51
|
Kern JK, Geier DA, Homme KG, Geier MR. A ten year longitudinal examination of the incidence rate and age of childhood encephalopathy diagnoses in an autism spectrum disorder diagnosed cohort. Acta Neurobiol Exp (Wars) 2020. [DOI: 10.21307/ane-2020-007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
52
|
Baclofen as an adjuvant therapy for autism: a randomized, double-blind, placebo-controlled trial. Eur Child Adolesc Psychiatry 2019; 28:1619-1628. [PMID: 30980177 DOI: 10.1007/s00787-019-01333-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 04/03/2019] [Indexed: 10/27/2022]
Abstract
Increasing evidence suggests that the function of the GABAergic system is abnormally low in autism spectrum disorder (ASD). Baclofen, which functions as a selective agonist for GABAB receptors, does appear promising for the treatment of ASD. We conducted a 10-week randomized-controlled study aimed at evaluating the potential of baclofen as an adjuvant therapy to enhance the effect of risperidone in children with ASD. Sixty-four children (3-12 years) with moderate-to-severe irritability symptoms of ASD were included. We used the Aberrant Behavior Checklist-Community Edition (ABC-C) for the outcome measures on each of the follow-up visits (weeks 0, 5, and 10). Analysis of the combined data revealed significant improvement for all the ABC subscales (irritability: F = 51.644, df = 1.66, p < 0.001, lethargy: F = 39.734, df = 1.38, p < 0.001, stereotypic behavior: F = 25.495, df = 1.56, p < 0.001, hyperactivity: F = 54.135, df = 1.35, p < 0.001, and inappropriate speech: F = 19.277, df = 1.47, p = 0.004). Combined treatment with baclofen and risperidone exerted a greater effect on improvement of hyperactivity symptoms at both midpoint [Cohen's d, 95% confidence interval (CI) = - 3.14, - 5.56 to - 0.72] and endpoint (d, 95% CI = - 4.45, - 8.74 to - 0.16) when compared with treatment with placebo plus risperidone. The two treatments achieved comparable results for other outcome measures. Our data support safety and efficacy of baclofen as an adjuvant to risperidone for improvement of hyperactivity symptoms in children with ASD.
Collapse
|
53
|
Al-Haddad BJS, Oler E, Armistead B, Elsayed NA, Weinberger DR, Bernier R, Burd I, Kapur R, Jacobsson B, Wang C, Mysorekar I, Rajagopal L, Adams Waldorf KM. The fetal origins of mental illness. Am J Obstet Gynecol 2019; 221:549-562. [PMID: 31207234 PMCID: PMC6889013 DOI: 10.1016/j.ajog.2019.06.013] [Citation(s) in RCA: 167] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 06/07/2019] [Accepted: 06/10/2019] [Indexed: 12/14/2022]
Abstract
The impact of infections and inflammation during pregnancy on the developing fetal brain remains incompletely defined, with important clinical and research gaps. Although the classic infectious TORCH pathogens (ie, Toxoplasma gondii, rubella virus, cytomegalovirus [CMV], herpes simplex virus) are known to be directly teratogenic, emerging evidence suggests that these infections represent the most extreme end of a much larger spectrum of injury. We present the accumulating evidence that prenatal exposure to a wide variety of viral and bacterial infections-or simply inflammation-may subtly alter fetal brain development, leading to neuropsychiatric consequences for the child later in life. The link between influenza infections in pregnant women and an increased risk for development of schizophrenia in their children was first described more than 30 years ago. Since then, evidence suggests that a range of infections during pregnancy may also increase risk for autism spectrum disorder and depression in the child. Subsequent studies in animal models demonstrated that both pregnancy infections and inflammation can result in direct injury to neurons and neural progenitor cells or indirect injury through activation of microglia and astrocytes, which can trigger cytokine production and oxidative stress. Infectious exposures can also alter placental serotonin production, which can perturb neurotransmitter signaling in the developing brain. Clinically, detection of these subtle injuries to the fetal brain is difficult. As the neuropsychiatric impact of perinatal infections or inflammation may not be known for decades after birth, our construct for defining teratogenic infections in pregnancy (eg, TORCH) based on congenital anomalies is insufficient to capture the full adverse impact on the child. We discuss the clinical implications of this body of evidence and how we might place greater emphasis on prevention of prenatal infections. For example, increasing uptake of the seasonal influenza vaccine is a key strategy to reduce perinatal infections and the risk for fetal brain injury. An important research gap exists in understanding how antibiotic therapy during pregnancy affects the fetal inflammatory load and how to avoid inflammation-mediated injury to the fetal brain. In summary, we discuss the current evidence and mechanisms linking infections and inflammation with the increased lifelong risk of neuropsychiatric disorders in the child, and how we might improve prenatal care to protect the fetal brain.
Collapse
Affiliation(s)
| | - Elizabeth Oler
- Department of Obstetrics & Gynecology, University of Washington, Seattle, WA
| | - Blair Armistead
- Department of Global Health, University of Washington Seattle, WA; Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA
| | - Nada A Elsayed
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Daniel R Weinberger
- Lieber Institute for Brain Development, Departments of Psychiatry, Neurology, Neuroscience, and McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine Baltimore, MD
| | - Raphael Bernier
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA
| | - Irina Burd
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Raj Kapur
- Department of Pediatrics, University of Washington, Seattle Children's Hospital, Seattle, WA
| | - Bo Jacobsson
- Department of Obstetrics and Gynecology, Institute of Clinical Science, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Genetics and Bioinformatics, Domain of Health Data and Digitalization, Institute of Public Health, Oslo, Norway
| | - Caihong Wang
- Department of Obstetrics and Gynecology, Center for Reproductive Health Sciences, Washington University School of Medicine, St. Louis, MO
| | - Indira Mysorekar
- Departments of Obstetrics and Gynecology and Pathology and Immunology, Center for Reproductive Health Sciences, Washington University School of Medicine, St. Louis, MO
| | - Lakshmi Rajagopal
- Center for Innate Immunity and Immune Disease, Department of Pediatrics, University of Washington, Seattle, WA; Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA
| | - Kristina M Adams Waldorf
- Department of Obstetrics & Gynecology and Global Health, Center for Innate Immunity and Immune Disease, Center for Emerging and Reemerging Infectious Diseases, University of Washington, Seattle, WA; Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
54
|
DiStasio MM, Nagakura I, Nadler MJ, Anderson MP. T lymphocytes and cytotoxic astrocyte blebs correlate across autism brains. Ann Neurol 2019; 86:885-898. [PMID: 31591744 PMCID: PMC7210715 DOI: 10.1002/ana.25610] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 09/24/2019] [Accepted: 09/25/2019] [Indexed: 01/10/2023]
Abstract
OBJECTIVE Autism spectrum disorder (ASD) affects 1 in 59 children, yet except for rare genetic causes, the etiology in most ASD remains unknown. In the ASD brain, inflammatory cytokine and transcript profiling shows increased expression of genes encoding mediators of the innate immune response. We evaluated postmortem brain tissue for adaptive immune cells and immune cell-mediated cytotoxic damage that could drive this innate immune response in the ASD brain. METHODS Standard neuropathology diagnostic methods including histology and immunohistochemistry were extended with automated image segmentation to quantify identified pathologic features in the postmortem brains. RESULTS We report multifocal perivascular lymphocytic cuffs contain increased numbers of lymphocytes in ~65% of ASD compared to control brains in males and females, across all ages, in most brain regions, and in white and gray matter, and leptomeninges. CD3+ T lymphocytes predominate over CD20+ B lymphocytes and CD8+ over CD4+ T lymphocytes in ASD brains. Importantly, the perivascular cuff lymphocyte numbers correlate to the quantity of astrocyte-derived round membranous blebs. Membranous blebs form as a cytotoxic reaction to lymphocyte attack. Consistent with multifocal immune cell-mediated injury at perivascular cerebrospinal fluid (CSF)-brain barriers, a subset of white matter vessels have increased perivascular space (with jagged contours) and collagen in ASD compared to control brains. CSF-brain barrier pathology is also evident at cerebral cortex pial and ventricular ependymal surfaces in ASD. INTERPRETATION The findings suggest dysregulated cellular immunity damages astrocytes at foci along the CSF-brain barrier in ASD. ANN NEUROL 2019;86:885-898.
Collapse
Affiliation(s)
- Marcello M. DiStasio
- Departments of Neurology and Pathology, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston, MA 02115, USA
| | - Ikue Nagakura
- Departments of Neurology and Pathology, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston, MA 02115, USA
| | - Monica J. Nadler
- Departments of Neurology and Pathology, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston, MA 02115, USA
| | - Matthew P. Anderson
- Departments of Neurology and Pathology, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston, MA 02115, USA
- Boston Children’s Hospital Intellectual and Developmental Disabilities Research Center, 300 Longwood Avenue, Boston, MA 02115, USA
- Program in Neuroscience, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| |
Collapse
|
55
|
Maternal and early postnatal immune activation produce sex-specific effects on autism-like behaviors and neuroimmune function in mice. Sci Rep 2019; 9:16928. [PMID: 31729416 PMCID: PMC6858355 DOI: 10.1038/s41598-019-53294-z] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 10/28/2019] [Indexed: 01/08/2023] Open
Abstract
Increasing evidence suggests a role for inflammation in neuropsychiatric conditions including autism spectrum disorder (ASD), a neurodevelopmental syndrome with higher prevalence in males than females. Here we examined the effects of early-life immune system activation (EIA)—comprising regimens of prenatal, early postnatal, or combined (“two-hit”) immune activation—on the core behavioral features of ASD (decreased social interaction, increased repetitive behavior, and aberrant communication) in C57BL/6J mice. We treated timed-pregnant mice with polyinosinic:polycytidylic acid (Poly I:C) on gestational day 12.5 to produce maternal immune activation (MIA). Some offspring also received lipopolysaccharide (LPS) on postnatal day 9 to produce postnatal immune activation (PIA). EIA produced disruptions in social behavior and increases in repetitive behaviors that were larger in males than in females. Ultrasonic vocalizations (USVs) were altered in both sexes. Molecular studies revealed that EIA also produced prominent sex-specific changes in inflammation-related gene expression in the brain. Whereas both sexes showed increases in pro-inflammatory factors, as reflected by levels of mRNA and protein, expression of anti-inflammatory factors was decreased in males but increased in females. Our findings demonstrate that EIA can produce sex-specific behavioral effects and immune responses in the brain, and identify molecular processes that may contribute to resilience in females.
Collapse
|
56
|
Yang G, Shcheglovitov A. Probing disrupted neurodevelopment in autism using human stem cell-derived neurons and organoids: An outlook into future diagnostics and drug development. Dev Dyn 2019; 249:6-33. [PMID: 31398277 DOI: 10.1002/dvdy.100] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Revised: 07/23/2019] [Accepted: 07/31/2019] [Indexed: 12/11/2022] Open
Abstract
Autism spectrum disorders (ASDs) represent a spectrum of neurodevelopmental disorders characterized by impaired social interaction, repetitive or restrictive behaviors, and problems with speech. According to a recent report by the Centers for Disease Control and Prevention, one in 68 children in the US is diagnosed with ASDs. Although ASD-related diagnostics and the knowledge of ASD-associated genetic abnormalities have improved in recent years, our understanding of the cellular and molecular pathways disrupted in ASD remains very limited. As a result, no specific therapies or medications are available for individuals with ASDs. In this review, we describe the neurodevelopmental processes that are likely affected in the brains of individuals with ASDs and discuss how patient-specific stem cell-derived neurons and organoids can be used for investigating these processes at the cellular and molecular levels. Finally, we propose a discovery pipeline to be used in the future for identifying the cellular and molecular deficits and developing novel personalized therapies for individuals with idiopathic ASDs.
Collapse
Affiliation(s)
- Guang Yang
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, Utah.,Neuroscience Graduate Program, University of Utah, Salt Lake City, Utah
| | - Alex Shcheglovitov
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, Utah.,Neuroscience Graduate Program, University of Utah, Salt Lake City, Utah
| |
Collapse
|
57
|
Park E, Elidrissi A, Schuller-Levis G, Chadman KK. Taurine Partially Improves Abnormal Anxiety in Taurine-Deficient Mice. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1155:905-921. [PMID: 31468456 DOI: 10.1007/978-981-13-8023-5_76] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
Taurine is abundant in various tissues including the brain, muscle, heart, spleen, liver and kidney with various physiological functions. Since taurine is produced by cysteine sulfinic acid decarboxylase (CSAD) in the liver and kidney, taurine-deficient mice without CSAD have been investigated for abnormal physiological functions such as retinal development, immune, pancreatic and liver function. In this study, the behavioral effects and abnormal brain development caused by low taurine in the developing brain were examined. In neonatal brains of homozygous CSAD knockout mice (HO), taurine was reduced by 85%, compared to wild-type mice (WT). Taurine was reduced by 35% in the brains of 2 month-old HO, compared to WT. Anxiety, motor coordination and autistic-like behaviors were evaluated at 2 months of age using five behavioral tests: elevated plus maze, open field, social approach, marble burying and accelerating rotarod. Mice were tested from 3 groups including WT, HO and HO with oral treatment of 0.2% taurine in the drinking water (HOT). HOT were born from HO dams treated with taurine from before pregnancy and were continuously treated with taurine in the drinking water after weaning. The taurine levels in the brain and plasma of HOT were restored to WT at 2 months of age. Taurine-deficiency did not lead to changes in autistic-like behaviors as the HO were not significantly different from WT in marble burying and social approach. However, taurine-deficiency increased anxiety-like behavior in HO in the elevated plus maze and open field, compared to WT. Taurine treatment significantly restored the HOT to WT levels of anxiety-like behavior in the elevated plus maze. However, changes in exploratory activity in the open field were not improved with taurine treatment. There was a slight difference in motor ability as the WT mice stayed on the accelerating rotarod longer that the HO and HOT, but the difference was significant in the HOT during the first trial only, compared to WT.These data support hypothesis that taurine is essential for the emotional development of the brain. First, taurine is remarkably low in the neonatal brain of HO, compared to the adult brain of HO. Second, taurine treatment in HO partially improves anxiety-like behavior to WT.
Collapse
Affiliation(s)
- Eunkyue Park
- Department of Developmental Neurobiology, New York Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA.
| | - Abdeslem Elidrissi
- Department of Biological Science, College of Staten Island, Staten Island, NY, USA
| | - Georgia Schuller-Levis
- Department of Developmental Neurobiology, New York Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - Kathryn K Chadman
- Department of Developmental Neurobiology, New York Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| |
Collapse
|
58
|
Mejias R, Chiu SL, Han M, Rose R, Gil-Infante A, Zhao Y, Huganir RL, Wang T. Purkinje cell-specific Grip1/2 knockout mice show increased repetitive self-grooming and enhanced mGluR5 signaling in cerebellum. Neurobiol Dis 2019; 132:104602. [PMID: 31476380 DOI: 10.1016/j.nbd.2019.104602] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 07/30/2019] [Accepted: 08/30/2019] [Indexed: 01/16/2023] Open
Abstract
Cerebellar Purkinje cell (PC) loss is a consistent pathological finding in autism. However, neural mechanisms of PC-dysfunction in autism remain poorly characterized. Glutamate receptor interacting proteins 1/2 (Grip1/2) regulate AMPA receptor (AMPAR) trafficking and synaptic strength. To evaluate role of PC-AMPAR signaling in autism, we produced PC-specific Grip1/2 knockout mice by crossing Grip2 conventional and Grip1 conditional KO with L7-Cre driver mice. PCs in the mutant mice showed normal morphology and number, and a lack of Grip1/2 expression. Rodent behavioral testing identified normal ambulation, anxiety, social interaction, and an increase in repetitive self-grooming. Electrophysiology studies revealed normal mEPSCs but an impaired mGluR-LTD at the Parallel Fiber-PC synapses. Immunoblots showed increased expression of mGluR5 and Arc, and enhanced phosphorylation of P38 and AKT in cerebellum of PC-specific Grip1/2 knockout mice. Results indicate that loss of Grip1/2 in PCs contributes to increased repetitive self-grooming, a core autism behavior in mice. Results support a role of AMPAR trafficking defects in PCs and disturbances of mGluR5 signaling in cerebellum in the pathogenesis of repetitive behaviors.
Collapse
Affiliation(s)
- Rebeca Mejias
- McKusick-Nathans Department of Genetic Medicine and Department of Pediatrics, Johns Hopkins University, Baltimore, MD 21205, USA; Department of Physiology, University of Seville, 41012 Seville, Spain.
| | - Shu-Ling Chiu
- Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Mei Han
- McKusick-Nathans Department of Genetic Medicine and Department of Pediatrics, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Rebecca Rose
- McKusick-Nathans Department of Genetic Medicine and Department of Pediatrics, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Ana Gil-Infante
- Department of Physiology, University of Seville, 41012 Seville, Spain
| | - Yifan Zhao
- McKusick-Nathans Department of Genetic Medicine and Department of Pediatrics, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Richard L Huganir
- Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Tao Wang
- McKusick-Nathans Department of Genetic Medicine and Department of Pediatrics, Johns Hopkins University, Baltimore, MD 21205, USA.
| |
Collapse
|
59
|
Hippocampal neurons in direct contact with astrocytes exposed to amyloid β 25-35 exhibit reduced excitatory synaptic transmission. IBRO Rep 2019; 7:34-41. [PMID: 31388597 PMCID: PMC6669318 DOI: 10.1016/j.ibror.2019.07.1719] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 07/16/2019] [Indexed: 12/28/2022] Open
Abstract
We exposed astrocytes to Aβ 25-35 and then co-cultured them with primary hippocampal neurons. The Aβ25-35-exposed astrocytes lowered excitatory postsynaptic release and the size of the readily releasable synaptic pool. The number of excitatory synapses was reduced by direct contact between Aβ25-35-exposed astrocytes and hippocampal neurons. The dendritic branching was decreased by direct contact between Aβ25-35-exposed astrocytes and hippocampal neurons. The number of excitatory synapses and dendrite branches were conserved by putting distance from Aβ25-35-exposed astrocytes.
Amyloid β protein (Aβ) is closely related to the progression of Alzheimer's disease because senile plaques consisting of Aβ cause synaptic depression and synaptic abnormalities. In the central nervous system, astrocytes are a major glial cell type that contribute to the modulation of synaptic transmission and synaptogenesis. In this study, we examined whether astrocytes exposed to Aβ fragment 25-35 (Aβ25-35) affect synaptic transmission. We show that synaptic transmission by hippocampal neurons was inhibited by astrocytes exposed to Aβ25-35. The Aβ25-35-exposed astrocytes lowered excitatory postsynaptic release and the size of the readily releasable synaptic pool. The number of excitatory synapses was also reduced. However, the number of excitatory synapses was unchanged unless there was direct contact between Aβ25-35-exposed astrocytes and hippocampal neurons. These data indicate that direct contact between Aβ25-35-exposed astrocytes and neurons is critical for inhibiting synaptic transmission in the progression of Alzheimer’s disease.
Collapse
|
60
|
Mazón-Cabrera R, Vandormael P, Somers V. Antigenic Targets of Patient and Maternal Autoantibodies in Autism Spectrum Disorder. Front Immunol 2019; 10:1474. [PMID: 31379804 PMCID: PMC6659315 DOI: 10.3389/fimmu.2019.01474] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 06/13/2019] [Indexed: 12/13/2022] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder whose behavioral symptoms become apparent in early childhood. The underlying pathophysiological mechanisms are only partially understood and the clinical manifestations are heterogeneous in nature, which poses a major challenge for diagnosis, prognosis and intervention. In the last years, an important role of a dysregulated immune system in ASD has emerged, but the mechanisms connecting this to a disruption of brain development are still largely unknown. Although ASD is not considered as a typical autoimmune disease, self-reactive antibodies or autoantibodies against a wide variety of targets have been found in a subset of ASD patients. In addition, autoantibodies reactive to fetal brain proteins have also been described in the prenatal stage of neurodevelopment, where they can be transferred from the mother to the fetus by transplacental transport. In this review, we give an extensive overview of the antibodies described in ASD according to their target antigens, their different origins, and timing of exposure during neurodevelopment.
Collapse
Affiliation(s)
| | | | - Veerle Somers
- Biomedical Research Institute, Faculty of Medicine and Life Science, Hasselt University, Diepenbeek, Belgium
| |
Collapse
|
61
|
Matta SM, Hill-Yardin EL, Crack PJ. The influence of neuroinflammation in Autism Spectrum Disorder. Brain Behav Immun 2019; 79:75-90. [PMID: 31029798 DOI: 10.1016/j.bbi.2019.04.037] [Citation(s) in RCA: 207] [Impact Index Per Article: 41.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 04/15/2019] [Accepted: 04/24/2019] [Indexed: 12/12/2022] Open
Abstract
Autism Spectrum Disorder (ASD) is a neurodevelopmental disorder characterised by deficits in social communication and restricted or repetitive behaviours. The clinical presentation of ASD is highly variable and diagnosis is based on the presence of impaired social communication and repetitive and/or restricted behaviours. Although the precise pathophysiologies underlying ASD are unclear, growing evidence supports a role for dysregulated neuroinflammation. The potential involvement of microglia and astrocytes reactive to inflammatory stimuli in ASD has generated much interest due to their varied roles including in mounting an immune response and regulating synaptic function. Increased numbers of reactive microglial and astrocytes in both ASD postmortem tissue and animal models have been reported. Whether dysregulation of glial subtypes exacerbates alterations in neural connectivity in the brain of autistic patients is not well explored. A role for the gut-brain axis involving microbial-immune-neuronal cross talk is also a growing area of neuroinflammation research. Greater understanding of these interactions under patho/physiological conditions and the identification of consistent immune profile abnormalities can potentially lead to more reliable diagnostic measures and treatments in ASD.
Collapse
Affiliation(s)
- Samantha M Matta
- Department of Pharmacology and Therapeutics, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Elisa L Hill-Yardin
- School of Health & Biomedical Sciences, RMIT University, Bundoora, VIC 3083, Australia; Department of Physiology, The University of Melbourne, Parkville, VIC 3010, Australia.
| | - Peter J Crack
- Department of Pharmacology and Therapeutics, The University of Melbourne, Parkville, VIC 3010, Australia.
| |
Collapse
|
62
|
Raciti M, Salma J, Spulber S, Gaudenzi G, Khalajzeyqami Z, Conti M, Anderlid BM, Falk A, Hermanson O, Ceccatelli S. NRXN1 Deletion and Exposure to Methylmercury Increase Astrocyte Differentiation by Different Notch-Dependent Transcriptional Mechanisms. Front Genet 2019; 10:593. [PMID: 31316548 PMCID: PMC6610538 DOI: 10.3389/fgene.2019.00593] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Accepted: 06/05/2019] [Indexed: 01/11/2023] Open
Abstract
Controversial evidence points to a possible involvement of methylmercury (MeHg) in the etiopathogenesis of autism spectrum disorders (ASD). In the present study, we used human neuroepithelial stem cells from healthy donors and from an autistic patient bearing a bi-allelic deletion in the gene encoding for NRXN1 to evaluate whether MeHg would induce cellular changes comparable to those seen in cells derived from the ASD patient. In healthy cells, a subcytotoxic concentration of MeHg enhanced astroglial differentiation similarly to what observed in the diseased cells (N1), as shown by the number of GFAP positive cells and immunofluorescence signal intensity. In both healthy MeHg-treated and N1 untreated cells, aberrations in Notch pathway activity seemed to play a critical role in promoting the differentiation toward glia. Accordingly, treatment with the established Notch inhibitor DAPT reversed the altered differentiation. Although our data are not conclusive since only one of the genes involved in ASD is considered, the results provide novel evidence suggesting that developmental exposure to MeHg, even at subcytotoxic concentrations, induces alterations in astroglial differentiation similar to those observed in ASD.
Collapse
Affiliation(s)
- Marilena Raciti
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Jahan Salma
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Stefan Spulber
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Giulia Gaudenzi
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | | | - Mirko Conti
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Britt-Marie Anderlid
- Centre for Molecular Medicine, Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden
| | - Anna Falk
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Ola Hermanson
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Sandra Ceccatelli
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
63
|
Propionic Acid Induces Gliosis and Neuro-inflammation through Modulation of PTEN/AKT Pathway in Autism Spectrum Disorder. Sci Rep 2019; 9:8824. [PMID: 31217543 PMCID: PMC6584527 DOI: 10.1038/s41598-019-45348-z] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 06/05/2019] [Indexed: 12/31/2022] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by glia over-proliferation, neuro-inflammation, perturbed neural circuitry, and gastrointestinal symptoms. The role of gut dys-biosis in ASD is intriguing and should be elucidated. We investigated the effect of Propionic acid (PPA), a short-chain fatty acid (SCFA) and a product of dys-biotic ASD gut, on human neural stem cells (hNSCs) proliferation, differentiation and inflammation. hNSCs proliferated to 66 neuropsheres when exposed to PPA versus 45 in control. The neurosphere diameter also increased at day 10 post PPA treatment to (Mean: 193.47 um ± SEM: 6.673 um) versus (154.16 um ± 9.95 um) in control, p < 0.001. Pre-treatment with β-HB, SCFA receptor inhibitor, hindered neurosphere expansion (p < 0.001). While hNSCs spontaneously differentiated to (48.38% ± 6.08%) neurons (Tubulin-IIIβ positive) and (46.63% ± 2.5%) glia (GFAP positive), PPA treatment drastically shifted differentiation to 80% GFAP cells (p < 0.05). Following 2 mM PPA exposure, TNF-α transcription increased 4.98 fold and the cytokine increased 3.29 fold compared to control (P < 0.001). Likewise, GPR41 (PPA receptor) and pro-survival p-Akt protein were elevated (p < 0.001). PTEN (Akt inhibitor) level decreased to (0.42 ug/ul ± 0.04 ug/ul) at 2 mM PPA compared to (0.83 ug/ul ± 0.09 ug/ul) in control (p < 0.001). PPA at 2 mM decreased neurite outgrowth to (80.70 um ± 5.5 um) compared to (194.93 um ± 19.7 um) in control. Clearly, the data supports a significant role for PPA in modulating hNSC patterning leading to gliosis, disturbed neuro-circuitry, and inflammatory response as seen in ASD.
Collapse
|
64
|
Lee FHF, Lai TKY, Su P, Liu F. Altered cortical Cytoarchitecture in the Fmr1 knockout mouse. Mol Brain 2019; 12:56. [PMID: 31200759 PMCID: PMC6570929 DOI: 10.1186/s13041-019-0478-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 06/03/2019] [Indexed: 12/15/2022] Open
Abstract
Fragile X syndrome (FXS) is a neurodevelopmental disorder caused by silencing of the FMR1 gene and subsequent loss of its protein product, fragile X retardation protein (FMRP). One of the most robust neuropathological findings in post-mortem human FXS and Fmr1 KO mice is the abnormal increase in dendritic spine densities, with the majority of spines showing an elongated immature morphology. However, the exact mechanisms of how FMRP can regulate dendritic spine development are still unclear. Abnormal dendritic spines can result from disturbances of multiple factors during neurodevelopment, such as alterations in neuron numbers, position and glial cells. In this study, we undertook a comprehensive histological analysis of the cerebral cortex in Fmr1 KO mice. They displayed significantly fewer neuron and PV-interneuron numbers, along with altered cortical lamination patterns. In terms of glial cells, Fmr1 KO mice exhibited an increase in Olig2-oligodendrocytes, which corresponded to the abnormally higher myelin expression in the corpus callosum. Iba1-microglia were significantly reduced but GFAP-astrocyte numbers and intensity were elevated. Using primary astrocytes derived from KO mice, we further demonstrated the presence of astrogliosis characterized by an increase in GFAP expression and astrocyte hypertrophy. Our findings provide important information on the cortical architecture of Fmr1 KO mice, and insights towards possible mechanisms associated with FXS.
Collapse
Affiliation(s)
- Frankie H F Lee
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, M5T 1R8, Canada
| | - Terence K Y Lai
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, M5T 1R8, Canada.,Department of Physiology, University of Toronto, Toronto, Ontario, M5T 1R8, Canada
| | - Ping Su
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, M5T 1R8, Canada
| | - Fang Liu
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, M5T 1R8, Canada. .,Department of Physiology, University of Toronto, Toronto, Ontario, M5T 1R8, Canada. .,Department of Psychiatry, University of Toronto, Toronto, Ontario, M5T 1R8, Canada.
| |
Collapse
|
65
|
Mohammad AS, Adkins CE, Shah N, Aljammal R, Griffith JIG, Tallman RM, Jarrell KL, Lockman PR. Permeability changes and effect of chemotherapy in brain adjacent to tumor in an experimental model of metastatic brain tumor from breast cancer. BMC Cancer 2018; 18:1225. [PMID: 30526520 PMCID: PMC6286543 DOI: 10.1186/s12885-018-5115-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 11/20/2018] [Indexed: 02/07/2023] Open
Abstract
Background Brain tumor vasculature can be significantly compromised and leakier than that of normal brain blood vessels. Little is known if there are vascular permeability alterations in the brain adjacent to tumor (BAT). Changes in BAT permeability may also lead to increased drug permeation in the BAT, which may exert toxicity on cells of the central nervous system. Herein, we studied permeation changes in BAT using quantitative fluorescent microscopy and autoradiography, while the effect of chemotherapy within the BAT region was determined by staining for activated astrocytes. Methods Human metastatic breast cancer cells (MDA-MB-231Br) were injected into left ventricle of female NuNu mice. Metastases were allowed to grow for 28 days, after which animals were injected fluorescent tracers Texas Red (625 Da) or Texas Red dextran (3 kDa) or a chemotherapeutic agent 14C-paclitaxel. The accumulation of tracers and 14C-paclitaxel in BAT were determined by using quantitative fluorescent microscopy and autoradiography respectively. The effect of chemotherapy in BAT was determined by staining for activated astrocytes. Results The mean permeability of texas Red (625 Da) within BAT region increased 1.0 to 2.5-fold when compared to normal brain, whereas, Texas Red dextran (3 kDa) demonstrated mean permeability increase ranging from 1.0 to 1.8-fold compared to normal brain. The Kin values in the BAT for both Texas Red (625 Da) and Texas Red dextran (3 kDa) were found to be 4.32 ± 0.2 × 105 mL/s/g and 1.6 ± 1.4 × 105 mL/s/g respectively and found to be significantly higher than the normal brain. We also found that there is significant increase in accumulation of 14C-Paclitaxel in BAT compared to the normal brain. We also observed animals treated with chemotherapy (paclitaxel (10 mg/kg), erubilin (1.5 mg/kg) and docetaxel (10 mg/kg)) showed activated astrocytes in BAT. Conclusions Our data showed increased permeation of fluorescent tracers and 14C-paclitaxel in the BAT. This increased permeation lead to elevated levels of activated astrocytes in BAT region in the animals treated with chemotherapy.
Collapse
Affiliation(s)
- Afroz S Mohammad
- Department of Pharmaceutical Sciences, West Virginia University Health Sciences Center, School of Pharmacy, 1 Medical Center Drive, Morgantown, West Virginia, 26506-9050, USA
| | - Chris E Adkins
- Department of Pharmaceutical Sciences, West Virginia University Health Sciences Center, School of Pharmacy, 1 Medical Center Drive, Morgantown, West Virginia, 26506-9050, USA
| | - Neal Shah
- Department of Pharmaceutical Sciences, West Virginia University Health Sciences Center, School of Pharmacy, 1 Medical Center Drive, Morgantown, West Virginia, 26506-9050, USA
| | - Rawaa Aljammal
- Department of Pharmaceutical Sciences, West Virginia University Health Sciences Center, School of Pharmacy, 1 Medical Center Drive, Morgantown, West Virginia, 26506-9050, USA
| | - Jessica I G Griffith
- Department of Pharmaceutical Sciences, West Virginia University Health Sciences Center, School of Pharmacy, 1 Medical Center Drive, Morgantown, West Virginia, 26506-9050, USA
| | - Rachel M Tallman
- Department of Pharmaceutical Sciences, West Virginia University Health Sciences Center, School of Pharmacy, 1 Medical Center Drive, Morgantown, West Virginia, 26506-9050, USA
| | - Katherine L Jarrell
- Department of Pharmaceutical Sciences, West Virginia University Health Sciences Center, School of Pharmacy, 1 Medical Center Drive, Morgantown, West Virginia, 26506-9050, USA
| | - Paul R Lockman
- Department of Pharmaceutical Sciences, West Virginia University Health Sciences Center, School of Pharmacy, 1 Medical Center Drive, Morgantown, West Virginia, 26506-9050, USA.
| |
Collapse
|
66
|
Azhari A, Azizan F, Esposito G. A systematic review of gut-immune-brain mechanisms in Autism Spectrum Disorder. Dev Psychobiol 2018; 61:752-771. [PMID: 30523646 DOI: 10.1002/dev.21803] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 09/10/2018] [Accepted: 10/09/2018] [Indexed: 12/13/2022]
Abstract
Despite decades of research, the etiological origins of Autism Spectrum Disorder (ASD) remain elusive. Recently, the mechanisms of ASD have encompassed emerging theories involving the gastrointestinal, immune, and nervous systems. While each of these perspectives presents its own set of supporting evidence, the field requires an integration of these modular concepts and an overarching view of how these subsystems intersect. In this systematic review, we have synthesized relevant evidences from the existing literature, evaluating them in an interdependent manner and in doing so, outlining their possible connections. Specifically, we first discussed gastrointestinal and immuno-inflammation pathways in-depth, exploring the relationships between microbial composition, bacterial metabolites, gut mucosa, and immune system constituents. Accounting for temporal differences in the mechanisms involved in neurodevelopment, prenatal and postnatal phases were further elucidated, where the former focused on maternal immune activation (MIA) and fetal development, while the latter addressed the role of immune dysregulation in contributing to atypical neurodevelopment. As autism remains, foremost, a neurodevelopmental disorder, this review presents an integration of disparate modules into a "Gut-Immune-Brain" paradigm. Existing gaps in the literature have been highlighted, and possible avenues for future research with an integrated physiological perspective underlying ASD have also been suggested.
Collapse
Affiliation(s)
- Atiqah Azhari
- Psychology Program, School of Social Sciences, Nanyang Technological University, Singapore, Singapore
| | - Farouq Azizan
- Psychology Program, School of Social Sciences, Nanyang Technological University, Singapore, Singapore
| | - Gianluca Esposito
- Psychology Program, School of Social Sciences, Nanyang Technological University, Singapore, Singapore.,Department of Psychology and Cognitive Science, University of Trento, Rovereto, TN, Italy
| |
Collapse
|
67
|
Scott H, Phillips T, Stuart G, Rogers M, Steinkraus B, Grant S, Case C. Preeclamptic placentae release factors that damage neurons: implications for foetal programming of disease. Neuronal Signal 2018; 2:NS20180139. [PMID: 32714596 PMCID: PMC7363326 DOI: 10.1042/ns20180139] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 09/03/2018] [Accepted: 09/18/2018] [Indexed: 12/14/2022] Open
Abstract
Prenatal development is a critical period for programming of neurological disease. Preeclampsia, a pregnancy complication involving oxidative stress in the placenta, has been associated with long-term health implications for the child, including an increased risk of developing schizophrenia and autism spectrum disorders in later life. To investigate if molecules released by the placenta may be important mediators in foetal programming of the brain, we analysed if placental tissue delivered from patients with preeclampsia secreted molecules that could affect cortical cells in culture. Application of culture medium conditioned by preeclamptic placentae to mixed cortical cultures caused changes in neurons and astrocytes that were related to key changes observed in brains of patients with schizophrenia and autism, including effects on dendrite lengths, astrocyte number as well as on levels of glutamate and γ-aminobutyric acid receptors. Treatment of the placental explants with an antioxidant prevented neuronal abnormalities. Furthermore, we identified that bidirectional communication between neurons and astrocytes, potentially via glutamate, is required to produce the effects of preeclamptic placenta medium on cortical cells. Analysis of possible signalling molecules in the placenta-conditioned medium showed that the secretion profile of extracellular microRNAs, small post-transcriptional regulators, was altered in preeclampsia and partially rescued by antioxidant treatment of the placental explants. Predicted targets of these differentially abundant microRNAs were linked to neurodevelopment and the placenta. The present study provides further evidence that the diseased placenta may release factors that damage cortical cells and suggests the possibility of targeted antioxidant treatment of the placenta to prevent neurodevelopmental disorders.
Collapse
Affiliation(s)
- Hannah Scott
- School of Clinical Sciences, University of Bristol, Learning & Research Building, Southmead Hospital, Bristol BS10 5NB, U.K
- UK Dementia Research Institute, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff CF24 4HQ, U.K
| | - Tom J. Phillips
- School of Clinical Sciences, University of Bristol, Learning & Research Building, Southmead Hospital, Bristol BS10 5NB, U.K
- UK Dementia Research Institute, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff CF24 4HQ, U.K
| | - Greer C. Stuart
- Department of Obstetrics, Southmead Hospital, Bristol BS10 5NB, U.K
| | - Mark F. Rogers
- Intelligent Systems Laboratory, University of Bristol, Merchant Venturers Building, Woodland Road, Bristol BS8 1UB, U.K
| | - Bruno R. Steinkraus
- Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DS, U.K
| | - Simon Grant
- Department of Obstetrics, Southmead Hospital, Bristol BS10 5NB, U.K
| | - C. Patrick Case
- School of Clinical Sciences, University of Bristol, Learning & Research Building, Southmead Hospital, Bristol BS10 5NB, U.K
| |
Collapse
|
68
|
Wegiel J, Brown WT, La Fauci G, Adayev T, Kascsak R, Kascsak R, Flory M, Kaczmarski W, Kuchna I, Nowicki K, Martinez-Cerdeno V, Wisniewski T, Wegiel J. The role of reduced expression of fragile X mental retardation protein in neurons and increased expression in astrocytes in idiopathic and syndromic autism (duplications 15q11.2-q13). Autism Res 2018; 11:1316-1331. [PMID: 30107092 DOI: 10.1002/aur.2003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 05/29/2018] [Accepted: 06/13/2018] [Indexed: 01/23/2023]
Abstract
Fragile X syndrome (FXS), caused by lack of fragile X mental retardation protein (FMRP), is associated with a high prevalence of autism. The deficit of FMRP reported in idiopathic autism suggests a mechanistic overlap between FXS and autism. The overall goal of this study is to detect neuropathological commonalities of FMRP deficits in the brains of people with idiopathic autism and with syndromic autism caused by dup15q11.2-q13 (dup15). This study tests the hypothesis based on our preliminary data that both idiopathic and syndromic autism are associated with brain region-specific deficits of neuronal FMRP and structural changes of the affected neurons. This immunocytochemical study revealed neuronal FMRP deficits and shrinkage of deficient neurons in the cerebral cortex, subcortical structures, and cerebellum in subjects with idiopathic and dup(15)/autism. Neuronal FMRP deficit coexists with surprising infiltration of the brains of autistic children and adults with FMRP-positive astrocytes known to be typical only for the fetal and short postnatal periods. In the examined autistic subjects, these astrocytes selectively infiltrate the border between white and gray matter in the cerebral and cerebellar cortex, the molecular layer of the cortex, part of the amygdala and thalamus, central cerebellar white matter, and dentate nucleus. Astrocyte pathology results in an additional local loss of FMRP in neurons and their shrinkage. Neuronal deficit of FMRP and shrinkage of affected neurons in structures free of FMRP-positive astrocytes and regions infiltrated with FMRP-expressing astrocytes appear to reflect mechanistic, neuropathological, and functional commonalities of FMRP abnormalities in FXS and autism spectrum disorder. Autism Res 2018, 11: 1316-1331. © 2018 International Society for Autism Research, Wiley Periodicals, Inc. LAY SUMMARY: Immunocytochemistry reveals a deficit of fragile X mental retardation protein (FMRP) in neurons of cortical and subcortical brain structures but increased FMRP expression in astrocytes infiltrating gray and white matter. The detected shrinkage of FMRP-deficient neurons may provide a mechanistic explanation of reported neuronal structural and functional changes in autism. This study contributes to growing evidence of mechanistic commonalities between fragile X syndrome and autism spectrum disorder.
Collapse
Affiliation(s)
- Jarek Wegiel
- Department of Developmental Neurobiology, NYS Institute for Basic Research in Developmental Disabilities, Staten Island, New York
| | - W Ted Brown
- Department of Human Genetics, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, New York
| | - Giuseppe La Fauci
- Department of Human Genetics, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, New York
| | - Tatyana Adayev
- Department of Human Genetics, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, New York
| | - Richard Kascsak
- Department of Developmental Biochemistry, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, New York
| | - Regina Kascsak
- Department of Developmental Biochemistry, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, New York
| | - Michael Flory
- Research Design and Analysis Service, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, New York
| | - Wojciech Kaczmarski
- Department of Developmental Neurobiology, NYS Institute for Basic Research in Developmental Disabilities, Staten Island, New York
| | - Izabela Kuchna
- Department of Developmental Neurobiology, NYS Institute for Basic Research in Developmental Disabilities, Staten Island, New York
| | - Krzysztof Nowicki
- Department of Developmental Neurobiology, NYS Institute for Basic Research in Developmental Disabilities, Staten Island, New York
| | - Veronica Martinez-Cerdeno
- Pathology and Laboratory Medicine, Institute for Pediatric Regenerative Medicine, MIND Institute, University of California, Davis, California
| | - Thomas Wisniewski
- Departments of Neurology, Pathology, and Psychiatry, NYU Langone Medical Center, New York, New York
| | - Jerzy Wegiel
- Department of Developmental Neurobiology, NYS Institute for Basic Research in Developmental Disabilities, Staten Island, New York
| |
Collapse
|
69
|
Mony TJ, Lee JW, Kim SS, Chun W, Lee HJ. Early Postnatal Valproic Acid Exposure Increase the Protein Level of Astrocyte Markers in Frontal Cortex of Rat. CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE 2018; 16:214-217. [PMID: 29739136 PMCID: PMC5953022 DOI: 10.9758/cpn.2018.16.2.214] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 03/05/2017] [Accepted: 03/29/2017] [Indexed: 11/18/2022]
Abstract
Objective In our previous study, it has been reported that valproic acid (VPA) effects gliogenesis and increases the number of glial precursor cells during the early postnatal period. However there is no specific report that whether this process is going on up to the age of mature brain development and the consequence effect of this ongoing gliogenesis process. Methods As an ongoing study, using Immunoblotting analysis, we checked the level of glial protein and glial-derived factor markers in the frontal cortex of a rat brain at postnatal day (PND) 21. Results The finding of the study suggests that, in the VPA group (p<0.05), early exposure elicited significantly to increase the expression level of glial protein cells at PND 21 in the frontal cortex of rat brain. Conclusion Therefore we suggest that, alter gliogenesis and abnormal number of glial cells modulate the neurobiological dysfunction and induces the risk of neurodevelopmental disorders.
Collapse
Affiliation(s)
- Tamanna Jahan Mony
- Department of Pharmacology, Kangwon National University School of Medicine, Chuncheon, Korea
| | - Jae Won Lee
- Department of Pharmacology, Kangwon National University School of Medicine, Chuncheon, Korea.,National Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungju, Korea
| | - Sung Soo Kim
- Department of Pharmacology, Kangwon National University School of Medicine, Chuncheon, Korea
| | - Wanjoo Chun
- Department of Pharmacology, Kangwon National University School of Medicine, Chuncheon, Korea
| | - Hee Jae Lee
- Department of Pharmacology, Kangwon National University School of Medicine, Chuncheon, Korea
| |
Collapse
|
70
|
Lee TT, Skafidas E, Dottori M, Zantomio D, Pantelis C, Everall I, Chana G. No preliminary evidence of differences in astrocyte density within the white matter of the dorsolateral prefrontal cortex in autism. Mol Autism 2017; 8:64. [PMID: 29234492 PMCID: PMC5721546 DOI: 10.1186/s13229-017-0181-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 11/28/2017] [Indexed: 12/27/2022] Open
Abstract
Background While evidence for white matter and astrocytic abnormalities exist in autism, a detailed investigation of astrocytes has not been conducted. Such an investigation is further warranted by an increasing role for neuroinflammation in autism pathogenesis, with astrocytes being key players in this process. We present the first study of astrocyte density and morphology within the white matter of the dorsolateral prefrontal cortex (DLPFC) in individuals with autism. Methods DLPFC formalin-fixed sections containing white matter from individuals with autism (n = 8, age = 4-51 years) and age-matched controls (n = 7, age = 4-46 years) were immunostained for glial fibrillary acidic protein (GFAP). Density of astrocytes and other glia were estimated via the optical fractionator, astrocyte somal size estimated via the nucleator, and astrocyte process length via the spaceballs probe. Results We found no evidence for alteration in astrocyte density within DLPFC white matter of individuals with autism versus controls, together with no differences in astrocyte somal size and process length. Conclusion Our results suggest that astrocyte abnormalities within the white matter in the DLPFC in autism may be less pronounced than previously thought. However, astrocytic dysregulation may still exist in autism, even in the absence of gross morphological changes. Our lack of evidence for astrocyte abnormalities could have been confounded to an extent by having a small sample size and wide age range, with pathological features potentially restricted to early stages of autism. Nonetheless, future investigations would benefit from assessing functional markers of astrocytes in light of the underlying pathophysiology of autism.
Collapse
Affiliation(s)
- Ting Ting Lee
- Department of Psychiatry, The University of Melbourne, Parkville, Australia.,Centre for Neural Engineering, The University of Melbourne, Parkville, Australia
| | - Efstratios Skafidas
- Department of Psychiatry, The University of Melbourne, Parkville, Australia.,Centre for Neural Engineering, The University of Melbourne, Parkville, Australia.,Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Australia
| | - Mirella Dottori
- Centre for Neural Engineering, The University of Melbourne, Parkville, Australia.,Department of Biomedical Engineering, The University of Melbourne, Parkville, Australia.,Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, Australia
| | - Daniela Zantomio
- Department of Clinical Haematology, Austin Hospital, Heidelberg, Australia
| | - Christos Pantelis
- Department of Psychiatry, The University of Melbourne, Parkville, Australia.,Centre for Neural Engineering, The University of Melbourne, Parkville, Australia.,Melbourne Neuropsychiatry Centre, The University of Melbourne & Melbourne Health, Parkville, Australia.,Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Australia
| | - Ian Everall
- Department of Psychiatry, The University of Melbourne, Parkville, Australia.,Centre for Neural Engineering, The University of Melbourne, Parkville, Australia.,Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Australia.,The Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Gursharan Chana
- Department of Psychiatry, The University of Melbourne, Parkville, Australia.,Centre for Neural Engineering, The University of Melbourne, Parkville, Australia.,Department of Medicine, The University of Melbourne, Parkville, Australia.,Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Australia
| |
Collapse
|
71
|
Chen J, He W, Hu X, Shen Y, Cao J, Wei Z, Luan Y, He L, Jiang F, Tao Y. A role for ErbB signaling in the induction of reactive astrogliosis. Cell Discov 2017; 3:17044. [PMID: 29238610 PMCID: PMC5717352 DOI: 10.1038/celldisc.2017.44] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 10/23/2017] [Indexed: 02/07/2023] Open
Abstract
Reactive astrogliosis is a hallmark of many neurological disorders, yet its functions and molecular mechanisms remain elusive. Particularly, the upstream signaling that regulates pathological responses of astrocytes is largely undetermined. We used a mouse traumatic brain injury model to induce astrogliosis and revealed activation of ErbB receptors in reactive astrocytes. Moreover, cell-autonomous inhibition of ErbB receptor activity in reactive astrocytes by a genetic approach suppressed hypertrophic remodeling possibly through the regulation of actin dynamics. However, inhibiting ErbB signaling in reactive astrocytes did not affect astrocyte proliferation after brain injury, although it aggravated local inflammation. In contrast, active ErbB signaling in mature astrocytes of various brain regions in mice was sufficient to initiate reactive responses, reproducing characterized molecular and cellular features of astrogliosis observed in injured or diseased brains. Further, prevalent astrogliosis in the brain induced by astrocytic ErbB activation caused anorexia in animals. Therefore, our findings defined an unrecognized role of ErbB signaling in inducing reactive astrogliosis. Mechanistically, inhibiting ErbB signaling in reactive astrocytes prominently reduced Src and focal adhesion kinase (FAK) activity that is important for actin remodeling, although ErbB signaling activated multiple downstream signaling proteins. The discrepancies between the results from loss- and gain-of-function studies indicated that ErbB signaling regulated hypertrophy and proliferation of reactive astrocytes by different downstream signaling pathways. Our work demonstrated an essential mechanism in the pathological regulation of astrocytes and provided novel insights into potential therapeutic targets for astrogliosis-implicated diseases.
Collapse
Affiliation(s)
- Jing Chen
- Institute of Life Sciences, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, China.,Key Lab of Organ Development and Regeneration of Zhejiang Province, Hangzhou, China.,Key Lab of GEM Resource and Model Research of Hangzhou, Hangzhou, Zhejiang, China
| | - Wanwan He
- Institute of Life Sciences, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, China
| | - Xu Hu
- Institute of Life Sciences, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, China
| | - Yuwen Shen
- Institute of Life Sciences, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, China
| | - Junyan Cao
- Institute of Life Sciences, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, China
| | - Zhengdong Wei
- Institute of Life Sciences, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, China
| | - Yifei Luan
- Institute of Life Sciences, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, China
| | - Li He
- Institute of Life Sciences, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, China
| | - Fangdun Jiang
- Institute of Life Sciences, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, China
| | - Yanmei Tao
- Institute of Life Sciences, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, China.,Key Lab of Organ Development and Regeneration of Zhejiang Province, Hangzhou, China.,Key Lab of GEM Resource and Model Research of Hangzhou, Hangzhou, Zhejiang, China
| |
Collapse
|
72
|
Li Y, Zhou Y, Peng L, Zhao Y. Reduced protein expressions of cytomembrane GABA ARβ3 at different postnatal developmental stages of rats exposed prenatally to valproic acid. Brain Res 2017; 1671:33-42. [PMID: 28641949 DOI: 10.1016/j.brainres.2017.06.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 05/15/2017] [Accepted: 06/13/2017] [Indexed: 11/20/2022]
Abstract
Decreased inhibition plays an extremely important role in pathogenesis of autism spectrum disorder (ASD). Therefore, we aimed to determine whether expression levels of the γ-aminobutyric acid type A receptor β3 subunit (GABAARβ3), K+-Cl- cotransporter 2 (KCC2), and Na+-K+-Cl- cotransporter 1 (NKCC1) related to inhibition transmission are changed in a sodium valproate-induced rat model of ASD. Decreased expression levels of membrane GABAARβ3 (m-GABAARβ3) and KCC2 as well as increased endocytosis of GABAARs were found in the model group. However, there were no significant differences in expression of total GABAARβ3 and NKCC1 between the control and model groups. In addition, we observed growth retardation, impaired spatial memory, limited exploration, increased anxiety, and reduced sociability in the model group. These results suggest alterations in m-GABAARβ3 levels, KCC2 levels, and trafficking of GABAARs in rats prenatally exposed to valproic acid and advance our understanding of the pathogenesis of ASD.
Collapse
Affiliation(s)
- Yixin Li
- Department of Pathology, Chongqing Medical University, Chongqing, People's Republic of China; Institute of Neuroscience, Chongqing Medical University, Chongqing, People's Republic of China; Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing, People's Republic of China
| | - Yang Zhou
- Department of Pathology, Chongqing Medical University, Chongqing, People's Republic of China; Institute of Neuroscience, Chongqing Medical University, Chongqing, People's Republic of China; Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing, People's Republic of China
| | - Li Peng
- Department of Pathology, Chongqing Medical University, Chongqing, People's Republic of China; Institute of Neuroscience, Chongqing Medical University, Chongqing, People's Republic of China; Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing, People's Republic of China
| | - Yong Zhao
- Department of Pathology, Chongqing Medical University, Chongqing, People's Republic of China; Institute of Neuroscience, Chongqing Medical University, Chongqing, People's Republic of China; Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing, People's Republic of China.
| |
Collapse
|
73
|
Allergic Inflammation Leads to Neuropathic Pain via Glial Cell Activation. J Neurosci 2017; 36:11929-11945. [PMID: 27881779 DOI: 10.1523/jneurosci.1981-16.2016] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 09/23/2016] [Accepted: 09/27/2016] [Indexed: 01/08/2023] Open
Abstract
Allergic and atopic disorders have increased over the past few decades and have been associated with neuropsychiatric conditions, such as autism spectrum disorder and asthmatic amyotrophy. Myelitis presenting with neuropathic pain can occur in patients with atopic disorder; however, the relationship between allergic inflammation and neuropathic pain, and the underlying mechanism, remains to be established. We studied whether allergic inflammation affects the spinal nociceptive system. We found that mice with asthma, atopic dermatitis, or atopic diathesis had widespread and significantly more activated microglia and astroglia in the spinal cord than those without atopy, and displayed tactile allodynia. Microarray analysis of isolated microglia revealed a dysregulated phenotype showing upregulation of M1 macrophage markers and downregulation of M2 markers in atopic mice. Among the cell surface protein genes, endothelin receptor type B (EDNRB) was most upregulated. Immunohistochemical analysis revealed that EDNRB expression was enhanced in microglia and astroglia, whereas endothelin-1, an EDNRB ligand, was increased in serum, lungs, and epidermis of atopic mice. No EDNRA expression was found in the spinal cord. Expression of FBJ murine osteosarcoma viral oncogene homolog B was significantly higher in the dorsal horn neurons of asthma mice than nonatopic mice. The EDNRB antagonist BQ788 abolished glial and neural activation and allodynia. We found increased serum endothelin-1 in atopic patients with myelitis and neuropathic pain, and activation of spinal microglia and astroglia with EDNRB upregulation in an autopsied case. These results suggest that allergic inflammation induces diffuse glial activation, influencing the nociceptive system via the EDNRB pathway. SIGNIFICANCE STATEMENT The prevalence of allergic disorders has markedly increased over the past few decades. Allergic disorders are associated with neuropsychiatric conditions; however, the relationship between allergic inflammation and CNS complications is unknown. A peculiar myelitis presenting with persistent neuropathic pain has been reported in patients with allergic disorders. We studied how atopy exerts substantial influence on the nociceptive system. We found that mice with allergic disorders had severe allodynia with activated astroglia and microglia, and showed marked upregulation of endothelin-1 (ET-1) receptor type B (EDNRB) in the spinal cord. A selective EDNRB antagonist prevented allodynia and glial activation. Our findings suggest a novel mechanism whereby atopy induces glial activation and neuropathic pain via an ET-1/EDNRB pathway.
Collapse
|
74
|
Naik AA, Patro N, Seth P, Patro IK. Intra-generational protein malnutrition impairs temporal astrogenesis in rat brain. Biol Open 2017; 6:931-942. [PMID: 28546341 PMCID: PMC5550907 DOI: 10.1242/bio.023432] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The lack of information on astrogenesis following stressor effect, notwithstanding the imperative roles of astroglia in normal physiology and pathophysiology, incited us to assess temporal astrogenesis and astrocyte density in an intra-generational protein malnutrition (PMN) rat model. Standard immunohistochemical procedures for glial lineage markers and their intensity measurements, and qRT-PCR studies, were performed to reveal the spatio-temporal origin and density of astrocytes. Reduced A2B5+ glia restricted precursor population in ventricles and caused poor dissemination to cortex at embryonic days (E)11-14, and low BLBP+ secondary radial glia in the subventricular zone (SVZ) of E16 low protein (LP) brains reflect compromised progenitor pooling. Contrary to large-sized BLBP+ gliospheres in high protein (HP) brains at E16, small gliospheres and discrete BLBP+ cells in LP brains evidence loss of colonization and low proliferative potential. Delayed emergence of GFAP expression, precocious astrocyte maturation and significantly reduced astrocyte number suggest impaired temporal and compromised astrogenesis within LP-F1 brains. Our findings of protein deprivation induced impairments in temporal astrogenesis, compromised density and astrocytic dysfunction, strengthen the hypothesis of astrocytes as possible drivers of neurodevelopmental disorders. This study may increase our understanding of stressor-associated brain development, opening up windows for effective therapeutic interventions against debilitating neurodevelopmental disorders. Summary: Maternal protein deprivation results in low progenitor pooling, and delayed and compromised astrogenesis, suggesting astrocyte impairment as a driver of neurological diseases owing to their imperative roles in normal and pathological situations.
Collapse
Affiliation(s)
- Aijaz Ahmad Naik
- School of Studies in Neuroscience, Jiwaji University, Gwalior 474011, India.,School of Studies in Zoology, Jiwaji University, Gwalior 474011, India
| | - Nisha Patro
- School of Studies in Neuroscience, Jiwaji University, Gwalior 474011, India
| | - Pankaj Seth
- National Brain Research Centre, Manesar, Haryana 122051, India
| | - Ishan K Patro
- School of Studies in Neuroscience, Jiwaji University, Gwalior 474011, India .,School of Studies in Zoology, Jiwaji University, Gwalior 474011, India
| |
Collapse
|
75
|
Esnafoglu E, Ayyıldız SN, Cırrık S, Erturk EY, Erdil A, Daglı A, Noyan T. Evaluation of serum Neuron‐specific enolase, S100B, myelin basic protein and glial fibrilliary acidic protein as brain specific proteins in children with autism spectrum disorder. Int J Dev Neurosci 2017; 61:86-91. [DOI: 10.1016/j.ijdevneu.2017.06.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 06/12/2017] [Accepted: 06/30/2017] [Indexed: 12/11/2022] Open
Affiliation(s)
- Erman Esnafoglu
- Department of Child and Adolescent PsychiatryTraining and Research HospitalFaculty of MedicineOrdu UniversityOrduTurkey
| | - Sema Nur Ayyıldız
- Department of BiochemistryTraining and Research HospitalFaculty of MedicineOrdu UniversityOrduTurkey
| | - Selma Cırrık
- Department of Medical Physiology, Faculty of MedicineOrdu UniversityOrduTurkey
| | - Emine Yurdakul Erturk
- Department of PediatryTraining and Research HospitalFaculty of MedicineOrdu UniversityOrduTurkey
| | - Abdullah Erdil
- Department of PediatryTraining and Research HospitalFaculty of MedicineOrdu UniversityOrduTurkey
| | - Abdullah Daglı
- Department of PediatryTraining and Research HospitalFaculty of MedicineOrdu UniversityOrduTurkey
| | - Tevfik Noyan
- Department of BiochemistryTraining and Research HospitalFaculty of MedicineOrdu UniversityOrduTurkey
| |
Collapse
|
76
|
Zuiki M, Chiyonobu T, Yoshida M, Maeda H, Yamashita S, Kidowaki S, Hasegawa T, Gotoh H, Nomura T, Ono K, Hosoi H, Morimoto M. Luteolin attenuates interleukin-6-mediated astrogliosis in human iPSC-derived neural aggregates: A candidate preventive substance for maternal immune activation-induced abnormalities. Neurosci Lett 2017; 653:296-301. [PMID: 28595950 DOI: 10.1016/j.neulet.2017.06.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 05/26/2017] [Accepted: 06/04/2017] [Indexed: 11/30/2022]
Abstract
Maternal infection during pregnancy increases the risk of neurodevelopmental conditions such as autism spectrum disorders and schizophrenia in offspring. Several previous animal studies have indicated that maternal immune activation (MIA), rather than a specific pathogen, alters fetal brain development. Among them, prenatal exposure to interleukin-6 (IL-6) has been associated with behavioral and neuropathological abnormalities, though such findings remain to be elucidated in humans. We developed a human cell-based model of MIA by exposing human induced pluripotent stem cells (hiPSCs)-derived neural aggregates to IL-6 and investigated whether luteolin-a naturally occurring flavonoid found in edible plants-could prevent MIA-induced abnormalities. We generated neural aggregates from hiPSCs using the serum-free floating culture of embryoid body-like aggregates with quick reaggregation (SFEBq) method, following which aggregates were cultured in suspension. We then exposed the aggregates to IL-6 (100ng/ml) for 24h at day 51. Transient IL-6 exposure significantly increased the area ratio of astrocytes (GFAP-positive area ratio) and decreased the area ratio of early-born neurons (TBR1-positive or CTIP2-positive area ratio) relative to controls. In addition, western blot analysis revealed that levels of phosphorylated STAT3 were significantly elevated in IL-6-exposed neural aggregates. Luteolin treatment inhibited STAT3 phosphorylation and counteracted IL-6-mediated increases of GFAP-positive cells and reductions of TBR1-positive and CTIP2-positive cells. Our observations suggest that the flavonoid luteolin may attenuate or prevent MIA-induced neural abnormalities. As we observed increased apoptosis at high concentrations of luteolin, further studies are required to determine the optimal intake dosage and duration for pregnant women.
Collapse
Affiliation(s)
- Masashi Zuiki
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo, Kyoto 602-8566, Japan
| | - Tomohiro Chiyonobu
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo, Kyoto 602-8566, Japan.
| | - Michiko Yoshida
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo, Kyoto 602-8566, Japan
| | - Hiroshi Maeda
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo, Kyoto 602-8566, Japan
| | - Satoshi Yamashita
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo, Kyoto 602-8566, Japan
| | - Satoshi Kidowaki
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo, Kyoto 602-8566, Japan
| | - Tatsuji Hasegawa
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo, Kyoto 602-8566, Japan
| | - Hitoshi Gotoh
- Department of Biology, Kyoto Prefectural University of Medicine, Inamori Memorial Building, 1-5 Shimogamo hangi-cho, Sakyo, Kyoto 606-0823, Japan
| | - Tadashi Nomura
- Department of Biology, Kyoto Prefectural University of Medicine, Inamori Memorial Building, 1-5 Shimogamo hangi-cho, Sakyo, Kyoto 606-0823, Japan
| | - Katsuhiko Ono
- Department of Biology, Kyoto Prefectural University of Medicine, Inamori Memorial Building, 1-5 Shimogamo hangi-cho, Sakyo, Kyoto 606-0823, Japan
| | - Hajime Hosoi
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo, Kyoto 602-8566, Japan
| | - Masafumi Morimoto
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo, Kyoto 602-8566, Japan
| |
Collapse
|
77
|
Magdalon J, Sánchez-Sánchez SM, Griesi-Oliveira K, Sertié AL. Dysfunctional mTORC1 Signaling: A Convergent Mechanism between Syndromic and Nonsyndromic Forms of Autism Spectrum Disorder? Int J Mol Sci 2017; 18:ijms18030659. [PMID: 28335463 PMCID: PMC5372671 DOI: 10.3390/ijms18030659] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 03/13/2017] [Accepted: 03/14/2017] [Indexed: 12/28/2022] Open
Abstract
Whereas autism spectrum disorder (ASD) exhibits striking heterogeneity in genetics and clinical presentation, dysfunction of mechanistic target of rapamycin complex 1 (mTORC1) signaling pathway has been identified as a molecular feature common to several well-characterized syndromes with high prevalence of ASD. Additionally, recent findings have also implicated mTORC1 signaling abnormalities in a subset of nonsyndromic ASD, suggesting that defective mTORC1 pathway may be a potential converging mechanism in ASD pathology across different etiologies. However, the mechanistic evidence for a causal link between aberrant mTORC1 pathway activity and ASD neurobehavioral features varies depending on the ASD form involved. In this review, we first discuss six monogenic ASD-related syndromes, including both classical and potentially novel mTORopathies, highlighting their contribution to our understanding of the neurobiological mechanisms underlying ASD, and then we discuss existing evidence suggesting that aberrant mTORC1 signaling may also play a role in nonsyndromic ASD.
Collapse
Affiliation(s)
- Juliana Magdalon
- Hospital Israelita Albert Einstein, Centro de Pesquisa Experimental, São Paulo 05652-900, Brazil.
| | - Sandra M Sánchez-Sánchez
- Hospital Israelita Albert Einstein, Centro de Pesquisa Experimental, São Paulo 05652-900, Brazil.
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo 05508-090, Brazil.
| | - Karina Griesi-Oliveira
- Hospital Israelita Albert Einstein, Centro de Pesquisa Experimental, São Paulo 05652-900, Brazil.
| | - Andréa L Sertié
- Hospital Israelita Albert Einstein, Centro de Pesquisa Experimental, São Paulo 05652-900, Brazil.
| |
Collapse
|
78
|
Hattori T, Kaji M, Ishii H, Jureepon R, Takarada-Iemata M, Minh Ta H, Manh Le T, Konno A, Hirai H, Shiraishi Y, Ozaki N, Yamamoto Y, Okamoto H, Yokoyama S, Higashida H, Kitao Y, Hori O. CD38 positively regulates postnatal development of astrocytes cell-autonomously and oligodendrocytes non-cell-autonomously. Glia 2017; 65:974-989. [PMID: 28295574 DOI: 10.1002/glia.23139] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 02/20/2017] [Accepted: 02/20/2017] [Indexed: 12/11/2022]
Abstract
Glial development is critical for the function of the central nervous system. CD38 is a multifunctional molecule with ADP-ribosyl cyclase activity. While critical roles of CD38 in the adult brain such as oxytocin release and social behavior have been reported, those in the developing brain remain largely unknown. Here we demonstrate that deletion of Cd38 leads to impaired development of astrocytes and oligodendrocytes in mice. CD38 is highly expressed in the developing brains between postnatal day 14 (P14) and day 28 (P28). In situ hybridization and FACS analysis revealed that CD38 is expressed predominantly in astrocytes in these periods. Analyses of the cortex of Cd38 knockout (Cd38-/- ) mice revealed delayed development of astrocytes and subsequently delayed differentiation of oligodendrocytes (OLs) at postnatal stages. In vitro experiments using primary OL cultures, mixed glial cultures, and astrocytic conditioned medium showed that astrocytic CD38 regulates the development of astrocytes in a cell-autonomous manner and the differentiation of OLs in a non-cell-autonomous manner. Further experiments revealed that connexin43 (Cx43) in astrocytes plays a promotive role for CD38-mediated OL differentiation. Finally, increased levels of NAD+ , caused by CD38 deficiency, are likely to be responsible for the suppression of astrocytic Cx43 expression and OL differentiation. Our data indicate that CD38 is a positive regulator of astrocyte and OL development.
Collapse
Affiliation(s)
- Tsuyoshi Hattori
- Department of Neuroanatomy, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Minoru Kaji
- Department of Neuroanatomy, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Hiroshi Ishii
- Department of Neuroanatomy, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Roboon Jureepon
- Department of Neuroanatomy, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Mika Takarada-Iemata
- Department of Neuroanatomy, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Hieu Minh Ta
- Department of Neuroanatomy, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Thuong Manh Le
- Department of Neuroanatomy, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Ayumu Konno
- Department of Neurophysiology and Neural Repair, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Hirokazu Hirai
- Department of Neurophysiology and Neural Repair, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Yoshitake Shiraishi
- Department of Functional Anatomy, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Noriyuki Ozaki
- Department of Functional Anatomy, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Yasuhiko Yamamoto
- Department of Biochemistry and Molecular Vascular Biology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Hiroshi Okamoto
- Department of Biochemistry and Molecular Vascular Biology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, Japan.,Department of Biochemistry, Tohoku University, Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Shigeru Yokoyama
- Department of Basic Research on Social Recognition and Memory, Research Center for Child Mental Development, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Haruhiro Higashida
- Department of Basic Research on Social Recognition and Memory, Research Center for Child Mental Development, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Yasuko Kitao
- Department of Neuroanatomy, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Osamu Hori
- Department of Neuroanatomy, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, Japan
| |
Collapse
|
79
|
Wu ZQ, Li D, Huang Y, Chen XP, Huang W, Liu CF, Zhao HQ, Xu RX, Cheng M, Schachner M, Ma QH. Caspr Controls the Temporal Specification of Neural Progenitor Cells through Notch Signaling in the Developing Mouse Cerebral Cortex. Cereb Cortex 2017; 27:1369-1385. [PMID: 26740489 DOI: 10.1093/cercor/bhv318] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The generation of layer-specific neurons and astrocytes by radial glial cells during development of the cerebral cortex follows a precise temporal sequence, which is regulated by intrinsic and extrinsic factors. The molecular mechanisms controlling the timely generation of layer-specific neurons and astrocytes remain not fully understood. In this study, we show that the adhesion molecule contactin-associated protein (Caspr), which is involved in the maintenance of the polarized domains of myelinated axons, is essential for the timing of generation of neurons and astrocytes in the developing mouse cerebral cortex. Caspr is expressed by radial glial cells, which are neural progenitor cells that generate both neurons and astrocytes. Absence of Caspr in neural progenitor cells delays the production cortical neurons and induces precocious formation of cortical astrocytes, without affecting the numbers of progenitor cells. At the molecular level, Caspr cooperates with the intracellular domain of Notch to repress transcription of the Notch effector Hes1. Suppression of Notch signaling via a Hes1 shRNA rescues the abnormal neurogenesis and astrogenesis in Caspr-deficient mice. These findings establish Caspr as a novel key regulator that controls the temporal specification of cell fate in radial glial cells of the developing cerebral cortex through Notch signaling.
Collapse
Affiliation(s)
- Zhi-Qiang Wu
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Institute of Neuroscience, Second Affiliated Hospital, Soochow University, Suzhou, Jiangsu Province 215123, China
| | - Di Li
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Institute of Neuroscience, Second Affiliated Hospital, Soochow University, Suzhou, Jiangsu Province 215123, China
| | - Ya Huang
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Institute of Neuroscience, Second Affiliated Hospital, Soochow University, Suzhou, Jiangsu Province 215123, China
| | - Xi-Ping Chen
- Department of Forensic Medicine, Soochow University, Suzhou, Jiangsu Province 215123, China
| | - Wenhui Huang
- Department of Molecular Physiology, Center for Integrative Physiology and Molecular Medicine, University of Saarland, Homburg D-66421, Germany
| | - Chun-Feng Liu
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Institute of Neuroscience, Second Affiliated Hospital, Soochow University, Suzhou, Jiangsu Province 215123, China
| | - He-Qing Zhao
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Institute of Neuroscience, Second Affiliated Hospital, Soochow University, Suzhou, Jiangsu Province 215123, China
| | - Ru-Xiang Xu
- Affiliated Bayi Brain Hospital, Beijing Military Hospital, Southern Medical University, Beijing 100070, China
| | - Mei Cheng
- Binzhou Medical University, Yantai, Shandong Province 264000, China
| | - Melitta Schachner
- Center for Neuroscience, Shantou University Medical College, Shantou, Guangdong Province 515041, China
| | - Quan-Hong Ma
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Institute of Neuroscience, Second Affiliated Hospital, Soochow University, Suzhou, Jiangsu Province 215123, China
| |
Collapse
|
80
|
Wang J, Zou Q, Han R, Li Y, Wang Y. Serum levels of Glial fibrillary acidic protein in Chinese children with autism spectrum disorders. Int J Dev Neurosci 2017; 57:41-45. [PMID: 28088366 DOI: 10.1016/j.ijdevneu.2017.01.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 01/05/2017] [Accepted: 01/06/2017] [Indexed: 01/08/2023] Open
Abstract
OBJECTIVE Glial fibrillary acidic protein (GFAP) has been studied in many neurological diseases. The purpose of this study is to investigate the potential role of GFAP in Chinese children with autism spectrum disorders (ASD) by measuring serum circulating levels of GFAP and comparing them with age and gender-matched typical development children. METHODS A total of one hundred and fifty 2-6 years old Chinese children (75 confirmed autism cases and 75 their age-gender matched typical development children) participated in this study. Serum levels of GFAP were assayed with enzyme-linked immunosorbent assay methods, and severity of ASD was evaluated with the Childhood Autism Rating Scale (CARS) Score. RESULTS The results indicated that the mean serum GFAP level was significantly (P<0.001) higher in autistic children as compared to controls (1.71±0.53ng/ml vs. 0.99±0.25ng/ml). There was a significant positive association between serum GFAP levels and CARS scores (r [Pearson]=0.390, P=0.001). Based on the Receiver operating characteristic (ROC) curve, the optimal cut-off value of serum GFAP levels as an indicator for auxiliary diagnosis of autism was projected to be 1.28ng/ml which yielded a sensitivity of 77.3% and a specificity of 88.4%, the area under the curve was 0.895(95%CI, 0.844-0.947). Further, an increased risk of ASD was associated with GFAP levels >1.28ng/ml (adjusted OR 9.88, 95% CI: 3.32-17.82) in the multivariate logistic analysis model. CONCLUSION The data indicates that serum GFAP levels may be associated with severity of ASD among Chinese children, suggesting the hypothesis that increased serum levels of GFAP could be implicated in the pathophysiology of autism in Chinese children.
Collapse
Affiliation(s)
- Jingwei Wang
- Department of Pediatrics, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, China; Department of Pediatrics, Yantaishan Hospital, Yantai 264001, China
| | - Qiuyan Zou
- Department of Children's Health Prevention, Zhangjiagang Women and Children Health Center, Zhangjiagang 215600, China
| | - Renfeng Han
- Department of Pediatrics, Yantaishan Hospital, Yantai 264001, China
| | - Yupeng Li
- Department of Pediatrics, People's Hospital of Rizhao, Rizhao 276500, China
| | - Yulin Wang
- Department of Pediatrics, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, China.
| |
Collapse
|
81
|
Ecker C, Schmeisser MJ, Loth E, Murphy DG. Neuroanatomy and Neuropathology of Autism Spectrum Disorder in Humans. ADVANCES IN ANATOMY, EMBRYOLOGY, AND CELL BIOLOGY 2017; 224:27-48. [PMID: 28551749 DOI: 10.1007/978-3-319-52498-6_2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Autism spectrum disorder (ASD) is a lifelong heterogeneous neurodevelopmental condition that is associated with differences in brain anatomy and connectivity. Yet, the molecular and cellular mechanisms that underpin the atypical developmental of the brain in ASD remain poorly understood. Here, we review the findings of in vivo neuroimaging studies examining the time course of atypical brain development in ASD and relate the different neurodevelopmental stages that are atypical in ASD to the known neurobiological mechanisms that drive the maturation of the typically developing brain. In particular, we focus on the notion of 'early brain overgrowth' in ASD, which may lead to differences in the formation of the brain's micro- and macro-circuitry. Moreover, we attempt to link the in vivo reports describing differences in brain anatomy and connectivity on the macroscopic level to the increasing number of post-mortem studies examining the neural architecture of the brain in ASD on the microscopic level. In addition, we discuss future directions and outstanding questions in this particular field of research and highlight the need for establishing the link between micro- and macro-pathology in the same set of individuals with ASD based on advances in genetic, molecular and imaging techniques. In combination, these may proof to be invaluable for patient stratification and the development of novel pharmacotherapies in the future.
Collapse
Affiliation(s)
- Christine Ecker
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital, Goethe University, Frankfurt am Main, Germany.
- Department of Forensic and Neurodevelopmental Sciences, Sackler Institute for Translational Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College, London, UK.
| | - Michael J Schmeisser
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany
- Division of Neuroanatomy, Institute of Anatomy, Otto-von-Guericke University, Magdeburg, Germany
- Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Eva Loth
- Department of Forensic and Neurodevelopmental Sciences, Sackler Institute for Translational Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College, London, UK
| | - Declan G Murphy
- Department of Forensic and Neurodevelopmental Sciences, Sackler Institute for Translational Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College, London, UK
| |
Collapse
|
82
|
Tripathi S, Kushwaha R, Mishra J, Gupta MK, Kumar H, Sanyal S, Singh D, Sanyal S, Sahasrabuddhe AA, Kamthan M, Mudiam MKR, Bandyopadhyay S. Docosahexaenoic acid up-regulates both PI3K/AKT-dependent FABP7-PPARγ interaction and MKP3 that enhance GFAP in developing rat brain astrocytes. J Neurochem 2016; 140:96-113. [DOI: 10.1111/jnc.13879] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 09/24/2016] [Accepted: 10/20/2016] [Indexed: 12/31/2022]
Affiliation(s)
- Sachin Tripathi
- Developmental Toxicology Laboratory; Systems Toxicology & Health Risk Assessment Group; CSIR-Indian Institute of Toxicology Research (IITR); Lucknow India
- Amity Institute of Biotechnology; Amity University (Lucknow campus); Lucknow India
| | - Rajesh Kushwaha
- Developmental Toxicology Laboratory; Systems Toxicology & Health Risk Assessment Group; CSIR-Indian Institute of Toxicology Research (IITR); Lucknow India
- Academy of Scientific and Innovative Research (AcSIR); CSIR-IITR campus; Lucknow India
| | - Juhi Mishra
- Developmental Toxicology Laboratory; Systems Toxicology & Health Risk Assessment Group; CSIR-Indian Institute of Toxicology Research (IITR); Lucknow India
- Babu Banarasi Das University; Lucknow India
| | - Manoj Kumar Gupta
- Academy of Scientific and Innovative Research (AcSIR); CSIR-IITR campus; Lucknow India
- Analytical Chemistry Laboratory and Regulatory Toxicology group; CSIR-IITR; Lucknow India
| | - Harish Kumar
- Division of Biochemistry; CSIR-Central Drug Research Institute (CDRI); Lucknow India
| | - Somali Sanyal
- Amity Institute of Biotechnology; Amity University (Lucknow campus); Lucknow India
| | | | - Sabyasachi Sanyal
- Division of Biochemistry; CSIR-Central Drug Research Institute (CDRI); Lucknow India
| | | | - Mohan Kamthan
- Environmental Biotechnology Laboratory; Environmental Toxicology Group; CSIR-IITR; Lucknow India
| | | | - Sanghamitra Bandyopadhyay
- Developmental Toxicology Laboratory; Systems Toxicology & Health Risk Assessment Group; CSIR-Indian Institute of Toxicology Research (IITR); Lucknow India
- Academy of Scientific and Innovative Research (AcSIR); CSIR-IITR campus; Lucknow India
| |
Collapse
|
83
|
Mony TJ, Lee JW, Dreyfus C, DiCicco-Bloom E, Lee HJ. Valproic Acid Exposure during Early Postnatal Gliogenesis Leads to Autistic-like Behaviors in Rats. CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE 2016; 14:338-344. [PMID: 27776385 PMCID: PMC5083944 DOI: 10.9758/cpn.2016.14.4.338] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 05/11/2016] [Accepted: 05/14/2016] [Indexed: 11/18/2022]
Abstract
Objective We reported that postnatal exposure of rats to valproic acid (VPA) stimulated proliferation of glial precursors during cortical gliogenesis. However, there are no reports whether enhanced postnatal gliogenesis affects behaviors related to neuropsychiatric disorders. Methods After VPA treatment during the postnatal day (PND) 2 to PND 4, four behavioral test, such as open field locomotor test, elevated plus maze test, three-chamber social interaction test, and passive avoidance test, were performed at PND 21 or 22. Results VPA treated rats showed significant hyperactive behavior in the open field locomotor test (p<0.05). Moreover, the velocity of movement in the VPA group was increased by 69.5% (p<0.01). In the elevated plus maze test, VPA exposed rats expressed significantly lower percentage of time spent on and of entries into open arms more than the control group (p<0.05). Also, both sociability and social preference indices with strangers in the three-chamber social interaction test were significantly lower in the VPA exposed rats (p<0.05). Conclusion Our results suggest that altered glial cell development is another locus at which pathogenetic factors can operate to contribute to the neurodevelopmental disorder.
Collapse
Affiliation(s)
- Tamanna Jahan Mony
- Department of Pharmacology, Kangwon National University School of Medicine, Chuncheon, Korea
| | - Jae Won Lee
- Department of Pharmacology, Kangwon National University School of Medicine, Chuncheon, Korea.,Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Korea
| | - Cheryl Dreyfus
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Rutgers, the State University of New Jersey, Piscataway, NJ, USA
| | - Emanuel DiCicco-Bloom
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Rutgers, the State University of New Jersey, Piscataway, NJ, USA.,Department of Pediatrics, Rutgers Robert Wood Johnson Medical School, Rutgers, the State University of New Jersey, New Brunswick, NJ, USA
| | - Hee Jae Lee
- Department of Pharmacology, Kangwon National University School of Medicine, Chuncheon, Korea
| |
Collapse
|
84
|
Sexual divergence in microtubule function: the novel intranasal microtubule targeting SKIP normalizes axonal transport and enhances memory. Mol Psychiatry 2016; 21:1467-76. [PMID: 26782054 DOI: 10.1038/mp.2015.208] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2015] [Revised: 11/17/2015] [Accepted: 11/24/2015] [Indexed: 01/21/2023]
Abstract
Activity-dependent neuroprotective protein (ADNP), essential for brain formation, is a frequent autism spectrum disorder (ASD)-mutated gene. ADNP associates with microtubule end-binding proteins (EBs) through its SxIP motif, to regulate dendritic spine formation and brain plasticity. Here, we reveal SKIP, a novel four-amino-acid peptide representing an EB-binding site, as a replacement therapy in an outbred Adnp-deficient mouse model. We discovered, for the first time, axonal transport deficits in Adnp(+/-) mice (measured by manganese-enhanced magnetic resonance imaging), with significant male-female differences. RNA sequencing evaluations showed major age, sex and genotype differences. Function enrichment and focus on major gene expression changes further implicated channel/transporter function and the cytoskeleton. In particular, a significant maturation change (1 month-five months) was observed in beta1 tubulin (Tubb1) mRNA, only in Adnp(+/+) males, and sex-dependent increase in calcium channel mRNA (Cacna1e) in Adnp(+/+) males compared with females. At the protein level, the Adnp(+/-) mice exhibited impaired hippocampal expression of the calcium channel (voltage-dependent calcium channel, Cacnb1) as well as other key ASD-linked genes including the serotonin transporter (Slc6a4), and the autophagy regulator, BECN1 (Beclin1), in a sex-dependent manner. Intranasal SKIP treatment normalized social memory in 8- to 9-month-old Adnp(+/-)-treated mice to placebo-control levels, while protecting axonal transport and ameliorating changes in ASD-like gene expression. The control, all d-amino analog D-SKIP, did not mimic SKIP activity. SKIP presents a novel prototype for potential ASD drug development, a prevalent unmet medical need.
Collapse
|
85
|
Kern JK, Geier DA, Sykes LK, Haley BE, Geier MR. The relationship between mercury and autism: A comprehensive review and discussion. J Trace Elem Med Biol 2016; 37:8-24. [PMID: 27473827 DOI: 10.1016/j.jtemb.2016.06.002] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 05/17/2016] [Accepted: 06/01/2016] [Indexed: 12/28/2022]
Abstract
The brain pathology in autism spectrum disorders (ASD) indicates marked and ongoing inflammatory reactivity with concomitant neuronal damage. These findings are suggestive of neuronal insult as a result of external factors, rather than some type of developmental mishap. Various xenobiotics have been suggested as possible causes of this pathology. In a recent review, the top ten environmental compounds suspected of causing autism and learning disabilities were listed and they included: lead, methyl-mercury, polychorinated biphenyls, organophosphate pesticides, organochlorine pesticides, endocrine disruptors, automotive exhaust, polycyclic aromatic hydrocarbons, polybrominated diphenyl ethers, and perfluorinated compounds. This current review, however, will focus specifically on mercury exposure and ASD by conducting a comprehensive literature search of original studies in humans that examine the potential relationship between mercury and ASD, categorizing, summarizing, and discussing the published research that addresses this topic. This review found 91 studies that examine the potential relationship between mercury and ASD from 1999 to February 2016. Of these studies, the vast majority (74%) suggest that mercury is a risk factor for ASD, revealing both direct and indirect effects. The preponderance of the evidence indicates that mercury exposure is causal and/or contributory in ASD.
Collapse
Affiliation(s)
- Janet K Kern
- Institute of Chronic Illnesses, Inc., 14 Redgate Court, Silver Spring, MD, 20905 USA; Council for Nutritional and Environmental Medicine, Mo i Rana, Norway; CoMeD, Inc., 14 Redgate Court, Silver Spring, MD, 20905 USA.
| | - David A Geier
- Institute of Chronic Illnesses, Inc., 14 Redgate Court, Silver Spring, MD, 20905 USA; CoMeD, Inc., 14 Redgate Court, Silver Spring, MD, 20905 USA
| | - Lisa K Sykes
- CoMeD, Inc., 14 Redgate Court, Silver Spring, MD, 20905 USA
| | - Boyd E Haley
- University of Kentucky, 410 Administration Drive, Lexington, KY, 40506 USA
| | - Mark R Geier
- Institute of Chronic Illnesses, Inc., 14 Redgate Court, Silver Spring, MD, 20905 USA; CoMeD, Inc., 14 Redgate Court, Silver Spring, MD, 20905 USA
| |
Collapse
|
86
|
Advancing the understanding of autism disease mechanisms through genetics. Nat Med 2016; 22:345-61. [PMID: 27050589 DOI: 10.1038/nm.4071] [Citation(s) in RCA: 530] [Impact Index Per Article: 66.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 02/26/2016] [Indexed: 12/11/2022]
Abstract
Progress in understanding the genetic etiology of autism spectrum disorders (ASD) has fueled remarkable advances in our understanding of its potential neurobiological mechanisms. Yet, at the same time, these findings highlight extraordinary causal diversity and complexity at many levels ranging from molecules to circuits and emphasize the gaps in our current knowledge. Here we review current understanding of the genetic architecture of ASD and integrate genetic evidence, neuropathology and studies in model systems with how they inform mechanistic models of ASD pathophysiology. Despite the challenges, these advances provide a solid foundation for the development of rational, targeted molecular therapies.
Collapse
|
87
|
Menassa DA, Sloan C, Chance SA. Primary olfactory cortex in autism and epilepsy: increased glial cells in autism. Brain Pathol 2016; 27:437-448. [PMID: 27409070 DOI: 10.1111/bpa.12415] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 07/12/2016] [Indexed: 01/06/2023] Open
Abstract
Autism Spectrum Disorder is characterized by sensory anomalies including impaired olfactory identification. Between 5 and 46 percent of individuals with autism have a clinical diagnosis of epilepsy. Primary olfactory cortex (piriform cortex) is central to olfactory identification and is an epileptogenic structure. Cytoarchitectural changes in olfactory cortex may underlie olfactory differences seen in autism. Primary olfactory cortex was sampled from 17 post-mortem autism cases with and without epilepsy, 11 epilepsy cases without autism and 11 typically developed cases. Stereological and neuropathological methods were used to quantify glial, pyramidal and non-pyramidal cell densities in layers of the piriform as well as identify pathological differences in this area and its neighbouring region, the olfactory tubercle. We found increased layer II glial cell densities in autism with and without epilepsy, which were negatively correlated with age and positively correlated with levels of corpora amylacea in layer I. These changes were also associated with greater symptom severity and did not extend to the olfactory tubercle. Glial cell organization may follow an altered trajectory of development with age in autism. The findings are consistent with other studies implicating increased glial cells in the autism brain. Altered cytoarchitecture may contribute to sensory deficits observed in affected individuals. This study provides evidence that autism is linked to alterations in the cytoarchitectural structure that underlies primary sensory processes and is not restricted to heteromodal ("higher") cognitive centers.
Collapse
Affiliation(s)
- David A Menassa
- Nuffield Department of Clinical Neurosciences, West Wing, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, United Kingdom
| | - Carolyn Sloan
- Nuffield Department of Clinical Neurosciences, West Wing, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, United Kingdom
| | - Steven A Chance
- Nuffield Department of Clinical Neurosciences, West Wing, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, United Kingdom
| |
Collapse
|
88
|
Cetin I, Tezdig I, Tarakcioglu MC, Kadak MT, Demirel OF, Ozer OF. Serum levels of glial fibrillary acidic protein and Nogo-A in children with autism spectrum disorders. Biomarkers 2016; 21:614-8. [DOI: 10.3109/1354750x.2016.1171901] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Ihsan Cetin
- Department of Nutrition and Dietetics, School of Health, Batman University, Batman, Turkey
| | - Ihsan Tezdig
- Department of Chemistry, Institute of Science, Batman University, Batman, Turkey
| | - Mahmut Cem Tarakcioglu
- Clinic of Child and Adolescent Psychiatry, Bakirköy Dr Sadi Konuk Training and Educatin Hospital, Istanbul University, İstanbul, Turkey
| | - Muhammed Tayyib Kadak
- Department of Child and Adolescent Psychiatry, Cerrahpaşa School of Medicine, Istanbul University, İstanbul, Turkey
| | - Omer Faruk Demirel
- Department of Psychiatry, Cerrahpaşa School of Medicine, Istanbul University, İstanbul, Turkey
| | - Omer Faruk Ozer
- Department of Biochemistry, BezmiAlem Vakif University, İstanbul, Turkey
| |
Collapse
|
89
|
Dallérac G, Rouach N. Astrocytes as new targets to improve cognitive functions. Prog Neurobiol 2016; 144:48-67. [PMID: 26969413 DOI: 10.1016/j.pneurobio.2016.01.003] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 01/07/2016] [Accepted: 01/24/2016] [Indexed: 01/09/2023]
Abstract
Astrocytes are now viewed as key elements of brain wiring as well as neuronal communication. Indeed, they not only bridge the gap between metabolic supplies by blood vessels and neurons, but also allow fine control of neurotransmission by providing appropriate signaling molecules and insulation through a tight enwrapping of synapses. Recognition that astroglia is essential to neuronal communication is nevertheless fairly recent and the large body of evidence dissecting such role has focused on the synaptic level by identifying neuro- and gliotransmitters uptaken and released at synaptic or extrasynaptic sites. Yet, more integrated research deciphering the impact of astroglial functions on neuronal network activity have led to the reasonable assumption that the role of astrocytes in supervising synaptic activity translates in influencing neuronal processing and cognitive functions. Several investigations using recent genetic tools now support this notion by showing that inactivating or boosting astroglial function directly affects cognitive abilities. Accordingly, brain diseases resulting in impaired cognitive functions have seen their physiopathological mechanisms revisited in light of this primary protagonist of brain processing. We here provide a review of the current knowledge on the role of astrocytes in cognition and in several brain diseases including neurodegenerative disorders, psychiatric illnesses, as well as other conditions such as epilepsy. Potential astroglial therapeutic targets are also discussed.
Collapse
Affiliation(s)
- Glenn Dallérac
- Neuroglial Interactions in Cerebral Physiopathology, Center for Interdisciplinary Research in Biology, Collège de France, Centre National de la Recherche Scientifique UMR 7241, Institut National de la Santé et de la Recherche Médicale U1050, Labex Memolife, PSL Research University, Paris, France.
| | - Nathalie Rouach
- Neuroglial Interactions in Cerebral Physiopathology, Center for Interdisciplinary Research in Biology, Collège de France, Centre National de la Recherche Scientifique UMR 7241, Institut National de la Santé et de la Recherche Médicale U1050, Labex Memolife, PSL Research University, Paris, France.
| |
Collapse
|
90
|
Banerjee A, Luong JA, Ho A, Saib AO, Ploski JE. Overexpression of Homer1a in the basal and lateral amygdala impairs fear conditioning and induces an autism-like social impairment. Mol Autism 2016; 7:16. [PMID: 26929812 PMCID: PMC4770673 DOI: 10.1186/s13229-016-0077-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 02/02/2016] [Indexed: 12/20/2022] Open
Abstract
Background Autism spectrum disorders (ASDs) represent a heterogeneous group of disorders with a wide range of behavioral impairments including social and communication deficits. Apart from these core symptoms, a significant number of ASD individuals display higher levels of anxiety, and some studies indicate that a subset of ASD individuals have a reduced ability to be fear conditioned. Deciphering the molecular basis of ASD has been considerably challenging and it currently remains poorly understood. In this study we examined the molecular basis of autism-like impairments in an environmentally induced animal model of ASD, where pregnant rats are exposed to the known teratogen, valproic acid (VPA), on day 12.5 of gestation and the subsequent progeny exhibit ASD-like symptoms. We focused our analysis on the basal and lateral nucleus of the amygdala (BLA), a region of the brain found to be associated with ASD pathology. Methods We performed whole genome gene expression analysis on the BLA using DNA microarrays to examine differences in gene expression within the amygdala of VPA-exposed animals. We validated one VPA-dysregulated candidate gene (Homer1a) using both quantitative PCR (qRT-PCR) and western blot. Finally, we overexpressed Homer1a within the basal and lateral amygdala of naïve animals utilizing adeno-associated viruses (AAV) and subsequently examined these animals in a battery of behavioral tests associated with ASD, including auditory fear conditioning, social interaction and open field. Results Our microarray data indicated that Homer1a was one of the genes which exhibited a significant upregulation within the amygdala. We observed an increase in Homer1a messenger RNA (mRNA) and protein in multiple cohorts of VPA-exposed animals indicating that dysregulation of Homer1a levels might underlie some of the symptoms exhibited by VPA-exposed animals. To test this hypothesis, we overexpressed Homer1a within BLA neurons utilizing a viral-mediated approach and found that overexpression of Homer1a impaired auditory fear conditioning and reduced social interaction, while having no influence on open-field behavior. Conclusions This study indicates that dysregulation of amygdala Homer1a might contribute to some autism-like symptoms induced by VPA exposure. These findings are interesting in part because Homer1a influences the functioning of Shank3, metabotropic glutamate receptors (mGluR5), and Homer1, and these proteins have previously been associated with ASD, indicating that these differing models of ASD may have a similar molecular basis. Electronic supplementary material The online version of this article (doi:10.1186/s13229-016-0077-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Anwesha Banerjee
- Department of Cell Biology, Emory University, 615 Michael St. WBRB #415, Atlanta, GA 30322 USA
| | - Jonathan A Luong
- School of Behavioral and Brain Sciences, University of Texas at Dallas, 800 West Campbell road, Richardson, TX 75080 USA
| | - Anthony Ho
- School of Behavioral and Brain Sciences, University of Texas at Dallas, 800 West Campbell road, Richardson, TX 75080 USA
| | - Aeshah O Saib
- School of Behavioral and Brain Sciences, University of Texas at Dallas, 800 West Campbell road, Richardson, TX 75080 USA
| | - Jonathan E Ploski
- School of Behavioral and Brain Sciences, University of Texas at Dallas, 800 West Campbell road, Richardson, TX 75080 USA
| |
Collapse
|
91
|
Skene NG, Grant SGN. Identification of Vulnerable Cell Types in Major Brain Disorders Using Single Cell Transcriptomes and Expression Weighted Cell Type Enrichment. Front Neurosci 2016; 10:16. [PMID: 26858593 PMCID: PMC4730103 DOI: 10.3389/fnins.2016.00016] [Citation(s) in RCA: 180] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 01/12/2016] [Indexed: 11/13/2022] Open
Abstract
The cell types that trigger the primary pathology in many brain diseases remain largely unknown. One route to understanding the primary pathological cell type for a particular disease is to identify the cells expressing susceptibility genes. Although this is straightforward for monogenic conditions where the causative mutation may alter expression of a cell type specific marker, methods are required for the common polygenic disorders. We developed the Expression Weighted Cell Type Enrichment (EWCE) method that uses single cell transcriptomes to generate the probability distribution associated with a gene list having an average level of expression within a cell type. Following validation, we applied EWCE to human genetic data from cases of epilepsy, Schizophrenia, Autism, Intellectual Disability, Alzheimer's disease, Multiple Sclerosis and anxiety disorders. Genetic susceptibility primarily affected microglia in Alzheimer's and Multiple Sclerosis; was shared between interneurons and pyramidal neurons in Autism and Schizophrenia; while intellectual disabilities and epilepsy were attributable to a range of cell-types, with the strongest enrichment in interneurons. We hypothesized that the primary cell type pathology could trigger secondary changes in other cell types and these could be detected by applying EWCE to transcriptome data from diseased tissue. In Autism, Schizophrenia and Alzheimer's disease we find evidence of pathological changes in all of the major brain cell types. These findings give novel insight into the cellular origins and progression in common brain disorders. The methods can be applied to any tissue and disorder and have applications in validating mouse models.
Collapse
Affiliation(s)
- Nathan G Skene
- Centre for Clinical Brain Sciences, Edinburgh University Edinburgh, UK
| | - Seth G N Grant
- Centre for Clinical Brain Sciences, Edinburgh University Edinburgh, UK
| |
Collapse
|
92
|
Skene NG, Grant SGN. Identification of Vulnerable Cell Types in Major Brain Disorders Using Single Cell Transcriptomes and Expression Weighted Cell Type Enrichment. Front Neurosci 2016; 10:16. [PMID: 26858593 DOI: 10.3389/fnins.2016.00016/bibtex] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 01/12/2016] [Indexed: 05/23/2023] Open
Abstract
The cell types that trigger the primary pathology in many brain diseases remain largely unknown. One route to understanding the primary pathological cell type for a particular disease is to identify the cells expressing susceptibility genes. Although this is straightforward for monogenic conditions where the causative mutation may alter expression of a cell type specific marker, methods are required for the common polygenic disorders. We developed the Expression Weighted Cell Type Enrichment (EWCE) method that uses single cell transcriptomes to generate the probability distribution associated with a gene list having an average level of expression within a cell type. Following validation, we applied EWCE to human genetic data from cases of epilepsy, Schizophrenia, Autism, Intellectual Disability, Alzheimer's disease, Multiple Sclerosis and anxiety disorders. Genetic susceptibility primarily affected microglia in Alzheimer's and Multiple Sclerosis; was shared between interneurons and pyramidal neurons in Autism and Schizophrenia; while intellectual disabilities and epilepsy were attributable to a range of cell-types, with the strongest enrichment in interneurons. We hypothesized that the primary cell type pathology could trigger secondary changes in other cell types and these could be detected by applying EWCE to transcriptome data from diseased tissue. In Autism, Schizophrenia and Alzheimer's disease we find evidence of pathological changes in all of the major brain cell types. These findings give novel insight into the cellular origins and progression in common brain disorders. The methods can be applied to any tissue and disorder and have applications in validating mouse models.
Collapse
Affiliation(s)
- Nathan G Skene
- Centre for Clinical Brain Sciences, Edinburgh University Edinburgh, UK
| | - Seth G N Grant
- Centre for Clinical Brain Sciences, Edinburgh University Edinburgh, UK
| |
Collapse
|
93
|
Sudo G, Kagawa T, Kokubu Y, Inazawa J, Taga T. Increase in GFAP-positive astrocytes in histone demethylase GASC1/KDM4C/JMJD2C hypomorphic mutant mice. Genes Cells 2016; 21:218-25. [PMID: 26805559 DOI: 10.1111/gtc.12331] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2015] [Accepted: 11/27/2015] [Indexed: 01/06/2023]
Abstract
GASC1, also known as KDM4C/JMJD2C, is a histone demethylase for histone H3 lysine 9 (H3K9) and H3K36. In this study, we observed an increase of GFAP-positive astrocytes in the brain of Gasc1 hypomorphic mutant mice at 2-3 months of age, but not at postnatal day 14 and day 30 by immunohistochemistry. Increases of GFAP-positive astrocytes were widely observed in the forebrain and prominent in such regions as cerebral cortex, caudate putamen, amygdala and diencephalon, but not obvious in hippocampus. Taken together with our observations to be published elsewhere that Gasc1 hypomorphic mutant mice exhibit abnormal behaviors including hyperactivity, persistence and many types of learning and memory deficits and abnormal synaptic functions such as prolonged long-term potentiation, the increase in GFAP-positive astrocytes may help understand their phenotypes, because astrocytes are known to affect synaptic plasticity.
Collapse
Affiliation(s)
- Genki Sudo
- Department of Stem Cell regulation, Medical Research Institute, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Tetsushi Kagawa
- Department of Stem Cell regulation, Medical Research Institute, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Yasuhiro Kokubu
- Department of Stem Cell regulation, Medical Research Institute, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Johji Inazawa
- Department of Molecular Cytogenetics, Medical Research Institute, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Tetsuya Taga
- Department of Stem Cell regulation, Medical Research Institute, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| |
Collapse
|
94
|
Kern JK, Geier DA, Sykes LK, Geier MR. Relevance of Neuroinflammation and Encephalitis in Autism. Front Cell Neurosci 2016; 9:519. [PMID: 26834565 PMCID: PMC4717322 DOI: 10.3389/fncel.2015.00519] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2015] [Accepted: 12/24/2015] [Indexed: 12/12/2022] Open
Abstract
In recent years, many studies indicate that children with an autism spectrum disorder (ASD) diagnosis have brain pathology suggestive of ongoing neuroinflammation or encephalitis in different regions of their brains. Evidence of neuroinflammation or encephalitis in ASD includes: microglial and astrocytic activation, a unique and elevated proinflammatory profile of cytokines, and aberrant expression of nuclear factor kappa-light-chain-enhancer of activated B cells. A conservative estimate based on the research suggests that at least 69% of individuals with an ASD diagnosis have microglial activation or neuroinflammation. Encephalitis, which is defined as inflammation of the brain, is medical diagnosis code G04.90 in the International Classification of Disease, 10th revision; however, children with an ASD diagnosis are not generally assessed for a possible medical diagnosis of encephalitis. This is unfortunate because if a child with ASD has neuroinflammation, then treating the underlying brain inflammation could lead to improved outcomes. The purpose of this review of the literature is to examine the evidence of neuroinflammation/encephalitis in those with an ASD diagnosis and to address how a medical diagnosis of encephalitis, when appropriate, could benefit these children by driving more immediate and targeted treatments.
Collapse
Affiliation(s)
- Janet K. Kern
- Institute of Chronic Illnesses, Inc., Silver SpringMD, USA
| | - David A. Geier
- Institute of Chronic Illnesses, Inc., Silver SpringMD, USA
| | | | - Mark R. Geier
- Institute of Chronic Illnesses, Inc., Silver SpringMD, USA
| |
Collapse
|
95
|
Developmental neurotoxicity of inhaled ambient ultrafine particle air pollution: Parallels with neuropathological and behavioral features of autism and other neurodevelopmental disorders. Neurotoxicology 2015; 59:140-154. [PMID: 26721665 DOI: 10.1016/j.neuro.2015.12.014] [Citation(s) in RCA: 153] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Revised: 12/18/2015] [Accepted: 12/18/2015] [Indexed: 12/25/2022]
Abstract
Accumulating evidence from both human and animal studies show that brain is a target of air pollution. Multiple epidemiological studies have now linked components of air pollution to diagnosis of autism spectrum disorder (ASD), a linkage with plausibility based on the shared mechanisms of inflammation. Additional plausibility appears to be provided by findings from our studies in mice of exposures from postnatal day (PND) 4-7 and 10-13 (human 3rd trimester equivalent), to concentrated ambient ultrafine (UFP) particles, considered the most reactive component of air pollution, at levels consistent with high traffic areas of major U.S. cities and thus highly relevant to human exposures. These exposures, occurring during a period of marked neuro- and gliogenesis, unexpectedly produced a pattern of developmental neurotoxicity notably similar to multiple hypothesized mechanistic underpinnings of ASD, including its greater impact in males. UFP exposures induced inflammation/microglial activation, reductions in size of the corpus callosum (CC) and associated hypomyelination, aberrant white matter development and/or structural integrity with ventriculomegaly (VM), elevated glutamate and excitatory/inhibitory imbalance, increased amygdala astrocytic activation, and repetitive and impulsive behaviors. Collectively, these findings suggest the human 3rd trimester equivalent as a period of potential vulnerability to neurodevelopmental toxicity to UFP, particularly in males, and point to the possibility that UFP air pollution exposure during periods of rapid neuro- and gliogenesis may be a risk factor not only for ASD, but also for other neurodevelopmental disorders that share features with ASD, such as schizophrenia, attention deficit disorder, and periventricular leukomalacia.
Collapse
|
96
|
Chen JA, Peñagarikano O, Belgard TG, Swarup V, Geschwind DH. The emerging picture of autism spectrum disorder: genetics and pathology. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2015; 10:111-44. [PMID: 25621659 DOI: 10.1146/annurev-pathol-012414-040405] [Citation(s) in RCA: 182] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Autism spectrum disorder (ASD) is defined by impaired social interaction and communication accompanied by stereotyped behaviors and restricted interests. Although ASD is common, its genetic and clinical features are highly heterogeneous. A number of recent breakthroughs have dramatically advanced our understanding of ASD from the standpoint of human genetics and neuropathology. These studies highlight the period of fetal development and the processes of chromatin structure, synaptic function, and neuron-glial signaling. The initial efforts to systematically integrate findings of multiple levels of genomic data and studies of mouse models have yielded new clues regarding ASD pathophysiology. This early work points to an emerging convergence of disease mechanisms in this complex and etiologically heterogeneous disorder.
Collapse
|
97
|
Elevated GFAP Protein in Anterior Cingulate Cortical White Matter in Males With Autism Spectrum Disorder. Autism Res 2015; 8:649-57. [DOI: 10.1002/aur.1480] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Accepted: 02/28/2015] [Indexed: 01/01/2023]
|
98
|
Vela G, Stark P, Socha M, Sauer AK, Hagmeyer S, Grabrucker AM. Zinc in gut-brain interaction in autism and neurological disorders. Neural Plast 2015; 2015:972791. [PMID: 25878905 PMCID: PMC4386645 DOI: 10.1155/2015/972791] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 03/05/2015] [Indexed: 12/27/2022] Open
Abstract
A growing amount of research indicates that abnormalities in the gastrointestinal (GI) system during development might be a common factor in multiple neurological disorders and might be responsible for some of the shared comorbidities seen among these diseases. For example, many patients with Autism Spectrum Disorder (ASD) have symptoms associated with GI disorders. Maternal zinc status may be an important factor given the multifaceted effect of zinc on gut development and morphology in the offspring. Zinc status influences and is influenced by multiple factors and an interdependence of prenatal and early life stress, immune system abnormalities, impaired GI functions, and zinc deficiency can be hypothesized. In line with this, systemic inflammatory events and prenatal stress have been reported to increase the risk for ASD. Thus, here, we will review the current literature on the role of zinc in gut formation, a possible link between gut and brain development in ASD and other neurological disorders with shared comorbidities, and tie in possible effects on the immune system. Based on these data, we present a novel model outlining how alterations in the maternal zinc status might pathologically impact the offspring leading to impairments in brain functions later in life.
Collapse
Affiliation(s)
- Guillermo Vela
- Zinpro Corporation, Eden Prairie, MN 55344, USA
- Autismo ABP, 64639 Monterrey, NL, Mexico
| | - Peter Stark
- Zinpro Corporation, Eden Prairie, MN 55344, USA
| | | | - Ann Katrin Sauer
- WG Molecular Analysis of Synaptopathies, Neurology Department, Neurocenter of Ulm University, 89081 Ulm, Germany
| | - Simone Hagmeyer
- WG Molecular Analysis of Synaptopathies, Neurology Department, Neurocenter of Ulm University, 89081 Ulm, Germany
| | - Andreas M. Grabrucker
- WG Molecular Analysis of Synaptopathies, Neurology Department, Neurocenter of Ulm University, 89081 Ulm, Germany
- Institute for Anatomy and Cell Biology, Ulm University, 89081 Ulm, Germany
| |
Collapse
|
99
|
Schitine C, Nogaroli L, Costa MR, Hedin-Pereira C. Astrocyte heterogeneity in the brain: from development to disease. Front Cell Neurosci 2015; 9:76. [PMID: 25852472 PMCID: PMC4367182 DOI: 10.3389/fncel.2015.00076] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 02/20/2015] [Indexed: 12/31/2022] Open
Abstract
In the last decades, astrocytes have risen from passive supporters of neuronal activity to central players in brain function and cognition. Likewise, the heterogeneity of astrocytes starts to become recognized in contrast to the homogeneous population previously predicted. In this review, we focused on astrocyte heterogeneity in terms of their morphological, protein expression and functional aspects, and debate in a historical perspective the diversity encountered in glial progenitors and how they may reflect mature astrocyte heterogeneity. We discussed data that show that different progenitors may have unsuspected roles in developmental processes. We have approached the functions of astrocyte subpopulations on the onset of psychiatric and neurological diseases.
Collapse
Affiliation(s)
- Clarissa Schitine
- Cellular Neuroanatomy Laboratory, Program in Neurobiology, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro Brazil
| | - Luciana Nogaroli
- Cellular Neuroanatomy Laboratory, Program in Neurobiology, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro Brazil
| | - Marcos R Costa
- Laboratory of Cellular Neurobiology, Brain Institute, Federal University of Rio Grande do Norte, Natal Brazil
| | - Cecilia Hedin-Pereira
- Cellular Neuroanatomy Laboratory, Program in Neurobiology, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro Brazil ; Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro Brazil
| |
Collapse
|
100
|
Kazim SF, Cardenas-Aguayo MDC, Arif M, Blanchard J, Fayyaz F, Grundke-Iqbal I, Iqbal K. Sera from children with autism induce autistic features which can be rescued with a CNTF small peptide mimetic in rats. PLoS One 2015; 10:e0118627. [PMID: 25769033 PMCID: PMC4359103 DOI: 10.1371/journal.pone.0118627] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 01/21/2015] [Indexed: 12/29/2022] Open
Abstract
Autism is a neurodevelopmental disorder characterized clinically by impairments in social interaction and verbal and non-verbal communication skills as well as restricted interests and repetitive behavior. It has been hypothesized that altered brain environment including an imbalance in neurotrophic support during early development contributes to the pathophysiology of autism. Here we report that sera from children with autism which exhibited abnormal levels of various neurotrophic factors induced cell death and oxidative stress in mouse primary cultured cortical neurons. The effects of sera from autistic children were rescued by pre-treatment with a ciliary neurotrophic factor (CNTF) small peptide mimetic, Peptide 6 (P6), which was previously shown to exert its neuroprotective effect by modulating CNTF/JAK/STAT pathway and LIF signaling and by enhancing brain derived neurotrophic factor (BDNF) expression. Similar neurotoxic effects and neuroinflammation were observed in young Wistar rats injected intracerebroventricularly with autism sera within hours after birth. The autism sera injected rats demonstrated developmental delay and deficits in social communication, interaction, and novelty. Both the neurobiological changes and the behavioral autistic phenotype were ameliorated by P6 treatment. These findings implicate the involvement of neurotrophic imbalance during early brain development in the pathophysiology of autism and a proof of principle of P6 as a potential therapeutic strategy for autism.
Collapse
Affiliation(s)
- Syed Faraz Kazim
- Inge Grundke-Iqbal Research Floor, Department of Neurochemistry, New York State Institute for Basic Research in Developmental Disabilities (NYSIBR), Staten Island, New York, United States of America
- Neural and Behavioral Science Graduate Program, State University of New York (SUNY) Downstate Medical Center, Brooklyn, New York, United States of America
- SUNY Downstate/NYSIBR Center for Developmental Neuroscience (CDN), Staten Island, New York, United States of America
| | - Maria del Carmen Cardenas-Aguayo
- Inge Grundke-Iqbal Research Floor, Department of Neurochemistry, New York State Institute for Basic Research in Developmental Disabilities (NYSIBR), Staten Island, New York, United States of America
| | - Mohammad Arif
- Inge Grundke-Iqbal Research Floor, Department of Neurochemistry, New York State Institute for Basic Research in Developmental Disabilities (NYSIBR), Staten Island, New York, United States of America
| | - Julie Blanchard
- Inge Grundke-Iqbal Research Floor, Department of Neurochemistry, New York State Institute for Basic Research in Developmental Disabilities (NYSIBR), Staten Island, New York, United States of America
| | - Fatima Fayyaz
- Inge Grundke-Iqbal Research Floor, Department of Neurochemistry, New York State Institute for Basic Research in Developmental Disabilities (NYSIBR), Staten Island, New York, United States of America
| | - Inge Grundke-Iqbal
- Inge Grundke-Iqbal Research Floor, Department of Neurochemistry, New York State Institute for Basic Research in Developmental Disabilities (NYSIBR), Staten Island, New York, United States of America
| | - Khalid Iqbal
- Inge Grundke-Iqbal Research Floor, Department of Neurochemistry, New York State Institute for Basic Research in Developmental Disabilities (NYSIBR), Staten Island, New York, United States of America
- SUNY Downstate/NYSIBR Center for Developmental Neuroscience (CDN), Staten Island, New York, United States of America
- * E-mail:
| |
Collapse
|