Skip to main content
Top
Published in: Journal of Translational Medicine 1/2022

Open Access 01-12-2022 | Autism Spectrum Disorder | Research

Continuous high-frequency deep brain stimulation of the anterior insula modulates autism-like behavior in a valproic acid-induced rat model

Authors: Lifei Xiao, Shucai Jiang, Yangyang Wang, Caibin Gao, Cuicui Liu, Xianhao Huo, Wenchao Li, Baorui Guo, Chaofan Wang, Yu Sun, Anni Wang, Yan Feng, Feng Wang, Tao Sun

Published in: Journal of Translational Medicine | Issue 1/2022

Login to get access

Abstract

Background

Until now, the treatment of patients with autism spectrum disorder (ASD) remain a difficult problem. The insula is involved in empathy and sensorimotor integration, which are often impaired in individuals with ASD. Deep brain stimulation, modulating neuronal activity in specific brain circuits, has recently been considered as a promising intervention for neuropsychiatric disorders. Valproic acid (VPA) is a potential teratogenic agent, and prenatal exposure can cause autism-like symptoms including repetitive behaviors and defective sociability. Herein, we investigated the effects of continuous high-frequency deep brain stimulation in the anterior insula of rats exposed to VPA and explored cognitive functions, behavior, and molecular proteins connected to autism spectrum disorder.

Methods

VPA-exposed offspring were bilaterally implanted with electrodes in the anterior insula (Day 0) with a recovery period of 1 week. (Day 0–7). High-frequency deep brain stimulation was applied from days 11 to 29. Three behavioral tests, including three-chamber social interaction test, were performed on days 7, 13, 18, 25 and 36, and several rats were used for analysis of immediate early genes and proteomic after deep brain stimulation intervention. Meanwhile, animals were subjected to a 20 day spatial learning and cognitive rigidity test using IntelliCage on day 11.

Results

Deep brain stimulation improved the sociability and social novelty preference at day 18 prior to those at day 13, and the improvement has reached the upper limit compared to day 25. As for repetitive/stereotypic-like behavior, self- grooming time were reduced at day 18 and reached the upper limit, and the numbers of burried marbles were reduced at day 13 prior to those at day 18 and day 25. The improvements of sociability and social novelty preference were persistent after the stimulation had ceased. Spatial learning ability and cognitive rigidity were unaffected. We identified 35 proteins in the anterior insula, some of which were intimately linked to autism, and their expression levels were reversed upon administration of deep brain stimulation.

Conclusions

Autism-like behavior was ameliorated and autism-related proteins were reversed in the insula by deep brain stimulation intervention, these findings reveal that the insula may be a potential target for DBS in the treatment of autism, which provide a theoretical basis for its clinical application., although future studies are still warranted.
Appendix
Available only for authorised users
Literature
1.
go back to reference Lord C, Brugha TS, Charman T, et al. Autism spectrum disorder. Nat Rev Dis Primers. 2020;6:5.CrossRef Lord C, Brugha TS, Charman T, et al. Autism spectrum disorder. Nat Rev Dis Primers. 2020;6:5.CrossRef
2.
go back to reference Robertson CE, Baron-Cohen S. Sensory perception in autism. Nat Rev Neurosci. 2017;18:671–84.CrossRef Robertson CE, Baron-Cohen S. Sensory perception in autism. Nat Rev Neurosci. 2017;18:671–84.CrossRef
3.
go back to reference Baxter AJ, Brugha TS, Erskine HE, et al. The epidemiology and global burden of autism spectrum disorders. Psychol Med. 2015;45:601–13.CrossRef Baxter AJ, Brugha TS, Erskine HE, et al. The epidemiology and global burden of autism spectrum disorders. Psychol Med. 2015;45:601–13.CrossRef
4.
go back to reference Chaste P, Leboyer M. Autism risk factors: genes, environment, and gene-environment interactions. Dialogues Clin Neurosci. 2012;14:281–92.CrossRef Chaste P, Leboyer M. Autism risk factors: genes, environment, and gene-environment interactions. Dialogues Clin Neurosci. 2012;14:281–92.CrossRef
5.
go back to reference Sharma SR, Gonda X, Tarazi FI. Autism spectrum disorder: classification, diagnosis and therapy. Pharmacol Ther. 2018;190:91–104.CrossRef Sharma SR, Gonda X, Tarazi FI. Autism spectrum disorder: classification, diagnosis and therapy. Pharmacol Ther. 2018;190:91–104.CrossRef
7.
go back to reference Herrington TM, Cheng JJ, Eskandar EN. Mechanisms of deep brain stimulation. J Neurophysiol. 2016;115:19–38.CrossRef Herrington TM, Cheng JJ, Eskandar EN. Mechanisms of deep brain stimulation. J Neurophysiol. 2016;115:19–38.CrossRef
8.
go back to reference Okun MS. Deep-brain stimulation for Parkinson’s disease. N Engl J Med. 2012;367:1529–38.CrossRef Okun MS. Deep-brain stimulation for Parkinson’s disease. N Engl J Med. 2012;367:1529–38.CrossRef
9.
go back to reference Pauls KA, Hammesfahr S, Moro E, et al. Deep brain stimulation in the ventrolateral thalamus/subthalamic area in dystonia with head tremor. Mov Disord. 2014;29:953–9.CrossRef Pauls KA, Hammesfahr S, Moro E, et al. Deep brain stimulation in the ventrolateral thalamus/subthalamic area in dystonia with head tremor. Mov Disord. 2014;29:953–9.CrossRef
10.
go back to reference Knotkova H, Hamani C, Sivanesan E, et al. Neuromodulation for chronic pain. Lancet. 2021;397:2111–24.CrossRef Knotkova H, Hamani C, Sivanesan E, et al. Neuromodulation for chronic pain. Lancet. 2021;397:2111–24.CrossRef
11.
go back to reference Lozano AM, Lipsman N, Bergman H, et al. Deep brain stimulation: current challenges and future directions. Nat Rev Neurol. 2019;15:148–60.CrossRef Lozano AM, Lipsman N, Bergman H, et al. Deep brain stimulation: current challenges and future directions. Nat Rev Neurol. 2019;15:148–60.CrossRef
13.
go back to reference Lin TC, Lo YC, Lin HC, et al. MR imaging central thalamic deep brain stimulation restored autistic-like social deficits in the rat. Brain Stimul. 2019;12:1410–20.CrossRef Lin TC, Lo YC, Lin HC, et al. MR imaging central thalamic deep brain stimulation restored autistic-like social deficits in the rat. Brain Stimul. 2019;12:1410–20.CrossRef
14.
go back to reference Chang AD, Berges VA, Chung SJ, et al. High-frequency stimulation at the subthalamic nucleus suppresses excessive self-grooming in autism-like mouse models. Neuropsychopharmacology. 2016;41:1813–21.CrossRef Chang AD, Berges VA, Chung SJ, et al. High-frequency stimulation at the subthalamic nucleus suppresses excessive self-grooming in autism-like mouse models. Neuropsychopharmacology. 2016;41:1813–21.CrossRef
17.
go back to reference Seeley WW. The salience network: a neural system for perceiving and responding to homeostatic demands. J Neurosci. 2019;39:9878–82.CrossRef Seeley WW. The salience network: a neural system for perceiving and responding to homeostatic demands. J Neurosci. 2019;39:9878–82.CrossRef
18.
go back to reference Tsai PJ, Keeley RJ, Carmack SA, et al. Converging structural and functional evidence for a rat salience network. Biol Psychiatry. 2020;88:867–78.CrossRef Tsai PJ, Keeley RJ, Carmack SA, et al. Converging structural and functional evidence for a rat salience network. Biol Psychiatry. 2020;88:867–78.CrossRef
19.
go back to reference Parellada M, Pina-Camacho L, Moreno C, et al. Insular pathology in young people with high-functioning autism and first-episode psychosis. Psychol Med. 2017;47:2472–82.CrossRef Parellada M, Pina-Camacho L, Moreno C, et al. Insular pathology in young people with high-functioning autism and first-episode psychosis. Psychol Med. 2017;47:2472–82.CrossRef
20.
go back to reference Xu J, Wang H, Zhang L, et al. Both hypo-connectivity and hyper-connectivity of the insular subregions associated with severity in children with autism spectrum disorders. Front Neurosci. 2018;12:234.CrossRef Xu J, Wang H, Zhang L, et al. Both hypo-connectivity and hyper-connectivity of the insular subregions associated with severity in children with autism spectrum disorders. Front Neurosci. 2018;12:234.CrossRef
21.
go back to reference Miura I, Sato M, Overton E, et al. Encoding of social exploration by neural ensembles in the insular cortex. PLoS Biol. 2020;18:e3000584.CrossRef Miura I, Sato M, Overton E, et al. Encoding of social exploration by neural ensembles in the insular cortex. PLoS Biol. 2020;18:e3000584.CrossRef
22.
go back to reference Rogers-Carter MM, Varela JA, Gribbons KB, et al. Insular cortex mediates approach and avoidance responses to social affective stimuli. Nat Neurosci. 2018;21:404–14.CrossRef Rogers-Carter MM, Varela JA, Gribbons KB, et al. Insular cortex mediates approach and avoidance responses to social affective stimuli. Nat Neurosci. 2018;21:404–14.CrossRef
23.
go back to reference Schneider T, Przewłocki R. Behavioral alterations in rats prenatally exposed to valproic acid: animal model of autism. Neuropsychopharmacology. 2005;30:80–9.CrossRef Schneider T, Przewłocki R. Behavioral alterations in rats prenatally exposed to valproic acid: animal model of autism. Neuropsychopharmacology. 2005;30:80–9.CrossRef
25.
go back to reference Vorhees CV. Teratogenicity and developmental toxicity of valproic acid in rats. Teratology. 1987;35:195–202.CrossRef Vorhees CV. Teratogenicity and developmental toxicity of valproic acid in rats. Teratology. 1987;35:195–202.CrossRef
26.
go back to reference Nicolini C, Fahnestock M. The valproic acid-induced rodent model of autism. Exp Neurol. 2018;299:217–27.CrossRef Nicolini C, Fahnestock M. The valproic acid-induced rodent model of autism. Exp Neurol. 2018;299:217–27.CrossRef
27.
go back to reference Paxinos G, Watson C. The Rat Brain in Stereotaxic Coordinates. London: Academic Press; 2013. Paxinos G, Watson C. The Rat Brain in Stereotaxic Coordinates. London: Academic Press; 2013.
28.
go back to reference Crawley JN. Designing mouse behavioral tasks relevant to autistic-like behaviors. Ment Retard Dev Disabil Res Rev. 2004;10:248–58.CrossRef Crawley JN. Designing mouse behavioral tasks relevant to autistic-like behaviors. Ment Retard Dev Disabil Res Rev. 2004;10:248–58.CrossRef
29.
go back to reference Hughes EM, Calcagno P, Clarke M, et al. Prenatal exposure to valproic acid reduces social responses and alters mRNA levels of opioid receptor and pre-pro-peptide in discrete brain regions of adolescent and adult male rats. Brain Res. 2020;1732:146675.CrossRef Hughes EM, Calcagno P, Clarke M, et al. Prenatal exposure to valproic acid reduces social responses and alters mRNA levels of opioid receptor and pre-pro-peptide in discrete brain regions of adolescent and adult male rats. Brain Res. 2020;1732:146675.CrossRef
30.
go back to reference Mohammadi S, Asadi-Shekaari M, Basiri M, et al. Improvement of autistic-like behaviors in adult rats prenatally exposed to valproic acid through early suppression of NMDA receptor function. Psychopharmacology. 2020;237:199–208.CrossRef Mohammadi S, Asadi-Shekaari M, Basiri M, et al. Improvement of autistic-like behaviors in adult rats prenatally exposed to valproic acid through early suppression of NMDA receptor function. Psychopharmacology. 2020;237:199–208.CrossRef
31.
go back to reference Campolongo M, Kazlauskas N, Falasco G, et al. Sociability deficits after prenatal exposure to valproic acid are rescued by early social enrichment. Mol Autism. 2018;9:36.CrossRef Campolongo M, Kazlauskas N, Falasco G, et al. Sociability deficits after prenatal exposure to valproic acid are rescued by early social enrichment. Mol Autism. 2018;9:36.CrossRef
32.
go back to reference McKinnell ZE, Maze T, Ramos A, et al. Valproic acid treated female Long-Evans rats are impaired on attentional set-shifting. Behav Brain Res. 2021;397:112966.CrossRef McKinnell ZE, Maze T, Ramos A, et al. Valproic acid treated female Long-Evans rats are impaired on attentional set-shifting. Behav Brain Res. 2021;397:112966.CrossRef
33.
go back to reference Zheng J, Jiang R, Chen M, et al. Multi-copper ferroxidase-deficient mice have increased brain iron concentrations and learning and memory deficits. J Nutr. 2018;148:643–9.CrossRef Zheng J, Jiang R, Chen M, et al. Multi-copper ferroxidase-deficient mice have increased brain iron concentrations and learning and memory deficits. J Nutr. 2018;148:643–9.CrossRef
34.
go back to reference Wu N, Wang F, Jin Z, et al. Effects of GABA(B) receptors in the insula on recognition memory observed with intellicage. Behav Brain Funct. 2017;13:7.CrossRef Wu N, Wang F, Jin Z, et al. Effects of GABA(B) receptors in the insula on recognition memory observed with intellicage. Behav Brain Funct. 2017;13:7.CrossRef
35.
go back to reference Puścian A, Lęski S, Górkiewicz T, et al. A novel automated behavioral test battery assessing cognitive rigidity in two genetic mouse models of autism. Front Behav Neurosci. 2014;8:140. Puścian A, Lęski S, Górkiewicz T, et al. A novel automated behavioral test battery assessing cognitive rigidity in two genetic mouse models of autism. Front Behav Neurosci. 2014;8:140.
36.
go back to reference Chang H, Gao C, Sun K, et al. Continuous high frequency deep brain stimulation of the rat anterior insula attenuates the relapse post withdrawal and strengthens the extinction of morphine seeking. Front Psychiatry. 2020;11:577155.CrossRef Chang H, Gao C, Sun K, et al. Continuous high frequency deep brain stimulation of the rat anterior insula attenuates the relapse post withdrawal and strengthens the extinction of morphine seeking. Front Psychiatry. 2020;11:577155.CrossRef
37.
go back to reference Bielas H, Arck P, Bruenahl CA, et al. Prenatal stress increases the striatal and hippocampal expression of correlating c-FOS and serotonin transporters in murine offspring. Int J Dev Neurosci. 2014;38:30–5.CrossRef Bielas H, Arck P, Bruenahl CA, et al. Prenatal stress increases the striatal and hippocampal expression of correlating c-FOS and serotonin transporters in murine offspring. Int J Dev Neurosci. 2014;38:30–5.CrossRef
38.
go back to reference Huguet G, Kadar E, Temel Y, et al. Electrical stimulation normalizes c-Fos expression in the deep cerebellar nuclei of depressive-like rats: implication of antidepressant activity. Cerebellum. 2017;16:398–410.CrossRef Huguet G, Kadar E, Temel Y, et al. Electrical stimulation normalizes c-Fos expression in the deep cerebellar nuclei of depressive-like rats: implication of antidepressant activity. Cerebellum. 2017;16:398–410.CrossRef
39.
go back to reference Hachem-Delaunay S, Fournier ML, Cohen C, et al. Subthalamic nucleus high-frequency stimulation modulates neuronal reactivity to cocaine within the reward circuit. Neurobiol Dis. 2015;80:54–62.CrossRef Hachem-Delaunay S, Fournier ML, Cohen C, et al. Subthalamic nucleus high-frequency stimulation modulates neuronal reactivity to cocaine within the reward circuit. Neurobiol Dis. 2015;80:54–62.CrossRef
40.
go back to reference Craig AD. Significance of the insula for the evolution of human awareness of feelings from the body. Ann N Y Acad Sci. 2011;1225:72–82.CrossRef Craig AD. Significance of the insula for the evolution of human awareness of feelings from the body. Ann N Y Acad Sci. 2011;1225:72–82.CrossRef
41.
go back to reference Etkin A, Büchel C, Gross JJ. The neural bases of emotion regulation. Nat Rev Neurosci. 2015;16:693–700.CrossRef Etkin A, Büchel C, Gross JJ. The neural bases of emotion regulation. Nat Rev Neurosci. 2015;16:693–700.CrossRef
42.
go back to reference Sun K, Xiao L, Wu Y, et al. GABAergic neurons in the insular cortex play an important role in cue-morphine reward memory reconsolidation. Life Sci. 2020;254:117655.CrossRef Sun K, Xiao L, Wu Y, et al. GABAergic neurons in the insular cortex play an important role in cue-morphine reward memory reconsolidation. Life Sci. 2020;254:117655.CrossRef
43.
go back to reference Belin-Rauscent A, Daniel ML, Puaud M, et al. From impulses to maladaptive actions: the insula is a neurobiological gate for the development of compulsive behavior. Mol Psychiatry. 2016;21:491–9.CrossRef Belin-Rauscent A, Daniel ML, Puaud M, et al. From impulses to maladaptive actions: the insula is a neurobiological gate for the development of compulsive behavior. Mol Psychiatry. 2016;21:491–9.CrossRef
45.
go back to reference Postorino V, Kerns CM, Vivanti G, et al. Anxiety disorders and obsessive-compulsive disorder in individuals with autism spectrum disorder. Curr Psychiatry Rep. 2017;19:92.CrossRef Postorino V, Kerns CM, Vivanti G, et al. Anxiety disorders and obsessive-compulsive disorder in individuals with autism spectrum disorder. Curr Psychiatry Rep. 2017;19:92.CrossRef
46.
go back to reference Elnahas EM, Abuelezz SA, Mohamad MI, et al. Validation of prenatal versus postnatal valproic acid rat models of autism: a behavioral and neurobiological study. Prog Neuropsychopharmacol Biol Psychiatry. 2021;108:110185.CrossRef Elnahas EM, Abuelezz SA, Mohamad MI, et al. Validation of prenatal versus postnatal valproic acid rat models of autism: a behavioral and neurobiological study. Prog Neuropsychopharmacol Biol Psychiatry. 2021;108:110185.CrossRef
47.
go back to reference Jiménez-Sánchez L, Linge R, Campa L, et al. Behavioral, neurochemical and molecular changes after acute deep brain stimulation of the infralimbic prefrontal cortex. Neuropharmacology. 2016;108:91–102.CrossRef Jiménez-Sánchez L, Linge R, Campa L, et al. Behavioral, neurochemical and molecular changes after acute deep brain stimulation of the infralimbic prefrontal cortex. Neuropharmacology. 2016;108:91–102.CrossRef
48.
go back to reference Pinhal CM, van den Boom B, Santana-Kragelund F, et al. Differential effects of deep brain stimulation of the internal capsule and the striatum on excessive grooming in sapap3 mutant mice. Biol Psychiatry. 2018;84:917–25.CrossRef Pinhal CM, van den Boom B, Santana-Kragelund F, et al. Differential effects of deep brain stimulation of the internal capsule and the striatum on excessive grooming in sapap3 mutant mice. Biol Psychiatry. 2018;84:917–25.CrossRef
49.
go back to reference Jensen AL, Durand DM. High frequency stimulation can block axonal conduction. Exp Neurol. 2009;220:57–70.CrossRef Jensen AL, Durand DM. High frequency stimulation can block axonal conduction. Exp Neurol. 2009;220:57–70.CrossRef
50.
go back to reference Rosenbaum R, Zimnik A, Zheng F, et al. Axonal and synaptic failure suppress the transfer of firing rate oscillations, synchrony and information during high frequency deep brain stimulation. Neurobiol Dis. 2014;62:86–99.CrossRef Rosenbaum R, Zimnik A, Zheng F, et al. Axonal and synaptic failure suppress the transfer of firing rate oscillations, synchrony and information during high frequency deep brain stimulation. Neurobiol Dis. 2014;62:86–99.CrossRef
51.
go back to reference Xia R, Berger F, Piallat B, et al. Alteration of hormone and neurotransmitter production in cultured cells by high and low frequency electrical stimulation. Acta Neurochir (Wien). 2007;149:67–73.CrossRef Xia R, Berger F, Piallat B, et al. Alteration of hormone and neurotransmitter production in cultured cells by high and low frequency electrical stimulation. Acta Neurochir (Wien). 2007;149:67–73.CrossRef
52.
go back to reference Ghahremani R, Mohammadkhani R, Salehi I, et al. Sex differences in spatial learning and memory in valproic acid rat model of autism: possible beneficial role of exercise interventions. Front Behav Neurosci. 2022;16:869792.CrossRef Ghahremani R, Mohammadkhani R, Salehi I, et al. Sex differences in spatial learning and memory in valproic acid rat model of autism: possible beneficial role of exercise interventions. Front Behav Neurosci. 2022;16:869792.CrossRef
53.
go back to reference Karvat G, Kimchi T. Acetylcholine elevation relieves cognitive rigidity and social deficiency in a mouse model of autism. Neuropsychopharmacology. 2014;39:831–40.CrossRef Karvat G, Kimchi T. Acetylcholine elevation relieves cognitive rigidity and social deficiency in a mouse model of autism. Neuropsychopharmacology. 2014;39:831–40.CrossRef
54.
go back to reference Suarez-Jimenez B, Bisby JA, Horner AJ, et al. Linked networks for learning and expressing location-specific threat. Proc Natl Acad Sci U S A. 2018;115:E1032-40.CrossRef Suarez-Jimenez B, Bisby JA, Horner AJ, et al. Linked networks for learning and expressing location-specific threat. Proc Natl Acad Sci U S A. 2018;115:E1032-40.CrossRef
55.
go back to reference Kim YS, Choi J, Yoon BE. Neuron-glia interactions in neurodevelopmental disorders. Cells. 2020;9:2176.CrossRef Kim YS, Choi J, Yoon BE. Neuron-glia interactions in neurodevelopmental disorders. Cells. 2020;9:2176.CrossRef
56.
go back to reference Chang Q, Yang H, Wang M, et al. Role of microtubule-associated protein in autism spectrum disorder. Neurosci Bull. 2018;34:1119–26.CrossRef Chang Q, Yang H, Wang M, et al. Role of microtubule-associated protein in autism spectrum disorder. Neurosci Bull. 2018;34:1119–26.CrossRef
57.
go back to reference DeMari J, Mroske C, Tang S, et al. CLTC as a clinically novel gene associated with multiple malformations and developmental delay. Am J Med Genet A. 2016;170A:958–66.CrossRef DeMari J, Mroske C, Tang S, et al. CLTC as a clinically novel gene associated with multiple malformations and developmental delay. Am J Med Genet A. 2016;170A:958–66.CrossRef
58.
go back to reference Nabais Sá MJ, Venselaar H, Wiel L, et al. De novo CLTC variants are associated with a variable phenotype from mild to severe intellectual disability, microcephaly, hypoplasia of the corpus callosum, and epilepsy. Genet Med. 2020;22:797–802.CrossRef Nabais Sá MJ, Venselaar H, Wiel L, et al. De novo CLTC variants are associated with a variable phenotype from mild to severe intellectual disability, microcephaly, hypoplasia of the corpus callosum, and epilepsy. Genet Med. 2020;22:797–802.CrossRef
59.
go back to reference Castroflorio E, den Hoed J, Svistunova D, et al. The Ncoa7 locus regulates V-ATPase formation and function, neurodevelopment and behaviour. Cell Mol Life Sci. 2021;78:3503–24.CrossRef Castroflorio E, den Hoed J, Svistunova D, et al. The Ncoa7 locus regulates V-ATPase formation and function, neurodevelopment and behaviour. Cell Mol Life Sci. 2021;78:3503–24.CrossRef
60.
go back to reference Aoto K, Kato M, Akita T, et al. ATP6V0A1 encoding the a1-subunit of the V0 domain of vacuolar H(+)-ATPases is essential for brain development in humans and mice. Nat Commun. 2021;12:2107.CrossRef Aoto K, Kato M, Akita T, et al. ATP6V0A1 encoding the a1-subunit of the V0 domain of vacuolar H(+)-ATPases is essential for brain development in humans and mice. Nat Commun. 2021;12:2107.CrossRef
61.
go back to reference Rizzo WB. Sjögren-larsson syndrome: molecular genetics and biochemical pathogenesis of fatty aldehyde dehydrogenase deficiency. Mol Genet Metab. 2007;90:1–9.CrossRef Rizzo WB. Sjögren-larsson syndrome: molecular genetics and biochemical pathogenesis of fatty aldehyde dehydrogenase deficiency. Mol Genet Metab. 2007;90:1–9.CrossRef
62.
go back to reference Toczylowska B, Zieminska E, Senator P, et al. Hippocampal metabolite profiles in two rat models of autism: NMR-based metabolomics studies. Mol Neurobiol. 2020;57:3089–105.CrossRef Toczylowska B, Zieminska E, Senator P, et al. Hippocampal metabolite profiles in two rat models of autism: NMR-based metabolomics studies. Mol Neurobiol. 2020;57:3089–105.CrossRef
63.
go back to reference Li C, Shen K, Chu L, et al. Decreased levels of urinary free amino acids in children with autism spectrum disorder. J Clin Neurosci. 2018;54:45–9.CrossRef Li C, Shen K, Chu L, et al. Decreased levels of urinary free amino acids in children with autism spectrum disorder. J Clin Neurosci. 2018;54:45–9.CrossRef
64.
go back to reference Witters P, Debbold E, Crivelly K, et al. Autism in patients with propionic acidemia. Mol Genet Metab. 2016;119:317–21.CrossRef Witters P, Debbold E, Crivelly K, et al. Autism in patients with propionic acidemia. Mol Genet Metab. 2016;119:317–21.CrossRef
65.
go back to reference Hirokawa N. Microtubule organization and dynamics dependent on microtubule-associated proteins. Curr Opin Cell Biol. 1994;6:74–81.CrossRef Hirokawa N. Microtubule organization and dynamics dependent on microtubule-associated proteins. Curr Opin Cell Biol. 1994;6:74–81.CrossRef
66.
go back to reference Takei Y, Teng J, Harada A, et al. Defects in axonal elongation and neuronal migration in mice with disrupted tau and map1b genes. J Cell Biol. 2000;150:989–1000.CrossRef Takei Y, Teng J, Harada A, et al. Defects in axonal elongation and neuronal migration in mice with disrupted tau and map1b genes. J Cell Biol. 2000;150:989–1000.CrossRef
67.
go back to reference Tai C, Chang CW, Yu GQ, et al. Tau reduction prevents key features of autism in mouse models. Neuron. 2020;106:421-37.e11.CrossRef Tai C, Chang CW, Yu GQ, et al. Tau reduction prevents key features of autism in mouse models. Neuron. 2020;106:421-37.e11.CrossRef
68.
go back to reference Pollard TD, Cooper JA. Actin, a central player in cell shape and movement. Science. 2009;326:1208–12.CrossRef Pollard TD, Cooper JA. Actin, a central player in cell shape and movement. Science. 2009;326:1208–12.CrossRef
70.
go back to reference Perrin BJ, Ervasti JM. The actin gene family: function follows isoform. Cytoskeleton (Hoboken). 2010;67:630–4.CrossRef Perrin BJ, Ervasti JM. The actin gene family: function follows isoform. Cytoskeleton (Hoboken). 2010;67:630–4.CrossRef
71.
go back to reference Verloes A, Di Donato N, Masliah-Planchon J, et al. Baraitser-winter cerebrofrontofacial syndrome: delineation of the spectrum in 42 cases. Eur J Hum Genet. 2015;23:292–301.CrossRef Verloes A, Di Donato N, Masliah-Planchon J, et al. Baraitser-winter cerebrofrontofacial syndrome: delineation of the spectrum in 42 cases. Eur J Hum Genet. 2015;23:292–301.CrossRef
72.
73.
go back to reference Tabrizi SJ, Flower MD, Ross CA, et al. Huntington disease: new insights into molecular pathogenesis and therapeutic opportunities. Nat Rev Neurol. 2020;16:529–46.CrossRef Tabrizi SJ, Flower MD, Ross CA, et al. Huntington disease: new insights into molecular pathogenesis and therapeutic opportunities. Nat Rev Neurol. 2020;16:529–46.CrossRef
74.
go back to reference Rojas-Charry L, Nardi L, Methner A, et al. Abnormalities of synaptic mitochondria in autism spectrum disorder and related neurodevelopmental disorders. J Mol Med (Berl). 2021;99:161–78.CrossRef Rojas-Charry L, Nardi L, Methner A, et al. Abnormalities of synaptic mitochondria in autism spectrum disorder and related neurodevelopmental disorders. J Mol Med (Berl). 2021;99:161–78.CrossRef
75.
go back to reference Lin J, Zhang K, Cao X, et al. iTRAQ-based proteomics analysis of rat cerebral cortex exposed to valproic acid before delivery. ACS Chem Neurosci. 2022;13:648–63.CrossRef Lin J, Zhang K, Cao X, et al. iTRAQ-based proteomics analysis of rat cerebral cortex exposed to valproic acid before delivery. ACS Chem Neurosci. 2022;13:648–63.CrossRef
76.
go back to reference Jiang H, Zhang XJ. Acetylcholinesterase and apoptosis. a novel perspective for an old enzyme. FEBS J. 2008;275:612–7.CrossRef Jiang H, Zhang XJ. Acetylcholinesterase and apoptosis. a novel perspective for an old enzyme. FEBS J. 2008;275:612–7.CrossRef
77.
go back to reference Suzuki K, Sugihara G, Ouchi Y, et al. Reduced acetylcholinesterase activity in the fusiform gyrus in adults with autism spectrum disorders. Arch Gen Psychiatry. 2011;68:306–13.CrossRef Suzuki K, Sugihara G, Ouchi Y, et al. Reduced acetylcholinesterase activity in the fusiform gyrus in adults with autism spectrum disorders. Arch Gen Psychiatry. 2011;68:306–13.CrossRef
78.
go back to reference Kinoshita PF, Leite JA, Orellana AM, et al. The influence of Na(+), K(+)-ATPase on glutamate signaling in neurodegenerative diseases and senescence. Front Physiol. 2016;7:195.CrossRef Kinoshita PF, Leite JA, Orellana AM, et al. The influence of Na(+), K(+)-ATPase on glutamate signaling in neurodegenerative diseases and senescence. Front Physiol. 2016;7:195.CrossRef
79.
go back to reference Petroff OA. GABA and glutamate in the human brain. Neuroscientist. 2002;8:562–73.CrossRef Petroff OA. GABA and glutamate in the human brain. Neuroscientist. 2002;8:562–73.CrossRef
80.
go back to reference Tang X, Jaenisch R, Sur M. The role of GABAergic signalling in neurodevelopmental disorders. Nat Rev Neurosci. 2021;22:290–307.CrossRef Tang X, Jaenisch R, Sur M. The role of GABAergic signalling in neurodevelopmental disorders. Nat Rev Neurosci. 2021;22:290–307.CrossRef
81.
go back to reference Cellot G, Cherubini E. GABAergic signaling as therapeutic target for autism spectrum disorders. Front Pediatr. 2014;2:70.CrossRef Cellot G, Cherubini E. GABAergic signaling as therapeutic target for autism spectrum disorders. Front Pediatr. 2014;2:70.CrossRef
82.
go back to reference Satterstrom FK, Kosmicki JA, Wang J, et al. Large-scale exome sequencing study implicates both developmental and functional changes in the neurobiology of autism. Cell. 2020;180:568-584.e23.CrossRef Satterstrom FK, Kosmicki JA, Wang J, et al. Large-scale exome sequencing study implicates both developmental and functional changes in the neurobiology of autism. Cell. 2020;180:568-584.e23.CrossRef
83.
go back to reference Hou Q, Wang Y, Li Y, et al. A developmental study of abnormal behaviors and altered GABAergic signaling in the VPA-treated rat model of autism. Front Behav Neurosci. 2018;12:182.CrossRef Hou Q, Wang Y, Li Y, et al. A developmental study of abnormal behaviors and altered GABAergic signaling in the VPA-treated rat model of autism. Front Behav Neurosci. 2018;12:182.CrossRef
84.
go back to reference Liu X, Bautista J, Liu E, et al. Imbalance of laminar-specific excitatory and inhibitory circuits of the orbitofrontal cortex in autism. Mol Autism. 2020;11:83.CrossRef Liu X, Bautista J, Liu E, et al. Imbalance of laminar-specific excitatory and inhibitory circuits of the orbitofrontal cortex in autism. Mol Autism. 2020;11:83.CrossRef
85.
go back to reference Hannan AJ, Blakemore C, Katsnelson A, et al. PLC-beta1, activated via mGluRs, mediates activity-dependent differentiation in cerebral cortex. Nat Neurosci. 2001;4:282–8.CrossRef Hannan AJ, Blakemore C, Katsnelson A, et al. PLC-beta1, activated via mGluRs, mediates activity-dependent differentiation in cerebral cortex. Nat Neurosci. 2001;4:282–8.CrossRef
86.
go back to reference Spires TL, Molnár Z, Kind PC, et al. Activity-dependent regulation of synapse and dendritic spine morphology in developing barrel cortex requires phospholipase C-beta1 signalling. Cereb Cortex. 2005;15:385–93.CrossRef Spires TL, Molnár Z, Kind PC, et al. Activity-dependent regulation of synapse and dendritic spine morphology in developing barrel cortex requires phospholipase C-beta1 signalling. Cereb Cortex. 2005;15:385–93.CrossRef
87.
go back to reference Chana G, Laskaris L, Pantelis C, et al. Decreased expression of mGluR5 within the dorsolateral prefrontal cortex in autism and increased microglial number in mGluR5 knockout mice: pathophysiological and neurobehavioral implications. Brain Behav Immun. 2015;49:197–205.CrossRef Chana G, Laskaris L, Pantelis C, et al. Decreased expression of mGluR5 within the dorsolateral prefrontal cortex in autism and increased microglial number in mGluR5 knockout mice: pathophysiological and neurobehavioral implications. Brain Behav Immun. 2015;49:197–205.CrossRef
88.
go back to reference Lee KW, Hong JH, Choi IY, et al. Impaired D2 dopamine receptor function in mice lacking type 5 adenylyl cyclase. J Neurosci. 2002;22:7931–40.CrossRef Lee KW, Hong JH, Choi IY, et al. Impaired D2 dopamine receptor function in mice lacking type 5 adenylyl cyclase. J Neurosci. 2002;22:7931–40.CrossRef
89.
go back to reference Kim H, Kim TK, Kim JE, et al. Adenylyl cyclase-5 in the dorsal striatum function as a molecular switch for the generation of behavioral preferences for cue-directed food choices. Mol Brain. 2014;7:77.CrossRef Kim H, Kim TK, Kim JE, et al. Adenylyl cyclase-5 in the dorsal striatum function as a molecular switch for the generation of behavioral preferences for cue-directed food choices. Mol Brain. 2014;7:77.CrossRef
90.
go back to reference Kim H, Lee Y, Park JY, et al. Loss of adenylyl cyclase type-5 in the dorsal striatum produces autistic-like behaviors. Mol Neurobiol. 2017;54:7994–8008.CrossRef Kim H, Lee Y, Park JY, et al. Loss of adenylyl cyclase type-5 in the dorsal striatum produces autistic-like behaviors. Mol Neurobiol. 2017;54:7994–8008.CrossRef
91.
go back to reference Leblond CS, Nava C, Polge A, et al. Meta-analysis of SHANK mutations in autism spectrum disorders: a gradient of severity in cognitive impairments. PLoS Genet. 2014;10:e1004580.CrossRef Leblond CS, Nava C, Polge A, et al. Meta-analysis of SHANK mutations in autism spectrum disorders: a gradient of severity in cognitive impairments. PLoS Genet. 2014;10:e1004580.CrossRef
92.
go back to reference Boccuto L, Lauri M, Sarasua SM, et al. Prevalence of SHANK3 variants in patients with different subtypes of autism spectrum disorders. Eur J Hum Genet. 2013;21:310–6.CrossRef Boccuto L, Lauri M, Sarasua SM, et al. Prevalence of SHANK3 variants in patients with different subtypes of autism spectrum disorders. Eur J Hum Genet. 2013;21:310–6.CrossRef
93.
go back to reference Monteiro P, Feng G. SHANK proteins: roles at the synapse and in autism spectrum disorder. Nat Rev Neurosci. 2017;18:147–57.CrossRef Monteiro P, Feng G. SHANK proteins: roles at the synapse and in autism spectrum disorder. Nat Rev Neurosci. 2017;18:147–57.CrossRef
94.
go back to reference Hung AY, Futai K, Sala C, et al. Smaller dendritic spines, weaker synaptic transmission, but enhanced spatial learning in mice lacking Shank1. J Neurosci. 2008;28:1697–708.CrossRef Hung AY, Futai K, Sala C, et al. Smaller dendritic spines, weaker synaptic transmission, but enhanced spatial learning in mice lacking Shank1. J Neurosci. 2008;28:1697–708.CrossRef
95.
go back to reference Mao W, Watanabe T, Cho S, et al. Shank1 regulates excitatory synaptic transmission in mouse hippocampal parvalbumin-expressing inhibitory interneurons. Eur J Neurosci. 2015;41:1025–35.CrossRef Mao W, Watanabe T, Cho S, et al. Shank1 regulates excitatory synaptic transmission in mouse hippocampal parvalbumin-expressing inhibitory interneurons. Eur J Neurosci. 2015;41:1025–35.CrossRef
96.
go back to reference Lai MC, Szatmari P. Sex and gender impacts on the behavioural presentation and recognition of autism. Curr Opin Psychiatry. 2020;33:117–23.CrossRef Lai MC, Szatmari P. Sex and gender impacts on the behavioural presentation and recognition of autism. Curr Opin Psychiatry. 2020;33:117–23.CrossRef
97.
go back to reference Roque C, Mendes-Oliveira J, Duarte-Chendo C, et al. The role of G protein-coupled estrogen receptor 1 on neurological disorders. Front Neuroendocrinol. 2019;55:100786.CrossRef Roque C, Mendes-Oliveira J, Duarte-Chendo C, et al. The role of G protein-coupled estrogen receptor 1 on neurological disorders. Front Neuroendocrinol. 2019;55:100786.CrossRef
99.
go back to reference Crider A, Thakkar R, Ahmed AO, et al. Dysregulation of estrogen receptor beta (ERβ), aromatase (CYP19A1), and ER co-activators in the middle frontal gyrus of autism spectrum disorder subjects. Mol Autism. 2014;5:46.CrossRef Crider A, Thakkar R, Ahmed AO, et al. Dysregulation of estrogen receptor beta (ERβ), aromatase (CYP19A1), and ER co-activators in the middle frontal gyrus of autism spectrum disorder subjects. Mol Autism. 2014;5:46.CrossRef
100.
go back to reference Altun H, Kurutaş EB, Şahin N, et al. Decreased levels of G protein-coupled estrogen receptor in children with autism spectrum disorders. Psychiatry Res. 2017;257:67–71.CrossRef Altun H, Kurutaş EB, Şahin N, et al. Decreased levels of G protein-coupled estrogen receptor in children with autism spectrum disorders. Psychiatry Res. 2017;257:67–71.CrossRef
Metadata
Title
Continuous high-frequency deep brain stimulation of the anterior insula modulates autism-like behavior in a valproic acid-induced rat model
Authors
Lifei Xiao
Shucai Jiang
Yangyang Wang
Caibin Gao
Cuicui Liu
Xianhao Huo
Wenchao Li
Baorui Guo
Chaofan Wang
Yu Sun
Anni Wang
Yan Feng
Feng Wang
Tao Sun
Publication date
01-12-2022
Publisher
BioMed Central
Published in
Journal of Translational Medicine / Issue 1/2022
Electronic ISSN: 1479-5876
DOI
https://doi.org/10.1186/s12967-022-03787-9

Other articles of this Issue 1/2022

Journal of Translational Medicine 1/2022 Go to the issue
Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine

Highlights from the ACC 2024 Congress

Year in Review: Pediatric cardiology

Watch Dr. Anne Marie Valente present the last year's highlights in pediatric and congenital heart disease in the official ACC.24 Year in Review session.

Year in Review: Pulmonary vascular disease

The last year's highlights in pulmonary vascular disease are presented by Dr. Jane Leopold in this official video from ACC.24.

Year in Review: Valvular heart disease

Watch Prof. William Zoghbi present the last year's highlights in valvular heart disease from the official ACC.24 Year in Review session.

Year in Review: Heart failure and cardiomyopathies

Watch this official video from ACC.24. Dr. Biykem Bozkurt discusses last year's major advances in heart failure and cardiomyopathies.