Treatment, Los Angeles, California; Program in Neurobehavioral Genetics, Center for Autism Research and Treatment, Los Angeles, California
| | - Karl Murray
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California, Davis, Davis, California
| | - David G Amaral
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California, Davis, Davis, California
| | - Judy A Van de Water
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California, Davis, Davis, California
| | - Cynthia M Schumann
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California, Davis, Davis, California
| | - Cameron S Carter
- Center for Neuroscience, School of Medicine, University of California, Davis, Davis, California; Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California, Davis, Davis, California
| | - Melissa D Bauman
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California, Davis, Davis, California
| | - A Kimberley McAllister
- Center for Neuroscience, School of Medicine, University of California, Davis, Davis, California; Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California, Davis, Davis, California
| | - Daniel H Geschwind
- Department of Psychiatry, Center for Autism Research and Treatment, Los Angeles, California; Program in Neurobehavioral Genetics, Center for Autism Research and Treatment, Los Angeles, California; Department of Neurology, Center for Autism Research and Treatment, Los Angeles, California; Department of Human Genetics, Semel Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California.
| |
Collapse
|
135
|
Meyer U. Maternal Immune Activation and Retrotransposition in Neurodevelopmental Disorders. Biol Psychiatry 2021; 89:842-844. [PMID: 33858590 DOI: 10.1016/j.biopsych.2021.02.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 02/09/2021] [Indexed: 12/31/2022]
Affiliation(s)
- Urs Meyer
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, and the Neuroscience Center Zurich, Zurich, Switzerland.
| |
Collapse
|
136
|
Inhibition of mechanistic target of rapamycin signaling decreases levels of O-GlcNAc transferase and increases serotonin release in the human placenta. Clin Sci (Lond) 2021; 134:3123-3136. [PMID: 33215629 DOI: 10.1042/cs20201050] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 11/02/2020] [Accepted: 11/20/2020] [Indexed: 12/13/2022]
Abstract
Changes in placental function, in particular down-regulation of placental O-linked N-acetylglucosamine (O-GlcNAc) transferase (OGT) in response to maternal stress and increased placental secretion of serotonin into the fetal circulation following maternal infection, have been mechanistically linked to adverse neurodevelopment in mice. We hypothesized that mechanistic target of rapamycin (mTOR) signaling is a key regulator of trophoblast serotonin synthesis and OGT protein expression and that serotonin is secreted by the human placenta into the fetal circulation. Placental homogenates (n=46) from elective terminations at 8-22 weeks of gestation and from healthy-term women were sexed and the protein levels of OGT and enzymes involved in serotonin synthesis was determined. Primary human trophoblast (PHT) cells were isolated from normal term placenta (n=27), cultured and transfected (n=8) with siRNA targeting a scramble sequence (control), raptor (inhibits mTOR Complex 1 (mTORC1)), or rictor (inhibits mTOR Complex 2 (mTORC2)). Subsequently, conditioned media and PHT cell lysates were collected. Free serotonin concentration was measured using ELISA in cell culture media and in platelet-depleted normal term umbilical vein and artery plasma (n=38). Both mTORC1 and mTORC2 inhibition down-regulated OGT levels in PHT cells. The level of serotonin synthesis enzyme tryptophan hydroxylase (TPH-1) was higher in early gestation female placentas and at term serotonin concentration was three-fold higher in the umbilical vein than in the umbilical artery. Inhibition of mTORC2, but not mTORC1, increased cultured PHT cell serotonin secretion. Our data are consistent with the model that mTOR signaling is a key regulator of trophoblast serotonin synthesis and OGT protein expression.
Collapse
|
137
|
Sheridan SD, Thanos JM, De Guzman RM, McCrea LT, Horng JE, Fu T, Sellgren CM, Perlis RH, Edlow AG. Umbilical cord blood-derived microglia-like cells to model COVID-19 exposure. Transl Psychiatry 2021; 11:179. [PMID: 33741894 PMCID: PMC7976669 DOI: 10.1038/s41398-021-01287-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 02/03/2021] [Indexed: 02/02/2023] Open
Abstract
Microglia, the resident brain immune cells, play a critical role in normal brain development, and are impacted by the intrauterine environment, including maternal immune activation and inflammatory exposures. The COVID-19 pandemic presents a potential developmental immune challenge to the fetal brain, in the setting of maternal SARS-CoV-2 infection with its attendant potential for cytokine production and, in severe cases, cytokine storming. There is currently no biomarker or model for in utero microglial priming and function that might aid in identifying the neonates and children most vulnerable to neurodevelopmental morbidity, as microglia remain inaccessible in fetal life and after birth. This study aimed to generate patient-derived microglial-like cell models unique to each neonate from reprogrammed umbilical cord blood mononuclear cells, adapting and extending a novel methodology previously validated for adult peripheral blood mononuclear cells. We demonstrate that umbilical cord blood mononuclear cells can be used to create microglial-like cell models morphologically and functionally similar to microglia observed in vivo. We illustrate the application of this approach by generating microglia from cells exposed and unexposed to maternal SARS-CoV-2 infection. Our ability to create personalized neonatal models of fetal brain immune programming enables non-invasive insights into fetal brain development and potential childhood neurodevelopmental vulnerabilities for a range of maternal exposures, including COVID-19.
Collapse
Affiliation(s)
- Steven D Sheridan
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Center for Genomic Medicine and Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Jessica M Thanos
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Center for Genomic Medicine and Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Rose M De Guzman
- Obstetrics, Gynecology, and Reproductive Biology, Harvard Medical School, Boston, MA, USA
- Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Liam T McCrea
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Center for Genomic Medicine and Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Joy E Horng
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Center for Genomic Medicine and Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Ting Fu
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Center for Genomic Medicine and Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Carl M Sellgren
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Roy H Perlis
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA.
- Center for Genomic Medicine and Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA.
| | - Andrea G Edlow
- Obstetrics, Gynecology, and Reproductive Biology, Harvard Medical School, Boston, MA, USA.
- Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
138
|
|
139
|
Gardner RM, Lee BK, Brynge M, Sjöqvist H, Dalman C, Karlsson H. Neonatal Levels of Acute Phase Proteins and Risk of Autism Spectrum Disorder. Biol Psychiatry 2021; 89:463-475. [PMID: 33187600 DOI: 10.1016/j.biopsych.2020.09.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 09/03/2020] [Accepted: 09/03/2020] [Indexed: 12/27/2022]
Abstract
BACKGROUND Immune signaling pathways influence neurodevelopment and are hypothesized to contribute to the etiology of autism spectrum disorder (ASD). We aimed to assess risk of ASD in relation to levels of neonatal acute phase proteins (APPs), key components of innate immune function, measured in neonatal dried blood spots. METHODS We included 924 ASD cases, 1092 unaffected population-based controls, and 203 unaffected siblings of ASD cases in this case-control study nested within the register-based Stockholm Youth Cohort. Concentrations of 9 different APPs were measured in eluates from neonatal dried blood spots from cases, controls, and siblings using a bead-based multiplex assay. RESULTS Neonatal C-reactive protein was consistently associated with odds of ASD in case-control comparisons, with higher odds associated with the highest quintile compared with the middle quintile (odds ratio [OR] = 1.50, 95% confidence interval [CI] = 1.10-2.04) in adjusted analyses. In contrast, the lowest quintiles of α-2-macroglobulin (OR = 3.71, CI = 1.21-11.33), ferritin (OR = 4.20, CI = 1.40-12.65), and serum amyloid P (OR = 3.05, CI = 1.16-8.01) were associated with odds of ASD in the matched sibling comparison. Neonatal APPs varied with perinatal environmental factors and maternal/fetal phenotypes. Significant interactions in terms of risk for ASD were observed between neonatal APPs and maternal infection during late pregnancy, maternal anemia, and maternal psychiatric history. CONCLUSIONS Indicators of the neonatal innate immune response are associated with risk of ASD, although the nature of these associations varies considerably with factors in the perinatal environment and the genetic background of the comparison group.
Collapse
Affiliation(s)
- Renee M Gardner
- Department of Global Public Health, Karolinska Institutet, Stockholm, Sweden.
| | - Brian K Lee
- Department of Global Public Health, Karolinska Institutet, Stockholm, Sweden; Department of Epidemiology and Biostatistics, Drexel University School of Public Health, Philadelphia, Pennsylvania; A.J. Drexel Autism Institute, Philadelphia, Pennsylvania
| | - Martin Brynge
- Department of Global Public Health, Karolinska Institutet, Stockholm, Sweden
| | - Hugo Sjöqvist
- Department of Global Public Health, Karolinska Institutet, Stockholm, Sweden
| | - Christina Dalman
- Department of Global Public Health, Karolinska Institutet, Stockholm, Sweden; Centre for Epidemiology and Community Medicine, Stockholm County Council, Stockholm, Sweden
| | - Håkan Karlsson
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
140
|
Leyrolle Q, Decoeur F, Briere G, Amadieu C, Quadros ARAA, Voytyuk I, Lacabanne C, Benmamar-Badel A, Bourel J, Aubert A, Sere A, Chain F, Schwendimann L, Matrot B, Bourgeois T, Grégoire S, Leblanc JG, De Moreno De Leblanc A, Langella P, Fernandes GR, Bretillon L, Joffre C, Uricaru R, Thebault P, Gressens P, Chatel JM, Layé S, Nadjar A. Maternal dietary omega-3 deficiency worsens the deleterious effects of prenatal inflammation on the gut-brain axis in the offspring across lifetime. Neuropsychopharmacology 2021; 46:579-602. [PMID: 32781459 PMCID: PMC8026603 DOI: 10.1038/s41386-020-00793-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 07/16/2020] [Accepted: 07/27/2020] [Indexed: 12/18/2022]
Abstract
Maternal immune activation (MIA) and poor maternal nutritional habits are risk factors for the occurrence of neurodevelopmental disorders (NDD). Human studies show the deleterious impact of prenatal inflammation and low n-3 polyunsaturated fatty acid (PUFA) intake on neurodevelopment with long-lasting consequences on behavior. However, the mechanisms linking maternal nutritional status to MIA are still unclear, despite their relevance to the etiology of NDD. We demonstrate here that low maternal n-3 PUFA intake worsens MIA-induced early gut dysfunction, including modification of gut microbiota composition and higher local inflammatory reactivity. These deficits correlate with alterations of microglia-neuron crosstalk pathways and have long-lasting effects, both at transcriptional and behavioral levels. This work highlights the perinatal period as a critical time window, especially regarding the role of the gut-brain axis in neurodevelopment, elucidating the link between MIA, poor nutritional habits, and NDD.
Collapse
Affiliation(s)
- Q. Leyrolle
- grid.488493.a0000 0004 0383 684XUniversity Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France ,Université de Paris, NeuroDiderot, Inserm, F-75019 Paris, France
| | - F. Decoeur
- grid.488493.a0000 0004 0383 684XUniversity Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - G. Briere
- grid.488493.a0000 0004 0383 684XUniversity Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France ,grid.503269.b0000 0001 2289 8198CNRS, Bordeaux INP, LaBRI, UMR 5800, F-33400 Talence, France
| | - C. Amadieu
- grid.488493.a0000 0004 0383 684XUniversity Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - A. R. A. A. Quadros
- grid.488493.a0000 0004 0383 684XUniversity Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - I. Voytyuk
- grid.488493.a0000 0004 0383 684XUniversity Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - C. Lacabanne
- grid.488493.a0000 0004 0383 684XUniversity Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - A. Benmamar-Badel
- grid.488493.a0000 0004 0383 684XUniversity Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - J. Bourel
- grid.488493.a0000 0004 0383 684XUniversity Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - A. Aubert
- grid.488493.a0000 0004 0383 684XUniversity Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - A. Sere
- grid.488493.a0000 0004 0383 684XUniversity Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - F. Chain
- grid.460789.40000 0004 4910 6535Micalis Institute, INRAE, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | - L. Schwendimann
- Université de Paris, NeuroDiderot, Inserm, F-75019 Paris, France
| | - B. Matrot
- Université de Paris, NeuroDiderot, Inserm, F-75019 Paris, France
| | - T. Bourgeois
- Université de Paris, NeuroDiderot, Inserm, F-75019 Paris, France
| | - S. Grégoire
- grid.462804.c0000 0004 0387 2525Centre des Sciences du Goût et de l’Alimentation, AgroSup Dijon, CNRS, INRAE, Université Bourgogne Franche-Comté, Dijon, France
| | - J. G. Leblanc
- CERELA-CONICET, San Miguel de Tucuman, 4000 Tucuman, Argentina
| | | | - P. Langella
- grid.460789.40000 0004 4910 6535Micalis Institute, INRAE, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | - G. R. Fernandes
- Rene Rachou Institute – Oswaldo Cruz Foundation, Belo Horizonte, MG Brazil
| | - L. Bretillon
- grid.462804.c0000 0004 0387 2525Centre des Sciences du Goût et de l’Alimentation, AgroSup Dijon, CNRS, INRAE, Université Bourgogne Franche-Comté, Dijon, France
| | - C. Joffre
- grid.488493.a0000 0004 0383 684XUniversity Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - R. Uricaru
- grid.503269.b0000 0001 2289 8198CNRS, Bordeaux INP, LaBRI, UMR 5800, F-33400 Talence, France
| | - P. Thebault
- grid.503269.b0000 0001 2289 8198CNRS, Bordeaux INP, LaBRI, UMR 5800, F-33400 Talence, France
| | - P. Gressens
- Université de Paris, NeuroDiderot, Inserm, F-75019 Paris, France ,grid.13097.3c0000 0001 2322 6764Centre for the Developing Brain, Department of Division of Imaging Sciences and Biomedical Engineering, King’s College London, King’s Health Partners, St. Thomas’ Hospital, London, SE1 7EH UK
| | - J. M. Chatel
- grid.460789.40000 0004 4910 6535Micalis Institute, INRAE, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | - S. Layé
- grid.488493.a0000 0004 0383 684XUniversity Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - A. Nadjar
- grid.488493.a0000 0004 0383 684XUniversity Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| |
Collapse
|
141
|
Mueller FS, Scarborough J, Schalbetter SM, Richetto J, Kim E, Couch A, Yee Y, Lerch JP, Vernon AC, Weber-Stadlbauer U, Meyer U. Behavioral, neuroanatomical, and molecular correlates of resilience and susceptibility to maternal immune activation. Mol Psychiatry 2021; 26:396-410. [PMID: 33230204 PMCID: PMC7850974 DOI: 10.1038/s41380-020-00952-8] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 10/24/2020] [Accepted: 11/04/2020] [Indexed: 12/13/2022]
Abstract
Infectious or noninfectious maternal immune activation (MIA) is an environmental risk factor for psychiatric and neurological disorders with neurodevelopmental etiologies. Whilst there is increasing evidence for significant health consequences, the effects of MIA on the offspring appear to be variable. Here, we aimed to identify and characterize subgroups of isogenic mouse offspring exposed to identical MIA, which was induced in C57BL6/N mice by administration of the viral mimetic, poly(I:C), on gestation day 12. Cluster analysis of behavioral data obtained from a first cohort containing >150 MIA and control offspring revealed that MIA offspring could be stratified into distinct subgroups that were characterized by the presence or absence of multiple behavioral dysfunctions. The two subgroups also differed in terms of their transcriptional profiles in cortical and subcortical brain regions and brain networks of structural covariance, as measured by ex vivo structural magnetic resonance imaging (MRI). In a second, independent cohort containing 50 MIA and control offspring, we identified a subgroup of MIA offspring that displayed elevated peripheral production of innate inflammatory cytokines, including IL-1β, IL-6, and TNF-α, in adulthood. This subgroup also showed significant impairments in social approach behavior and sensorimotor gating, whereas MIA offspring with a low inflammatory cytokine status did not. Taken together, our results highlight the existence of subgroups of MIA-exposed offspring that show dissociable behavioral, transcriptional, brain network, and immunological profiles even under conditions of genetic homogeneity. These data have relevance for advancing our understanding of the variable neurodevelopmental effects induced by MIA and for biomarker-guided approaches in preclinical psychiatric research.
Collapse
Affiliation(s)
- Flavia S Mueller
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland
| | - Joseph Scarborough
- 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
| | - Juliet Richetto
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Eugene Kim
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Amalie Couch
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Yohan Yee
- Mouse Imaging Centre, Hospital for Sick Children, Toronto, ON, Canada
| | - Jason P Lerch
- Mouse Imaging Centre, Hospital for Sick Children, Toronto, ON, Canada
- Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford, UK
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Anthony C Vernon
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - 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
| | - Urs Meyer
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland.
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
142
|
Cruz-Carrillo G, Camacho-Morales A. Metabolic Flexibility Assists Reprograming of Central and Peripheral Innate Immunity During Neurodevelopment. Mol Neurobiol 2021; 58:703-718. [PMID: 33006752 DOI: 10.1007/s12035-020-02154-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 09/28/2020] [Indexed: 01/03/2023]
Abstract
Central innate immunity assists time-dependent neurodevelopment by recruiting and interacting with peripheral immune cells. Microglia are the major player of central innate immunity integrating peripheral signals arising from the circumventricular regions lacking the blood-brain barrier (BBB), via neural afferent pathways such as the vagal nerve and also by choroid plexus into the brain ventricles. Defective and/or unrestrained activation of central and peripheral immunity during embryonic development might set an aberrant connectome establishment and brain function, leading to major psychiatric disorders in postnatal stages. Molecular candidates leading to central and peripheral innate immune overactivation identified metabolic substrates and lipid species as major contributors of immunological priming, supporting the role of a metabolic flexibility node during trained immunity. Mechanistically, trained immunity is established by an epigenetic program including DNA methylation and histone acetylation, as the major molecular epigenetic signatures to set immune phenotypes. By definition, immunological training sets reprogramming of innate immune cells, enhancing or repressing immune responses towards a second challenge which potentially might contribute to neurodevelopment disorders. Notably, the innate immune training might be set during pregnancy by maternal immune activation stimuli. In this review, we integrate the most valuable scientific evidence supporting the role of metabolic cues assisting metabolic flexibility, leading to innate immune training during development and its effects on aberrant neurological phenotypes in the offspring. We also add reports supporting the role of methylation and histone acetylation signatures as a major epigenetic mechanism regulating immune training.
Collapse
Affiliation(s)
- Gabriela Cruz-Carrillo
- Departamento de Bioquímica. Facultad de Medicina,, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Mexico
- Neurometabolism Unit, Center for Research and Development in Health Sciences, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, NL, Mexico
| | - Alberto Camacho-Morales
- Departamento de Bioquímica. Facultad de Medicina,, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Mexico.
- Neurometabolism Unit, Center for Research and Development in Health Sciences, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, NL, Mexico.
| |
Collapse
|
143
|
Zhang X, Ibi M, Haga R, Iwata K, Matsumoto M, Asaoka N, Liu J, Katsuyama M, Yabe-Nishimura C. NOX1/NADPH oxidase affects the development of autism-like behaviors in a maternal immune activation model. Biochem Biophys Res Commun 2021; 534:59-66. [PMID: 33310189 DOI: 10.1016/j.bbrc.2020.11.070] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 11/16/2020] [Indexed: 12/26/2022]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder caused by genetic and environmental factors. Among the environmental factors, maternal infection is known as one of the principal risk factors for ASD. On the other hand, postmortem studies suggested the relationship of oxidative stress with ASD etiology. However, the role of oxidative stress in the development of ASD remains unclear. Here, we report the involvement of NOX1/NADPH oxidase, an enzyme generating reactive oxygen species (ROS), in behavioral and anatomical abnormalities in a maternal immune activation (MIA) model. In the MIA model of gestational polyinosinic-polycytidylic acid (poly(I:C)) exposure, increased serum levels of IL-6 were observed in both wild-type (WT) and Nox1-deficient mice (Nox1KO). Following the comparable induction of MIA in the two genotypes, impairment of social preference and defects in motor coordination were observed in WT offspring but not in offspring deficient in Nox1. MIA up-regulated NOX1 mRNA in the cerebral cortex and cerebellum of the fetus but not in the adult offspring. Although the development of cortical neurons was unaffected by MIA in either genotype, the dropout of Purkinje cells in lobule VII of MIA-affected offspring was significantly ameliorated in Nox1KO. Taken together, these results suggested that NOX1/NADPH oxidase plays an essential role in some behavioral phenotypes observed in ASD, possibly by promoting the loss of Purkinje cells in the cerebellum.
Collapse
Affiliation(s)
- Xueqing Zhang
- Department of Pharmacology, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Masakazu Ibi
- Department of Pharmacology, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Ryu Haga
- Department of Pharmacology, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Kazumi Iwata
- Department of Pharmacology, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Misaki Matsumoto
- Department of Pharmacology, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Nozomi Asaoka
- Department of Pharmacology, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Junjie Liu
- Department of Pharmacology, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Masato Katsuyama
- Radioisotope Center, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Chihiro Yabe-Nishimura
- Department of Pharmacology, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan.
| |
Collapse
|
144
|
Healing autism spectrum disorder with cannabinoids: a neuroinflammatory story. Neurosci Biobehav Rev 2020; 121:128-143. [PMID: 33358985 DOI: 10.1016/j.neubiorev.2020.12.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 10/28/2020] [Accepted: 12/10/2020] [Indexed: 02/07/2023]
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder with a multifactorial etiology. Latest researches are raising the hypothesis of a link between the onset of the main behavioral symptoms of ASD and the chronic neuroinflammatory condition of the autistic brain; increasing evidence of this connection is shedding light on new possible players in the pathogenesis of ASD. The endocannabinoid system (ECS) has a key role in neurodevelopment as well as in normal inflammatory responses and it is not surprising that many preclinical and clinical studies account for alterations of the endocannabinoid signaling in ASD. These findings lay the foundation for a better understanding of the neurochemical mechanisms underlying ASD and for new therapeutic attempts aimed at exploiting the renowned anti-inflammatory properties of cannabinoids to treat pathologies encompassed in the autistic spectrum. This review discusses the current preclinical and clinical evidence supporting a key role of the ECS in the neuroinflammatory state that characterizes ASD, providing hints to identify new biomarkers in ASD and promising therapies for the future.
Collapse
|
145
|
Casanova EL, Baeza-Velasco C, Buchanan CB, Casanova MF. The Relationship between Autism and Ehlers-Danlos Syndromes/Hypermobility Spectrum Disorders. J Pers Med 2020; 10:E260. [PMID: 33271870 PMCID: PMC7711487 DOI: 10.3390/jpm10040260] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 11/29/2020] [Accepted: 11/29/2020] [Indexed: 12/12/2022] Open
Abstract
Considerable interest has arisen concerning the relationship between hereditary connective tissue disorders such as the Ehlers-Danlos syndromes (EDS)/hypermobility spectrum disorders (HSD) and autism, both in terms of their comorbidity as well as co-occurrence within the same families. This paper reviews our current state of knowledge, as well as highlighting unanswered questions concerning this remarkable patient group, which we hope will attract further scientific interest in coming years. In particular, patients themselves are demanding more research into this growing area of interest, although science has been slow to answer that call. Here, we address the overlap between these two spectrum conditions, including neurobehavioral, psychiatric, and neurological commonalities, shared peripheral neuropathies and neuropathologies, and similar autonomic and immune dysregulation. Together, these data highlight the potential relatedness of these two conditions and suggest that EDS/HSD may represent a subtype of autism.
Collapse
Affiliation(s)
- Emily L. Casanova
- School of Medicine Greenville, University of South Carolina, Greenville, SC 29615, USA;
| | - Carolina Baeza-Velasco
- Laboratory of Psychopathology and Health Processes, University of Paris, 92100 Boulogne Billancourt, France;
- Department of Emergency Psychiatry and Acute Care, CHU Montpellier, 34000 Montpellier, France
| | | | - Manuel F. Casanova
- School of Medicine Greenville, University of South Carolina, Greenville, SC 29615, USA;
- Department of Psychiatry and Behavioral Sciences, University of Louisville, Louisville, KY 40292, USA
| |
Collapse
|
146
|
Sandini TM, Greba Q, Lins BR, Howland JG. Maternal Immune Activation with the Viral Mimetic Poly:IC in Pregnant Rats. Bio Protoc 2020; 10:e3817. [PMID: 33659469 DOI: 10.21769/bioprotoc.3817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 08/24/2020] [Accepted: 08/31/2020] [Indexed: 11/02/2022] Open
Abstract
Maternal immune activation (MIA) is increasingly well appreciated as an environmental risk factor for some psychiatric disorders. Administration of proinflammatory compounds such as the synthetic double-stranded RNA molecule polyinosinic-polycytidylic acid (polyI:C) to pregnant rodents results in the release of proinflammatory cytokines in the maternal circulation. Various behavioural and brain changes are produced in the offspring that are associated with psychiatric disorders such as autism and schizophrenia. This protocol describes the steps necessary for inducing MIA in pregnant rat dams, which will allow for investigations into the mechanisms in the dam and offspring that mediate the long-term effects of exposure to inflammation while in utero. Increasing our understanding of these mechanisms may provide new insights for the diagnosis, treatment, and prevention of psychiatric disorders. This protocol has been developed and improved over the years by various researchers in Dr. Howland's laboratory at the University of Saskatchewan.
Collapse
Affiliation(s)
- Thaísa Meira Sandini
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, Canada
| | - Quentin Greba
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, Canada
| | - Brittney Rose Lins
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, Canada
| | - John George Howland
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, Canada
| |
Collapse
|
147
|
Holingue C, Brucato M, Ladd-Acosta C, Hong X, Volk H, Mueller NT, Wang X, Fallin MD. Interaction between Maternal Immune Activation and Antibiotic Use during Pregnancy and Child Risk of Autism Spectrum Disorder. Autism Res 2020; 13:2230-2241. [PMID: 33067915 DOI: 10.1002/aur.2411] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 09/28/2020] [Accepted: 09/29/2020] [Indexed: 12/11/2022]
Abstract
Prenatal exposure to maternal immune activation (MIA) has been implicated as a risk factor for the development of autism spectrum disorder (ASD), though the conditions under which this elevated risk occurs are unclear. Animal literature demonstrates that antibiotic use, which affects the composition of the maternal gut microbiota, modifies the effect of MIA on neurodevelopmental outcomes in the offspring. The aim of this study was to assess whether antibiotic use during pregnancy modifies the association between MIA and subsequent risk of ASD, in a prospective birth cohort with 116 ASD cases and 860 typically developing (TD) child controls. There was no evidence of interaction between fever or genitourinary infection and antibiotic use on the odds of ASD in unadjusted or adjusted analyzes. However, we found evidence of an interaction between flu, specifically in second trimester, and antibiotic use at any point during pregnancy on the odds of ASD in the child. Among women who received an antibiotic during pregnancy, flu in trimester two was not associated with ASD (adjusted odds ratio [aOR] = 0.99 [0.43-2.28]). Among women who were not exposed to an antibiotic at any point during pregnancy, flu in second trimester was significantly associated with increased odds of ASD (aOR = 4.05 [1.14-14.38], P = .03), after adjustment for child sex, child birth year, maternal age, gestational age, C-section delivery, and low birthweight. These findings should be treated as hypothesis-generating and suggest that antibiotic use may modify the influence that MIA has on autism risk in the child. LAY SUMMARY: We looked at whether the association between activation of the immune system during pregnancy and risk of the child developing autism spectrum disorder (ASD) differed among women who did or did not take an antibiotic at any point during pregnancy. We examined 116 children with ASD and 860 without ASD and found that flu in second trimester was associated with increased ASD, but only among women who did not take an antibiotic during pregnancy. No other immune activation exposures seemed to interact with antibiotic use.
Collapse
Affiliation(s)
- Calliope Holingue
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA.,Wendy Klag Center for Autism and Developmental Disabilities, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Martha Brucato
- Wendy Klag Center for Autism and Developmental Disabilities, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA.,Medical Scientist Training Program, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Christine Ladd-Acosta
- Wendy Klag Center for Autism and Developmental Disabilities, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA.,Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Xiumei Hong
- Department of Population, Family and Reproductive Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA.,The Center on the Early Life Origins of Disease, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Heather Volk
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA.,Wendy Klag Center for Autism and Developmental Disabilities, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Noel T Mueller
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Xiaobin Wang
- Department of Population, Family and Reproductive Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA.,The Center on the Early Life Origins of Disease, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - M Daniele Fallin
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA.,Wendy Klag Center for Autism and Developmental Disabilities, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| |
Collapse
|
148
|
Increased RNA editing in maternal immune activation model of neurodevelopmental disease. Nat Commun 2020; 11:5236. [PMID: 33067431 PMCID: PMC7567798 DOI: 10.1038/s41467-020-19048-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 09/23/2020] [Indexed: 12/19/2022] Open
Abstract
The etiology of major neurodevelopmental disorders such as schizophrenia and autism is unclear, with evidence supporting a combination of genetic factors and environmental insults, including viral infection during pregnancy. Here we utilized a mouse model of maternal immune activation (MIA) with the viral mimic PolyI:C infection during early gestation. We investigated the transcriptional changes in the brains of mouse fetuses following MIA during the prenatal period, and evaluated the behavioral and biochemical changes in the adult brain. The results reveal an increase in RNA editing levels and dysregulation in brain development-related gene pathways in the fetal brains of MIA mice. These MIA-induced brain editing changes are not observed in adulthood, although MIA-induced behavioral deficits are observed. Taken together, our findings suggest that MIA induces transient dysregulation of RNA editing at a critical time in brain development.
Collapse
|
149
|
Sheridan SD, Thanos JM, De Guzman RM, McCrea LT, Horng J, Fu T, Sellgren CM, Perlis RH, Edlow AG. Umbilical cord blood derived microglia-like cells to model COVID-19 exposure. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020:2020.10.07.329748. [PMID: 33052344 PMCID: PMC7553171 DOI: 10.1101/2020.10.07.329748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Microglia, the resident brain immune cells, play a critical role in normal brain development, and are impacted by the intrauterine environment, including maternal immune activation and inflammatory exposures. The COVID-19 pandemic presents a potential developmental immune challenge to the fetal brain, in the setting of maternal SARS-CoV-2 infection with its attendant potential for cytokine production and, in severe cases, cytokine storming. There is currently no biomarker or model for in utero microglial priming and function that might aid in identifying the neonates and children most vulnerable to neurodevelopmental morbidity, as microglia remain inaccessible in fetal life and after birth. This study aimed to generate patient-derived microglial-like cell models unique to each neonate from reprogrammed umbilical cord blood mononuclear cells, adapting and extending a novel methodology previously validated for adult peripheral blood mononuclear cells. We demonstrate that umbilical cord blood mononuclear cells can be used to create microglial-like cell models morphologically and functionally similar to microglia observed in vivo . We illustrate the application of this approach by generating microglia from cells exposed and unexposed to maternal SARS-CoV-2 infection. Our ability to create personalized neonatal models of fetal brain immune programming enables non-invasive insights into fetal brain development and potential childhood neurodevelopmental vulnerabilities for a range of maternal exposures, including COVID-19.
Collapse
|
150
|
Maternal immune activation alters visual acuity and retinogeniculate axon pruning in offspring mice. Brain Behav Immun 2020; 89:518-523. [PMID: 32827701 DOI: 10.1016/j.bbi.2020.08.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 08/16/2020] [Accepted: 08/17/2020] [Indexed: 01/29/2023] Open
Abstract
Individuals with autism spectrum disorder (ASD) have been found to have a variety of sensory processing deficits. Here we report that maternal immune activation, a known factor for ASD, alters visual acuity in the offspring mice. By intraperitoneally injecting polyinosinic-polycytidylic acid (polyI:C) to induce maternal immune activation during embryonic days 10 to 14, we found that polyI:C treatment impairs visual acuity in young adult offspring mice as examined by their optomotor responses. Concurrently, polyI:C treatment suppresses retinogeniculate axon elimination, resulting in a high fraction of weak optical fibers innervating the relay neurons in the visual thalamus. The results link in-utero maternal inflammation to defective optical fiber pruning and arrested developmental strengthening of single optic fibers which may underlie impaired visual acuity.
Collapse
|