1
|
Wegiel J, Chadman K, London E, Wisniewski T, Wegiel J. Contribution of the serotonergic system to developmental brain abnormalities in autism spectrum disorder. Autism Res 2024; 17:1300-1321. [PMID: 38500252 PMCID: PMC11272444 DOI: 10.1002/aur.3123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 02/28/2024] [Indexed: 03/20/2024]
Abstract
This review highlights a key role of the serotonergic system in brain development and in distortions of normal brain development in early stages of fetal life resulting in cascades of abnormalities, including defects of neurogenesis, neuronal migration, neuronal growth, differentiation, and arborization, as well as defective neuronal circuit formation in the cortex, subcortical structures, brainstem, and cerebellum of autistic subjects. In autism, defects in regulation of neuronal growth are the most frequent and ubiquitous developmental changes associated with impaired neuron differentiation, smaller size, distorted shape, loss of spatial orientation, and distortion of cortex organization. Common developmental defects of the brain in autism include multiregional focal dysplastic changes contributing to local neuronal circuit distortion, epileptogenic activity, and epilepsy. There is a discrepancy between more than 500 reports demonstrating the contribution of the serotonergic system to autism's behavioral anomalies, highlighted by lack of studies of autistic subjects' brainstem raphe nuclei, the center of brain serotonergic innervation, and of the contribution of the serotonergic system to the diagnostic features of autism spectrum disorder (ASD). Discovery of severe fetal brainstem auditory system neuronal deficits and other anomalies leading to a spectrum of hearing deficits contributing to a cascade of behavioral alterations, including deficits of social and verbal communication in individuals with autism, is another argument to intensify postmortem studies of the type and topography of, and the severity of developmental defects in raphe nuclei and their contribution to abnormal brain development and to the broad spectrum of functional deficits and comorbid conditions in ASD.
Collapse
Affiliation(s)
- Jarek Wegiel
- Department of Developmental Neurobiology, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, New York, USA
| | - Kathryn Chadman
- Department of Developmental Neurobiology, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, New York, USA
| | - Eric London
- Department of Psychology, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, New York, USA
| | - Thomas Wisniewski
- Department of Developmental Neurobiology, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, New York, USA
- Center for Cognitive Neurology, Department of Neurology, Pathology and Psychiatry, NYU Grossman School of Medicine, New York, New York, USA
| | - Jerzy Wegiel
- Department of Developmental Neurobiology, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, New York, USA
| |
Collapse
|
2
|
Adiguzel E, Bozkurt NM, Unal G. Independent and combined effects of astaxanthin and omega-3 on behavioral deficits and molecular changes in a prenatal valproic acid model of autism in rats. Nutr Neurosci 2024; 27:590-606. [PMID: 37534957 DOI: 10.1080/1028415x.2023.2239575] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/04/2023]
Abstract
Objectives: Autism is a devastating neurodevelopmental disorder and recent studies showed that omega-3 or astaxanthin might reduce autistic symptoms due to their anti-inflammatory properties. Therefore, we investigated the effects of omega-3 and astaxanthin on the VPA-induced autism model of rats.Material and Methods: Female Wistar albino pups (n = 40) were grouped as control, autistic, astaxanthin (2 mg/kg), omega-3 (200 mg/kg), and astaxanthin (2 mg/kg)+omega-3 (200 mg/kg). All groups except the control were prenatally exposed to VPA. Astaxanthin and omega-3 were orally administered from the postnatal day 41 to 68 and behavioral tests were performed between day 69 and 73. The rats were decapitated 24 h after the behavioral tests and hippocampal and prefrontal cytokines and 5-HT levels were analyzed by ELISA.Results: VPA rats have increased grooming behavior while decreased sociability (SI), social preference index (SPI), discrimination index (DI), and prepulse inhibition (PPI) compared to control. Additionally, IL-1β, IL-6, TNF-α, and IFN-γ levels increased while IL-10 and 5-HT levels decreased in both brain regions. Astaxanthin treatment raised SI, SPI, DI, PPI, and prefrontal IL-10 levels. It also raised 5-HT levels and decreased IL-6 levels in both brain regions. Omega-3 and astaxanthin + omega-3 increased the SI, SPI, DI, and PPI and decreased grooming behavior. Moreover, they increased IL-10 and 5-HT levels whereas decreased IL-1β, IL-6, TNF-α, IFN-γ levels in both brain regions.Conclusions: Our results showed that VPA administration mimicked the behavioral and molecular changes of autism in rats. Single and combined administration of astaxanthin and omega-3 improved the autistic-like behavioral and molecular changes in the VPA model of rats.
Collapse
Affiliation(s)
- Emre Adiguzel
- Faculty of Health Sciences, Department of Nutrition and Dietetics, Karamanoğlu Mehmetbey University, Karaman, Türkiye
| | - Nuh Mehmet Bozkurt
- Faculty of Pharmacy, Department of Pharmacology, Erciyes University, Kayseri, Türkiye
- Experimental Research and Application Center (DEKAM), Brain Research Unit, Erciyes University, Kayseri, Türkiye
- e-Neuro Lab, Drug Application and Research Center (ERFARMA), Erciyes University, Kayseri, Türkiye
| | - Gokhan Unal
- Faculty of Pharmacy, Department of Pharmacology, Erciyes University, Kayseri, Türkiye
- Experimental Research and Application Center (DEKAM), Brain Research Unit, Erciyes University, Kayseri, Türkiye
- e-Neuro Lab, Drug Application and Research Center (ERFARMA), Erciyes University, Kayseri, Türkiye
| |
Collapse
|
3
|
Jannati A, Oberman LM, Rotenberg A, Pascual-Leone A. Assessing the mechanisms of brain plasticity by transcranial magnetic stimulation. Neuropsychopharmacology 2023; 48:191-208. [PMID: 36198876 PMCID: PMC9700722 DOI: 10.1038/s41386-022-01453-8] [Citation(s) in RCA: 54] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 09/01/2022] [Accepted: 09/02/2022] [Indexed: 11/10/2022]
Abstract
Transcranial magnetic stimulation (TMS) is a non-invasive technique for focal brain stimulation based on electromagnetic induction where a fluctuating magnetic field induces a small intracranial electric current in the brain. For more than 35 years, TMS has shown promise in the diagnosis and treatment of neurological and psychiatric disorders in adults. In this review, we provide a brief introduction to the TMS technique with a focus on repetitive TMS (rTMS) protocols, particularly theta-burst stimulation (TBS), and relevant rTMS-derived metrics of brain plasticity. We then discuss the TMS-EEG technique, the use of neuronavigation in TMS, the neural substrate of TBS measures of plasticity, the inter- and intraindividual variability of those measures, effects of age and genetic factors on TBS aftereffects, and then summarize alterations of TMS-TBS measures of plasticity in major neurological and psychiatric disorders including autism spectrum disorder, schizophrenia, depression, traumatic brain injury, Alzheimer's disease, and diabetes. Finally, we discuss the translational studies of TMS-TBS measures of plasticity and their therapeutic implications.
Collapse
Affiliation(s)
- Ali Jannati
- Neuromodulation Program, Division of Epilepsy and Clinical Neurophysiology, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
- F. M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
- Berenson-Allen Center for Noninvasive Brain Stimulation, Division of Cognitive Neurology, Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| | - Lindsay M Oberman
- Center for Neuroscience and Regenerative Medicine, Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Alexander Rotenberg
- Neuromodulation Program, Division of Epilepsy and Clinical Neurophysiology, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- F. M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Berenson-Allen Center for Noninvasive Brain Stimulation, Division of Cognitive Neurology, Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Alvaro Pascual-Leone
- Department of Neurology, Harvard Medical School, Boston, MA, USA.
- Hinda and Arthur Marcus Institute for Aging Research and Deanna and Sidney Wolk Center for Memory Health, Hebrew SeniorLife, Boston, MA, USA.
- Guttmann Brain Health Institute, Institut Guttmann, Barcelona, Spain.
| |
Collapse
|
4
|
NMR-Based Metabolomics of Rat Hippocampus, Serum, and Urine in Two Models of Autism. Mol Neurobiol 2022; 59:5452-5475. [PMID: 35715683 DOI: 10.1007/s12035-022-02912-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 06/03/2022] [Indexed: 10/18/2022]
Abstract
Autism spectrum disorders (ASDs) are increasingly diagnosed as developmental disabilities of unclear etiology related to genetic, epigenetic, or environmental factors. The diagnosis of ASD in children is based on the recognition of typical behavioral symptoms, while no reliable biomarkers are available. Rats in whom ASD-like symptoms are due to maternal administration of the teratogenic drugs valproate or thalidomide on critical day 11 of pregnancy are widely used models in autism research. The present studies, aimed at detecting changes in the levels of hydrophilic and hydrophobic metabolites, were carried out on 1-month-old rats belonging to the abovementioned two ASD models and on a control group. Analysis of both hydrophilic and hydrophobic metabolite levels gives a broader view of possible mechanisms involved in the pathogenesis of autism. Hippocampal proton magnetic resonance (MRS) spectroscopy and ex vivo nuclear magnetic resonance (NMR) analysis of serum and urine samples were used. The results were analyzed using advanced statistical tests. Both the results of our present MRS studies of the hippocampus and of the NMR studies of body fluids in both ASD models, particularly from the THAL model, appeared to be consistent with previously published NMR results of hippocampal homogenates and data from the literature on autistic children. We detected symptoms of disturbances in neurotransmitter metabolism, energy deficit, and oxidative stress, as well as intestinal malfunction, which shed light on the pathogenesis of ASD and could be used for diagnostic purposes. These results confirm the usefulness of the noninvasive techniques used in ASD studies.
Collapse
|
5
|
Mehra S, Ul Ahsan A, Seth E, Chopra M. Critical Evaluation of Valproic Acid-Induced Rodent Models of Autism: Current and Future Perspectives. J Mol Neurosci 2022; 72:1259-1273. [DOI: 10.1007/s12031-022-02033-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 05/21/2022] [Indexed: 11/29/2022]
|
6
|
Therapeutic Effects of a Novel Form of Biotin on Propionic Acid-Induced Autistic Features in Rats. Nutrients 2022; 14:nu14061280. [PMID: 35334937 PMCID: PMC8955994 DOI: 10.3390/nu14061280] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 03/15/2022] [Accepted: 03/16/2022] [Indexed: 12/04/2022] Open
Abstract
Magnesium biotinate (MgB) is a novel biotin complex with superior absorption and anti-inflammatory effects in the brain than D-Biotin. This study aimed to investigate the impact of different doses of MgB on social behavior deficits, learning and memory alteration, and inflammatory markers in propionic acid (PPA)-exposed rats. In this case, 35 Wistar rats (3 weeks old) were distributed into five groups: 1, Control; 2, PPA treated group; 3, PPA+MgBI (10 mg, HED); 4, PPA+MgBII (100 mg, HED); 5, PPA+MgBIII (500 mg, HED). PPA was given subcutaneously at 500 mg/kg/day for five days, followed by MgB for two weeks. PPA-exposed rats showed poor sociability and a high level of anxiety-like behaviors and cognitive impairments (p < 0.001). In a dose-dependent manner, behavioral and learning-memory disorders were significantly improved by MgB supplementation (p < 0.05). PPA decreased both the numbers and the sizes of Purkinje cells in the cerebellum. However, MgB administration increased the sizes and the densities of Purkinje cells. MgB improved the brain and serum Mg, biotin, serotonin, and dopamine concentrations, as well as antioxidant enzymes (CAT, SOD, GPx, and GSH) (p < 0.05). In addition, MgB treatment significantly regulated the neurotoxicity-related cytokines and neurotransmission-related markers. For instance, MgB significantly decreased the expression level of TNF-α, IL-6, IL-17, CCL-3, CCL-5, and CXCL-16 in the brain, compared to the control group (p < 0.05). These data demonstrate that MgB may ameliorate dysfunctions in social behavior, learning and memory and reduce the oxidative stress and inflammation indexes of the brain in a rat model.
Collapse
|
7
|
Kuo HY, Liu FC. Pathophysiological Studies of Monoaminergic Neurotransmission Systems in Valproic Acid-Induced Model of Autism Spectrum Disorder. Biomedicines 2022; 10:560. [PMID: 35327362 PMCID: PMC8945169 DOI: 10.3390/biomedicines10030560] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 02/21/2022] [Accepted: 02/22/2022] [Indexed: 01/27/2023] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder with complex etiology. The core syndromes of ASD are deficits in social communication and self-restricted interests and repetitive behaviors. Social communication relies on the proper integration of sensory and motor functions, which is tightly interwoven with the limbic function of reward, motivation, and emotion in the brain. Monoamine neurotransmitters, including serotonin, dopamine, and norepinephrine, are key players in the modulation of neuronal activity. Owing to their broad distribution, the monoamine neurotransmitter systems are well suited to modulate social communication by coordinating sensory, motor, and limbic systems in different brain regions. The complex and diverse functions of monoamine neurotransmission thus render themselves as primary targets of pathophysiological investigation of the etiology of ASD. Clinical studies have reported that children with maternal exposure to valproic acid (VPA) have an increased risk of developing ASD. Extensive animal studies have confirmed that maternal treatments of VPA include ASD-like phenotypes, including impaired social communication and repetitive behavior. Here, given that ASD is a neurodevelopmental disorder, we begin with an overview of the neural development of monoaminergic systems with their neurochemical properties in the brain. We then review and discuss the evidence of human clinical and animal model studies of ASD with a focus on the VPA-induced pathophysiology of monoamine neurotransmitter systems. We also review the potential interactions of microbiota and monoamine neurotransmitter systems in ASD pathophysiology. Widespread and complex changes in monoamine neurotransmitters are detected in the brains of human patients with ASD and validated in animal models. ASD animal models are not only essential to the characterization of pathogenic mechanisms, but also provide a preclinical platform for developing therapeutic approaches to ASD.
Collapse
Affiliation(s)
- Hsiao-Ying Kuo
- Institute of Anatomy and Cell Biology, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Fu-Chin Liu
- Institute of Neuroscience, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| |
Collapse
|
8
|
Jannati A, Ryan MA, Block G, Kayarian FB, Oberman LM, Rotenberg A, Pascual-Leone A. Modulation of motor cortical excitability by continuous theta-burst stimulation in adults with autism spectrum disorder. Clin Neurophysiol 2021; 132:1647-1662. [PMID: 34030059 PMCID: PMC8197744 DOI: 10.1016/j.clinph.2021.03.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 02/23/2021] [Accepted: 03/15/2021] [Indexed: 11/21/2022]
Abstract
OBJECTIVE To test whether change in motor evoked potential (ΔMEP) induced by continuous theta-burst stimulation (cTBS) of motor cortex (M1) distinguishes adults with autism spectrum disorder (ASD) from neurotypicals, and to explore the contribution of two common polymorphisms related to neuroplasticity. METHODS 44 adult neurotypical (NT) participants (age 21-65, 34 males) and 19 adults with ASD (age 21-58, 17 males) prospectively underwent M1 cTBS. Their data were combined with previously obtained results from 35 NT and 35 ASD adults. RESULTS ΔMEP at 15 minutes post-cTBS (T15) was a significant predictor of diagnosis (p = 0.04) in the present sample (n=63). T15 remained a significant predictor in a larger sample (n=91) and when partially imputed based on T10-T20 from a yet-greater sample (N=133). T15 also remained a significant predictor of diagnosis among brain-derived neurotrophic factor (BDNF) Met+ and apolipoprotein E (APOE) ε4- subjects (p's < 0.05), but not among Met- or ε4+ subjects (p's > 0.19). CONCLUSIONS ΔMEP at T15 post-cTBS is a significant biomarker for adults with ASD, and its utility is modulated by BDNF and APOE polymorphisms. SIGNIFICANCE M1 cTBS response is a physiologic biomarker for adults with ASD in large samples, and controlling for BDNF and APOE polymorphisms can improve its diagnostic utility.
Collapse
Affiliation(s)
- Ali Jannati
- Berenson-Allen Center for Noninvasive Brain Stimulation and Division of Cognitive Neurology, Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, USA; Neuromodulation Program and Division of Epilepsy and Clinical Neurophysiology, Department of Neurology, Boston Children's Hospital, Boston, MA, USA; Department of Neurology, Harvard Medical School, Boston, MA, USA; F.M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Mary A Ryan
- Berenson-Allen Center for Noninvasive Brain Stimulation and Division of Cognitive Neurology, Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, USA; Neuromodulation Program and Division of Epilepsy and Clinical Neurophysiology, Department of Neurology, Boston Children's Hospital, Boston, MA, USA
| | - Gabrielle Block
- Berenson-Allen Center for Noninvasive Brain Stimulation and Division of Cognitive Neurology, Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, USA; Neuromodulation Program and Division of Epilepsy and Clinical Neurophysiology, Department of Neurology, Boston Children's Hospital, Boston, MA, USA
| | - Fae B Kayarian
- Berenson-Allen Center for Noninvasive Brain Stimulation and Division of Cognitive Neurology, Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Lindsay M Oberman
- Berenson-Allen Center for Noninvasive Brain Stimulation and Division of Cognitive Neurology, Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, USA; Neuromodulation Program and Division of Epilepsy and Clinical Neurophysiology, Department of Neurology, Boston Children's Hospital, Boston, MA, USA; Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - Alexander Rotenberg
- Berenson-Allen Center for Noninvasive Brain Stimulation and Division of Cognitive Neurology, Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, USA; Neuromodulation Program and Division of Epilepsy and Clinical Neurophysiology, Department of Neurology, Boston Children's Hospital, Boston, MA, USA; Department of Neurology, Harvard Medical School, Boston, MA, USA; F.M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Alvaro Pascual-Leone
- Department of Neurology, Harvard Medical School, Boston, MA, USA; Hinda and Arthur Marcus Institute for Aging Research and Deanna and Sidney Wolk Center for Memory Health, Hebrew SeniorLife, Boston, MA, USA; Guttman Brain Health Institute, Institut Guttman de Neurorehabilitació, Universitat Autónoma de Barcelona, Badalona, Barcelona, Spain.
| |
Collapse
|
9
|
Manduca A, Carbone E, Schiavi S, Cacchione C, Buzzelli V, Campolongo P, Trezza V. The neurochemistry of social reward during development: What have we learned from rodent models? J Neurochem 2021; 157:1408-1435. [PMID: 33569830 DOI: 10.1111/jnc.15321] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 02/02/2021] [Accepted: 02/07/2021] [Indexed: 12/14/2022]
Abstract
Social rewards are fundamental to survival and overall health. Several studies suggest that adequate social stimuli during early life are critical for developing appropriate socioemotional and cognitive skills, whereas adverse social experiences negatively affect the proper development of brain and behavior, by increasing the susceptibility to develop neuropsychiatric conditions. Therefore, a better understanding of the neural mechanisms underlying social interactions, and their rewarding components in particular, is an important challenge of current neuroscience research. In this context, preclinical research has a crucial role: Animal models allow to investigate the neurobiological aspects of social reward in order to shed light on possible neurochemical alterations causing aberrant social reward processing in neuropsychiatric diseases, and they allow to test the validity and safety of innovative therapeutic strategies. Here, we discuss preclinical research that has investigated the rewarding properties of two forms of social interaction that occur in different phases of the lifespan of mammals, that is, mother-infant interaction and social interactions with peers, by focusing on the main neurotransmitter systems mediating their rewarding components. Together, the research performed so far helped to elucidate the mechanisms of social reward and its psychobiological components throughout development, thus increasing our understanding of the neurobiological substrates sustaining social functioning in health conditions and social dysfunction in major psychiatric disorders.
Collapse
Affiliation(s)
- Antonia Manduca
- Department of Science, Section of Biomedical Sciences and Technologies, Roma Tre University, Rome, Italy.,Neuroendocrinology, Metabolism and Neuropharmacology Unit, IRCSS Fondazione Santa Lucia, Rome, Italy
| | - Emilia Carbone
- Department of Science, Section of Biomedical Sciences and Technologies, Roma Tre University, Rome, Italy
| | - Sara Schiavi
- Department of Science, Section of Biomedical Sciences and Technologies, Roma Tre University, Rome, Italy
| | - Claudia Cacchione
- Department of Science, Section of Biomedical Sciences and Technologies, Roma Tre University, Rome, Italy
| | - Valeria Buzzelli
- Department of Science, Section of Biomedical Sciences and Technologies, Roma Tre University, Rome, Italy.,Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Patrizia Campolongo
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy.,Neurobiology of Behavior Laboratory, IRCSS Fondazione Santa Lucia, Rome, Italy
| | - Viviana Trezza
- Department of Science, Section of Biomedical Sciences and Technologies, Roma Tre University, Rome, Italy
| |
Collapse
|
10
|
Loss CM, Teodoro L, Rodrigues GD, Moreira LR, Peres FF, Zuardi AW, Crippa JA, Hallak JEC, Abílio VC. Is Cannabidiol During Neurodevelopment a Promising Therapy for Schizophrenia and Autism Spectrum Disorders? Front Pharmacol 2021; 11:635763. [PMID: 33613289 PMCID: PMC7890086 DOI: 10.3389/fphar.2020.635763] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 12/24/2020] [Indexed: 01/22/2023] Open
Abstract
Schizophrenia and autism spectrum disorders (ASD) are psychiatric neurodevelopmental disorders that cause high levels of functional disabilities. Also, the currently available therapies for these disorders are limited. Therefore, the search for treatments that could be beneficial for the altered course of the neurodevelopment associated with these disorders is paramount. Preclinical and clinical evidence points to cannabidiol (CBD) as a promising strategy. In this review, we discuss clinical and preclinical studies on schizophrenia and ASD investigating the behavioral, molecular, and functional effects of chronic treatment with CBD (and with cannabidivarin for ASD) during neurodevelopment. In summary, the results point to CBD's beneficial potential for the progression of these disorders supporting further investigations to strengthen its use.
Collapse
Affiliation(s)
- Cássio Morais Loss
- Molecular and Behavioral Neuroscience Laboratory, Departamento de Farmacologia, Universidade Federal de São Paulo, São Paulo, Brazil.,National Institute for Translational Medicine (INCT-TM), National Council for Scientific and Technological Development (CNPq/CAPES/FAPESP), Ribeirão Preto, Brazil
| | - Lucas Teodoro
- Molecular and Behavioral Neuroscience Laboratory, Departamento de Farmacologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Gabriela Doná Rodrigues
- Molecular and Behavioral Neuroscience Laboratory, Departamento de Farmacologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Lucas Roberto Moreira
- Molecular and Behavioral Neuroscience Laboratory, Departamento de Farmacologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Fernanda Fiel Peres
- Molecular and Behavioral Neuroscience Laboratory, Departamento de Farmacologia, Universidade Federal de São Paulo, São Paulo, Brazil.,National Institute for Translational Medicine (INCT-TM), National Council for Scientific and Technological Development (CNPq/CAPES/FAPESP), Ribeirão Preto, Brazil
| | - Antonio Waldo Zuardi
- National Institute for Translational Medicine (INCT-TM), National Council for Scientific and Technological Development (CNPq/CAPES/FAPESP), Ribeirão Preto, Brazil.,Department of Neuroscience and Behavior, Ribeirão Preto Medical School, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - José Alexandre Crippa
- National Institute for Translational Medicine (INCT-TM), National Council for Scientific and Technological Development (CNPq/CAPES/FAPESP), Ribeirão Preto, Brazil.,Department of Neuroscience and Behavior, Ribeirão Preto Medical School, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Jaime Eduardo Cecilio Hallak
- National Institute for Translational Medicine (INCT-TM), National Council for Scientific and Technological Development (CNPq/CAPES/FAPESP), Ribeirão Preto, Brazil.,Department of Neuroscience and Behavior, Ribeirão Preto Medical School, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Vanessa Costhek Abílio
- Molecular and Behavioral Neuroscience Laboratory, Departamento de Farmacologia, Universidade Federal de São Paulo, São Paulo, Brazil.,National Institute for Translational Medicine (INCT-TM), National Council for Scientific and Technological Development (CNPq/CAPES/FAPESP), Ribeirão Preto, Brazil
| |
Collapse
|
11
|
Zürcher NR, Walsh EC, Phillips RD, Cernasov PM, Tseng CEJ, Dharanikota A, Smith E, Li Z, Kinard JL, Bizzell JC, Greene RK, Dillon D, Pizzagalli DA, Izquierdo-Garcia D, Truong K, Lalush D, Hooker JM, Dichter GS. A simultaneous [ 11C]raclopride positron emission tomography and functional magnetic resonance imaging investigation of striatal dopamine binding in autism. Transl Psychiatry 2021; 11:33. [PMID: 33431841 PMCID: PMC7801430 DOI: 10.1038/s41398-020-01170-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 11/30/2020] [Accepted: 12/07/2020] [Indexed: 01/10/2023] Open
Abstract
The social motivation hypothesis of autism posits that autism spectrum disorder (ASD) is characterized by impaired motivation to seek out social experience early in life that interferes with the development of social functioning. This framework suggests that impaired mesolimbic dopamine function underlies compromised responses to social rewards in ASD. Although this hypothesis is supported by functional magnetic resonance imaging (fMRI) studies, no molecular imaging study has evaluated striatal dopamine functioning in response to rewards in ASD. Here, we examined striatal functioning during monetary incentive processing in ASD and controls using simultaneous positron emission tomography (PET) and fMRI. Using a bolus + infusion protocol with the D2/D3 dopamine receptor antagonist [11C]raclopride, voxel-wise binding potential (BPND) was compared between groups (controls = 12, ASD = 10) in the striatum. Striatal clusters showing significant between-group BPND differences were used as seeds in whole-brain fMRI general functional connectivity analyses. Relative to controls, the ASD group demonstrated decreased phasic dopamine release to incentives in the bilateral putamen and left caudate, as well as increased functional connectivity between a PET-derived right putamen seed and the precuneus and insula. Within the ASD group, decreased phasic dopamine release in the putamen was related to poorer theory-of-mind skills. Our findings that ASD is characterized by impaired striatal phasic dopamine release to incentives provide support for the social motivation hypothesis of autism. PET-fMRI may be a suitable tool to evaluate novel ASD therapeutics targeting the striatal dopamine system.
Collapse
Affiliation(s)
- Nicole R. Zürcher
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129 USA
| | - Erin C. Walsh
- grid.10698.360000000122483208Department of Psychiatry, University of North Carolina-Chapel Hill, Chapel Hill, NC 27514 USA
| | - Rachel D. Phillips
- grid.10698.360000000122483208Department of Psychology and Neuroscience, University of North Carolina-Chapel Hill, Chapel Hill, NC 27514 USA
| | - Paul M. Cernasov
- grid.10698.360000000122483208Department of Psychology and Neuroscience, University of North Carolina-Chapel Hill, Chapel Hill, NC 27514 USA
| | - Chieh-En J. Tseng
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129 USA
| | - Ayarah Dharanikota
- grid.10698.360000000122483208Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC USA
| | - Eric Smith
- grid.10698.360000000122483208UNC-Chapel Hill Department of Radiology and Biomedical Research Imaging Center (BRIC), Chapel Hill, NC 27514 USA
| | - Zibo Li
- grid.10698.360000000122483208UNC-Chapel Hill Department of Radiology and Biomedical Research Imaging Center (BRIC), Chapel Hill, NC 27514 USA
| | - Jessica L. Kinard
- grid.10698.360000000122483208Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, Chapel Hill, NC 27510 USA
| | - Joshua C. Bizzell
- grid.10698.360000000122483208Department of Psychiatry, University of North Carolina-Chapel Hill, Chapel Hill, NC 27514 USA
| | - Rachel K. Greene
- grid.10698.360000000122483208Department of Psychology and Neuroscience, University of North Carolina-Chapel Hill, Chapel Hill, NC 27514 USA
| | - Daniel Dillon
- grid.240206.20000 0000 8795 072XCenter for Depression, Anxiety and Stress Research, McLean Hospital, Belmont, MA USA
| | - Diego A. Pizzagalli
- grid.240206.20000 0000 8795 072XCenter for Depression, Anxiety and Stress Research, McLean Hospital, Belmont, MA USA
| | - David Izquierdo-Garcia
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129 USA
| | - Kinh Truong
- grid.10698.360000000122483208Department of Biostatistics, University of North Carolina-Chapel Hill, Chapel Hill, NC 27514 USA
| | - David Lalush
- grid.10698.360000000122483208Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC USA
| | - Jacob M. Hooker
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129 USA
| | - Gabriel S. Dichter
- grid.10698.360000000122483208Department of Psychiatry, University of North Carolina-Chapel Hill, Chapel Hill, NC 27514 USA ,grid.10698.360000000122483208Department of Psychology and Neuroscience, University of North Carolina-Chapel Hill, Chapel Hill, NC 27514 USA ,grid.10698.360000000122483208Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, Chapel Hill, NC 27510 USA
| |
Collapse
|
12
|
Al‐Salem HS, Al‐Yousef HM, Ashour AE, Ahmed AF, Amina M, Issa IS, Bhat RS. Antioxidant and hepatorenal protective effects of bee pollen fractions against propionic acid-induced autistic feature in rats. Food Sci Nutr 2020; 8:5114-5127. [PMID: 32994972 PMCID: PMC7500755 DOI: 10.1002/fsn3.1813] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 07/14/2020] [Accepted: 07/18/2020] [Indexed: 01/08/2023] Open
Abstract
In the brain, propionic acid (PA) can cross cell membranes and accumulate within cells, leading to intracellular acidification, which may alter neurotransmitter release (NT), communication between neurons, and behavior. Such elevation in levels of PA constitutes a neurodevelopmental metabolic disorder called propionic acidemia, which could clinically manifest as autism. The purpose of this study was to investigate the protective effects of different fractions of bee pollen (BP) on PA-induced autism in rats, and to evaluate their effects on the expression of liver and renal biomarkers. Groups of rats received treatments of different fractions of BP at a dose of 250 mg/kg of body weight/day for a period of 1 month. Normal control group I and group II were orally administered with phosphate-buffered saline and propionic acid, respectively, for 3 days. BP contains various health-promoting phenolic components. Different fractions of BP administered pre- and post-treatment with PA showed significant reduction in the levels of liver and renal biomarkers (p < .05). Also, a significant enhancement in the levels of glutathione S-transferase (GST), catalase CAT), and ascorbic acid (VIT C) was observed. Supplementation with BP significantly reduced biochemical changes in the liver, kidneys, and brain of rats with PA-induced toxicity. It exhibited protective effects against oxidative damage and reactive oxygen species produced by PA-induced adverse reactions in rats. Taken together, our study shows that BP possesses protective effects in PA-induced liver and kidney damage.
Collapse
Affiliation(s)
- Huda S. Al‐Salem
- Pharmaceutical Chemistry DepartmentCollege of PharmacyKing Saud UniversityRiyadhSaudi Arabia
| | - Hanan M. Al‐Yousef
- Pharmacognosy DepartmentCollege of PharmacyKing Saud UniversityRiyadhSaudi Arabia
| | - Abdelkader E. Ashour
- Department of Basic Medical SciencesKulliyyah of MedicineInternational Islamic University MalaysiaKuantanMalaysia
| | - Atallah F. Ahmed
- Pharmacognosy DepartmentCollege of PharmacyKing Saud UniversityRiyadhSaudi Arabia
- Department of PharmacognosyFaculty of PharmacyMansoura UniversityMansouraEgypt
| | - Musarat Amina
- Pharmacognosy DepartmentCollege of PharmacyKing Saud UniversityRiyadhSaudi Arabia
| | - Iman S. Issa
- Pharmaceutical Chemistry DepartmentCollege of PharmacyKing Saud UniversityRiyadhSaudi Arabia
| | - Ramesa Shafi Bhat
- Biochemistry DepartmentScience CollegeKing Saud UniversityRiyadhSaudi Arabia
| |
Collapse
|
13
|
Hippocampal Metabolite Profiles in Two Rat Models of Autism: NMR-Based Metabolomics Studies. Mol Neurobiol 2020; 57:3089-3105. [PMID: 32468248 PMCID: PMC7320041 DOI: 10.1007/s12035-020-01935-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 05/13/2020] [Indexed: 12/22/2022]
Abstract
Autism spectrum disorders (ASDs) are increasingly being diagnosed. Hypotheses link ASD to genetic, epigenetic, or environmental factors. The role of oxidative stress and the imbalance between excitatory and inhibitory neurotransmission in the pathogenesis of ASD has been suggested. Rats in which ASD symptoms are induced by valproate (VPA) or thalidomide (THAL) application in utero are useful models in ASD studies. Our study investigated whether rats in ASD models show changes in metabolite levels in the brain consistent with the hypothetical pathomechanisms of ASD. Female rats were fed one dose of 800 mg/kg VPA or 500 mg/kg THAL orally on the 11th day of gestation, and 1-month offspring were used for the experiments. Metabolic profiles from proton nuclear magnetic resonance spectroscopy of hydrophilic and hydrophobic extracts of rat hippocampi were subjected to OPLS-DA statistical analysis. Large differences between both models in the content of several metabolites in the rat hippocampus were noticed. The following metabolic pathways were identified as being disturbed in both ASD models: steroid hormone biosynthesis; fatty acid biosynthesis; the synthesis and degradation of ketone bodies; glycerophospholipid metabolism; cholesterol metabolism; purine metabolism; arginine and proline metabolism; valine, leucine, and isoleucine biosynthesis and degradation. These results indicate disorders of energy metabolism, altered structure of cell membranes, changes in neurotransmission, and the induction of oxidative stress in the hippocampus. Our data, consistent with hypotheses of ASD pathomechanisms, may be useful in future ASD studies, especially for the interpretation of the results of metabolomics analysis of body fluids in rat ASD models.
Collapse
|
14
|
Changes in the Expression of SNAP-25 Protein in the Brain of Juvenile Rats in Two Models of Autism. J Mol Neurosci 2020; 70:1313-1320. [PMID: 32367505 PMCID: PMC7399687 DOI: 10.1007/s12031-020-01543-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 04/13/2020] [Indexed: 12/13/2022]
Abstract
The results of genetic studies suggest a possible role for SNAP-25 polymorphism in the development of autism spectrum disorders (ASDs); however, there are no data available on whether changes in SNAP-25 expression also affect animals in rodent models of ASD. The aim of the present study was to explore this issue. The studies included 1-month-old rats representing valproic acid (VPA)- and thalidomide (THAL)-induced models of autism. Their mothers received single doses of VPA (800 mg/kg) or THAL (500 mg/kg) per os on the 11th day of gestation. SNAP-25 protein content in the cerebellum, hippocampus, and frontal lobe was determined using Western blotting, while changes of mRNA levels of Snap25 gene were determined using real-time polymerase chain reaction. Compared to controls, SNAP-25 content was decreased by approximately 35% in all brain structures tested, in both males and females, exclusively in the VPA group. In contrast to this, Snap25 expression, studied in males, was increased in the hippocampus and cerebellum in both, VPA- and THAL-treated rats. We discuss the compliance of these results with the hypothesized role of SNAP-25 in the pathophysiology of ASD and the adequacy of the experimental models used.
Collapse
|
15
|
Tsugiyama LE, Ida-Eto M, Ohkawara T, Noro Y, Narita M. Altered neuronal activity in the auditory brainstem following sound stimulation in thalidomide-induced autism model rats. Congenit Anom (Kyoto) 2020; 60:82-86. [PMID: 31373055 DOI: 10.1111/cga.12353] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 07/04/2019] [Accepted: 07/22/2019] [Indexed: 11/28/2022]
Abstract
Auditory hypersensitivity in autism is frequently observed in clinics. Dysfunction in the auditory brainstem has been suspected. We have established autism model rats using prenatal thalidomide exposure. Here we investigated whether abnormal response occurs in the brainstem following sound stimulus in autism model rats. Autism model rats were prepared by prenatal exposure to thalidomide on embryonic days 9 and 10 in pregnant rats. Then, the animals were exposed to 16-kHz pure tone auditory stimulus and c-Fos immunostaining was performed to examine the neuronal activity on postnatal day 49 to 51. Following sound stimulus, increased number of c-Fos-positive neurons was observed in the medial nucleus of the trapezoid body of autism model rats compared with the control rats. These results suggest that prenatal thalidomide might cause altered processing of auditory stimulus, leading to the characteristics of auditory hypersensitivity in autism.
Collapse
Affiliation(s)
- Lucila E Tsugiyama
- Department of Developmental and Regenerative Medicine, Mie University, Graduate School of Medicine, Mie, Japan
| | - Michiru Ida-Eto
- Department of Developmental and Regenerative Medicine, Mie University, Graduate School of Medicine, Mie, Japan
| | - Takeshi Ohkawara
- Department of Developmental and Regenerative Medicine, Mie University, Graduate School of Medicine, Mie, Japan
| | - Yuichi Noro
- Department of Physics Engineering, Mie University, Graduate School of Engineering, Mie, Japan
| | - Masaaki Narita
- Department of Developmental and Regenerative Medicine, Mie University, Graduate School of Medicine, Mie, Japan
| |
Collapse
|
16
|
Wang J, Zheng B, Zhou D, Xing J, Li H, Li J, Zhang Z, Zhang B, Li P. Supplementation of Diet With Different n-3/n-6 PUFA Ratios Ameliorates Autistic Behavior, Reduces Serotonin, and Improves Intestinal Barrier Impairments in a Valproic Acid Rat Model of Autism. Front Psychiatry 2020; 11:552345. [PMID: 33033482 PMCID: PMC7509584 DOI: 10.3389/fpsyt.2020.552345] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 08/24/2020] [Indexed: 12/29/2022] Open
Abstract
The implication of different dietary n-3/n-6 polyunsaturated fatty acids (PUFAs) ratios has been investigated in some neurodevelopmental disorders (including autism and depression). However, the mechanisms underlying the effects of different PUFAs ratios on the autism are still poorly understood. In the present study, a valproic acid (VPA) rat model of autism was used to study the effects of diet with different n-3/n-6 PUFA ratios on the autism, and the underlying mechanisms explored. Our results showed that rats with prenatal administration of VPA took less response time to sniff three odorants in the olfactory habituation/dishabituation tests, had lower frequency of pinning and following patterns, and had decreased hippocampal 5-hydroxytryptamine (5-HT), increased serum 5-HT and downregulated expression of tight junction protein (occludin and claudin-1) in the colon. However, supplementation of n-3/n-6 PUFAs (1:5) in the VPA treated rats ameliorated the autistic behaviors, increased hippocampal 5-HT and tight junction expression in the colon, and decreased serum 5-HT. In conclusion, dietary supplementation of n-3/n-6 PUFAs (1:5) significantly improves VPA-induced autism-like behaviors in rats, which may be, at least partially, related to the increased hippocampal 5-HT. Furthermore, this diet can increase the expression of tight junction proteins to improve intestinal barrier impairment.
Collapse
Affiliation(s)
- Jinpeng Wang
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, China
| | - Baihong Zheng
- Department of Pediatrics, The Second Hospital of Jilin University, Changchun, China
| | - Dan Zhou
- Department of Pediatrics, The Second Hospital of Jilin University, Changchun, China
| | - Jie Xing
- Department of Developmental Pediatrics, The Second Hospital of Jilin University, Changchun, China
| | - Honghua Li
- Department of Developmental and Behavioral Pediatrics, The First Hospital of Jilin University, Changchun, China
| | - Jiayu Li
- Department of Physiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Zehui Zhang
- Department of Developmental and Behavioral Pediatrics, The First Hospital of Jilin University, Changchun, China
| | - Beilin Zhang
- Department of Physiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Ping Li
- Department of Developmental Pediatrics, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
17
|
Schiavi S, Iezzi D, Manduca A, Leone S, Melancia F, Carbone C, Petrella M, Mannaioni G, Masi A, Trezza V. Reward-Related Behavioral, Neurochemical and Electrophysiological Changes in a Rat Model of Autism Based on Prenatal Exposure to Valproic Acid. Front Cell Neurosci 2019; 13:479. [PMID: 31708750 PMCID: PMC6824319 DOI: 10.3389/fncel.2019.00479] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 10/10/2019] [Indexed: 12/17/2022] Open
Abstract
Prenatal exposure to the antiepileptic drug valproic acid (VPA) induces autism spectrum disorder (ASD) in humans and autistic-like behaviors in rodents, which makes it a good model to study the neural underpinnings of ASD. Rats prenatally exposed to VPA show profound deficits in the social domain. The altered social behavior displayed by VPA-exposed rats may be due to either a deficit in social reward processing or to a more general inability to properly understand and respond to social signals. To address this issue, we performed behavioral, electrophysiological and neurochemical experiments and tested the involvement of the brain reward system in the social dysfunctions displayed by rats prenatally exposed to VPA (500 mg/kg). We found that, compared to control animals, VPA-exposed rats showed reduced play responsiveness together with impaired sociability in the three-chamber test and altered social discrimination abilities. In addition, VPA-exposed rats showed altered expression of dopamine receptors together with inherent hyperexcitability of medium spiny neurons (MSNs) in the nucleus accumbens (NAc). However, when tested for socially-induced conditioned place preference, locomotor response to amphetamine and sucrose preference, control and VPA-exposed rats performed similarly, indicating normal responses to social, drug and food rewards. On the basis of the results obtained, we hypothesize that social dysfunctions displayed by VPA-exposed rats are more likely caused by alterations in cognitive aspects of the social interaction, such as the interpretation and reciprocation of social stimuli and/or the ability to adjust the social behavior of the individual to the changing circumstances in the social and physical environment, rather than to inability to enjoy the pleasurable aspects of the social interaction. The observed neurochemical and electrophysiological alterations in the NAc may contribute to the inability of VPA-exposed rats to process and respond to social cues, or, alternatively, represent a compensatory mechanism towards VPA-induced neurodevelopmental insults.
Collapse
Affiliation(s)
- Sara Schiavi
- Department of Science, Section of Biomedical Sciences and Technologies, University "Roma Tre", Rome, Italy
| | - Daniela Iezzi
- Department of Neuroscience, Psychology, Drug Research and Child Health -NEUROFARBA-, Section of Pharmacology and Toxicology, School of Psychology, University of Florence, Florence, Italy
| | - Antonia Manduca
- Department of Science, Section of Biomedical Sciences and Technologies, University "Roma Tre", Rome, Italy
| | - Stefano Leone
- Department of Science, Section of Biomedical Sciences and Technologies, University "Roma Tre", Rome, Italy
| | - Francesca Melancia
- Department of Science, Section of Biomedical Sciences and Technologies, University "Roma Tre", Rome, Italy
| | - Carmen Carbone
- Department of Neuroscience, Psychology, Drug Research and Child Health -NEUROFARBA-, Section of Pharmacology and Toxicology, School of Psychology, University of Florence, Florence, Italy
| | | | - Guido Mannaioni
- Department of Neuroscience, Psychology, Drug Research and Child Health -NEUROFARBA-, Section of Pharmacology and Toxicology, School of Psychology, University of Florence, Florence, Italy
| | - Alessio Masi
- Department of Neuroscience, Psychology, Drug Research and Child Health -NEUROFARBA-, Section of Pharmacology and Toxicology, School of Psychology, University of Florence, Florence, Italy.,School of Pharmacy, University of Camerino, Camerino, Italy
| | - Viviana Trezza
- Department of Science, Section of Biomedical Sciences and Technologies, University "Roma Tre", Rome, Italy
| |
Collapse
|
18
|
Greene RK, Walsh E, Mosner MG, Dichter GS. A potential mechanistic role for neuroinflammation in reward processing impairments in autism spectrum disorder. Biol Psychol 2019; 142:1-12. [PMID: 30552950 PMCID: PMC6401269 DOI: 10.1016/j.biopsycho.2018.12.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 12/11/2018] [Accepted: 12/11/2018] [Indexed: 01/18/2023]
Abstract
Accumulating evidence suggests that autism spectrum disorder (ASD) may be conceptualized within a framework of reward processing impairments. The Social Motivation Theory of Autism posits that reduced motivation to interact with people and decreased pleasure derived from social interactions may derail typical social development and contribute to the emergence of core social communication deficits in ASD. Neuroinflammation may disrupt the development of mesolimbic dopaminergic systems that are critical for optimal functioning of social reward processing systems. This neuroinflammation-induced disturbance of mesolimbic dopaminergic functioning has been substantiated using maternal immune activation rodent models whose offspring show aberrant dopaminergic corticostriatal function, as well as behavioral characteristics of ASD model systems. Preclinical findings are in turn supported by clinical evidence of increased mesolimbic neuroinflammatory responses in individuals with ASD. This review summarizes evidence for reward processing deficits and neuroinflammatory impairments in ASD and examines how immune inflammatory dysregulation may impair the development of dopaminergic mesolimbic circuitry in ASD. Finally, future research directions examining neuroinflammatory effects on reward processing in ASD are proposed.
Collapse
Affiliation(s)
- Rachel K Greene
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA.
| | - Erin Walsh
- Department of Psychiatry, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27514, USA.
| | - Maya G Mosner
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA.
| | - Gabriel S Dichter
- Department of Psychology and Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA; Department of Psychiatry, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27514, USA; Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27514, USA.
| |
Collapse
|
19
|
Liu Q, Chen MX, Sun L, Wallis CU, Zhou JS, Ao LJ, Li Q, Sham PC. Rational use of mesenchymal stem cells in the treatment of autism spectrum disorders. World J Stem Cells 2019; 11:55-72. [PMID: 30842805 PMCID: PMC6397804 DOI: 10.4252/wjsc.v11.i2.55] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 12/30/2018] [Accepted: 01/23/2019] [Indexed: 02/06/2023] Open
Abstract
Autism and autism spectrum disorders (ASD) refer to a range of conditions characterized by impaired social and communication skills and repetitive behaviors caused by different combinations of genetic and environmental influences. Although the pathophysiology underlying ASD is still unclear, recent evidence suggests that immune dysregulation and neuroinflammation play a role in the etiology of ASD. In particular, there is direct evidence supporting a role for maternal immune activation during prenatal life in neurodevelopmental conditions. Currently, the available options of behavioral therapies and pharmacological and supportive nutritional treatments in ASD are only symptomatic. Given the disturbing rise in the incidence of ASD, and the fact that there is no effective pharmacological therapy for ASD, there is an urgent need for new therapeutic options. Mesenchymal stem cells (MSCs) possess immunomodulatory properties that make them relevant to several diseases associated with inflammation and tissue damage. The paracrine regenerative mechanisms of MSCs are also suggested to be therapeutically beneficial for ASD. Thus the underlying pathology in ASD, including immune system dysregulation and inflammation, represent potential targets for MSC therapy. This review will focus on immune dysfunction in the pathogenesis of ASD and will further discuss the therapeutic potential for MSCs in mediating ASD-related immunological disorders.
Collapse
Affiliation(s)
- Qiang Liu
- Department of Surgery, The Chinese University of Hong Kong, Hong Kong, China
| | - Mo-Xian Chen
- School of Rehabilitation, Kunming Medical University, Kunming 650500, Yunnan Province, China
| | - Lin Sun
- Department of Psychology, Weifang Medical University, Weifang 261053, Shandong Province, China
| | - Chloe U Wallis
- Medical Sciences Division, University of Oxford, Oxford OX3 9DU, United Kingdom
| | - Jian-Song Zhou
- Mental Health Institute of the Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Li-Juan Ao
- School of Rehabilitation, Kunming Medical University, Kunming 650500, Yunnan Province, China
| | - Qi Li
- Department of Psychiatry, the University of Hong Kong, Hong Kong, China
| | - Pak C Sham
- Department of Psychiatry, the University of Hong Kong, Hong Kong, China
- State Key Laboratory of Brain and Cognitive Sciences, Center for Genomic Sciences, the University of Hong Kong, Hong Kong, China
| |
Collapse
|
20
|
Garbarino VR, Gilman TL, Daws LC, Gould GG. Extreme enhancement or depletion of serotonin transporter function and serotonin availability in autism spectrum disorder. Pharmacol Res 2019; 140:85-99. [PMID: 30009933 PMCID: PMC6345621 DOI: 10.1016/j.phrs.2018.07.010] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 06/22/2018] [Accepted: 07/12/2018] [Indexed: 12/15/2022]
Abstract
A variety of human and animal studies support the hypothesis that serotonin (5-hydroxytryptamine or 5-HT) system dysfunction is a contributing factor to the development of autism in some patients. However, many questions remain about how developmental manipulation of various components that influence 5-HT signaling (5-HT synthesis, transport, metabolism) persistently impair social behaviors. This review will summarize key aspects of central 5-HT function important for normal brain development, and review evidence implicating perinatal disruptions in 5-HT signaling in the pathophysiology of autism spectrum disorder. We discuss the importance, and relative dearth, of studies that explore the possible correlation to autism in the interactions between important intrinsic and extrinsic factors that may disrupt 5-HT homeostasis during development. In particular, we focus on exposure to 5-HT transport altering mechanisms such as selective serotonin-reuptake inhibitors or genetic polymorphisms in primary or auxiliary transporters of 5-HT, and how they relate to neurological stores of serotonin and its precursors. A deeper understanding of the many mechanisms by which 5-HT signaling can be disrupted, alone and in concert, may contribute to an improved understanding of the etiologies and heterogeneous nature of this disorder. We postulate that extreme bidirectional perturbations of these factors during development likely compound or synergize to facilitate enduring neurochemical changes resulting in insufficient or excessive 5-HT signaling, that could underlie the persistent behavioral characteristics of autism spectrum disorder.
Collapse
Affiliation(s)
- Valentina R Garbarino
- Department of Cellular and Integrative Physiology, United States; The Sam and Ann Barshop Institute for Longevity and Aging Studies, United States.
| | - T Lee Gilman
- Department of Cellular and Integrative Physiology, United States; Addiction Research, Treatment & Training Center of Excellence, United States.
| | - Lynette C Daws
- Department of Cellular and Integrative Physiology, United States; Addiction Research, Treatment & Training Center of Excellence, United States; Department of Pharmacology, United States.
| | - Georgianna G Gould
- Department of Cellular and Integrative Physiology, United States; Center for Biomedical Neuroscience, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA.
| |
Collapse
|
21
|
Epigenetic and Neurological Impairments Associated with Early Life Exposure to Persistent Organic Pollutants. Int J Genomics 2019; 2019:2085496. [PMID: 30733955 PMCID: PMC6348822 DOI: 10.1155/2019/2085496] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Revised: 09/14/2018] [Accepted: 10/31/2018] [Indexed: 12/31/2022] Open
Abstract
The incidence of neurodevelopmental and neurodegenerative diseases worldwide has dramatically increased over the last decades. Although the aetiology remains uncertain, evidence is now growing that exposure to persistent organic pollutants during sensitive neurodevelopmental periods such as early life may be a strong risk factor, predisposing the individual to disease development later in life. Epidemiological studies have associated environmentally persistent organic pollutant exposure to brain disorders including neuropathies, cognitive, motor, and sensory impairments; neurodevelopmental disorders such as autism spectrum disorder (ASD) and attention-deficit hyperactivity disorder (ADHD); and neurodegenerative diseases including Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis (ALS). In many ways, this expands the classical “Developmental Origins of Health and Disease” paradigm to include exposure to pollutants. This model has been refined over the years to give the current “three-hit” model that considers the individual's genetic factors as a first “hit.” It has an immediate interaction with the early-life exposome (including persistent organic pollutants) that can be considered to be a second “hit.” Together, these first two “hits” produce a quiescent or latent phenotype, most probably encoded in the epigenome, which has become susceptible to a third environmental “hit” in later life. It is only after the third “hit” that the increased risk of disease symptoms is crystallised. However, if the individual is exposed to a different environment in later life, they would be expected to remain healthy. In this review, we examine the effect of exposure to persistent organic pollutants and particulate matters in early life and the relationship to subsequent neurodevelopmental and neurodegenerative disorders. The roles of those environmental factors which may affect epigenetic DNA methylation and therefore influence normal neurodevelopment are then evaluated.
Collapse
|
22
|
Schiavi S, Iezzi D, Manduca A, Leone S, Melancia F, Carbone C, Petrella M, Mannaioni G, Masi A, Trezza V. Reward-Related Behavioral, Neurochemical and Electrophysiological Changes in a Rat Model of Autism Based on Prenatal Exposure to Valproic Acid. Front Cell Neurosci 2019; 13:479. [PMID: 31708750 DOI: 10.3389/fncel.2019.00479/bibtex] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 10/10/2019] [Indexed: 05/20/2023] Open
Abstract
Prenatal exposure to the antiepileptic drug valproic acid (VPA) induces autism spectrum disorder (ASD) in humans and autistic-like behaviors in rodents, which makes it a good model to study the neural underpinnings of ASD. Rats prenatally exposed to VPA show profound deficits in the social domain. The altered social behavior displayed by VPA-exposed rats may be due to either a deficit in social reward processing or to a more general inability to properly understand and respond to social signals. To address this issue, we performed behavioral, electrophysiological and neurochemical experiments and tested the involvement of the brain reward system in the social dysfunctions displayed by rats prenatally exposed to VPA (500 mg/kg). We found that, compared to control animals, VPA-exposed rats showed reduced play responsiveness together with impaired sociability in the three-chamber test and altered social discrimination abilities. In addition, VPA-exposed rats showed altered expression of dopamine receptors together with inherent hyperexcitability of medium spiny neurons (MSNs) in the nucleus accumbens (NAc). However, when tested for socially-induced conditioned place preference, locomotor response to amphetamine and sucrose preference, control and VPA-exposed rats performed similarly, indicating normal responses to social, drug and food rewards. On the basis of the results obtained, we hypothesize that social dysfunctions displayed by VPA-exposed rats are more likely caused by alterations in cognitive aspects of the social interaction, such as the interpretation and reciprocation of social stimuli and/or the ability to adjust the social behavior of the individual to the changing circumstances in the social and physical environment, rather than to inability to enjoy the pleasurable aspects of the social interaction. The observed neurochemical and electrophysiological alterations in the NAc may contribute to the inability of VPA-exposed rats to process and respond to social cues, or, alternatively, represent a compensatory mechanism towards VPA-induced neurodevelopmental insults.
Collapse
Affiliation(s)
- Sara Schiavi
- Department of Science, Section of Biomedical Sciences and Technologies, University "Roma Tre", Rome, Italy
| | - Daniela Iezzi
- Department of Neuroscience, Psychology, Drug Research and Child Health -NEUROFARBA-, Section of Pharmacology and Toxicology, School of Psychology, University of Florence, Florence, Italy
| | - Antonia Manduca
- Department of Science, Section of Biomedical Sciences and Technologies, University "Roma Tre", Rome, Italy
| | - Stefano Leone
- Department of Science, Section of Biomedical Sciences and Technologies, University "Roma Tre", Rome, Italy
| | - Francesca Melancia
- Department of Science, Section of Biomedical Sciences and Technologies, University "Roma Tre", Rome, Italy
| | - Carmen Carbone
- Department of Neuroscience, Psychology, Drug Research and Child Health -NEUROFARBA-, Section of Pharmacology and Toxicology, School of Psychology, University of Florence, Florence, Italy
| | | | - Guido Mannaioni
- Department of Neuroscience, Psychology, Drug Research and Child Health -NEUROFARBA-, Section of Pharmacology and Toxicology, School of Psychology, University of Florence, Florence, Italy
| | - Alessio Masi
- Department of Neuroscience, Psychology, Drug Research and Child Health -NEUROFARBA-, Section of Pharmacology and Toxicology, School of Psychology, University of Florence, Florence, Italy
- School of Pharmacy, University of Camerino, Camerino, Italy
| | - Viviana Trezza
- Department of Science, Section of Biomedical Sciences and Technologies, University "Roma Tre", Rome, Italy
| |
Collapse
|
23
|
Hamzawy MA, El-Ghandour YB, Abdel-Aziem SH, Ali ZH. Leptin and camel milk abate oxidative stress status, genotoxicity induced in valproic acid rat model of autism. Int J Immunopathol Pharmacol 2018; 32:2058738418785514. [PMID: 30004275 PMCID: PMC6047246 DOI: 10.1177/2058738418785514] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
The aspect of treatment of autistic behaviour was investigated using valproic
acid rat model of pregnant female rats. Two main groups (10 male rats/group)
were treated for 6 days and then divided into six subgroups. The first group of
normal rats was divided into three subgroups: (A) – control group, (B) – treated
with camel milk (CAM; 2 mL/p.o) and (C) – treated with leptin (1000 µg/kg i.p)
twice daily. The second group of autistic rats was randomly distributed into
four subgroups as follows: (D) – positive control (autistics rats), (E) –
treated with CAM, (F) – treated with a moderate dose of leptin and (G) – treated
with a higher dose of leptin. Autistic behaviours of male offspring were checked
by grooming and elevated pulz maze tests. Valproic acid (VPA)-induced autistic
rats showed severe changes in oxidative stress markers, neurotransmitters and
inflammatory cytokines, besides genotoxic manifestation of expression of tumour
necrosis factor (TNF)-α, Bax and caspase-3. Leptin or CAM alone showed no signs
of toxicity. CAM showed pronounced improvement in control rats than control
itself. Leptin or CAM treatment of autistic animals showed a significant
improvement of all measured parameters and genetic expression values. The
improvement was pronounced in animals treated with CAM. These results suggest
that CAM is a potential therapeutic candidate for autism via regulation of
inflammatory and apoptotic pathways. Leptin plays an essential role in
alleviation of autistic behaviour through antioxidant effects.
Collapse
Affiliation(s)
- Mohamed A Hamzawy
- 1 Pharmacology & Toxicology Department, Faculty of Pharmacy, Fayoum University, Fayoum, Egypt
| | - Yasmin B El-Ghandour
- 2 Oral Biology Department, Faculty Oral and Dental Medicine, Cairo University, Egypt
| | | | - Zoba H Ali
- 2 Oral Biology Department, Faculty Oral and Dental Medicine, Cairo University, Egypt
| |
Collapse
|
24
|
Kuo HY, Liu FC. Molecular Pathology and Pharmacological Treatment of Autism Spectrum Disorder-Like Phenotypes Using Rodent Models. Front Cell Neurosci 2018; 12:422. [PMID: 30524240 PMCID: PMC6262306 DOI: 10.3389/fncel.2018.00422] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 10/29/2018] [Indexed: 12/13/2022] Open
Abstract
Autism spectrum disorder (ASD) is a heterogeneous neurodevelopmental disorder with a high prevalence rate. The core symptoms of ASD patients are impaired social communication and repetitive behavior. Genetic and environmental factors contribute to pathophysiology of ASD. Regarding environmental risk factors, it is known that valproic acid (VPA) exposure during pregnancy increases the chance of ASD among offspring. Over a decade of animal model studies have shown that maternal treatment with VPA in rodents recapitulates ASD-like pathophysiology at a molecular, cellular and behavioral level. Here, we review the prevailing theories of ASD pathogenesis, including excitatory/inhibitory imbalance, neurotransmitter dysfunction, dysfunction of mTOR and endocannabinoid signaling pathways, neuroinflammation and epigenetic alterations that have been associated with ASD. We also describe the evidence linking neuropathological changes to ASD-like behavioral abnormalities in maternal VPA-treated rodents. In addition to obtaining an understanding of the neuropathological mechanisms, the VPA-induced ASD-like animal models also serve as a good platform for testing pharmacological reagents that might be use treating ASD. We therefore have summarized the various pharmacological studies that have targeted the classical neurotransmitter systems, the endocannabinoids, the Wnt signal pathway and neuroinflammation. These approaches have been shown to often be able to ameliorate the ASD-like phenotypes induced by maternal VPA treatments.
Collapse
Affiliation(s)
- Hsiao-Ying Kuo
- Institute of Neuroscience, National Yang-Ming University, Taipei, Taiwan
| | - Fu-Chin Liu
- Institute of Neuroscience, National Yang-Ming University, Taipei, Taiwan
- Brain Research Center, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
25
|
Zieminska E, Toczylowska B, Diamandakis D, Hilgier W, Filipkowski RK, Polowy R, Orzel J, Gorka M, Lazarewicz JW. Glutamate, Glutamine and GABA Levels in Rat Brain Measured Using MRS, HPLC and NMR Methods in Study of Two Models of Autism. Front Mol Neurosci 2018; 11:418. [PMID: 30505268 PMCID: PMC6250849 DOI: 10.3389/fnmol.2018.00418] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Accepted: 10/26/2018] [Indexed: 11/13/2022] Open
Abstract
The disorders of the glutamatergic neurotransmission have been associated with pathogenesis of autism. In this study we evaluated the impact of the in vivo and ex vivo test methodology on measurements of levels of neurotransmitter amino acids in hippocampus of rats for valproic acid- (VPA) and thalidomide- (THAL) induced models of autism. The main goal was to compare the changes in concentrations of glutamate (Glu), glutamine (Gln) and GABA between both autistic groups and the control, measured in vivo and ex vivo in homogenates. The rat pups underwent three in vivo tests: ultrasonic vocalization (USV), magnetic resonance spectroscopy (MRS) and unilateral microdialysis of the hippocampus. Analyses of homogenates of rat hippocampus were performed using high-performance liquid chromatography (HPLC) and nuclear magnetic resonance (NMR) spectroscopy. For the statistical analysis, we performed univariate and multivariate tests. USV test, which is considered in rodents as an indicator of pathology similar to autism, showed decreased USV in VPA and THAL groups. In vivo MRS studies demonstrated increases of Glu content in male rat's hippocampus in VPA and THAL groups, while the microdialysis, which allows examination of the contents in the extracellular space, detected decreases in the basal level of Gln concentrations in VPA and THAL groups. Ex vivo HPLC studies showed that levels of Glu, Gln and GABA significantly increased in male rat's hippocampus in the VPA and THAL groups, while NMR studies showed increased levels of Gln and GABA in the VPA group. Collectively, these results are consistent with the hypothesis suggesting the role of the glutamatergic disturbances on the pathogenesis of autism. For all methods used, the values of measured changes were in the same direction. The orthogonal partial least square discriminant analysis confirmed that both animal models of autism tested here can be used to trace neurochemical changes in the brain.
Collapse
Affiliation(s)
- Elzbieta Zieminska
- Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Beata Toczylowska
- Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Warsaw, Poland
| | - Dominik Diamandakis
- Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Wojciech Hilgier
- Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | | | - Rafal Polowy
- Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Jaroslaw Orzel
- Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland.,Faculty of Electronics and Information Technology, Warsaw University of Technology, Warsaw, Poland
| | - Michal Gorka
- Faculty of Physics, University of Warsaw, Warsaw, Poland
| | | |
Collapse
|
26
|
Wang R, Hausknecht K, Shen RY, Haj-Dahmane S. Potentiation of Glutamatergic Synaptic Transmission Onto Dorsal Raphe Serotonergic Neurons in the Valproic Acid Model of Autism. Front Pharmacol 2018; 9:1185. [PMID: 30459605 PMCID: PMC6232663 DOI: 10.3389/fphar.2018.01185] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 09/28/2018] [Indexed: 11/17/2022] Open
Abstract
Autism spectrum disorder (ASD) is characterized by social and communicative impairments and increased repetitive behaviors. These symptoms are often comorbid with increased anxiety. Prenatal exposure to valproic acid (VPA), an anti-seizure and mood stabilizer medication, is a major environmental risk factor of ASD. Given the important role of the serotonergic (5-HT) system in anxiety, we examined the impact of prenatal VPA exposure on the function of dorsal raphe nucleus (DRn) 5-HT neurons. We found that male rats prenatally exposed to VPA exhibited increased anxiety-like behaviors revealed by a decreased time spent on the open arms of the elevated plus maze. Prenatal VPA exposed rats also exhibited a stereotypic behavior as indicated by excessive self-grooming in a novel environment. These behavioral phenotypes were associated with increased electrical activity of putative DRn 5-HT neurons recorded in vitro. Examination of underlying mechanisms revealed that prenatal VPA exposure increased excitation/inhibition ratio in synapses onto these neurons. The effect was mainly mediated by enhanced glutamate but not GABA release. We found reduced paired-pulse ratio (PPR) of evoked excitatory postsynaptic currents (EPSCs) and increased frequency but not amplitude of miniature EPSCs in VPA exposed rats. On the other hand, presynaptic GABA release did not change in VPA exposed rats, as the PPR of evoked inhibitory postsynaptic currents was unaltered. Furthermore, the spike-timing-dependent long-term potentiation at the glutamatergic synapses was occluded, indicating glutamatergic synaptic transmission is maximized. Lastly, VPA exposure did not alter the intrinsic membrane properties of DRn 5-HT neurons. Taken together, these results indicate that prenatal VPA exposure profoundly enhances glutamatergic synaptic transmission in the DRn and increases spontaneous firing in DRn 5-HT neurons, which could lead to increased serotonergic tone and underlie the increased anxiety and stereotypy after prenatal VPA exposure.
Collapse
Affiliation(s)
- Ruixiang Wang
- Research Institute on Addictions, University at Buffalo, The State University of New York, Buffalo, NY, United States.,Department of Psychology, University at Buffalo, The State University of New York, Buffalo, NY, United States
| | - Kathryn Hausknecht
- Research Institute on Addictions, University at Buffalo, The State University of New York, Buffalo, NY, United States
| | - Roh-Yu Shen
- Research Institute on Addictions, University at Buffalo, The State University of New York, Buffalo, NY, United States.,Department of Pharmacology and Toxicology, The Jacob School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, United States.,Neuroscience Program, University at Buffalo, The State University of New York, Buffalo, NY, United States
| | - Samir Haj-Dahmane
- Research Institute on Addictions, University at Buffalo, The State University of New York, Buffalo, NY, United States.,Department of Pharmacology and Toxicology, The Jacob School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, United States.,Neuroscience Program, University at Buffalo, The State University of New York, Buffalo, NY, United States
| |
Collapse
|
27
|
Dadalko OI, Travers BG. Evidence for Brainstem Contributions to Autism Spectrum Disorders. Front Integr Neurosci 2018; 12:47. [PMID: 30337860 PMCID: PMC6180283 DOI: 10.3389/fnint.2018.00047] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 09/18/2018] [Indexed: 12/27/2022] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental condition that affects one in 59 children in the United States. Although there is a mounting body of knowledge of cortical and cerebellar contributions to ASD, our knowledge about the early developing brainstem in ASD is only beginning to accumulate. Understanding how brainstem neurotransmission is implicated in ASD is important because many of this condition’s sensory and motor symptoms are consistent with brainstem pathology. Therefore, the purpose of this review was to integrate epidemiological, behavioral, histological, neuroimaging, and animal evidence of brainstem contributions to ASD. Because ASD is a neurodevelopmental condition, we examined the available data through a lens of hierarchical brain development. The review of the literature suggests that developmental alterations of the brainstem could have potential cascading effects on cortical and cerebellar formation, ultimately leading to ASD symptoms. This view is supported by human epidemiology findings and data from animal models of ASD, showing that perturbed development of the brainstem substructures, particularly during the peak formation of the brainstem’s monoaminergic centers, may relate to ASD or ASD-like behaviors. Furthermore, we review evidence from human histology, psychophysiology, and neuroimaging suggesting that brainstem development and maturation may be atypical in ASD and may be related to key ASD symptoms, such as atypical sensorimotor features and social responsiveness. From this review there emerges the need of future research to validate early detection of the brainstem-based somatosensory and psychophysiological behaviors that emerge in infancy, and to examine the brainstem across the life span, while accounting for age. In all, there is preliminary evidence for brainstem involvement in ASD, but a better understanding of the brainstem’s role would likely pave the way for earlier diagnosis and treatment of ASD.
Collapse
Affiliation(s)
- Olga I Dadalko
- Motor and Brain Development Lab, Waisman Center, University of Wisconsin-Madison, Madison, WI, United States
| | - Brittany G Travers
- Motor and Brain Development Lab, Occupational Therapy Program in the Department of Kinesiology, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
28
|
Hennessey T, Andari E, Rainnie DG. RDoC-based categorization of amygdala functions and its implications in autism. Neurosci Biobehav Rev 2018; 90:115-129. [PMID: 29660417 PMCID: PMC6250055 DOI: 10.1016/j.neubiorev.2018.04.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 03/09/2018] [Accepted: 04/09/2018] [Indexed: 12/28/2022]
Abstract
Confusion endures as to the exact role of the amygdala in relation to autism. To help resolve this we turned to the NIMH's Research Domain Criteria (RDoC) which provides a classification schema that identifies different categories of behaviors that can turn pathologic in mental health disorders, e.g. autism. While RDoC incorporates all the known neurobiological substrates for each domain, this review will focus primarily on the amygdala. We first consider the amygdala from an anatomical, historical, and developmental perspective. Next, we examine the different domains and constructs of RDoC that the amygdala is involved in: Negative Valence Systems, Positive Valence Systems, Cognitive Systems, Social Processes, and Arousal and Regulatory Systems. Then the evidence for a dysfunctional amygdala in autism is presented with a focus on alterations in development, prenatal valproic acid exposure as a model for ASD, and changes in the oxytocin system therein. Finally, a synthesis of RDoC, the amygdala, and autism is offered, emphasizing the task of disambiguation and suggestions for future research.
Collapse
Affiliation(s)
- Thomas Hennessey
- Department of Behavioral Neuroscience and Psychiatric Disorders, Yerkes National Primate Research Center, United States; Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, 30329, United States
| | - Elissar Andari
- Silvio O. Conte Center for Oxytocin and Social Cognition, Department of Psychiatry and Behavioral Sciences, Division of Behavioral Neuroscience and Psychiatric Disorders, Yerkes National Primate Research Center, Emory University, United States
| | - Donald G Rainnie
- Department of Behavioral Neuroscience and Psychiatric Disorders, Yerkes National Primate Research Center, United States; Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, 30329, United States.
| |
Collapse
|
29
|
Al-Hamdan AZ, Preetha PP, Albashaireh RN, Al-Hamdan MZ, Crosson WL. Investigating the effects of environmental factors on autism spectrum disorder in the USA using remotely sensed data. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2018; 25:7924-7936. [PMID: 29299867 DOI: 10.1007/s11356-017-1114-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 12/20/2017] [Indexed: 06/07/2023]
Abstract
This study aimed to assess the association between exposures to outdoor environmental factors and autism spectrum disorder (ASD) prevalence in a diverse and spatially distributed population of 8-year-old children from the USA (n = 2,097,188) using the air quality index (AQI) of the US Environmental Protection Agency as well as satellite-derived data of PM2.5 concentrations, sunlight, and maximum heat index. Multivariable logistic regression analyses were performed to determine whether the unhealthy AQI, PM2.5, sunlight, and maximum heat index were related to the odds of ASD prevalence based on gender and race and taking into consideration the confounding factors of smoking and socioeconomic status. The logistic regression odds ratios for ASD per 10% increase in the unhealthy AQI were greater than 1 for all categories, indicating that unhealthy AQI is related to the odds of ASD prevalence. The odds ratio of ASD due to the exposure to the unhealthy AQI was higher for Asians (OR = 2.96, 95% CI = 1.11-7.88) than that for Hispanics (OR = 1.308, 95% CI = 0.607-2.820), and it was higher for Blacks (OR = 1.398, 95% CI = 0.827-2.364) than that for Whites (OR = 1.219, 95% CI = 0.760-1.954). The odds ratio of ASD due to the unhealthy AQI was slightly higher for males (OR = 1.123, 95% CI = 0.771-1.635) than that for females (OR = 1.117, 95% CI = 0.789-1.581). The effects of the unhealthy environmental exposures on the odds ratios of ASD of this study were inconclusive (i.e., statically insignificant; p value > 0.05) for all categories except for Asians. The odds ratios of ASD for Asians were increased by 5, 12, and 14% with increased levels of the environmental exposures of 10 μg/m3 of PM2.5, 1000 kJ/m2 of sunlight, and 1 °F of maximum heat index, respectively. The odds ratios of ASD prevalence for all categories, except for Asians, were increased with the inclusion of the smoking covariate, reflecting the effect of smoking on ASD prevalence besides the unhealthy environmental factors.
Collapse
Affiliation(s)
- Ashraf Z Al-Hamdan
- Department of Civil and Environmental Engineering, University of Alabama in Huntsville, 301 Sparkman Drive, Huntsville, AL, 35899, USA.
| | - Pooja P Preetha
- Department of Civil and Environmental Engineering, University of Alabama in Huntsville, 301 Sparkman Drive, Huntsville, AL, 35899, USA
| | - Reem N Albashaireh
- Department of Mathematics, Alabama Agricultural and Mechanical University, Normal, AL, 35762, USA
| | - Mohammad Z Al-Hamdan
- Universities Space Research Association, NASA Marshall Space Flight Center, National Space Science and Technology Center, 320 Sparkman Drive, Huntsville, AL, 35805, USA
| | - William L Crosson
- Universities Space Research Association, NASA Marshall Space Flight Center, National Space Science and Technology Center, 320 Sparkman Drive, Huntsville, AL, 35805, USA
| |
Collapse
|
30
|
Liu T, Guo F, Zhu X, He X, Xie L. Thalidomide and its analogues: A review of the potential for immunomodulation of fibrosis diseases and opthalmopathy. Exp Ther Med 2017; 14:5251-5257. [PMID: 29285050 DOI: 10.3892/etm.2017.5209] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 02/14/2017] [Indexed: 12/11/2022] Open
Abstract
The US Food and Drug Administration approved thalidomide and its analogues for the treatment of erythema nodosum leprosum, in spite of the notoriety of reports of severe birth defects in the middle of the last century. As immunomodulatory drugs, thalidomide and its analogues have been used to effectively treat various diseases. In the present review, preclinical data about the effects of thalidomide and its analogues on the immune system are integrated, including the effects of cytokines on transdifferentiation, the anti-inflammatory effect, immune cell function regulation and angiogenesis. The present review also investigates the latest developments of thalidomide as a therapeutic option for the treatment of idiopathic pulmonary fibrosis, skin fibrosis, and ophthalmopathies.
Collapse
Affiliation(s)
- Ting Liu
- Department of Ophthalmology, Daping Hospital and Research Institute of Surgery, The Third Military Medical University, People's Liberation Army, Chongqing 400042, P.R. China
| | - Feng Guo
- Department of Ophthalmology, Daping Hospital and Research Institute of Surgery, The Third Military Medical University, People's Liberation Army, Chongqing 400042, P.R. China
| | - Xiaomin Zhu
- Department of Ophthalmology, Daping Hospital and Research Institute of Surgery, The Third Military Medical University, People's Liberation Army, Chongqing 400042, P.R. China
| | - Xiangge He
- Department of Ophthalmology, Daping Hospital and Research Institute of Surgery, The Third Military Medical University, People's Liberation Army, Chongqing 400042, P.R. China
| | - Lin Xie
- Department of Ophthalmology, Daping Hospital and Research Institute of Surgery, The Third Military Medical University, People's Liberation Army, Chongqing 400042, P.R. China
| |
Collapse
|
31
|
Ellenbroek BA, August C, Youn J. Does Prenatal Valproate Interact with a Genetic Reduction in the Serotonin Transporter? A Rat Study on Anxiety and Cognition. Front Neurosci 2016; 10:424. [PMID: 27708559 PMCID: PMC5030776 DOI: 10.3389/fnins.2016.00424] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 08/30/2016] [Indexed: 12/22/2022] Open
Abstract
There is ample evidence that prenatal exposure to valproate (or valproic acid, VPA) enhances the risk of developing Autism Spectrum Disorders (ASD). In line with this, a single injection of VPA induces a multitude of ASD-like symptoms in animals, such as rats and mice. However, there is equally strong evidence that genetic factors contribute significantly to the risk of ASD and indeed, like most other psychiatric disorders, ASD is now generally thought to results from an interaction between genetic and environmental factors. Given that VPA significantly impacts on the serotonergic system, and serotonin has strong biochemical and genetic links to ASD, we aimed to investigate the interaction between genetic reduction in the serotonin transporter and prenatal valproate administration. More specifically, we exposed both wildtype (SERT+/+) rats and rats heterozygous for the serotonin transporter deletion (SERT+/-) to a single injection of 400 mg/kg VPA at gestational day (GD) 12. The offspring, in adulthood, was assessed in four different tests: Elevated Plus Maze and Novelty Suppressed Feeding as measures for anxiety and prepulse inhibition (PPI) and latent inhibition as measures for cognition and information processing. The results show that prenatal VPA significantly increased anxiety in both paradigm, reduced PPI and reduced conditioning in the latent inhibition paradigm. However, we failed to find a significant gene-environment interaction. We propose that this may be related to the timing of the VPA injection and suggest that whereas GD12 might be optimal for affecting normal rat, rats with a genetically compromised serotonergic system may be more sensitive to VPA at earlier time points during gestation. Overall our data are the first to investigate gene * environmental interactions in a genetic rat model for ASD and suggest that timing may be of crucial importance to the long-term outcome.
Collapse
Affiliation(s)
- Bart A Ellenbroek
- School of Psychology, Victoria University of Wellington Wellington, New Zealand
| | - Caren August
- School of Psychology, Victoria University of Wellington Wellington, New Zealand
| | - Jiun Youn
- School of Psychology, Victoria University of Wellington Wellington, New Zealand
| |
Collapse
|
32
|
Guo YP, Commons KG. Serotonin neuron abnormalities in the BTBR mouse model of autism. Autism Res 2016; 10:66-77. [PMID: 27478061 DOI: 10.1002/aur.1665] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 06/08/2016] [Accepted: 06/13/2016] [Indexed: 12/21/2022]
Abstract
The inbred mouse strain BTBR T+ Itpr3tf /J (BTBR) is studied as a model of idiopathic autism because they are less social and more resistant to change than other strains. Forebrain serotonin receptors and the response to serotonin drugs are altered in BTBR mice, yet it remains unknown if serotonin neurons themselves are abnormal. In this study, we found that serotonin tissue content and the density of serotonin axons is reduced in the hippocampus of BTBR mice in comparison to C57BL/6J (C57) mice. This was accompanied by possible compensatory changes in serotonin neurons that were most pronounced in regions known to provide innervation to the hippocampus: the caudal dorsal raphe (B6) and the median raphe. These changes included increased numbers of serotonin neurons and hyperactivation of Fos expression. Metrics of serotonin neurons in the rostral 2/3 of the dorsal raphe and serotonin content of the prefrontal cortex were less impacted. Thus, serotonin neurons exhibit region-dependent abnormalities in the BTBR mouse that may contribute to their altered behavioral profile. Autism Res 2017, 10: 66-77. © 2016 International Society for Autism Research, Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Yue-Ping Guo
- Department of Anesthesiology, Second Affiliated Hospital, Harbin Medical University, Harbin, China.,Department of Anesthesiology, Perioperative, and Pain Medicine, Boston Children's Hospital; Department of Anaesthesia, Harvard Medical School, Boston, Massachusetts
| | - Kathryn G Commons
- Department of Anesthesiology, Perioperative, and Pain Medicine, Boston Children's Hospital; Department of Anaesthesia, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
33
|
Perez-Pouchoulen M, Miquel M, Saft P, Brug B, Toledo R, Hernandez ME, Manzo J. Prenatal exposure to sodium valproate alters androgen receptor expression in the developing cerebellum in a region and age specific manner in male and female rats. Int J Dev Neurosci 2016; 53:46-52. [PMID: 27423376 DOI: 10.1016/j.ijdevneu.2016.07.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Revised: 06/22/2016] [Accepted: 07/12/2016] [Indexed: 01/31/2023] Open
Abstract
Valproic acid (VPA) is an anti-epileptic drug with teratogenicity activity that has been related to autism. In rodents, exposure to VPA in utero leads to brain abnormalities similar than those reported in the autistic brain. Particularly, VPA reduces the number of Purkinje neurons in the rat cerebellum parallel to cerebellar abnormalities found in autism. Thus, we injected pregnant females on embryonic day 12 either with VPA (600mg/kg, i.p.) or 0.9% saline solution and obtained the cerebellum from their offspring at different postnatal time points. Testosterone has been linked to autism and plays an important role during brain development. Therefore, we identified and analyzed the androgen receptor (AR) by immunohistochemistry and densitometry, respectively. We found VPA decreases AR density in the superficial Purkinje layer only in cerebellar lobule 8 at PN7, but increased it at PN14 compared to control in males. In females, VPA decreased AR density in the superficial Purkinje layer in cerebellar lobule 6 at PN14, but increased it in lobule 9 at the same time point. No differences were found in the deep Purkinje layer of any cerebellar lobule in terms of AR density neither in males nor females. We additionally found a particular AR density decreasing in both superficial and deep regions across development in the majority of cerebellar lobules in males, but in all cerebellar lobules in females. Thus, our results indicate that VPA disrupts the AR ontogeny in the developing cerebellum in an age and region specific manner in male and female rats. Future epigenetic studies including the evaluation of histone deacetylases (HDAC's) might shed light these results as HDAC's are expressed by Purkinje neurons, interact with the AR and are VPA targets. This work contributes to the understanding of the cerebellar development and it might help to understand the role of the cerebellum in neurodevelopmental disorders such as autism.
Collapse
Affiliation(s)
| | - Marta Miquel
- Area de Psicobiologia, Universidad Jaume I, Castellon de la Plana, Spain.
| | - Paul Saft
- Centro de Investigaciones Cerebrales, Universidad Veracruzana, Xalapa, Ver, Mexico.
| | - Brenda Brug
- Centro de Investigaciones Cerebrales, Universidad Veracruzana, Xalapa, Ver, Mexico.
| | - Rebeca Toledo
- Centro de Investigaciones Cerebrales, Universidad Veracruzana, Xalapa, Ver, Mexico.
| | | | - Jorge Manzo
- Centro de Investigaciones Cerebrales, Universidad Veracruzana, Xalapa, Ver, Mexico.
| |
Collapse
|
34
|
Al-Salem HS, Bhat RS, Al-Ayadhi L, El-Ansary A. Therapeutic potency of bee pollen against biochemical autistic features induced through acute and sub-acute neurotoxicity of orally administered propionic acid. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2016; 16:120. [PMID: 27107819 PMCID: PMC4842259 DOI: 10.1186/s12906-016-1099-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2015] [Accepted: 04/16/2016] [Indexed: 01/17/2023]
Abstract
Background It is now well documented that postnatal exposure to certain chemicals has been reported to increase the risk of autism spectrum disorder. Propionic acid (PA), as a metabolic product of gut microbiotaandas a commonly used food additive, has been reported to mediate the effects of autism. Results from animal studies may help to identify environmental neurotoxic agents and drugs that can ameliorate neurotoxicity and may thereby aid in the treatment of autism. The present study investigated the ameliorative effects of natural bee pollen against acute and sub-acute brain intoxication induced by (PA) in rats. Methods Twenty-four young male Western Albino ratswere enrolled in the present study. They were classified into four equal groups, eachwith6 rats. The control group received only phosphate buffered saline; the oral buffered PA-treated groups (II and III) received a neurotoxic dose of 750 mg/kg body weight divided in 3 dose of 250 mg/kg body weight/day serving asthe acute group and 750 mg/kg body weight divided in 10 equal dose of 75 mg/kg body weight/day as the sub-acute group. The fourth group received 50 mg bee pollen for 30 days after PA-acute intoxication. Results The obtained data showed that the PA-treated groups demonstrated multiple signs of brain toxicity, as indicated by a depletion of serotonin (5HT), dopamine and nor-adrenaline, together withan increase in IFN-γ and caspase 3. Bee pollen was effective in ameliorating the neurotoxic effect of PA. All measured parameters demonstrated minimal alteration in comparison with thecontrol animal than did those of acute and sub-acute PA-treated animals. Conclusions In conclusion, bee pollen demonstrates anti-inflammatory and anti-apoptotic effects while ameliorating the impaired neurochemistry of PA-intoxicated rats.
Collapse
|
35
|
Mabunga DFN, Gonzales ELT, Kim JW, Kim KC, Shin CY. Exploring the Validity of Valproic Acid Animal Model of Autism. Exp Neurobiol 2015; 24:285-300. [PMID: 26713077 PMCID: PMC4688329 DOI: 10.5607/en.2015.24.4.285] [Citation(s) in RCA: 149] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 12/01/2015] [Accepted: 12/02/2015] [Indexed: 02/06/2023] Open
Abstract
The valproic acid (VPA) animal model of autism spectrum disorder (ASD) is one of the most widely used animal model in the field. Like any other disease models, it can't model the totality of the features seen in autism. Then, is it valid to model autism? This model demonstrates many of the structural and behavioral features that can be observed in individuals with autism. These similarities enable the model to define relevant pathways of developmental dysregulation resulting from environmental manipulation. The uncovering of these complex pathways resulted to the growing pool of potential therapeutic candidates addressing the core symptoms of ASD. Here, we summarize the validity points of VPA that may or may not qualify it as a valid animal model of ASD.
Collapse
Affiliation(s)
- Darine Froy N Mabunga
- Department of Neuroscience, School of Medicine, and Neuroscience Research Center, SMART-IABS and KU Open Innovation Center, Konkuk University, Seoul 05029, Korea
| | - Edson Luck T Gonzales
- Department of Neuroscience, School of Medicine, and Neuroscience Research Center, SMART-IABS and KU Open Innovation Center, Konkuk University, Seoul 05029, Korea
| | - Ji-Woon Kim
- Department of Neuroscience, School of Medicine, and Neuroscience Research Center, SMART-IABS and KU Open Innovation Center, Konkuk University, Seoul 05029, Korea
| | - Ki Chan Kim
- Department of Neuroscience, School of Medicine, and Neuroscience Research Center, SMART-IABS and KU Open Innovation Center, Konkuk University, Seoul 05029, Korea
| | - Chan Young Shin
- Department of Neuroscience, School of Medicine, and Neuroscience Research Center, SMART-IABS and KU Open Innovation Center, Konkuk University, Seoul 05029, Korea. ; Department of Pharmacology, School of Medicine, Konkuk University, Seoul 05029, Korea
| |
Collapse
|
36
|
Lipopolysaccharide Exposure Induces Maternal Hypozincemia, and Prenatal Zinc Treatment Prevents Autistic-Like Behaviors and Disturbances in the Striatal Dopaminergic and mTOR Systems of Offspring. PLoS One 2015. [PMID: 26218250 PMCID: PMC4517817 DOI: 10.1371/journal.pone.0134565] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Autism is characterized by social deficits, repetitive behaviors, and cognitive inflexibility. The risk factors appear to include genetic and environmental conditions, such as prenatal infections and maternal dietary factors. Previous investigations by our group have demonstrated that prenatal exposure to lipopolysaccharide (LPS), which mimics infection by gram-negative bacteria, induces autistic-like behaviors. To understand the causes of autistic-like behaviors, we evaluated maternal serum metal concentrations, which are involved in intrauterine development and infection/inflammation. We identified reduced maternal levels of zinc, magnesium, selenium and manganese after LPS exposure. Because LPS induced maternal hypozincemia, we treated dams with zinc in an attempt to prevent or ease the impairments in the offspring. We evaluated the social and cognitive autistic-like behaviors and brain tissues of the offspring to identify the central mechanism that triggers the development of autism. Prenatal LPS exposure impaired play behaviors and T-maze spontaneous alternations, i.e., it induced autistic-like behaviors. Prenatal LPS also decreased tyrosine hydroxylase levels and increased the levels of mammalian target of rapamycin (mTOR) in the striatum. Thus, striatal dopaminergic impairments may be related to autism. Moreover, excessive signaling through the mTOR pathway has been considered a biomarker of autism, corroborating our rat model of autism. Prenatal zinc treatment prevented these autistic-like behaviors and striatal dopaminergic and mTOR disturbances in the offspring induced by LPS exposure. The present findings revealed a possible relation between maternal hypozincemia during gestation and the onset of autism. Furthermore, prenatal zinc administration appears to have a beneficial effect on the prevention of autism.
Collapse
|
37
|
Mottron L, Duret P, Mueller S, Moore RD, Forgeot d'Arc B, Jacquemont S, Xiong L. Sex differences in brain plasticity: a new hypothesis for sex ratio bias in autism. Mol Autism 2015; 6:33. [PMID: 26052415 PMCID: PMC4456778 DOI: 10.1186/s13229-015-0024-1] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2014] [Accepted: 04/27/2015] [Indexed: 01/13/2023] Open
Abstract
Several observations support the hypothesis that differences in synaptic and regional cerebral plasticity between the sexes account for the high ratio of males to females in autism. First, males are more susceptible than females to perturbations in genes involved in synaptic plasticity. Second, sex-related differences in non-autistic brain structure and function are observed in highly variable regions, namely, the heteromodal associative cortices, and overlap with structural particularities and enhanced activity of perceptual associative regions in autistic individuals. Finally, functional cortical reallocations following brain lesions in non-autistic adults (for example, traumatic brain injury, multiple sclerosis) are sex-dependent. Interactions between genetic sex and hormones may therefore result in higher synaptic and consecutively regional plasticity in perceptual brain areas in males than in females. The onset of autism may largely involve mutations altering synaptic plasticity that create a plastic reaction affecting the most variable and sexually dimorphic brain regions. The sex ratio bias in autism may arise because males have a lower threshold than females for the development of this plastic reaction following a genetic or environmental event.
Collapse
Affiliation(s)
- Laurent Mottron
- Centre d'excellence en Troubles envahissants du dévelopement de l'Université de Montréal (CETEDUM), Montréal, Canada.,Hôpital Rivière-des-Prairies, Département de Psychiatrie, Montréal, Canada.,Centre de Recherche de l'Institut Universitaire en Santé Mentale de Montréal, Québec, Canada.,Department of Psychiatry, University of Montreal, Québec, Canada
| | - Pauline Duret
- Centre d'excellence en Troubles envahissants du dévelopement de l'Université de Montréal (CETEDUM), Montréal, Canada.,Hôpital Rivière-des-Prairies, Département de Psychiatrie, Montréal, Canada.,Centre de Recherche de l'Institut Universitaire en Santé Mentale de Montréal, Québec, Canada.,Department of Psychiatry, University of Montreal, Québec, Canada.,Département de Biologie, École Normale Supérieure de Lyon, Lyon, CEDEX 07 France
| | - Sophia Mueller
- Institute of Clinical Radiology, University Hospitals, Munich, Germany.,Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA 02129 USA.,Harvard University, Center for Brain Science, Cambridge, MA 02138 USA
| | - Robert D Moore
- Department of Psychiatry, University of Montreal, Québec, Canada.,Department of Health Sciences, University of Montreal, Montreal, Canada.,College of Applied Health Sciences, University of Illinois, Urbana-Champaign, USA
| | - Baudouin Forgeot d'Arc
- Centre d'excellence en Troubles envahissants du dévelopement de l'Université de Montréal (CETEDUM), Montréal, Canada.,Hôpital Rivière-des-Prairies, Département de Psychiatrie, Montréal, Canada.,Centre de Recherche de l'Institut Universitaire en Santé Mentale de Montréal, Québec, Canada.,Department of Psychiatry, University of Montreal, Québec, Canada
| | - Sebastien Jacquemont
- Department of Psychiatry, University of Montreal, Québec, Canada.,Centre de recherche, Centre Hospitalier Universitaire Sainte Justine, Montréal, Canada.,Service of Medical Genetics, University Hospital of Lausanne, University of Lausanne, Lausanne, 1011 Switzerland
| | - Lan Xiong
- Centre de Recherche de l'Institut Universitaire en Santé Mentale de Montréal, Québec, Canada.,Department of Psychiatry, University of Montreal, Québec, Canada
| |
Collapse
|
38
|
Kang J, Kim E. Suppression of NMDA receptor function in mice prenatally exposed to valproic acid improves social deficits and repetitive behaviors. Front Mol Neurosci 2015; 8:17. [PMID: 26074764 PMCID: PMC4444740 DOI: 10.3389/fnmol.2015.00017] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 05/09/2015] [Indexed: 12/12/2022] Open
Abstract
Animals prenatally exposed to valproic acid (VPA), an antiepileptic agent, have been used as a model for autism spectrum disorders (ASDs). Previous studies have identified enhanced NMDA receptor (NMDAR) function in the brain of VPA rats, and demonstrated that pharmacological suppression of NMDAR function normalizes social deficits in these animals. However, whether repetitive behavior, another key feature of ASDs, can be rescued by NMDAR inhibition remains unknown. We report here that memantine, an NMDAR antagonist, administered to VPA mice rescues both social deficits and repetitive behaviors such as self-grooming and jumping. These results suggest that suppression of elevated NMDAR function in VPA animals normalizes repetitive behaviors in addition to social deficits.
Collapse
Affiliation(s)
- Jaeseung Kang
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology Daejeon, Korea
| | - Eunjoon Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology Daejeon, Korea ; Center for Synaptic Brain Dysfunctions, Institute for Basic Science Daejeon, Korea
| |
Collapse
|
39
|
Nicolini C, Ahn Y, Michalski B, Rho JM, Fahnestock M. Decreased mTOR signaling pathway in human idiopathic autism and in rats exposed to valproic acid. Acta Neuropathol Commun 2015; 3:3. [PMID: 25627160 PMCID: PMC4307681 DOI: 10.1186/s40478-015-0184-4] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Accepted: 01/04/2015] [Indexed: 12/20/2022] Open
Abstract
Background The molecular mechanisms underlying autistic behaviors remain to be elucidated. Mutations in genes linked to autism adversely affect molecules regulating dendritic spine formation, function and plasticity, and some increase the mammalian target of rapamycin, mTOR, a regulator of protein synthesis at spines. Here, we investigated whether the Akt/mTOR pathway is disrupted in idiopathic autism and in rats exposed to valproic acid, an animal model exhibiting autistic-like behavior. Methods Components of the mTOR pathway were assayed by Western blotting in postmortem fusiform gyrus samples from 11 subjects with idiopathic autism and 13 controls and in valproic acid versus saline-exposed rat neocortex. Additionally, protein levels of brain-derived neurotrophic factor receptor (TrkB) isoforms and the postsynaptic organizing molecule PSD-95 were measured in autistic versus control subjects. Results Full-length TrkB, PI3K, Akt, phosphorylated and total mTOR, p70S6 kinase, eIF4B and PSD-95 were reduced in autistic versus control fusiform gyrus. Similarly, phosphorylated and total Akt, mTOR and 4E-BP1 and phosphorylated S6 protein were decreased in valproic acid- versus saline-exposed rats. However, no changes in 4E-BP1 or eIF4E were found in autistic brains. Conclusions In contrast to some monogenic disorders with high rates of autism, our data demonstrate down-regulation of the Akt/mTOR pathway, specifically via p70S6K/eIF4B, in idiopathic autism. These findings suggest that disruption of this pathway in either direction is widespread in autism and can have adverse consequences for synaptic function. The use of valproic acid, a histone deacetylase inhibitor, in rats successfully modeled these changes, implicating an epigenetic mechanism in these pathway disruptions.
Collapse
|
40
|
Choi CS, Hong M, Kim KC, Kim JW, Yang SM, Seung H, Ko MJ, Choi DH, You JS, Shin CY, Bahn GH. Effects of atomoxetine on hyper-locomotive activity of the prenatally valproate-exposed rat offspring. Biomol Ther (Seoul) 2014; 22:406-13. [PMID: 25414770 PMCID: PMC4201219 DOI: 10.4062/biomolther.2014.027] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Revised: 06/23/2014] [Accepted: 07/21/2014] [Indexed: 01/08/2023] Open
Abstract
A substantial proportion of patients with autism spectrum disorder (ASD) display hyperactivity as a comorbid symptom. Exposure to valproic acid (VPA) during pregnancy produces ASD-like core behavioral phenotypes as well as hyperactivity in offspring both in human and experimental animals, which makes it a plausible model to study ASD-related neurobiological processes. In this study, we examined the effects of two of currently available attention defecit hyperactivity disorder (ADHD) medications, methylphenidate (MPH) and atomoxetine (ATX) targeting dopamine and norepinephrine transporters (DAT and NET), respectively, on hyperactive behavior of prenatally VPA-exposed rat offspring. In the prefrontal cortex of VPA exposed rat offspring, both mRNA and protein expression of DAT was increased as compared with control. VPA function as a histone deacetylase inhibitor (HDACi) and chromatin immunoprecipitation experiments demonstrated that the acetylation of histone bound to DAT gene promoter was increased in VPA-exposed rat offspring suggesting epigenetic mechanism of DAT regulation. Similarly, the expression of NET was increased, possibly via increased histone acetylation in prefrontal cortex of VPA-exposed rat offspring. When we treated the VPA-exposed rat offspring with ATX, a NET selective inhibitor, hyperactivity was reversed to control level. In contrast, MPH that inhibits both DAT and NET, did not produce inhibitory effects against hyperactivity. The results suggest that NET abnormalities may underlie the hyperactive phenotype in VPA animal model of ASD. Profiling the pharmacological responsiveness as well as investigating underlying mechanism in multiple models of ASD and ADHD may provide more insights into the neurobiological correlates regulating the behavioral abnormalities.
Collapse
Affiliation(s)
- Chang Soon Choi
- Department of Neuroscience and Center for Neuroscience Research, SMART Institute of Advanced Biomedical Sciences, Seoul 143-701 ; Department of Advanced Translational Medical Science, School of Medicine, Konkuk University, Seoul 143-701
| | - Minha Hong
- Department of Psychiatry, School of Medicine, Dankook University Hospital, Cheonan 330-715 ; Department of Neuropsychiatry, School of Medicine, Kyung Hee University, Seoul 130-702, Republic of Korea
| | - Ki Chan Kim
- Department of Neuroscience and Center for Neuroscience Research, SMART Institute of Advanced Biomedical Sciences, Seoul 143-701 ; Department of Pharmacology, College of Pharmacy, Seoul National University, Seoul 151-742
| | - Ji-Woon Kim
- Department of Neuroscience and Center for Neuroscience Research, SMART Institute of Advanced Biomedical Sciences, Seoul 143-701 ; Department of Advanced Translational Medical Science, School of Medicine, Konkuk University, Seoul 143-701
| | - Sung Min Yang
- Department of Neuroscience and Center for Neuroscience Research, SMART Institute of Advanced Biomedical Sciences, Seoul 143-701 ; Department of Advanced Translational Medical Science, School of Medicine, Konkuk University, Seoul 143-701
| | - Hana Seung
- Department of Neuroscience and Center for Neuroscience Research, SMART Institute of Advanced Biomedical Sciences, Seoul 143-701 ; Department of Advanced Translational Medical Science, School of Medicine, Konkuk University, Seoul 143-701
| | - Mee Jung Ko
- Department of Neuroscience and Center for Neuroscience Research, SMART Institute of Advanced Biomedical Sciences, Seoul 143-701 ; Department of Advanced Translational Medical Science, School of Medicine, Konkuk University, Seoul 143-701
| | - Dong-Hee Choi
- Department of Neuroscience and Center for Neuroscience Research, SMART Institute of Advanced Biomedical Sciences, Seoul 143-701
| | - Jueng Soo You
- Department of Neuroscience and Center for Neuroscience Research, SMART Institute of Advanced Biomedical Sciences, Seoul 143-701
| | - Chan Young Shin
- Department of Neuroscience and Center for Neuroscience Research, SMART Institute of Advanced Biomedical Sciences, Seoul 143-701 ; Department of Advanced Translational Medical Science, School of Medicine, Konkuk University, Seoul 143-701
| | - Geon Ho Bahn
- Department of Neuropsychiatry, School of Medicine, Kyung Hee University, Seoul 130-702, Republic of Korea
| |
Collapse
|
41
|
Nankova BB, Agarwal R, MacFabe DF, La Gamma EF. Enteric bacterial metabolites propionic and butyric acid modulate gene expression, including CREB-dependent catecholaminergic neurotransmission, in PC12 cells--possible relevance to autism spectrum disorders. PLoS One 2014; 9:e103740. [PMID: 25170769 PMCID: PMC4149359 DOI: 10.1371/journal.pone.0103740] [Citation(s) in RCA: 175] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Accepted: 07/01/2014] [Indexed: 12/11/2022] Open
Abstract
Alterations in gut microbiome composition have an emerging role in health and disease including brain function and behavior. Short chain fatty acids (SCFA) like propionic (PPA), and butyric acid (BA), which are present in diet and are fermentation products of many gastrointestinal bacteria, are showing increasing importance in host health, but also may be environmental contributors in neurodevelopmental disorders including autism spectrum disorders (ASD). Further to this we have shown SCFA administration to rodents over a variety of routes (intracerebroventricular, subcutaneous, intraperitoneal) or developmental time periods can elicit behavioral, electrophysiological, neuropathological and biochemical effects consistent with findings in ASD patients. SCFA are capable of altering host gene expression, partly due to their histone deacetylase inhibitor activity. We have previously shown BA can regulate tyrosine hydroxylase (TH) mRNA levels in a PC12 cell model. Since monoamine concentration is known to be elevated in the brain and blood of ASD patients and in many ASD animal models, we hypothesized that SCFA may directly influence brain monoaminergic pathways. When PC12 cells were transiently transfected with plasmids having a luciferase reporter gene under the control of the TH promoter, PPA was found to induce reporter gene activity over a wide concentration range. CREB transcription factor(s) was necessary for the transcriptional activation of TH gene by PPA. At lower concentrations PPA also caused accumulation of TH mRNA and protein, indicative of increased cell capacity to produce catecholamines. PPA and BA induced broad alterations in gene expression including neurotransmitter systems, neuronal cell adhesion molecules, inflammation, oxidative stress, lipid metabolism and mitochondrial function, all of which have been implicated in ASD. In conclusion, our data are consistent with a molecular mechanism through which gut related environmental signals such as increased levels of SCFA's can epigenetically modulate cell function further supporting their role as environmental contributors to ASD.
Collapse
Affiliation(s)
- Bistra B. Nankova
- New York Medical College, Department of Pediatrics/Maria Fareri Children's Hospital, Valhalla, New York, United States of America
- * E-mail:
| | - Raj Agarwal
- New York Medical College, Department of Pediatrics/Maria Fareri Children's Hospital, Valhalla, New York, United States of America
| | - Derrick F. MacFabe
- The Kilee Patchell-Evans Autism Research Group, Departments of Psychology (Neuroscience) and Psychiatry, Division of Developmental Disabilities, The University of Western Ontario, London, Ontario, Canada
| | - Edmund F. La Gamma
- New York Medical College, Department of Pediatrics/Maria Fareri Children's Hospital, Valhalla, New York, United States of America
| |
Collapse
|
42
|
Efficient reprogramming of mouse fibroblasts to neuronal cells including dopaminergic neurons. ScientificWorldJournal 2014; 2014:957548. [PMID: 24991651 PMCID: PMC4058809 DOI: 10.1155/2014/957548] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 05/08/2014] [Indexed: 11/18/2022] Open
Abstract
Somatic cells were directly converted to functional neurons through the use of a combination of transcription factors, including Ascl1, Brn2, and Myt1l. However, a major limitation is the lack of a reliable source of cell-replacement therapy for neurological diseases. Here, we show that a combination of the transcription factors Ascl1 and Nurr1 (AN) and neurotrophic factors including SHH and FGF8b directly reprogrammed embryonic mouse fibroblasts to induced neuronal (iN) cells: pan-neuronal cells and dopaminergic (DA) neurons under our systematic cell culture conditions. Reprogrammed cells showed the morphological properties of neuronal cells. Additionally, cells were analyzed using various markers, including Tuj1 and Map2 for neuronal cells and Lmx1a, Th, Aadc and Vmat2 for DA neurons in our immunostaining and reverse transcription (RT)-PCR experiments. We found that a combination of transcription factors and neurotrophic factors could directly reprogram fibroblasts to neuronal cells including DA neurons. Various types of reprogrammed cells are promising cell sources for cell-based therapy of neurological disorders like Parkinson's disease and spinal cord injury.
Collapse
|
43
|
Jacob J, Ribes V, Moore S, Constable SC, Sasai N, Gerety SS, Martin DJ, Sergeant CP, Wilkinson DG, Briscoe J. Valproic acid silencing of ascl1b/Ascl1 results in the failure of serotonergic differentiation in a zebrafish model of fetal valproate syndrome. Dis Model Mech 2013; 7:107-17. [PMID: 24135485 PMCID: PMC3882053 DOI: 10.1242/dmm.013219] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Fetal valproate syndrome (FVS) is caused by in utero exposure to the drug sodium valproate. Valproate is used worldwide for the treatment of epilepsy, as a mood stabiliser and for its pain-relieving properties. In addition to birth defects, FVS is associated with an increased risk of autism spectrum disorder (ASD), which is characterised by abnormal behaviours. Valproate perturbs multiple biochemical pathways and alters gene expression through its inhibition of histone deacetylases. Which, if any, of these mechanisms is relevant to the genesis of its behavioural side effects is unclear. Neuroanatomical changes associated with FVS have been reported and, among these, altered serotonergic neuronal differentiation is a consistent finding. Altered serotonin homeostasis is also associated with autism. Here we have used a chemical-genetics approach to investigate the underlying molecular defect in a zebrafish FVS model. Valproate causes the selective failure of zebrafish central serotonin expression. It does so by downregulating the proneural gene ascl1b, an ortholog of mammalian Ascl1, which is a known determinant of serotonergic identity in the mammalian brainstem. ascl1b is sufficient to rescue serotonin expression in valproate-treated embryos. Chemical and genetic blockade of the histone deacetylase Hdac1 downregulates ascl1b, consistent with the Hdac1-mediated silencing of ascl1b expression by valproate. Moreover, tonic Notch signalling is crucial for ascl1b repression by valproate. Concomitant blockade of Notch signalling restores ascl1b expression and serotonin expression in both valproate-exposed and hdac1 mutant embryos. Together, these data provide a molecular explanation for serotonergic defects in FVS and highlight an epigenetic mechanism for genome-environment interaction in disease.
Collapse
Affiliation(s)
- John Jacob
- Division of Developmental Biology, MRC National Institute for Medical Research, The Ridgeway, Mill Hill, London, NW7 1AA, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Neuropathology and animal models of autism: genetic and environmental factors. AUTISM RESEARCH AND TREATMENT 2013; 2013:731935. [PMID: 24151553 PMCID: PMC3787615 DOI: 10.1155/2013/731935] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Revised: 08/07/2013] [Accepted: 08/09/2013] [Indexed: 02/05/2023]
Abstract
Autism is a heterogeneous behaviorally defined neurodevelopmental disorder. It is defined by the presence of marked social deficits, specific language abnormalities, and stereotyped repetitive patterns of behavior. Because of the variability in the behavioral phenotype of the disorder among patients, the term autism spectrum disorder has been established. In the first part of this review, we provide an overview of neuropathological findings from studies of autism postmortem brains and identify the cerebellum as one of the key brain regions that can play a role in the autism phenotype. We review research findings that indicate possible links between the environment and autism including the role of mercury and immune-related factors. Because both genes and environment can alter the structure of the developing brain in different ways, it is not surprising that there is heterogeneity in the behavioral and neuropathological phenotypes of autism spectrum disorders. Finally, we describe animal models of autism that occur following insertion of different autism-related genes and exposure to environmental factors, highlighting those models which exhibit both autism-like behavior and neuropathology.
Collapse
|
45
|
Won H, Mah W, Kim E. Autism spectrum disorder causes, mechanisms, and treatments: focus on neuronal synapses. Front Mol Neurosci 2013; 6:19. [PMID: 23935565 PMCID: PMC3733014 DOI: 10.3389/fnmol.2013.00019] [Citation(s) in RCA: 129] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Accepted: 07/16/2013] [Indexed: 12/24/2022] Open
Abstract
Autism spectrum disorder (ASD) is a group of developmental disabilities characterized by impairments in social interaction and communication and restricted and repetitive interests/behaviors. Advances in human genomics have identified a large number of genetic variations associated with ASD. These associations are being rapidly verified by a growing number of studies using a variety of approaches, including mouse genetics. These studies have also identified key mechanisms underlying the pathogenesis of ASD, many of which involve synaptic dysfunctions, and have investigated novel, mechanism-based therapeutic strategies. This review will try to integrate these three key aspects of ASD research: human genetics, animal models, and potential treatments. Continued efforts in this direction should ultimately reveal core mechanisms that account for a larger fraction of ASD cases and identify neural mechanisms associated with specific ASD symptoms, providing important clues to efficient ASD treatment.
Collapse
Affiliation(s)
- Hyejung Won
- Department of Biological Sciences, Korea Advanced Institute of Science and TechnologyDaejeon, South Korea
| | - Won Mah
- Department of Biological Sciences, Korea Advanced Institute of Science and TechnologyDaejeon, South Korea
- Center for Synaptic Brain Dysfunctions, Institute for Basic ScienceDaejeon, South Korea
| | - Eunjoon Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and TechnologyDaejeon, South Korea
- Center for Synaptic Brain Dysfunctions, Institute for Basic ScienceDaejeon, South Korea
| |
Collapse
|
46
|
Trent S, Dean R, Veit B, Cassano T, Bedse G, Ojarikre OA, Humby T, Davies W. Biological mechanisms associated with increased perseveration and hyperactivity in a genetic mouse model of neurodevelopmental disorder. Psychoneuroendocrinology 2013; 38:1370-80. [PMID: 23276394 PMCID: PMC3690523 DOI: 10.1016/j.psyneuen.2012.12.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Revised: 12/03/2012] [Accepted: 12/04/2012] [Indexed: 12/23/2022]
Abstract
Chromosomal deletions at Xp22.3 appear to influence vulnerability to the neurodevelopmental disorders attention deficit hyperactivity disorder (ADHD) and autism. 39,X(Y*)O mice, which lack the murine orthologue of the Xp22.3 ADHD candidate gene STS (encoding steroid sulfatase), exhibit behavioural phenotypes relevant to such disorders (e.g. hyperactivity), elevated hippocampal serotonin (5-HT) levels, and reduced serum levels of dehydroepiandrosterone (DHEA). Here we initially show that 39,X(Y*)O mice are also deficient for the recently-characterised murine orthologue of the Xp22.3 autism candidate gene ASMT (encoding acetylserotonin-O-methyltransferase). Subsequently, to specify potential behavioural correlates of elevated hippocampal 5-HT arising due to the genetic lesion, we compared 39,X(Y*)O MF1 mice to 40,XY MF1 mice on behavioural tasks taxing hippocampal and/or 5-HT function (a 'foraging' task, an object-location task, and the 1-choice serial reaction time task of impulsivity). Although Sts/Asmt deficiency did not influence foraging behaviour, reactivity to familiar objects in novel locations, or 'ability to wait', it did result in markedly increased response rates; these rates correlated with hippocampal 5-HT levels and are likely to index behavioural perseveration, a frequent feature of neurodevelopmental disorders. Additionally, we show that whilst there was no systematic relationship between serum DHEA levels and hippocampal 5-HT levels across 39,X(Y*)O and 40,XY mice, there was a significant inverse linear correlation between serum DHEA levels and activity. Our data suggest that deficiency for genes within Xp22.3 could influence core behavioural features of neurodevelopmental disorders via dissociable effects on hippocampal neurochemistry and steroid hormone levels, and that the mediating neurobiological mechanisms may be investigated in the 39,X(Y*)O model.
Collapse
Affiliation(s)
- Simon Trent
- Behavioural Genetics Group and Neuroscience and Mental Health Research Institute, Schools of Psychology and Medicine, Cardiff University, Cardiff, UK,Institute of Psychological Medicine and Clinical Neurosciences and MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, UK
| | - Rachel Dean
- School of Psychology, Cardiff University, Cardiff, UK
| | - Bonnie Veit
- School of Psychology, Cardiff University, Cardiff, UK
| | - Tommaso Cassano
- Department of Clinical and Experimental Medicine, Medical School, University of Foggia, Foggia, Italy
| | - Gaurav Bedse
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Obah A. Ojarikre
- Division of Stem Cell Biology and Developmental Genetics, MRC National Institute for Medical Research, London, UK
| | - Trevor Humby
- Behavioural Genetics Group and Neuroscience and Mental Health Research Institute, Schools of Psychology and Medicine, Cardiff University, Cardiff, UK,Institute of Psychological Medicine and Clinical Neurosciences and MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, UK
| | - William Davies
- Behavioural Genetics Group and Neuroscience and Mental Health Research Institute, Schools of Psychology and Medicine, Cardiff University, Cardiff, UK,Institute of Psychological Medicine and Clinical Neurosciences and MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, UK,Corresponding author at: Henry Wellcome Building, Heath Park Campus, Cardiff CF14 4XN, UK. Tel.: +44 0 29 2068 7047; fax: +44 0 29 2068 7068.
| |
Collapse
|
47
|
Kinast K, Peeters D, Kolk SM, Schubert D, Homberg JR. Genetic and pharmacological manipulations of the serotonergic system in early life: neurodevelopmental underpinnings of autism-related behavior. Front Cell Neurosci 2013; 7:72. [PMID: 23781172 PMCID: PMC3679613 DOI: 10.3389/fncel.2013.00072] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2013] [Accepted: 04/30/2013] [Indexed: 12/28/2022] Open
Abstract
Serotonin, in its function as neurotransmitter, is well-known for its role in depression, autism and other neuropsychiatric disorders, however, less known as a neurodevelopmental factor. The serotonergic system is one of the earliest to develop during embryogenesis and early changes in serotonin levels can have large consequences for the correct development of specific brain areas. The regulation and functioning of serotonin is influenced by genetic risk factors, such as the serotonin transporter polymorphism in humans. This polymorphism is associated with anxiety-related symptoms, changes in social behavior, and cortical gray and white matter changes also seen in patients suffering from autism spectrum disorders (ASD). The human polymorphism can be mimicked by the knockout of the serotonin transporter in rodents, which are as a model system therefore vital to explore the precise neurobiological mechanisms. Moreover, there are pharmacological challenges influencing serotonin in early life, like prenatal/neonatal exposure to selective serotonin reuptake inhibitors (SSRI) in depressed pregnant women. There is accumulating evidence that this dysregulation of serotonin during critical phases of brain development can lead to ASD-related symptoms in children, and reduced social behavior and increased anxiety in rodents. Furthermore, prenatal valproic acid (VPA) exposure, a mood stabilizing drug which is also thought to interfere with serotonin levels, has the potency to induce ASD-like symptoms and to affect the development of the serotonergic system. Here, we review and compare the neurodevelopmental and behavioral consequences of serotonin transporter gene variation, and prenatal SSRI and VPA exposure in the context of ASD.
Collapse
Affiliation(s)
- Karsten Kinast
- Behavioural Neurogenetics, Department of Cognitive Neuroscience, Centre for Neuroscience, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Nijmegen Medical Centre Nijmegen, Netherlands
| | | | | | | | | |
Collapse
|
48
|
Bath KG, Scharfman HE. Impact of early life exposure to antiepileptic drugs on neurobehavioral outcomes based on laboratory animal and clinical research. Epilepsy Behav 2013; 26:427-39. [PMID: 23305780 PMCID: PMC3925312 DOI: 10.1016/j.yebeh.2012.10.031] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2012] [Accepted: 10/30/2012] [Indexed: 12/13/2022]
Abstract
Epilepsy affects approximately 1% of children under the age of 15, making it a very common neurological disorder in the pediatric population (Russ et al., 2012). In addition, ~0.4-0.8% of all pregnant women have some form of epilepsy (Hauser et al., 1996a,b; Borthen et al., 2009; Krishnamurthy, 2012). Despite the potential deleterious effects of antiepileptic drugs (AEDs) on the developing brain, their use is still required for seizure control in pregnant women (Krishnamurthy, 2012), and they represent the standard approach for treating children with epilepsy (Chu-Shore and Thiele, 2010; Quach et al., 2010; Verrotti et al., 2011). Even when AEDs are effective, there are potential side effects, including cognitive and affective changes or altered sleep and appetite. The consequences of AED exposure in development have been studied extensively (Canger et al., 1999; Modi et al., 2011a,b; Oguni, 2011). Despite intensive study, there is still debate about the long-term consequences of early life AED exposure. Here, we consider the evidence to date that AED exposure, either prenatally or in early postnatal life, has significant adverse effects on the developing brain and incorporate studies of laboratory animals as well as those of patients. We also note the areas of research where greater clarity seems critical in order to make significant advances. A greater understanding of the impact of AEDs on somatic, cognitive and behavioral development has substantial value because it has the potential to inform clinical practice and guide studies aimed at understanding the genetic and molecular bases of comorbid pathologies associated with common treatment regimens. Understanding these effects has the potential to lead to AEDs with fewer side effects. Such advances would expand treatment options, diminish the risk associated with AED exposure in susceptible populations, and improve the quality of life and health outcomes of children with epilepsy and children born to women who took AEDs during pregnancy.
Collapse
Affiliation(s)
- Kevin G. Bath
- Department of Neuroscience, Brown University, Box GL-N, 185 Meeting St., Providence, RI 02912, USA,Corresponding author. (K.G. Bath)
| | - Helen E. Scharfman
- The Nathan Kline Institute for Psychiatric Research, 140 Old Orangeburg Rd., Bldg. 35, Orangeburg, NY 10962, USA,New York University Langone Medical Center, 550 First Ave., New York, NY 10016, USA
| |
Collapse
|
49
|
Oyabu A, Narita M, Tashiro Y. The effects of prenatal exposure to valproic acid on the initial development of serotonergic neurons. Int J Dev Neurosci 2013; 31:202-8. [DOI: 10.1016/j.ijdevneu.2013.01.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Revised: 01/23/2013] [Accepted: 01/23/2013] [Indexed: 10/27/2022] Open
Affiliation(s)
- Akiko Oyabu
- Department of Developmental and Regenerative MedicineMie University Graduate School of MedicineJapan
| | - Masaaki Narita
- Department of Developmental and Regenerative MedicineMie University Graduate School of MedicineJapan
| | - Yasura Tashiro
- Department of Developmental and Regenerative MedicineMie University Graduate School of MedicineJapan
| |
Collapse
|
50
|
Morphology of the facial motor nuclei in a rat model of autism during early development. Int J Dev Neurosci 2012; 31:138-44. [PMID: 23253376 DOI: 10.1016/j.ijdevneu.2012.12.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Revised: 11/22/2012] [Accepted: 12/08/2012] [Indexed: 11/22/2022] Open
Abstract
The development of facial nuclei in animal models of disease is poorly understood, but autism is sometimes associated with facial palsy. In the present study, to investigate migration of facial neurons and initial facial nucleus formation in an animal model of autism, rat embryos were treated with valproic acid (VPA) in utero at embryonic day (E) 9.5 and their facial nuclei were analyzed by in situ hybridization at E13.5, E14.5 and E15.5. Signals for Tbx20, which is expressed in early motor neurons, appeared near the floor plate at the level of the vestibular ganglion and extended caudolaterally, where they became ovoid in shape. This pattern of development was similar between control and VPA-exposed embryos. However, measurements of the migratory pathway and the size of the facial nuclei revealed that exposure to VPA hindered the caudal migration of neurons to the facial nuclei. Signals for cadherin 8, which is expressed in mature facial nuclei, revealed that exposure to VPA caused a significant reduction in the size of the facial nuclei. Our findings provide the first quantitative description of tangential migration and nucleus formation in the developing hindbrain in a rat model of autism.
Collapse
|