151
|
Bateup HS, Denefrio CL, Johnson CA, Saulnier JL, Sabatini BL. Temporal dynamics of a homeostatic pathway controlling neural network activity. Front Mol Neurosci 2013; 6:28. [PMID: 24065881 PMCID: PMC3776619 DOI: 10.3389/fnmol.2013.00028] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Accepted: 08/24/2013] [Indexed: 11/13/2022] Open
Abstract
Neurons use a variety of mechanisms to homeostatically regulate neural network activity in order to maintain firing in a bounded range. One such process involves the bi-directional modulation of excitatory synaptic drive in response to chronic changes in network activity. Down-scaling of excitatory synapses in response to high activity requires Arc-dependent endocytosis of glutamate receptors. However, the temporal dynamics and signaling pathways regulating Arc during homeostatic plasticity are not well understood. Here we determine the relative contribution of transcriptional and translational control in the regulation of Arc, the signaling pathways responsible for the activity-dependent production of Arc, and the time course of these signaling events as they relate to the homeostatic adjustment of network activity in hippocampal neurons. We find that an ERK1/2-dependent transcriptional pathway active within 1-2 h of up-regulated network activity induces Arc leading to a restoration of network spiking rates within 12 h. Under basal and low activity conditions, specialized mechanisms are in place to rapidly degrade Arc mRNA and protein such that they have half-lives of less than 1 h. In addition, we find that while mTOR signaling is regulated by network activity on a similar time scale, mTOR-dependent translational control is not a major regulator of Arc production or degradation suggesting that the signaling pathways underlying homeostatic plasticity are distinct from those mediating synapse-specific forms of synaptic depression.
Collapse
Affiliation(s)
- Helen S Bateup
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School Boston, MA, USA
| | | | | | | | | |
Collapse
|
152
|
Eran A, Li JB, Vatalaro K, McCarthy J, Rahimov F, Collins C, Markianos K, Margulies DM, Brown EN, Calvo SE, Kohane IS, Kunkel LM. Comparative RNA editing in autistic and neurotypical cerebella. Mol Psychiatry 2013; 18:1041-8. [PMID: 22869036 PMCID: PMC3494744 DOI: 10.1038/mp.2012.118] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2012] [Revised: 06/29/2012] [Accepted: 06/29/2012] [Indexed: 01/03/2023]
Abstract
Adenosine-to-inosine (A-to-I) RNA editing is a neurodevelopmentally regulated epigenetic modification shown to modulate complex behavior in animals. Little is known about human A-to-I editing, but it is thought to constitute one of many molecular mechanisms connecting environmental stimuli and behavioral outputs. Thus, comprehensive exploration of A-to-I RNA editing in human brains may shed light on gene-environment interactions underlying complex behavior in health and disease. Synaptic function is a main target of A-to-I editing, which can selectively recode key amino acids in synaptic genes, directly altering synaptic strength and duration in response to environmental signals. Here, we performed a high-resolution survey of synaptic A-to-I RNA editing in a human population, and examined how it varies in autism, a neurodevelopmental disorder in which synaptic abnormalities are a common finding. Using ultra-deep (>1000 × ) sequencing, we quantified the levels of A-to-I editing of 10 synaptic genes in postmortem cerebella from 14 neurotypical and 11 autistic individuals. A high dynamic range of editing levels was detected across individuals and editing sites, from 99.6% to below detection limits. In most sites, the extreme ends of the population editing distributions were individuals with autism. Editing was correlated with isoform usage, clusters of correlated sites were identified, and differential editing patterns examined. Finally, a dysfunctional form of the editing enzyme adenosine deaminase acting on RNA B1 was found more commonly in postmortem cerebella from individuals with autism. These results provide a population-level, high-resolution view of A-to-I RNA editing in human cerebella and suggest that A-to-I editing of synaptic genes may be informative for assessing the epigenetic risk for autism.
Collapse
Affiliation(s)
- Alal Eran
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA 02139, USA,Program in Genomics, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Jin Billy Li
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Kayla Vatalaro
- Program in Genomics, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Jillian McCarthy
- Program in Genomics, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Fedik Rahimov
- Program in Genomics, Boston Children’s Hospital, Boston, MA 02115, USA,Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Christin Collins
- Program in Genomics, Boston Children’s Hospital, Boston, MA 02115, USA,Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Kyriacos Markianos
- Program in Genomics, Boston Children’s Hospital, Boston, MA 02115, USA,Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - David M. Margulies
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA,Correlagen Diagnostics, Waltham, MA 02452, USA,Center for Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
| | - Emery N. Brown
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA 02139, USA,Neuroscience Statistics Research Laboratory, Department of Anesthesia and Critical Care, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA,Department of Brain and Cognitive Sciences, MIT, Cambridge, MA 02139, USA
| | - Sarah E. Calvo
- Broad Institute of Harvard and MIT, Cambridge, MA 02139, USA
| | - Isaac S. Kohane
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA 02139, USA,Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA,Center for Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA,Correspondence: Louis Kunkel, Program in Genomics, Department of Genetics, Boston Children’s Hospital, 3 Blackfan Circle, CLS 15027.1, Boston, MA 02115, USA. Telephone: (617) 355-6279, fax: (617) 730-0253, , Isaac Kohane, Department of Pediatrics, Boston Children's Hospital, 300 Longwood Ave., Enders 144, Boston, MA 02115, USA. Telephone: (617) 919-2182, fax: (617) 730-0921,
| | - Louis M. Kunkel
- Program in Genomics, Boston Children’s Hospital, Boston, MA 02115, USA,Department of Genetics, Harvard Medical School, Boston, MA 02115, USA,Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA,The Manton Center for Orphan Disease Research, Boston, MA 02115, USA,Correspondence: Louis Kunkel, Program in Genomics, Department of Genetics, Boston Children’s Hospital, 3 Blackfan Circle, CLS 15027.1, Boston, MA 02115, USA. Telephone: (617) 355-6279, fax: (617) 730-0253, , Isaac Kohane, Department of Pediatrics, Boston Children's Hospital, 300 Longwood Ave., Enders 144, Boston, MA 02115, USA. Telephone: (617) 919-2182, fax: (617) 730-0921,
| |
Collapse
|
153
|
Ey E, Torquet N, Le Sourd AM, Leblond CS, Boeckers TM, Faure P, Bourgeron T. The Autism ProSAP1/Shank2 mouse model displays quantitative and structural abnormalities in ultrasonic vocalisations. Behav Brain Res 2013; 256:677-89. [PMID: 23994547 DOI: 10.1016/j.bbr.2013.08.031] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2013] [Revised: 07/23/2013] [Accepted: 08/19/2013] [Indexed: 02/07/2023]
Abstract
Mouse ultrasonic vocalisations have been often used as a paradigm to extrapolate vocal communication defects observed in patients with autism spectrum disorders (ASD). The role of these vocalisations as well as their development, structure and informational content, however, remain largely unknown. In the present study, we characterised in depth the emission of pup and adult ultrasonic vocalisations of wild-type mice and their ProSAP1/Shank2(-/-) littermates lacking a synaptic scaffold protein mutated in ASD. We hypothesised that the vocal behaviour of ProSAP1/Shank2(-/-) mice not only differs from the vocal behaviour of their wild-type littermates in a quantitative way, but also presents more qualitative abnormalities in temporal organisation and acoustic structure. We first quantified the rate of emission of ultrasonic vocalisations, and analysed the organisation of vocalisations sequences using Markov models. We subsequently measured duration and peak frequency characteristics of each ultrasonic vocalisation, to characterise their acoustic structure. In wild-type mice, we found a high level of organisation in sequences of ultrasonic vocalisations, suggesting a communicative function in this complex system. Very limited significant sex-related variations were detected in their usage and acoustic structure, even in adult mice. In adult ProSAP1/Shank2(-/-) mice, we found abnormalities in the call usage and the structure of ultrasonic vocalisations. Both ProSAP1/Shank2(-/-) male and female mice uttered less vocalisations with a different call distribution and at lower peak frequency in comparison with wild-type littermates. This study provides a comprehensive framework to characterise abnormalities of ultrasonic vocalisations in mice and confirms that ProSAP1/Shank2(-/-) mice represent a relevant model to study communication defects.
Collapse
Affiliation(s)
- Elodie Ey
- Human Genetics and Cognitive Functions, Institut Pasteur, Paris, France; CNRS URA 2182 'Genes, synapses and cognition', Institut Pasteur, Paris, France; University Paris Diderot, Sorbonne Paris Cité, Human Genetics and Cognitive Functions, Paris, France.
| | | | | | | | | | | | | |
Collapse
|
154
|
Lv YT, Zhang Y, Liu M, Qiuwaxi JNT, Ashwood P, Cho SC, Huan Y, Ge RC, Chen XW, Wang ZJ, Kim BJ, Hu X. Transplantation of human cord blood mononuclear cells and umbilical cord-derived mesenchymal stem cells in autism. J Transl Med 2013; 11:196. [PMID: 23978163 PMCID: PMC3765833 DOI: 10.1186/1479-5876-11-196] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Accepted: 08/23/2013] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Autism is a pervasive neurodevelopmental disorder. At present there are no defined mechanisms of pathogenesis and therapy is mostly limited to behavioral interventions. Stem cell transplantation may offer a unique treatment strategy for autism due to immune and neural dysregulation observed in this disease. This non-randomized, open-label, single center phase I/II trial investigated the safety and efficacy of combined transplantation of human cord blood mononuclear cells (CBMNCs) and umbilical cord-derived mesenchymal stem cells (UCMSCs) in treating children with autism. METHODS 37 subjects diagnosed with autism were enrolled into this study and divided into three groups: CBMNC group (14 subjects, received CBMNC transplantation and rehabilitation therapy), Combination group (9 subjects, received both CBMNC and UCMSC transplantation and rehabilitation therapy), and Control group (14 subjects, received only rehabilitation therapy). Transplantations included four stem cell infusions through intravenous and intrathecal injections once a week. Treatment safety was evaluated with laboratory examinations and clinical assessment of adverse effects. The Childhood Autism Rating Scale (CARS), Clinical Global Impression (CGI) scale and Aberrant Behavior Checklist (ABC) were adopted to assess the therapeutic efficacy at baseline (pre-treatment) and following treatment. RESULTS There were no significant safety issues related to the treatment and no observed severe adverse effects. Statistically significant differences were shown on CARS, ABC scores and CGI evaluation in the two treatment groups compared to the control at 24 weeks post-treatment (p < 0.05). CONCLUSIONS Transplantation of CBMNCs demonstrated efficacy compared to the control group; however, the combination of CBMNCs and UCMSCs showed larger therapeutic effects than the CBMNC transplantation alone. There were no safety issues noted during infusion and the whole monitoring period. TRIAL REGISTRATION ClinicalTrials.gov: NCT01343511, Title "Safety and Efficacy of Stem Cell Therapy in Patients with Autism".
Collapse
Affiliation(s)
- Yong-Tao Lv
- Shandong Jiaotong Hospital, Jinan, Shandong, China
| | - Yun Zhang
- Shenzhen Beike Cell Engineering Research Institute, 2/F, Yuanxing Technology Building, #1 Songpingshan Street, Nanshan District, Shenzhen, Guangdong 518057, China
| | - Min Liu
- Shandong Jiaotong Hospital, Jinan, Shandong, China
| | - Jia-na-ti Qiuwaxi
- Shenzhen Beike Cell Engineering Research Institute, 2/F, Yuanxing Technology Building, #1 Songpingshan Street, Nanshan District, Shenzhen, Guangdong 518057, China
| | - Paul Ashwood
- Department of Medical Microbiology & Immunology, University of California Davis, Davis, CA, USA
| | - Sungho Charles Cho
- Department of Neurology and Neurosurgery, Stanford University, Stanford, CA, USA
| | - Ying Huan
- Shandong Jiaotong Hospital, Jinan, Shandong, China
| | - Ru-Cun Ge
- Shandong Jiaotong Hospital, Jinan, Shandong, China
| | | | - Zhao-Jing Wang
- Shenzhen Beike Cell Engineering Research Institute, 2/F, Yuanxing Technology Building, #1 Songpingshan Street, Nanshan District, Shenzhen, Guangdong 518057, China
| | - Byung-Jo Kim
- Department of Neurology, Korea University Anam Medical Center, Seoul, Korea
| | - Xiang Hu
- Shenzhen Beike Cell Engineering Research Institute, 2/F, Yuanxing Technology Building, #1 Songpingshan Street, Nanshan District, Shenzhen, Guangdong 518057, China
| |
Collapse
|
155
|
Benchoua A, Peschanski M. Pluripotent stem cells as a model to study non-coding RNAs function in human neurogenesis. Front Cell Neurosci 2013; 7:140. [PMID: 23986659 PMCID: PMC3753451 DOI: 10.3389/fncel.2013.00140] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Accepted: 08/12/2013] [Indexed: 12/22/2022] Open
Abstract
As fine regulators of gene expression, non-coding RNAs, and more particularly micro-RNAs (miRNAs), have emerged as key players in the development of the nervous system. In vivo experiments manipulating miRNAs expression as neurogenesis proceeds are very challenging in the mammalian embryo and totally impossible in the human. Human pluripotent stem cells (hPSCs), from embryonic origin (hESCs) or induced from adult somatic cells (iPSCs), represent an opportunity to study the role of miRNAs in the earliest steps of human neurogenesis in both physiological and pathological contexts. Robust protocols are now available to convert pluripotent stem cells into several sub-types of fully functional neurons, recapitulating key developmental milestones along differentiation. This provides a convenient cellular system for dissecting the role of miRNAs in phenotypic transitions critical to brain development and plasticity that may be impaired in neurological diseases with onset during development. The aim of this review is to illustrate how hPSCs can be used to recapitulate early steps of human neurogenesis and summarize recent reports of their contribution to the study of the role of miRNA in regulating development of the nervous system.
Collapse
Affiliation(s)
- Alexandra Benchoua
- Centre d'Etude des Cellules Souches, Institut des cellules Souches pour le Traitement et l'Étude des Maladies monogéniques, Association Française contre les Myopathies Evry, France
| | | |
Collapse
|
156
|
Nair A, Treiber JM, Shukla DK, Shih P, Müller RA. Impaired thalamocortical connectivity in autism spectrum disorder: a study of functional and anatomical connectivity. ACTA ACUST UNITED AC 2013; 136:1942-55. [PMID: 23739917 DOI: 10.1093/brain/awt079] [Citation(s) in RCA: 258] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The thalamus plays crucial roles in the development and mature functioning of numerous sensorimotor, cognitive and attentional circuits. Currently limited evidence suggests that autism spectrum disorder may be associated with thalamic abnormalities, potentially related to sociocommunicative and other impairments in this disorder. We used functional connectivity magnetic resonance imaging and diffusion tensor imaging probabilistic tractography to study the functional and anatomical integrity of thalamo-cortical connectivity in children and adolescents with autism spectrum disorder and matched typically developing children. For connectivity with five cortical seeds (prefontal, parieto-occipital, motor, somatosensory and temporal), we found evidence of both anatomical and functional underconnectivity. The only exception was functional connectivity with the temporal lobe, which was increased in the autism spectrum disorders group, especially in the right hemisphere. However, this effect was robust only in partial correlation analyses (partialling out time series from other cortical seeds), whereas findings from total correlation analyses suggest that temporo-thalamic overconnectivity in the autism group was only relative to the underconnectivity found for other cortical seeds. We also found evidence of microstructural compromise within the thalamic motor parcel, associated with compromise in tracts between thalamus and motor cortex, suggesting that the thalamus may play a role in motor abnormalities reported in previous autism studies. More generally, a number of correlations of diffusion tensor imaging and functional connectivity magnetic resonance imaging measures with diagnostic and neuropsychological scores indicate involvement of abnormal thalamocortical connectivity in sociocommunicative and cognitive impairments in autism spectrum disorder.
Collapse
Affiliation(s)
- Aarti Nair
- Brain Development Imaging Laboratory, Department of Psychology, San Diego State University, San Diego, CA 92120, USA
| | | | | | | | | |
Collapse
|
157
|
Cook D, Nuro E, Murai KK. Increasing our understanding of human cognition through the study of Fragile X Syndrome. Dev Neurobiol 2013; 74:147-77. [PMID: 23723176 PMCID: PMC4216185 DOI: 10.1002/dneu.22096] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Accepted: 05/17/2013] [Indexed: 12/16/2022]
Abstract
Fragile X Syndrome (FXS) is considered the most common form of inherited intellectual disability. It is caused by reductions in the expression level or function of a single protein, the Fragile X Mental Retardation Protein (FMRP), a translational regulator which binds to approximately 4% of brain messenger RNAs. Accumulating evidence suggests that FXS is a complex disorder of cognition, involving interactions between genetic and environmental influences, leading to difficulties in acquiring key life skills including motor skills, language, and proper social behaviors. Since many FXS patients also present with one or more features of autism spectrum disorders (ASDs), insights gained from studying the monogenic basis of FXS could pave the way to a greater understanding of underlying features of multigenic ASDs. Here we present an overview of the FXS and FMRP field with the goal of demonstrating how loss of a single protein involved in translational control affects multiple stages of brain development and leads to debilitating consequences on human cognition. We also focus on studies which have rescued or improved FXS symptoms in mice using genetic or therapeutic approaches to reduce protein expression. We end with a brief description of how deficits in translational control are implicated in FXS and certain cases of ASDs, with many recent studies demonstrating that ASDs are likely caused by increases or decreases in the levels of certain key synaptic proteins. The study of FXS and its underlying single genetic cause offers an invaluable opportunity to study how a single gene influences brain development and behavior.
Collapse
Affiliation(s)
- Denise Cook
- Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, The Research Institute of the McGill University Health Centre, Montreal General Hospital, Montreal, Quebec, Canada
| | | | | |
Collapse
|
158
|
Abstract
The autism spectrum disorders (ASD) are characterized by impairments in social interaction and stereotyped behaviors. For the majority of individuals with ASD, the causes of the disorder remain unknown; however, in up to 25% of cases, a genetic cause can be identified. Chromosomal rearrangements as well as rare and de novo copy-number variants are present in ∼10-20% of individuals with ASD, compared with 1-2% in the general population and/or unaffected siblings. Rare and de novo coding-sequence mutations affecting neuronal genes have also been identified in ∼5-10% of individuals with ASD. Common variants such as single-nucleotide polymorphisms seem to contribute to ASD susceptibility, but, taken individually, their effects appear to be small. Despite a heterogeneous genetic landscape, the genes implicated thus far-which are involved in chromatin remodeling, metabolism, mRNA translation, and synaptic function-seem to converge in common pathways affecting neuronal and synaptic homeostasis. Animal models developed to study these genes should lead to a better understanding of the diversity of the genetic landscapes of ASD.
Collapse
Affiliation(s)
- Guillaume Huguet
- Human Genetics and Cognitive Functions Unit, Institut Pasteur, 75015 Paris, France;
| | | | | |
Collapse
|
159
|
Siniscalco D, Bradstreet JJ, Antonucci N. Therapeutic role of hematopoietic stem cells in autism spectrum disorder-related inflammation. Front Immunol 2013; 4:140. [PMID: 23772227 PMCID: PMC3677147 DOI: 10.3389/fimmu.2013.00140] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Accepted: 05/26/2013] [Indexed: 12/24/2022] Open
Abstract
Autism and autism spectrum disorders (ASDs) are heterogeneous, severe neuro-developmental disorders with core symptoms of dysfunctions in social interactions and communication skills, restricted interests, repetitive – stereotypic verbal and non-verbal behaviors. Biomolecular evidence points to complex gene-environmental interactions in ASDs. Several biochemical processes are associated with ASDs: oxidative stress (including endoplasmic reticulum stress), decreased methylation capacity, limited production of glutathione; mitochondrial dysfunction, intestinal dysbiosis, increased toxic metal burden, and various immune abnormalities. The known immunological disorders include: T-lymphocyte populations and function, gene expression changes in monocytes, several autoimmune-related findings, high levels of N-acetylgalactosaminidase (which precludes macrophage activation), and primary immune deficiencies. These immunological observations may result in minicolumn structural changes in the brain, as well as, abnormal immune mediation of synaptic functions. Equally, these immune dysregulations serve as the rationale for immune-directed interventions such as hematopoietic stem cells (HSCs), which are pivotal in controlling chronic inflammation and in the restoration of immunological balance. These properties make them intriguing potential agents for ASD treatments. This prospective review will focus on the current state-of-the-art knowledge and challenges intrinsic in the application of HSCs for ASD-related immunological disorders.
Collapse
Affiliation(s)
- Dario Siniscalco
- Department of Experimental Medicine, Second University of Naples , Naples , Italy ; Centre for Autism - La Forza del Silenzio , Caserta , Italy ; Cancellautismo , Florence , Italy
| | | | | |
Collapse
|
160
|
Progress toward treatments for synaptic defects in autism. Nat Med 2013; 19:685-94. [PMID: 23744158 DOI: 10.1038/nm.3193] [Citation(s) in RCA: 134] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Accepted: 04/11/2013] [Indexed: 12/11/2022]
Abstract
Autism spectrum disorder (ASD) encompasses a range of disorders that are characterized by social and communication deficits and repetitive behaviors. For the majority of affected individuals, the cause of ASD remains unknown, but in at least 20% of the cases, a genetic cause can be identified. There is currently no cure for ASD; however, results from mouse models indicate that some forms of the disorder could be alleviated even at the adult stage. Genes involved in ASD seem to converge on common pathways altering synaptic homeostasis. We propose, given the clinical heterogeneity of ASD, that specific 'synaptic clinical trials' should be designed and launched with the aim of establishing whether phenotype 'reversals' could also occur in humans.
Collapse
|
161
|
Eps8 controls dendritic spine density and synaptic plasticity through its actin-capping activity. EMBO J 2013; 32:1730-44. [PMID: 23685357 DOI: 10.1038/emboj.2013.107] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2013] [Accepted: 04/15/2013] [Indexed: 12/13/2022] Open
Abstract
Actin-based remodelling underlies spine structural changes occurring during synaptic plasticity, the process that constantly reshapes the circuitry of the adult brain in response to external stimuli, leading to learning and memory formation. A positive correlation exists between spine shape and synaptic strength and, consistently, abnormalities in spine number and morphology have been described in a number of neurological disorders. In the present study, we demonstrate that the actin-regulating protein, Eps8, is recruited to the spine head during chemically induced long-term potentiation in culture and that inhibition of its actin-capping activity impairs spine enlargement and plasticity. Accordingly, mice lacking Eps8 display immature spines, which are unable to undergo potentiation, and are impaired in cognitive functions. Additionally, we found that reduction in the levels of Eps8 occurs in brains of patients affected by autism compared to controls. Our data reveal the key role of Eps8 actin-capping activity in spine morphogenesis and plasticity and indicate that reductions in actin-capping proteins may characterize forms of intellectual disabilities associated with spine defects.
Collapse
|
162
|
Norton WHJ. Toward developmental models of psychiatric disorders in zebrafish. Front Neural Circuits 2013; 7:79. [PMID: 23637652 PMCID: PMC3636468 DOI: 10.3389/fncir.2013.00079] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Accepted: 04/09/2013] [Indexed: 12/20/2022] Open
Abstract
Psychiatric disorders are a diverse set of diseases that affect all aspects of mental function including social interaction, thinking, feeling, and mood. Although psychiatric disorders place a large economic burden on society, the drugs available to treat them are often palliative with variable efficacy and intolerable side-effects. The development of novel drugs has been hindered by a lack of knowledge about the etiology of these diseases. It is thus necessary to further investigate psychiatric disorders using a combination of human molecular genetics, gene-by-environment studies, in vitro pharmacological and biochemistry experiments, animal models, and investigation of the non-biological basis of these diseases, such as environmental effects. Many psychiatric disorders, including autism spectrum disorder, attention-deficit/hyperactivity disorder, mental retardation, and schizophrenia can be triggered by alterations to neural development. The zebrafish is a popular model for developmental biology that is increasingly used to study human disease. Recent work has extended this approach to examine psychiatric disorders as well. However, since psychiatric disorders affect complex mental functions that might be human specific, it is not possible to fully model them in fish. In this review, I will propose that the suitability of zebrafish for developmental studies, and the genetic tools available to manipulate them, provide a powerful model to study the roles of genes that are linked to psychiatric disorders during neural development. The relative speed and ease of conducting experiments in zebrafish can be used to address two areas of future research: the contribution of environmental factors to disease onset, and screening for novel therapeutic compounds.
Collapse
Affiliation(s)
- William H J Norton
- Department of Biology, College of Medicine, Biological Sciences and Psychiatry, University of Leicester Leicester, UK
| |
Collapse
|
163
|
Leung LC, Wang GX, Mourrain P. Imaging zebrafish neural circuitry from whole brain to synapse. Front Neural Circuits 2013; 7:76. [PMID: 23630470 PMCID: PMC3634052 DOI: 10.3389/fncir.2013.00076] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Accepted: 04/03/2013] [Indexed: 12/20/2022] Open
Abstract
Recent advances in imaging tools are inspiring zebrafish researchers to tackle ever more ambitious questions in the neurosciences. Behaviorally fundamental conserved neural networks can now be potentially studied using zebrafish from a brain-wide scale to molecular resolution. In this perspective, we offer a roadmap by which a zebrafish researcher can navigate the course from collecting neural activities across the brain associated with a behavior, to unraveling molecular identities and testing the functional relevance of active neurons. In doing so, important insights will be gained as to how neural networks generate behaviors and assimilate changes in synaptic connectivity.
Collapse
Affiliation(s)
- Louis C Leung
- Department of Psychiatry and Behavioral Sciences, Center for Sleep Sciences, Beckman Center, Stanford University Palo Alto, CA, USA
| | | | | |
Collapse
|
164
|
Mosca-Boidron AL, Valduga M, Thauvin-Robinet C, Lagarde N, Marle N, Henry C, Pinoit JM, Huet F, Béri-Deixheimer M, Ragon C, Gueneau L, Payet M, Callier P, Mugneret F, Jonveaux P, Faivre L. Additional evidence to support the role of the 20q13.33 region in susceptibility to autism. Am J Med Genet A 2013; 161A:1505-7. [PMID: 23613186 DOI: 10.1002/ajmg.a.35878] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2012] [Accepted: 12/19/2012] [Indexed: 11/10/2022]
|
165
|
Possible use of Trichuris suis ova in autism spectrum disorders therapy. Med Hypotheses 2013; 81:1-4. [PMID: 23597946 DOI: 10.1016/j.mehy.2013.03.024] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Revised: 03/01/2013] [Accepted: 03/17/2013] [Indexed: 12/24/2022]
Abstract
Autism and autism spectrum disorders (ASDs) are heterogeneous, severe neurodevelopmental pathologies. The main core symptoms are: dysfunctions in social interactions and communication skills, restricted interests, repetitive and stereotypic verbal and non-verbal behaviors. Several biochemical processes are associated with ASDs: oxidative stress; endoplasmic reticulum stress; decreased methylation capacity; limited production of glutathione; mitochondrial dysfunction; intestinal dysbiosis; increased toxic metal burden; immune dysregulation. Current available treatments for ASDs can be divided into behavioral, nutritional and medical approaches, although no defined standard approach exists. Current drugs fail to benefit the ASD core symptoms and can have marked adverse effects, are mainly palliative and only sometimes efficacy in attenuating specific autistic behaviors. Helminthic therapy shows potential for application as anti-inflammatory agent. Several human diseases can be treated by helminths (i.e. inflammatory bowel disease, asthma, multiple sclerosis and autoimmune diabetes). Trichuris suis ova (TSO) show strong immunomodulatory properties. Authors hypothesize that TSO could be useful in addressing ASD immune dysregulations. TSO could be a novel therapeutic option for ASD management.
Collapse
|
166
|
Poot M. Towards identification of individual etiologies by resolving genomic and biological conundrums in patients with autism spectrum disorders. Mol Syndromol 2013; 4:213-26. [PMID: 23885228 DOI: 10.1159/000350041] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/04/2013] [Indexed: 01/11/2023] Open
Abstract
Recent genomic research into autism spectrum disorders (ASD) has revealed a remarkably complex genetic architecture. Large numbers of common variants, copy number variations and single nucleotide variants have been identified, yet each of them individually afforded only a small phenotypic impact. A polygenic model in which multiple genes interact either in an additive or a synergistic way appears the most plausible for the majority of ASD patients. Based on recently identified ASD candidate genes, transgenic mouse models for neuroligins/neurorexins and genes such as Cntnap2, Cntn5, Tsc1, Tsc2, Akt3, Cyfip1, Scn1a, En2, Slc6a4, and Bckdk have been generated and studied with respect to behavioral and neuroanatomical phenotypes and sensitivity to drug treatments. From these models, a few clues for potential pharmacologic intervention emerged. The Fmr1, Shank2 and Cntn5 knockout mice exhibited alterations of glutamate receptors, which may become a target for pharmacologic modulation. Some of the phenotypes of Mecp2 knockout mice can be ameliorated by administering IGF1. In the near future, comprehensive genotyping of individual patients and siblings combined with the novel insights generated from the transgenic animal studies may provide us with personalized treatment options. Eventually, autism may indeed turn out to be a phenotypically heterogeneous group of disorders ('autisms') caused by combinations of changes in multiple possible candidate genes, being different in each patient and requiring for each combination of mutations a distinct, individually tailored treatment.
Collapse
Affiliation(s)
- M Poot
- Department of Medical Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
167
|
Bahl S, Chiang C, Beauchamp RL, Neale BM, Daly MJ, Gusella JF, Talkowski ME, Ramesh V. Lack of association of rare functional variants in TSC1/TSC2 genes with autism spectrum disorder. Mol Autism 2013; 4:5. [PMID: 23514105 PMCID: PMC3610211 DOI: 10.1186/2040-2392-4-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2012] [Accepted: 03/05/2013] [Indexed: 01/05/2023] Open
Abstract
Background Autism spectrum disorder (ASD) is reported in 30 to 60% of patients with tuberous sclerosis complex (TSC) but shared genetic mechanisms that exist between TSC-associated ASD and idiopathic ASD have yet to be determined. Through the small G-protein Rheb, the TSC proteins, hamartin and tuberin, negatively regulate mammalian target of rapamycin complex 1 (mTORC1) signaling. It is well established that mTORC1 plays a pivotal role in neuronal translation and connectivity, so dysregulation of mTORC1 signaling could be a common feature in many ASDs. Pam, an E3 ubiquitin ligase, binds to TSC proteins and regulates mTORC1 signaling in the CNS, and the FBXO45-Pam ubiquitin ligase complex plays an essential role in neurodevelopment by regulating synapse formation and growth. Since mounting evidence has established autism as a disorder of the synapses, we tested whether rare genetic variants in TSC1, TSC2, MYCBP2, RHEB and FBXO45, genes that regulate mTORC1 signaling and/or play a role in synapse development and function, contribute to the pathogenesis of idiopathic ASD. Methods Exons and splice junctions of TSC1, TSC2, MYCBP2, RHEB and FBXO45 were resequenced for 300 ASD trios from the Simons Simplex Collection (SSC) using a pooled PCR amplification and next-generation sequencing strategy, targeted to the discovery of deleterious coding variation. These detected, potentially functional, variants were confirmed by Sanger sequencing of the individual samples comprising the pools in which they were identified. Results We identified a total of 23 missense variants in MYCBP2, TSC1 and TSC2. These variants exhibited a near equal distribution between the proband and parental pools, with no statistical excess in ASD cases (P > 0.05). All proband variants were inherited. No putative deleterious variants were confirmed in RHEB and FBXO45. Three intronic variants, identified as potential splice defects in MYCBP2 did not show aberrant splicing upon RNA assay. Overall, we did not find an over-representation of ASD causal variants in the genes studied to support them as contributors to autism susceptibility. Conclusions We did not observe an enrichment of rare functional variants in TSC1 and TSC2 genes in our sample set of 300 trios.
Collapse
Affiliation(s)
- Samira Bahl
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA 02114, USA.
| | | | | | | | | | | | | | | |
Collapse
|
168
|
Sudarov A, Gooden F, Tseng D, Gan WB, Ross ME. Lis1 controls dynamics of neuronal filopodia and spines to impact synaptogenesis and social behaviour. EMBO Mol Med 2013; 5:591-607. [PMID: 23483716 PMCID: PMC3628102 DOI: 10.1002/emmm.201202106] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Revised: 01/29/2013] [Accepted: 01/31/2013] [Indexed: 11/08/2022] Open
Abstract
LIS1 (PAFAH1B1) mutation can impair neuronal migration, causing lissencephaly in humans. LIS1 loss is associated with dynein protein motor dysfunction, and disrupts the actin cytoskeleton through disregulated RhoGTPases. Recently, LIS1 was implicated as an important protein-network interaction node with high-risk autism spectrum disorder genes expressed in the synapse. How LIS1 might participate in this disorder has not been investigated. We examined the role of LIS1 in synaptogenesis of post-migrational neurons and social behaviour in mice. Two-photon imaging of actin-rich dendritic filopodia and spines in vivo showed significant reductions in elimination and turnover rates of dendritic protrusions of layer V pyramidal neurons in adolescent Lis1+/− mice. Lis1+/− filopodia on immature hippocampal neurons in vitro exhibited reduced density, length and RhoA dependent impaired dynamics compared to Lis1+/+. Moreover, Lis1+/− adolescent mice exhibited deficits in social interaction. Lis1 inactivation restricted to the postnatal hippocampus resulted in similar deficits in dendritic protrusion density and social interactions. Thus, LIS1 plays prominently in dendritic filopodia dynamics and spine turnover implicating reduced dendritic spine plasticity as contributing to developmental autistic-like behaviour.
Collapse
Affiliation(s)
- Anamaria Sudarov
- Brain and Mind Research Institute, Weill Medical College of Cornell University, New York, USA
| | | | | | | | | |
Collapse
|
169
|
van Alphen B, van Swinderen B. Drosophila strategies to study psychiatric disorders. Brain Res Bull 2013; 92:1-11. [DOI: 10.1016/j.brainresbull.2011.09.007] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2011] [Revised: 09/08/2011] [Accepted: 09/09/2011] [Indexed: 01/03/2023]
|
170
|
Lionel AC, Vaags AK, Sato D, Gazzellone MJ, Mitchell EB, Chen HY, Costain G, Walker S, Egger G, Thiruvahindrapuram B, Merico D, Prasad A, Anagnostou E, Fombonne E, Zwaigenbaum L, Roberts W, Szatmari P, Fernandez BA, Georgieva L, Brzustowicz LM, Roetzer K, Kaschnitz W, Vincent JB, Windpassinger C, Marshall CR, Trifiletti RR, Kirmani S, Kirov G, Petek E, Hodge JC, Bassett AS, Scherer SW. Rare exonic deletions implicate the synaptic organizer Gephyrin (GPHN) in risk for autism, schizophrenia and seizures. Hum Mol Genet 2013; 22:2055-66. [DOI: 10.1093/hmg/ddt056] [Citation(s) in RCA: 112] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
171
|
Kim KC, Kim P, Go HS, Choi CS, Park JH, Kim HJ, Jeon SJ, dela Pena IC, Han SH, Cheong JH, Ryu JH, Shin CY. Male-specific alteration in excitatory post-synaptic development and social interaction in pre-natal valproic acid exposure model of autism spectrum disorder. J Neurochem 2013; 124:832-43. [DOI: 10.1111/jnc.12147] [Citation(s) in RCA: 140] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Revised: 12/17/2012] [Accepted: 01/07/2013] [Indexed: 11/28/2022]
Affiliation(s)
- Ki Chan Kim
- Department of Pharmacology; College of Pharmacy; Seoul National University; Seoul Korea
- Center for Neuroscience Research; SMART Institute of Advanced Biomedical Sciences; Konkuk University; Seoul Korea
| | - Pitna Kim
- Center for Neuroscience Research; SMART Institute of Advanced Biomedical Sciences; Konkuk University; Seoul Korea
- School of Medicine; Konkuk University; Seoul Korea
| | - Hyo Sang Go
- Department of Pharmacology; College of Pharmacy; Seoul National University; Seoul Korea
- Center for Neuroscience Research; SMART Institute of Advanced Biomedical Sciences; Konkuk University; Seoul Korea
| | - Chang Soon Choi
- Center for Neuroscience Research; SMART Institute of Advanced Biomedical Sciences; Konkuk University; Seoul Korea
- School of Medicine; Konkuk University; Seoul Korea
| | - Jin Hee Park
- School of Medicine; Konkuk University; Seoul Korea
| | - Hee Jin Kim
- Department of Pharmacy; Sahmyook University; Seoul Korea
| | - Se Jin Jeon
- Department of Psychiatry; School of Medicine; University of California; Los Angeles California USA
| | | | - Seol-Heui Han
- Center for Neuroscience Research; SMART Institute of Advanced Biomedical Sciences; Konkuk University; Seoul Korea
- School of Medicine; Konkuk University; Seoul Korea
| | | | - Jong Hoon Ryu
- Department of Oriental Pharmaceutical Science; College of Pharmacy; Kyung Hee University; Seoul Korea
| | - Chan Young Shin
- Center for Neuroscience Research; SMART Institute of Advanced Biomedical Sciences; Konkuk University; Seoul Korea
- School of Medicine; Konkuk University; Seoul Korea
| |
Collapse
|
172
|
Abstract
Autism spectrum disorder (ASD) is a highly heterogeneous disorder diagnosed based on the presence and severity of core abnormalities in social communication and repetitive behavior, yet several studies converge on immune dysregulation as a feature of ASD. Widespread alterations in immune molecules and responses are seen in the brains and periphery of ASD individuals, and early life immune disruptions are associated with ASD. This chapter discusses immune-related environmental and genetic risk factors for ASD, emphasizing population-wide studies and animal research that reveal potential mechanistic pathways involved in the development of ASD-related symptoms. It further reviews immunologic pathologies seen in ASD individuals and how such abnormalities can impact neurodevelopment and behavior. Finally, it evaluates emerging evidence for an immune contribution to the pathogenesis of ASD and a potential role for immunomodulatory effects in current treatments for ASD.
Collapse
Affiliation(s)
- Elaine Y Hsiao
- Division of Biology and Biological Engineering, Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, California, USA.
| |
Collapse
|
173
|
Palumbo O, D'Agruma L, Minenna AF, Palumbo P, Stallone R, Palladino T, Zelante L, Carella M. 3p14.1 de novo microdeletion involving the FOXP1 gene in an adult patient with autism, severe speech delay and deficit of motor coordination. Gene 2012; 516:107-13. [PMID: 23287644 DOI: 10.1016/j.gene.2012.12.073] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2012] [Revised: 10/09/2012] [Accepted: 12/02/2012] [Indexed: 01/01/2023]
Abstract
Interstitial deletion of chromosome region 3p14.1, including FOXP1 gene, is relatively rare and, until recently, there were no strong evidences to support the hypothesis that this microdeletion could play a role in the etiology of genomic disorders. Here, we report on an adult patient with a recognizable phenotype of autism, severe speech delay, deficit of motor coordination and typical dysmorphic features. Analysis of a dense whole genome single-nucleotide polymorphism (SNP) array showed a 1Mb interstitial deletion of chromosome region 3p14.1 including the entire coding region of FOXP1 (MIM 605515) gene. In order to study the parental origin of the deletion, we analyzed selected SNPs in the deleted area in the proband and his parents showing Mendelian incompatibilities suggesting a de novo deletion on the chromosome of paternal origin. Despite the frequency of this genomic alteration has not been estimated, our patient confirm the hypothesis that microdeletion of 3p14.1 seems to be a rare cause of cognitive disorders and that haploinsufficiency of FOXP1 may play a role in neurological and language deficits in patients carrying a 3p14.1 deletion. Finally, our patient is also important because useful to further delineate the clinical spectrum secondary to the 3p14.1 microdeletions.
Collapse
Affiliation(s)
- Orazio Palumbo
- Medical Genetics Unit, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo (FG), Italy
| | | | | | | | | | | | | | | |
Collapse
|
174
|
Millan MJ. An epigenetic framework for neurodevelopmental disorders: from pathogenesis to potential therapy. Neuropharmacology 2012; 68:2-82. [PMID: 23246909 DOI: 10.1016/j.neuropharm.2012.11.015] [Citation(s) in RCA: 151] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2012] [Revised: 11/11/2012] [Accepted: 11/22/2012] [Indexed: 12/12/2022]
Abstract
Neurodevelopmental disorders (NDDs) are characterized by aberrant and delayed early-life development of the brain, leading to deficits in language, cognition, motor behaviour and other functional domains, often accompanied by somatic symptoms. Environmental factors like perinatal infection, malnutrition and trauma can increase the risk of the heterogeneous, multifactorial and polygenic disorders, autism and schizophrenia. Conversely, discrete genetic anomalies are involved in Down, Rett and Fragile X syndromes, tuberous sclerosis and neurofibromatosis, the less familiar Phelan-McDermid, Sotos, Kleefstra, Coffin-Lowry and "ATRX" syndromes, and the disorders of imprinting, Angelman and Prader-Willi syndromes. NDDs have been termed "synaptopathies" in reference to structural and functional disturbance of synaptic plasticity, several involve abnormal Ras-Kinase signalling ("rasopathies"), and many are characterized by disrupted cerebral connectivity and an imbalance between excitatory and inhibitory transmission. However, at a different level of integration, NDDs are accompanied by aberrant "epigenetic" regulation of processes critical for normal and orderly development of the brain. Epigenetics refers to potentially-heritable (by mitosis and/or meiosis) mechanisms controlling gene expression without changes in DNA sequence. In certain NDDs, prototypical epigenetic processes of DNA methylation and covalent histone marking are impacted. Conversely, others involve anomalies in chromatin-modelling, mRNA splicing/editing, mRNA translation, ribosome biogenesis and/or the regulatory actions of small nucleolar RNAs and micro-RNAs. Since epigenetic mechanisms are modifiable, this raises the hope of novel therapy, though questions remain concerning efficacy and safety. The above issues are critically surveyed in this review, which advocates a broad-based epigenetic framework for understanding and ultimately treating a diverse assemblage of NDDs ("epigenopathies") lying at the interface of genetic, developmental and environmental processes. This article is part of the Special Issue entitled 'Neurodevelopmental Disorders'.
Collapse
Affiliation(s)
- Mark J Millan
- Unit for Research and Discovery in Neuroscience, IDR Servier, 125 chemin de ronde, 78290 Croissy sur Seine, Paris, France.
| |
Collapse
|
175
|
Lanore F, Labrousse VF, Szabo Z, Normand E, Blanchet C, Mulle C. Deficits in morphofunctional maturation of hippocampal mossy fiber synapses in a mouse model of intellectual disability. J Neurosci 2012; 32:17882-93. [PMID: 23223307 PMCID: PMC6621665 DOI: 10.1523/jneurosci.2049-12.2012] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2012] [Revised: 10/16/2012] [Accepted: 10/18/2012] [Indexed: 01/04/2023] Open
Abstract
The grik2 gene, coding for the kainate receptor subunit GluK2 (formerly GluR6), is associated with autism spectrum disorders and intellectual disability. Here, we tested the hypothesis that GluK2 could play a role in the appropriate maturation of synaptic circuits involved in learning and memory. We show that both the functional and morphological maturation of hippocampal mossy fiber to CA3 pyramidal cell (mf-CA3) synapses is delayed in mice deficient for the GluK2 subunit (GluK2⁻/⁻). In GluK2⁻/⁻ mice this deficit is manifested by a transient reduction in the amplitude of AMPA-EPSCs at a critical time point of postnatal development, whereas the NMDA component is spared. By combining multiple probability peak fluctuation analysis and immunohistochemistry, we have provided evidence that the decreased amplitude reflects a decrease in the quantal size per mf-CA3 synapse and in the number of active synaptic sites. Furthermore, we analyzed the time course of structural maturation of CA3 synapses by confocal imaging of YFP-expressing cells followed by tridimensional (3D) anatomical reconstruction of thorny excrescences and presynaptic boutons. We show that major changes in synaptic structures occur subsequently to the sharp increase in synaptic transmission, and more importantly that the course of structural maturation of synaptic elements is impaired in GluK2⁻/⁻ mice. This study highlights how a mutation in a gene linked to intellectual disability in the human may lead to a transient reduction of synaptic strength during postnatal development, impacting on the proper formation of neural circuits linked to memory.
Collapse
MESH Headings
- Animals
- Animals, Outbred Strains
- Disease Models, Animal
- Excitatory Amino Acid Agonists/pharmacology
- Excitatory Postsynaptic Potentials/genetics
- Excitatory Postsynaptic Potentials/physiology
- Intellectual Disability/genetics
- Intellectual Disability/metabolism
- Intellectual Disability/pathology
- Intellectual Disability/physiopathology
- Mice
- Mice, Knockout
- Mossy Fibers, Hippocampal/drug effects
- Mossy Fibers, Hippocampal/growth & development
- Mossy Fibers, Hippocampal/pathology
- Mossy Fibers, Hippocampal/physiopathology
- N-Methylaspartate/pharmacology
- Presynaptic Terminals/pathology
- Receptors, Kainic Acid/agonists
- Receptors, Kainic Acid/genetics
- Receptors, Kainic Acid/physiology
- Synapses/pathology
- alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/pharmacology
- GluK2 Kainate Receptor
Collapse
Affiliation(s)
- Frederic Lanore
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, and
- CNRS UMR 5297, F-33000 Bordeaux, France
| | - Virginie F. Labrousse
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, and
- CNRS UMR 5297, F-33000 Bordeaux, France
| | - Zsolt Szabo
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, and
- CNRS UMR 5297, F-33000 Bordeaux, France
| | - Elisabeth Normand
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, and
- CNRS UMR 5297, F-33000 Bordeaux, France
| | - Christophe Blanchet
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, and
- CNRS UMR 5297, F-33000 Bordeaux, France
| | - Christophe Mulle
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, and
- CNRS UMR 5297, F-33000 Bordeaux, France
| |
Collapse
|
176
|
Wagnon JL, Briese M, Sun W, Mahaffey CL, Curk T, Rot G, Ule J, Frankel WN. CELF4 regulates translation and local abundance of a vast set of mRNAs, including genes associated with regulation of synaptic function. PLoS Genet 2012; 8:e1003067. [PMID: 23209433 PMCID: PMC3510034 DOI: 10.1371/journal.pgen.1003067] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2012] [Accepted: 09/20/2012] [Indexed: 11/25/2022] Open
Abstract
RNA–binding proteins have emerged as causal agents of complex neurological diseases. Mice deficient for neuronal RNA–binding protein CELF4 have a complex neurological disorder with epilepsy as a prominent feature. Human CELF4 has recently been associated with clinical features similar to those seen in mutant mice. CELF4 is expressed primarily in excitatory neurons, including large pyramidal cells of the cerebral cortex and hippocampus, and it regulates excitatory but not inhibitory neurotransmission. We examined mechanisms underlying neuronal hyperexcitability in Celf4 mutants by identifying CELF4 target mRNAs and assessing their fate in the absence of CELF4 in view of their known functions. CELF4 binds to at least 15%–20% of the transcriptome, with striking specificity for the mRNA 3′ untranslated region. CELF4 mRNA targets encode a variety of proteins, many of which are well established in neuron development and function. While the overall abundance of these mRNA targets is often dysregulated in Celf4 deficient mice, the actual expression changes are modest at the steady-state level. In contrast, by examining the transcriptome of polysome fractions and the mRNA distribution along the neuronal cell body-neuropil axis, we found that CELF4 is critical for maintaining mRNA stability and availability for translation. Among biological processes associated with CELF4 targets that accumulate in neuropil of mutants, regulation of synaptic plasticity and transmission are the most prominent. Together with a related study of the impact of CELF4 loss on sodium channel Nav1.6 function, we suggest that CELF4 deficiency leads to abnormal neuronal function by combining a specific effect on neuronal excitation with a general impairment of synaptic transmission. These results also expand our understanding of the vital roles RNA–binding proteins play in regulating and shaping the activity of neural circuits. Epilepsy is a devastating brain disorder whereby a loss of regulation of electrochemical signals between neurons causes too much excitation and ultimately results in an “electrical storm” known as a seizure. Epilepsy can be heritable, but it is usually genetically complex, resulting from a collaboration of many genes. It is also a frequent feature of other common brain diseases, such as autism spectrum disorder and intellectual disability, likely because these diseases have a similar dysregulation of neuronal communication. To understand more about how the brain regulates electrical activity, we focused on an RNA–binding protein called CELF4, because a) mice that lack CELF4 have a complex form of epilepsy that includes features of other neurological diseases and b) this kind of protein has the potential to be a master regulator. We show that CELF4 binds to a vast array of mRNAs, and without CELF4 these mRNAs accumulate in the wrong places and can produce the wrong amount of protein. Moreover, many of these mRNAs encode key players in electrochemical signaling between neurons. Although the defects in individual mRNAs are modest, like a genetically complex disease, together these alterations collude to cause neurological symptoms including recurrent seizures.
Collapse
Affiliation(s)
- Jacy L. Wagnon
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
| | - Michael Briese
- MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - Wenzhi Sun
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
| | | | - Tomaž Curk
- Faculty of Computer and Information Science, University of Ljubljana, Ljubljana, Slovenia
| | - Gregor Rot
- Faculty of Computer and Information Science, University of Ljubljana, Ljubljana, Slovenia
| | - Jernej Ule
- MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - Wayne N. Frankel
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
- * E-mail:
| |
Collapse
|
177
|
Potential opposite roles of the extracellular signal-regulated kinase (ERK) pathway in autism spectrum and bipolar disorders. Neurosci Biobehav Rev 2012; 36:2206-13. [DOI: 10.1016/j.neubiorev.2012.07.008] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Revised: 07/20/2012] [Accepted: 07/28/2012] [Indexed: 11/22/2022]
|
178
|
The expression of caspases is enhanced in peripheral blood mononuclear cells of autism spectrum disorder patients. J Autism Dev Disord 2012; 42:1403-10. [PMID: 21969075 DOI: 10.1007/s10803-011-1373-z] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Autism and autism spectrum disorders (ASDs) are heterogeneous complex neuro-developmental disorders characterized by dysfunctions in social interaction and communication skills. Their pathogenesis has been linked to interactions between genes and environmental factors. Consistent with the evidence of certain similarities between immune cells and neurons, autistic children also show an altered immune response of peripheral blood mononuclear cells (PBMCs). In this study, we investigated the activation of caspases, cysteinyl aspartate-specific proteases involved in apoptosis and several other cell functions in PBMCs from 15 ASD children compared to age-matched normal healthy developing controls. The mRNA levels for caspase-1, -2, -4, -5 were significantly increased in ASD children as compared to healthy subjects. Protein levels of Caspase-3, -7, -12 were also increased in ASD patients. Our data are suggestive of a possible role of the caspase pathway in ASD clinical outcome and of the use of caspase as potential diagnostic and/or therapeutic tools in ASD management.
Collapse
|
179
|
Kalkman HO. A review of the evidence for the canonical Wnt pathway in autism spectrum disorders. Mol Autism 2012; 3:10. [PMID: 23083465 PMCID: PMC3492093 DOI: 10.1186/2040-2392-3-10] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2012] [Accepted: 10/04/2012] [Indexed: 12/21/2022] Open
Abstract
Microdeletion and microduplication copy number variations are found in patients with autism spectrum disorder and in a number of cases they include genes that are involved in the canonical Wnt signaling pathway (for example, FZD9, BCL9 or CDH8). Association studies investigating WNT2, DISC1, MET, DOCK4 or AHI1 also provide evidence that the canonical Wnt pathway might be affected in autism. Prenatal medication with sodium-valproate or antidepressant drugs increases autism risk. In animal studies, it has been found that these medications promote Wnt signaling, including among others an increase in Wnt2 gene expression. Notably, the available genetic information indicates that not only canonical Wnt pathway activation, but also inhibition seems to increase autism risk. The canonical Wnt pathway plays a role in dendrite growth and suboptimal activity negatively affects the dendritic arbor. In principle, this provides a logical explanation as to why both hypo- and hyperactivity may generate a similar set of behavioral and cognitive symptoms. However, without a validated biomarker to stratify for deviant canonical Wnt pathway activity, it is probably too dangerous to treat patients with compounds that modify pathway activity.
Collapse
Affiliation(s)
- Hans Otto Kalkman
- Neuroscience Department, Novartis Institute of Biomedical Research, Building 386-14,22,15, Basel, CH 4002, Switzerland.
| |
Collapse
|
180
|
Ey E, Yang M, Katz AM, Woldeyohannes L, Silverman JL, Leblond CS, Faure P, Torquet N, Le Sourd AM, Bourgeron T, Crawley JN. Absence of deficits in social behaviors and ultrasonic vocalizations in later generations of mice lacking neuroligin4. GENES BRAIN AND BEHAVIOR 2012; 11:928-941. [PMID: 22989184 DOI: 10.1111/j.1601-183x.2012.00849.x] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Revised: 08/03/2012] [Accepted: 09/04/2012] [Indexed: 11/30/2022]
Abstract
Mutations in NLGN4X have been identified in individuals with autism spectrum disorders and other neurodevelopmental disorders. A previous study reported that adult male mice lacking neuroligin4 (Nlgn4) displayed social approach deficits in the three-chambered test, altered aggressive behaviors and reduced ultrasonic vocalizations. To replicate and extend these findings, independent comprehensive analyses of autism-relevant behavioral phenotypes were conducted in later generations of the same line of Nlgn4 mutant mice at the National Institute of Mental Health in Bethesda, MD, USA and at the Institut Pasteur in Paris, France. Adult social approach was normal in all three genotypes of Nlgn4 mice tested at both sites. Reciprocal social interactions in juveniles were similarly normal across genotypes. No genotype differences were detected in ultrasonic vocalizations in pups separated from the nest or in adults during reciprocal social interactions. Anxiety-like behaviors, self-grooming, rotarod and open field exploration did not differ across genotypes, and measures of developmental milestones and general health were normal. Our findings indicate an absence of autism-relevant behavioral phenotypes in subsequent generations of Nlgn4 mice tested at two locations. Testing environment and methods differed from the original study in some aspects, although the presence of normal sociability was seen in all genotypes when methods taken from Jamain et al. (2008) were used. The divergent results obtained from this study indicate that phenotypes may not be replicable across breeding generations, and highlight the significant roles of environmental, generational and/or procedural factors on behavioral phenotypes.
Collapse
Affiliation(s)
- E Ey
- Human Genetics and Cognitive Functions, URA 2182 'Genes, synapses and cognition', Institut Pasteur, Paris, France.,CNRS, URA 2182 'Genes, synapses and cognition', Institut Pasteur, Paris, France.,Human Genetics and Cognitive Functions, Sorbonne Paris Cité, University Paris Diderot, Paris, France
| | - M Yang
- Laboratory of Behavioral Neuroscience, National Institute of Mental Health, Bethesda, MD, USA
| | - A M Katz
- Laboratory of Behavioral Neuroscience, National Institute of Mental Health, Bethesda, MD, USA
| | - L Woldeyohannes
- Laboratory of Behavioral Neuroscience, National Institute of Mental Health, Bethesda, MD, USA
| | - J L Silverman
- Laboratory of Behavioral Neuroscience, National Institute of Mental Health, Bethesda, MD, USA
| | - C S Leblond
- Human Genetics and Cognitive Functions, URA 2182 'Genes, synapses and cognition', Institut Pasteur, Paris, France.,CNRS, URA 2182 'Genes, synapses and cognition', Institut Pasteur, Paris, France.,Human Genetics and Cognitive Functions, Sorbonne Paris Cité, University Paris Diderot, Paris, France
| | - P Faure
- CNRS, UMR 7102, University Paris 06, Paris, France
| | - N Torquet
- Human Genetics and Cognitive Functions, URA 2182 'Genes, synapses and cognition', Institut Pasteur, Paris, France.,CNRS, URA 2182 'Genes, synapses and cognition', Institut Pasteur, Paris, France.,Human Genetics and Cognitive Functions, Sorbonne Paris Cité, University Paris Diderot, Paris, France
| | - A-M Le Sourd
- Human Genetics and Cognitive Functions, URA 2182 'Genes, synapses and cognition', Institut Pasteur, Paris, France.,CNRS, URA 2182 'Genes, synapses and cognition', Institut Pasteur, Paris, France.,Human Genetics and Cognitive Functions, Sorbonne Paris Cité, University Paris Diderot, Paris, France
| | - T Bourgeron
- Human Genetics and Cognitive Functions, URA 2182 'Genes, synapses and cognition', Institut Pasteur, Paris, France.,CNRS, URA 2182 'Genes, synapses and cognition', Institut Pasteur, Paris, France.,Human Genetics and Cognitive Functions, Sorbonne Paris Cité, University Paris Diderot, Paris, France
| | - J N Crawley
- Laboratory of Behavioral Neuroscience, National Institute of Mental Health, Bethesda, MD, USA
| |
Collapse
|
181
|
Swillen A, de Ravel T, Oliver C. Social phenotypes in genetic syndromes. JOURNAL OF INTELLECTUAL DISABILITY RESEARCH : JIDR 2012; 56:919-921. [PMID: 22973833 DOI: 10.1111/j.1365-2788.2012.01620.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
|
182
|
Baudouin SJ, Gaudias J, Gerharz S, Hatstatt L, Zhou K, Punnakkal P, Tanaka KF, Spooren W, Hen R, De Zeeuw CI, Vogt K, Scheiffele P. Shared Synaptic Pathophysiology in Syndromic and Nonsyndromic Rodent Models of Autism. Science 2012; 338:128-32. [DOI: 10.1126/science.1224159] [Citation(s) in RCA: 235] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The genetic heterogeneity of autism poses a major challenge for identifying mechanism-based treatments. A number of rare mutations are associated with autism, and it is unclear whether these result in common neuronal alterations. Monogenic syndromes, such as fragile X, include autism as one of their multifaceted symptoms and have revealed specific defects in synaptic plasticity. We discovered an unexpected convergence of synaptic pathophysiology in a nonsyndromic form of autism with those in fragile X syndrome. Neuroligin-3 knockout mice (a model for nonsyndromic autism) exhibited disrupted heterosynaptic competition and perturbed metabotropic glutamate receptor–dependent synaptic plasticity, a hallmark of fragile X. These phenotypes could be rescued by reexpression of neuroligin-3 in juvenile mice, highlighting the possibility of reverting neuronal circuit alterations in autism after the completion of development.
Collapse
|
183
|
Eriksson MA, Westerlund J, Anderlid BM, Gillberg C, Fernell E. First-degree relatives of young children with autism spectrum disorders: some gender aspects. RESEARCH IN DEVELOPMENTAL DISABILITIES 2012; 33:1642-1648. [PMID: 22554810 DOI: 10.1016/j.ridd.2012.03.025] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2012] [Revised: 03/10/2012] [Accepted: 03/12/2012] [Indexed: 05/31/2023]
Abstract
Prenatal risk factors, with special focus on gender distribution of neurodevelopmental and psychiatric conditions were analysed in first-degree relatives in a population-based group of young children with autism spectrum disorders (ASD). Multiple information sources were combined. This group was contrasted with the general population regarding data from the Swedish Medical Birth register. In the ASD group, information was also obtained at parental interviews focusing on developmental and psychiatric disorders in the family. Compared to the general population, fathers of children with ASD were older and parents more often of non-European origin. Mothers of children with ASD had an increased rate of antidepressant and psychoactive medication use, and of scheduled caesarean sections. Fathers and brothers of children with ASD had high rates of ASD including the broader phenotype. Mothers of children with ASD had high rates of depression and other psychiatric disorders. These findings, hypothetically, could reflect a different ASD phenotype and difficulties diagnosing ASD in females or be an example of the close genetic relation between ASD and other psychiatric disorders. The results suggest that, in clinical and research settings, the familial background in ASD should be reviewed with a broader approach, and not be restricted to "looking out" only for diagnoses and symptoms traditionally accepted as being part of or typical of ASD. The high rate of parents of non-European origin has been noted in many Swedish studies of ASD, but the reason for this association, remains unclear.
Collapse
Affiliation(s)
- Mats Anders Eriksson
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden; Karolinska Institutet, Center of Neurodevelopmental Disorders, Stockholm, Sweden.
| | | | | | | | | |
Collapse
|
184
|
Xu X, Xu Q, Zhang Y, Zhang X, Cheng T, Wu B, Ding Y, Lu P, Zheng J, Zhang M, Qiu Z, Yu X. A case report of Chinese brothers with inherited MECP2-containing duplication: autism and intellectual disability, but not seizures or respiratory infections. BMC MEDICAL GENETICS 2012; 13:75. [PMID: 22909152 PMCID: PMC3506511 DOI: 10.1186/1471-2350-13-75] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2012] [Accepted: 08/15/2012] [Indexed: 11/16/2022]
Abstract
Background Autistic spectrum disorders (ASDs) are a family of neurodevelopmental disorders with strong genetic components. Recent studies have shown that copy number variations in dosage sensitive genes can contribute significantly to these disorders. One such gene is the transcription factor MECP2, whose loss of function in females results in Rett syndrome, while its duplication in males results in developmental delay and autism. Case presentation Here, we identified a Chinese family with two brothers both inheriting a 2.2 Mb MECP2-containing duplication (151,369,305 – 153,589,577) from their mother. In addition, both brothers also had a 213.7 kb duplication on Chromosome 2, inherited from their father. The older brother also carried a 48.4 kb duplication on Chromosome 2 inherited from the mother, and a 8.2 kb deletion at 11q13.5 inherited from the father. Based on the published literature, MECP2 is the most autism-associated gene among the identified CNVs. Consistently, the boys displayed clinical features in common with other patients carrying MECP2 duplications, including intellectual disability, autism, lack of speech, slight hypotonia and unsteadiness of movement. They also had slight dysmorphic features including a depressed nose bridge, large ears and midface hypoplasia. Interestingly, they did not exhibit other clinical features commonly observed in American-European patients with MECP2 duplication, including recurrent respiratory infections and epilepsy. Conclusions To our knowledge, this is the first identification and characterization of Chinese Han patients with MECP2-containing duplications. Further cases are required to determine if the above described clinical differences are due to individual variations or related to the genetic background of the patients.
Collapse
Affiliation(s)
- Xiu Xu
- Department of Child Healthcare, Children's Hospital of Fudan University, Shanghai, China.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
185
|
Dombret C, Nguyen T, Schakman O, Michaud JL, Hardin-Pouzet H, Bertrand MJ, De Backer O. Loss of Maged1 results in obesity, deficits of social interactions, impaired sexual behavior and severe alteration of mature oxytocin production in the hypothalamus. Hum Mol Genet 2012; 21:4703-17. [DOI: 10.1093/hmg/dds310] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
186
|
Wöhr M, Silverman JL, Scattoni ML, Turner SM, Harris MJ, Saxena R, Crawley JN. Developmental delays and reduced pup ultrasonic vocalizations but normal sociability in mice lacking the postsynaptic cell adhesion protein neuroligin2. Behav Brain Res 2012; 251:50-64. [PMID: 22820233 DOI: 10.1016/j.bbr.2012.07.024] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2012] [Revised: 07/12/2012] [Accepted: 07/12/2012] [Indexed: 12/12/2022]
Abstract
Mutations in neurexin and neuroligin genes have been associated with neurodevelopmental disabilities including autism. Autism spectrum disorder is diagnosed by aberrant reciprocal social interactions, deficits in social communication, and repetitive, stereotyped patterns of behaviors, along with narrow restricted interests. Mouse models have been successfully used to study physiological and behavioral outcomes of mutations in the trans-synaptic neurexin-neuroligin complex. To further understand the behavioral consequences of Neuroligin2 (NLGN2) mutations, we assessed several behavioral phenotypes relevant to autism in neuroligin2 null (Nlgn2(-/-)), heterozygote (Nlgn2(+/-)), and wildtype (Nlgn2(+/+)) littermate control mice. Reduced breeding efficiency and high reactivity to handling was observed in Nlgn2(-/-) mice, resulting in low numbers of adult mice available for behavioral assessment. Consistent with previous findings, Nlgn2(-/-) mice displayed normal social behaviors, concomitant with reduced exploratory activity, impaired rotarod performance, and delays on several developmental milestones. No spontaneous stereotypies or repetitive behaviors were detected. Acoustic, tactile, and olfactory sensory information processing as well as sensorimotor gating were not affected. Nlgn2(-/-) pups isolated from mother and littermates emitted fewer ultrasonic vocalizations and spent less time calling than Nlgn2(+/+) littermate controls. The present findings add to the growing literature on the role of neurexins and neuroligins in physiology and behavior relevant to neurodevelopmental disorders.
Collapse
Affiliation(s)
- Markus Wöhr
- Laboratory of Behavioral Neuroscience, Intramural Research Program, National Institute of Mental Health, NIH, Bethesda, MD 20892, USA.
| | | | | | | | | | | | | |
Collapse
|
187
|
Kim KY, Jung YW, Sullivan GJ, Chung L, Park IH. Cellular reprogramming: a novel tool for investigating autism spectrum disorders. Trends Mol Med 2012; 18:463-71. [PMID: 22771169 DOI: 10.1016/j.molmed.2012.06.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2012] [Revised: 06/05/2012] [Accepted: 06/11/2012] [Indexed: 12/19/2022]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by impairment in reciprocal social interaction and communication, as well as the manifestation of stereotyped behaviors. Despite much effort, ASDs are not yet fully understood. Advanced genetics and genomics technologies have recently identified novel ASD genes, and approaches using genetically engineered murine models or postmortem human brain have facilitated understanding ASD. Reprogramming somatic cells into induced pluripotent stem cells (iPSCs) provides unprecedented opportunities in generating human disease models. Here, we present an overview of applying iPSCs in developing cellular models for understanding ASD. We also discuss future perspectives in the use of iPSCs as a source of cell therapy and as a screening platform for identifying small molecules with efficacy for alleviating ASD.
Collapse
Affiliation(s)
- Kun-Yong Kim
- Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, 10 Amistad, 201B, New Haven, CT 06520, USA
| | | | | | | | | |
Collapse
|
188
|
Camacho-Garcia RJ, Planelles MI, Margalef M, Pecero ML, Martínez-Leal R, Aguilera F, Vilella E, Martinez-Mir A, Scholl FG. Mutations affecting synaptic levels of neurexin-1β in autism and mental retardation. Neurobiol Dis 2012; 47:135-43. [DOI: 10.1016/j.nbd.2012.03.031] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2011] [Revised: 03/19/2012] [Accepted: 03/24/2012] [Indexed: 11/26/2022] Open
|
189
|
Section summary and perspectives: Translational medicine in psychiatry. Transl Neurosci 2012. [DOI: 10.1017/cbo9780511980053.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
|
190
|
Lesca G, Rudolf G, Labalme A, Hirsch E, Arzimanoglou A, Genton P, Motte J, de Saint Martin A, Valenti MP, Boulay C, De Bellescize J, Kéo-Kosal P, Boutry-Kryza N, Edery P, Sanlaville D, Szepetowski P. Epileptic encephalopathies of the Landau-Kleffner and continuous spike and waves during slow-wave sleep types: genomic dissection makes the link with autism. Epilepsia 2012; 53:1526-38. [PMID: 22738016 DOI: 10.1111/j.1528-1167.2012.03559.x] [Citation(s) in RCA: 122] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
PURPOSE The continuous spike and waves during slow-wave sleep syndrome (CSWSS) and the Landau-Kleffner (LKS) syndrome are two rare epileptic encephalopathies sharing common clinical features including seizures and regression. Both CSWSS and LKS can be associated with the electroencephalography pattern of electrical status epilepticus during slow-wave sleep and are part of a clinical continuum that at its benign end also includes rolandic epilepsy (RE) with centrotemporal spikes. The CSWSS and LKS patients can also have behavioral manifestations that overlap the spectrum of autism disorders (ASD). An impairment of brain development and/or maturation with complex interplay between genetic predisposition and nongenetic factors has been suspected. A role for autoimmunity has been proposed but the pathophysiology of CSWSS and of LKS remains uncharacterized. METHODS In recent years, the participation of rare genomic alterations in the susceptibility to epileptic and autistic disorders has been demonstrated. The involvement of copy number variations (CNVs) in 61 CSWSS and LKS patients was questioned using comparative genomic hybridization assays coupled with validation by quantitative polymerase chain reaction (PCR). KEY FINDINGS Whereas the patients showed highly heterogeneous in genomic architecture, several potentially pathogenic alterations were detected. A large number of these corresponded to genomic regions or genes (ATP13A4, CDH9, CDH13, CNTNAP2, CTNNA3, DIAPH3, GRIN2A, MDGA2, SHANK3) that have been either associated with ASD for most of them, or involved in speech or language impairment, or in RE. Particularly, CNVs encoding cell adhesion proteins (cadherins, protocadherins, contactins, catenins) were detected with high frequency (≈20% of the patients) and significant enrichment (cell adhesion: p = 0.027; cell adhesion molecule binding: p = 9.27 × 10(-7)). SIGNIFICANCE Overall our data bring the first insights into the possible molecular pathophysiology of CSWSS and LKS. The overrepresentation of cell adhesion genes and the strong overlap with the genetic, genomic and molecular ASD networks, provide an exciting and unifying view on the clinical links among CSWSS, LKS, and ASD.
Collapse
Affiliation(s)
- Gaetan Lesca
- Department of Constitutional Cytogenetics, Lyon Hospices Civils, Lyon, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
191
|
Ruan YW, Han XJ, Shi ZS, Lei ZG, Xu ZC. Remodeling of synapses in the CA1 area of the hippocampus after transient global ischemia. Neuroscience 2012; 218:268-77. [PMID: 22634576 DOI: 10.1016/j.neuroscience.2012.05.035] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2012] [Revised: 04/30/2012] [Accepted: 05/14/2012] [Indexed: 10/28/2022]
Abstract
Synapses are essential to neuronal functions. Synaptic changes occur under physiological and pathological conditions. Here we report the remodeling of synapses in the CA1 area of the hippocampus after transient global ischemia using electron microscopy. Much electron-dense material appeared in the cytoplasm of dendrites at 24h after ischemia. Many dark axons or terminals were found in the CA1 neuropil; some of which were phagocytized by dendrites. Interestingly autophagosomes appeared in many axons or dendrites at 48 h after ischemia. In addition, postsynaptic density (PSD) - like structures or synaptic - like structures were found inside spines and dendrites. Statistical analysis demonstrated that the thickness of PSDs in the CA1 neuropil increased from 12 to 48 h after ischemia. The frequency of autophagosomes appeared to escalate from 12 to 48 h after ischemia. The frequency of asymmetric synapses was significantly increased at 12h and 24h after ischemia in stratum oriens, proximal and distal stratum radiatum. Among asymmetric synapses, the number of perforated synapses consistently increased and reached a peak (approximately 10-fold increase) at 48 h after ischemia. On the other hand, the number of multiple synaptic boutons decreased after ischemia reaching a two to fourfold decrease at 48 h after ischemia. These results have shown that ischemia induces an increase of asymmetric synapses as well as synaptic autophagy, which may contribute to the neuronal death in the CA1 area after transient global ischemia.
Collapse
Affiliation(s)
- Y-W Ruan
- Joint Laboratory for Brain Health and Function of Jinan University, The University of Hong Kong, Jinan University School of Medicine, Guangzhou 510632, China.
| | | | | | | | | |
Collapse
|
192
|
Qiu S, Aldinger KA, Levitt P. Modeling of autism genetic variations in mice: focusing on synaptic and microcircuit dysfunctions. Dev Neurosci 2012; 34:88-100. [PMID: 22572629 DOI: 10.1159/000336644] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2011] [Accepted: 12/21/2011] [Indexed: 12/24/2022] Open
Abstract
Autism spectrum disorders (ASD) are heterogeneous neurodevelopmental disorders that are characterized by deficits in social interaction, verbal and nonverbal communication, and restrictive interests and repetitive behaviors. While human genetic studies have revealed marked heritability in ASD, it has been challenging to translate this genetic risk into a biological mechanism that influences brain development relevant to the disorder phenotypes. This is partly due to the complex genetic architecture of ASD, which involves de novo gene mutations, genomic abnormalities, and common genetic variants. Rather than trying to reconstitute the clinical disorder, using genetic model animals to examine specific features of core ASD pathophysiology offers unique opportunities for refining our understanding of neurodevelopmental mechanisms in ASD. A variety of ASD-relevant phenotypes can now be investigated in rodents, including stereotyped and repetitive behaviors, and deficits in social interaction and communication. In this review, we focus on several prevailing mouse models and discuss how studies have advanced our understanding of synaptic mechanisms that may underlie ASD pathophysiology. Although synaptic perturbations are not the only alterations relevant for ASD, we reason that understanding the synaptic underpinnings of ASD using mouse models may provide mechanistic insights into its etiology and lead to novel therapeutic and interventional strategies.
Collapse
Affiliation(s)
- Shenfeng Qiu
- Department of Cell and Neurobiology, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, Calif., USA
| | | | | |
Collapse
|
193
|
Sato D, Lionel AC, Leblond CS, Prasad A, Pinto D, Walker S, O'Connor I, Russell C, Drmic IE, Hamdan FF, Michaud JL, Endris V, Roeth R, Delorme R, Huguet G, Leboyer M, Rastam M, Gillberg C, Lathrop M, Stavropoulos DJ, Anagnostou E, Weksberg R, Fombonne E, Zwaigenbaum L, Fernandez BA, Roberts W, Rappold GA, Marshall CR, Bourgeron T, Szatmari P, Scherer SW. SHANK1 Deletions in Males with Autism Spectrum Disorder. Am J Hum Genet 2012; 90:879-87. [PMID: 22503632 DOI: 10.1016/j.ajhg.2012.03.017] [Citation(s) in RCA: 250] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2012] [Revised: 03/09/2012] [Accepted: 03/19/2012] [Indexed: 12/11/2022] Open
Abstract
Recent studies have highlighted the involvement of rare (<1% frequency) copy-number variations and point mutations in the genetic etiology of autism spectrum disorder (ASD); these variants particularly affect genes involved in the neuronal synaptic complex. The SHANK gene family consists of three members (SHANK1, SHANK2, and SHANK3), which encode scaffolding proteins required for the proper formation and function of neuronal synapses. Although SHANK2 and SHANK3 mutations have been implicated in ASD and intellectual disability, the involvement of SHANK1 is unknown. Here, we assess microarray data from 1,158 Canadian and 456 European individuals with ASD to discover microdeletions at the SHANK1 locus on chromosome 19. We identify a hemizygous SHANK1 deletion that segregates in a four-generation family in which male carriers--but not female carriers--have ASD with higher functioning. A de novo SHANK1 deletion was also detected in an unrelated male individual with ASD with higher functioning, and no equivalent SHANK1 mutations were found in >15,000 controls (p = 0.009). The discovery of apparent reduced penetrance of ASD in females bearing inherited autosomal SHANK1 deletions provides a possible contributory model for the male gender bias in autism. The data are also informative for clinical-genetics interpretations of both inherited and sporadic forms of ASD involving SHANK1.
Collapse
Affiliation(s)
- Daisuke Sato
- The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
194
|
Liu Y, Hu Z, Xun G, Peng Y, Lu L, Xu X, Xiong Z, Xia L, Liu D, Li W, Zhao J, Xia K. Mutation analysis of the NRXN1 gene in a Chinese autism cohort. J Psychiatr Res 2012; 46:630-4. [PMID: 22405623 DOI: 10.1016/j.jpsychires.2011.10.015] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2011] [Revised: 10/31/2011] [Accepted: 10/31/2011] [Indexed: 10/28/2022]
Abstract
Autism is a brain developmental disorder characterized by impaired social interaction and communication, as well as restricted and repetitive behaviors. The neurexin-1(NRXN1) gene mapped on chromosome 2p16.3 encodes neurexin, a cell adhesion molecule and receptor in the vertebrate nervous system. Rare de novo alterations and copy number variations (CNVs) suggested neurexin-1 as a candidate gene for the pathogenesis of autism, but data on the gene mutation of neurexin-1 in Chinese Han population with autism are limited. By direct sequencing, we analyzed the entire coding regions and associated splice junctions of neurexin-1 in 313 Chinese autism patients. For exons in which non-synonymous variants were identified, sequencing was performed in 500 healthy controls. We identified 22 variants in the neurexin-1 coding regions, including 7 missense variants, 3 deletions, and 12 synonymous mutations. Among them, 3 missense and 3 synonymous variants were not reported in the dbSNP database and absent in 500 control subjects; whereas 4 missense variants, 3 deletions and 3 synonymous mutations were not reported in the dbSNP database but were identified in the control subjects. However, there is no significant association of these mutations with autism risk. Interestingly, there was a statistically significant association of neurexin-1 SNP P300P (rs2303298) with risk of autism (26.2% vs. 13.8%; χ(2) = 22.487; p = 3.45E-006; OR = 2.152 (1.559-2.970)). Our data suggest a possible association of neurexin-1 with autism risk in Chinese Han population, warranting further large-scale study on this gene.
Collapse
Affiliation(s)
- Yalan Liu
- School of Biological Science and Technology, Central South University, Changsha, Hunan, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
195
|
Schmeisser MJ, Ey E, Wegener S, Bockmann J, Stempel AV, Kuebler A, Janssen AL, Udvardi PT, Shiban E, Spilker C, Balschun D, Skryabin BV, Dieck ST, Smalla KH, Montag D, Leblond CS, Faure P, Torquet N, Le Sourd AM, Toro R, Grabrucker AM, Shoichet SA, Schmitz D, Kreutz MR, Bourgeron T, Gundelfinger ED, Boeckers TM. Autistic-like behaviours and hyperactivity in mice lacking ProSAP1/Shank2. Nature 2012; 486:256-60. [PMID: 22699619 DOI: 10.1038/nature11015] [Citation(s) in RCA: 480] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2011] [Accepted: 03/08/2012] [Indexed: 01/22/2023]
Abstract
Autism spectrum disorders comprise a range of neurodevelopmental disorders characterized by deficits in social interaction and communication, and by repetitive behaviour. Mutations in synaptic proteins such as neuroligins, neurexins, GKAPs/SAPAPs and ProSAPs/Shanks were identified in patients with autism spectrum disorder, but the causative mechanisms remain largely unknown. ProSAPs/Shanks build large homo- and heteromeric protein complexes at excitatory synapses and organize the complex protein machinery of the postsynaptic density in a laminar fashion. Here we demonstrate that genetic deletion of ProSAP1/Shank2 results in an early, brain-region-specific upregulation of ionotropic glutamate receptors at the synapse and increased levels of ProSAP2/Shank3. Moreover, ProSAP1/Shank2(-/-) mutants exhibit fewer dendritic spines and show reduced basal synaptic transmission, a reduced frequency of miniature excitatory postsynaptic currents and enhanced N-methyl-d-aspartate receptor-mediated excitatory currents at the physiological level. Mutants are extremely hyperactive and display profound autistic-like behavioural alterations including repetitive grooming as well as abnormalities in vocal and social behaviours. By comparing the data on ProSAP1/Shank2(-/-) mutants with ProSAP2/Shank3αβ(-/-) mice, we show that different abnormalities in synaptic glutamate receptor expression can cause alterations in social interactions and communication. Accordingly, we propose that appropriate therapies for autism spectrum disorders are to be carefully matched to the underlying synaptopathic phenotype.
Collapse
|
196
|
Kelleher RJ, Geigenmüller U, Hovhannisyan H, Trautman E, Pinard R, Rathmell B, Carpenter R, Margulies D. High-throughput sequencing of mGluR signaling pathway genes reveals enrichment of rare variants in autism. PLoS One 2012; 7:e35003. [PMID: 22558107 PMCID: PMC3338748 DOI: 10.1371/journal.pone.0035003] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2011] [Accepted: 03/08/2012] [Indexed: 12/11/2022] Open
Abstract
Identification of common molecular pathways affected by genetic variation in autism is important for understanding disease pathogenesis and devising effective therapies. Here, we test the hypothesis that rare genetic variation in the metabotropic glutamate-receptor (mGluR) signaling pathway contributes to autism susceptibility. Single-nucleotide variants in genes encoding components of the mGluR signaling pathway were identified by high-throughput multiplex sequencing of pooled samples from 290 non-syndromic autism cases and 300 ethnically matched controls on two independent next-generation platforms. This analysis revealed significant enrichment of rare functional variants in the mGluR pathway in autism cases. Higher burdens of rare, potentially deleterious variants were identified in autism cases for three pathway genes previously implicated in syndromic autism spectrum disorder, TSC1, TSC2, and SHANK3, suggesting that genetic variation in these genes also contributes to risk for non-syndromic autism. In addition, our analysis identified HOMER1, which encodes a postsynaptic density-localized scaffolding protein that interacts with Shank3 to regulate mGluR activity, as a novel autism-risk gene. Rare, potentially deleterious HOMER1 variants identified uniquely in the autism population affected functionally important protein regions or regulatory sequences and co-segregated closely with autism among children of affected families. We also identified rare ASD-associated coding variants predicted to have damaging effects on components of the Ras/MAPK cascade. Collectively, these findings suggest that altered signaling downstream of mGluRs contributes to the pathogenesis of non-syndromic autism.
Collapse
Affiliation(s)
- Raymond J Kelleher
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, Massachusetts, United States of America.
| | | | | | | | | | | | | | | |
Collapse
|
197
|
Newschaffer CJ, Croen LA, Fallin MD, Hertz-Picciotto I, Nguyen DV, Lee NL, Berry CA, Farzadegan H, Hess HN, Landa RJ, Levy SE, Massolo ML, Meyerer SC, Mohammed SM, Oliver MC, Ozonoff S, Pandey J, Schroeder A, Shedd-Wise KM. Infant siblings and the investigation of autism risk factors. J Neurodev Disord 2012; 4:7. [PMID: 22958474 PMCID: PMC3436647 DOI: 10.1186/1866-1955-4-7] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2011] [Accepted: 04/18/2012] [Indexed: 12/31/2022] Open
Abstract
Infant sibling studies have been at the vanguard of autism spectrum disorders (ASD) research over the past decade, providing important new knowledge about the earliest emerging signs of ASD and expanding our understanding of the developmental course of this complex disorder. Studies focused on siblings of children with ASD also have unrealized potential for contributing to ASD etiologic research. Moving targeted time of enrollment back from infancy toward conception creates tremendous opportunities for optimally studying risk factors and risk biomarkers during the pre-, peri- and neonatal periods. By doing so, a traditional sibling study, which already incorporates close developmental follow-up of at-risk infants through the third year of life, is essentially reconfigured as an enriched-risk pregnancy cohort study. This review considers the enriched-risk pregnancy cohort approach of studying infant siblings in the context of current thinking on ASD etiologic mechanisms. It then discusses the key features of this approach and provides a description of the design and implementation strategy of one major ASD enriched-risk pregnancy cohort study: the Early Autism Risk Longitudinal Investigation (EARLI).
Collapse
Affiliation(s)
- Craig J Newschaffer
- Department of Epidemiology and Biostatistics, Drexel School of Public Health, 1505 Race Street, Mail Stop 1033, Philadelphia, PA 19102, USA
| | - Lisa A Croen
- Kaiser Permanente Division of Research, 2000 Broadway, Oakland, CA 94612, USA
| | - M Daniele Fallin
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, 615 N Wolfe Street, Baltimore, MD 21205, USA
| | - Irva Hertz-Picciotto
- Department of Public Health Sciences, University of California, Davis, CA 95616, USA
| | - Danh V Nguyen
- Department of Public Health Sciences, University of California, Davis, CA 95616, USA
| | - Nora L Lee
- Department of Epidemiology and Biostatistics, Drexel School of Public Health, 1505 Race Street, Mail Stop 1033, Philadelphia, PA 19102, USA
| | - Carmen A Berry
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, 615 N Wolfe Street, Baltimore, MD 21205, USA
| | - Homayoon Farzadegan
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, 615 N Wolfe Street, Baltimore, MD 21205, USA
| | - H Nicole Hess
- Kaiser Permanente San Jose Medical Center, 6620 Via Del Oro, San Jose, CA 95119, USA
| | - Rebecca J Landa
- Kennedy Krieger Institute, 3901 Greenspring Avenue, 2nd Floor, Baltimore, MD 21211, USA
| | - Susan E Levy
- Center for Autism Research, The Children's Hospital of Philadelphia, 3535 Market Street, Suite 860, Philadelphia, PA 19104, USA
| | - Maria L Massolo
- Kaiser Permanente Division of Research, 2000 Broadway, Oakland, CA 94612, USA
| | - Stacey C Meyerer
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, 615 N Wolfe Street, Baltimore, MD 21205, USA
| | - Sandra M Mohammed
- Department of Public Health Sciences, University of California, Davis, CA 95616, USA
| | - McKenzie C Oliver
- Department of Public Health Sciences, University of California, Davis, CA 95616, USA
| | - Sally Ozonoff
- The MIND Institute, UC Davis Medical Center, 2825 50th Street, Sacramento, CA 95817, USA
| | - Juhi Pandey
- Center for Autism Research, The Children's Hospital of Philadelphia, 3535 Market Street, Suite 860, Philadelphia, PA 19104, USA
| | - Adam Schroeder
- Department of Public Health Sciences, University of California, Davis, CA 95616, USA
| | - Kristine M Shedd-Wise
- Department of Public Health Sciences, University of California, Davis, CA 95616, USA
| |
Collapse
|
198
|
Devlin B, Scherer SW. Genetic architecture in autism spectrum disorder. Curr Opin Genet Dev 2012; 22:229-37. [PMID: 22463983 DOI: 10.1016/j.gde.2012.03.002] [Citation(s) in RCA: 338] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2012] [Revised: 03/02/2012] [Accepted: 03/05/2012] [Indexed: 01/08/2023]
Abstract
Autism spectrum disorder (ASD) is characterized by impairments in reciprocal social interaction and communication, and by restricted and repetitive behaviors. Family studies indicate a significant genetic basis for ASD susceptibility, and genomic scanning is beginning to elucidate the underlying genetic architecture. Some 5-15% of individuals with ASD have an identifiable genetic etiology corresponding to known chromosomal rearrangements or single gene disorders. Rare (<1% frequency) de novo or inherited copy number variations (CNVs) (especially those that affect genes with synaptic function) are observed in 5-10% of idiopathic ASD cases. These findings, coupled with genome sequencing data suggest the existence of hundreds of ASD risk genes. Common variants, yet unidentified, exert only small effects on risk. Identification of ASD risk genes with high penetrance will broaden the targets amenable to genetic testing; while the biological pathways revealed by the deeper list of ASD genes should narrow the targets for therapeutic intervention.
Collapse
Affiliation(s)
- Bernie Devlin
- Department of Psychiatry, University of Pittsburgh School of Medicine, 3811 O'Hara St., Pittsburgh, PA 15213, USA
| | | |
Collapse
|
199
|
Autism spectrum disorders: is mesenchymal stem cell personalized therapy the future? J Biomed Biotechnol 2012; 2012:480289. [PMID: 22496609 PMCID: PMC3303614 DOI: 10.1155/2012/480289] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2011] [Accepted: 09/29/2011] [Indexed: 12/16/2022] Open
Abstract
Autism and autism spectrum disorders (ASDs) are heterogeneous neurodevelopmental disorders. They are enigmatic conditions that have their origins in the interaction of genes and environmental factors. ASDs are characterized by dysfunctions in social interaction and communication skills, in addition to repetitive and stereotypic verbal and nonverbal behaviours. Immune dysfunction has been confirmed with autistic children. There are no defined mechanisms of pathogenesis or curative therapy presently available. Indeed, ASDs are still untreatable. Available treatments for autism can be divided into behavioural, nutritional, and medical approaches, although no defined standard approach exists. Nowadays, stem cell therapy represents the great promise for the future of molecular medicine. Among the stem cell population, mesenchymal stem cells (MSCs) show probably best potential good results in medical research. Due to the particular immune and neural dysregulation observed in ASDs, mesenchymal stem cell transplantation could offer a unique tool to provide better resolution for this disease.
Collapse
|
200
|
Leblond CS, Heinrich J, Delorme R, Proepper C, Betancur C, Huguet G, Konyukh M, Chaste P, Ey E, Rastam M, Anckarsäter H, Nygren G, Gillberg IC, Melke J, Toro R, Regnault B, Fauchereau F, Mercati O, Lemière N, Skuse D, Poot M, Holt R, Monaco AP, Järvelä I, Kantojärvi K, Vanhala R, Curran S, Collier DA, Bolton P, Chiocchetti A, Klauck SM, Poustka F, Freitag CM, Waltes R, Kopp M, Duketis E, Bacchelli E, Minopoli F, Ruta L, Battaglia A, Mazzone L, Maestrini E, Sequeira AF, Oliveira B, Vicente A, Oliveira G, Pinto D, Scherer SW, Zelenika D, Delepine M, Lathrop M, Bonneau D, Guinchat V, Devillard F, Assouline B, Mouren MC, Leboyer M, Gillberg C, Boeckers TM, Bourgeron T. Genetic and functional analyses of SHANK2 mutations suggest a multiple hit model of autism spectrum disorders. PLoS Genet 2012; 8:e1002521. [PMID: 22346768 PMCID: PMC3276563 DOI: 10.1371/journal.pgen.1002521] [Citation(s) in RCA: 308] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2011] [Accepted: 12/11/2011] [Indexed: 01/15/2023] Open
Abstract
Autism spectrum disorders (ASD) are a heterogeneous group of neurodevelopmental disorders with a complex inheritance pattern. While many rare variants in synaptic proteins have been identified in patients with ASD, little is known about their effects at the synapse and their interactions with other genetic variations. Here, following the discovery of two de novo SHANK2 deletions by the Autism Genome Project, we identified a novel 421 kb de novo SHANK2 deletion in a patient with autism. We then sequenced SHANK2 in 455 patients with ASD and 431 controls and integrated these results with those reported by Berkel et al. 2010 (n = 396 patients and n = 659 controls). We observed a significant enrichment of variants affecting conserved amino acids in 29 of 851 (3.4%) patients and in 16 of 1,090 (1.5%) controls (P = 0.004, OR = 2.37, 95% CI = 1.23-4.70). In neuronal cell cultures, the variants identified in patients were associated with a reduced synaptic density at dendrites compared to the variants only detected in controls (P = 0.0013). Interestingly, the three patients with de novo SHANK2 deletions also carried inherited CNVs at 15q11-q13 previously associated with neuropsychiatric disorders. In two cases, the nicotinic receptor CHRNA7 was duplicated and in one case the synaptic translation repressor CYFIP1 was deleted. These results strengthen the role of synaptic gene dysfunction in ASD but also highlight the presence of putative modifier genes, which is in keeping with the "multiple hit model" for ASD. A better knowledge of these genetic interactions will be necessary to understand the complex inheritance pattern of ASD.
Collapse
Affiliation(s)
- Claire S. Leblond
- Human Genetics and Cognitive Functions, Institut Pasteur, Paris, France
- CNRS URA 2182 “Genes, synapses and cognition,” Institut Pasteur, Paris, France
- University Denis Diderot Paris 7, Paris, France
| | - Jutta Heinrich
- Institute of Anatomy and Cell Biology, Ulm University, Ulm, Germany
| | - Richard Delorme
- Human Genetics and Cognitive Functions, Institut Pasteur, Paris, France
- CNRS URA 2182 “Genes, synapses and cognition,” Institut Pasteur, Paris, France
- Assistance Publique-Hôpitaux de Paris, Robert Debré Hospital, Department of Child and Adolescent Psychiatry, Paris, France
| | | | - Catalina Betancur
- INSERM, U952, Paris, France
- CNRS, UMR 7224, Paris, France
- UPMC Univ Paris 06, Paris, France
| | - Guillaume Huguet
- Human Genetics and Cognitive Functions, Institut Pasteur, Paris, France
- CNRS URA 2182 “Genes, synapses and cognition,” Institut Pasteur, Paris, France
- University Denis Diderot Paris 7, Paris, France
| | - Marina Konyukh
- Human Genetics and Cognitive Functions, Institut Pasteur, Paris, France
- CNRS URA 2182 “Genes, synapses and cognition,” Institut Pasteur, Paris, France
- University Denis Diderot Paris 7, Paris, France
| | - Pauline Chaste
- Human Genetics and Cognitive Functions, Institut Pasteur, Paris, France
- CNRS URA 2182 “Genes, synapses and cognition,” Institut Pasteur, Paris, France
- University Denis Diderot Paris 7, Paris, France
| | - Elodie Ey
- Human Genetics and Cognitive Functions, Institut Pasteur, Paris, France
- CNRS URA 2182 “Genes, synapses and cognition,” Institut Pasteur, Paris, France
- University Denis Diderot Paris 7, Paris, France
| | - Maria Rastam
- Department of Clinical Sciences in Lund, Lund University, Lund, Sweden
| | | | - Gudrun Nygren
- Gillberg Neuropsychiatry Centre, University of Gothenburg, Göteborg, Sweden
| | - I. Carina Gillberg
- Gillberg Neuropsychiatry Centre, University of Gothenburg, Göteborg, Sweden
| | - Jonas Melke
- Institute of Neuroscience and Physiology, Department of Pharmacology, Gothenburg University, Göteborg, Sweden
| | - Roberto Toro
- Human Genetics and Cognitive Functions, Institut Pasteur, Paris, France
- CNRS URA 2182 “Genes, synapses and cognition,” Institut Pasteur, Paris, France
- University Denis Diderot Paris 7, Paris, France
| | - Beatrice Regnault
- Eukaryote Genotyping Platform, Genopole, Institut Pasteur, Paris, France
| | - Fabien Fauchereau
- Human Genetics and Cognitive Functions, Institut Pasteur, Paris, France
- CNRS URA 2182 “Genes, synapses and cognition,” Institut Pasteur, Paris, France
- University Denis Diderot Paris 7, Paris, France
| | - Oriane Mercati
- Human Genetics and Cognitive Functions, Institut Pasteur, Paris, France
- CNRS URA 2182 “Genes, synapses and cognition,” Institut Pasteur, Paris, France
- University Denis Diderot Paris 7, Paris, France
| | - Nathalie Lemière
- Human Genetics and Cognitive Functions, Institut Pasteur, Paris, France
- CNRS URA 2182 “Genes, synapses and cognition,” Institut Pasteur, Paris, France
- University Denis Diderot Paris 7, Paris, France
| | - David Skuse
- Behavioural and Brain Sciences Unit, Institute of Child Health, University College London, London, United Kingdom
| | - Martin Poot
- Department of Medical Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Richard Holt
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Anthony P. Monaco
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Irma Järvelä
- Department of Medical Genetics, University of Helsinki, Helsinki, Finland
| | - Katri Kantojärvi
- Department of Medical Genetics, University of Helsinki, Helsinki, Finland
| | - Raija Vanhala
- Department of Medical Genetics, University of Helsinki, Helsinki, Finland
| | - Sarah Curran
- Academic Department of Child and Adolescent Psychiatry, Institute of Psychiatry, King's College London, London, United Kingdom
| | - David A. Collier
- Social Genetic Developmental Psychiatry Centre, Institute of Psychiatry, King's College London, London, United Kingdom
| | - Patrick Bolton
- Academic Department of Child and Adolescent Psychiatry, Institute of Psychiatry, King's College London, London, United Kingdom
- Social Genetic Developmental Psychiatry Centre, Institute of Psychiatry, King's College London, London, United Kingdom
| | - Andreas Chiocchetti
- Division of Molecular Genome Analysis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sabine M. Klauck
- Division of Molecular Genome Analysis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Fritz Poustka
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Goethe University, Frankfurt am Main, Germany
| | - Christine M. Freitag
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Goethe University, Frankfurt am Main, Germany
| | - Regina Waltes
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Goethe University, Frankfurt am Main, Germany
| | - Marnie Kopp
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Goethe University, Frankfurt am Main, Germany
| | - Eftichia Duketis
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Goethe University, Frankfurt am Main, Germany
| | - Elena Bacchelli
- Department of Biology, University of Bologna, Bologna, Italy
| | | | - Liliana Ruta
- Division of Child Neurology and Psychiatry, Department of Paediatrics, University of Catania, Catania, Italy
| | - Agatino Battaglia
- Stella Maris Clinical Research Institute for Child and Adolescent Neuropsychiatry, Pisa, Italy
| | - Luigi Mazzone
- Division of Child Neurology and Psychiatry, Department of Pediatrics, University of Catania, Catania, Italy
| | - Elena Maestrini
- Department of Biology, University of Bologna, Bologna, Italy
| | - Ana F. Sequeira
- Instituto Nacional de Saude Dr Ricardo Jorge, Lisbon, Portugal
- Instituto Gulbenkian de Ciencia, Oeiras, Portugal
- Center for Biodiversity, Functional and Integrative Genomics, Faculdade de Ciências da Universidade de Lisboa, Lisboa, Portugal
| | - Barbara Oliveira
- Instituto Nacional de Saude Dr Ricardo Jorge, Lisbon, Portugal
- Instituto Gulbenkian de Ciencia, Oeiras, Portugal
- Center for Biodiversity, Functional and Integrative Genomics, Faculdade de Ciências da Universidade de Lisboa, Lisboa, Portugal
| | - Astrid Vicente
- Instituto Nacional de Saude Dr Ricardo Jorge, Lisbon, Portugal
- Instituto Gulbenkian de Ciencia, Oeiras, Portugal
- Center for Biodiversity, Functional and Integrative Genomics, Faculdade de Ciências da Universidade de Lisboa, Lisboa, Portugal
| | - Guiomar Oliveira
- Unidade Neurodesenvolvimento e Autismo, Centro Investigação e Formação Clinica, Hospital Pediátrico Coimbra e Faculdade Medicina, Universidade Coimbra, Coimbra, Portugal
| | - Dalila Pinto
- The Centre for Applied Genomics and Program in Genetics and Genomic Biology, The Hospital for Sick Children, Toronto, Canada
| | - Stephen W. Scherer
- The Centre for Applied Genomics and Program in Genetics and Genomic Biology, The Hospital for Sick Children, Toronto, Canada
| | | | | | | | - Dominique Bonneau
- INSERM U771 and CNRS 6214, Angers, France
- Département de Biochimie et Génétique, Centre Hospitalier Universitaire, Angers, France
| | - Vincent Guinchat
- CADIPA–Centre de Ressources Autisme Rhône-Alpes, Saint Egrève, France
| | | | | | - Marie-Christine Mouren
- Assistance Publique-Hôpitaux de Paris, Robert Debré Hospital, Department of Child and Adolescent Psychiatry, Paris, France
| | - Marion Leboyer
- INSERM, U955, Psychiatrie Génétique, Créteil, France
- Université Paris Est, Faculté de Médecine, Créteil, France
- AP-HP, Hôpital H. Mondor–A. Chenevier, Département de Psychiatrie, Créteil, France
| | - Christopher Gillberg
- Gillberg Neuropsychiatry Centre, University of Gothenburg, Göteborg, Sweden
- Institute of Child Health, University College London, London, United Kingdom
| | | | - Thomas Bourgeron
- Human Genetics and Cognitive Functions, Institut Pasteur, Paris, France
- CNRS URA 2182 “Genes, synapses and cognition,” Institut Pasteur, Paris, France
- University Denis Diderot Paris 7, Paris, France
- * E-mail:
| |
Collapse
|