51
|
Weiss JT, Donlea JM. Roles for Sleep in Neural and Behavioral Plasticity: Reviewing Variation in the Consequences of Sleep Loss. Front Behav Neurosci 2022; 15:777799. [PMID: 35126067 PMCID: PMC8810646 DOI: 10.3389/fnbeh.2021.777799] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 12/16/2021] [Indexed: 12/13/2022] Open
Abstract
Sleep is a vital physiological state that has been broadly conserved across the evolution of animal species. While the precise functions of sleep remain poorly understood, a large body of research has examined the negative consequences of sleep loss on neural and behavioral plasticity. While sleep disruption generally results in degraded neural plasticity and cognitive function, the impact of sleep loss can vary widely with age, between individuals, and across physiological contexts. Additionally, several recent studies indicate that sleep loss differentially impacts distinct neuronal populations within memory-encoding circuitry. These findings indicate that the negative consequences of sleep loss are not universally shared, and that identifying conditions that influence the resilience of an organism (or neuron type) to sleep loss might open future opportunities to examine sleep's core functions in the brain. Here, we discuss the functional roles for sleep in adaptive plasticity and review factors that can contribute to individual variations in sleep behavior and responses to sleep loss.
Collapse
Affiliation(s)
- Jacqueline T. Weiss
- Department of Neurobiology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA, United States
- Neuroscience Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA, United States
| | - Jeffrey M. Donlea
- Department of Neurobiology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA, United States
- *Correspondence: Jeffrey M. Donlea
| |
Collapse
|
52
|
Sato A, Kotajima-Murakami H, Tanaka M, Katoh Y, Ikeda K. Influence of Prenatal Drug Exposure, Maternal Inflammation, and Parental Aging on the Development of Autism Spectrum Disorder. Front Psychiatry 2022; 13:821455. [PMID: 35222122 PMCID: PMC8863673 DOI: 10.3389/fpsyt.2022.821455] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 01/12/2022] [Indexed: 12/17/2022] Open
Abstract
Autism spectrum disorder (ASD) affects reciprocal social interaction and produces abnormal repetitive, restrictive behaviors and interests. The diverse causes of ASD are divided into genetic alterations and environmental risks. The prevalence of ASD has been rising for several decades, which might be related to environmental risks as it is difficult to consider that the prevalence of genetic disorders related to ASD would increase suddenly. The latter includes (1) exposure to medications, such as valproic acid (VPA) and selective serotonin reuptake inhibitors (SSRIs) (2), maternal complications during pregnancy, including infection and hypertensive disorders of pregnancy, and (3) high parental age. Epidemiological studies have indicated a pathogenetic role of prenatal exposure to VPA and maternal inflammation in the development of ASD. VPA is considered to exert its deleterious effects on the fetal brain through several distinct mechanisms, such as alterations of γ-aminobutyric acid signaling, the inhibition of histone deacetylase, the disruption of folic acid metabolism, and the activation of mammalian target of rapamycin. Maternal inflammation that is caused by different stimuli converges on a higher load of proinflammatory cytokines in the fetal brain. Rodent models of maternal exposure to SSRIs generate ASD-like behavior in offspring, but clinical correlations with these preclinical findings are inconclusive. Hypertensive disorders of pregnancy and advanced parental age increase the risk of ASD in humans, but the mechanisms have been poorly investigated in animal models. Evidence of the mechanisms by which environmental factors are related to ASD is discussed, which may contribute to the development of preventive and therapeutic interventions for ASD.
Collapse
Affiliation(s)
- Atsushi Sato
- Department of Pediatrics, The University of Tokyo Hospital, Tokyo, Japan.,Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | | | - Miho Tanaka
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.,Department of Psychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yoshihisa Katoh
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.,Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kazutaka Ikeda
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| |
Collapse
|
53
|
Rocha-Gomes A, Teixeira AE, Santiago CMO, Oliveira DGD, Silva AAD, Lacerda ACR, Riul TR, Mendonça VA, Rocha-Vieira E, Leite HR. Prenatal LPS exposure increases hippocampus IL-10 and prevents short-term memory loss in the male adolescent offspring of high-fat diet fed dams. Physiol Behav 2022; 243:113628. [PMID: 34695488 DOI: 10.1016/j.physbeh.2021.113628] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 10/18/2021] [Accepted: 10/20/2021] [Indexed: 11/25/2022]
Abstract
Lipopolysaccharide (LPS) tolerance can reduce the neuroinflammation caused by high fat maternal diets; however, there are no reports that have evaluated the effects of prenatal LPS exposure on the memories of the offspring of high-fat diet fed dams. This study evaluated the effects of prenatal LPS exposure on the inflammatory parameters and redox status in the brain, as well as the object recognition memory of adolescent offspring of Wistar rat dams that were treated with a high-fat diet during gestation and lactation. Female pregnant Wistar rats randomly received a standard diet (17.5% fat) or a high-fat diet (45.0% fat) during gestation and lactation. On gestation days 8, 10, and 12, half of the females in each group were intraperitoneally treated with LPS (0.1 mg.kg-1). After weaning, the male offspring were placed in cages in standard conditions, and at 6 weeks old, animals underwent the novel object recognition test (for short- and long-term memory). The offspring of the high-fat diet fed dams showed increased hippocampus IL-6 levels (21-days-old) and impaired short-term memories. These effects were avoided in the offspring of high-fat diet fed dams submitted to prenatal LPS exposure, which showed greater hippocampus IL-10 levels (at 21- and 50-days-old), increased antioxidant activity (50-days-old) in the hippocampus and prefrontal cortex, without memory impairments (short- and long-term memory). IL-6 has been consistently implicated in memory deficits and as an endogenous mechanism for limiting plasticity, while IL-10 regulates glial activation and has a strong association with improvements in cognitive function. Prenatal LPS exposure preventing the increase of IL-6 in the hippocampus and the impairment to short-term object recognition memory caused by the high-fat maternal diet.
Collapse
Affiliation(s)
- Arthur Rocha-Gomes
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Sociedade Brasileira de Fisiologia, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, 39100-000 Brasil; Laboratório de Nutrição Experimental - LabNutrex - Departamento de Nutrição. Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brasil.
| | - Amanda Escobar Teixeira
- Laboratório de Nutrição Experimental - LabNutrex - Departamento de Nutrição. Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brasil
| | - Camilla Mainy Oliveira Santiago
- Laboratório de Nutrição Experimental - LabNutrex - Departamento de Nutrição. Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brasil; Programa de Pós-Graduação em Ciências da Nutrição, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brasil
| | - Dalila Gomes de Oliveira
- Laboratório de Nutrição Experimental - LabNutrex - Departamento de Nutrição. Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brasil
| | - Alexandre Alves da Silva
- Laboratório de Nutrição Experimental - LabNutrex - Departamento de Nutrição. Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brasil
| | - Ana Cristina Rodrigues Lacerda
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Sociedade Brasileira de Fisiologia, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, 39100-000 Brasil
| | - Tania Regina Riul
- Laboratório de Nutrição Experimental - LabNutrex - Departamento de Nutrição. Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brasil; Programa de Pós-Graduação em Ciências da Nutrição, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, Brasil
| | - Vanessa Amaral Mendonça
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Sociedade Brasileira de Fisiologia, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, 39100-000 Brasil
| | - Etel Rocha-Vieira
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Sociedade Brasileira de Fisiologia, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, 39100-000 Brasil
| | - Hércules Ribeiro Leite
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Sociedade Brasileira de Fisiologia, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG, 39100-000 Brasil; Departamento de Fisioterapia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, 31270-901 Brasil.
| |
Collapse
|
54
|
Erbescu A, Papuc SM, Budisteanu M, Arghir A, Neagu M. Re-emerging concepts of immune dysregulation in autism spectrum disorders. Front Psychiatry 2022; 13:1006612. [PMID: 36339838 PMCID: PMC9626859 DOI: 10.3389/fpsyt.2022.1006612] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 09/23/2022] [Indexed: 11/17/2022] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental condition characterized by communication and social interaction deficits, and by restricted interests and stereotyped, repetitive behavior patterns. ASD has a strong genetic component and a complex architecture characterized by the interplay of rare and common genetic variants. Recently, increasing evidence suggest a significant contribution of immune system dysregulation in ASD. The present paper reviews the latest updates regarding the altered immune landscape of this complex disorder highlighting areas with potential for biomarkers discovery as well as personalization of therapeutic approaches. Cross-talk between the central nervous system and immune system has long been envisaged and recent evidence brings insights into the pathways connecting the brain to the immune system. Disturbance of cytokine levels plays an important role in the establishment of a neuroinflammatory milieu in ASD. Several other immune molecules involved in antigen presentation and inflammatory cellular phenotypes are also at play in ASD. Maternal immune activation, the presence of brain-reactive antibodies and autoimmunity are other potential prenatal and postnatal contributors to ASD pathophysiology. The molecular players involved in oxidative-stress response and mitochondrial system function, are discussed as contributors to the pro-inflammatory pattern. The gastrointestinal inflammation pathways proposed to play a role in ASD are also discussed. Moreover, the body of evidence regarding some of the genetic factors linked to the immune system dysregulation is reviewed and discussed. Last, but not least, the epigenetic traits and their interactions with the immune system are reviewed as an expanding field in ASD research. Understanding the immune-mediated pathways that influence brain development and function, metabolism, and intestinal homeostasis, may lead to the identification of robust diagnostic or predictive biomarkers for ASD individuals. Thus, novel therapeutic approaches could be developed, ultimately aiming to improve their quality of life.
Collapse
Affiliation(s)
- Alina Erbescu
- Victor Babes National Institute of Pathology, Bucharest, Romania.,Faculty of Biology, Doctoral School, University of Bucharest, Bucharest, Romania
| | | | - Magdalena Budisteanu
- Victor Babes National Institute of Pathology, Bucharest, Romania.,Prof. Dr. Alex. Obregia Clinical Hospital of Psychiatry, Bucharest, Romania.,Faculty of Medicine, Titu Maiorescu University, Bucharest, Romania
| | - Aurora Arghir
- Victor Babes National Institute of Pathology, Bucharest, Romania
| | - Monica Neagu
- Victor Babes National Institute of Pathology, Bucharest, Romania.,Faculty of Biology, Doctoral School, University of Bucharest, Bucharest, Romania.,Colentina Clinical Hospital, Bucharest, Romania
| |
Collapse
|
55
|
Ayash TA, Vancolen SY, Segura M, Allard MJ, Sebire G. Protective Effects of Interleukin-1 Blockade on Group B Streptococcus-Induced Chorioamnionitis and Subsequent Neurobehavioral Impairments of the Offspring. Front Endocrinol (Lausanne) 2022; 13:833121. [PMID: 35846278 PMCID: PMC9283950 DOI: 10.3389/fendo.2022.833121] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 04/26/2022] [Indexed: 11/24/2022] Open
Abstract
Group B Streptococcus (GBS) is one of the most common bacteria isolated in human chorioamnionitis. Placental infection due to GBS is a major risk factor for fetal organ injuries, preterm birth, perinatal morbidity and mortality, and life-long multiorgan morbidities. Preclinical and clinical studies have shown that GBS-induced infection drives polymorphonuclear (PMN) cell infiltration within the placenta, the hallmark of human chorioamnionitis. In preclinical and clinical studies, the upregulation of interleukin(IL)-1β in the placenta and maternal/fetal blood was associated with a high risk of neurodevelopmental impairments in the progeny. We hypothesized that targeted IL-1 blockade administered to the dam alleviates GBS-induced chorioamnionitis and the downstream fetal inflammatory response syndrome (FIRS). IL-1 receptor antagonist (IL-1Ra) improved the gestational weight gain of GBS-infected dams and did not worsen the infectious manifestations. IL-1Ra reduced the IL-1β titer in the maternal sera of GBS-infected dams. IL-1Ra decreased the levels of IL-1β, IL-6, chemokine (C-X-C motif) ligand 1 (CXCL1), and polymorphonuclear (PMN) infiltration in GBS-infected placenta. IL-1Ra treatment reduced the IL-1β titer in the fetal sera of GBS-exposed fetuses. IL-1 blockade also alleviated GBS-induced FIRS and subsequent neurobehavioral impairments of the offspring without worsening the outcome of GBS infection. Altogether, these results showed that IL-1 plays a key role in the physiopathology of live GBS-induced chorioamnionitis and consequent neurobehavioral impairments.
Collapse
Affiliation(s)
| | | | - Mariela Segura
- Faculty of Veterinary Medicine, Université de Montreal, St-Hyacinthe, QC, Canada
| | | | - Guillaume Sebire
- Department of Pediatrics, McGill University, Montreal, QC, Canada
- *Correspondence: Guillaume Sebire,
| |
Collapse
|
56
|
Reicher V, Bunford N, Kis A, Carreiro C, Csibra B, Kratz L, Gácsi M. Developmental features of sleep electrophysiology in family dogs. Sci Rep 2021; 11:22760. [PMID: 34815446 PMCID: PMC8611005 DOI: 10.1038/s41598-021-02117-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 11/10/2021] [Indexed: 12/20/2022] Open
Abstract
Age-related differences in dog sleep and the age at which dogs reach adulthood as indexed by sleep electrophysiology are unknown. We assessed, in (1) a Juvenile sample (n = 60) of 2-14-month-old dogs (weight range: 4-68 kg), associations between age, sleep macrostructure, and non-rapid eye movement (NREM) EEG power spectrum, whether weight moderates associations, and (2) an extended sample (n = 91) of 2-30-months-old dogs, when sleep parameters stabilise. In Juvenile dogs, age was positively associated with time in drowsiness between 2 and 8 months, and negatively with time in rapid eye movement (REM) sleep between 2 and 6 months. Age was negatively associated with delta and positively with theta and alpha power activity, between 8 and 14 months. Older dogs exhibited greater sigma and beta power activity. Larger, > 8-month-old dogs had less delta and more alpha and beta activity. In extended sample, descriptive data suggest age-related power spectrum differences do not stabilise by 14 months. Drowsiness, REM, and delta power findings are consistent with prior results. Sleep electrophysiology is a promising index of dog neurodevelopment; some parameters stabilise in adolescence and some later than one year. Determination of the effect of weight and timing of power spectrum stabilisation needs further inquiry. The dog central nervous system is not fully mature by 12 months of age.
Collapse
Affiliation(s)
- Vivien Reicher
- Department of Ethology, Institute of Biology, Eötvös Loránd University, Pázmány Péter sétány 1/C, Budapest, 1117, Hungary.
- MTA-ELTE Comparative Ethology Research Group, Budapest, Hungary.
| | - Nóra Bunford
- Department of Ethology, Institute of Biology, Eötvös Loránd University, Pázmány Péter sétány 1/C, Budapest, 1117, Hungary
- Developmental and Translational Neuroscience Research Group, Institute of Cognitive Neuroscience and Psychology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Anna Kis
- Department of Ethology, Institute of Biology, Eötvös Loránd University, Pázmány Péter sétány 1/C, Budapest, 1117, Hungary
- Institute of Cognitive Neuroscience and Psychology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Cecília Carreiro
- Department of Ethology, Institute of Biology, Eötvös Loránd University, Pázmány Péter sétány 1/C, Budapest, 1117, Hungary
| | - Barbara Csibra
- Department of Ethology, Institute of Biology, Eötvös Loránd University, Pázmány Péter sétány 1/C, Budapest, 1117, Hungary
| | - Lorraine Kratz
- Department of Ethology, Institute of Biology, Eötvös Loránd University, Pázmány Péter sétány 1/C, Budapest, 1117, Hungary
| | - Márta Gácsi
- Department of Ethology, Institute of Biology, Eötvös Loránd University, Pázmány Péter sétány 1/C, Budapest, 1117, Hungary
- MTA-ELTE Comparative Ethology Research Group, Budapest, Hungary
| |
Collapse
|
57
|
Pietropaolo S, Marsicano G. The role of the endocannabinoid system as a therapeutic target for autism spectrum disorder: Lessons from behavioral studies on mouse models. Neurosci Biobehav Rev 2021; 132:664-678. [PMID: 34813825 DOI: 10.1016/j.neubiorev.2021.11.031] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 11/02/2021] [Accepted: 11/19/2021] [Indexed: 12/17/2022]
Abstract
Recent years have seen an impressive amount of research devoted to understanding the etiopathology of Autism Spectrum Disorder (ASD) and developing therapies for this syndrome. Because of the lack of biomarkers of ASD, this work has been largely based on the behavioral characterization of rodent models, based on a multitude of genetic and environmental manipulations. Here we highlight how the endocannabinoid system (ECS) has recently emerged within this context of mouse behavioral studies as an etiopathological factor in ASD and a valid potential therapeutic target. We summarize the most recent results showing alterations of the ECS in rodent models of ASD, and demonstrating ASD-like behaviors in mice with altered ECS, induced either by genetic or pharmacological manipulations. We also give a critical overview of the most relevant advances in designing treatments and novel mouse models for ASD targeting the ECS, highlighting the relevance of thorough and innovative behavioral approaches to investigate the mechanisms acting underneath the complex features of ASD.
Collapse
Affiliation(s)
| | - Giovanni Marsicano
- INSERM, U1215 NeuroCentre Magendie, 146 rue Léo Saignat, 33077, Bordeaux Cedex, France
| |
Collapse
|
58
|
Parrott JM, Oster T, Lee HY. Altered inflammatory response in FMRP-deficient microglia. iScience 2021; 24:103293. [PMID: 34820601 PMCID: PMC8602000 DOI: 10.1016/j.isci.2021.103293] [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] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 02/08/2021] [Accepted: 10/14/2021] [Indexed: 02/04/2023] Open
Abstract
Fragile X syndrome (FXS) is an inherited intellectual disability with a high risk for comorbid autism spectrum disorders. Since FXS is a genetic disease, patients are more susceptible to environmental factors aggravating symptomatology. However, this confounding interaction between FXS environmental and genetic risk factors is under-investigated. Here, Fmr1 knock-out (KO) mice and the immune stimulus lipopolysaccharide (LPS) were used to explore this interaction between FXS development and inflammation in microglia, the brain’s primary immune cell. Our results demonstrate that Fmr1 KO and wild-type (WT) microglia are not different in inflammatory outcomes without LPS. However, Fmr1 KO microglia produces an elevated pro-inflammatory and phagocytic response following LPS treatment when compared to WT microglia. Our experiments also revealed baseline differences in mitochondrial function and morphology between WT and Fmr1 KO microglia, which LPS treatment exaggerated. Our data suggest an altered inflammatory mechanism in Fmr1 KO microglia implicating a gene and environment interaction. Fmr1 KO microglia display elevated LPS-induced pro-inflammatory gene expressions Fmr1 KO microglia display elevated LPS-induced pro-inflammatory cytokine releases Fmr1 KO microglia demonstrate increased LPS-induced phagocytic responses Fmr1 KO microglial mitochondria have altered properties and LPS-stimulated responses
Collapse
Affiliation(s)
- Jennifer M Parrott
- The Department of Cellular and Integrative Physiology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Thomas Oster
- The Department of Cellular and Integrative Physiology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Hye Young Lee
- The Department of Cellular and Integrative Physiology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| |
Collapse
|
59
|
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: 5.0] [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
|
60
|
Faust TE, Gunner G, Schafer DP. Mechanisms governing activity-dependent synaptic pruning in the developing mammalian CNS. Nat Rev Neurosci 2021; 22:657-673. [PMID: 34545240 PMCID: PMC8541743 DOI: 10.1038/s41583-021-00507-y] [Citation(s) in RCA: 160] [Impact Index Per Article: 53.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/27/2021] [Indexed: 02/08/2023]
Abstract
Almost 60 years have passed since the initial discovery by Hubel and Wiesel that changes in neuronal activity can elicit developmental rewiring of the central nervous system (CNS). Over this period, we have gained a more comprehensive picture of how both spontaneous neural activity and sensory experience-induced changes in neuronal activity guide CNS circuit development. Here we review activity-dependent synaptic pruning in the mammalian CNS, which we define as the removal of a subset of synapses, while others are maintained, in response to changes in neural activity in the developing nervous system. We discuss the mounting evidence that immune and cell-death molecules are important mechanistic links by which changes in neural activity guide the pruning of specific synapses, emphasizing the role of glial cells in this process. Finally, we discuss how these developmental pruning programmes may go awry in neurodevelopmental disorders of the human CNS, focusing on autism spectrum disorder and schizophrenia. Together, our aim is to give an overview of how the field of activity-dependent pruning research has evolved, led to exciting new questions and guided the identification of new, therapeutically relevant mechanisms that result in aberrant circuit development in neurodevelopmental disorders.
Collapse
Affiliation(s)
- Travis E Faust
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Medical School, Worcester, MA, USA
| | - Georgia Gunner
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Medical School, Worcester, MA, USA
| | - Dorothy P Schafer
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Medical School, Worcester, MA, USA.
| |
Collapse
|
61
|
Guma E, Snook E, Spring S, Lerch JP, Nieman BJ, Devenyi GA, Chakravarty MM. Subtle alterations in neonatal neurodevelopment following early or late exposure to prenatal maternal immune activation in mice. Neuroimage Clin 2021; 32:102868. [PMID: 34749289 PMCID: PMC8573196 DOI: 10.1016/j.nicl.2021.102868] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 10/25/2021] [Accepted: 10/26/2021] [Indexed: 11/21/2022]
Abstract
Prenatal exposure to maternal immune activation (MIA) is a risk factor for a variety of neurodevelopmental and psychiatric disorders. The timing of MIA-exposure has been shown to affect adolescent and adult offspring neurodevelopment, however, less is known about these effects in the neonatal period. To better understand the impact of MIA-exposure on neonatal brain development in a mouse model, we assess neonate communicative abilities with the ultrasonic vocalization task, followed by high-resolution ex vivo magnetic resonance imaging (MRI) on the neonatal (postnatal day 8) mouse brain. Early exposed offspring displayed decreased communicative ability, while brain anatomy appeared largely unaffected, apart from some subtle alterations. By integrating MRI and behavioural assays to investigate the effects of MIA-exposure on neonatal neurodevelopment we show that offspring neuroanatomy and behaviour are only subtly affected by both early and late exposure. This suggests that the deficits often observed in later stages of life may be dormant, not yet developed in the neonatal period, or not as easily detectable using a cross-sectional approach.
Collapse
Affiliation(s)
- Elisa Guma
- Computational Brain Anatomy Laboratory, Cerebral Imaging Center, Douglas Mental Health University Institute, Montreal, Quebec, Canada; Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada.
| | - Emily Snook
- Computational Brain Anatomy Laboratory, Cerebral Imaging Center, Douglas Mental Health University Institute, Montreal, Quebec, Canada; Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Shoshana Spring
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Jason P Lerch
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada; Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford, United Kingdom
| | - Brian J Nieman
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada; Translational Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada; Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Gabriel A Devenyi
- Computational Brain Anatomy Laboratory, Cerebral Imaging Center, Douglas Mental Health University Institute, Montreal, Quebec, Canada; Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| | - M Mallar Chakravarty
- Computational Brain Anatomy Laboratory, Cerebral Imaging Center, Douglas Mental Health University Institute, Montreal, Quebec, Canada; Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada; Department of Psychiatry, McGill University, Montreal, Quebec, Canada; Department of Biological and Biomedical Engineering, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
62
|
Fernández de Cossío L, Lacabanne C, Bordeleau M, Castino G, Kyriakakis P, Tremblay MÈ. Lipopolysaccharide-induced maternal immune activation modulates microglial CX3CR1 protein expression and morphological phenotype in the hippocampus and dentate gyrus, resulting in cognitive inflexibility during late adolescence. Brain Behav Immun 2021; 97:440-454. [PMID: 34343619 DOI: 10.1016/j.bbi.2021.07.025] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 07/25/2021] [Accepted: 07/28/2021] [Indexed: 12/26/2022] Open
Abstract
Inflammation during pregnancy can disturb brain development and lead to disorders in the progeny, including autism spectrum disorder and schizophrenia. However, the mechanism by which a prenatal, short-lived increase of cytokines results in adverse neurodevelopmental outcomes remains largely unknown. Microglia-the brain's resident immune-cells-stand as fundamental cellular mediators, being highly sensitive and responsive to immune signals, which also play key roles during normal development. The fractalkine signaling axis is a neuron-microglia communication mechanism used to regulate neurogenesis and network formation. Previously, we showed hippocampal reduction of fractalkine receptor (Cx3cr1) mRNA at postnatal day (P) 15 in male offspring exposed to maternal immune activation induced with lipopolysaccharide (LPS) during late gestation, which was concomitant to an increased dendritic spine density in the dentate gyrus, a neurogenic niche. The current study sought to evaluate the origin and impact of this reduced hippocampal Cx3cr1 mRNA expression on microglia and cognition. We found that microglial total cell number and density are not affected in the dorsal hippocampus and dentate gyrus, respectively, but that the microglial CX3CR1 protein is decreased in the hippocampus of LPS-male offspring at P15. Further characterization of microglial morphology in the dentate gyrus identified a more ameboid phenotype in LPS-exposed offspring, predominantly in males, at P15. We thus explored maternal plasma and fetal brain cytokines to understand the mechanism behind microglial priming, showing a robust immune activation in the mother at 2 and 4 hrs after LPS administration, while only IL-10 tended towards upregulation at 2 hrs after LPS in fetal brains. To evaluate the functional long-term consequences, we assessed learning and cognitive flexibility behavior during late adolescence, finding that LPS affects only the latter with a male predominance on perseveration. A CX3CR1 gene variant in humans that results in disrupted fractalkine signaling has been recently associated with an increased risk for neurodevelopmental disorders. We show that an acute immune insult during late gestation can alter fractalkine signaling by reducing the microglial CX3CR1 protein expression, highlighting neuron-microglial fractalkine signaling as a relevant target underlying the outcomes of environmental risk factors on neurodevelopmental disorders.
Collapse
Affiliation(s)
- Lourdes Fernández de Cossío
- Department of Neurosciences, University of California, La Jolla, CA, USA; Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada.
| | - Chloé Lacabanne
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Maude Bordeleau
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada; Axe Neurosciences, Centre de Recherche du CHU de Québec - Université Laval, Québec, QC, Canada
| | - Garance Castino
- Department of Biology, École Normale Supérieure de Lyon, Université de Lyon, Lyon, France
| | | | - Marie-Ève Tremblay
- Axe Neurosciences, Centre de Recherche du CHU de Québec - Université Laval, Québec, QC, Canada; Département de médecine moléculaire, Université Laval, Québec, QC, Canada; Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada; Division of Medical Sciences, University of Victoria, Victoria, BC, Canada; Biochemistry and Molecular Biology, Faculty of Medicine, The University of British Colombia, Vancouver, BC, Canada.
| |
Collapse
|
63
|
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: 3.3] [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
|
64
|
Prieto-Villalobos J, Alvear TF, Liberona A, Lucero CM, Martínez-Araya CJ, Balmazabal J, Inostroza CA, Ramírez G, Gómez GI, Orellana JA. Astroglial Hemichannels and Pannexons: The Hidden Link between Maternal Inflammation and Neurological Disorders. Int J Mol Sci 2021; 22:ijms22179503. [PMID: 34502412 PMCID: PMC8430734 DOI: 10.3390/ijms22179503] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 08/30/2021] [Accepted: 08/30/2021] [Indexed: 12/11/2022] Open
Abstract
Maternal inflammation during pregnancy causes later-in-life alterations of the offspring’s brain structure and function. These abnormalities increase the risk of developing several psychiatric and neurological disorders, including schizophrenia, intellectual disability, bipolar disorder, autism spectrum disorder, microcephaly, and cerebral palsy. Here, we discuss how astrocytes might contribute to postnatal brain dysfunction following maternal inflammation, focusing on the signaling mediated by two families of plasma membrane channels: hemi-channels and pannexons. [Ca2+]i imbalance linked to the opening of astrocytic hemichannels and pannexons could disturb essential functions that sustain astrocytic survival and astrocyte-to-neuron support, including energy and redox homeostasis, uptake of K+ and glutamate, and the delivery of neurotrophic factors and energy-rich metabolites. Both phenomena could make neurons more susceptible to the harmful effect of prenatal inflammation and the experience of a second immune challenge during adulthood. On the other hand, maternal inflammation could cause excitotoxicity by producing the release of high amounts of gliotransmitters via astrocytic hemichannels/pannexons, eliciting further neuronal damage. Understanding how hemichannels and pannexons participate in maternal inflammation-induced brain abnormalities could be critical for developing pharmacological therapies against neurological disorders observed in the offspring.
Collapse
Affiliation(s)
- Juan Prieto-Villalobos
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; (J.P.-V.); (T.F.A.); (A.L.); (C.J.M.-A.); (J.B.); (C.A.I.); (G.R.)
| | - Tanhia F. Alvear
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; (J.P.-V.); (T.F.A.); (A.L.); (C.J.M.-A.); (J.B.); (C.A.I.); (G.R.)
| | - Andrés Liberona
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; (J.P.-V.); (T.F.A.); (A.L.); (C.J.M.-A.); (J.B.); (C.A.I.); (G.R.)
| | - Claudia M. Lucero
- Institute of Biomedical Sciences, Faculty of Health Sciences, Universidad Autónoma de Chile, Santiago 8910060, Chile; (C.M.L.); (G.I.G.)
| | - Claudio J. Martínez-Araya
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; (J.P.-V.); (T.F.A.); (A.L.); (C.J.M.-A.); (J.B.); (C.A.I.); (G.R.)
| | - Javiera Balmazabal
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; (J.P.-V.); (T.F.A.); (A.L.); (C.J.M.-A.); (J.B.); (C.A.I.); (G.R.)
| | - Carla A. Inostroza
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; (J.P.-V.); (T.F.A.); (A.L.); (C.J.M.-A.); (J.B.); (C.A.I.); (G.R.)
| | - Gigliola Ramírez
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; (J.P.-V.); (T.F.A.); (A.L.); (C.J.M.-A.); (J.B.); (C.A.I.); (G.R.)
| | - Gonzalo I. Gómez
- Institute of Biomedical Sciences, Faculty of Health Sciences, Universidad Autónoma de Chile, Santiago 8910060, Chile; (C.M.L.); (G.I.G.)
| | - Juan A. Orellana
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; (J.P.-V.); (T.F.A.); (A.L.); (C.J.M.-A.); (J.B.); (C.A.I.); (G.R.)
- Correspondence: ; Tel.: +56-23548105
| |
Collapse
|
65
|
Guma E, Bordignon PDC, Devenyi GA, Gallino D, Anastassiadis C, Cvetkovska V, Barry AD, Snook E, Germann J, Greenwood CMT, Misic B, Bagot RC, Chakravarty MM. Early or Late Gestational Exposure to Maternal Immune Activation Alters Neurodevelopmental Trajectories in Mice: An Integrated Neuroimaging, Behavioral, and Transcriptional Study. Biol Psychiatry 2021; 90:328-341. [PMID: 34053674 DOI: 10.1016/j.biopsych.2021.03.017] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/23/2021] [Accepted: 03/15/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND Exposure to maternal immune activation (MIA) in utero is a risk factor for neurodevelopmental disorders later in life. The impact of the gestational timing of MIA exposure on downstream development remains unclear. METHODS We characterized neurodevelopmental trajectories of mice exposed to the viral mimetic poly I:C (polyinosinic:polycytidylic acid) either on gestational day 9 (early) or on day 17 (late) using longitudinal structural magnetic resonance imaging from weaning to adulthood. Using multivariate methods, we related neuroimaging and behavioral variables for the time of greatest alteration (adolescence/early adulthood) and identified regions for further investigation using RNA sequencing. RESULTS Early MIA exposure was associated with accelerated brain volume increases in adolescence/early adulthood that normalized in later adulthood in the striatum, hippocampus, and cingulate cortex. Similarly, alterations in anxiety-like, stereotypic, and sensorimotor gating behaviors observed in adolescence normalized in adulthood. MIA exposure in late gestation had less impact on anatomical and behavioral profiles. Multivariate maps associated anxiety-like, social, and sensorimotor gating deficits with volume of the dorsal and ventral hippocampus and anterior cingulate cortex, among others. The most transcriptional changes were observed in the dorsal hippocampus, with genes enriched for fibroblast growth factor regulation, autistic behaviors, inflammatory pathways, and microRNA regulation. CONCLUSIONS Leveraging an integrated hypothesis- and data-driven approach linking brain-behavior alterations to the transcriptome, we found that MIA timing differentially affects offspring development. Exposure in late gestation leads to subthreshold deficits, whereas exposure in early gestation perturbs brain development mechanisms implicated in neurodevelopmental disorders.
Collapse
Affiliation(s)
- Elisa Guma
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada; Computational Brain Imaging Lab, Cerebral Imaging Center, Douglas Mental Health University Institute, Montreal, Quebec, Canada.
| | - Pedro do Couto Bordignon
- Department of Psychology, McGill University, Montreal, Quebec, Canada; Ludmer Center for Neuroinformatics and Mental Health, Montreal, Quebec, Canada
| | - Gabriel A Devenyi
- Department of Psychiatry, McGill University, Montreal, Quebec, Canada; Computational Brain Imaging Lab, Cerebral Imaging Center, Douglas Mental Health University Institute, Montreal, Quebec, Canada
| | - Daniel Gallino
- Computational Brain Imaging Lab, Cerebral Imaging Center, Douglas Mental Health University Institute, Montreal, Quebec, Canada
| | - Chloe Anastassiadis
- Computational Brain Imaging Lab, Cerebral Imaging Center, Douglas Mental Health University Institute, Montreal, Quebec, Canada; Institute of Medical Science & Collaborative Program in Neuroscience, University of Toronto, Toronto, Ontario, Canada
| | | | - Amadou D Barry
- Departments of Human Genetics and Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Quebec, Canada; Ludmer Center for Neuroinformatics and Mental Health, Montreal, Quebec, Canada
| | - Emily Snook
- Computational Brain Imaging Lab, Cerebral Imaging Center, Douglas Mental Health University Institute, Montreal, Quebec, Canada; Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Jurgen Germann
- Computational Brain Imaging Lab, Cerebral Imaging Center, Douglas Mental Health University Institute, Montreal, Quebec, Canada; University Health Network, Toronto, Ontario, Canada
| | - Celia M T Greenwood
- Gerald Bronfman Department of Oncology, McGill University, Montreal, Quebec, Canada; Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University, Montreal, Quebec, Canada; Departments of Human Genetics and Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Quebec, Canada; Ludmer Center for Neuroinformatics and Mental Health, Montreal, Quebec, Canada
| | - Bratislav Misic
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Rosemary C Bagot
- Department of Psychology, McGill University, Montreal, Quebec, Canada; Ludmer Center for Neuroinformatics and Mental Health, Montreal, Quebec, Canada
| | - M Mallar Chakravarty
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada; Department of Psychiatry, McGill University, Montreal, Quebec, Canada; Department of Biological and Biomedical Engineering, McGill University, Montreal, Quebec, Canada; Computational Brain Imaging Lab, Cerebral Imaging Center, Douglas Mental Health University Institute, Montreal, Quebec, Canada.
| |
Collapse
|
66
|
Notarangelo FM, Schwarcz R. A single prenatal lipopolysaccharide injection has acute, but not long-lasting, effects on cerebral kynurenine pathway metabolism in mice. Eur J Neurosci 2021; 54:5968-5981. [PMID: 34363411 DOI: 10.1111/ejn.15416] [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: 04/29/2021] [Revised: 07/26/2021] [Accepted: 08/03/2021] [Indexed: 11/28/2022]
Abstract
In rodents, a single injection of lipopolysaccharide (LPS) during gestation causes chemical and functional abnormalities in the offspring. These effects may involve changes in the kynurenine pathway (KP) of tryptophan degradation and may provide insights into the pathophysiology of psychiatric diseases. Using CD1 mice, we examined acute and long-term effects of prenatal LPS treatment on the levels of kynurenine and its neuroactive downstream products kynurenic acid (KYNA), 3-hydroxykynurenine (3-HK) and quinolinic acid. To this end, LPS (100 μg/kg, i.p.) was administered on gestational day 15, and KP metabolites were measured 4 and 24 h later or in adulthood. After 4 h, kynurenine, KYNA and 3-HK levels were elevated in the fetal brain, 3-HK and KYNA levels were increased in the maternal plasma, and kynurenine was increased in the maternal brain, whereas no changes were seen in the placenta. These effects were less prominent after 24 h, and prenatal LPS did not affect the basal levels of KP metabolites in the forebrain of adult animals. In addition, a second LPS injection (1 mg/kg) in adulthood in the offspring of prenatally saline- and LPS-treated mice caused a similar elevation in 3-HK levels in both groups after 24 h, but the effect was significantly more pronounced in male mice. Thus, acute immune activation during pregnancy has only short-lasting effects on KP metabolism and does not cause cerebral KP metabolites to be disproportionally affected by a second immune challenge in adulthood. However, prenatal KYNA elevations still contribute to functional abnormalities in the offspring.
Collapse
Affiliation(s)
- Francesca M Notarangelo
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Robert Schwarcz
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
67
|
Neuroinflammation in autism spectrum disorders: Exercise as a "pharmacological" tool. Neurosci Biobehav Rev 2021; 129:63-74. [PMID: 34310976 DOI: 10.1016/j.neubiorev.2021.07.023] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 03/26/2021] [Accepted: 07/21/2021] [Indexed: 02/06/2023]
Abstract
The worldwide prevalence of ASD is around 1%. Although the pathogenesis of ASD is not entirely understood, it is recognized that a combination of genetic, epigenetics, environmental factors and immune system dysfunction can play an essential role in its development. It has been suggested that autism results from the central nervous system derangements due to low-grade chronic inflammatory reactions associated with the immune system activation. ASD individuals have increased microglial activation, density, and increased proinflammatory cytokines in the several brain regions. Autism has no available pharmacological treatments, however there are pedagogical and psychotherapeutic therapies, and pharmacological treatment, that help to control behavioral symptoms. Recent data indicate that exercise intervention programs may improve cognitive and behavioral symptoms in children with ASD. Exercise can also modify inflammatory profiles that will ameliorate associated metabolic disorders. This review highlights the involvement of neuroinflammation in ASD and the beneficial effects of physical exercise on managing ASD symptoms and associated comorbidities.
Collapse
|
68
|
Solek CM, Farooqi NAI, Brake N, Kesner P, Schohl A, Antel JP, Ruthazer ES. Early Inflammation Dysregulates Neuronal Circuit Formation In Vivo via Upregulation of IL-1β. J Neurosci 2021; 41:6353-6366. [PMID: 34103360 PMCID: PMC8287996 DOI: 10.1523/jneurosci.2159-20.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 04/04/2021] [Accepted: 05/29/2021] [Indexed: 12/18/2022] Open
Abstract
Neuroimmune interaction during development is strongly implicated in the pathogenesis of neurodevelopmental disorders, but the mechanisms that cause neuronal circuit dysregulation are not well understood. We performed in vivo imaging of the developing retinotectal system in the larval zebrafish to characterize the effects of immune system activation on refinement of an archetypal sensory processing circuit. Acute inflammatory insult induced hyperdynamic remodeling of developing retinal axons in larval fish and increased axon arbor elaboration over days. Using calcium imaging in GCaMP6s transgenic fish, we showed that these morphologic changes were accompanied by a shift toward decreased visual acuity in tectal cells. This finding was supported by poorer performance in a visually guided behavioral task. We further found that the pro-inflammatory cytokine, interleukin-1β (IL-1β), is upregulated by the inflammatory insult, and that downregulation of IL-1β abrogated the effects of inflammation on axonal dynamics and growth. Moreover, baseline branching of the retinal ganglion cell arbors in IL-1β morphant animals was significantly different from that in control larvae, and their performance in a predation assay was impaired, indicating a role for this cytokine in normal neuronal development. This work establishes a simple and powerful non-mammalian model of developmental immune activation and demonstrates a role for IL-1β in mediating the pathologic effects of inflammation on neuronal circuit development.SIGNIFICANCE STATEMENT Maternal immune activation can increase the risk of neurodevelopmental disorders in offspring; however, the mechanisms involved are not fully understood. Using a non-mammalian vertebrate model of developmental immune activation, we show that even brief activation of inflammatory pathways has immediate and long-term effects on the arborization of axons, and that these morphologic changes have functional and behavioral consequences. Finally, we show that the pro-inflammatory cytokine IL-1β plays an essential role in both the effects of inflammation on circuit formation and normal axonal development. Our data add to a growing body of evidence supporting epidemiological studies linking immune activation to neurodevelopmental disorders, and help shed light on the molecular and cellular processes that contribute to the etiology of these disorders.
Collapse
Affiliation(s)
- Cynthia M Solek
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Nasr A I Farooqi
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Niklas Brake
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Philip Kesner
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Anne Schohl
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Jack P Antel
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Edward S Ruthazer
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec H3A 2B4, Canada
| |
Collapse
|
69
|
Woods RM, Lorusso JM, Potter HG, Neill JC, Glazier JD, Hager R. Maternal immune activation in rodent models: A systematic review of neurodevelopmental changes in gene expression and epigenetic modulation in the offspring brain. Neurosci Biobehav Rev 2021; 129:389-421. [PMID: 34280428 DOI: 10.1016/j.neubiorev.2021.07.015] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 05/11/2021] [Accepted: 07/11/2021] [Indexed: 01/06/2023]
Abstract
Maternal immune activation (mIA) during pregnancy is hypothesised to disrupt offspring neurodevelopment and predispose offspring to neurodevelopmental disorders such as schizophrenia. Rodent models of mIA have explored possible mechanisms underlying this paradigm and provide a vital tool for preclinical research. However, a comprehensive analysis of the molecular changes that occur in mIA-models is lacking, hindering identification of robust clinical targets. This systematic review assesses mIA-driven transcriptomic and epigenomic alterations in specific offspring brain regions. Across 118 studies, we focus on 88 candidate genes and show replicated changes in expression in critical functional areas, including elevated inflammatory markers, and reduced myelin and GABAergic signalling proteins. Further, disturbed epigenetic markers at nine of these genes support mIA-driven epigenetic modulation of transcription. Overall, our results demonstrate that current outcome measures have direct relevance for the hypothesised pathology of schizophrenia and emphasise the importance of mIA-models in contributing to the understanding of biological pathways impacted by mIA and the discovery of new drug targets.
Collapse
Affiliation(s)
- Rebecca M Woods
- Division of Evolution & Genomic Sciences, School of Biological Sciences, Manchester Academic Health Science Center, Faculty of Biology, Medicine & Health, University of Manchester, Manchester, M13 9PT, United Kingdom.
| | - Jarred M Lorusso
- Division of Evolution & Genomic Sciences, School of Biological Sciences, Manchester Academic Health Science Center, Faculty of Biology, Medicine & Health, University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Harry G Potter
- Division of Evolution & Genomic Sciences, School of Biological Sciences, Manchester Academic Health Science Center, Faculty of Biology, Medicine & Health, University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Joanna C Neill
- Division of Pharmacy & Optometry, School of Health Sciences, Manchester Academic Health Science Center, Faculty of Biology, Medicine & Health, University of Manchester, Manchester, M13 9PL, United Kingdom
| | - Jocelyn D Glazier
- Division of Evolution & Genomic Sciences, School of Biological Sciences, Manchester Academic Health Science Center, Faculty of Biology, Medicine & Health, University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Reinmar Hager
- Division of Evolution & Genomic Sciences, School of Biological Sciences, Manchester Academic Health Science Center, Faculty of Biology, Medicine & Health, University of Manchester, Manchester, M13 9PT, United Kingdom
| |
Collapse
|
70
|
Sandre PC, da Silva Chagas L, de Velasco PC, Galvani RG, Dias Fraga KY, Tavares do Carmo MDG, Vianna PHO, Bonomo AC, Serfaty CA. Chronic nutritional restriction of omega-3 fatty acids induces a pro-inflammatory profile during the development of the rat visual system. Brain Res Bull 2021; 174:366-378. [PMID: 34237395 DOI: 10.1016/j.brainresbull.2021.07.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 06/19/2021] [Accepted: 07/02/2021] [Indexed: 12/24/2022]
Abstract
Modern western diets have been associated with a reduced proportion of dietary omega-3 fatty acids leading to decreased levels of DHA (docosahexaenoic acid) in the brain. Low DHA content has been associated with altered development of visual acuity in infants and also with an altered time course of synapse elimination and plasticity in subcortical visual nuclei in rodents. Microglia has an active role in normal developmental processes such as circuitry refinement and plasticity, and its activation status can be modulated by omega-3 (ω3) and omega-6 (ω6) essential fatty acids. In the present study, we investigated the impact of dietary restriction of DHA (ω3-), through the chronic administration of a coconut-based diet as the only fat source. This dietary protocol resulted in a reduction in DHA content in the retina and superior colliculus (SC) and in a neuroinflammatory outcome during the development of the rodent visual system. The ω3- group showed changes in microglial morphology in the retina and SC and a corresponding altered pattern of pro-inflammatory cytokine expression. Early and late fish oil protocols supplementation were able to restore DHA levels. The early supplementation also decreased neuroinflammatory markers in the visual system. The present study indicates that a chronic dietary restriction of omega-3 fatty acids and the resulting deficits in DHA content, commonly observed in Western diets, interferes with the microglial profile leading to an inflamed microenvironment which may underlie a disruption of synapse elimination, altered topographical organization, abnormal plasticity, and duration of critical periods during brain development.
Collapse
Affiliation(s)
- Poliana Capucho Sandre
- Laboratory of Neural Plasticity Neurobiology Department, Biology Institute, Federal Fluminense University, Niteroi, Brazil
| | - Luana da Silva Chagas
- Laboratory of Neural Plasticity Neurobiology Department, Biology Institute, Federal Fluminense University, Niteroi, Brazil
| | - Patricia Coelho de Velasco
- Josué Castro Nutrition Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil; Department of Applied Nutrition, Institute of Nutrition, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Rômulo Gonçalves Galvani
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Karla Yasmin Dias Fraga
- Josué Castro Nutrition Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | - Adriana Cesar Bonomo
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Claudio Alberto Serfaty
- Laboratory of Neural Plasticity Neurobiology Department, Biology Institute, Federal Fluminense University, Niteroi, Brazil; National Institute of Science and Technology on Neuroimmunomodulation - INCT-NIM, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, 21040-360, Brazil.
| |
Collapse
|
71
|
Ikezu S, Yeh H, Delpech JC, Woodbury ME, Van Enoo AA, Ruan Z, Sivakumaran S, You Y, Holland C, Guillamon-Vivancos T, Yoshii-Kitahara A, Botros MB, Madore C, Chao PH, Desani A, Manimaran S, Kalavai SV, Johnson WE, Butovsky O, Medalla M, Luebke JI, Ikezu T. Inhibition of colony stimulating factor 1 receptor corrects maternal inflammation-induced microglial and synaptic dysfunction and behavioral abnormalities. Mol Psychiatry 2021; 26:1808-1831. [PMID: 32071385 PMCID: PMC7431382 DOI: 10.1038/s41380-020-0671-2] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 01/21/2020] [Accepted: 01/29/2020] [Indexed: 12/23/2022]
Abstract
Maternal immune activation (MIA) disrupts the central innate immune system during a critical neurodevelopmental period. Microglia are primary innate immune cells in the brain although their direct influence on the MIA phenotype is largely unknown. Here we show that MIA alters microglial gene expression with upregulation of cellular protrusion/neuritogenic pathways, concurrently causing repetitive behavior, social deficits, and synaptic dysfunction to layer V intrinsically bursting pyramidal neurons in the prefrontal cortex of mice. MIA increases plastic dendritic spines of the intrinsically bursting neurons and their interaction with hyper-ramified microglia. Treating MIA offspring by colony stimulating factor 1 receptor inhibitors induces depletion and repopulation of microglia, and corrects protein expression of the newly identified MIA-associated neuritogenic molecules in microglia, which coalesces with correction of MIA-associated synaptic, neurophysiological, and behavioral abnormalities. Our study demonstrates that maternal immune insults perturb microglial phenotypes and influence neuronal functions throughout adulthood, and reveals a potent effect of colony stimulating factor 1 receptor inhibitors on the correction of MIA-associated microglial, synaptic, and neurobehavioral dysfunctions.
Collapse
Affiliation(s)
- Seiko Ikezu
- Departments of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA.
| | - Hana Yeh
- Departments of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
- Graduate Program in Neuroscience, Boston University, Boston, MA, USA
| | - Jean-Christophe Delpech
- Departments of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Maya E Woodbury
- Departments of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
- Graduate Program in Neuroscience, Boston University, Boston, MA, USA
| | - Alicia A Van Enoo
- Departments of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Zhi Ruan
- Departments of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Sudhir Sivakumaran
- Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA, USA
| | - Yang You
- Departments of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Carl Holland
- Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA, USA
| | | | - Asuka Yoshii-Kitahara
- Departments of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Mina B Botros
- Departments of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Charlotte Madore
- Ann Romney Center for Neurologic Diseases, Department of Neurology and Evergrande Center for Immunologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Pin-Hao Chao
- Departments of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Ankita Desani
- Departments of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Solaiappan Manimaran
- Computational Biomedicine, Boston University School of Medicine, Boston, MA, USA
| | - Srinidhi Venkatesan Kalavai
- Departments of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - W Evan Johnson
- Computational Biomedicine, Boston University School of Medicine, Boston, MA, USA
| | - Oleg Butovsky
- Ann Romney Center for Neurologic Diseases, Department of Neurology and Evergrande Center for Immunologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Maria Medalla
- Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA, USA
| | - Jennifer I Luebke
- Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA, USA
- Center for Systems Neuroscience, Boston University, Boston, MA, USA
| | - Tsuneya Ikezu
- Departments of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA.
- Center for Systems Neuroscience, Boston University, Boston, MA, USA.
- Department of Neurology and Alzheimer's Disease Center, Boston University School of Medicine, Boston, MA, USA.
| |
Collapse
|
72
|
Ye J, Wang H, Cui L, Chu S, Chen N. The progress of chemokines and chemokine receptors in autism spectrum disorders. Brain Res Bull 2021; 174:268-280. [PMID: 34077795 DOI: 10.1016/j.brainresbull.2021.05.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 05/19/2021] [Accepted: 05/27/2021] [Indexed: 12/16/2022]
Abstract
Autism spectrum disorders (ASD) are a group of neurodevelopmental disorders and the main symptoms of ASD are impairments in social communication and abnormal behavioral patterns. Studies have shown that immune dysfunction and neuroinflammation play a key role in ASD patients and experimental models. Chemokines are groups of small proteins that regulate cell migration and mediate inflammation responses via binding to chemokine receptors. Thus, chemokines/chemokine receptors may be involved in neurodevelopmental disorders and associated with ASD. In this review, we summarize the research progress of chemokine aberrations in ASD and also review the recent progress of clinical treatment of ASD and pharmacological research related to chemokines/chemokine receptors. This review highlights the possible connection between chemokines/chemokine receptors and ASD, and provides novel potential targets for drug discovery of ASD.
Collapse
Affiliation(s)
- Junrui Ye
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica and Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Hongyun Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica and Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Liyuan Cui
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica and Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Shifeng Chu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica and Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| | - Naihong Chen
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica and Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| |
Collapse
|
73
|
Quagliato LA, de Matos U, Nardi AE. Maternal immune activation generates anxiety in offspring: A translational meta-analysis. Transl Psychiatry 2021; 11:245. [PMID: 33903587 PMCID: PMC8076195 DOI: 10.1038/s41398-021-01361-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 04/12/2021] [Indexed: 02/07/2023] Open
Abstract
Maternal immune activation (MIA) during pregnancy is recognized as an etiological risk factor for various psychiatric disorders, such as schizophrenia, major depressive disorder, and autism. Prenatal immune challenge may serve as a "disease primer" for alteration of the trajectory of fetal brain development that, in combination with other genetic and environmental factors, may ultimately result in the emergence of different psychiatric conditions. However, the association between MIA and an offspring's chance of developing anxiety disorders is less clear. To evaluate the effect of MIA on offspring anxiety, a systematic review and meta-analysis of the preclinical literature was conducted. We performed a systematic search of the PubMed, Web of Science, PsycINFO, and Cochrane Library electronic databases using the PRISMA and World Health Organization (WHO) methodologies for systematic reviews. Studies that investigated whether MIA during pregnancy could cause anxiety symptoms in rodent offspring were included. Overall, the meta-analysis showed that MIA induced anxiety behavior in offspring. The studies provide strong evidence that prenatal immune activation impacts specific molecular targets and synapse formation and function and induces an imbalance in neurotransmission that could be related to the generation of anxiety in offspring. Future research should further explore the role of MIA in anxiety endophenotypes. According to this meta-analysis, MIA plays an important role in the pathophysiological mechanisms of anxiety disorders and is a promising therapeutic target.
Collapse
Affiliation(s)
- Laiana A Quagliato
- Laboratory of Panic & Respiration, Institute of Psychiatry, Federal University of Rio de Janeiro, 22270-010, Rio de Janeiro, Brazil.
| | - Ursula de Matos
- Laboratory of Panic & Respiration, Institute of Psychiatry, Federal University of Rio de Janeiro, 22270-010, Rio de Janeiro, Brazil
| | - Antonio E Nardi
- Laboratory of Panic & Respiration, Institute of Psychiatry, Federal University of Rio de Janeiro, 22270-010, Rio de Janeiro, Brazil
| |
Collapse
|
74
|
Haq SU, Bhat UA, Kumar A. Prenatal stress effects on offspring brain and behavior: Mediators, alterations and dysregulated epigenetic mechanisms. J Biosci 2021. [DOI: 10.1007/s12038-021-00153-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
75
|
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: 3.7] [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
|
76
|
Feng K, Zhao Y, Yu Q, Deng J, Wu J, Liu L. Can probiotic supplements improve the symptoms of autism spectrum disorder in children?: A protocol for systematic review and meta analysis. Medicine (Baltimore) 2021; 100:e18621. [PMID: 33725807 PMCID: PMC7969216 DOI: 10.1097/md.0000000000018621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 12/06/2019] [Indexed: 01/05/2023] Open
Abstract
INTRODUCTION Autism spectrum disorder (ASD) is a neurodevelopmental disorder with increasing incidence. The externalizing and internalizing problems among children with ASD often persistent and highly impair functioning of both the child and the family. Children with ASD often develop gut-related comorbidities and dysbiosis can have negative effects on not only the gastrointestinal (GI) tract, but also psychological symptoms. Dietary exclusions and probiotic supplements also have been investigated in the management of ASD symptoms. Especially, there is some anecdotal evidence that probiotics supplements are able to alleviate GI symptoms as well as improve behaviors in children with ASD. METHOD AND ANALYSIS This review will report on overall studies that include randomized control trials, randomized cross-over studies and cluster-randomized trials designs that consider curative effect in children with ASD by probiotic supplements. We will search 6 databases: MEDLINE, Embase, Scopus, PubMed, The Cochrane Library, and Web of Science and we will perform a manual search the journal Autism and information of ongoing or unpublished studies. The Mixed Methods Appraisal Tool (MMAT) will be used to assess quality of articles and the Jadad scale will be used to assess for bias. Assessment of publication bias will be performed using funnel plots generated by Comprehensive Meta-Analysis (CMA) 3.0 software. Clarifying the evidence in this area will be important for future research directions when reformulating and promoting the therapeutic regime in the field. ETHICS AND DISSEMINATION There are no human participants, data, or tissue being directly studied for the purposes of the review; therefore, ethics approval and consent to participate are not applicable. The results of this study will be presented at conferences and published in peer-reviewed journals. REGISTRATION AND STATUS PROSPERO 2019 CRD42019132754.
Collapse
Affiliation(s)
- Kai Feng
- Beijing University of Chinese Medicine, Beijing
| | - Ying Zhao
- Chengdu University of Chinese Medicine, Chengdu
| | - Qingyang Yu
- Beijing University of Chinese Medicine, Beijing
| | - Jialin Deng
- Beijing University of Chinese Medicine, Beijing
| | - Jingjing Wu
- Beijing University of Chinese Medicine, Beijing
| | - Lingjia Liu
- Department of Pediatrics, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
77
|
Xiao L, Yan J, Feng D, Ye S, Yang T, Wei H, Li T, Sun W, Chen J. Critical Role of TLR4 on the Microglia Activation Induced by Maternal LPS Exposure Leading to ASD-Like Behavior of Offspring. Front Cell Dev Biol 2021; 9:634837. [PMID: 33748121 PMCID: PMC7969707 DOI: 10.3389/fcell.2021.634837] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Accepted: 02/15/2021] [Indexed: 12/28/2022] Open
Abstract
Objective: To investigate the role of TLR4 on the microglia activation in the pre-frontal cortex, which leads to autism-like behavior of the offspring induced by maternal lipopolysaccharide (LPS) exposure. Methods: Pregnant TLR4-/- (knockout, KO) and WT (wild type, WT) dams were intraperitoneally injected with LPS or PBS, respectively. The levels of TNFα, IL-1β, and IL-6 in the maternal serum and fetal brain were assessed with ELISA following LPS exposure. The gestation period, litter size and weight of the offspring were evaluated. Three-chamber sociability test, open field test and olfactory habituation/dishabituation test were used to assess the offspring's autism-like behavior at 7 weeks of age. Western blotting was performed to examine the levels of TLR4, Phospho-NFκB p65, IKKα, IBA-1, iNOS, Arg-1, C3, CR3A, NMDAR2A, and Syn-1 expression in the pre-frontal cortex. The morphological changes in the microglia, the distribution and expression of TLR4 were observed by immunofluorescence staining. Golgi-Cox staining was conducted to evaluate the dendritic length and spine density of the neurons in 2-week-old offspring. Results: Maternal LPS stimulation increased serum TNFα and IL-6, as well as fetal brain TNFα in the WT mice. The litter size and the weight of the WT offspring were significantly reduced following maternal LPS treatment. LPS-treated WT offspring had lower social and self-exploration behavior, and greater anxiety and repetitive behaviors. The protein expression levels of TLR4 signaling pathways, including TLR4, Phospho-NFκB p65, IKKα, and IBA-1, iNOS expression were increased in the LPS-treated WT offspring, whereas Arg-1 was decreased. Maternal LPS treatment resulted in the significant reduction in the levels of the synaptic pruning-related proteins, C3 and CR3A. Moreover, the neuronal dendritic length and spine density, as well as the expression levels of the synaptic plasticity-related proteins, NMDAR2A and Syn-1 were reduced in the WT offspring; however, gestational LPS exposure had no effect on the TLR4-/- offspring. Conclusion: Activation of TLR4 signaling pathway following maternal LPS exposure induced the abnormal activation of microglia, which in turn was involved in excessive synaptic pruning to decrease synaptic plasticity in the offspring. This may be one of the reasons for the autism-like behavior in the offspring mice.
Collapse
Affiliation(s)
- Lu Xiao
- Chongqing Key Laboratory of Childhood Nutrition and Health, Children's Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing, China
| | - Junyan Yan
- Chongqing Key Laboratory of Childhood Nutrition and Health, Children's Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing, China
| | - Di Feng
- Chongqing Key Laboratory of Childhood Nutrition and Health, Children's Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing, China
| | - Shasha Ye
- Chongqing Key Laboratory of Childhood Nutrition and Health, Children's Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing, China
| | - Ting Yang
- Chongqing Key Laboratory of Childhood Nutrition and Health, Children's Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing, China
| | - Hua Wei
- Chongqing Key Laboratory of Childhood Nutrition and Health, Children's Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing, China
| | - Tingyu Li
- Chongqing Key Laboratory of Childhood Nutrition and Health, Children's Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing, China
| | - Wuqing Sun
- Information Technological Service Center, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Jie Chen
- Chongqing Key Laboratory of Childhood Nutrition and Health, Children's Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing, China
| |
Collapse
|
78
|
Moradi K, Ashraf-Ganjouei A, Tavolinejad H, Bagheri S, Akhondzadeh S. The interplay between gut microbiota and autism spectrum disorders: A focus on immunological pathways. Prog Neuropsychopharmacol Biol Psychiatry 2021; 106:110091. [PMID: 32891667 DOI: 10.1016/j.pnpbp.2020.110091] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 08/21/2020] [Accepted: 08/30/2020] [Indexed: 12/23/2022]
Abstract
Autism spectrum disorders (ASD) are a group of neurodevelopmental disorders characterized by impairments in social and cognitive activities, stereotypical and repetitive behaviors and restricted areas of interest. A remarkable proportion of ASD patients represent immune dysregulation as well as gastrointestinal complications. Hence, a novel concept has recently emerged, addressing the possible intercommunication between the brain, the immune system, the gut and its commensals. Here, we provide an overview of how gut microbes and their metabolites are associated with neurobehavioral features of ASD through various immunologic mechanisms. Moreover, we discuss the potential therapeutic options that could modify these features.
Collapse
Affiliation(s)
- Kamyar Moradi
- Psychiatric Research Center, Roozbeh Psychiatric Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Ashraf-Ganjouei
- Psychiatric Research Center, Roozbeh Psychiatric Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamed Tavolinejad
- Psychiatric Research Center, Roozbeh Psychiatric Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Sayna Bagheri
- Psychiatric Research Center, Roozbeh Psychiatric Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Shahin Akhondzadeh
- Psychiatric Research Center, Roozbeh Psychiatric Hospital, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
79
|
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: 2.3] [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
|
80
|
Pekala M, Doliwa M, Kalita K. Impact of maternal immune activation on dendritic spine development. Dev Neurobiol 2021; 81:524-545. [PMID: 33382515 DOI: 10.1002/dneu.22804] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 11/26/2020] [Accepted: 12/28/2020] [Indexed: 01/08/2023]
Abstract
Dendritic spines are small dendritic protrusions that harbor most excitatory synapses in the brain. The proper generation and maturation of dendritic spines are crucial for the regulation of synaptic transmission and formation of neuronal circuits. Abnormalities in dendritic spine density and morphology are common pathologies in autism and schizophrenia. According to epidemiological studies, one risk factor for these neurodevelopmental disorders is maternal infection during pregnancy. This review discusses spine alterations in animal models of maternal immune activation in the context of neurodevelopmental disorders. We describe potential mechanisms that might be responsible for prenatal infection-induced changes in the dendritic spine phenotype and behavior in offspring.
Collapse
Affiliation(s)
- Martyna Pekala
- Laboratory of Neurobiology, BRAINCITY, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Marta Doliwa
- Laboratory of Neurobiology, BRAINCITY, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Katarzyna Kalita
- Laboratory of Neurobiology, BRAINCITY, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
81
|
Bordeleau M, Fernández de Cossío L, Chakravarty MM, Tremblay MÈ. From Maternal Diet to Neurodevelopmental Disorders: A Story of Neuroinflammation. Front Cell Neurosci 2021; 14:612705. [PMID: 33536875 PMCID: PMC7849357 DOI: 10.3389/fncel.2020.612705] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 12/07/2020] [Indexed: 12/13/2022] Open
Abstract
Providing the appropriate quantity and quality of food needed for both the mother's well-being and the healthy development of the offspring is crucial during pregnancy. However, the macro- and micronutrient intake also impacts the body's regulatory supersystems of the mother, such as the immune, endocrine, and nervous systems, which ultimately influence the overall development of the offspring. Of particular importance is the association between unhealthy maternal diet and neurodevelopmental disorders in the offspring. Epidemiological studies have linked neurodevelopmental disorders like autism spectrum disorders, attention-deficit-hyperactivity disorder, and schizophrenia, to maternal immune activation (MIA) during gestation. While the deleterious consequences of diet-induced MIA on offspring neurodevelopment are increasingly revealed, neuroinflammation is emerging as a key underlying mechanism. In this review, we compile the evidence available on how the mother and offspring are both impacted by maternal dietary imbalance. We specifically explore the various inflammatory and anti-inflammatory effects of dietary components and discuss how changes in inflammatory status can prime the offspring brain development toward neurodevelopmental disorders. Lastly, we discuss research evidence on the mechanisms that sustain the relationship between maternal dietary imbalance and offspring brain development, involving altered neuroinflammatory status in the offspring, as well as genetic to cellular programming notably of microglia, and the evidence that the gut microbiome may act as a key mediator.
Collapse
Affiliation(s)
- Maude Bordeleau
- Integrated Program in Neuroscience, McGill University, Montréal, QC, Canada
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada
| | | | - M. Mallar Chakravarty
- Integrated Program in Neuroscience, McGill University, Montréal, QC, Canada
- Cerebral Imaging Centre, Douglas Mental Health University, McGill University, Montréal, QC, Canada
- Department of Psychiatry, McGill University, Montréal, QC, Canada
- Department of Biological and Biomedical Engineering, McGill University, Montréal, QC, Canada
| | - Marie-Ève Tremblay
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada
- Département de Médecine Moléculaire, Université Laval, Québec, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Biochemistry and Molecular Biology, Faculty of Medicine, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
82
|
Kalavai SV, Ikezu S. Neuritogenic function of microglia in maternal immune activation and autism spectrum disorders. Neural Regen Res 2021; 16:1436-1437. [PMID: 33318443 PMCID: PMC8284274 DOI: 10.4103/1673-5374.301012] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Affiliation(s)
- Srinidhi Venkatesan Kalavai
- Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Seiko Ikezu
- Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
83
|
Virulence factor-related gut microbiota genes and immunoglobulin A levels as novel markers for machine learning-based classification of autism spectrum disorder. Comput Struct Biotechnol J 2020; 19:545-554. [PMID: 33510860 PMCID: PMC7809157 DOI: 10.1016/j.csbj.2020.12.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 12/10/2020] [Accepted: 12/13/2020] [Indexed: 02/07/2023] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental condition for which early identification and intervention is crucial for optimum prognosis. Our previous work showed gut Immunoglobulin A (IgA) to be significantly elevated in the gut lumen of children with ASD compared to typically developing (TD) children. Gut microbiota variations have been reported in ASD, yet not much is known about virulence factor-related gut microbiota (VFGM) genes. Upon determining the VFGM genes distinguishing ASD from TD, this study is the first to utilize VFGM genes and IgA levels for a machine learning-based classification of ASD. Sequence comparisons were performed of metagenome datasets from children with ASD (n = 43) and TD children (n = 31) against genes in the virulence factor database. VFGM gene composition was associated with ASD phenotype. VFGM gene diversity was higher in children with ASD and positively correlated with IgA content. As Group B streptococcus (GBS) genes account for the highest proportion of 24 different VFGMs between ASD and TD and positively correlate with gut IgA, GBS genes were used in combination with IgA and VFGMs diversity to distinguish ASD from TD. Given that VFGM diversity, increases in IgA, and ASD-enriched VFGM genes were independent of sex and gastrointestinal symptoms, a classification method utilizing them will not pertain only to a specific subgroup of ASD. By introducing the classification value of VFGM genes and considering that VFs can be isolated in pregnant women and newborns, these findings provide a novel machine learning-based early risk identification method for ASD.
Collapse
|
84
|
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: 2.0] [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
|
85
|
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
|
86
|
Fernandez A, Dumon C, Guimond D, Tyzio R, Bonifazi P, Lozovaya N, Burnashev N, Ferrari DC, Ben-Ari Y. The GABA Developmental Shift Is Abolished by Maternal Immune Activation Already at Birth. Cereb Cortex 2020; 29:3982-3992. [PMID: 30395185 DOI: 10.1093/cercor/bhy279] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 09/14/2018] [Accepted: 10/11/2018] [Indexed: 01/27/2023] Open
Abstract
Epidemiological and experimental studies suggest that maternal immune activation (MIA) leads to developmental brain disorders, but whether the pathogenic mechanism impacts neurons already at birth is not known. We now report that MIA abolishes in mice the oxytocin-mediated delivery γ-aminobutyric acid (GABA) shift from depolarizing to hyperpolarizing in CA3 pyramidal neurons, and this is restored by the NKCC1 chloride importer antagonist bumetanide. Furthermore, MIA hippocampal pyramidal neurons at birth have a more exuberant apical arbor organization and increased apical dendritic length than age-matched controls. The frequency of spontaneous glutamatergic postsynaptic currents is also increased in MIA offspring, as well as the pairwise correlation of the synchronized firing of active cells in CA3. These alterations produced by MIA persist, since at P14-15 GABA action remains depolarizing, produces excitatory action, and network activity remains elevated with a higher frequency of spontaneous glutamatergic postsynaptic currents. Therefore, the pathogenic actions of MIA lead to important morphophysiological and network alterations in the hippocampus already at birth.
Collapse
Affiliation(s)
- Amandine Fernandez
- Neurochlore, Ben-Ari Institute of Neuroarcheology (IBEN), Bâtiment Beret-Delaage, Parc Scientifique et Technologique de Luminy, MARSEILLE Cedex 09, France.,Mediterranean Institute of Neurobiology (INMED), INSERM UMR1249, Marseille, France.,Aix-Marseille University UMR 1249, Marseille, France
| | - Camille Dumon
- Neurochlore, Ben-Ari Institute of Neuroarcheology (IBEN), Bâtiment Beret-Delaage, Parc Scientifique et Technologique de Luminy, MARSEILLE Cedex 09, France.,Aix-Marseille University UMR 1249, Marseille, France
| | - Damien Guimond
- Neurochlore, Ben-Ari Institute of Neuroarcheology (IBEN), Bâtiment Beret-Delaage, Parc Scientifique et Technologique de Luminy, MARSEILLE Cedex 09, France
| | - Roman Tyzio
- Neurochlore, Ben-Ari Institute of Neuroarcheology (IBEN), Bâtiment Beret-Delaage, Parc Scientifique et Technologique de Luminy, MARSEILLE Cedex 09, France.,Mediterranean Institute of Neurobiology (INMED), INSERM UMR1249, Marseille, France.,Aix-Marseille University UMR 1249, Marseille, France
| | - Paolo Bonifazi
- Biocruces Health Research Institute, Barakaldo, Spain.,IKERBASQUE: The Basque Foundation for Science, Bilbao, Spain
| | - Natalia Lozovaya
- Neurochlore, Ben-Ari Institute of Neuroarcheology (IBEN), Bâtiment Beret-Delaage, Parc Scientifique et Technologique de Luminy, MARSEILLE Cedex 09, France
| | - Nail Burnashev
- Mediterranean Institute of Neurobiology (INMED), INSERM UMR1249, Marseille, France.,Aix-Marseille University UMR 1249, Marseille, France
| | - Diana C Ferrari
- Neurochlore, Ben-Ari Institute of Neuroarcheology (IBEN), Bâtiment Beret-Delaage, Parc Scientifique et Technologique de Luminy, MARSEILLE Cedex 09, France
| | - Yehezkel Ben-Ari
- Neurochlore, Ben-Ari Institute of Neuroarcheology (IBEN), Bâtiment Beret-Delaage, Parc Scientifique et Technologique de Luminy, MARSEILLE Cedex 09, France.,Mediterranean Institute of Neurobiology (INMED), INSERM UMR1249, Marseille, France
| |
Collapse
|
87
|
Laue HE, Korrick SA, Baker ER, Karagas MR, Madan JC. Prospective associations of the infant gut microbiome and microbial function with social behaviors related to autism at age 3 years. Sci Rep 2020; 10:15515. [PMID: 32968156 PMCID: PMC7511970 DOI: 10.1038/s41598-020-72386-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 08/31/2020] [Indexed: 12/22/2022] Open
Abstract
The hypothesized link between gut bacteria and autism spectrum disorder (ASD) has been explored through animal models and human studies with microbiome assessment after ASD presentation. We aimed to prospectively characterize the association between the infant/toddler gut microbiome and ASD-related social behaviors at age 3 years. As part of an ongoing birth cohort gut bacterial diversity, structure, taxa, and function at 6 weeks (n = 166), 1 year (n = 158), 2 years (n = 129), and 3 years (n = 140) were quantified with 16S rRNA gene and shotgun metagenomic sequencing (n = 101 six weeks, n = 103 one year). ASD-related social behavior was assessed at age 3 years using Social Responsiveness Scale (SRS-2) T-scores. Covariate-adjusted linear and permutation-based models were implemented. Microbiome structure at 1 year was associated with SRS-2 total T-scores (p = 0.01). Several taxa at 1, 2, and 3 years were associated with SRS-2 performance, including many in the Lachnospiraceae family. Higher relative abundance of Adlercreutzia equolifaciens and Ruminococcus torques at 1 year related to poorer SRS-2 performance. Two functional pathways, L-ornithine and vitamin B6 biosynthesis, were associated with better social skills at 3 years. Our results support potential associations between early-childhood gut microbiome and social behaviors. Future mechanistic studies are warranted to pinpoint sensitive targets for intervention.
Collapse
Affiliation(s)
- Hannah E Laue
- Department of Epidemiology, Geisel School of Medicine at Dartmouth College, Hanover, NH, USA.
| | - Susan A Korrick
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Emily R Baker
- Department of Obstetrics and Gynecology, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
| | - Margaret R Karagas
- Department of Epidemiology, Geisel School of Medicine at Dartmouth College, Hanover, NH, USA
| | - Juliette C Madan
- Department of Epidemiology, Geisel School of Medicine at Dartmouth College, Hanover, NH, USA
- Department of Pediatrics, Children's Hospital at Dartmouth, Lebanon, NH, USA
- Department of Psychiatry, Children's Hospital at Dartmouth, Lebanon, NH, USA
| |
Collapse
|
88
|
Oschwald A, Petry P, Kierdorf K, Erny D. CNS Macrophages and Infant Infections. Front Immunol 2020; 11:2123. [PMID: 33072074 PMCID: PMC7531029 DOI: 10.3389/fimmu.2020.02123] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 08/05/2020] [Indexed: 12/11/2022] Open
Abstract
The central nervous system (CNS) harbors its own immune system composed of microglia in the parenchyma and CNS-associated macrophages (CAMs) in the perivascular space, leptomeninges, dura mater, and choroid plexus. Recent advances in understanding the CNS resident immune cells gave new insights into development, maturation and function of its immune guard. Microglia and CAMs undergo essential steps of differentiation and maturation triggered by environmental factors as well as intrinsic transcriptional programs throughout embryonic and postnatal development. These shaping steps allow the macrophages to adapt to their specific physiological function as first line of defense of the CNS and its interfaces. During infancy, the CNS might be targeted by a plethora of different pathogens which can cause severe tissue damage with potentially long reaching defects. Therefore, an efficient immune response of infant CNS macrophages is required even at these early stages to clear the infections but may also lead to detrimental consequences for the developing CNS. Here, we highlight the recent knowledge of the infant CNS immune system during embryonic and postnatal infections and the consequences for the developing CNS.
Collapse
Affiliation(s)
- Alexander Oschwald
- Faculty of Medicine, Institute of Neuropathology, University of Freiburg, Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Philippe Petry
- Faculty of Medicine, Institute of Neuropathology, University of Freiburg, Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Katrin Kierdorf
- Faculty of Medicine, Institute of Neuropathology, University of Freiburg, Freiburg, Germany.,CIBBS Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany.,Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Daniel Erny
- Faculty of Medicine, Institute of Neuropathology, University of Freiburg, Freiburg, Germany
| |
Collapse
|
89
|
Brain Structural and Functional Alterations in Mice Prenatally Exposed to LPS Are Only Partially Rescued by Anti-Inflammatory Treatment. Brain Sci 2020; 10:brainsci10090620. [PMID: 32906830 PMCID: PMC7564777 DOI: 10.3390/brainsci10090620] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 08/30/2020] [Accepted: 09/03/2020] [Indexed: 12/12/2022] Open
Abstract
Aberrant immune activity during neurodevelopment could participate in the generation of neurological dysfunctions characteristic of several neurodevelopmental disorders (NDDs). Numerous epidemiological studies have shown a link between maternal infections and NDDs risk; animal models of maternal immune activation (MIA) have confirmed this association. Activation of maternal immune system during pregnancy induces behavioral and functional alterations in offspring but the biological mechanisms at the basis of these effects are still poorly understood. In this study, we investigated the effects of prenatal lipopolysaccharide (LPS) exposure in peripheral and central inflammation, cortical cytoarchitecture and behavior of offspring (LPS-mice). LPS-mice reported a significant increase in interleukin-1β (IL-1β) serum level, glial fibrillary acidic protein (GFAP)- and ionized calcium-binding adapter molecule 1 (Iba1)-positive cells in the cortex. Furthermore, cytoarchitecture analysis in specific brain areas, showed aberrant alterations in minicolumns’ organization in LPS-mice adult brain. In addition, we demonstrated that LPS-mice presented behavioral alterations throughout life. In order to better understand biological mechanisms whereby LPS induced these alterations, dams were treated with meloxicam. We demonstrated for the first time that exposure to LPS throughout pregnancy induces structural permanent alterations in offspring brain. LPS-mice also present severe behavioral impairments. Preventive treatment with meloxicam reduced inflammation in offspring but did not rescue them from structural and behavioral alterations.
Collapse
|
90
|
Hao X, Pan J, Gao X, Zhang S, Li Y. Gut microbiota on gender bias in autism spectrum disorder. Rev Neurosci 2020; 32:/j/revneuro.ahead-of-print/revneuro-2020-0042/revneuro-2020-0042.xml. [PMID: 32887209 DOI: 10.1515/revneuro-2020-0042] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Accepted: 05/29/2020] [Indexed: 12/15/2022]
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder. Its three core symptoms are social communication disorder, communication disorder, narrow interest and stereotyped repetitive behavior. The proportion of male and female autistic patients is 4:1. Many researchers have studied this phenomenon, but the mechanism is still unclear. This review mainly discusses the related mechanism from the perspective of gut microbiota and introduces the influence of gut microbiota on the difference of ASD between men and women, as well as how gut microbiota may affect the gender dimorphism of ASD through metabolite of microbiota, immunity, and genetics, which provide some useful information for those who are interested in this research and find more gender-specific treatment for autistic men and women.
Collapse
Affiliation(s)
- Xia Hao
- State Key Laboratory of Component-Based Chinese Medicine, Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medicine Formulae, Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin301617,China
- College of Traditional Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin301617,China
| | - Jiao Pan
- Department of Microbiology, Ministry of Education Key Laboratory of Molecular Microbiology and Technology, Nankai University, Tianjin300071,China
| | - Xiumei Gao
- State Key Laboratory of Component-Based Chinese Medicine, Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medicine Formulae, Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin301617,China
| | - Shiyu Zhang
- College of Traditional Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin301617,China
| | - Yue Li
- State Key Laboratory of Component-Based Chinese Medicine, Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medicine Formulae, Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin301617,China
| |
Collapse
|
91
|
Comer AL, Carrier M, Tremblay MÈ, Cruz-Martín A. The Inflamed Brain in Schizophrenia: The Convergence of Genetic and Environmental Risk Factors That Lead to Uncontrolled Neuroinflammation. Front Cell Neurosci 2020; 14:274. [PMID: 33061891 PMCID: PMC7518314 DOI: 10.3389/fncel.2020.00274] [Citation(s) in RCA: 119] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 08/03/2020] [Indexed: 12/12/2022] Open
Abstract
Schizophrenia is a disorder with a heterogeneous etiology involving complex interplay between genetic and environmental risk factors. The immune system is now known to play vital roles in nervous system function and pathology through regulating neuronal and glial development, synaptic plasticity, and behavior. In this regard, the immune system is positioned as a common link between the seemingly diverse genetic and environmental risk factors for schizophrenia. Synthesizing information about how the immune-brain axis is affected by multiple factors and how these factors might interact in schizophrenia is necessary to better understand the pathogenesis of this disease. Such knowledge will aid in the development of more translatable animal models that may lead to effective therapeutic interventions. Here, we provide an overview of the genetic risk factors for schizophrenia that modulate immune function. We also explore environmental factors for schizophrenia including exposure to pollution, gut dysbiosis, maternal immune activation and early-life stress, and how the consequences of these risk factors are linked to microglial function and dysfunction. We also propose that morphological and signaling deficits of the blood-brain barrier, as observed in some individuals with schizophrenia, can act as a gateway between peripheral and central nervous system inflammation, thus affecting microglia in their essential functions. Finally, we describe the diverse roles that microglia play in response to neuroinflammation and their impact on brain development and homeostasis, as well as schizophrenia pathophysiology.
Collapse
Affiliation(s)
- Ashley L. Comer
- Graduate Program for Neuroscience, Boston University, Boston, MA, United States
- Department of Biology, Boston University, Boston, MA, United States
- Neurophotonics Center, Boston University, Boston, MA, United States
- Center for Systems Neuroscience, Boston University, Boston, MA, United States
| | - Micaël Carrier
- Axe Neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Québec City, QC, Canada
| | - Marie-Ève Tremblay
- Axe Neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Québec City, QC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada
| | - Alberto Cruz-Martín
- Graduate Program for Neuroscience, Boston University, Boston, MA, United States
- Department of Biology, Boston University, Boston, MA, United States
- Neurophotonics Center, Boston University, Boston, MA, United States
- Center for Systems Neuroscience, Boston University, Boston, MA, United States
- Department of Pharmacology and Experimental Therapeutics, Boston University, Boston, MA, United States
| |
Collapse
|
92
|
Hanger B, Couch A, Rajendran L, Srivastava DP, Vernon AC. Emerging Developments in Human Induced Pluripotent Stem Cell-Derived Microglia: Implications for Modelling Psychiatric Disorders With a Neurodevelopmental Origin. Front Psychiatry 2020; 11:789. [PMID: 32848951 PMCID: PMC7433763 DOI: 10.3389/fpsyt.2020.00789] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 07/23/2020] [Indexed: 12/15/2022] Open
Abstract
Microglia, the resident tissue macrophages of the brain, are increasingly implicated in the pathophysiology of psychiatric disorders with a neurodevelopmental origin, including schizophrenia. To date, however, our understanding of the potential role for these cells in schizophrenia has been informed by studies of aged post-mortem samples, low resolution in vivo neuroimaging and rodent models. Whilst these have provided important insights, including signs of the heterogeneous nature of microglia, we currently lack a validated human in vitro system to characterize microglia in the context of brain health and disease during neurodevelopment. Primarily, this reflects a lack of access to human primary tissue during developmental stages. In this review, we first describe microglia, including their ontogeny and heterogeneity and consider their role in brain development. We then provide an evaluation of the potential for differentiating microglia from human induced pluripotent stem cells (hiPSCs) as a robust in vitro human model system to study these cells. We find the majority of protocols for hiPSC-derived microglia generate cells characteristically similar to foetal stage microglia when exposed to neuronal environment-like cues. This may represent a robust and relevant model for the study of cellular and molecular mechanisms in schizophrenia. Each protocol however, provides unique benefits as well as shortcomings, highlighting the need for context-dependent protocol choice and cross-lab collaboration and communication to identify the most robust and translatable microglia model.
Collapse
Affiliation(s)
- Bjørn Hanger
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King’s College London, London, United Kingdom
| | - Amalie Couch
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King’s College London, London, United Kingdom
| | - Lawrence Rajendran
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King’s College London, London, United Kingdom
- UK Dementia Research Institute, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, United Kingdom
| | - Deepak P. Srivastava
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King’s College London, London, United Kingdom
- MRC Centre for Neurodevelopmental Disorders, King’s College London, London, United Kingdom
| | - Anthony C. Vernon
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King’s College London, London, United Kingdom
- MRC Centre for Neurodevelopmental Disorders, King’s College London, London, United Kingdom
| |
Collapse
|
93
|
Maternal Immunity in Autism Spectrum Disorders: Questions of Causality, Validity, and Specificity. J Clin Med 2020; 9:jcm9082590. [PMID: 32785127 PMCID: PMC7464885 DOI: 10.3390/jcm9082590] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/05/2020] [Accepted: 08/06/2020] [Indexed: 02/07/2023] Open
Abstract
Autism spectrum disorders (ASD) are complex neurodevelopmental disorders with unknown heterogeneous aetiologies. Epidemiological studies have found an association between maternal infection and development of ASD in the offspring, and clinical findings reveal a state of immune dysregulation in the pre- and postnatal period of affected subjects. Maternal immune activation (MIA) has been proposed to mediate this association by altering fetal neurodevelopment and leading to autism. Although animal models have supported a causal link between MIA and development of ASD, their validity needs to be explored. Moreover, considering that only a small proportion of affected offspring develop autism, and that MIA has been implicated in related diseases such as schizophrenia, a key unsolved question is how disease specificity and phenotypic outcome are determined. Here, we have integrated preclinical and clinical evidence, including the use of animal models for establishing causality, to explore the role of maternal infections in ASD. A proposed priming/multi-hit model may offer insights into the clinical heterogeneity of ASD, its convergence with related disorders, and therapeutic strategies.
Collapse
|
94
|
Andoh M, Shibata K, Okamoto K, Onodera J, Morishita K, Miura Y, Ikegaya Y, Koyama R. Exercise Reverses Behavioral and Synaptic Abnormalities after Maternal Inflammation. Cell Rep 2020; 27:2817-2825.e5. [PMID: 31167129 DOI: 10.1016/j.celrep.2019.05.015] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 03/11/2019] [Accepted: 05/01/2019] [Indexed: 12/17/2022] Open
Abstract
Abnormal behaviors in individuals with neurodevelopmental disorders are generally believed to be irreversible. Here, we show that voluntary wheel running ameliorates the abnormalities in sociability, repetitiveness, and anxiety observed in a mouse model of a neurodevelopmental disorder induced by maternal immune activation (MIA). Exercise activates a portion of dentate granule cells, normalizing the density of hippocampal CA3 synapses, which is excessive in the MIA-affected offspring. The synaptic surplus in the MIA offspring is induced by deficits in synapse engulfment by microglia, which is normalized by exercise through microglial activation. Finally, chemogenetically induced activation of granule cells promotes the engulfment of CA3 synapses. Thus, our study proposes a role of voluntary exercise in the modulation of behavioral and synaptic abnormalities in neurodevelopmental disorders.
Collapse
Affiliation(s)
- Megumi Andoh
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Kazuki Shibata
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Kazuki Okamoto
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Junya Onodera
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Kohei Morishita
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Yuki Miura
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Yuji Ikegaya
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan; Center for Information and Neural Networks, 1-4 Yamadaoka, Suita City, Osaka 565-0871, Japan
| | - Ryuta Koyama
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan.
| |
Collapse
|
95
|
Zhang ZZ, Zhuang ZQ, Sun SY, Ge HH, Wu YF, Cao L, Xia L, Yang QG, Wang F, Chen GH. Effects of Prenatal Exposure to Inflammation Coupled With Stress Exposure During Adolescence on Cognition and Synaptic Protein Levels in Aged CD-1 Mice. Front Aging Neurosci 2020; 12:157. [PMID: 32774299 PMCID: PMC7381390 DOI: 10.3389/fnagi.2020.00157] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Accepted: 05/07/2020] [Indexed: 12/18/2022] Open
Abstract
Age-associated impairment of spatial learning and memory (AISLM) presents substantial challenges to our health and society. Increasing evidence has indicated that embryonic exposure to inflammation accelerates the AISLM, and this can be attributable, at least partly, to changed synaptic plasticity associated with the activities of various proteins. However, it is still uncertain whether social psychological factors affect this AISLM and/or the expression of synaptic protein-associated genes. Synaptotagmin-1 (Syt1) and activity-regulated cytoskeleton-associated protein (Arc) are two synaptic proteins closely related to cognitive functions. In this study, pregnant CD-1 mice received daily intraperitoneal injections of lipopolysaccharide (LPS) (50 μg/kg) or normal saline at days 15-17 of gestation, and half of the offspring of each group were then subjected to stress for 28 days in adolescence. The Morris water maze (MWM) test was used to separately evaluate spatial learning and memory at 3 and 15 months of age, while western blotting and RNAscope assays were used to measure the protein and mRNA levels of Arc and Syt1 in the hippocampus. The results showed that, at 15 months of age, control mice had worse cognitive ability and higher protein and mRNA levels of Arc and Syt1 than their younger counterparts. Embryonic exposure to inflammation or exposure to stress in adolescence aggravated the AISLM, as well as the age-related increase in Arc and Syt1 expression. Moreover, the hippocampal protein and mRNA levels of Arc and Syt1 were significantly correlated with the performance in the learning and memory periods of the MWM test, especially in the mice that had suffered adverse insults in early life. Our findings indicated that prenatal exposure to inflammation or stress exposure in adolescence exacerbated the AISLM and age-related upregulation of Arc and Syt1 expression, and these effects were linked to cognitive impairments in CD-1 mice exposed to adverse factors in early life.
Collapse
Affiliation(s)
- Zhe-Zhe Zhang
- Department of Neurology or Department of Critical Care, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zhan-Qiang Zhuang
- Department of Neurology (Sleep Disorders), The Affiliated Chaohu Hospital of Anhui Medical University, Hefei, China
| | - Shi-Yu Sun
- Department of Neurology (Sleep Disorders), The Affiliated Chaohu Hospital of Anhui Medical University, Hefei, China
| | - He-Hua Ge
- Department of Neurology (Sleep Disorders), The Affiliated Chaohu Hospital of Anhui Medical University, Hefei, China
| | - Yong-Fang Wu
- Department of Neurology (Sleep Disorders), The Affiliated Chaohu Hospital of Anhui Medical University, Hefei, China
| | - Lei Cao
- Department of Neurology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Lan Xia
- Department of Neurology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Qi-Gang Yang
- Department of Neurology or Department of Critical Care, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Fang Wang
- Department of Neurology or Department of Critical Care, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Gui-Hai Chen
- Department of Neurology (Sleep Disorders), The Affiliated Chaohu Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
96
|
Cieślik M, Gąssowska-Dobrowolska M, Jęśko H, Czapski GA, Wilkaniec A, Zawadzka A, Dominiak A, Polowy R, Filipkowski RK, Boguszewski PM, Gewartowska M, Frontczak-Baniewicz M, Sun GY, Beversdorf DQ, Adamczyk A. Maternal Immune Activation Induces Neuroinflammation and Cortical Synaptic Deficits in the Adolescent Rat Offspring. Int J Mol Sci 2020; 21:E4097. [PMID: 32521803 PMCID: PMC7312084 DOI: 10.3390/ijms21114097] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 06/03/2020] [Accepted: 06/05/2020] [Indexed: 01/01/2023] Open
Abstract
Maternal immune activation (MIA), induced by infection during pregnancy, is an important risk factor for neuro-developmental disorders, such as autism. Abnormal maternal cytokine signaling may affect fetal brain development and contribute to neurobiological and behavioral changes in the offspring. Here, we examined the effect of lipopolysaccharide-induced MIA on neuro-inflammatory changes, as well as synaptic morphology and key synaptic protein level in cerebral cortex of adolescent male rat offspring. Adolescent MIA offspring showed elevated blood cytokine levels, microglial activation, increased pro-inflammatory cytokines expression and increased oxidative stress in the cerebral cortex. Moreover, pathological changes in synaptic ultrastructure of MIA offspring was detected, along with presynaptic protein deficits and down-regulation of postsynaptic scaffolding proteins. Consequently, ability to unveil MIA-induced long-term alterations in synapses structure and protein level may have consequences on postnatal behavioral changes, associated with, and predisposed to, the development of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Magdalena Cieślik
- Department of Cellular Signalling, Mossakowski Medical Research Centre, Polish Academy of Sciences, Pawińskiego 5, 02-106 Warsaw, Poland; (M.G.-D.); (H.J.); (G.A.C.); (A.W.); (A.Z.)
| | - Magdalena Gąssowska-Dobrowolska
- Department of Cellular Signalling, Mossakowski Medical Research Centre, Polish Academy of Sciences, Pawińskiego 5, 02-106 Warsaw, Poland; (M.G.-D.); (H.J.); (G.A.C.); (A.W.); (A.Z.)
| | - Henryk Jęśko
- Department of Cellular Signalling, Mossakowski Medical Research Centre, Polish Academy of Sciences, Pawińskiego 5, 02-106 Warsaw, Poland; (M.G.-D.); (H.J.); (G.A.C.); (A.W.); (A.Z.)
| | - Grzegorz A. Czapski
- Department of Cellular Signalling, Mossakowski Medical Research Centre, Polish Academy of Sciences, Pawińskiego 5, 02-106 Warsaw, Poland; (M.G.-D.); (H.J.); (G.A.C.); (A.W.); (A.Z.)
| | - Anna Wilkaniec
- Department of Cellular Signalling, Mossakowski Medical Research Centre, Polish Academy of Sciences, Pawińskiego 5, 02-106 Warsaw, Poland; (M.G.-D.); (H.J.); (G.A.C.); (A.W.); (A.Z.)
| | - Aleksandra Zawadzka
- Department of Cellular Signalling, Mossakowski Medical Research Centre, Polish Academy of Sciences, Pawińskiego 5, 02-106 Warsaw, Poland; (M.G.-D.); (H.J.); (G.A.C.); (A.W.); (A.Z.)
| | - Agnieszka Dominiak
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medical University of Warsaw, Żwirki i Wigury 61, 02-097 Warsaw, Poland;
| | - Rafał Polowy
- Behavior and Metabolism Research Laboratory, Mossakowski Medical Research Centre, Polish Academy of Sciences, Pawińskiego 5, 02-106 Warsaw, Poland; (R.P.); (R.K.F.)
| | - Robert K. Filipkowski
- Behavior and Metabolism Research Laboratory, Mossakowski Medical Research Centre, Polish Academy of Sciences, Pawińskiego 5, 02-106 Warsaw, Poland; (R.P.); (R.K.F.)
| | - Paweł M. Boguszewski
- Laboratory of Animal Models, Neurobiology Center, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteur 3, 02-093 Warsaw, Poland;
| | - Magdalena Gewartowska
- Electron Microscopy Platform, Mossakowski Medical Research Centre, Polish Academy of Sciences, Pawińskiego 5, 02-106 Warsaw, Poland; (M.G.); (M.F.-B.)
| | - Małgorzata Frontczak-Baniewicz
- Electron Microscopy Platform, Mossakowski Medical Research Centre, Polish Academy of Sciences, Pawińskiego 5, 02-106 Warsaw, Poland; (M.G.); (M.F.-B.)
| | - Grace Y. Sun
- Department of Biochemistry, University of Missouri, 117 Schweitzer Hall, Columbia, MO 65201, USA;
| | - David Q. Beversdorf
- Departments of Radiology, Neurology, and Psychological Sciences, William and Nancy Thompson Endowed Chair in Radiology, DC069.10, One Hospital Drive, University of Missouri, Columbia, MO 65211, USA;
| | - Agata Adamczyk
- Department of Cellular Signalling, Mossakowski Medical Research Centre, Polish Academy of Sciences, Pawińskiego 5, 02-106 Warsaw, Poland; (M.G.-D.); (H.J.); (G.A.C.); (A.W.); (A.Z.)
| |
Collapse
|
97
|
Arambula SE, McCarthy MM. Neuroendocrine-Immune Crosstalk Shapes Sex-Specific Brain Development. Endocrinology 2020; 161:bqaa055. [PMID: 32270188 PMCID: PMC7217281 DOI: 10.1210/endocr/bqaa055] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 04/06/2020] [Indexed: 12/11/2022]
Abstract
Sex is an essential biological variable that significantly impacts multiple aspects of neural functioning in both the healthy and diseased brain. Sex differences in brain structure and function are organized early in development during the critical period of sexual differentiation. While decades of research establish gonadal hormones as the primary modulators of this process, new research has revealed a critical, and perhaps underappreciated, role of the neuroimmune system in sex-specific brain development. The immune and endocrine systems are tightly intertwined and share processes and effector molecules that influence the nervous system. Thus, a natural question is whether endocrine-immune crosstalk contributes to sexual differentiation of the brain. In this mini-review, we first provide a conceptual framework by classifying the major categories of neural sex differences and review the concept of sexual differentiation of the brain, a process occurring early in development and largely controlled by steroid hormones. Next, we describe developmental sex differences in the neuroimmune system, which may represent targets or mediators of the sexual differentiation process. We then discuss the overwhelming evidence in support of crosstalk between the neuroendocrine and immune systems and highlight recent examples that shape sex differences in the brain. Finally, we review how early life events can perturb sex-specific neurodevelopment via aberrant immune activation.
Collapse
Affiliation(s)
- Sheryl E Arambula
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD
| | - Margaret M McCarthy
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD
| |
Collapse
|
98
|
Carroll L, Braeutigam S, Dawes JM, Krsnik Z, Kostovic I, Coutinho E, Dewing JM, Horton CA, Gomez-Nicola D, Menassa DA. Autism Spectrum Disorders: Multiple Routes to, and Multiple Consequences of, Abnormal Synaptic Function and Connectivity. Neuroscientist 2020; 27:10-29. [PMID: 32441222 PMCID: PMC7804368 DOI: 10.1177/1073858420921378] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Autism spectrum disorders (ASDs) are a heterogeneous group of
neurodevelopmental disorders of genetic and environmental etiologies.
Some ASD cases are syndromic: associated with clinically defined
patterns of somatic abnormalities and a neurobehavioral phenotype
(e.g., Fragile X syndrome). Many cases, however, are idiopathic or
non-syndromic. Such disorders present themselves during the early
postnatal period when language, speech, and personality start to
develop. ASDs manifest by deficits in social communication and
interaction, restricted and repetitive patterns of behavior across
multiple contexts, sensory abnormalities across multiple modalities
and comorbidities, such as epilepsy among many others. ASDs are
disorders of connectivity, as synaptic dysfunction is common to both
syndromic and idiopathic forms. While multiple theories have been
proposed, particularly in idiopathic ASDs, none address why certain
brain areas (e.g., frontotemporal) appear more vulnerable than others
or identify factors that may affect phenotypic specificity. In this
hypothesis article, we identify possible routes leading to, and the
consequences of, altered connectivity and review the evidence of
central and peripheral synaptic dysfunction in ASDs. We postulate that
phenotypic specificity could arise from aberrant experience-dependent
plasticity mechanisms in frontal brain areas and peripheral sensory
networks and propose why the vulnerability of these areas could be
part of a model to unify preexisting pathophysiological theories.
Collapse
Affiliation(s)
- Liam Carroll
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, Oxfordshire, UK
| | - Sven Braeutigam
- Oxford Centre for Human Brain Activity, Wellcome Centre for Integrative Neuroimaging, Department of Psychiatry, University of Oxford, Oxford, Oxfordshire, UK
| | - John M Dawes
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, Oxfordshire, UK
| | - Zeljka Krsnik
- Croatian Institute for Brain Research, Centre of Research Excellence for Basic, Clinical and Translational Neuroscience, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Ivica Kostovic
- Croatian Institute for Brain Research, Centre of Research Excellence for Basic, Clinical and Translational Neuroscience, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Ester Coutinho
- Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK
| | - Jennifer M Dewing
- Faculty of Medicine, University of Southampton, Southampton, Hampshire, UK
| | - Christopher A Horton
- Sir William Dunn School of Pathology, University of Oxford, Oxford, Oxfordshire, UK
| | - Diego Gomez-Nicola
- Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, UK
| | - David A Menassa
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, Oxfordshire, UK.,Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, UK
| |
Collapse
|
99
|
Andoh M, Ikegaya Y, Koyama R. Microglia in animal models of autism spectrum disorders. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020; 173:239-273. [PMID: 32711812 DOI: 10.1016/bs.pmbts.2020.04.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Various genetic and environmental factors have been suggested to cause autism spectrum disorders (ASDs). A variety of animal models of ASDs have been developed and used to investigate the mechanisms underlying the pathogenesis of ASDs. These animal models have contributed to clarifying that abnormalities in neuronal morphology and neurotransmission are responsible for the onset of ASDs. In recent years, researchers have started to focus not only on neurons but also on glial cells, particularly microglia. This is because microglial malfunction is strongly associated with structural and functional abnormalities of neurons, as well as the inflammation that is commonly observed both in the brains of patients with ASDs and in animal models of ASDs. In this chapter, we first introduce a list of commonly available animal models of ASDs and describe the validity of each model from the viewpoint of behaviors and neuroanatomy. We next detail the malfunction of microglia that has been reported in animal models of ASDs and discuss the roles of microglia in ASD pathogenesis. We will further propose possible therapeutic strategies to tackle ASDs by controlling microglial functions.
Collapse
Affiliation(s)
- Megumi Andoh
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Yuji Ikegaya
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Ryuta Koyama
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, Japan.
| |
Collapse
|
100
|
Haddad FL, Patel SV, Schmid S. Maternal Immune Activation by Poly I:C as a preclinical Model for Neurodevelopmental Disorders: A focus on Autism and Schizophrenia. Neurosci Biobehav Rev 2020; 113:546-567. [PMID: 32320814 DOI: 10.1016/j.neubiorev.2020.04.012] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 01/28/2020] [Accepted: 04/09/2020] [Indexed: 12/18/2022]
Abstract
Maternal immune activation (MIA) in response to a viral infection during early and mid-gestation has been linked through various epidemiological studies to a higher risk for the child to develop autism or schizophrenia-related symptoms.. This has led to the establishment of the pathogen-free poly I:C-induced MIA animal model for neurodevelopmental disorders, which shows relatively high construct and face validity. Depending on the experimental variables, particularly the timing of poly I:C administration, different behavioural and molecular phenotypes have been described that relate to specific symptoms of neurodevelopmental disorders such as autism spectrum disorder and/or schizophrenia. We here review and summarize epidemiological evidence for the effects of maternal infection and immune activation, as well as major findings in different poly I:C MIA models with a focus on poly I:C exposure timing, behavioural and molecular changes in the offspring, and characteristics of the model that relate it to autism spectrum disorder and schizophrenia.
Collapse
Affiliation(s)
- Faraj L Haddad
- Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON, Canada.
| | - Salonee V Patel
- Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON, Canada.
| | - Susanne Schmid
- Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON, Canada.
| |
Collapse
|