51
|
Fueta Y, Sekino Y, Yoshida S, Kanda Y, Ueno S. Prenatal exposure to valproic acid alters the development of excitability in the postnatal rat hippocampus. Neurotoxicology 2018; 65:1-8. [DOI: 10.1016/j.neuro.2018.01.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 12/03/2017] [Accepted: 01/04/2018] [Indexed: 11/16/2022]
|
52
|
Hajizadeh-Zaker R, Ghajar A, Mesgarpour B, Afarideh M, Mohammadi MR, Akhondzadeh S. l-Carnosine As an Adjunctive Therapy to Risperidone in Children with Autistic Disorder: A Randomized, Double-Blind, Placebo-Controlled Trial. J Child Adolesc Psychopharmacol 2018; 28:74-81. [PMID: 29027815 DOI: 10.1089/cap.2017.0026] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
OBJECTIVES This study aimed at investigating the efficacy and tolerability of l-carnosine as an add-on to risperidone in the management of children with autism. METHODS This was a 10-week, randomized, double-blind, placebo-controlled study. Seventy drug-free children aged 4-12 years old with a diagnosis of autism spectrum disorder (ASD), according to the Diagnostic and Statistical Manual of Mental Disorders, fifth edition. (DSM-5) who had an Aberrant Behavior Checklist-Community (ABC-C) scale irritability subscale score of ≥12, entered the study. The patients were randomly assigned to l-carnosine (800 mg/day in 2 divided doses) or placebo in addition to risperidone titrated up to 2 mg/day (based on body weight) for 10 weeks. The children were assessed by using ABC-C at baseline and weeks 5 and 10 post-baseline. The primary outcome measure was the mean change in the ABC-C irritability subscale score, and other subscale scores were defined as secondary outcomes. RESULTS Using the general linear model repeated measures, no significant effect was observed for time × treatment interaction on the irritability subscale scores. However, significant effect was detected on the hyperactivity/noncompliance subscale [F (1.62, 64.96) = 3.53, p-value = 0.044]. No significant improvements were obtained on the lethargy/social withdrawal, stereotypic behavior, and inappropriate speech subscale scores. Significantly greater score reduction in the hyperactivity/noncompliance subscale occurred in the l-carnosine group compared with the placebo group at the end of the trial. Extrapyramidal Symptom Rating Scale Scores and its changes did not differ between the two groups. The frequency of other side effects was not significantly different between the two groups. CONCLUSIONS Although no significant difference was detected on the irritability subscale scores, l-carnosine add-on can improve hyperactivity/noncompliance subscales of the ABC-C rating scale in patients with ASD.
Collapse
Affiliation(s)
- Reihaneh Hajizadeh-Zaker
- 1 Psychiatric Research Center, Roozbeh Hospital, Tehran University of Medical Sciences , Tehran, Iran
| | - Alireza Ghajar
- 1 Psychiatric Research Center, Roozbeh Hospital, Tehran University of Medical Sciences , Tehran, Iran
| | - Bita Mesgarpour
- 2 National Institute for Medical Research Development (NIMAD) , Tehran, Iran
| | - Mohsen Afarideh
- 1 Psychiatric Research Center, Roozbeh Hospital, Tehran University of Medical Sciences , Tehran, Iran
| | - Mohammad-Reza Mohammadi
- 1 Psychiatric Research Center, Roozbeh Hospital, Tehran University of Medical Sciences , Tehran, Iran
| | - Shahin Akhondzadeh
- 1 Psychiatric Research Center, Roozbeh Hospital, Tehran University of Medical Sciences , Tehran, Iran
| |
Collapse
|
53
|
Inui K, Takeuchi N, Sugiyama S, Motomura E, Nishihara M. GABAergic mechanisms involved in the prepulse inhibition of auditory evoked cortical responses in humans. PLoS One 2018; 13:e0190481. [PMID: 29298327 PMCID: PMC5752037 DOI: 10.1371/journal.pone.0190481] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 12/17/2017] [Indexed: 11/30/2022] Open
Abstract
Despite their essential roles in signal processing in the brain, the functions of interneurons currently remain unclear in humans. We recently developed a method using the prepulse inhibition of sensory evoked cortical responses for functional measurements of interneurons. When a sensory feature is abruptly changed in a continuous sensory stimulus, change-related cortical responses are recorded using MEG. By inserting a weak change stimulus (prepulse) before the test change stimulus, it is possible to observe the inhibition of the test response. By manipulating the prepulse–test interval (PTI), several peaks appear in inhibition, suggesting the existence of temporally distinct mechanisms. We herein attempted to separate these components through the oral administration of diazepam and baclofen. The test stimulus and prepulse were an abrupt increase in sound pressure in a continuous click train of 10 and 5 dB, respectively. The results obtained showed that the inhibition at PTIs of 10 and 20 ms was significantly greater with diazepam than with the placebo administration, suggesting increased GABAA-mediated inhibition. Baclofen decreased inhibition at PTIs of 40 and 50 ms, which may have been due to the activation of GABAB autoreceptors. Therefore, the present study separated at least two inhibitory mechanisms pharmacologically.
Collapse
Affiliation(s)
- Koji Inui
- Institute for Developmental Research, Aichi Human Service Center, Kasugai, Japan
- Department of Integrative Physiology, National Institute for Physiological Sciences, Okazaki, Japan
- * E-mail:
| | | | - Shunsuke Sugiyama
- Department of Psychiatry, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Eishi Motomura
- Department of Neuropsychiatry, Mie University Graduate School of Medicine, Tsu, Japan
| | - Makoto Nishihara
- Multidisciplinary Pain Center, Aichi Medical University, Nagakute, Japan
| |
Collapse
|
54
|
Cuevas-Olguin R, Roychowdhury S, Banerjee A, Garcia-Oscos F, Esquivel-Rendon E, Bringas ME, Kilgard MP, Flores G, Atzori M. Cerebrolysin prevents deficits in social behavior, repetitive conduct, and synaptic inhibition in a rat model of autism. J Neurosci Res 2017; 95:2456-2468. [PMID: 28609577 DOI: 10.1002/jnr.24072] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 04/02/2017] [Accepted: 04/05/2017] [Indexed: 12/11/2022]
Abstract
Autism spectrum disorder (ASD) is a syndrome of diverse neuropsychiatric diseases of growing incidence characterized by repetitive conduct and impaired social behavior and communication for which effective pharmacological treatment is still unavailable. While the mechanisms and etiology of ASD are still unknown, a consensus is emerging about the synaptic nature of the syndrome, suggesting a possible avenue for pharmacological treatment with synaptogenic compounds. The peptidic mixture cerebrolysin (CBL) has been successfully used during the last three decades in the treatment of stroke and neurodegenerative disease. Animal experiments indicate that at least one possible mechanism of action of CBL is through neuroprotection and/or synaptogenesis. In the present study, we tested the effect of CBL treatment (daily injection of 2.5 mL/Kg i.p. during 15 days) on a rat model of ASD. This was based on the offspring (43 male and 51 female pups) of a pregnant female rat injected with valproic acid (VPA, 600 mg/Kg) at the embryonic day 12.5, which previous work has shown to display extensive behavioral, as well as synaptic impairment. Comparison between saline vs. CBL-injected VPA animals shows that CBL treatment improves behavioral as well as synaptic impairments, measured by behavioral performance (social interaction, Y-maze, plus-maze), maximal response of inhibitory γ-amino butyric acid type A receptor (GABAA R)-mediated synaptic currents, as well as their kinetic properties and adrenergic and muscarinic modulation. We speculate that CBL might be a viable and effective candidate for pharmacological treatment or co-treatment of ASD patients. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Roberto Cuevas-Olguin
- Facultad de Ciencias, Programa de Biología, Universidad Autónoma de San Luis Potosí, San Luis Potosí, SLP, 78290, México
| | - Swagata Roychowdhury
- National Institute of Children and Human Development, NIH, Bethesda, MD, 20892, USA
| | - Anwesha Banerjee
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, 75080, USA
- Emory School of Medicine, Department of Cell Biology, Emory University, 615 Michael St. WBRB #415, Atlanta, GA 30322, USA
| | | | - Eric Esquivel-Rendon
- Facultad de Ciencias, Programa de Biología, Universidad Autónoma de San Luis Potosí, San Luis Potosí, SLP, 78290, México
| | - María Elena Bringas
- Benemerita Universidad Autónoma de Puebla, Instituto de Fisiología, Puebla Pue., 72000, México
| | - Michael P Kilgard
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Gonzalo Flores
- Benemerita Universidad Autónoma de Puebla, Instituto de Fisiología, Puebla Pue., 72000, México
| | - Marco Atzori
- Facultad de Ciencias, Programa de Biología, Universidad Autónoma de San Luis Potosí, San Luis Potosí, SLP, 78290, México
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, 75080, USA
| |
Collapse
|
55
|
Increased GABAA receptor binding in amygdala after prenatal administration of valproic acid to rats. Acta Neuropsychiatr 2017; 29:309-314. [PMID: 27938419 DOI: 10.1017/neu.2016.59] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Prenatal exposure to valproic acid (VPA) enhances the risk for later development of autism spectrum disorders (ASD). An altered gamma-aminobutyric acid (GABA) system may be a key factor in ASD. Here we investigated possible changes in the GABA system in rats exposed to a low dose of prenatal VPA. METHOD We performed autoradiography with [3H]muscimol, (a GABAA receptor agonist), and [11C]Ro15-4513 (a partial agonist of the GABAA α1+5 receptor subtypes), in brain sections containing amygdala, thalamus and hippocampus of rats treated prenatally with 20 mg/kg VPA or saline from the 12th day of gestation. Result Prenatal VPA significantly increased [11C]Ro15-4513 binding in the left amygdala compared with controls (p<0.05). This difference was not observed in the hippocampus, thalamus or right amygdala. No differences were observed in [3H]muscimol binding. CONCLUSION We observed an asymmetric increase in GABAA receptor binding. Disturbances in the GABAA receptor system have also been detected in human autism with [11C]Ro15-4513.
Collapse
|
56
|
Li Y, Zhou Y, Peng L, Zhao Y. Reduced protein expressions of cytomembrane GABA ARβ3 at different postnatal developmental stages of rats exposed prenatally to valproic acid. Brain Res 2017; 1671:33-42. [PMID: 28641949 DOI: 10.1016/j.brainres.2017.06.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 05/15/2017] [Accepted: 06/13/2017] [Indexed: 11/20/2022]
Abstract
Decreased inhibition plays an extremely important role in pathogenesis of autism spectrum disorder (ASD). Therefore, we aimed to determine whether expression levels of the γ-aminobutyric acid type A receptor β3 subunit (GABAARβ3), K+-Cl- cotransporter 2 (KCC2), and Na+-K+-Cl- cotransporter 1 (NKCC1) related to inhibition transmission are changed in a sodium valproate-induced rat model of ASD. Decreased expression levels of membrane GABAARβ3 (m-GABAARβ3) and KCC2 as well as increased endocytosis of GABAARs were found in the model group. However, there were no significant differences in expression of total GABAARβ3 and NKCC1 between the control and model groups. In addition, we observed growth retardation, impaired spatial memory, limited exploration, increased anxiety, and reduced sociability in the model group. These results suggest alterations in m-GABAARβ3 levels, KCC2 levels, and trafficking of GABAARs in rats prenatally exposed to valproic acid and advance our understanding of the pathogenesis of ASD.
Collapse
Affiliation(s)
- Yixin Li
- Department of Pathology, Chongqing Medical University, Chongqing, People's Republic of China; Institute of Neuroscience, Chongqing Medical University, Chongqing, People's Republic of China; Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing, People's Republic of China
| | - Yang Zhou
- Department of Pathology, Chongqing Medical University, Chongqing, People's Republic of China; Institute of Neuroscience, Chongqing Medical University, Chongqing, People's Republic of China; Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing, People's Republic of China
| | - Li Peng
- Department of Pathology, Chongqing Medical University, Chongqing, People's Republic of China; Institute of Neuroscience, Chongqing Medical University, Chongqing, People's Republic of China; Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing, People's Republic of China
| | - Yong Zhao
- Department of Pathology, Chongqing Medical University, Chongqing, People's Republic of China; Institute of Neuroscience, Chongqing Medical University, Chongqing, People's Republic of China; Key Laboratory of Neurobiology, Chongqing Medical University, Chongqing, People's Republic of China.
| |
Collapse
|
57
|
Brizuela M, Blizzard CA, Chuckowree JA, Pitman KA, Young KM, Dickson T. Mild Traumatic Brain Injury Leads to Decreased Inhibition and a Differential Response of Calretinin Positive Interneurons in the Injured Cortex. J Neurotrauma 2017; 34:2504-2517. [DOI: 10.1089/neu.2017.4977] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Affiliation(s)
- Mariana Brizuela
- Menzies Institute for Medical Research, University of Tasmania, Tasmania, Australia
| | | | - Jyoti A. Chuckowree
- Menzies Institute for Medical Research, University of Tasmania, Tasmania, Australia
| | - Kimberley A. Pitman
- Menzies Institute for Medical Research, University of Tasmania, Tasmania, Australia
| | - Kaylene M. Young
- Menzies Institute for Medical Research, University of Tasmania, Tasmania, Australia
| | - Tracey Dickson
- Menzies Institute for Medical Research, University of Tasmania, Tasmania, Australia
| |
Collapse
|
58
|
Port RG, Gajewski C, Krizman E, Dow HC, Hirano S, Brodkin ES, Carlson GC, Robinson MB, Roberts TPL, Siegel SJ. Protocadherin 10 alters γ oscillations, amino acid levels, and their coupling; baclofen partially restores these oscillatory deficits. Neurobiol Dis 2017; 108:324-338. [PMID: 28844789 DOI: 10.1016/j.nbd.2017.08.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 08/06/2017] [Accepted: 08/22/2017] [Indexed: 11/20/2022] Open
Abstract
Approximately one in 45 children have been diagnosed with Autism Spectrum Disorder (ASD), which is characterized by social/communication impairments. Recent studies have linked a subset of familial ASD to mutations in the Protocadherin 10 (Pcdh10) gene. Additionally, Pcdh10's expression pattern, as well as its known role within protein networks, implicates the gene in ASD. Subsequently, the neurobiology of mice heterozygous for Pcdh10 (Pcdh10+/-) has been investigated as a proxy for ASD. Male Pcdh10+/- mice have demonstrated sex-specific deficits in social behavior, recapitulating the gender bias observed in ASD. Furthermore, in vitro slice preparations of these Pcdh10+/- mice demonstrate selective decreases to high frequency electrophysiological responses, mimicking clinical observations. The direct in vivo ramifications of such decreased in vitro high frequency responses are unclear. As such, Pcdh10+/- mice and their wild-type (WT) littermates underwent in vivo electrocorticography (ECoG), as well as ex vivo amino acid concentration quantification using High Performance Liquid Chromatography (HPLC). Similar to the previously observed reductions to in vitro high frequency electrophysiological responses in Pcdh10+/- mice, male Pcdh10+/- mice exhibited reduced gamma-band (30-80Hz), but not lower frequency (10 and 20Hz), auditory steady state responses (ASSR). In addition, male Pcdh10+/- mice exhibited decreased signal-to-noise-ratio (SNR) for high gamma-band (60-100Hz) activity. These gamma-band perturbations for both ASSR and SNR were not observed in females. Administration of a GABAB agonist remediated these electrophysiological alterations among male Pcdh10+/-mice. Pcdh10+/- mice demonstrated increased concentrations of GABA and glutamine. Of note, a correlation of auditory gamma-band responses with underlying GABA concentrations was observed in WT mice. This correlation was not present in Pcdh10+/- mice. This study demonstrates the role of Pcdh10 in the regulation of excitatory-inhibitory balance as a function of GABA in ASD.
Collapse
Affiliation(s)
- Russell G Port
- Department of Psychiatry, University of Pennsylvania Perelman, School of Medicine, Philadelphia, PA 19104, USA; Lurie Family Foundations MEG Imaging Center, Department of Radiology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Christopher Gajewski
- Department of Psychiatry, University of Pennsylvania Perelman, School of Medicine, Philadelphia, PA 19104, USA
| | - Elizabeth Krizman
- Children's Hospital of Philadelphia Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Pediatric, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Holly C Dow
- Department of Psychiatry, University of Pennsylvania Perelman, School of Medicine, Philadelphia, PA 19104, USA
| | - Shinji Hirano
- Department of Cell Biology, Kansai Medical University, 2-5-1 Shinmachi, Hirakata, Osaka 573-1010, Japan
| | - Edward S Brodkin
- Department of Psychiatry, University of Pennsylvania Perelman, School of Medicine, Philadelphia, PA 19104, USA
| | - Gregory C Carlson
- Department of Psychiatry, University of Pennsylvania Perelman, School of Medicine, Philadelphia, PA 19104, USA
| | - Michael B Robinson
- Children's Hospital of Philadelphia Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Pediatric, University of Pennsylvania, Philadelphia, PA 19104, USA; Systems Pharmacology and Experimental Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Timothy P L Roberts
- Lurie Family Foundations MEG Imaging Center, Department of Radiology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Steven J Siegel
- Department of Psychiatry, University of Pennsylvania Perelman, School of Medicine, Philadelphia, PA 19104, USA.
| |
Collapse
|
59
|
Ben-Ari Y. NKCC1 Chloride Importer Antagonists Attenuate Many Neurological and Psychiatric Disorders. Trends Neurosci 2017; 40:536-554. [PMID: 28818303 DOI: 10.1016/j.tins.2017.07.001] [Citation(s) in RCA: 148] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 07/03/2017] [Accepted: 07/10/2017] [Indexed: 12/23/2022]
Abstract
In physiological conditions, adult neurons have low intracellular Cl- [(Cl-)I] levels underlying the γ-aminobutyric acid (GABA)ergic inhibitory drive. In contrast, neurons have high (Cl-)I levels and excitatory GABA actions in a wide range of pathological conditions including spinal cord lesions, chronic pain, brain trauma, cerebrovascular infarcts, autism, Rett and Down syndrome, various types of epilepsies, and other genetic or environmental insults. The diuretic highly specific NKCC1 chloride importer antagonist bumetanide (PubChem CID: 2461) efficiently restores low (Cl-)I levels and attenuates many disorders in experimental conditions and in some clinical trials. Here, I review the mechanisms of action, therapeutic effects, promises, and pitfalls of bumetanide.
Collapse
Affiliation(s)
- Yehezkel Ben-Ari
- New INMED, Aix-Marseille University, Campus Scientifique de Luminy, Marseilles, France.
| |
Collapse
|
60
|
Barrett CE, Hennessey TM, Gordon KM, Ryan SJ, McNair ML, Ressler KJ, Rainnie DG. Developmental disruption of amygdala transcriptome and socioemotional behavior in rats exposed to valproic acid prenatally. Mol Autism 2017; 8:42. [PMID: 28775827 PMCID: PMC5539636 DOI: 10.1186/s13229-017-0160-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 07/19/2017] [Indexed: 12/12/2022] Open
Abstract
Background The amygdala controls socioemotional behavior and has consistently been implicated in the etiology of autism spectrum disorder (ASD). Precocious amygdala development is commonly reported in ASD youth with the degree of overgrowth positively correlated to the severity of ASD symptoms. Prenatal exposure to VPA leads to an ASD phenotype in both humans and rats and has become a commonly used tool to model the complexity of ASD symptoms in the laboratory. Here, we examined abnormalities in gene expression in the amygdala and socioemotional behavior across development in the valproic acid (VPA) rat model of ASD. Methods Rat dams received oral gavage of VPA (500 mg/kg) or saline daily between E11 and 13. Socioemotional behavior was tracked across development in both sexes. RNA sequencing and proteomics were performed on amygdala samples from male rats across development. Results Effects of VPA on time spent in social proximity and anxiety-like behavior were sex dependent, with social abnormalities presenting in males and heightened anxiety in females. Across time VPA stunted developmental and immune, but enhanced cellular death and disorder, pathways in the amygdala relative to saline controls. At postnatal day 10, gene pathways involved in nervous system and cellular development displayed predicted activations in prenatally exposed VPA amygdala samples. By juvenile age, however, transcriptomic and proteomic pathways displayed reductions in cellular growth and neural development. Alterations in immune pathways, calcium signaling, Rho GTPases, and protein kinase A signaling were also observed. Conclusions As behavioral, developmental, and genomic alterations are similar to those reported in ASD, these results lend support to prenatal exposure to VPA as a useful tool for understanding how developmental insults to molecular pathways in the amygdala give rise to ASD-related syndromes. Electronic supplementary material The online version of this article (doi:10.1186/s13229-017-0160-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Catherine E Barrett
- Silvio O. Conte Center for Oxytocin and Social Cognition, Division of Behavioral Neuroscience and Psychiatric Disorders, Yerkes National Primate Research Center, Emory University, 954 Gatewood Rd, 30329 Atlanta, GA USA.,Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, 30329 Atlanta, GA USA
| | - Thomas M Hennessey
- Silvio O. Conte Center for Oxytocin and Social Cognition, Division of Behavioral Neuroscience and Psychiatric Disorders, Yerkes National Primate Research Center, Emory University, 954 Gatewood Rd, 30329 Atlanta, GA USA.,Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, 30329 Atlanta, GA USA
| | - Katelyn M Gordon
- Silvio O. Conte Center for Oxytocin and Social Cognition, Division of Behavioral Neuroscience and Psychiatric Disorders, Yerkes National Primate Research Center, Emory University, 954 Gatewood Rd, 30329 Atlanta, GA USA.,Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, 30329 Atlanta, GA USA
| | - Steve J Ryan
- Silvio O. Conte Center for Oxytocin and Social Cognition, Division of Behavioral Neuroscience and Psychiatric Disorders, Yerkes National Primate Research Center, Emory University, 954 Gatewood Rd, 30329 Atlanta, GA USA.,Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, 30329 Atlanta, GA USA
| | - Morgan L McNair
- Silvio O. Conte Center for Oxytocin and Social Cognition, Division of Behavioral Neuroscience and Psychiatric Disorders, Yerkes National Primate Research Center, Emory University, 954 Gatewood Rd, 30329 Atlanta, GA USA.,Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, 30329 Atlanta, GA USA
| | - Kerry J Ressler
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA 02478 USA
| | - Donald G Rainnie
- Silvio O. Conte Center for Oxytocin and Social Cognition, Division of Behavioral Neuroscience and Psychiatric Disorders, Yerkes National Primate Research Center, Emory University, 954 Gatewood Rd, 30329 Atlanta, GA USA.,Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, 30329 Atlanta, GA USA
| |
Collapse
|
61
|
Bozzi Y, Provenzano G, Casarosa S. Neurobiological bases of autism-epilepsy comorbidity: a focus on excitation/inhibition imbalance. Eur J Neurosci 2017; 47:534-548. [PMID: 28452083 DOI: 10.1111/ejn.13595] [Citation(s) in RCA: 171] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 03/18/2017] [Accepted: 04/21/2017] [Indexed: 12/13/2022]
Abstract
Autism spectrum disorders (ASD) and epilepsy are common neurological diseases of childhood, with an estimated incidence of approximately 0.5-1% of the worldwide population. Several genetic, neuroimaging and neuropathological studies clearly showed that both ASD and epilepsy have developmental origins and a substantial degree of heritability. Most importantly, ASD and epilepsy frequently coexist in the same individual, suggesting a common neurodevelopmental basis for these disorders. Genome-wide association studies recently allowed for the identification of a substantial number of genes involved in ASD and epilepsy, some of which are mutated in syndromes presenting both ASD and epilepsy clinical features. At the cellular level, both preclinical and clinical studies indicate that the different genetic causes of ASD and epilepsy may converge to perturb the excitation/inhibition (E/I) balance, due to the dysfunction of excitatory and inhibitory circuits in various brain regions. Metabolic and immune dysfunctions, as well as environmental causes also contribute to ASD pathogenesis. Thus, an E/I imbalance resulting from neurodevelopmental deficits of multiple origins might represent a common pathogenic mechanism for both diseases. Here, we will review the most significant studies supporting these hypotheses. A deeper understanding of the molecular and cellular determinants of autism-epilepsy comorbidity will pave the way to the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Yuri Bozzi
- Neurodevelopmental Disorders Research Group, Centre for Mind/Brain Sciences, University of Trento, via Sommarive 9, 38123, Povo, Trento, Italy.,CNR Neuroscience Institute, Pisa, Italy
| | - Giovanni Provenzano
- Laboratory of Molecular Neuropathology, Centre for Integrative Biology, University of Trento, Trento, Italy
| | - Simona Casarosa
- CNR Neuroscience Institute, Pisa, Italy.,Laboratory of Neural Development and Regeneration, Centre for Integrative Biology, University of Trento, Trento, Italy
| |
Collapse
|
62
|
Cai Y, Wang L, Xiao R, Li X, He X, Gao J, Xu H, Fan X. Autism-like behavior in the BTBR mouse model of autism is improved by propofol. Neuropharmacology 2017; 118:175-187. [PMID: 28341205 DOI: 10.1016/j.neuropharm.2017.03.021] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Revised: 02/28/2017] [Accepted: 03/20/2017] [Indexed: 01/01/2023]
Abstract
Autism spectrum disorder (ASD) is a developmental disorder that is characterized by symptoms of impaired social interactions, restricted interests and repetitive behaviors. Recent studies in humans and animal-models suggest that reduced GABAergic neurotransmission in the brain may underlie autism-related behavioral symptoms. It has been shown that propofol, a commonly used anesthetic, facilitates γ-aminobutyric acid-mediated inhibitory synaptic transmission. The present study investigated whether propofol improved autistic phenotypes in BTBR T + Itpr3tf/J (BTBR) mice, a model of idiopathic autism. We found that i.p. injection of propofol in BTBR mice significantly improved aspects of social approach and repetitive behaviors without affecting reciprocal social interactions and without any detrimental effects in C57BL/6J mice. The ability of propofol to improve autistic phenotypes in BTBR mice through GABAergic neurotransmission suggests a potential pharmacological target for interventions to treat symptoms of autism.
Collapse
Affiliation(s)
- Yulong Cai
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Chongqing, 400038, PR China
| | - Lian Wang
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Chongqing, 400038, PR China
| | - Rui Xiao
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Chongqing, 400038, PR China
| | - Xin Li
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Chongqing, 400038, PR China
| | - Xie He
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Chongqing, 400038, PR China
| | - Junwei Gao
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Chongqing, 400038, PR China
| | - Haiwei Xu
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University, Chongqing, 400038, PR China
| | - Xiaotang Fan
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Chongqing, 400038, PR China.
| |
Collapse
|
63
|
Cuevas-Olguin R, Esquivel-Rendon E, Vargas-Mireles J, Garcia-Oscos F, Miranda-Morales M, Salgado H, Rose-John S, Atzori M. Interleukin 6 trans-signaling regulates basal synaptic transmission and sensitivity to pentylenetetrazole-induced seizures in mice. Synapse 2017; 71. [PMID: 28481031 DOI: 10.1002/syn.21984] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 04/30/2017] [Accepted: 05/01/2017] [Indexed: 11/12/2022]
Abstract
The pro-inflammatory cytokine interleukin 6 (IL-6) interacts with the central nervous system in a largely unknown manner. We used a genetically modified mouse strain (GFAP-sgp130Fc, TG) and wild type (WT) mice to determine whether IL-6 trans-signaling contributes to basal properties of synaptic transmission. Postsynaptic currents (PSCs) were studied by patch-clamp recording in cortical layer 5 of a mouse prefrontal cortex brain slice preparation. TG and WT animals displayed differences mainly (but not exclusively) in excitatory synaptic responses. The frequency of both action potential-independent (miniature) and action potential-dependent (spontaneous) excitatory PSCs (EPSCs) were higher for TG vs. WT animals. No differences were observed in inhibitory miniature, spontaneous, or tonic inhibitory currents. The pair pulse ratio (PPR) of electrically evoked inhibitory as well as of excitatory PSCs were also larger in TG animals vs. WT ones, while no changes were detected in electrically evoked excitatory-inhibitory synaptic ratio (eEPSC/eIPSC), nor in the ratio between the amino-propionic acid receptor (AMPAR)-mediated and N-methyl D aspartate-R (NMDAR)-mediated components of eEPSCs (IAMPA /INMDA ). Evoked IPSC rise times were shorter for TG vs. WT animals. We also compared the sensitivity of TG and WT animals to pentylenetetrazole (PTZ)-induced seizures. We found that TG animals were more sensitive to PTZ injections, as they displayed longer and more severe seizures. We conclude that the absence of basal IL-6 trans-signaling contributes to increase the basal excitability of the central nervous system, at the system level as well at the synaptic level, at least in the prefrontal cortex.
Collapse
Affiliation(s)
- Roberto Cuevas-Olguin
- Faculty of Science, Universidad Autónoma de San Luis Potosí, Av. Salvador Nava S/N, San Luis Potosí, San Luis Potosí, 78290, México
| | - Eric Esquivel-Rendon
- Faculty of Science, Universidad Autónoma de San Luis Potosí, Av. Salvador Nava S/N, San Luis Potosí, San Luis Potosí, 78290, México
| | - Jorge Vargas-Mireles
- Faculty of Science, Universidad Autónoma de San Luis Potosí, Av. Salvador Nava S/N, San Luis Potosí, San Luis Potosí, 78290, México
| | - Francisco Garcia-Oscos
- Department of Neuroscience, Southwestern University, 5323 Harry Hines Boulevard, Dallas, Texas, 75390
| | - Marcela Miranda-Morales
- Faculty of Science, Universidad Autónoma de San Luis Potosí, Av. Salvador Nava S/N, San Luis Potosí, San Luis Potosí, 78290, México
| | - Humberto Salgado
- Centro de Investigaciones Regionales Hideyo Noguchi, Universidad Autonoma de Yucatan, Avenida Itzáes número 490 por calle 59, colonia Centro, Mérida Yucatán, México. CP 97000
| | - Stefan Rose-John
- Department of Biochemistry, Christian Albrecht Universitet, Christian-Albrechts-Platz 4, Kiel, 24118, Germany
| | - Marco Atzori
- Faculty of Science, Universidad Autónoma de San Luis Potosí, Av. Salvador Nava S/N, San Luis Potosí, San Luis Potosí, 78290, México.,School of Behavioral and Brain Sciences, University of Texas, 800 West Campbell Road, Richardson, Dallas, Texas, 75080
| |
Collapse
|
64
|
Lauber E, Filice F, Schwaller B. Prenatal Valproate Exposure Differentially Affects Parvalbumin-Expressing Neurons and Related Circuits in the Cortex and Striatum of Mice. Front Mol Neurosci 2016; 9:150. [PMID: 28066177 PMCID: PMC5174119 DOI: 10.3389/fnmol.2016.00150] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 12/05/2016] [Indexed: 12/21/2022] Open
Abstract
Autism spectrum disorders (ASD) comprise a number of heterogeneous neurodevelopmental diseases characterized by core behavioral symptoms in the domains of social interaction, language/communication and repetitive or stereotyped patterns of behavior. In utero exposure to valproic acid (VPA) has evolved as a highly recognized rodent ASD model due to the robust behavioral phenotype observed in the offspring and the proven construct-, face- and predictive validity of the model. The number of parvalbumin-immunoreactive (PV+) GABAergic interneurons has been consistently reported to be decreased in human ASD subjects and in ASD animal models. The presumed loss of this neuron subpopulation hereafter termed Pvalb neurons and/or PV deficits were proposed to result in an excitation/inhibition imbalance often observed in ASD. Importantly, loss of Pvalb neurons and decreased/absent PV protein levels have two fundamentally different consequences. Thus, Pvalb neurons were investigated in in utero VPA-exposed male ("VPA") mice in the striatum, medial prefrontal cortex (mPFC) and somatosensory cortex (SSC), three ASD-associated brain regions. Unbiased stereology of PV+ neurons and Vicia Villosa Agglutinin-positive (VVA+) perineuronal nets, which specifically enwrap Pvalb neurons, was carried out. Analyses of PV protein expression and mRNA levels for Pvalb, Gad67, Kcnc1, Kcnc2, Kcns3, Hcn1, Hcn2, and Hcn4 were performed. We found a ∼15% reduction in the number of PV+ cells and decreased Pvalb mRNA and PV protein levels in the striatum of VPA mice compared to controls, while the number of VVA+ cells was unchanged, indicating that Pvalb neurons were affected at the level of the transcriptome. In selected cortical regions (mPFC, SSC) of VPA mice, no quantitative loss/decrease of PV+ cells was observed. However, expression of Kcnc1, coding for the voltage-gated potassium channel Kv3.1 specifically expressed in Pvalb neurons, was decreased by ∼40% in forebrain lysates of VPA mice. Moreover, hyperpolarization-activated cyclic nucleotide-gated channel (HCN) 1 expression was increased by ∼40% in the same samples from VPA mice. We conclude that VPA leads to alterations that are brain region- and gene-specific including Pvalb, Kcnc1, and Hcn1 possibly linked to homeostatic mechanisms. Striatal PV down-regulation appears as a common feature in a subset of genetic (Shank3B-/-) and environmental ASD models.
Collapse
Affiliation(s)
| | | | - Beat Schwaller
- Anatomy, Department of Medicine, University of FribourgFribourg, Switzerland
| |
Collapse
|
65
|
Brondino N, Fusar-Poli L, Panisi C, Damiani S, Barale F, Politi P. Pharmacological Modulation of GABA Function in Autism Spectrum Disorders: A Systematic Review of Human Studies. J Autism Dev Disord 2016; 46:825-39. [PMID: 26443675 DOI: 10.1007/s10803-015-2619-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Autism spectrum disorders are an emerging health problem worldwide, but little is known about their pathogenesis. It has been hypothesized that autism may result from an imbalance between excitatory glutamatergic and inhibitory GABAergic pathways. Commonly used medications such as valproate, acamprosate, and arbaclofen may act on the GABAergic system and be a potential treatment for people with ASD. The present systematic review aimed at evaluating the state-of-the-art of clinical trials of GABA modulators in autism. To date there is insufficient evidence to suggest the use of these drugs in autistic subjects, even if data are promising. Of note, short-term use of all the reviewed medications appears to be safe. Future well designed trials are needed to elucidate these preliminary findings.
Collapse
Affiliation(s)
- Natascia Brondino
- Department of Brain and Behavioral Sciences, University of Pavia, Via Bassi 21, 27100, Pavia, Italy.
| | - Laura Fusar-Poli
- Department of Brain and Behavioral Sciences, University of Pavia, Via Bassi 21, 27100, Pavia, Italy
| | - Cristina Panisi
- Department of Brain and Behavioral Sciences, University of Pavia, Via Bassi 21, 27100, Pavia, Italy
| | - Stefano Damiani
- Department of Brain and Behavioral Sciences, University of Pavia, Via Bassi 21, 27100, Pavia, Italy
| | - Francesco Barale
- Department of Brain and Behavioral Sciences, University of Pavia, Via Bassi 21, 27100, Pavia, Italy
| | - Pierluigi Politi
- Department of Brain and Behavioral Sciences, University of Pavia, Via Bassi 21, 27100, Pavia, Italy
| |
Collapse
|
66
|
Maternal Exposure to Valproic Acid Primarily Targets Interneurons Followed by Late Effects on Neurogenesis in the Hippocampal Dentate Gyrus in Rat Offspring. Neurotox Res 2016; 31:46-62. [DOI: 10.1007/s12640-016-9660-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 07/21/2016] [Accepted: 08/11/2016] [Indexed: 12/21/2022]
|
67
|
A single low dose of valproic acid in late prenatal life alters postnatal behavior and glutamic acid decarboxylase levels in the mouse. Behav Brain Res 2016; 314:190-8. [PMID: 27498245 DOI: 10.1016/j.bbr.2016.08.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 08/01/2016] [Accepted: 08/03/2016] [Indexed: 12/25/2022]
Abstract
RATIONALE Rodents exposed to valproic acid (VPA) in prenatal life exhibit post-natal characteristics analogous to autism spectrum disorder (ASD). Many previous studies used relatively high doses of VPA during early pregnancy, potentially confounding interpretation because the offspring are the 'survivors' of a toxic insult. Low dose or late gestation exposure has not been widely studied. OBJECTIVES We examined the behavioral sequelae of late gestation exposure to low dose VPA in the mouse. We also examined postnatal levels of glutamic acid decarboxylase (GAD65 and GAD67) as markers for GABA neurons, because GABA pathology and subsequent excitatory/inhibitory imbalance is strongly implicated in ASD. METHODS Pregnant C57BL/6N mice received a single subcutaneous injection of 100 or 200mg/kg on gestation day 17. The control group received a saline injection on the same day. The offspring were tested in a battery of behavioral tests in adolescence and adulthood. Six brain regions were harvested and GAD65 and GAD67 were measured by western blotting. RESULTS Compared to saline-exposed controls, adult mice exposed to prenatal VPA had impaired novel object exploration and fear conditioning anomalies. GAD67 was decreased in midbrain, olfactory bulb, prefrontal cortex and increased in cerebellum, hippocampus and striatum; GAD65 was decreased in all 6 regions. CONCLUSIONS Our results suggest that a low dose of VPA in late pregnancy has persistent effects on brain development, and in particular the GABA system, which may be relevant to ASD. Further attention to the impact of gestation time and dose of exposure in VPA-induced ASD models is encouraged.
Collapse
|
68
|
Olexová L, Štefánik P, Kršková L. Increased anxiety-like behaviour and altered GABAergic system in the amygdala and cerebellum of VPA rats - An animal model of autism. Neurosci Lett 2016; 629:9-14. [PMID: 27353514 DOI: 10.1016/j.neulet.2016.06.035] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 06/01/2016] [Accepted: 06/20/2016] [Indexed: 02/06/2023]
Abstract
Anxiety is one of the associated symptoms of autism spectrum disorder. According to the literature, increases in anxiety are accompanied by GABAergic system deregulation. The aim of our study, performed using an animal model of autism in the form of rats prenatally treated with valproic acid (VPA rats), was to investigate changes in anxiety-like behaviour and the gene expression of molecules that control levels of the inhibitory neurotransmitter γ-aminobutyric acid (GABA) in the brain. Anxiety-like behaviours were investigated using zone preferences in the open field test. The levels of the 65 and 67kDa enzymes of l-glutamic acid decarboxylase (GAD) mRNAs and type 1 GABA transporter (GAT1) were evaluated in the amygdala, as well as GABA producing enzymes in the cortex layer of the cerebellum. Our research showed that adult VPA rats spent less time in the inner zone of the testing chamber and more time in the outer zone of the testing chamber in the open field test. We also found that adult VPA rats had increased expression of GAT1 in the amygdala, as well as decreased levels of GAD65 and GAD67 mRNA in the cerebellum compared to control animals. These findings support the existence of a relationship between increased anxiety-like behaviour and changes in the regulation of the GABAergic system in VPA rats.
Collapse
Affiliation(s)
- Lucia Olexová
- Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University in Bratislava, Slovakia.
| | - Peter Štefánik
- Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University in Bratislava, Slovakia
| | - Lucia Kršková
- Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University in Bratislava, Slovakia
| |
Collapse
|
69
|
Distinct Defects in Synaptic Differentiation of Neocortical Neurons in Response to Prenatal Valproate Exposure. Sci Rep 2016; 6:27400. [PMID: 27264355 PMCID: PMC4893673 DOI: 10.1038/srep27400] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 05/18/2016] [Indexed: 01/17/2023] Open
Abstract
Autism spectrum disorders (ASDs) are a heterogeneous group of neurodevelopmental disorders characterized by impairments in social interactions and stereotyped behaviors. Valproic acid (VPA) is frequently used to treat epilepsy and bipolar disorders. When taken during pregnancy, VPA increases the risk of the unborn child to develop an ASD. In rodents, in utero VPA exposure can precipitate behavioral phenotypes related to ASD in the offspring. Therefore, such rodent models may allow for identification of synaptic pathophysiology underlying ASD risk. Here, we systematically probed alterations in synaptic proteins that might contribute to autism-related behavior in the offspring of in utero VPA-exposed mice. Moreover, we tested whether direct VPA exposure of cultured neocortical neurons may recapitulate the molecular alterations seen in vivo. VPA-exposed neurons in culture exhibit a significant increase in the number of glutamatergic synapses accompanied by a significant decrease in the number of GABAergic synapses. This shift in excitatory/inhibitory balance results in substantially increased spontaneous activity in neuronal networks arising from VPA-exposed neurons. Pharmacological experiments demonstrate that the alterations in GABAergic and glutamatergic synaptic proteins and structures are largely caused by inhibition of histone deacetylases. Therefore, our study highlights an epigenetic mechanism underlying the synaptic pathophysiology in this ASD model.
Collapse
|
70
|
Analysis of Extracellular Nucleotide Metabolism in Adult Zebrafish After Embryological Exposure to Valproic Acid. Mol Neurobiol 2016; 54:3542-3553. [PMID: 27189619 DOI: 10.1007/s12035-016-9917-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2016] [Accepted: 05/03/2016] [Indexed: 10/21/2022]
Abstract
Autism is a neurodevelopmental disorder characterized by symptoms related to stereotyped movements, deficits in social interaction, impaired communication, anxiety, hyperactivity, and the presence of restricted interests. Evidence indicates an important role of extracellular ATP and adenosine as signaling molecules in autism. ATP hydrolysis by ectonucleotidases is an important source of adenosine, and adenosine deaminase (ADA) contributes to the control of the nucleoside concentrations. Considering zebrafish is an animal model that may contribute towards to understanding the mechanisms that underlie social behavior, we investigated the purinergic signaling in a model of embryological exposure to valproic acid (VPA) that induces social interaction deficit in adult zebrafish. We demonstrated embryological exposure to VPA did not change ATP and ADP hydrolysis in zebrafish at 120 dpf, and the cytosolic (soluble) ADA activity was not altered. However, we observed an increase of AMP hydrolysis (12.5 %) whereas the ecto-ADA activity was decreased (19.2 %) in adult zebrafish submitted to embryological exposure to VPA. Quantitative reverse transcription PCR (RT-PCR) analysis showed changes on ntpd8, ADA 2.1, and A2a1 mRNA transcript levels. Brain ATP metabolism showed a rapid catabolism of ATP and ADP, whereas the extracellular metabolism of AMP and adenosine (ADO) occurred slowly. We demonstrated that embryological exposure to VPA altered biochemical and molecular parameters related to purinergic system in adult zebrafish. These findings indicate that the enzyme activities involved in the control of ATP and adenosine levels may be involved in the pathophysiological mechanisms of diseases related to the impairment of social interaction, such as autism.
Collapse
|
71
|
Port RG, Edgar JC, Ku M, Bloy L, Murray R, Blaskey L, Levy SE, Roberts TPL. Maturation of auditory neural processes in autism spectrum disorder - A longitudinal MEG study. Neuroimage Clin 2016; 11:566-577. [PMID: 27158589 PMCID: PMC4844592 DOI: 10.1016/j.nicl.2016.03.021] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 03/20/2016] [Accepted: 03/29/2016] [Indexed: 11/23/2022]
Abstract
BACKGROUND Individuals with autism spectrum disorder (ASD) show atypical brain activity, perhaps due to delayed maturation. Previous studies examining the maturation of auditory electrophysiological activity have been limited due to their use of cross-sectional designs. The present study took a first step in examining magnetoencephalography (MEG) evidence of abnormal auditory response maturation in ASD via the use of a longitudinal design. METHODS Initially recruited for a previous study, 27 children with ASD and nine typically developing (TD) children, aged 6- to 11-years-old, were re-recruited two to five years later. At both timepoints, MEG data were obtained while participants passively listened to sinusoidal pure-tones. Bilateral primary/secondary auditory cortex time domain (100 ms evoked response latency (M100)) and spectrotemporal measures (gamma-band power and inter-trial coherence (ITC)) were examined. MEG measures were also qualitatively examined for five children who exhibited "optimal outcome", participants who were initially on spectrum, but no longer met diagnostic criteria at follow-up. RESULTS M100 latencies were delayed in ASD versus TD at the initial exam (~ 19 ms) and at follow-up (~ 18 ms). At both exams, M100 latencies were associated with clinical ASD severity. In addition, gamma-band evoked power and ITC were reduced in ASD versus TD. M100 latency and gamma-band maturation rates did not differ between ASD and TD. Of note, the cohort of five children that demonstrated "optimal outcome" additionally exhibited M100 latency and gamma-band activity mean values in-between TD and ASD at both timepoints. Though justifying only qualitative interpretation, these "optimal outcome" related data are presented here to motivate future studies. CONCLUSIONS Children with ASD showed perturbed auditory cortex neural activity, as evidenced by M100 latency delays as well as reduced transient gamma-band activity. Despite evidence for maturation of these responses in ASD, the neural abnormalities in ASD persisted across time. Of note, data from the five children whom demonstrated "optimal outcome" qualitatively suggest that such clinical improvements may be associated with auditory brain responses intermediate between TD and ASD. These "optimal outcome" related results are not statistically significant though, likely due to the low sample size of this cohort, and to be expected as a result of the relatively low proportion of "optimal outcome" in the ASD population. Thus, further investigations with larger cohorts are needed to determine if the above auditory response phenotypes have prognostic utility, predictive of clinical outcome.
Collapse
Affiliation(s)
- Russell G Port
- Lurie Family Foundations MEG Imaging Center, Department of Radiology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - J Christopher Edgar
- Lurie Family Foundations MEG Imaging Center, Department of Radiology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Matthew Ku
- Lurie Family Foundations MEG Imaging Center, Department of Radiology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Luke Bloy
- Lurie Family Foundations MEG Imaging Center, Department of Radiology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Rebecca Murray
- Lurie Family Foundations MEG Imaging Center, Department of Radiology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Lisa Blaskey
- Lurie Family Foundations MEG Imaging Center, Department of Radiology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Susan E Levy
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Timothy P L Roberts
- Lurie Family Foundations MEG Imaging Center, Department of Radiology, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
| |
Collapse
|
72
|
Uzunova G, Pallanti S, Hollander E. Excitatory/inhibitory imbalance in autism spectrum disorders: Implications for interventions and therapeutics. World J Biol Psychiatry 2016; 17:174-86. [PMID: 26469219 DOI: 10.3109/15622975.2015.1085597] [Citation(s) in RCA: 127] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
OBJECTIVES Imbalance between excitation and inhibition and increased excitatory-inhibitory (E-I) ratio is a common mechanism in autism spectrum disorders (ASD) that is responsible for the learning and memory, cognitive, sensory, motor deficits, and seizures occurring in these disorders. ASD are very heterogeneous and better understanding of E-I imbalance in brain will lead to better diagnosis and treatments. METHODS We perform a critical literature review of the causes and presentations of E-I imbalance in ASD. RESULTS E-I imbalance in ASD is due primarily to abnormal glutamatergic and GABAergic neurotransmission in key brain regions such as neocortex, hippocampus, amygdala, and cerebellum. Other causes are due to dysfunction of neuropeptides (oxytocin), synaptic proteins (neuroligins), and immune system molecules (cytokines). At the neuropathological level E-I imbalance in ASD is presented as a "minicolumnopathy". E-I imbalance alters the manner by which the brain processes information and regulates behaviour. New developments for investigating E-I imbalance such as optogenetics and transcranial magnetic stimulation (TMS) are presented. Non-invasive brain stimulation methods such as TMS for treatment of the core symptoms of ASD are discussed. CONCLUSIONS Understanding E-I imbalance has important implications for developing better pharmacological and behavioural treatments for ASD, including TMS, new drugs, biomarkers and patient stratification.
Collapse
Affiliation(s)
- Genoveva Uzunova
- a Albert Einstein College of Medicine and Montefiore Medical Center , Bronx , NY , USA
| | - Stefano Pallanti
- a Albert Einstein College of Medicine and Montefiore Medical Center , Bronx , NY , USA.,b Psychiatry and Behavioural Sciences, UC Davis Health System , CA , USA.,c Department Psychiatry , University of Florence , Florence , Italy.,d Icahn School of Medicine at Mount Sinai , New York , NY , USA
| | - Eric Hollander
- a Albert Einstein College of Medicine and Montefiore Medical Center , Bronx , NY , USA
| |
Collapse
|
73
|
Banerjee A, Luong JA, Ho A, Saib AO, Ploski JE. Overexpression of Homer1a in the basal and lateral amygdala impairs fear conditioning and induces an autism-like social impairment. Mol Autism 2016; 7:16. [PMID: 26929812 PMCID: PMC4770673 DOI: 10.1186/s13229-016-0077-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 02/02/2016] [Indexed: 12/20/2022] Open
Abstract
Background Autism spectrum disorders (ASDs) represent a heterogeneous group of disorders with a wide range of behavioral impairments including social and communication deficits. Apart from these core symptoms, a significant number of ASD individuals display higher levels of anxiety, and some studies indicate that a subset of ASD individuals have a reduced ability to be fear conditioned. Deciphering the molecular basis of ASD has been considerably challenging and it currently remains poorly understood. In this study we examined the molecular basis of autism-like impairments in an environmentally induced animal model of ASD, where pregnant rats are exposed to the known teratogen, valproic acid (VPA), on day 12.5 of gestation and the subsequent progeny exhibit ASD-like symptoms. We focused our analysis on the basal and lateral nucleus of the amygdala (BLA), a region of the brain found to be associated with ASD pathology. Methods We performed whole genome gene expression analysis on the BLA using DNA microarrays to examine differences in gene expression within the amygdala of VPA-exposed animals. We validated one VPA-dysregulated candidate gene (Homer1a) using both quantitative PCR (qRT-PCR) and western blot. Finally, we overexpressed Homer1a within the basal and lateral amygdala of naïve animals utilizing adeno-associated viruses (AAV) and subsequently examined these animals in a battery of behavioral tests associated with ASD, including auditory fear conditioning, social interaction and open field. Results Our microarray data indicated that Homer1a was one of the genes which exhibited a significant upregulation within the amygdala. We observed an increase in Homer1a messenger RNA (mRNA) and protein in multiple cohorts of VPA-exposed animals indicating that dysregulation of Homer1a levels might underlie some of the symptoms exhibited by VPA-exposed animals. To test this hypothesis, we overexpressed Homer1a within BLA neurons utilizing a viral-mediated approach and found that overexpression of Homer1a impaired auditory fear conditioning and reduced social interaction, while having no influence on open-field behavior. Conclusions This study indicates that dysregulation of amygdala Homer1a might contribute to some autism-like symptoms induced by VPA exposure. These findings are interesting in part because Homer1a influences the functioning of Shank3, metabotropic glutamate receptors (mGluR5), and Homer1, and these proteins have previously been associated with ASD, indicating that these differing models of ASD may have a similar molecular basis. Electronic supplementary material The online version of this article (doi:10.1186/s13229-016-0077-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Anwesha Banerjee
- Department of Cell Biology, Emory University, 615 Michael St. WBRB #415, Atlanta, GA 30322 USA
| | - Jonathan A Luong
- School of Behavioral and Brain Sciences, University of Texas at Dallas, 800 West Campbell road, Richardson, TX 75080 USA
| | - Anthony Ho
- School of Behavioral and Brain Sciences, University of Texas at Dallas, 800 West Campbell road, Richardson, TX 75080 USA
| | - Aeshah O Saib
- School of Behavioral and Brain Sciences, University of Texas at Dallas, 800 West Campbell road, Richardson, TX 75080 USA
| | - Jonathan E Ploski
- School of Behavioral and Brain Sciences, University of Texas at Dallas, 800 West Campbell road, Richardson, TX 75080 USA
| |
Collapse
|
74
|
Dubiel A, Kulesza RJ. Prenatal valproic acid exposure disrupts tonotopic c-Fos expression in the rat brainstem. Neuroscience 2016; 324:511-23. [PMID: 27094734 DOI: 10.1016/j.neuroscience.2016.01.030] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Autism spectrum disorder (ASD) is a group of neurodevelopmental conditions characterized by difficulties in communication and social interactions, restricted, repetitive behaviors and sensory abnormalities. Notably, the vast majority of individuals with ASD experience some degree of auditory dysfunction and we have recently reported consistent hypoplasia and dysmorphology in auditory brainstem centers in individuals with ASD. Prenatal exposure to the antiepileptic drug valproic acid (VPA) is associated with an increased risk of ASD. In rodents, prenatal exposure to VPA is employed as an animal model of ASD and is associated with a number of anatomical, physiological and behavioral deficits, including hypoplasia and dysmorphology of auditory brainstem centers. Based on these observations, we hypothesized that such dysmorphology in VPA-exposed animals would translate into abnormal neuronal activity in brainstem circuits and irregular tonotopic maps. Herein, we have subjected control and VPA-exposed animals to 4- or 16-kHz tones and examined neuronal activation with immunohistochemistry for c-Fos. After these exposures, we identified significantly more c-Fos-positive neurons in the auditory brainstem of VPA-exposed animals. Additionally, we observed a larger dispersion of c-Fos-positive neurons and shifted tonotopic bands in VPA-exposed rats. We interpret these findings to suggest hyper-responsiveness to sounds and disrupted mapping of sound frequencies after prenatal VPA exposure. Based on these findings, we suggest that such abnormal patterns of activation may play a role in auditory processing deficits in ASD.
Collapse
Affiliation(s)
- A Dubiel
- Auditory Research Center, Lake Erie College of Osteopathic Medicine, Erie, PA 16509, United States
| | - R J Kulesza
- Auditory Research Center, Department of Anatomy, Lake Erie College of Osteopathic Medicine, Erie, PA 16509, United States.
| |
Collapse
|
75
|
Anomal RF, de Villers-Sidani E, Brandão JA, Diniz R, Costa MR, Romcy-Pereira RN. Impaired Processing in the Primary Auditory Cortex of an Animal Model of Autism. Front Syst Neurosci 2015; 9:158. [PMID: 26635548 PMCID: PMC4644803 DOI: 10.3389/fnsys.2015.00158] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 10/30/2015] [Indexed: 12/27/2022] Open
Abstract
Autism is a neurodevelopmental disorder clinically characterized by deficits in communication, lack of social interaction and repetitive behaviors with restricted interests. A number of studies have reported that sensory perception abnormalities are common in autistic individuals and might contribute to the complex behavioral symptoms of the disorder. In this context, hearing incongruence is particularly prevalent. Considering that some of this abnormal processing might stem from the unbalance of inhibitory and excitatory drives in brain circuitries, we used an animal model of autism induced by valproic acid (VPA) during pregnancy in order to investigate the tonotopic organization of the primary auditory cortex (AI) and its local inhibitory circuitry. Our results show that VPA rats have distorted primary auditory maps with over-representation of high frequencies, broadly tuned receptive fields and higher sound intensity thresholds as compared to controls. However, we did not detect differences in the number of parvalbumin-positive interneurons in AI of VPA and control rats. Altogether our findings show that neurophysiological impairments of hearing perception in this autism model occur independently of alterations in the number of parvalbumin-expressing interneurons. These data support the notion that fine circuit alterations, rather than gross cellular modification, could lead to neurophysiological changes in the autistic brain.
Collapse
Affiliation(s)
| | | | | | - Rebecca Diniz
- Brain Institute, Federal University of Rio Grande do Norte Natal, Brazil
| | - Marcos R Costa
- Brain Institute, Federal University of Rio Grande do Norte Natal, Brazil
| | | |
Collapse
|
76
|
Dubiel A, Kulesza RJ. Prenatal valproic acid exposure disrupts tonotopic c-Fos expression in the rat brainstem. Neuroscience 2015; 311:349-61. [PMID: 26518464 DOI: 10.1016/j.neuroscience.2015.10.043] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2015] [Revised: 10/05/2015] [Accepted: 10/22/2015] [Indexed: 12/13/2022]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental condition characterized by difficulties with communication and social interactions, restricted, repetitive behaviors and sensory abnormalities. Additionally, the vast majority of subjects with ASD suffer some degree of auditory dysfunction and we have previously identified significant hypoplasia and dysmorphology in auditory brainstem centers in individuals with ASD. Prenatal exposure to the antiepileptic drug valproic acid (VPA) is associated with an increased risk of ASD. In rodents, prenatal exposure to VPA is utilized as an animal model of ASD and is associated with a number of anatomical, physiological and behavioral deficits, including hypoplasia and dysmorphology in the auditory brainstem. Based on these observations, we hypothesized that such dysmorphology in VPA-exposed animals would translate into abnormal activity in brainstem circuits and irregular tonotopic maps. Herein, we have subjected control and VPA-exposed animals to 4 or 16 kHz tones and examined neuronal activation with immunohistochemistry for c-Fos. After these sound exposures, we found significantly more c-Fos-positive neurons in the auditory brainstem of VPA-exposed animals. Further, we found a larger dispersion of c-Fos-positive neurons and shifted tonotopic bands in VPA-exposed rats. We interpret these findings to suggest hyper-responsiveness to sounds and disrupted mapping of sound frequencies after prenatal VPA exposure. Based on these findings, we suggest that such abnormal patterns of activation may play a role in auditory processing deficits in ASD.
Collapse
Affiliation(s)
- A Dubiel
- Auditory Research Center, Lake Erie College of Osteopathic Medicine, Erie, PA 16509, United States
| | - R J Kulesza
- Auditory Research Center, Department of Anatomy, Lake Erie College of Osteopathic Medicine, Erie, PA 16509, United States.
| |
Collapse
|
77
|
Sesarini CV, Costa L, Grañana N, Coto MG, Pallia RC, Argibay PF. Association between GABA(A) receptor subunit polymorphisms and autism spectrum disorder (ASD). Psychiatry Res 2015; 229:580-2. [PMID: 26239769 DOI: 10.1016/j.psychres.2015.07.077] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Revised: 07/14/2015] [Accepted: 07/25/2015] [Indexed: 01/30/2023]
Abstract
ASD might be associated with alterations in excitation/inhibition ratio and GABA(A) has been implicated since it mediates synaptic inhibition. Polymorphisms in GABA receptor (GABAR) were studied: significant differences in allele and genotype frequencies observed between cases and controls (rs1912960, GABRA4). Haplotype analysis: rs1912960 (GABRA4) and rs211037 (GABRG2) overrepresented in cases. Rs1912960 has been associated with ASD and rs211037 with epilepsy. GABRA4 is associated with autism in the Argentinean dataset independently or in combination with GABRG2.
Collapse
Affiliation(s)
- Carla V Sesarini
- Instituto de Ciencias Básicas y Medicina Experimental (ICBME), Instituto Universitario del Hospital Italiano de Buenos Aires (HIBA), Potosi 4240 (C1199ACL), CABA, Argentina.
| | - Lucas Costa
- Instituto de Ciencias Básicas y Medicina Experimental (ICBME), Instituto Universitario del Hospital Italiano de Buenos Aires (HIBA), Potosi 4240 (C1199ACL), CABA, Argentina
| | - Nora Grañana
- Hospital Durand, Diaz Velez 5044 (C1405DCS), CABA, Argentina
| | - Miguel Garcia Coto
- Centro de Investigaciones del Desarrollo Psiconeurológico (CIDEP), Av. Libertador 6049 (C1428AAG), CABA, Argentina
| | - Roberto C Pallia
- Pediatric Mental Health, Hospital Italiano de Buenos Aires (HIBA), Potosi 4060 (C1181ACH), CABA, Argentina
| | - Pablo F Argibay
- Instituto de Ciencias Básicas y Medicina Experimental (ICBME), Instituto Universitario del Hospital Italiano de Buenos Aires (HIBA), Potosi 4240 (C1199ACL), CABA, Argentina
| |
Collapse
|
78
|
Reduced prefrontal dopaminergic activity in valproic acid-treated mouse autism model. Behav Brain Res 2015; 289:39-47. [DOI: 10.1016/j.bbr.2015.04.022] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2015] [Revised: 04/08/2015] [Accepted: 04/11/2015] [Indexed: 01/23/2023]
|
79
|
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: 58] [Impact Index Per Article: 5.8] [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
|
80
|
Port RG, Anwar AR, Ku M, Carlson GC, Siegel SJ, Roberts TP. Prospective MEG biomarkers in ASD: pre-clinical evidence and clinical promise of electrophysiological signatures. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2015; 88:25-36. [PMID: 25745372 PMCID: PMC4345535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Autism spectrum disorders (ASD) are characterized by social impairments and restricted/stereotyped behaviors and currently affect an estimated 1 in 68 children aged 8 years old. While there has been substantial recent focus on ASD in research, both the biological pathology and, perhaps consequently, a fully effective treatment have yet to be realized. What has remained throughout is the hypothesis that ASD has neurobiological underpinnings and the observation that both the phenotypic expression and likely the underlying etiology is highly heterogeneous. Given the neurodevelopmental basis of ASD, a biologically based marker (biomarker) could prove useful not only for diagnostic and prognostic purposes, but also for stratification and response indices for pharmaceutical development. In this review, we examine the current state of the field for MEG-related biomarkers in ASD. We describe several potential biomarkers (middle latency delays [M50/M100], mismatch negativity latency, gamma-band oscillatory activity), and investigate their relation to symptomology, core domains of dysfunction (e.g., language impairment), and putative biological underpinnings.
Collapse
Affiliation(s)
- Russell G. Port
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania,Lurie Family Foundations MEG Imaging Center, Department of Radiology, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania,Neurosciences Graduate Group, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ayesha R. Anwar
- Lurie Family Foundations MEG Imaging Center, Department of Radiology, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Matthew Ku
- Lurie Family Foundations MEG Imaging Center, Department of Radiology, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Gregory C. Carlson
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania,Neurosciences Graduate Group, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Steven J. Siegel
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania,Neurosciences Graduate Group, University of Pennsylvania, Philadelphia, Pennsylvania,Bioengineering Graduate Group, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Timothy P.L. Roberts
- Lurie Family Foundations MEG Imaging Center, Department of Radiology, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania,Neurosciences Graduate Group, University of Pennsylvania, Philadelphia, Pennsylvania,To whom all correspondence should be addressed: Timothy P.L. Roberts, Department of Radiology, Lurie Family Foundations MEG Imaging Center, The Children’s Hospital of Philadelphia, Philadelphia, PA. Fax: 215-590-1345;
| |
Collapse
|
81
|
Vagal nerve stimulation blocks interleukin 6-dependent synaptic hyperexcitability induced by lipopolysaccharide-induced acute stress in the rodent prefrontal cortex. Brain Behav Immun 2015; 43:149-58. [PMID: 25128387 PMCID: PMC4727901 DOI: 10.1016/j.bbi.2014.07.020] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Revised: 07/29/2014] [Accepted: 07/29/2014] [Indexed: 01/24/2023] Open
Abstract
The ratio between synaptic inhibition and excitation (sI/E) is a critical factor in the pathophysiology of neuropsychiatric disease. We recently described a stress-induced interleukin-6 dependent mechanism leading to a decrease in sI/E in the rodent temporal cortex. The aim of the present study was to determine whether a similar mechanism takes place in the prefrontal cortex, and to elaborate strategies to prevent or attenuate it. We used aseptic inflammation (single acute injections of lipopolysaccharide, LPS, 10mg/kg) as stress model, and patch-clamp recording on a prefrontal cortical slice preparation from wild-type rat and mice, as well as from transgenic mice in which the inhibitor of IL-6 trans-signaling sgp130Fc was produced in a brain-specific fashion (sgp130Fc mice). The anti-inflammatory reflex was activated either by vagal nerve stimulation or peripheral administration of the nicotinic α7 receptor agonist PHA543613. We found that the IL-6-dependent reduction in prefrontal cortex synaptic inhibition was blocked in sgp130Fc mice, or - in wild-type animals - upon application sgp130Fc. Similar results were obtained by activating the "anti-inflammatory reflex" - a neural circuit regulating peripheral immune response - by stimulation of the vagal nerve or through peripheral administration of the α7 nicotinic receptor agonist PHA543613. Our results indicate that the prefrontal cortex is an important potential target of IL-6 mediated trans-signaling, and suggest a potential new avenue in the treatment of a large class of hyperexcitable neuropsychiatric conditions, including epilepsy, schizophrenic psychoses, anxiety disorders, autism spectrum disorders, and depression.
Collapse
|
82
|
Raza S, Harker A, Richards S, Kolb B, Gibb R. Tactile stimulation improves neuroanatomical pathology but not behavior in rats prenatally exposed to valproic acid. Behav Brain Res 2014; 282:25-36. [PMID: 25557797 DOI: 10.1016/j.bbr.2014.12.055] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Revised: 12/20/2014] [Accepted: 12/25/2014] [Indexed: 12/11/2022]
Abstract
Autism is a severe neurodevelopmental disorder with a population prevalence of 1 in 68, and dramatically increasing. While no single pharmacologic intervention has successfully targeted the core symptoms of autism, emerging evidence suggests that postnatal environmental manipulations may offer greater therapeutic efficacy. Massage therapy, or tactile stimulation (TS), early in life has repeatedly been shown to be an effective, low-cost, therapeutic approach in ameliorating the cognitive, social, and emotional symptoms of autism. While early TS treatment attenuates many of the behavioral aberrations among children with autism, the neuroanatomical correlates driving such changes are unknown. The present study assessed the therapeutic effects of early TS treatment on behavior and neuroanatomy using the valproic acid (VPA) rodent model of autism. Rats were prenatally exposed to VPA on gestational day 12.5 and received TS shortly following birth. Whereas TS reversed almost all the VPA-induced alterations in neuroanatomy, it failed to do so behaviorally. The TS VPA animals, when compared to VPA animals, did not exhibit altered or improved behavior in the delayed non-match-to-sample T-maze, Whishaw tray reaching, activity box, or elevated plus maze tasks. Anatomically, however, there were significant increases in dendritic branching and spine density in the medial prefrontal cortex, orbital frontal cortex, and amygdala in VPA animals following early TS treatment, suggesting a complete reversal or remediation of the VPA-induced effects in these regions. The results suggest that postnatal TS, during a critical period in development, acts as a powerful reorganization tool that can ameliorate the neuroanatomical consequences of prenatal VPA exposure.
Collapse
Affiliation(s)
- S Raza
- Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Canada.
| | - A Harker
- Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Canada
| | - S Richards
- Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Canada
| | - B Kolb
- Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Canada; Canadian Institute for Advanced Research Program in Child Brain Development, Canada
| | - R Gibb
- Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Canada
| |
Collapse
|
83
|
Fatemi SH, Reutiman TJ, Folsom TD, Rustan OG, Rooney RJ, Thuras PD. Downregulation of GABAA receptor protein subunits α6, β2, δ, ε, γ2, θ, and ρ2 in superior frontal cortex of subjects with autism. J Autism Dev Disord 2014; 44:1833-45. [PMID: 24668190 DOI: 10.1007/s10803-014-2078-x] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
We measured protein and mRNA levels for nine gamma-aminobutyric acid A (GABAA) receptor subunits in three brain regions (cerebellum, superior frontal cortex, and parietal cortex) in subjects with autism versus matched controls. We observed changes in mRNA for a number of GABAA and GABAB subunits and overall reduced protein expression for GABAA receptor alpha 6 (GABRα6), GABAA receptor beta 2 (GABRβ2), GABAA receptor delta (GABRδ), GABAA receptor epsilon (GABRε), GABAA receptor gamma 2 (GABRγ2), GABAA receptor theta (GABRθ), and GABAA receptor rho 2 (GABRρ2) in superior frontal cortex from subjects with autism. Our data demonstrate systematic changes in GABAA&B subunit expression in brains of subjects with autism, which may help explain the presence of cognitive abnormalities in subjects with autism.
Collapse
Affiliation(s)
- S Hossein Fatemi
- Division of Neuroscience Research, Department of Psychiatry, University of Minnesota Medical School, 420 Delaware St SE, MMC 392, Minneapolis, MN, 55455, USA,
| | | | | | | | | | | |
Collapse
|
84
|
Engineer CT, Centanni TM, Im KW, Kilgard MP. Speech sound discrimination training improves auditory cortex responses in a rat model of autism. Front Syst Neurosci 2014; 8:137. [PMID: 25140133 PMCID: PMC4122159 DOI: 10.3389/fnsys.2014.00137] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Accepted: 07/14/2014] [Indexed: 11/28/2022] Open
Abstract
Children with autism often have language impairments and degraded cortical responses to speech. Extensive behavioral interventions can improve language outcomes and cortical responses. Prenatal exposure to the antiepileptic drug valproic acid (VPA) increases the risk for autism and language impairment. Prenatal exposure to VPA also causes weaker and delayed auditory cortex responses in rats. In this study, we document speech sound discrimination ability in VPA exposed rats and document the effect of extensive speech training on auditory cortex responses. VPA exposed rats were significantly impaired at consonant, but not vowel, discrimination. Extensive speech training resulted in both stronger and faster anterior auditory field (AAF) responses compared to untrained VPA exposed rats, and restored responses to control levels. This neural response improvement generalized to non-trained sounds. The rodent VPA model of autism may be used to improve the understanding of speech processing in autism and contribute to improving language outcomes.
Collapse
Affiliation(s)
- Crystal T Engineer
- Cortical Plasticity Laboratory, School of Behavioral and Brain Sciences, The University of Texas at Dallas Richardson, TX, USA
| | - Tracy M Centanni
- Cortical Plasticity Laboratory, School of Behavioral and Brain Sciences, The University of Texas at Dallas Richardson, TX, USA
| | - Kwok W Im
- Cortical Plasticity Laboratory, School of Behavioral and Brain Sciences, The University of Texas at Dallas Richardson, TX, USA
| | - Michael P Kilgard
- Cortical Plasticity Laboratory, School of Behavioral and Brain Sciences, The University of Texas at Dallas Richardson, TX, USA
| |
Collapse
|
85
|
Kumamaru E, Egashira Y, Takenaka R, Takamori S. Valproic acid selectively suppresses the formation of inhibitory synapses in cultured cortical neurons. Neurosci Lett 2014; 569:142-7. [PMID: 24708928 DOI: 10.1016/j.neulet.2014.03.066] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Revised: 12/19/2013] [Accepted: 03/27/2014] [Indexed: 12/15/2022]
Abstract
Valproic acid (VPA) has been used to treat epileptic patients because of its ability to potentiate GABA signaling in the brain. Despite its clinical significance, VPA administration during pregnancy increases the risk of congenital abnormalities, such as neural tube defects and neurodevelopmental disorders including autism. Furthermore, recent studies revealed that early postnatal administration of VPA also leads to neurodevelopmental deficits in rodents. Here, using cultured cortical neurons derived from postnatal day 1 rats, we examined whether exposure to VPA would affect synapse formation. When neurons were exposed to 1mM VPA during early development, expression of the vesicular GABA transporter (VGAT) was selectively reduced, whereas other synaptic markers, including the vesicular glutamate transporters 1 and 2 (VGLUT1 and 2), were not affected. This VPA effect was mediated through inhibition of histone deacetylases (HDACs), since the effects were mostly recapitulated by an HDAC inhibitor, trichostatin A, but not by a VPA derivative, valpromide, which lacks HDAC inhibitor activity. Immunocytochemical analysis demonstrated that VPA exposure resulted in a retardation of axonal growth specific to GABAergic neurons and a decrease in VGAT-positive synapses. Since disturbance of the excitatory and inhibitory (E-I) balance has been implicated as a potential cause of multiple psychiatric disorders, our results may account for one of the cellular mechanisms underlying the pathogenesis of VPA-induced neurodevelopmental impairments.
Collapse
Affiliation(s)
- Emi Kumamaru
- Laboratory of Neural Membrane Biology, Graduate School of Brain Science, Doshisha University, 1-3 Miyakodani, Tatara, Kyotanabe-shi, Kyoto 610-0394, Japan.
| | - Yoshihiro Egashira
- Laboratory of Neural Membrane Biology, Graduate School of Brain Science, Doshisha University, 1-3 Miyakodani, Tatara, Kyotanabe-shi, Kyoto 610-0394, Japan.
| | - Rie Takenaka
- Laboratory of Neural Membrane Biology, Graduate School of Brain Science, Doshisha University, 1-3 Miyakodani, Tatara, Kyotanabe-shi, Kyoto 610-0394, Japan.
| | - Shigeo Takamori
- Laboratory of Neural Membrane Biology, Graduate School of Brain Science, Doshisha University, 1-3 Miyakodani, Tatara, Kyotanabe-shi, Kyoto 610-0394, Japan.
| |
Collapse
|
86
|
Engineer CT, Centanni TM, Im KW, Borland MS, Moreno NA, Carraway RS, Wilson LG, Kilgard MP. Degraded auditory processing in a rat model of autism limits the speech representation in non-primary auditory cortex. Dev Neurobiol 2014; 74:972-86. [PMID: 24639033 DOI: 10.1002/dneu.22175] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Revised: 02/17/2014] [Accepted: 03/07/2014] [Indexed: 01/22/2023]
Abstract
Although individuals with autism are known to have significant communication problems, the cellular mechanisms responsible for impaired communication are poorly understood. Valproic acid (VPA) is an anticonvulsant that is a known risk factor for autism in prenatally exposed children. Prenatal VPA exposure in rats causes numerous neural and behavioral abnormalities that mimic autism. We predicted that VPA exposure may lead to auditory processing impairments which may contribute to the deficits in communication observed in individuals with autism. In this study, we document auditory cortex responses in rats prenatally exposed to VPA. We recorded local field potentials and multiunit responses to speech sounds in primary auditory cortex, anterior auditory field, ventral auditory field. and posterior auditory field in VPA exposed and control rats. Prenatal VPA exposure severely degrades the precise spatiotemporal patterns evoked by speech sounds in secondary, but not primary auditory cortex. This result parallels findings in humans and suggests that secondary auditory fields may be more sensitive to environmental disturbances and may provide insight into possible mechanisms related to auditory deficits in individuals with autism.
Collapse
Affiliation(s)
- C T Engineer
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, Texas, 75080
| | | | | | | | | | | | | | | |
Collapse
|
87
|
Chomiak T, Hung J, Cihal A, Dhaliwal J, Baghdadwala MI, Dzwonek A, Podgorny P, Hu B. Auditory-cued sensorimotor task reveals disengagement deficits in rats exposed to the autism-associated teratogen valproic acid. Neuroscience 2014; 268:212-20. [PMID: 24631679 DOI: 10.1016/j.neuroscience.2014.02.049] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Revised: 02/26/2014] [Accepted: 02/27/2014] [Indexed: 10/25/2022]
Abstract
Autism Spectrum Disorder (ASD) is often found to co-exist with non-core behavioral manifestations that include difficulties in disengagement of attention to sensory cues. Here we examined whether this behavioral abnormality can be induced in rats prenatally exposed to valproic acid (VPA), a well-established teratogen associated with ASD animal models. We tested rats using an auditory-cued sensorimotor task (ACST) based on the premise that ACST will be more sensitive to developmental changes in temporal association cortex (TeA) of the posterior attention system. We show that VPA rats learned the ACST markedly faster than control animals, but they exhibited a profound preoccupation with cues associated with the expectancy at the reward location such that disengagement was disrupted. Control rats on the other hand were able to disengage and utilize auditory cues for re-engagement. However, both control and VPA-treated rats performed similarly when tested on novel object recognition (NOR) and novel context mismatch (NOCM) behavioral tasks that are known to be sensitive to normal perirhinal and prefrontal network functioning respectively. Consistent with disrupted posterior rather than frontal networks, we also report that VPA can selectively act on deep-layer TeA cortical neurons by showing that VPA increased dendritic density in isolated deep-layer TeA but not frontal neurons. These results describe a useful approach to examine the role of cue-dependent control of attention systems in rodent models of autism and suggest that disengagement impairments may arise from an inability to modify behavior through the appropriate use of sensory cue associations.
Collapse
Affiliation(s)
- T Chomiak
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, Faculty of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada.
| | - J Hung
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, Faculty of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - A Cihal
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, Faculty of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - J Dhaliwal
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, Faculty of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - M I Baghdadwala
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, Faculty of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - A Dzwonek
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, Faculty of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - P Podgorny
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, Faculty of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - B Hu
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, Faculty of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada.
| |
Collapse
|
88
|
Napoli E, Dueñas N, Giulivi C. Potential therapeutic use of the ketogenic diet in autism spectrum disorders. Front Pediatr 2014; 2:69. [PMID: 25072037 PMCID: PMC4074854 DOI: 10.3389/fped.2014.00069] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Accepted: 06/17/2014] [Indexed: 11/13/2022] Open
Abstract
The ketogenic diet (KGD) has been recognized as an effective treatment for individuals with glucose transporter 1 (GLUT1) and pyruvate dehydrogenase (PDH) deficiencies as well as with epilepsy. More recently, its use has been advocated in a number of neurological disorders prompting a newfound interest in its possible therapeutic use in autism spectrum disorders (ASD). One study and one case report indicated that children with ASD treated with a KGD showed decreased seizure frequencies and exhibited behavioral improvements (i.e., improved learning abilities and social skills). The KGD could benefit individuals with ASD affected with epileptic episodes as well as those with either PDH or mild respiratory chain (RC) complex deficiencies. Given that the mechanism of action of the KGD is not fully understood, caution should be exercised in ASD cases lacking a careful biochemical and metabolic characterization to avoid deleterious side effects or refractory outcomes.
Collapse
Affiliation(s)
- Eleonora Napoli
- Department of Molecular Biosciences, University of California Davis , Davis, CA , USA
| | - Nadia Dueñas
- Department of Molecular Biosciences, University of California Davis , Davis, CA , USA
| | - Cecilia Giulivi
- Department of Molecular Biosciences, University of California Davis , Davis, CA , USA ; Medical Investigations of Neurodevelopmental Disorders (M. I. N. D.) Institute , Sacramento, CA , USA
| |
Collapse
|
89
|
Cellot G, Cherubini E. GABAergic signaling as therapeutic target for autism spectrum disorders. Front Pediatr 2014; 2:70. [PMID: 25072038 PMCID: PMC4085902 DOI: 10.3389/fped.2014.00070] [Citation(s) in RCA: 191] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Accepted: 06/23/2014] [Indexed: 11/13/2022] Open
Abstract
γ-Aminobutyric acid (GABA), the main inhibitory neurotransmitter in the adult brain, early in postnatal life exerts a depolarizing and excitatory action. This depends on accumulation of chloride inside the cell via the cation-chloride importer NKCC1, being the expression of the chloride exporter KCC2 very low at birth. The developmentally regulated expression of KCC2 results in extrusion of chloride with age and a shift of GABA from the depolarizing to the hyperpolarizing direction. The depolarizing action of GABA leads to intracellular calcium rise through voltage-dependent calcium channels and/or N-methyl-d-aspartate receptors. GABA-mediated calcium signals regulate a variety of developmental processes from cell proliferation migration, differentiation, synapse maturation, and neuronal wiring. Therefore, it is not surprising that some forms of neuro-developmental disorders such as autism spectrum disorders (ASDs) are associated with alterations of GABAergic signaling and impairment of the excitatory/inhibitory balance in selective neuronal circuits. In this review, we will discuss how changes of GABAA-mediated neurotransmission affect several forms of ASDs including the Fragile X, the Angelman, and Rett syndromes. Then, we will describe various animal models of ASDs with GABAergic dysfunctions, highlighting their behavioral deficits and the possibility to rescue them by targeting selective components of the GABAergic synapse. In particular, we will discuss how in some cases, reverting the polarity of GABA responses from the depolarizing to the hyperpolarizing direction with the diuretic bumetanide, a selective blocker of NKCC1, may have beneficial effects on ASDs, thus opening new therapeutic perspectives for the treatment of these devastating disorders.
Collapse
Affiliation(s)
- Giada Cellot
- Department of Neuroscience, Scuola Internazionale Superiore di Studi Avanzati , Trieste , Italy
| | - Enrico Cherubini
- Department of Neuroscience, Scuola Internazionale Superiore di Studi Avanzati , Trieste , Italy ; European Brain Research Institute , Rome , Italy
| |
Collapse
|
90
|
What We Have Learned about Autism Spectrum Disorder from Valproic Acid. PATHOLOGY RESEARCH INTERNATIONAL 2013; 2013:712758. [PMID: 24381784 PMCID: PMC3871912 DOI: 10.1155/2013/712758] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Revised: 10/16/2013] [Accepted: 10/17/2013] [Indexed: 12/29/2022]
Abstract
Two recent epidemiological investigations in children exposed to valproic acid (VPA) treatment in utero have reported a significant risk associated with neurodevelopmental disorders and autism spectrum disorder (ASD) in particular. Parallel to this work, there is a growing body of animal research literature using VPA as an animal model of ASD. In this focused review we first summarize the epidemiological evidence linking VPA to ASD and then comment on two important neurobiological findings linking VPA to ASD clinicopathology, namely, accelerated or early brain overgrowth and hyperexcitable networks. Improving our understanding of how the drug VPA can alter early development of neurological systems will ultimately improve our understanding of ASD.
Collapse
|
91
|
Bringas M, Carvajal-Flores F, López-Ramírez T, Atzori M, Flores G. Rearrangement of the dendritic morphology in limbic regions and altered exploratory behavior in a rat model of autism spectrum disorder. Neuroscience 2013; 241:170-87. [DOI: 10.1016/j.neuroscience.2013.03.030] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Revised: 02/25/2013] [Accepted: 03/15/2013] [Indexed: 12/30/2022]
|