151
|
Advancing the understanding of autism disease mechanisms through genetics. Nat Med 2016; 22:345-61. [PMID: 27050589 DOI: 10.1038/nm.4071] [Citation(s) in RCA: 560] [Impact Index Per Article: 62.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 02/26/2016] [Indexed: 12/11/2022]
Abstract
Progress in understanding the genetic etiology of autism spectrum disorders (ASD) has fueled remarkable advances in our understanding of its potential neurobiological mechanisms. Yet, at the same time, these findings highlight extraordinary causal diversity and complexity at many levels ranging from molecules to circuits and emphasize the gaps in our current knowledge. Here we review current understanding of the genetic architecture of ASD and integrate genetic evidence, neuropathology and studies in model systems with how they inform mechanistic models of ASD pathophysiology. Despite the challenges, these advances provide a solid foundation for the development of rational, targeted molecular therapies.
Collapse
|
152
|
Abstract
Abstract
ASD research is at an important crossroads. The ASD diagnosis is important for assigning a child to early behavioral intervention and explaining a child’s condition. But ASD research has not provided a diagnosis-specific medical treatment, or a consistent early predictor, or a unified life course. If the ASD diagnosis also lacks biological and construct validity, a shift away from studying ASD-defined samples would be warranted. Consequently, this paper reviews recent findings for the neurobiological validity of ASD, the construct validity of ASD diagnostic criteria, and the construct validity of ASD spectrum features. The findings reviewed indicate that the ASD diagnosis lacks biological and construct validity. The paper concludes with proposals for research going forward.
Collapse
|
153
|
Mullins C, Fishell G, Tsien RW. Unifying Views of Autism Spectrum Disorders: A Consideration of Autoregulatory Feedback Loops. Neuron 2016; 89:1131-1156. [PMID: 26985722 DOI: 10.1016/j.neuron.2016.02.017] [Citation(s) in RCA: 135] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/08/2016] [Indexed: 12/31/2022]
Abstract
Understanding the mechanisms underlying autism spectrum disorders (ASDs) is a challenging goal. Here we review recent progress on several fronts, including genetics, proteomics, biochemistry, and electrophysiology, that raise motivation for forming a viable pathophysiological hypothesis. In place of a traditionally unidirectional progression, we put forward a framework that extends homeostatic hypotheses by explicitly emphasizing autoregulatory feedback loops and known synaptic biology. The regulated biological feature can be neuronal electrical activity, the collective strength of synapses onto a dendritic branch, the local concentration of a signaling molecule, or the relative strengths of synaptic excitation and inhibition. The sensor of the biological variable (which we have termed the homeostat) engages mechanisms that operate as negative feedback elements to keep the biological variable tightly confined. We categorize known ASD-associated gene products according to their roles in such feedback loops and provide detailed commentary for exemplar genes within each module.
Collapse
Affiliation(s)
- Caitlin Mullins
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Langone Medical Center, New York, NY 10016, USA
| | - Gord Fishell
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Langone Medical Center, New York, NY 10016, USA
| | - Richard W Tsien
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Langone Medical Center, New York, NY 10016, USA.
| |
Collapse
|
154
|
Duringer J, Fombonne E, Craig M. No Association between Mycotoxin Exposure and Autism: A Pilot Case-Control Study in School-Aged Children. Toxins (Basel) 2016; 8:toxins8070224. [PMID: 27447670 PMCID: PMC4963856 DOI: 10.3390/toxins8070224] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2016] [Revised: 07/06/2016] [Accepted: 07/08/2016] [Indexed: 01/11/2023] Open
Abstract
Evaluation of environmental risk factors in the development of autism spectrum disorder (ASD) is needed for a more complete understanding of disease etiology and best approaches for prevention, diagnosis, and treatment. A pilot experiment in 54 children (n = 25 ASD, n = 29 controls; aged 12.4 ± 3.9 years) screened for 87 urinary mycotoxins via liquid chromatography-tandem mass spectrometry to assess current exposure. Zearalenone, zearalenone-4-glucoside, 3-acetyldeoxynivalenol, and altenuene were detected in 9/54 (20%) samples, most near the limit of detection. No mycotoxin/group of mycotoxins was associated with ASD-diagnosed children. To identify potential correlates of mycotoxin presence in urine, we further compared the nine subjects where a urinary mycotoxin was confirmed to the remaining 45 participants and found no difference based on the presence or absence of mycotoxin for age (t-test; p = 0.322), gender (Fisher’s exact test; p = 0.456), exposure or not to selective serotonin reuptake inhibitors (Fisher’s exact test; p = 0.367), or to other medications (Fisher’s exact test; p = 1.00). While no positive association was found, more sophisticated sample preparation techniques and instrumentation, coupled with selectivity for a smaller group of mycotoxins, could improve sensitivity and detection. Further, broadening sampling to in utero (mothers) and newborn-toddler years would cover additional exposure windows.
Collapse
Affiliation(s)
- Jennifer Duringer
- Department of Environmental & Molecular Toxicology, Oregon State University, 139 Oak Creek Building, Corvallis, OR 97331, USA.
| | - Eric Fombonne
- Department of Psychiatry, Institute for Development & Disability, Oregon Health & Science University, 840 SW Gaines St., Portland, OR 97239, USA.
| | - Morrie Craig
- College of Veterinary Medicine, Oregon State University, 105 Magruder Hall, Corvallis, OR 97331, USA.
| |
Collapse
|
155
|
Noroozi R, Taheri M, Movafagh A, Mirfakhraie R, Solgi G, Sayad A, Mazdeh M, Darvish H. Glutamate receptor, metabotropic 7 (GRM7) gene variations and susceptibility to autism: A case-control study. Autism Res 2016; 9:1161-1168. [DOI: 10.1002/aur.1640] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 04/04/2016] [Indexed: 12/29/2022]
Affiliation(s)
- Rezvan Noroozi
- From the Department of Medical Genetics, Faculty of Medicine; Shahid Beheshti University of Medical Sciences; Tehran Iran
| | - Mohammad Taheri
- From the Department of Medical Genetics, Faculty of Medicine; Shahid Beheshti University of Medical Sciences; Tehran Iran
| | - Abolfazl Movafagh
- From the Department of Medical Genetics, Faculty of Medicine; Shahid Beheshti University of Medical Sciences; Tehran Iran
| | - Reza Mirfakhraie
- From the Department of Medical Genetics, Faculty of Medicine; Shahid Beheshti University of Medical Sciences; Tehran Iran
| | - Ghasem Solgi
- Department of Immunology, School of Medicine; Hamadan University of Medical Sciences; Hamadan Iran
| | - Arezou Sayad
- From the Department of Medical Genetics, Faculty of Medicine; Shahid Beheshti University of Medical Sciences; Tehran Iran
| | - Mehrdokht Mazdeh
- Department of Neurology, School of Medicine; Hamadan University of Medical Sciences; Hamadan Iran
| | - Hossein Darvish
- From the Department of Medical Genetics, Faculty of Medicine; Shahid Beheshti University of Medical Sciences; Tehran Iran
| |
Collapse
|
156
|
Haas MA, Ngo L, Li SS, Schleich S, Qu Z, Vanyai HK, Cullen HD, Cardona-Alberich A, Gladwyn-Ng IE, Pagnamenta AT, Taylor JC, Stewart H, Kini U, Duncan KE, Teleman AA, Keays DA, Heng JIT. De Novo Mutations in DENR Disrupt Neuronal Development and Link Congenital Neurological Disorders to Faulty mRNA Translation Re-initiation. Cell Rep 2016; 15:2251-2265. [PMID: 27239039 PMCID: PMC4906373 DOI: 10.1016/j.celrep.2016.04.090] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 03/23/2016] [Accepted: 04/30/2016] [Indexed: 02/05/2023] Open
Abstract
Disruptions to neuronal mRNA translation are hypothesized to underlie human neurodevelopmental syndromes. Notably, the mRNA translation re-initiation factor DENR is a regulator of eukaryotic translation and cell growth, but its mammalian functions are unknown. Here, we report that Denr influences the migration of murine cerebral cortical neurons in vivo with its binding partner Mcts1, whereas perturbations to Denr impair the long-term positioning, dendritic arborization, and dendritic spine characteristics of postnatal projection neurons. We characterized de novo missense mutations in DENR (p.C37Y and p.P121L) detected in two unrelated human subjects diagnosed with brain developmental disorder to find that each variant impairs the function of DENR in mRNA translation re-initiation and disrupts the migration and terminal branching of cortical neurons in different ways. Thus, our findings link human brain disorders to impaired mRNA translation re-initiation through perturbations in DENR (OMIM: 604550) function in neurons.
Collapse
Affiliation(s)
- Matilda A Haas
- EMBL Australia, The Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia
| | - Linh Ngo
- EMBL Australia, The Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia; The Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, the University of Western Australia, Nedlands, WA 6009, Australia
| | - Shan Shan Li
- EMBL Australia, The Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia
| | - Sibylle Schleich
- German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, 69120 Heidelberg, Germany
| | - Zhengdong Qu
- EMBL Australia, The Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia
| | - Hannah K Vanyai
- The Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, the University of Western Australia, Nedlands, WA 6009, Australia
| | - Hayley D Cullen
- The Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, the University of Western Australia, Nedlands, WA 6009, Australia
| | - Aida Cardona-Alberich
- Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany
| | - Ivan E Gladwyn-Ng
- EMBL Australia, The Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia; The Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, the University of Western Australia, Nedlands, WA 6009, Australia
| | - Alistair T Pagnamenta
- National Institute for Health Research Biomedical Research Centre, Wellcome Trust Centre for Human Genetics, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Jenny C Taylor
- National Institute for Health Research Biomedical Research Centre, Wellcome Trust Centre for Human Genetics, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Helen Stewart
- Department of Clinical Genetics, Churchill Hospital, Old Road, Headington, Oxford OX3 7LE, UK
| | - Usha Kini
- Department of Clinical Genetics, Churchill Hospital, Old Road, Headington, Oxford OX3 7LE, UK
| | - Kent E Duncan
- Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany
| | - Aurelio A Teleman
- German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, 69120 Heidelberg, Germany
| | - David A Keays
- Institute of Molecular Pathology, Dr Bohr-Gasse, Vienna 1030, Austria
| | - Julian I-T Heng
- EMBL Australia, The Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia; The Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, the University of Western Australia, Nedlands, WA 6009, Australia.
| |
Collapse
|
157
|
Eilam-Stock T, Wu T, Spagna A, Egan LJ, Fan J. Neuroanatomical Alterations in High-Functioning Adults with Autism Spectrum Disorder. Front Neurosci 2016; 10:237. [PMID: 27313505 PMCID: PMC4889574 DOI: 10.3389/fnins.2016.00237] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 05/12/2016] [Indexed: 12/14/2022] Open
Abstract
Autism spectrum disorder (ASD) is a pervasive neurodevelopmental condition, affecting cognition and behavior throughout the life span. With recent advances in neuroimaging techniques and analytical approaches, a considerable effort has been directed toward identifying the neuroanatomical underpinnings of ASD. While gray-matter abnormalities have been found throughout cortical, subcortical, and cerebellar regions of affected individuals, there is currently little consistency across findings, partly due to small sample-sizes and great heterogeneity among participants in previous studies. Here, we report voxel-based morphometry of structural magnetic resonance images in a relatively large sample of high-functioning adults with ASD (n = 66) and matched typically-developing controls (n = 66) drawn from multiple studies. We found decreased gray-matter volume in posterior brain regions, including the posterior hippocampus and cuneus, as well as increased gray-matter volume in frontal brain regions, including the medial prefrontal cortex, superior and inferior frontal gyri, and middle temporal gyrus in individuals with ASD. We discuss our results in relation to findings obtained in previous studies, as well as their potential clinical implications.
Collapse
Affiliation(s)
- Tehila Eilam-Stock
- Department of Psychiatry, Icahn School of Medicine at Mount SinaiNew York, NY, USA; Department of Psychology, Queens College, City University of New YorkFlushing, NY, USA; The Graduate Center, City University of New YorkNew York, NY, USA
| | - Tingting Wu
- Department of Psychology, Queens College, City University of New York Flushing, NY, USA
| | - Alfredo Spagna
- Department of Psychiatry, Icahn School of Medicine at Mount SinaiNew York, NY, USA; Department of Psychology, Queens College, City University of New YorkFlushing, NY, USA
| | - Laura J Egan
- Department of Psychiatry, Icahn School of Medicine at Mount SinaiNew York, NY, USA; Department of Psychology, Queens College, City University of New YorkFlushing, NY, USA
| | - Jin Fan
- Department of Psychiatry, Icahn School of Medicine at Mount SinaiNew York, NY, USA; Department of Psychology, Queens College, City University of New YorkFlushing, NY, USA; The Graduate Center, City University of New YorkNew York, NY, USA; Department of Neuroscience, Icahn School of Medicine at Mount SinaiNew York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount SinaiNew York, NY, USA
| |
Collapse
|
158
|
Ergaz Z, Weinstein-Fudim L, Ornoy A. Genetic and non-genetic animal models for autism spectrum disorders (ASD). Reprod Toxicol 2016; 64:116-40. [PMID: 27142188 DOI: 10.1016/j.reprotox.2016.04.024] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 04/18/2016] [Accepted: 04/25/2016] [Indexed: 12/13/2022]
Abstract
Autism spectrum disorder (ASD) is associated, in addition to complex genetic factors, with a variety of prenatal, perinatal and postnatal etiologies. We discuss the known animal models, mostly in mice and rats, of ASD that helps us to understand the etiology, pathogenesis and treatment of human ASD. We describe only models where behavioral testing has shown autistic like behaviors. Some genetic models mimic known human syndromes like fragile X where ASD is part of the clinical picture, and others are without defined human syndromes. Among the environmentally induced ASD models in rodents, the most common model is the one induced by valproic acid (VPA) either prenatally or early postnatally. VPA induces autism-like behaviors following single exposure during different phases of brain development, implying that the mechanism of action is via a general biological mechanism like epigenetic changes. Maternal infection and inflammation are also associated with ASD in man and animal models.
Collapse
Affiliation(s)
- Zivanit Ergaz
- Laboratory of Teratology, Department of Medical Neurobiology Hebrew University Hadassah Medical School and Hadassah Hospital, Jerusalem, Israel
| | - Liza Weinstein-Fudim
- Laboratory of Teratology, Department of Medical Neurobiology Hebrew University Hadassah Medical School and Hadassah Hospital, Jerusalem, Israel
| | - Asher Ornoy
- Laboratory of Teratology, Department of Medical Neurobiology Hebrew University Hadassah Medical School and Hadassah Hospital, Jerusalem, Israel.
| |
Collapse
|
159
|
Ben-Reuven L, Reiner O. Modeling the autistic cell: iPSCs recapitulate developmental principles of syndromic and nonsyndromic ASD. Dev Growth Differ 2016; 58:481-91. [PMID: 27111774 DOI: 10.1111/dgd.12280] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 02/26/2016] [Accepted: 03/01/2016] [Indexed: 12/13/2022]
Abstract
The opportunity to model autism spectrum disorders (ASD) through generation of patient-derived induced pluripotent stem cells (iPSCs) is currently an emerging topic. Wide-scale research of altered brain circuits in syndromic ASD, including Rett Syndrome, Fragile X Syndrome, Angelman's Syndrome and sporadic Schizophrenia, was made possible through animal models. However, possibly due to species differences, and to the possible contribution of epigenetics in the pathophysiology of these diseases, animal models fail to recapitulate many aspects of ASD. With the advent of iPSCs technology, 3D cultures of patient-derived cells are being used to study complex neuronal phenotypes, including both syndromic and nonsyndromic ASD. Here, we review recent advances in using iPSCs to study various aspects of the ASD neuropathology, with emphasis on the efforts to create in vitro model systems for syndromic and nonsyndromic ASD. We summarize the main cellular activity phenotypes and aberrant genetic interaction networks that were found in iPSC-derived neurons of syndromic and nonsyndromic autistic patients.
Collapse
Affiliation(s)
- Lihi Ben-Reuven
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Orly Reiner
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
160
|
Abstract
PURPOSE OF REVIEW Many studies have reported that individuals with autism spectrum disorder (ASD) have different brain connectivity patterns compared with typically developing individuals. However, the results of more recent studies do not unanimously support the traditional view in which individuals with ASD have lower connectivity between distant brain regions and increased connectivity within local brain regions. In this review, we discuss different methods for measuring brain connectivity and how the use of different metrics may contribute to the lack of convergence of investigations of connectivity in ASD. RECENT FINDINGS The discrepancy in brain connectivity results across studies may be due to important methodological factors, such as the connectivity measure applied, the age of patients studied, the brain region(s) examined, and the time interval and frequency band(s) in which connectivity was analyzed. SUMMARY We conclude that more sophisticated electroencephalography analytic approaches should be utilized to more accurately infer causation and directionality of information transfer between brain regions, which may show dynamic changes of functional connectivity in the brain. Moreover, further investigations of connectivity with respect to behavior and clinical phenotype are needed to probe underlying brain networks implicated in core deficits of ASD.
Collapse
Affiliation(s)
| | | | - Sandra K. Loo
- UCLA Semel Institute of Neuroscience and Human Behavior, David Geffen School of Medicine, Los Angeles, California, USA
| | - Shafali S. Jeste
- UCLA Semel Institute of Neuroscience and Human Behavior, David Geffen School of Medicine, Los Angeles, California, USA
| |
Collapse
|
161
|
Pearson BL, Simon JM, McCoy ES, Salazar G, Fragola G, Zylka MJ. Identification of chemicals that mimic transcriptional changes associated with autism, brain aging and neurodegeneration. Nat Commun 2016; 7:11173. [PMID: 27029645 PMCID: PMC4821887 DOI: 10.1038/ncomms11173] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 02/29/2016] [Indexed: 12/13/2022] Open
Abstract
Environmental factors, including pesticides, have been linked to autism and neurodegeneration risk using retrospective epidemiological studies. Here we sought to prospectively identify chemicals that share transcriptomic signatures with neurological disorders, by exposing mouse cortical neuron-enriched cultures to hundreds of chemicals commonly found in the environment and on food. We find that rotenone, a pesticide associated with Parkinson's disease risk, and certain fungicides, including pyraclostrobin, trifloxystrobin, famoxadone and fenamidone, produce transcriptional changes in vitro that are similar to those seen in brain samples from humans with autism, advanced age and neurodegeneration (Alzheimer's disease and Huntington's disease). These chemicals stimulate free radical production and disrupt microtubules in neurons, effects that can be reduced by pretreating with a microtubule stabilizer, an antioxidant, or with sulforaphane. Our study provides an approach to prospectively identify environmental chemicals that transcriptionally mimic autism and other brain disorders.
Collapse
Affiliation(s)
- Brandon L. Pearson
- Department of Cell Biology and Physiology, UNC Neuroscience Center, University of North Carolina at Chapel Hill, 111 Mason Farm Road, Chapel Hill, North Carolina 27599-7545, USA
- Carolina Institute for Developmental Disabilities, University of North Carolina Chapel Hill, Chapel Hill, North Carolina 27599-7255, USA
| | - Jeremy M. Simon
- Department of Cell Biology and Physiology, UNC Neuroscience Center, University of North Carolina at Chapel Hill, 111 Mason Farm Road, Chapel Hill, North Carolina 27599-7545, USA
- Carolina Institute for Developmental Disabilities, University of North Carolina Chapel Hill, Chapel Hill, North Carolina 27599-7255, USA
| | - Eric S. McCoy
- Department of Cell Biology and Physiology, UNC Neuroscience Center, University of North Carolina at Chapel Hill, 111 Mason Farm Road, Chapel Hill, North Carolina 27599-7545, USA
| | - Gabriela Salazar
- Department of Cell Biology and Physiology, UNC Neuroscience Center, University of North Carolina at Chapel Hill, 111 Mason Farm Road, Chapel Hill, North Carolina 27599-7545, USA
| | - Giulia Fragola
- Department of Cell Biology and Physiology, UNC Neuroscience Center, University of North Carolina at Chapel Hill, 111 Mason Farm Road, Chapel Hill, North Carolina 27599-7545, USA
| | - Mark J. Zylka
- Department of Cell Biology and Physiology, UNC Neuroscience Center, University of North Carolina at Chapel Hill, 111 Mason Farm Road, Chapel Hill, North Carolina 27599-7545, USA
- Carolina Institute for Developmental Disabilities, University of North Carolina Chapel Hill, Chapel Hill, North Carolina 27599-7255, USA
| |
Collapse
|
162
|
Gangi DN, Messinger DS, Martin ER, Cuccaro ML. Dopaminergic variants in siblings at high risk for autism: Associations with initiating joint attention. Autism Res 2016; 9:1142-1150. [PMID: 26990357 DOI: 10.1002/aur.1623] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Accepted: 02/16/2016] [Indexed: 12/22/2022]
Abstract
Younger siblings of children with autism spectrum disorder (ASD; high-risk siblings) exhibit lower levels of initiating joint attention (IJA; sharing an object or experience with a social partner through gaze and/or gesture) than low-risk siblings of children without ASD. However, high-risk siblings also exhibit substantial variability in this domain. The neurotransmitter dopamine is linked to brain areas associated with reward, motivation, and attention, and common dopaminergic variants have been associated with attention difficulties. We examined whether these common dopaminergic variants, DRD4 and DRD2, explain variability in IJA in high-risk (n = 55) and low-risk (n = 38) siblings. IJA was assessed in the first year during a semi-structured interaction with an examiner. DRD4 and DRD2 genotypes were coded according to associated dopaminergic functioning to create a gene score, with higher scores indicating more genotypes associated with less efficient dopaminergic functioning. Higher dopamine gene scores (indicative of less efficient dopaminergic functioning) were associated with lower levels of IJA in the first year for high-risk siblings, while the opposite pattern emerged in low-risk siblings. Findings suggest differential susceptibility-IJA was differentially associated with dopaminergic functioning depending on familial ASD risk. Understanding genes linked to ASD-relevant behaviors in high-risk siblings will aid in early identification of children at greatest risk for difficulties in these behavioral domains, facilitating targeted prevention and intervention. Autism Res 2016, 9: 1142-1150. © 2016 International Society for Autism Research, Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Devon N Gangi
- Department of Psychology, University of Miami, Florida.,MIND Institute, University of California, Davis, California
| | - Daniel S Messinger
- Departments of Psychology, Pediatrics, Electrical & Computer Engineering, and Music Engineering, University of Miami, Florida
| | - Eden R Martin
- University of Miami Miller School of Medicine and Hussman Institute for Human Genomics, Florida
| | - Michael L Cuccaro
- University of Miami Miller School of Medicine and Hussman Institute for Human Genomics, Florida
| |
Collapse
|
163
|
Grabrucker S, Boeckers TM, Grabrucker AM. Gender Dependent Evaluation of Autism like Behavior in Mice Exposed to Prenatal Zinc Deficiency. Front Behav Neurosci 2016; 10:37. [PMID: 26973485 PMCID: PMC4776245 DOI: 10.3389/fnbeh.2016.00037] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 02/19/2016] [Indexed: 01/09/2023] Open
Abstract
Zinc deficiency has recently been linked to the etiology of autism spectrum disorders (ASD) as environmental risk factor. With an estimated 17% of the world population being at risk of zinc deficiency, especially zinc deficiency during pregnancy might be a common occurrence, also in industrialized nations. On molecular level, zinc deficiency has been shown to affect a signaling pathway at glutamatergic synapses that has previously been identified through genetic mutations in ASD patients, the Neurexin-Neuroligin-Shank pathway, via altering zinc binding Shank family members. In particular, prenatal zinc deficient but not acute zinc deficient animals have been reported to display autism like behavior in some behavioral tests. However, a full behavioral analysis of a possible autism like behavior has been lacking so far. Here, we performed an extensive behavioral phenotyping of mice born from mothers with mild zinc deficiency during all trimesters of pregnancy. Prenatal zinc deficient animals were investigated as adults and gender differences were assessed. Our results show that prenatal zinc deficient mice display increased anxiety, deficits in nest building and various social interaction paradigm, as well as mild alterations in ultrasonic vocalizations. A gender specific analysis revealed only few sex specific differences. Taken together, given that similar behavioral abnormalities as reported here are frequently observed in ASD mouse models, we conclude that prenatal zinc deficient animals even without specific genetic susceptibility for ASD, already show some features of ASD like behavior.
Collapse
Affiliation(s)
| | | | - Andreas M Grabrucker
- Institute for Anatomy and Cell Biology, Ulm UniversityUlm, Germany; WG Molecular Analysis of Synaptopathies, Neurology Department, Neurocenter of Ulm UniversityUlm, Germany
| |
Collapse
|
164
|
Molenhuis RT, Bruining H, Remmelink E, de Visser L, Loos M, Burbach JPH, Kas MJH. Limited impact of Cntn4 mutation on autism-related traits in developing and adult C57BL/6J mice. J Neurodev Disord 2016; 8:6. [PMID: 26958094 PMCID: PMC4782374 DOI: 10.1186/s11689-016-9140-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 02/21/2016] [Indexed: 11/15/2022] Open
Abstract
Background Mouse models offer an essential tool to unravel the impact of genetic mutations on autism-related phenotypes. The behavioral impact of some important candidate gene models for autism spectrum disorder (ASD) has not yet been studied, and existing characterizations mostly describe behavioral phenotypes at adult ages, disregarding the developmental nature of the disorder. In this context, the behavioral influence of CNTN4, one of the strongest suggested ASD candidate genes, is unknown. Here, we used our recently established developmental test battery to characterize the consequences of disruption of contactin 4 (Cntn4) on neurological, sensory, cognitive, and behavioral phenotypes across different developmental stages. Methods C57BL/6J mice with heterozygous and homozygous disruption of Cntn4 were studied through an extensive, partially longitudinal, test battery at various developmental stages, including various paradigms testing social and restricted repetitive behaviors. Results Developmental neurological and cognitive screenings revealed no significant differences between genotypes, and ASD-related behavioral domains were also unchanged in Cntn4-deficient versus wild-type mice. The impact of Cntn4-deficiency was found to be limited to increased startle responsiveness following auditory stimuli of different high amplitudes in heterozygous and homozygous Cntn4-deficient mice and enhanced acquisition in a spatial learning task in homozygous mice. Conclusions Disruption of Cntn4 in the C57BL/6J background does not affect specific autism-related phenotypes in developing or adult mice but causes subtle non-disorder specific changes in sensory behavioral responses and cognitive performance. Electronic supplementary material The online version of this article (doi:10.1186/s11689-016-9140-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Remco T Molenhuis
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Hilgo Bruining
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands ; Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Esther Remmelink
- Sylics (Synaptologics BV), Amsterdam, The Netherlands ; Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, VU University, Amsterdam, The Netherlands ; Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, VU University, Amsterdam, The Netherlands
| | - Leonie de Visser
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Maarten Loos
- Sylics (Synaptologics BV), Amsterdam, The Netherlands
| | - J Peter H Burbach
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Martien J H Kas
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
165
|
Moos WH, Maneta E, Pinkert CA, Irwin MH, Hoffman ME, Faller DV, Steliou K. Epigenetic Treatment of Neuropsychiatric Disorders: Autism and Schizophrenia. Drug Dev Res 2016; 77:53-72. [PMID: 26899191 DOI: 10.1002/ddr.21295] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Neuropsychiatric disorders are a heterogeneous group of conditions that often share underlying mitochondrial dysfunction and biological pathways implicated in their pathogenesis, progression, and treatment. To date, these disorders have proven notoriously resistant to molecular-targeted therapies, and clinical options are relegated to interventional types, which do not address the core symptoms of the disease. In this review, we discuss emerging epigenetic-driven approaches using novel acylcarnitine esters (carnitinoids) that act on master regulators of antioxidant and cytoprotective genes and mitophagic pathways. These carnitinoids are actively transported, mitochondria-localizing, biomimetic coenzyme A surrogates of short-chain fatty acids, which inhibit histone deacetylase and may reinvigorate synaptic plasticity and protect against neuronal damage. We outline these neuroprotective effects in the context of treatment of neuropsychiatric disorders such as autism spectrum disorder and schizophrenia.
Collapse
Affiliation(s)
- Walter H Moos
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of California San Francisco, San Francisco, CA, USA.,SRI Biosciences, A Division of SRI International, Menlo Park, CA, USA
| | - Eleni Maneta
- Department of Psychiatry, Boston Children's Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - Carl A Pinkert
- Department of Biological Sciences, College of Arts and Sciences, The University of Alabama, Tuscaloosa, AL, USA.,Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA
| | - Michael H Irwin
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA
| | - Michelle E Hoffman
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA
| | - Douglas V Faller
- Cancer Research Center, Boston University School of Medicine, Boston, MA, USA
| | - Kosta Steliou
- Cancer Research Center, Boston University School of Medicine, Boston, MA, USA.,PhenoMatriX, Inc., Boston, MA, USA
| |
Collapse
|
166
|
Rothwell PE. Autism Spectrum Disorders and Drug Addiction: Common Pathways, Common Molecules, Distinct Disorders? Front Neurosci 2016; 10:20. [PMID: 26903789 PMCID: PMC4742554 DOI: 10.3389/fnins.2016.00020] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 01/15/2016] [Indexed: 11/17/2022] Open
Abstract
Autism spectrum disorders (ASDs) and drug addiction do not share substantial comorbidity or obvious similarities in etiology or symptomatology. It is thus surprising that a number of recent studies implicate overlapping neural circuits and molecular signaling pathways in both disorders. The purpose of this review is to highlight this emerging intersection and consider implications for understanding the pathophysiology of these seemingly distinct disorders. One area of overlap involves neural circuits and neuromodulatory systems in the striatum and basal ganglia, which play an established role in addiction and reward but are increasingly implicated in clinical and preclinical studies of ASDs. A second area of overlap relates to molecules like Fragile X mental retardation protein (FMRP) and methyl CpG-binding protein-2 (MECP2), which are best known for their contribution to the pathogenesis of syndromic ASDs, but have recently been shown to regulate behavioral and neurobiological responses to addictive drug exposure. These shared pathways and molecules point to common dimensions of behavioral dysfunction, including the repetition of behavioral patterns and aberrant reward processing. The synthesis of knowledge gained through parallel investigations of ASDs and addiction may inspire the design of new therapeutic interventions to correct common elements of striatal dysfunction.
Collapse
Affiliation(s)
- Patrick E Rothwell
- Department of Neuroscience, University of Minnesota Minneapolis, MN, USA
| |
Collapse
|
167
|
Prevalence of Autism Spectrum Disorders in Guanajuato, Mexico: The Leon survey. J Autism Dev Disord 2016; 46:1669-85. [DOI: 10.1007/s10803-016-2696-6] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
168
|
Young AMH, Chakrabarti B, Roberts D, Lai MC, Suckling J, Baron-Cohen S. From molecules to neural morphology: understanding neuroinflammation in autism spectrum condition. Mol Autism 2016; 7:9. [PMID: 26793298 PMCID: PMC4719563 DOI: 10.1186/s13229-016-0068-x] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 01/04/2016] [Indexed: 12/31/2022] Open
Abstract
Growing evidence points toward a critical role for early (prenatal) atypical neurodevelopmental processes in the aetiology of autism spectrum condition (ASC). One such process that could impact early neural development is inflammation. We review the evidence for atypical expression of molecular markers in the amniotic fluid, serum, cerebrospinal fluid (CSF), and the brain parenchyma that suggest a role for inflammation in the emergence of ASC. This is complemented with a number of neuroimaging and neuropathological studies describing microglial activation. Implications for treatment are discussed.
Collapse
Affiliation(s)
- Adam M H Young
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Douglas House, 18B Trumpington Road, Cambridge, UK ; School of Clinical Medicine, Addenbrookes Hospital, University of Cambridge, Cambridge, UK
| | - Bhismadev Chakrabarti
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Douglas House, 18B Trumpington Road, Cambridge, UK ; Centre for Integrative Neuroscience and Neurodynamics, School of Psychology and Clinical Language Science, University of Reading, Reading, UK
| | - David Roberts
- School of Clinical Medicine, Addenbrookes Hospital, University of Cambridge, Cambridge, UK
| | - Meng-Chuan Lai
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Douglas House, 18B Trumpington Road, Cambridge, UK ; Centre for Addiction and Mental Health and Department of Psychiatry, University of Toronto, Toronto, Canada ; Department of Psychiatry, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - John Suckling
- Brain Mapping Unit, Department of Psychiatry, University of Cambridge, Cambridge, UK ; Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK
| | - Simon Baron-Cohen
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Douglas House, 18B Trumpington Road, Cambridge, UK ; CLASS Clinic, Cambridgeshire and Peterborough NHS Foundation Trust, Cambridge, UK
| |
Collapse
|
169
|
Affiliation(s)
- Enrique F. Schisterman
- Epidemiology Branch; Division of Intramural Population Health Research; Eunice Kennedy Shriver National Institute of Child Health and Human Development; Rockville MD USA
| | - Lindsey A. Sjaarda
- Epidemiology Branch; Division of Intramural Population Health Research; Eunice Kennedy Shriver National Institute of Child Health and Human Development; Rockville MD USA
| |
Collapse
|
170
|
Peñagarikano O. New Therapeutic Options for Autism Spectrum Disorder: Experimental Evidences. Exp Neurobiol 2015; 24:301-11. [PMID: 26713078 PMCID: PMC4688330 DOI: 10.5607/en.2015.24.4.301] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 11/25/2015] [Accepted: 11/25/2015] [Indexed: 02/07/2023] Open
Abstract
Autism spectrum disorder (ASD) is characterized by impairment in two behavioral domains: social interaction/communication together with the presence of stereotyped behaviors and restricted interests. The heterogeneity in the phenotype among patients and the complex etiology of the disorder have long impeded the advancement of the development of successful pharmacotherapies. However, in the recent years, the integration of findings of multiple levels of research, from human genetics to mouse models, have made considerable progress towards the understanding of ASD pathophysiology, allowing the development of more effective targeted drug therapies. The present review discusses the current state of pharmacological research in ASD based on the emerging common pathophysiology signature.
Collapse
Affiliation(s)
- Olga Peñagarikano
- Department of Pharmacology, School of Medicine, University of the Basque Country, Sarriena s/n, Leioa 48940, Spain
| |
Collapse
|
171
|
Gordon A, Salomon D, Barak N, Pen Y, Tsoory M, Kimchi T, Peles E. Expression of Cntnap2 (Caspr2) in multiple levels of sensory systems. Mol Cell Neurosci 2015; 70:42-53. [PMID: 26647347 DOI: 10.1016/j.mcn.2015.11.012] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 10/01/2015] [Accepted: 11/27/2015] [Indexed: 01/09/2023] Open
Abstract
Genome-wide association studies and copy number variation analyses have linked contactin associated protein 2 (Caspr2, gene name Cntnap2) with autism spectrum disorder (ASD). In line with these findings, mice lacking Caspr2 (Cntnap2(-/-)) were shown to have core autism-like deficits including abnormal social behavior and communication, and behavior inflexibility. However the role of Caspr2 in ASD pathogenicity remains unclear. Here we have generated a new Caspr2:tau-LacZ knock-in reporter line (Cntnap2(tlacz/tlacz)), which enabled us to monitor the neuronal circuits in the brain expressing Caspr2. We show that Caspr2 is expressed in many brain regions and produced a comprehensive report of Caspr2 expression. Moreover, we found that Caspr2 marks all sensory modalities: it is expressed in distinct brain regions involved in different sensory processings and is present in all primary sensory organs. Olfaction-based behavioral tests revealed that mice lacking Caspr2 exhibit abnormal response to sensory stimuli and lack preference for novel odors. These results suggest that loss of Caspr2 throughout the sensory system may contribute to the sensory manifestations frequently observed in ASD.
Collapse
Affiliation(s)
- Aaron Gordon
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Daniela Salomon
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Noy Barak
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Yefim Pen
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Michael Tsoory
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Tali Kimchi
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Elior Peles
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel.
| |
Collapse
|
172
|
D'Gama AM, Pochareddy S, Li M, Jamuar SS, Reiff RE, Lam ATN, Sestan N, Walsh CA. Targeted DNA Sequencing from Autism Spectrum Disorder Brains Implicates Multiple Genetic Mechanisms. Neuron 2015; 88:910-917. [PMID: 26637798 PMCID: PMC4672379 DOI: 10.1016/j.neuron.2015.11.009] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 09/16/2015] [Accepted: 10/27/2015] [Indexed: 11/18/2022]
Abstract
Single nucleotide variants (SNVs), particularly loss-of-function mutations, are significant contributors to autism spectrum disorder (ASD) risk. Here we report the first systematic deep sequencing study of 55 postmortem ASD brains for SNVs in 78 known ASD candidate genes. Remarkably, even without parental samples, we find more ASD brains with mutations that are protein-altering (26/55 cases versus 12/50 controls, p = 0.015), deleterious (16/55 versus 5/50, p = 0.016), or loss-of-function (6/55 versus 0/50, p = 0.028) compared to controls, with recurrent deleterious mutations in ARID1B, SCN1A, SCN2A, and SETD2, suggesting these mutations contribute to ASD risk. In several cases, the identified mutations and medical records suggest syndromic ASD diagnoses. Two ASD and one Fragile X premutation case showed deleterious somatic mutations, providing evidence that somatic mutations occur in ASD cases, and supporting a model in which a combination of germline and/or somatic mutations may contribute to ASD risk on a case-by-case basis.
Collapse
Affiliation(s)
- Alissa M D'Gama
- Division of Genetics and Genomics, Manton Center for Orphan Disease, and Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA 02115, USA; Departments of Pediatrics and Neurology, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Sirisha Pochareddy
- Departments of Neuroscience, Genetics, and Psychiatry, and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Mingfeng Li
- Departments of Neuroscience, Genetics, and Psychiatry, and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Saumya S Jamuar
- Division of Genetics and Genomics, Manton Center for Orphan Disease, and Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA 02115, USA; Departments of Pediatrics and Neurology, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Paediatrics, KK Women's and Children's Hospital, Singapore; Paediatrics Academic Clinical Programme, Duke-NUS Graduate School of Medicine, Singapore
| | - Rachel E Reiff
- Division of Genetics and Genomics, Manton Center for Orphan Disease, and Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA 02115, USA; Departments of Pediatrics and Neurology, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Anh-Thu N Lam
- Division of Genetics and Genomics, Manton Center for Orphan Disease, and Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA 02115, USA; Departments of Pediatrics and Neurology, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Nenad Sestan
- Departments of Neuroscience, Genetics, and Psychiatry, and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Christopher A Walsh
- Division of Genetics and Genomics, Manton Center for Orphan Disease, and Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA 02115, USA; Departments of Pediatrics and Neurology, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| |
Collapse
|
173
|
Reiner O, Karzbrun E, Kshirsagar A, Kaibuchi K. Regulation of neuronal migration, an emerging topic in autism spectrum disorders. J Neurochem 2015; 136:440-56. [PMID: 26485324 DOI: 10.1111/jnc.13403] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 09/04/2015] [Accepted: 10/09/2015] [Indexed: 12/14/2022]
Abstract
Autism spectrum disorders (ASD) encompass a group of neurodevelopmental diseases that demonstrate strong heritability, however, the inheritance is not simple and many genes have been associated with these disorders. ASD is regarded as a neurodevelopmental disorder, and abnormalities at different developmental stages are part of the disease etiology. This review provides a general background on neuronal migration during brain development and discusses recent advancements in the field connecting ASD and aberrant neuronal migration. We propose that neuronal migration impairment may be an important common pathophysiology in autism spectrum disorders (ASD). This review provides a general background on neuronal migration during brain development and discusses recent advancements in the field connecting ASD and aberrant neuronal migration.
Collapse
Affiliation(s)
- Orly Reiner
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Eyal Karzbrun
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Aditya Kshirsagar
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Kozo Kaibuchi
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Showa, Nagoya, Japan
| |
Collapse
|
174
|
CNTNAP2 gene in high functioning autism: no association according to family and meta-analysis approaches. J Neural Transm (Vienna) 2015; 123:353-63. [PMID: 26559825 DOI: 10.1007/s00702-015-1458-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 09/07/2015] [Indexed: 10/22/2022]
Abstract
The Contactin Associated Protein-like 2 (CNTNAP2) gene has been discussed to be associated with different symptoms of autism spectrum disorders (ASDs) and other neurodevelopmental disorders. We aimed to elucidate the genetic association of CNTNAP2 within high functioning ASD (HFA), focusing on autism specific symptoms and reducing intelligence related factors. Furthermore, we compared our findings conducting a meta-analysis in patients with ASD and HFA only. A case-control association study was performed for HFA (HFA, n = 105; controls, n = 133). Moreover, we performed a family-based association study (DFAM) analysis (HFA, n = 44; siblings, n = 57). Individuals were genotyped for the two most frequently reported single nucleotide polymorphisms (SNPs) in the CNTNAP2 gene (rs2710102, rs7794745). Furthermore, a meta-analysis using the MIX2 software integrated our results with previously published data. A significant association for the carriers of the T-allele of the rs7794745 with HFA was found in the case-control sample [OR = 1.547; (95 % CI 1.056-2.266); p = 0.025]. No association could be found by DFAM with any of the CNTNAP2 SNPs with HFA. The meta-analysis of both SNPs did not show a significant association with either ASD or with HFA. Overall, including case-control, sibs, and meta-analysis, we could not detect any significant association with the CNTNAP2 gene and HFA. Our results point in the direction that CNTNAP2 may not play a major role in HFA, but rather seems to have a significance in neurodevelopmental disorders or in individuals displaying intellectual delays.
Collapse
|
175
|
Targeted Gene Resequencing (Astrochip) to Explore the Tripartite Synapse in Autism-Epilepsy Phenotype with Macrocephaly. Neuromolecular Med 2015; 18:69-80. [PMID: 26537360 DOI: 10.1007/s12017-015-8378-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 10/27/2015] [Indexed: 12/26/2022]
Abstract
The frequent co-occurrence of autism spectrum disorders (ASD) and epilepsy, or paroxysmal EEG abnormalities, defines a condition termed autism-epilepsy phenotype (AEP). This condition results, in some cases , from dysfunctions of glial inwardly rectifying potassium channels (Kir), which are mainly expressed in astrocytes where they mediate neuron-glia communication. Macrocephaly is also often comorbid with autism-epilepsy (autism-epilepsy phenotype with macrocephaly, MAEP), and it is tempting to hypothesize that shared pathogenic mechanisms might explain concurrence of these conditions. In the present study, we assessed whether protein pathways involved, along with Kir channels, in astrocyte-neuron interaction at the tripartite synapse play a role in the etiopathogenesis of MAEP. Using a targeted resequencing methodology, we investigated the coding regions of 35 genes in 61 patients and correlated genetic results with clinical features. Variants were subdivided into 12 classes and clustered into four groups. We detected rare or previously unknown predicted deleterious missense changes in GJA1, SLC12A2, SNTA1, EFNA3, CNTNAP2, EPHA4, and STXBP1 in seven patients and two high-frequency variants in DLG1 in six individuals. We also found that a group of variants (predicted deleterious and non-coding), segregating with the comorbid MAEP/AEP subgroups, belong to proteins specifically involved in glutamate transport and metabolism (namely, SLC17A6, GRM8, and GLUL), as well as in potassium conductance (KCNN3). This "endophenotype-oriented" study, performed using a targeted strategy, helped to further delineate part of the complex genetic background of ASD, particularly in the presence of coexisting macrocephaly and/or epilepsy/paroxysmal EEG, and suggests that use of stringent clinical clustering might be an approach worth adopting in order to unravel the complex genomic data in neurodevelopmental disorders.
Collapse
|
176
|
Affiliation(s)
- Victoria E Brings
- Department of Cell Biology and Physiology, UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Mark J Zylka
- Department of Cell Biology and Physiology, UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
177
|
Sullivan JM, Badimon A, Schaefer U, Ayata P, Gray J, Chung CW, von Schimmelmann M, Zhang F, Garton N, Smithers N, Lewis H, Tarakhovsky A, Prinjha RK, Schaefer A. Autism-like syndrome is induced by pharmacological suppression of BET proteins in young mice. J Exp Med 2015; 212:1771-81. [PMID: 26392221 PMCID: PMC4612093 DOI: 10.1084/jem.20151271] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 08/25/2015] [Indexed: 01/09/2023] Open
Abstract
Studies investigating the causes of autism spectrum disorder (ASD) point to genetic, as well as epigenetic, mechanisms of the disease. Identification of epigenetic processes that contribute to ASD development and progression is of major importance and may lead to the development of novel therapeutic strategies. Here, we identify the bromodomain and extraterminal domain-containing proteins (BETs) as epigenetic regulators of genes involved in ASD-like behaviors in mice. We found that the pharmacological suppression of BET proteins in the brain of young mice, by the novel, highly specific, brain-permeable inhibitor I-BET858 leads to selective suppression of neuronal gene expression followed by the development of an autism-like syndrome. Many of the I-BET858-affected genes have been linked to ASD in humans, thus suggesting the key role of the BET-controlled gene network in the disorder. Our studies suggest that environmental factors controlling BET proteins or their target genes may contribute to the epigenetic mechanism of ASD.
Collapse
Affiliation(s)
- Josefa M Sullivan
- Department of Neuroscience and Department of Psychiatry, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 Department of Neuroscience and Department of Psychiatry, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Ana Badimon
- Department of Neuroscience and Department of Psychiatry, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 Department of Neuroscience and Department of Psychiatry, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Uwe Schaefer
- Laboratory of Immune Cell Epigenetics and Signaling, The Rockefeller University, New York, NY 10065
| | - Pinar Ayata
- Department of Neuroscience and Department of Psychiatry, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 Department of Neuroscience and Department of Psychiatry, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - James Gray
- Epinova DPU, Quantitative Pharmacology, and Platform Technology and Science, Immuno-Inflammation Therapy Area, GlaxoSmithKline, Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, England, UK Epinova DPU, Quantitative Pharmacology, and Platform Technology and Science, Immuno-Inflammation Therapy Area, GlaxoSmithKline, Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, England, UK
| | - Chun-wa Chung
- Epinova DPU, Quantitative Pharmacology, and Platform Technology and Science, Immuno-Inflammation Therapy Area, GlaxoSmithKline, Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, England, UK
| | - Melanie von Schimmelmann
- Department of Neuroscience and Department of Psychiatry, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 Department of Neuroscience and Department of Psychiatry, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Fan Zhang
- Department of Neuroscience and Department of Psychiatry, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 Department of Neuroscience and Department of Psychiatry, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Neil Garton
- Epinova DPU, Quantitative Pharmacology, and Platform Technology and Science, Immuno-Inflammation Therapy Area, GlaxoSmithKline, Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, England, UK
| | - Nicholas Smithers
- Epinova DPU, Quantitative Pharmacology, and Platform Technology and Science, Immuno-Inflammation Therapy Area, GlaxoSmithKline, Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, England, UK
| | - Huw Lewis
- Epinova DPU, Quantitative Pharmacology, and Platform Technology and Science, Immuno-Inflammation Therapy Area, GlaxoSmithKline, Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, England, UK
| | - Alexander Tarakhovsky
- Laboratory of Immune Cell Epigenetics and Signaling, The Rockefeller University, New York, NY 10065
| | - Rab K Prinjha
- Epinova DPU, Quantitative Pharmacology, and Platform Technology and Science, Immuno-Inflammation Therapy Area, GlaxoSmithKline, Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, England, UK
| | - Anne Schaefer
- Department of Neuroscience and Department of Psychiatry, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 Department of Neuroscience and Department of Psychiatry, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| |
Collapse
|
178
|
Hammer M, Krueger-Burg D, Tuffy LP, Cooper BH, Taschenberger H, Goswami SP, Ehrenreich H, Jonas P, Varoqueaux F, Rhee JS, Brose N. Perturbed Hippocampal Synaptic Inhibition and γ-Oscillations in a Neuroligin-4 Knockout Mouse Model of Autism. Cell Rep 2015; 13:516-523. [PMID: 26456829 PMCID: PMC5862414 DOI: 10.1016/j.celrep.2015.09.011] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 08/23/2015] [Accepted: 09/02/2015] [Indexed: 12/02/2022] Open
Abstract
Loss-of-function mutations in the synaptic adhesion protein Neuroligin-4 are among the most common genetic abnormalities associated with autism spectrum disorders, but little is known about the function of Neuroligin-4 and the consequences of its loss. We assessed synaptic and network characteristics in Neuroligin-4 knockout mice, focusing on the hippocampus as a model brain region with a critical role in cognition and memory, and found that Neuroligin-4 deletion causes subtle defects of the protein composition and function of GABAergic synapses in the hippocampal CA3 region. Interestingly, these subtle synaptic changes are accompanied by pronounced perturbations of γ-oscillatory network activity, which has been implicated in cognitive function and is altered in multiple psychiatric and neurodevelopmental disorders. Our data provide important insights into the mechanisms by which Neuroligin-4-dependent GABAergic synapses may contribute to autism phenotypes and indicate new strategies for therapeutic approaches.
Collapse
Affiliation(s)
- Matthieu Hammer
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Hermann-Rein-Straße 3, 37075 Göttingen, Germany
| | - Dilja Krueger-Burg
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Hermann-Rein-Straße 3, 37075 Göttingen, Germany
| | - Liam Patrick Tuffy
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Hermann-Rein-Straße 3, 37075 Göttingen, Germany
| | - Benjamin Hillman Cooper
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Hermann-Rein-Straße 3, 37075 Göttingen, Germany
| | - Holger Taschenberger
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Hermann-Rein-Straße 3, 37075 Göttingen, Germany
| | - Sarit Pati Goswami
- Institute of Science and Technology Austria, Am Campus 1, 3400 Klosterneuburg, Austria
| | - Hannelore Ehrenreich
- Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Hermann-Rein-Straße 3, 37075 Göttingen, Germany
| | - Peter Jonas
- Institute of Science and Technology Austria, Am Campus 1, 3400 Klosterneuburg, Austria
| | - Frederique Varoqueaux
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Hermann-Rein-Straße 3, 37075 Göttingen, Germany
| | - Jeong-Seop Rhee
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Hermann-Rein-Straße 3, 37075 Göttingen, Germany.
| | - Nils Brose
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Hermann-Rein-Straße 3, 37075 Göttingen, Germany.
| |
Collapse
|
179
|
Abstract
Large-scale genomic investigations have just begun to illuminate the molecular genetic contributions to major psychiatric illnesses, ranging from small-effect-size common variants to larger-effect-size rare mutations. The findings provide causal anchors from which to understand their neurobiological basis. Although these studies represent enormous success, they highlight major challenges reflected in the heterogeneity and polygenicity of all of these conditions and the difficulty of connecting multiple levels of molecular, cellular, and circuit functions to complex human behavior. Nevertheless, these advances place us on the threshold of a new frontier in the pathophysiological understanding, diagnosis, and treatment of psychiatric disease.
Collapse
Affiliation(s)
- Daniel H Geschwind
- Departments of Neurology, Psychiatry, and Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Jonathan Flint
- Wellcome Trust Center for Human Genetics, University of Oxford, Oxford, UK.
| |
Collapse
|
180
|
Yi JJ, Berrios J, Newbern JM, Snider WD, Philpot BD, Hahn KM, Zylka MJ. An Autism-Linked Mutation Disables Phosphorylation Control of UBE3A. Cell 2015; 162:795-807. [PMID: 26255772 DOI: 10.1016/j.cell.2015.06.045] [Citation(s) in RCA: 122] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Revised: 03/06/2015] [Accepted: 06/18/2015] [Indexed: 12/31/2022]
Abstract
Deletion of UBE3A causes the neurodevelopmental disorder Angelman syndrome (AS), while duplication or triplication of UBE3A is linked to autism. These genetic findings suggest that the ubiquitin ligase activity of UBE3A must be tightly maintained to promote normal brain development. Here, we found that protein kinase A (PKA) phosphorylates UBE3A in a region outside of the catalytic domain at residue T485 and inhibits UBE3A activity toward itself and other substrates. A de novo autism-linked missense mutation disrupts this phosphorylation site, causing enhanced UBE3A activity in vitro, enhanced substrate turnover in patient-derived cells, and excessive dendritic spine development in the brain. Our study identifies PKA as an upstream regulator of UBE3A activity and shows that an autism-linked mutation disrupts this phosphorylation control. Moreover, our findings implicate excessive UBE3A activity and the resulting synaptic dysfunction to autism pathogenesis.
Collapse
Affiliation(s)
- Jason J Yi
- Department of Cell Biology and Physiology and UNC Neuroscience Center, The University of North Carolina, Chapel Hill, NC 27599, USA; Department of Pharmacology, The University of North Carolina, Chapel Hill, NC 27599, USA; Carolina Institute for Developmental Disabilities, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Janet Berrios
- Department of Cell Biology and Physiology and UNC Neuroscience Center, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Jason M Newbern
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - William D Snider
- Department of Cell Biology and Physiology and UNC Neuroscience Center, The University of North Carolina, Chapel Hill, NC 27599, USA; Carolina Institute for Developmental Disabilities, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Benjamin D Philpot
- Department of Cell Biology and Physiology and UNC Neuroscience Center, The University of North Carolina, Chapel Hill, NC 27599, USA; Carolina Institute for Developmental Disabilities, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Klaus M Hahn
- Department of Pharmacology, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Mark J Zylka
- Department of Cell Biology and Physiology and UNC Neuroscience Center, The University of North Carolina, Chapel Hill, NC 27599, USA; Carolina Institute for Developmental Disabilities, The University of North Carolina, Chapel Hill, NC 27599, USA.
| |
Collapse
|
181
|
Abstract
A recent study by Gabel et al. (2015) found that Mecp2, the gene mutated in Rett syndrome, represses long (> 100 kb) genes associated with neuronal physiology and connectivity by binding to methylated CA sites in DNA. This study adds to a growing body of literature implicating gene length and transcriptional mechanisms in neurodevelopmental and neurodegenerative disorders.
Collapse
|
182
|
Lin HY, Ni HC, Lai MC, Tseng WYI, Gau SSF. Regional brain volume differences between males with and without autism spectrum disorder are highly age-dependent. Mol Autism 2015; 6:29. [PMID: 26045942 PMCID: PMC4455336 DOI: 10.1186/s13229-015-0022-3] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 04/21/2015] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Neuroanatomical differences between individuals with and without autism spectrum disorder (ASD) were inconsistent in the literature. Such heterogeneity may substantially originate from age-differential effects. METHODS Voxel-based morphometry was applied in 86 males with ASD and 90 typically developing control (TDC) males (aged 7 to 29 years). Three steps of statistical modeling (model 1, multiple regression with age as a covariate; model 2, multiple regression further considering diagnosis-by-age interaction; model 3, age-stratified analyses) were performed to dissect the moderating effects of age on diagnostic group differences in neuroanatomy. RESULTS Across ages, males with and without ASD did not differ significantly in total gray matter (GM) or white matter (WM) volumes. For both groups, total GM volumes decreased and WM volumes increased with age. For regional volume, comparing with the model only held the age constant (model 1), the main effect of group altered when diagnosis-by-age interaction effects were considered (model 2). Here, participants with ASD had significantly greater relative regional GM volumes than TDC in the right inferior orbitofrontal cortex and bilateral thalamus; for WM, participants with ASD were larger than TDC in the bilateral splenium of corpus callosum and right anterior corona radiata. Importantly, significant diagnosis-by-age interactions were identified at the bilateral anterior prefrontal cortex, bilateral cuneus, bilateral caudate, and the left cerebellum Crus I for GM and left forceps minor for WM. Finally, age-stratified analyses (model 3) showed distinct patterns in GM and WM volumetric alterations in ASD among subsamples of children, adolescents, and adults. CONCLUSIONS Our findings suggest that the heterogeneous reports on the atypical neuroanatomy of ASD may substantially originate from age variation in the study samples. Age variation and its methodological and biological implications have to be carefully delineated in future studies of the neurobiology of ASD.
Collapse
Affiliation(s)
- Hsiang-Yuan Lin
- Department of Psychiatry, National Taiwan University Hospital and College of Medicine, No. 7 Chung-Shan South Road, Taipei, 10002 Taiwan
| | - Hsing-Chang Ni
- Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, No. 1, Jen-Ai Road Section 1, Taipei, 10051 Taiwan.,Department of Child Psychiatry, Chang Gung Memorial Hospital at Linkou, No. 5 Fu-Hsing St., Taoyuan, 33305 Taiwan
| | - Meng-Chuan Lai
- Department of Psychiatry, National Taiwan University Hospital and College of Medicine, No. 7 Chung-Shan South Road, Taipei, 10002 Taiwan.,Autism Research Centre, Department of Psychiatry, University of Cambridge, Douglas House, 18b Trumpington Road, Cambridge, CB2 8AH UK.,Centre for Addiction and Mental Health, Hospital for Sick Children, and Department of Psychiatry, University of Toronto, 250 College St., Toronto, M5T 1R8 Canada
| | - Wen-Yih Isaac Tseng
- Graduate Institute of Brain and Mind Sciences, National Taiwan University College of Medicine, No. 1, Jen-Ai Road Section 1, Taipei, 10051 Taiwan.,Center for Optoelectronic Medicine, National Taiwan University College of Medicine, No. 1, Jen-Ai Road Section 1, Taipei, 10051 Taiwan
| | - Susan Shur-Fen Gau
- Department of Psychiatry, National Taiwan University Hospital and College of Medicine, No. 7 Chung-Shan South Road, Taipei, 10002 Taiwan.,Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, No. 1, Jen-Ai Road Section 1, Taipei, 10051 Taiwan.,Graduate Institute of Brain and Mind Sciences, National Taiwan University College of Medicine, No. 1, Jen-Ai Road Section 1, Taipei, 10051 Taiwan
| |
Collapse
|
183
|
Gene length matters in neurons. Neuron 2015. [PMID: 25905808 DOI: 10.1016/j.neuron.2015.03.059.] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
A recent study by Gabel et al. (2015) found that Mecp2, the gene mutated in Rett syndrome, represses long (> 100 kb) genes associated with neuronal physiology and connectivity by binding to methylated CA sites in DNA. This study adds to a growing body of literature implicating gene length and transcriptional mechanisms in neurodevelopmental and neurodegenerative disorders.
Collapse
|
184
|
Desarkar P, Rajji TK, Ameis SH, Daskalakis ZJ. Assessing and Stabilizing Aberrant Neuroplasticity in Autism Spectrum Disorder: The Potential Role of Transcranial Magnetic Stimulation. Front Psychiatry 2015; 6:124. [PMID: 26441685 PMCID: PMC4563147 DOI: 10.3389/fpsyt.2015.00124] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 08/25/2015] [Indexed: 11/13/2022] Open
Abstract
Exciting developments have taken place in the neuroscience research in autism spectrum disorder (ASD), and results from these studies indicate that brain in ASD is associated with aberrant neuroplasticity. Transcranial magnetic stimulation (TMS) has rapidly evolved to become a widely used, safe, and non-invasive neuroscientific tool to investigate a variety of neurophysiological processes, including neuroplasticity. The diagnostic and therapeutic potential of TMS in ASD is beginning to be realized. In this article, we briefly reviewed evidence of aberrant neuroplasticity in ASD, suggested future directions in assessing neuroplasticity using repetitive TMS (rTMS), and discussed the potential of rTMS in rectifying aberrant neuroplasticity in ASD.
Collapse
Affiliation(s)
- Pushpal Desarkar
- Department of Psychiatry, Centre for Addiction and Mental Health, University of Toronto , Toronto, ON , Canada ; Temerty Centre for Therapeutic Brain Intervention, Centre for Addiction and Mental Health , Toronto, ON , Canada
| | - Tarek K Rajji
- Department of Psychiatry, Centre for Addiction and Mental Health, University of Toronto , Toronto, ON , Canada ; Temerty Centre for Therapeutic Brain Intervention, Centre for Addiction and Mental Health , Toronto, ON , Canada
| | - Stephanie H Ameis
- Department of Psychiatry, Centre for Addiction and Mental Health, University of Toronto , Toronto, ON , Canada ; Temerty Centre for Therapeutic Brain Intervention, Centre for Addiction and Mental Health , Toronto, ON , Canada ; Department of Psychiatry, The Hospital for Sick Children, University of Toronto , Toronto, ON , Canada ; Research Imaging Centre, Campbell Family Mental Health Research Institute, The Centre for Addiction and Mental Health (CAMH) , Toronto, ON , Canada
| | - Zafiris Jeff Daskalakis
- Department of Psychiatry, Centre for Addiction and Mental Health, University of Toronto , Toronto, ON , Canada ; Temerty Centre for Therapeutic Brain Intervention, Centre for Addiction and Mental Health , Toronto, ON , Canada
| |
Collapse
|