1
|
Lu X, Ni P, Suarez-Meade P, Ma Y, Forrest EN, Wang G, Wang Y, Quiñones-Hinojosa A, Gerstein M, Jiang YH. Transcriptional Determinism and Stochasticity Contribute to the Complexity of Autism Associated SHANK Family Genes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.18.585480. [PMID: 38562714 PMCID: PMC10983920 DOI: 10.1101/2024.03.18.585480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Precision of transcription is critical because transcriptional dysregulation is disease causing. Traditional methods of transcriptional profiling are inadequate to elucidate the full spectrum of the transcriptome, particularly for longer and less abundant mRNAs. SHANK3 is one of the most common autism causative genes. Twenty-four Shank3 mutant animal lines have been developed for autism modeling. However, their preclinical validity has been questioned due to incomplete Shank3 transcript structure. We applied an integrative approach combining cDNA-capture and long-read sequencing to profile the SHANK3 transcriptome in human and mice. We unexpectedly discovered an extremely complex SHANK3 transcriptome. Specific SHANK3 transcripts were altered in Shank3 mutant mice and postmortem brains tissues from individuals with ASD. The enhanced SHANK3 transcriptome significantly improved the detection rate for potential deleterious variants from genomics studies of neuropsychiatric disorders. Our findings suggest the stochastic transcription of genome associated with SHANK family genes.
Collapse
Affiliation(s)
- Xiaona Lu
- Department of Genetics, Yale University School of Medicine New Haven, CT, 06520 USA
| | - Pengyu Ni
- Biomedical Informatics & Data Science, Yale University School of Medicine New Haven, CT, 06520 USA
| | | | - Yu Ma
- Department of Neurology, Children’s Hospital of Fudan University, Shanghai, 201102 China
| | | | - Guilin Wang
- Yale Center for Genome Analysis, Yale University School of Medicine New Haven, CT, 06520 USA
| | - Yi Wang
- Department of Neurology, Children’s Hospital of Fudan University, Shanghai, 201102 China
| | | | - Mark Gerstein
- Biomedical Informatics & Data Science, Yale University School of Medicine New Haven, CT, 06520 USA
- Yale Center for Genome Analysis, Yale University School of Medicine New Haven, CT, 06520 USA
| | - Yong-hui Jiang
- Department of Genetics, Yale University School of Medicine New Haven, CT, 06520 USA
- Neuroscienc, Yale University School of Medicine New Haven, CT, 06520 USA
- Pediatrics, Yale University School of Medicine New Haven, CT, 06520 USA
| |
Collapse
|
2
|
Savage MC, Bliss G, Buxbaum JD, Farrell JS, Levin AR, Srivastava S, Berry-Kravis E, Holder JL, Sahin M. A roadmap for SHANK3-related Epilepsy Research: recommendations from the 2023 strategic planning workshop. THERAPEUTIC ADVANCES IN RARE DISEASE 2024; 5:26330040241273464. [PMID: 39295819 PMCID: PMC11409305 DOI: 10.1177/26330040241273464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 07/10/2024] [Indexed: 09/21/2024]
Abstract
On September 27, 2023, the CureSHANK nonprofit foundation sponsored a conference in Boston, Massachusetts, to identify gaps in knowledge surrounding SHANK3-related epilepsy with the goal of determining future research priorities and recommendations. In addition to patient families and members of the CureSHANK community, participants in the conference included a broad cross-section of preclinical and clinical researchers and clinicians with expertise in SHANK3-related epilepsy as well as representatives from the pharmaceutical industry. Here we summarize the outcomes from comprehensive premeeting deliberations and the final conference recommendations, including (1) gaps in knowledge related to clinical science, (2) gaps in knowledge related to preclinical science, and (3) research priorities moving forward.
Collapse
Affiliation(s)
| | | | - Joseph D Buxbaum
- Seaver Autism Center for Research and Treatment, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jordan S Farrell
- Department of Neurology, Harvard Medical School, Boston, MA, USA
- Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Boston, MA, USA
- Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
| | - April R Levin
- Department of Neurology, Harvard Medical School, Boston, MA, USA
| | | | - Elizabeth Berry-Kravis
- Departments of Pediatrics, Neurological Sciences, Anatomy and Cell Biology, Rush Medical Center, Chicago, IL, USA
| | - J Lloyd Holder
- Department of Pediatrics, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, USA
| | - Mustafa Sahin
- Department of Neurology, Harvard Medical School, Boston, MA, USA
- Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Boston, MA, USA
- Kirby Neurobiology Center, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| |
Collapse
|
3
|
Ryndych D, Sebold A, Strassburg A, Li Y, Ramos RL, Otazu GH. Haploinsufficiency of Shank3 in Mice Selectively Impairs Target Odor Recognition in Novel Background Odors. J Neurosci 2023; 43:7799-7811. [PMID: 37739796 PMCID: PMC10648539 DOI: 10.1523/jneurosci.0255-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 07/30/2023] [Accepted: 09/13/2023] [Indexed: 09/24/2023] Open
Abstract
Individuals with mutations in a single copy of the SHANK3 gene present with social interaction deficits. Although social behavior in mice depends on olfaction, mice with mutations in a single copy of the Shank3 gene do not have olfactory deficits in simple odor identification tasks (Drapeau et al., 2018). Here, we tested olfaction in mice with mutations in a single copy of the Shank3 gene (Peça et al., 2011) using a complex odor task and imaging in awake mice. Average glomerular responses in the olfactory bulb of Shank3B +/- were correlated with WT mice. However, there was increased trial-to-trial variability in the odor responses for Shank3B +/- mice. Simulations demonstrated that this increased variability could affect odor detection in novel environments. To test whether performance was affected by the increased variability, we tested target odor recognition in the presence of novel background odors using a recently developed task (Li et al., 2023). Head-fixed mice were trained to detect target odors in the presence of known background odors. Performance was tested using catch trials where the known background odors were replaced by novel background odors. We compared the performance of eight Shank3B +/- mice (five males, three females) on this task with six WT mice (three males, three females). Performance for known background odors and learning rates were similar between Shank3B +/- and WT mice. However, when tested with novel background odors, the performance of Shank3B +/- mice dropped to almost chance levels. Thus, haploinsufficiency of the Shank3 gene causes a specific deficit in odor detection in novel environments. Our results are discussed in the context of other Shank3 mouse models and have implications for understanding olfactory function in neurodevelopmental disorders.SIGNIFICANCE STATEMENT People and mice with mutations in a single copy in the synaptic gene Shank3 show features seen in autism spectrum disorders, including social interaction deficits. Although mice social behavior uses olfaction, mice with mutations in a single copy of Shank3 have so far not shown olfactory deficits when tested using simple tasks. Here, we used a recently developed task to show that these mice could identify odors in the presence of known background odors as well as wild-type mice. However, their performance fell below that of wild-type mice when challenged with novel background odors. This deficit was also previously reported in the Cntnap2 mouse model of autism, suggesting that odor detection in novel backgrounds is a general deficit across mouse models of autism.
Collapse
Affiliation(s)
- Darya Ryndych
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, New York 11568
| | - Alison Sebold
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, New York 11568
| | - Alyssa Strassburg
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, New York 11568
| | - Yan Li
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, New York 11568
| | - Raddy L Ramos
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, New York 11568
| | - Gonzalo H Otazu
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, New York 11568
| |
Collapse
|
4
|
Landry O, François A, Oye Mintsa Mi-Mba MF, Traversy MT, Tremblay C, Emond V, Bennett DA, Gylys KH, Buxbaum JD, Calon F. Postsynaptic Protein Shank3a Deficiency Synergizes with Alzheimer's Disease Neuropathology to Impair Cognitive Performance in the 3xTg-AD Murine Model. J Neurosci 2023; 43:4941-4954. [PMID: 37253603 PMCID: PMC10312061 DOI: 10.1523/jneurosci.1945-22.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 04/17/2023] [Accepted: 05/04/2023] [Indexed: 06/01/2023] Open
Abstract
Synaptic loss is intrinsically linked to Alzheimer's disease (AD) neuropathology and symptoms, but its direct impact on clinical symptoms remains elusive. The postsynaptic protein Shank3 (SH3 and multiple ankyrin repeat domains) is of particular interest, as the loss of a single allele of the SHANK3 gene is sufficient to cause profound cognitive symptoms in children. We thus sought to determine whether a SHANK3 deficiency could contribute to the emergence or worsening of AD symptoms and neuropathology. We first found a 30%-50% postmortem loss of SHANK3a associated with cognitive decline in the parietal cortex of individuals with AD. To further probe the role of SHANK3 in AD, we crossed male and female 3xTg-AD mice modelling Aβ and tau pathologies with Shank3a-deficient mice (Shank3Δex4-9). We observed synergistic deleterious effects of Shank3a deficiency and AD neuropathology on object recognition memory at 9, 12, and 18 months of age and on anxious behavior at 9 and 12 months of age in hemizygous Shank3Δex4-9-3xTg-AD mice. In addition to the expected 50% loss of Shank3a, levels of other synaptic proteins, such as PSD-95, drebrin, and homer1, remained unchanged in the parietotemporal cortex of hemizygous Shank3Δex4-9 animals. However, Shank3a deficiency increased the levels of soluble Aβ42 and human tau at 18 months of age compared with 3xTg-AD mice with normal Shank3 expression. The results of this study in human brain samples and in transgenic mice are consistent with the hypothesis that Shank3 deficiency makes a key contribution to cognitive impairment in AD.SIGNIFICANCE STATEMENT Although the loss of several synaptic proteins has been described in Alzheimer's disease (AD), it remains unclear whether their reduction contributes to clinical symptoms. The results of this study in human samples show lower levels of SHANK3a in AD brain, correlating with cognitive decline. Data gathered in a novel transgenic mouse suggest that Shank3a deficiency synergizes with AD neuropathology to induce cognitive impairment, consistent with a causal role in AD. Therefore, treatment aiming at preserving Shank3 in the aging brain may be beneficial to prevent AD.
Collapse
Affiliation(s)
- Olivier Landry
- Faculté de pharmacie, Université Laval, Quebec G1V 0A6, Quebec, Canada
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Quebec G1V 4G2, Quebec, Canada
| | - Arnaud François
- Faculté de pharmacie, Université Laval, Quebec G1V 0A6, Quebec, Canada
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Quebec G1V 4G2, Quebec, Canada
| | - Méryl-Farelle Oye Mintsa Mi-Mba
- Faculté de pharmacie, Université Laval, Quebec G1V 0A6, Quebec, Canada
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Quebec G1V 4G2, Quebec, Canada
| | - Marie-Therese Traversy
- Faculté de pharmacie, Université Laval, Quebec G1V 0A6, Quebec, Canada
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Quebec G1V 4G2, Quebec, Canada
| | - Cyntia Tremblay
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Quebec G1V 4G2, Quebec, Canada
| | - Vincent Emond
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Quebec G1V 4G2, Quebec, Canada
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois 60612
| | - Karen H Gylys
- School of Nursing, University of California, Los Angeles, California 90095
| | - Joseph D Buxbaum
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York 10029, New York
| | - Frédéric Calon
- Faculté de pharmacie, Université Laval, Quebec G1V 0A6, Quebec, Canada
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Quebec G1V 4G2, Quebec, Canada
| |
Collapse
|
5
|
Riemersma IW, Havekes R, Kas MJH. Spatial and Temporal Gene Function Studies in Rodents: Towards Gene-Based Therapies for Autism Spectrum Disorder. Genes (Basel) 2021; 13:28. [PMID: 35052369 PMCID: PMC8774890 DOI: 10.3390/genes13010028] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/06/2021] [Accepted: 12/20/2021] [Indexed: 12/26/2022] Open
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental condition that is characterized by differences in social interaction, repetitive behaviors, restricted interests, and sensory differences beginning early in life. Especially sensory symptoms are highly correlated with the severity of other behavioral differences. ASD is a highly heterogeneous condition on multiple levels, including clinical presentation, genetics, and developmental trajectories. Over a thousand genes have been implicated in ASD. This has facilitated the generation of more than two hundred genetic mouse models that are contributing to understanding the biological underpinnings of ASD. Since the first symptoms already arise during early life, it is especially important to identify both spatial and temporal gene functions in relation to the ASD phenotype. To further decompose the heterogeneity, ASD-related genes can be divided into different subgroups based on common functions, such as genes involved in synaptic function. Furthermore, finding common biological processes that are modulated by this subgroup of genes is essential for possible patient stratification and the development of personalized early treatments. Here, we review the current knowledge on behavioral rodent models of synaptic dysfunction by focusing on behavioral phenotypes, spatial and temporal gene function, and molecular targets that could lead to new targeted gene-based therapy.
Collapse
Affiliation(s)
| | | | - Martien J. H. Kas
- Groningen Institute for Evolutionary Life Sciences, Neurobiology, University of Groningen, Nijenborgh 7, 9747 AG Groningen, The Netherlands; (I.W.R.); (R.H.)
| |
Collapse
|
6
|
Moutin E, Sakkaki S, Compan V, Bouquier N, Giona F, Areias J, Goyet E, Hemonnot-Girard AL, Seube V, Glasson B, Benac N, Chastagnier Y, Raynaud F, Audinat E, Groc L, Maurice T, Sala C, Verpelli C, Perroy J. Restoring glutamate receptosome dynamics at synapses rescues autism-like deficits in Shank3-deficient mice. Mol Psychiatry 2021; 26:7596-7609. [PMID: 34331007 DOI: 10.1038/s41380-021-01230-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 07/02/2021] [Accepted: 07/06/2021] [Indexed: 11/09/2022]
Abstract
Shank3 monogenic mutations lead to autism spectrum disorders (ASD). Shank3 is part of the glutamate receptosome that physically links ionotropic NMDA receptors to metabotropic mGlu5 receptors through interactions with scaffolding proteins PSD95-GKAP-Shank3-Homer. A main physiological function of the glutamate receptosome is to control NMDA synaptic function that is required for plasticity induction. Intact glutamate receptosome supports glutamate receptors activation and plasticity induction, while glutamate receptosome disruption blocks receptors activity, preventing the induction of subsequent plasticity. Despite possible impact on metaplasticity and cognitive behaviors, scaffold interaction dynamics and their consequences are poorly defined. Here, we used mGlu5-Homer interaction as a biosensor of glutamate receptosome integrity to report changes in synapse availability for plasticity induction. Combining BRET imaging and electrophysiology, we show that a transient neuronal depolarization inducing NMDA-dependent plasticity disrupts glutamate receptosome in a long-lasting manner at synapses and activates signaling pathways required for the expression of the initiated neuronal plasticity, such as ERK and mTOR pathways. Glutamate receptosome disruption also decreases the NMDA/AMPA ratio, freezing the sensitivity of the synapse to subsequent changes of neuronal activity. These data show the importance of a fine-tuning of protein-protein interactions within glutamate receptosome, driven by changes of neuronal activity, to control plasticity. In a mouse model of ASD, a truncated mutant form of Shank3 prevents the integrity of the glutamate receptosome. These mice display altered plasticity, anxiety-like, and stereotyped behaviors. Interestingly, repairing the integrity of glutamate receptosome and its sensitivity to the neuronal activity rescued synaptic transmission, plasticity, and some behavioral traits of Shank3∆C mice. Altogether, our findings characterize mechanisms by which Shank3 mutations cause ASD and highlight scaffold dynamics as new therapeutic target.
Collapse
Affiliation(s)
- Enora Moutin
- IGF, University of Montpellier, CNRS, INSERM, Montpellier, France.
| | - Sophie Sakkaki
- IGF, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Vincent Compan
- IGF, University of Montpellier, CNRS, INSERM, Montpellier, France
| | | | | | - Julie Areias
- IGF, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Elise Goyet
- Interdisciplinary Institute for NeuroScience, CNRS, UMR 5297, Centre Broca Nouvelle-Aquitaine, 33076, Bordeaux, France
| | | | - Vincent Seube
- IGF, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Bastien Glasson
- IGF, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Nathan Benac
- Interdisciplinary Institute for NeuroScience, CNRS, UMR 5297, Centre Broca Nouvelle-Aquitaine, 33076, Bordeaux, France
| | - Yan Chastagnier
- IGF, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Fabrice Raynaud
- IGF, University of Montpellier, CNRS, INSERM, Montpellier, France.,PhyMedExp, Univ Montpellier, INSERM, CNRS, CHU de Montpellier, France
| | - Etienne Audinat
- IGF, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Laurent Groc
- Interdisciplinary Institute for NeuroScience, CNRS, UMR 5297, Centre Broca Nouvelle-Aquitaine, 33076, Bordeaux, France
| | - Tangui Maurice
- MMDN, Univ Montpellier, EPHE, INSERM, Montpellier, France
| | - Carlo Sala
- Cnr Neuroscience Institute, 3220129, Milan, Italy
| | | | - Julie Perroy
- IGF, University of Montpellier, CNRS, INSERM, Montpellier, France.
| |
Collapse
|
7
|
Delling JP, Boeckers TM. Comparison of SHANK3 deficiency in animal models: phenotypes, treatment strategies, and translational implications. J Neurodev Disord 2021; 13:55. [PMID: 34784886 PMCID: PMC8594088 DOI: 10.1186/s11689-021-09397-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 09/27/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Autism spectrum disorder (ASD) is a neurodevelopmental condition, which is characterized by clinical heterogeneity and high heritability. Core symptoms of ASD include deficits in social communication and interaction, as well as restricted, repetitive patterns of behavior, interests, or activities. Many genes have been identified that are associated with an increased risk for ASD. Proteins encoded by these ASD risk genes are often involved in processes related to fetal brain development, chromatin modification and regulation of gene expression in general, as well as the structural and functional integrity of synapses. Genes of the SH3 and multiple ankyrin repeat domains (SHANK) family encode crucial scaffolding proteins (SHANK1-3) of excitatory synapses and other macromolecular complexes. SHANK gene mutations are highly associated with ASD and more specifically the Phelan-McDermid syndrome (PMDS), which is caused by heterozygous 22q13.3-deletion resulting in SHANK3-haploinsufficiency, or by SHANK3 missense variants. SHANK3 deficiency and potential treatment options have been extensively studied in animal models, especially in mice, but also in rats and non-human primates. However, few of the proposed therapeutic strategies have translated into clinical practice yet. MAIN TEXT This review summarizes the literature concerning SHANK3-deficient animal models. In particular, the structural, behavioral, and neurological abnormalities are described and compared, providing a broad and comprehensive overview. Additionally, the underlying pathophysiologies and possible treatments that have been investigated in these models are discussed and evaluated with respect to their effect on ASD- or PMDS-associated phenotypes. CONCLUSIONS Animal models of SHANK3 deficiency generated by various genetic strategies, which determine the composition of the residual SHANK3-isoforms and affected cell types, show phenotypes resembling ASD and PMDS. The phenotypic heterogeneity across multiple models and studies resembles the variation of clinical severity in human ASD and PMDS patients. Multiple therapeutic strategies have been proposed and tested in animal models, which might lead to translational implications for human patients with ASD and/or PMDS. Future studies should explore the effects of new therapeutic approaches that target genetic haploinsufficiency, like CRISPR-mediated activation of promotors.
Collapse
Affiliation(s)
- Jan Philipp Delling
- Institute for Anatomy and Cell Biology, Ulm University, Albert-Einstein-Allee 11, Ulm, 89081, Germany.
| | - Tobias M Boeckers
- Institute for Anatomy and Cell Biology, Ulm University, Albert-Einstein-Allee 11, Ulm, 89081, Germany. .,Ulm Site, DZNE, Ulm, Germany.
| |
Collapse
|
8
|
de Chaumont F, Lemière N, Coqueran S, Bourgeron T, Ey E. LMT USV Toolbox, a Novel Methodological Approach to Place Mouse Ultrasonic Vocalizations in Their Behavioral Contexts-A Study in Female and Male C57BL/6J Mice and in Shank3 Mutant Females. Front Behav Neurosci 2021; 15:735920. [PMID: 34720899 PMCID: PMC8548730 DOI: 10.3389/fnbeh.2021.735920] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Accepted: 09/20/2021] [Indexed: 11/13/2022] Open
Abstract
Ultrasonic vocalizations (USVs) are used as a phenotypic marker in mouse models of neuropsychiatric disorders. Nevertheless, current methodologies still require time-consuming manual input or sound recordings clean of any background noise. We developed a method to overcome these two restraints to boost knowledge on mouse USVs. The methods are freely available and the USV analysis runs online at https://usv.pasteur.cloud. As little is currently known about usage and structure of ultrasonic vocalizations during social interactions over the long-term and in unconstrained context, we investigated mouse spontaneous communication by coupling the analysis of USVs with automatic labeling of behaviors. We continuously recorded during 3 days undisturbed interactions of same-sex pairs of C57BL/6J sexually naive males and females at 5 weeks and 3 and 7 months of age. In same-sex interactions, we observed robust differences between males and females in the amount of USVs produced, in the acoustic structure and in the contexts of emission. The context-specific acoustic variations emerged with increasing age. The emission of USVs also reflected a high level of excitement during social interactions. We finally highlighted the importance of studying long-term spontaneous communication by investigating female mice lacking Shank3, a synaptic protein associated with autism. While the previous short-time constrained investigations could not detect USV emission abnormalities, our analysis revealed robust differences in the usage and structure of the USVs emitted by mutant mice compared to wild-type female pairs.
Collapse
Affiliation(s)
- Fabrice de Chaumont
- Human Genetics and Cognitive Functions, Institut Pasteur, UMR 3571 CNRS, Université de Paris, Paris, France
| | - Nathalie Lemière
- Human Genetics and Cognitive Functions, Institut Pasteur, UMR 3571 CNRS, Université de Paris, Paris, France
| | - Sabrina Coqueran
- Human Genetics and Cognitive Functions, Institut Pasteur, UMR 3571 CNRS, Université de Paris, Paris, France
| | - Thomas Bourgeron
- Human Genetics and Cognitive Functions, Institut Pasteur, UMR 3571 CNRS, Université de Paris, Paris, France
| | - Elodie Ey
- Human Genetics and Cognitive Functions, Institut Pasteur, UMR 3571 CNRS, Université de Paris, Paris, France
| |
Collapse
|
9
|
Gandhi T, Lee CC. Neural Mechanisms Underlying Repetitive Behaviors in Rodent Models of Autism Spectrum Disorders. Front Cell Neurosci 2021; 14:592710. [PMID: 33519379 PMCID: PMC7840495 DOI: 10.3389/fncel.2020.592710] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 12/09/2020] [Indexed: 12/15/2022] Open
Abstract
Autism spectrum disorder (ASD) is comprised of several conditions characterized by alterations in social interaction, communication, and repetitive behaviors. Genetic and environmental factors contribute to the heterogeneous development of ASD behaviors. Several rodent models display ASD-like phenotypes, including repetitive behaviors. In this review article, we discuss the potential neural mechanisms involved in repetitive behaviors in rodent models of ASD and related neuropsychiatric disorders. We review signaling pathways, neural circuits, and anatomical alterations in rodent models that display robust stereotypic behaviors. Understanding the mechanisms and circuit alterations underlying repetitive behaviors in rodent models of ASD will inform translational research and provide useful insight into therapeutic strategies for the treatment of repetitive behaviors in ASD and other neuropsychiatric disorders.
Collapse
Affiliation(s)
- Tanya Gandhi
- Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA, United States
| | | |
Collapse
|
10
|
Rein B, Ma K, Yan Z. A standardized social preference protocol for measuring social deficits in mouse models of autism. Nat Protoc 2020; 15:3464-3477. [PMID: 32895524 DOI: 10.1038/s41596-020-0382-9] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 07/07/2020] [Indexed: 01/13/2023]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by social communication deficits and other behavioral abnormalities. The three-chamber social preference test is often used to assess social deficits in mouse models of ASD. However, varying and often contradicting phenotypic descriptions of ASD mouse models can be found in the scientific literature, and the substantial variability in the methods used by researchers to assess social deficits in mice could be a contributing factor. Here we describe a standardized three-chamber social preference protocol, which is sensitive and reliable at detecting social preference deficits in several mouse models of ASD. This protocol comprises three phases that can all be completed within 1 d. The test mouse is first habituated to the apparatus containing two empty cups in the side chambers, followed by the pre-test phase in which the mouse can interact with two identical inanimate objects placed in the cups. During the test phase, the mouse is allowed to interact with a social stimulus (an unfamiliar wild-type (WT) mouse) contained in one cup and a novel non-social stimulus contained in the other cup. The protocol is thus designed to assess preference between social and non-social stimuli under conditions of equal salience. The broad implementation of the three-chamber social preference protocol presented here should improve the accuracy and consistency of assessments for social preference deficits associated with ASD and other psychiatric disorders.
Collapse
Affiliation(s)
- Benjamin Rein
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Kaijie Ma
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Zhen Yan
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA.
| |
Collapse
|
11
|
Vitrac A, Pons S, Balkota M, Lemière N, Raïs C, Bourgeois JP, Maskos U, Bourgeron T, Cloëz-Tayarani I. A chimeric mouse model to study human iPSC-derived neurons: the case of a truncating SHANK3 mutation. Sci Rep 2020; 10:13315. [PMID: 32769989 PMCID: PMC7414912 DOI: 10.1038/s41598-020-70056-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 07/17/2020] [Indexed: 11/09/2022] Open
Abstract
Using human induced pluripotent stem cells (iPSC), recent studies have shown that the events underlying autism spectrum disorders (ASD) can occur during neonatal development. We previously analyzed the iPSC-derived pyramidal cortical neurons of a subset of patients with ASD carrying de novo heterozygous mutations in postsynaptic SHANK3 protein, in culture. We reported altered spinogenesis of those neurons. The transplantation of human iPSC-derived neuronal precursors into mouse brain represents a novel option for in vivo analysis of mutations affecting the human brain. In this study, we transplanted the neuronal precursor cells (NPC) into the cortex of newborn mice to analyze their integration and maturation at early stages of development and studied axonal projections of transplanted human neurons into adult mouse brain. We then co-transplanted NPC from a control individual and from a patient carrying a de novo heterozygous SHANK3 mutation. We observed a reduction in cell soma size of selective neuronal categories and in axonal projections at 30 days post-transplantation. In contrast to previous in vitro studies, we did not observe any alteration in spinogenesis at this early age. The humanized chimeric mouse models offer the means to analyze ASD-associated mutations further and provide the opportunity to visualize phenotypes in vivo.
Collapse
Affiliation(s)
- Aline Vitrac
- Human Genetics and Cognitive Functions, CNRS UMR 3571 « Genes, Synapses and Cognition », Université de Paris, Institut Pasteur, Paris, France
| | - Stéphanie Pons
- Integrative Neurobiology of Cholinergic Systems, CNRS UMR 3571 « Genes, Synapses and Cognition », Institut Pasteur, Paris, France
| | - Marta Balkota
- Integrative Neurobiology of Cholinergic Systems, CNRS UMR 3571 « Genes, Synapses and Cognition », Institut Pasteur, Paris, France
| | - Nathalie Lemière
- Human Genetics and Cognitive Functions, CNRS UMR 3571 « Genes, Synapses and Cognition », Université de Paris, Institut Pasteur, Paris, France
| | - Célia Raïs
- Integrative Neurobiology of Cholinergic Systems, CNRS UMR 3571 « Genes, Synapses and Cognition », Institut Pasteur, Paris, France.,Collège Doctoral, Sorbonne Université, Paris, France
| | - Jean-Pierre Bourgeois
- Human Genetics and Cognitive Functions, CNRS UMR 3571 « Genes, Synapses and Cognition », Université de Paris, Institut Pasteur, Paris, France
| | - Uwe Maskos
- Integrative Neurobiology of Cholinergic Systems, CNRS UMR 3571 « Genes, Synapses and Cognition », Institut Pasteur, Paris, France
| | - Thomas Bourgeron
- Human Genetics and Cognitive Functions, CNRS UMR 3571 « Genes, Synapses and Cognition », Université de Paris, Institut Pasteur, Paris, France
| | - Isabelle Cloëz-Tayarani
- Integrative Neurobiology of Cholinergic Systems, CNRS UMR 3571 « Genes, Synapses and Cognition », Institut Pasteur, Paris, France.
| |
Collapse
|
12
|
Dunn JT, Mroczek J, Patel HR, Ragozzino ME. Tandospirone, a Partial 5-HT1A Receptor Agonist, Administered Systemically or Into Anterior Cingulate Attenuates Repetitive Behaviors in Shank3B Mice. Int J Neuropsychopharmacol 2020; 23:533-542. [PMID: 32619232 PMCID: PMC7689206 DOI: 10.1093/ijnp/pyaa047] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 04/29/2020] [Accepted: 06/30/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Several cases of autism spectrum disorder have been linked to mutations in the SHANK3 gene. Haploinsufficiency of the SHANK3 gene contributes to Phelan-McDermid syndrome, which often presents an autism spectrum disorder phenotype along with moderate to severe intellectual disability. A SHANK3 gene deletion in mice results in elevated excitation of cortical pyramidal neurons that alters signaling to other brain areas. Serotonin 1A receptors are highly expressed on layer 2 cortical neurons and are known to have inhibitory actions. Serotonin 1A receptor agonist treatment in autistic cases with SHANK3 mutations and possibly other cases may restore excitatory and inhibitory balance that attenuates core symptoms. METHODS A series of experiments investigated the effects of acute tandospirone treatment on spatial learning and self-grooming, subchronic treatment of tandospirone on self-grooming behavior, and the effect of tandospirone infusion into the anterior cingulate on self-grooming behavior. RESULTS Only male Shank3B+/- mice exhibited a spatial learning deficit and elevated self-grooming. Acute i.p. injection of tandospirone, 0.01 and 0.06 mg/kg in male Shank3B+/- mice, attenuated a spatial acquisition deficit by improving sensitivity to positive reinforcement and reduced elevated self-grooming behavior. Repeated tandospirone (0.06 mg/kg) treatment attenuated elevated self-grooming behavior in male Shank3B+/- mice. Tandospirone injected into the anterior cingulate/premotor area reduced self-grooming behavior in male Shank3B+/- mice. CONCLUSIONS These results suggest that stimulation of cortical serotonin 1A receptors may reduce repetitive behaviors and cognitive impairments as observed in autism spectrum disorder, possibly by attenuating an excitation/inhibition imbalance. Further, tandospirone may serve as a treatment in autism spectrum disorder and other disorders associated with SHANK3 mutations.
Collapse
Affiliation(s)
- Jeffrey T Dunn
- Department of Psychology, University of Illinois at Chicago, Chicago, Illinois
| | - Jessica Mroczek
- Department of Psychology, University of Illinois at Chicago, Chicago, Illinois
| | - Harsh R Patel
- Department of Psychology, University of Illinois at Chicago, Chicago, Illinois
| | - Michael E Ragozzino
- Department of Psychology, University of Illinois at Chicago, Chicago, Illinois,Correspondence: Michael E. Ragozzino, PhD, Department of Psychology, 1007 West Harrison Street, Chicago, IL 60607 ()
| |
Collapse
|
13
|
Takumi T, Tamada K, Hatanaka F, Nakai N, Bolton PF. Behavioral neuroscience of autism. Neurosci Biobehav Rev 2019; 110:60-76. [PMID: 31059731 DOI: 10.1016/j.neubiorev.2019.04.012] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 04/03/2019] [Accepted: 04/22/2019] [Indexed: 12/29/2022]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder. Several genetic causes of ASD have been identified and this has enabled researchers to construct mouse models. Mouse behavioral tests reveal impaired social interaction and communication, as well as increased repetitive behavior and behavioral inflexibility in these mice, which correspond to core behavioral deficits observed in individuals with ASD. However, the connection between these behavioral abnormalities and the underlying dysregulation in neuronal circuits and synaptic function is poorly understood. Moreover, different components of the ASD phenotype may be linked to dysfunction in different brain regions, making it even more challenging to chart the pathophysiological mechanisms involved in ASD. Here we summarize the research on mouse models of ASD and their contribution to understanding pathophysiological mechanisms. Specifically, we emphasize abnormal serotonin production and regulation, as well as the disruption in circadian rhythms and sleep that are observed in a subset of ASD, and propose that spatiotemporal disturbances in brainstem development may be a primary cause of ASD that propagates towards the cerebral cortex.
Collapse
Affiliation(s)
- Toru Takumi
- RIKEN Brain Science Institute, Wako, Saitama 351-0198, Japan.
| | - Kota Tamada
- RIKEN Brain Science Institute, Wako, Saitama 351-0198, Japan
| | | | - Nobuhiro Nakai
- RIKEN Brain Science Institute, Wako, Saitama 351-0198, Japan
| | - Patrick F Bolton
- Institute of Psychiatry, King's College London, London, SE5 8AF, UK
| |
Collapse
|
14
|
Angelakos CC, Tudor JC, Ferri SL, Jongens TA, Abel T. Home-cage hypoactivity in mouse genetic models of autism spectrum disorder. Neurobiol Learn Mem 2019; 165:107000. [PMID: 30797034 DOI: 10.1016/j.nlm.2019.02.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 11/28/2018] [Accepted: 02/19/2019] [Indexed: 11/16/2022]
Abstract
Genome-wide association and whole exome sequencing studies from Autism Spectrum Disorder (ASD) patient populations have implicated numerous risk factor genes whose mutation or deletion results in significantly increased incidence of ASD. Behavioral studies of monogenic mutant mouse models of ASD-associated genes have been useful for identifying aberrant neural circuitry. However, behavioral results often differ from lab to lab, and studies incorporating both males and females are often not performed despite the significant sex-bias of ASD. In this study, we sought to investigate the simple, passive behavior of home-cage activity monitoring across multiple 24-h days in four different monogenic mouse models of ASD: Shank3b-/-, Cntnap2-/-, Pcdh10+/-, and Fmr1 knockout mice. Relative to sex-matched wildtype (WT) littermates, we discovered significant home-cage hypoactivity, particularly in the dark (active) phase of the light/dark cycle, in male mice of all four ASD-associated transgenic models. For Cntnap2-/- and Pcdh10+/- mice, these activity alterations were sex-specific, as female mice did not exhibit home-cage activity differences relative to sex-matched WT controls. These home-cage hypoactivity alterations differ from activity findings previously reported using short-term activity measurements in a novel open field. Despite circadian problems reported in human ASD patients, none of the mouse models studied had alterations in free-running circadian period. Together, these findings highlight a shared phenotype across several monogenic mouse models of ASD, outline the importance of methodology on behavioral interpretation, and in some genetic lines parallel the male-enhanced phenotypic presentation observed in human ASDs.
Collapse
Affiliation(s)
- Christopher C Angelakos
- Neuroscience Graduate Group, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Jennifer C Tudor
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, United States; Department of Biology, Saint Joseph's University, Philadelphia, PA 19131, United States
| | - Sarah L Ferri
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, United States; Molecular Physiology and Biophysics, Iowa Neuroscience Institute, University of Iowa, Iowa City, IA 52242, United States
| | - Thomas A Jongens
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Ted Abel
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, United States; Molecular Physiology and Biophysics, Iowa Neuroscience Institute, University of Iowa, Iowa City, IA 52242, United States.
| |
Collapse
|
15
|
Transcriptome analysis of Shank3-overexpressing mice reveals unique molecular changes in the hypothalamus. Mol Brain 2018; 11:71. [PMID: 30482234 PMCID: PMC6257967 DOI: 10.1186/s13041-018-0413-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 11/08/2018] [Indexed: 11/20/2022] Open
Abstract
Various mutations in the SH3 and multiple ankyrin repeat domains 3 (SHANK3) gene are associated with neurodevelopmental and neuropsychiatric disorders. Thus far, synaptic abnormalities in multiple brain regions, including the hippocampus, prefrontal cortex, striatum, and ventral tegmental area, have been investigated in several lines of Shank3 mutant mice. However, although some reports have shown loss and gain of body weight in Shank3 knock-out and overexpressing transgenic (TG) mice, respectively, the potential functions of Shank3 in the hypothalamus, a brain region critically involved in energy intake and expenditure, are unknown. Hence, we first characterized endogenous Shank3 mRNA and protein expression in the hypothalamus of adult wild-type mice. Thereafter, we performed transcriptome analysis (RNA-sequencing) in the hypothalamus of adult Shank3 TG mice which mildly overexpress Shank3 proteins. By comparing the 174 differentially expressed genes in the hypothalamus with those previously reported in the striatum and medial prefrontal cortex (mPFC) of Shank3 TG mice, we found that 159 were hypothalamus-specific while only 15 were also observed in either the striatum or mPFC. Furthermore, gene set enrichment analysis of the RNA-sequencing analysis revealed that ribosome-related genes were enriched especially in the up-regulated genes of Shank3 TG hypothalamus, which is in contrast to the results of the Shank3 TG striatum and mPFC analyses, where ribosome-related genes were enriched in the down-regulated genes. Beyond revealing endogenous Shank3 mRNA and protein expression in the hypothalamus, our results suggest unique molecular changes in the hypothalamus of Shank3 TG mice compared with those in the striatum and mPFC.
Collapse
|
16
|
Hulbert SW, Bey AL, Jiang YH. Environmental enrichment has minimal effects on behavior in the Shank3 complete knockout model of autism spectrum disorder. Brain Behav 2018; 8:e01107. [PMID: 30317697 PMCID: PMC6236244 DOI: 10.1002/brb3.1107] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 08/01/2018] [Accepted: 08/05/2018] [Indexed: 01/24/2023] Open
Abstract
INTRODUCTION Several studies have supported the use of enriched environments to prevent the manifestation of ASD-like phenotypes in laboratory rodents. While the translational value of such experiments is unknown, the findings have been relatively consistent across many different models. METHODS In the current study, we tested the effects of early environmental enrichment on a mouse model of ASD with high construct validity, the Shank3 ∆e4-22 mice our laboratory previously generated and characterized. RESULTS Contrary to previous reports, we found no benefits of enriched rearing, including no change in repetitive self-grooming or hole-board exploration. Instead, we found that early environmental enrichment increased anxiety-like behavior in all mice regardless of genotype and decreased motor performance specifically in wild-type mice. CONCLUSIONS Although using a different enrichment protocol may have rescued the phenotypes in our mouse model, these results suggest that a "one-size fits all" approach may not be the best when it comes to behavioral intervention for ASD and underscores the need for effective pharmaceutical development in certain genetic syndromes with severe symptom presentation.
Collapse
Affiliation(s)
- Samuel W Hulbert
- Department of Neurobiology, Duke University School of Medicine, Durham, North Carolina
| | - Alexandra L Bey
- Department of Neurobiology, Duke University School of Medicine, Durham, North Carolina
| | - Yong-Hui Jiang
- Department of Neurobiology, Duke University School of Medicine, Durham, North Carolina.,Department of Pediatrics, Duke University School of Medicine, Durham, North Carolina.,Duke Institute of Brain Science, Duke University School of Medicine, Durham, North Carolina
| |
Collapse
|
17
|
Bariselli S, Contestabile A, Tzanoulinou S, Musardo S, Bellone C. SHANK3 Downregulation in the Ventral Tegmental Area Accelerates the Extinction of Contextual Associations Induced by Juvenile Non-familiar Conspecific Interaction. Front Mol Neurosci 2018; 11:360. [PMID: 30364266 PMCID: PMC6193109 DOI: 10.3389/fnmol.2018.00360] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 09/12/2018] [Indexed: 01/14/2023] Open
Abstract
Haploinsufficiency of the SHANK3 gene, encoding for a scaffolding protein located in the postsynaptic density of glutamatergic synapse, has been linked to forms of autism spectrum disorders (ASDs). It has been shown that SHANK3 controls the maturation of social reward circuits in the ventral tegmental area (VTA). Whether the impairments in associative learning observed in ASD relate to SHANK3 insufficiency restricted to the reward system is still an open question. Here, we first characterize a social-conditioned place preference (CPP) paradigm based on the direct and free interaction with a juvenile and non-familiar conspecific. In both group- and single-housed C57Bl6/j late adolescence male mice, this CPP protocol promotes the formation of social-induced contextual associations that undergo extinction. Interestingly, the downregulation of Shank3 expression in the VTA altered the habituation to a non-familiar conspecific during conditioning and accelerated the extinction of social-induced conditioned responses. Thus, inspired by the literature on drugs of abuse-induced contextual learning, we propose that acquisition and extinction of CPP might be used as behavioral assays to assess social-induced contextual association and “social-seeking” dysfunctions in animal models of psychiatric disorders.
Collapse
Affiliation(s)
- Sebastiano Bariselli
- Department of Fundamental Neuroscience, University of Geneva, Centre Médical Universitaire (CMU), Geneva, Switzerland
| | - Alessandro Contestabile
- Department of Fundamental Neuroscience, University of Geneva, Centre Médical Universitaire (CMU), Geneva, Switzerland
| | - Stamatina Tzanoulinou
- Department of Fundamental Neuroscience, University of Geneva, Centre Médical Universitaire (CMU), Geneva, Switzerland
| | - Stefano Musardo
- Department of Fundamental Neuroscience, University of Geneva, Centre Médical Universitaire (CMU), Geneva, Switzerland
| | - Camilla Bellone
- Department of Fundamental Neuroscience, University of Geneva, Centre Médical Universitaire (CMU), Geneva, Switzerland
| |
Collapse
|
18
|
Behavioral Phenotyping of an Improved Mouse Model of Phelan-McDermid Syndrome with a Complete Deletion of the Shank3 Gene. eNeuro 2018; 5:eN-CFN-0046-18. [PMID: 30302388 PMCID: PMC6175061 DOI: 10.1523/eneuro.0046-18.2018] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 05/07/2018] [Accepted: 05/28/2018] [Indexed: 11/26/2022] Open
Abstract
Phelan–McDermid syndrome (PMS) is a rare genetic disorder in which one copy of the SHANK3 gene is missing or mutated, leading to a global developmental delay, intellectual disability (ID), and autism. Multiple intragenic promoters and alternatively spliced exons are responsible for the formation of numerous isoforms. Many genetically-modified mouse models of PMS have been generated but most disrupt only some of the isoforms. In contrast, the vast majority of known SHANK3 mutations found in patients involve deletions that disrupt all isoforms. Here, we report the production and thorough behavioral characterization of a new mouse model in which all Shank3 isoforms are disrupted. Domains and tasks examined in adults included measures of general health, neurological reflexes, motor abilities, sensory reactivity, social behavior, repetitive behaviors, cognition and behavioral inflexibility, and anxiety. Our mice are more severely affected than previously published models. While the deficits were typically more pronounced in homozygotes, an intermediate phenotype was observed for heterozygotes in many paradigms. As in other Shank3 mouse models, stereotypies, including increased grooming, were observed. Additionally, sensory alterations were detected in both neonatal and adult mice, and motor behavior was strongly altered, especially in the open field and rotarod locomotor tests. While social behaviors measured with the three-chambered social approach and male-female interaction tests were not strongly impacted, Shank3-deficient mice displayed a strong escape behavior and avoidance of inanimate objects in novel object recognition, repetitive novel object contact, marble burying, and nest building tasks, indicating increased novelty-induced anxiety. Similarly, increased freezing was observed during fear conditioning training and amygdala-dependent cued retrieval. Finally, deficits were observed in both initial training and reversal in the Barnes maze and in contextual fear testing, which are memory tasks involving hippocampal-prefrontal circuits. In contrast, working memory in the Y-maze spontaneous alternation test was not altered. This new mouse model of PMS, engineered to most closely represent human mutations, recapitulates core symptoms of PMS providing improvements for both construct and face validity, compared to previous models.
Collapse
|
19
|
Ueno H, Suemitsu S, Murakami S, Kitamura N, Wani K, Okamoto M, Matsumoto Y, Aoki S, Ishihara T. Empathic behavior according to the state of others in mice. Brain Behav 2018; 8:e00986. [PMID: 29845752 PMCID: PMC6043708 DOI: 10.1002/brb3.986] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Accepted: 03/24/2018] [Indexed: 01/11/2023] Open
Abstract
INTRODUCTION Empathic behavior is essential for social activities in social animals. Therefore, lack of empathy is a feature of several neuropsychiatric disorders. However, the underlying mechanisms of empathy and which animals possess it remain unclear. In this study, we investigated whether mice show empathic behavior. METHODS We tested mice for empathy-like behaviors toward conspecifics who were distressed. We investigated behavioral changes in cage-mate or stranger mice. RESULTS When the conspecific mice were in a painful state, subject mice showed preferential approach behavior toward them, presumably recognizing the state. Both visual information and olfactory information are indispensable for this empathic behavior. CONCLUSIONS These results suggest that the mouse recognizes the emotional state of the conspecific and engages in social interaction. The results of this study are useful for the elucidation of the causal mechanisms involved in neuropsychiatric disorders and may contribute in the development of novel treatment targets.
Collapse
Affiliation(s)
- Hiroshi Ueno
- Department of Medical Technology, Kawasaki University of Medical Welfare, Okayama, Japan.,Department of Medical Technology, Graduate School of Health Sciences, Okayama University, Okayama, Japan
| | | | - Shinji Murakami
- Department of Psychiatry, Kawasaki Medical School, Okayama, Japan
| | - Naoya Kitamura
- Department of Psychiatry, Kawasaki Medical School, Okayama, Japan
| | - Kenta Wani
- Department of Psychiatry, Kawasaki Medical School, Okayama, Japan
| | - Motoi Okamoto
- Department of Medical Technology, Graduate School of Health Sciences, Okayama University, Okayama, Japan
| | - Yosuke Matsumoto
- Department of Neuropsychiatry, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Shozo Aoki
- Department of Psychiatry, Kawasaki Medical School, Okayama, Japan
| | - Takeshi Ishihara
- Department of Psychiatry, Kawasaki Medical School, Okayama, Japan
| |
Collapse
|
20
|
Luo J, Norris RH, Gordon SL, Nithianantharajah J. Neurodevelopmental synaptopathies: Insights from behaviour in rodent models of synapse gene mutations. Prog Neuropsychopharmacol Biol Psychiatry 2018; 84:424-439. [PMID: 29217145 DOI: 10.1016/j.pnpbp.2017.12.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 11/28/2017] [Accepted: 12/03/2017] [Indexed: 11/15/2022]
Abstract
The genomic revolution has begun to unveil the enormous complexity and heterogeneity of the genetic basis of neurodevelopmental disorders such as such epilepsy, intellectual disability, autism spectrum disorder and schizophrenia. Increasingly, human mutations in synapse genes are being identified across these disorders. These neurodevelopmental synaptopathies highlight synaptic homeostasis pathways as a convergence point underlying disease mechanisms. Here, we review some of the key pre- and postsynaptic genes in which penetrant human mutations have been identified in neurodevelopmental disorders for which genetic rodent models have been generated. Specifically, we focus on the main behavioural phenotypes that have been documented in these animal models, to consolidate our current understanding of how synapse genes regulate key behavioural and cognitive domains. These studies provide insights into better understanding the basis of the overlapping genetic and cognitive heterogeneity observed in neurodevelopmental disorders.
Collapse
Affiliation(s)
- J Luo
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria 3052, Australia
| | - R H Norris
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria 3052, Australia
| | - S L Gordon
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria 3052, Australia
| | - J Nithianantharajah
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria 3052, Australia.
| |
Collapse
|
21
|
Ferhat AT, Halbedl S, Schmeisser MJ, Kas MJ, Bourgeron T, Ey E. Behavioural Phenotypes and Neural Circuit Dysfunctions in Mouse Models of Autism Spectrum Disorder. ADVANCES IN ANATOMY EMBRYOLOGY AND CELL BIOLOGY 2018; 224:85-101. [PMID: 28551752 DOI: 10.1007/978-3-319-52498-6_5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental condition primarily characterised by alterations in social interaction and communication combined with the presence of restricted interests and stereotyped behaviours. Mutations in several genes have been associated with ASD resulting in the generation of corresponding mouse models. Here, we focus on the behavioural (social and stereotyped behaviours), functional and structural traits of mice with mutations in genes encoding defined synaptic proteins including adhesion proteins, scaffolding proteins and subunits of channels and receptors. A meta-analysis on ASD mouse models shows that they can be divided into two subgroups. Cluster I gathered models highly impaired in social interest, stereotyped behaviours, synaptic physiology and protein composition, while Cluster II regrouped much less impaired models, with typical social interactions. This distribution was not related to gene families. Even within the large panel of mouse models carrying mutations in Shank3, the number of mutated isoforms was not related to the severity of the phenotype. Our study points that the majority of structural or functional analyses were performed in the hippocampus. However, to robustly link the structural and functional impairments with the behavioural deficits observed, brain structures forming relevant nodes in networks involved in social and stereotyped behaviours should be targeted in the future. In addition, the characterisation of core ASD-like behaviours needs to be more detailed using new approaches quantifying the variations in social motivation, recognition and stereotyped behaviours.
Collapse
Affiliation(s)
- Allain-Thibeault Ferhat
- Human Genetics and Cognitive Functions Unit, Institut Pasteur, Paris, France.,CNRS UMR 3571 Genes, Synapses and Cognition, Institut Pasteur, Paris, France.,Human Genetics and Cognitive Functions, University Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Sonja Halbedl
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany.,International Graduate School in Molecular Medicine, Ulm University, Ulm, Germany
| | - Michael J Schmeisser
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany.,Division of Neuroanatomy, Institute of Anatomy, Otto-von-Guericke University Magdeburg, Magdeburg, Germany.,Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Martien J Kas
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands.,Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, The Netherlands
| | - Thomas Bourgeron
- Human Genetics and Cognitive Functions Unit, Institut Pasteur, Paris, France.,CNRS UMR 3571 Genes, Synapses and Cognition, Institut Pasteur, Paris, France.,Human Genetics and Cognitive Functions, University Paris Diderot, Sorbonne Paris Cité, Paris, France.,FondaMental Foundation, Créteil, France.,Gillberg Neuropsychiatry Centre, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Elodie Ey
- Human Genetics and Cognitive Functions Unit, Institut Pasteur, Paris, France. .,CNRS UMR 3571 Genes, Synapses and Cognition, Institut Pasteur, Paris, France. .,Human Genetics and Cognitive Functions, University Paris Diderot, Sorbonne Paris Cité, Paris, France.
| |
Collapse
|
22
|
Hiroi N. Critical reappraisal of mechanistic links of copy number variants to dimensional constructs of neuropsychiatric disorders in mouse models. Psychiatry Clin Neurosci 2018; 72:301-321. [PMID: 29369447 PMCID: PMC5935536 DOI: 10.1111/pcn.12641] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 12/27/2017] [Accepted: 01/19/2018] [Indexed: 12/17/2022]
Abstract
Copy number variants are deletions and duplications of a few thousand to million base pairs and are associated with extraordinarily high levels of autism spectrum disorder, schizophrenia, intellectual disability, or attention-deficit hyperactivity disorder. The unprecedented levels of robust and reproducible penetrance of copy number variants make them one of the most promising and reliable entry points to delve into the mechanistic bases of many mental disorders. However, the precise mechanistic bases of these associations still remain elusive in humans due to the many genes encoded in each copy number variant and the diverse associated phenotypic features. Genetically engineered mice have provided a technical means to ascertain precise genetic mechanisms of association between copy number variants and dimensional aspects of mental illnesses. Molecular, cellular, and neuronal phenotypes can be detected as potential mechanistic substrates for various behavioral constructs of mental illnesses. However, mouse models come with many technical pitfalls. Genetic background is not well controlled in many mouse models, leading to rather obvious interpretative issues. Dose alterations of many copy number variants and single genes within copy number variants result in some molecular, cellular, and neuronal phenotypes without a behavioral phenotype or with a behavioral phenotype opposite to what is seen in humans. In this review, I discuss technical and interpretative pitfalls of mouse models of copy number variants and highlight well-controlled studies to suggest potential neuronal mechanisms of dimensional aspects of mental illnesses. Mouse models of copy number variants represent toeholds to achieve a better understanding of the mechanistic bases of dimensions of neuropsychiatric disorders and thus for development of mechanism-based therapeutic options in humans.
Collapse
Affiliation(s)
- Noboru Hiroi
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, New York, USA.,Department of Neuroscience, Albert Einstein College of Medicine, New York, USA.,Department of Genetics, Albert Einstein College of Medicine, New York, USA
| |
Collapse
|
23
|
Yin J, Chen W, Chao ES, Soriano S, Wang L, Wang W, Cummock SE, Tao H, Pang K, Liu Z, Pereira FA, Samaco RC, Zoghbi HY, Xue M, Schaaf CP. Otud7a Knockout Mice Recapitulate Many Neurological Features of 15q13.3 Microdeletion Syndrome. Am J Hum Genet 2018; 102:296-308. [PMID: 29395075 DOI: 10.1016/j.ajhg.2018.01.005] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 01/10/2018] [Indexed: 10/18/2022] Open
Abstract
15q13.3 microdeletion syndrome is characterized by a wide spectrum of neurodevelopmental disorders, including developmental delay, intellectual disability, epilepsy, language impairment, abnormal behaviors, neuropsychiatric disorders, and hypotonia. This syndrome is caused by a deletion on chromosome 15q, which typically encompasses six genes. Here, through studies on OTU deubiquitinase 7A (Otud7a) knockout mice, we identify OTUD7A as a critical gene responsible for many of the cardinal phenotypes associated with 15q13.3 microdeletion syndrome. Otud7a-null mice show reduced body weight, developmental delay, abnormal electroencephalography patterns and seizures, reduced ultrasonic vocalizations, decreased grip strength, impaired motor learning/motor coordination, and reduced acoustic startle. We show that OTUD7A localizes to dendritic spines and that Otud7a-null mice have decreased dendritic spine density compared to their wild-type littermates. Furthermore, frequency of miniature excitatory postsynaptic currents (mEPSCs) is reduced in the frontal cortex of Otud7a-null mice, suggesting a role of Otud7a in regulation of dendritic spine density and glutamatergic synaptic transmission. Taken together, our results suggest decreased OTUD7A dosage as a major contributor to the neurodevelopmental phenotypes associated with 15q13.3 microdeletion syndrome, through the misregulation of dendritic spine density and activity.
Collapse
|
24
|
Dhamne SC, Silverman JL, Super CE, Lammers SHT, Hameed MQ, Modi ME, Copping NA, Pride MC, Smith DG, Rotenberg A, Crawley JN, Sahin M. Replicable in vivo physiological and behavioral phenotypes of the Shank3B null mutant mouse model of autism. Mol Autism 2017. [PMID: 28638591 PMCID: PMC5472997 DOI: 10.1186/s13229-017-0142-z] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Autism spectrum disorder (ASD) is a clinically and biologically heterogeneous condition characterized by social, repetitive, and sensory behavioral abnormalities. No treatments are approved for the core diagnostic symptoms of ASD. To enable the earliest stages of therapeutic discovery and development for ASD, robust and reproducible behavioral phenotypes and biological markers are essential to establish in preclinical animal models. The goal of this study was to identify electroencephalographic (EEG) and behavioral phenotypes that are replicable between independent cohorts in a mouse model of ASD. The larger goal of our strategy is to empower the preclinical biomedical ASD research field by generating robust and reproducible behavioral and physiological phenotypes in animal models of ASD, for the characterization of mechanistic underpinnings of ASD-relevant phenotypes, and to ensure reliability for the discovery of novel therapeutics. Genetic disruption of the SHANK3 gene, a scaffolding protein involved in the stability of the postsynaptic density in excitatory synapses, is thought to be responsible for a relatively large number of cases of ASD. Therefore, we have thoroughly characterized the robustness of ASD-relevant behavioral phenotypes in two cohorts, and for the first time quantified translational EEG activity in Shank3B null mutant mice. METHODS In vivo physiology and behavioral assays were conducted in two independently bred and tested full cohorts of Shank3B null mutant (Shank3B KO) and wildtype littermate control (WT) mice. EEG was recorded via wireless implanted telemeters for 7 days of baseline followed by 20 min of recording following pentylenetetrazol (PTZ) challenge. Behaviors relevant to the diagnostic and associated symptoms of ASD were tested on a battery of established behavioral tests. Assays were designed to reproduce and expand on the original behavioral characterization of Shank3B KO mice. Two or more corroborative tests were conducted within each behavioral domain, including social, repetitive, cognitive, anxiety-related, sensory, and motor categories of assays. RESULTS Relative to WT mice, Shank3B KO mice displayed a dramatic resistance to PTZ seizure induction and an enhancement of gamma band oscillatory EEG activity indicative of enhanced inhibitory tone. These findings replicated in two separate cohorts. Behaviorally, Shank3B KO mice exhibited repetitive grooming, deficits in aspects of reciprocal social interactions and vocalizations, and reduced open field activity, as well as variable deficits in sensory responses, anxiety-related behaviors, learning and memory. CONCLUSIONS Robust animal models and quantitative, replicable biomarkers of neural dysfunction are needed to decrease risk and enable successful drug discovery and development for ASD and other neurodevelopmental disorders. Complementary to the replicated behavioral phenotypes of the Shank3B mutant mouse is the new identification of a robust, translational in vivo neurophysiological phenotype. Our findings provide strong evidence for robustness and replicability of key translational phenotypes in Shank3B mutant mice and support the usefulness of this mouse model of ASD for therapeutic discovery.
Collapse
Affiliation(s)
- Sameer C Dhamne
- F.M. Kirby Neurobiology Center, Translational Neuroscience Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115 USA
| | - Jill L Silverman
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA 95821 USA
| | - Chloe E Super
- F.M. Kirby Neurobiology Center, Translational Neuroscience Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115 USA
| | - Stephen H T Lammers
- F.M. Kirby Neurobiology Center, Translational Neuroscience Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115 USA
| | - Mustafa Q Hameed
- F.M. Kirby Neurobiology Center, Translational Neuroscience Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115 USA
| | - Meera E Modi
- F.M. Kirby Neurobiology Center, Translational Neuroscience Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115 USA
| | - Nycole A Copping
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA 95821 USA
| | - Michael C Pride
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA 95821 USA
| | - Daniel G Smith
- Autism Speaks, Inc., Boston, MA USA.,Present address: BlackThorn Therapeutics, Inc., Cambridge, MA USA
| | - Alexander Rotenberg
- F.M. Kirby Neurobiology Center, Translational Neuroscience Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115 USA
| | - Jacqueline N Crawley
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA 95821 USA
| | - Mustafa Sahin
- F.M. Kirby Neurobiology Center, Translational Neuroscience Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115 USA
| |
Collapse
|
25
|
Vicidomini C, Ponzoni L, Lim D, Schmeisser M, Reim D, Morello N, Orelanna D, Tozzi A, Durante V, Scalmani P, Mantegazza M, Genazzani AA, Giustetto M, Sala M, Calabresi P, Boeckers TM, Sala C, Verpelli C. Pharmacological enhancement of mGlu5 receptors rescues behavioral deficits in SHANK3 knock-out mice. Mol Psychiatry 2017; 22:689-702. [PMID: 27021819 PMCID: PMC5014121 DOI: 10.1038/mp.2016.30] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 12/23/2015] [Accepted: 01/25/2016] [Indexed: 02/08/2023]
Abstract
SHANK3 (also called PROSAP2) genetic haploinsufficiency is thought to be the major cause of neuropsychiatric symptoms in Phelan-McDermid syndrome (PMS). PMS is a rare genetic disorder that causes a severe form of intellectual disability (ID), expressive language delays and other autistic features. Furthermore, a significant number of SHANK3 mutations have been identified in patients with autism spectrum disorders (ASD), and SHANK3 truncating mutations are associated with moderate to profound ID. The Shank3 protein is a scaffold protein that is located in the postsynaptic density (PSD) of excitatory synapses and is crucial for synapse development and plasticity. In this study, we investigated the molecular mechanisms associated with the ASD-like behaviors observed in Shank3Δ11-/- mice, in which exon 11 has been deleted. Our results indicate that Shank3 is essential to mediating metabotropic glutamate receptor 5 (mGlu5)-receptor signaling by recruiting Homer1b/c to the PSD, specifically in the striatum and cortex. Moreover, augmenting mGlu5-receptor activity by administering 3-Cyano-N-(1,3-diphenyl-1H-pyrazol-5-yl)benzamide ameliorated the functional and behavioral defects that were observed in Shank3Δ11-/- mice, suggesting that pharmaceutical treatments that increase mGlu5 activity may represent a new approach for treating patients that are affected by PMS and SHANK3 mutations.
Collapse
Affiliation(s)
| | | | - Dmitry Lim
- Department of Pharmaceutical Sciences, Università degli Studi
del Piemonte Orientale “Amedeo Avogadro”, Novara
| | | | | | - Noemi Morello
- Department of Neuroscience, University of Turin, Torino
| | | | - Alessandro Tozzi
- University of Perugia, Department of Experimental Medicine,
Perugia
| | - Valentina Durante
- Department of Medicine, University of Perugia and Clinica
Neurologica, Santa Maria della Misericordia Hospital, Perugia, Italy
| | - Paolo Scalmani
- U.O. of Neurophysiopathology and Diagnostic Epileptology, Foundation
Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS) Neurological Institute
Carlo Besta, Milan
| | - Massimo Mantegazza
- Institute of Molecular and Cellular Pharmacology (IPMC), Laboratory
of Excellence Ion Channel Science and Therapeutics (LabEx ICST), CNRS UMR7275 and
University of Nice-Sophia Antipolis, Valbonne
| | - Armando A. Genazzani
- Department of Pharmaceutical Sciences, Università degli Studi
del Piemonte Orientale “Amedeo Avogadro”, Novara
| | | | - Mariaelvina Sala
- CNR Neuroscience Institute, Milan, Milano,Institute for Anatomy and Cell Biology, Ulm University, Ulm
| | - Paolo Calabresi
- Department of Medicine, University of Perugia and Clinica
Neurologica, Santa Maria della Misericordia Hospital, Perugia, Italy
| | | | - Carlo Sala
- CNR Neuroscience Institute, Milan, Milano
| | | |
Collapse
|
26
|
Descalzi G, Mitsi V, Purushothaman I, Gaspari S, Avrampou K, Loh YHE, Shen L, Zachariou V. Neuropathic pain promotes adaptive changes in gene expression in brain networks involved in stress and depression. Sci Signal 2017; 10:10/471/eaaj1549. [PMID: 28325815 DOI: 10.1126/scisignal.aaj1549] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Neuropathic pain is a complex chronic condition characterized by various sensory, cognitive, and affective symptoms. A large percentage of patients with neuropathic pain are also afflicted with depression and anxiety disorders, a pattern that is also seen in animal models. Furthermore, clinical and preclinical studies indicate that chronic pain corresponds with adaptations in several brain networks involved in mood, motivation, and reward. Chronic stress is also a major risk factor for depression. We investigated whether chronic pain and stress affect similar molecular mechanisms and whether chronic pain can affect gene expression patterns that are involved in depression. Using two mouse models of neuropathic pain and depression [spared nerve injury (SNI) and chronic unpredictable stress (CUS)], we performed next-generation RNA sequencing and pathway analysis to monitor changes in gene expression in the nucleus accumbens (NAc), the medial prefrontal cortex (mPFC), and the periaqueductal gray (PAG). In addition to finding unique transcriptome profiles across these regions, we identified a substantial number of signaling pathway-associated genes with similar changes in expression in both SNI and CUS mice. Many of these genes have been implicated in depression, anxiety, and chronic pain in patients. Our study provides a resource of the changes in gene expression induced by long-term neuropathic pain in three distinct brain regions and reveals molecular connections between pain and chronic stress.
Collapse
Affiliation(s)
- Giannina Descalzi
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Vasiliki Mitsi
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Immanuel Purushothaman
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sevasti Gaspari
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kleopatra Avrampou
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Yong-Hwee Eddie Loh
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Li Shen
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Venetia Zachariou
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
27
|
Harony-Nicolas H, Kay M, du Hoffmann J, Klein ME, Bozdagi-Gunal O, Riad M, Daskalakis NP, Sonar S, Castillo PE, Hof PR, Shapiro ML, Baxter MG, Wagner S, Buxbaum JD. Oxytocin improves behavioral and electrophysiological deficits in a novel Shank3-deficient rat. eLife 2017; 6. [PMID: 28139198 PMCID: PMC5283828 DOI: 10.7554/elife.18904] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 01/02/2017] [Indexed: 11/13/2022] Open
Abstract
Mutations in the synaptic gene SHANK3 lead to a neurodevelopmental disorder known as Phelan-McDermid syndrome (PMS). PMS is a relatively common monogenic and highly penetrant cause of autism spectrum disorder (ASD) and intellectual disability (ID), and frequently presents with attention deficits. The underlying neurobiology of PMS is not fully known and pharmacological treatments for core symptoms do not exist. Here, we report the production and characterization of a Shank3-deficient rat model of PMS, with a genetic alteration similar to a human SHANK3 mutation. We show that Shank3-deficient rats exhibit impaired long-term social recognition memory and attention, and reduced synaptic plasticity in the hippocampal-medial prefrontal cortex pathway. These deficits were attenuated with oxytocin treatment. The effect of oxytocin on reversing non-social attention deficits is a particularly novel finding, and the results implicate an oxytocinergic contribution in this genetically defined subtype of ASD and ID, suggesting an individualized therapeutic approach for PMS. DOI:http://dx.doi.org/10.7554/eLife.18904.001 Phelan-McDermid syndrome is a genetic disorder on the autism spectrum that affects how children develop in several ways, with additional symptoms including attention deficits, delays in learning to speak and motor problems. This syndrome is known to be caused by changes in a single gene known as SHANK3 that disrupt communication between brain cells involved in memory and learning. However, we do not know how these changes relate to the symptoms of Phelan-McDermid syndrome. To understand how genetic changes affect the human brain, researchers often carry out experiments in rats or other small rodents because they have brains that are similar to ours. Harony-Nicolas et al. genetically modified rats to carry changes in the SHANK3 gene that reflect those found in people with Phelan-McDermid syndrome. The rats had disabilities related to those seen in Phelan-McDermid syndrome, including limits in long-term social memory and reduced attention span. They also showed changes in the connections between important parts of the brain. Therefore, studying these rats could help us to understand the link between molecular and cellular changes in the brain and how they affect people with Phelan-McDermid syndrome, and associated symptoms. Previous studies have shown that a chemical called oxytocin, which is naturally produced by the brain, helps to form bonds between individuals and can cause positive feelings in relation to certain memories. Harony-Nicolas et al. found treating the rats with oxytocin boosted social memory and led to improvements in other symptoms of Phelan-McDermid syndrome. In particular, oxytocin treatment helped to increase the attention span of the rats. Rats with changes in the SHANK3 gene will be a useful tool for future research into Phelan-McDermid syndrome, particularly in understanding how it affects the connections between brain cells, leading to the symptoms of Phelan-McDermid syndrome. A future challenge will be to find out whether oxytocin has the potential to be developed into a therapy to treat Phelan-McDermid syndrome in humans. Since there is evidence that SHANK3 is involved in other forms of autism, these rats will also be useful in understanding the other ways in which autism can develop. DOI:http://dx.doi.org/10.7554/eLife.18904.002
Collapse
Affiliation(s)
- Hala Harony-Nicolas
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, United States.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Maya Kay
- Sagol Department of Neurobiology, University of Haifa, Haifa, Israel
| | - Johann du Hoffmann
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, United States.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Matthew E Klein
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, United States
| | - Ozlem Bozdagi-Gunal
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, United States.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Mohammed Riad
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, United States.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Nikolaos P Daskalakis
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Sankalp Sonar
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, United States.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Pablo E Castillo
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, United States
| | - Patrick R Hof
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, United States.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, United States.,Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Matthew L Shapiro
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, United States.,Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Mark G Baxter
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, United States.,Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Shlomo Wagner
- Sagol Department of Neurobiology, University of Haifa, Haifa, Israel
| | - Joseph D Buxbaum
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, United States.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, United States.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, United States.,Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, United States.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, United States.,The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, United States
| |
Collapse
|
28
|
van de Lagemaat LN, Stanford LE, Pettit CM, Strathdee DJ, Strathdee KE, Elsegood KA, Fricker DG, Croning MDR, Komiyama NH, Grant SGN. Standardized experiments in mutant mice reveal behavioural similarity on 129S5 and C57BL/6J backgrounds. GENES BRAIN AND BEHAVIOR 2017; 16:409-418. [DOI: 10.1111/gbb.12364] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 09/12/2016] [Accepted: 11/21/2016] [Indexed: 01/27/2023]
Affiliation(s)
- L. N. van de Lagemaat
- Centre for Clinical Brain Sciences, Chancellor's Building; University of Edinburgh; Edinburgh UK
- Genes to Cognition Programme; Wellcome Trust Sanger Institute; Cambridge UK
| | - L. E. Stanford
- Genes to Cognition Programme; Wellcome Trust Sanger Institute; Cambridge UK
| | - C. M. Pettit
- Genes to Cognition Programme; Wellcome Trust Sanger Institute; Cambridge UK
| | - D. J. Strathdee
- Genes to Cognition Programme; Wellcome Trust Sanger Institute; Cambridge UK
- Transgenic Technology Division; CRUK Beatson Institute; Glasgow UK
| | - K. E. Strathdee
- Genes to Cognition Programme; Wellcome Trust Sanger Institute; Cambridge UK
| | - K. A. Elsegood
- Centre for Clinical Brain Sciences, Chancellor's Building; University of Edinburgh; Edinburgh UK
- Genes to Cognition Programme; Wellcome Trust Sanger Institute; Cambridge UK
| | - D. G. Fricker
- Centre for Clinical Brain Sciences, Chancellor's Building; University of Edinburgh; Edinburgh UK
- Genes to Cognition Programme; Wellcome Trust Sanger Institute; Cambridge UK
| | - M. D. R. Croning
- Centre for Clinical Brain Sciences, Chancellor's Building; University of Edinburgh; Edinburgh UK
- Genes to Cognition Programme; Wellcome Trust Sanger Institute; Cambridge UK
| | - N. H. Komiyama
- Centre for Clinical Brain Sciences, Chancellor's Building; University of Edinburgh; Edinburgh UK
- Genes to Cognition Programme; Wellcome Trust Sanger Institute; Cambridge UK
| | - S. G. N. Grant
- Centre for Clinical Brain Sciences, Chancellor's Building; University of Edinburgh; Edinburgh UK
- Genes to Cognition Programme; Wellcome Trust Sanger Institute; Cambridge UK
| |
Collapse
|
29
|
Copping NA, Berg EL, Foley GM, Schaffler MD, Onaga BL, Buscher N, Silverman JL, Yang M. Touchscreen learning deficits and normal social approach behavior in the Shank3B model of Phelan-McDermid Syndrome and autism. Neuroscience 2016; 345:155-165. [PMID: 27189882 DOI: 10.1016/j.neuroscience.2016.05.016] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 05/02/2016] [Accepted: 05/05/2016] [Indexed: 11/24/2022]
Abstract
SHANK3 is a synaptic scaffolding protein localized in the postsynaptic density and has a crucial role in synaptogenesis and neural physiology. Deletions and point mutations in SHANK3 cause Phelan-McDermid Syndrome (PMS), and have also been implicated in autism spectrum disorder (ASD) and intellectual disabilities, leading to the hypothesis that reduced SHANK3 expression impairs basic brain functions that are important for social communication and cognition. Several mouse models of Shank3 deletions have been generated, varying in the specific domain deleted. Here we report impairments in cognitive function in mice heterozygous for exon 13-16 (coding for the PDZ domain) deletion. The touchscreen pairwise discrimination task was chosen by virtue of its: (a) conceptual and technical similarities to the Cambridge Neuropsychological Test Automated Battery (CANTAB) and NIH Toolbox Cognition Battery used for testing cognitive functions in humans, (b) minimal demand on motor abilities, and (c) capability to measure many aspects of learning and memory and complex cognitive functions, including cognitive flexibility. The similarity between our mouse tasks and human cognitive assays means a high translational validity in future intervention studies using preclinical models. Our study revealed that Shank3B heterozygous mice (+/-) were slower to reach criterion in the pairwise visual discrimination task, and exhibited trends toward making more errors (first trial errors) and more correction errors than wildtype mice (+/+). Open field activity was normal in +/-, ruling out hypo- or hyperactivity as potential confounds in the touchscreen test. Sociability in the three chamber test was also normal in both +/+ and +/-. These results indicate a deficit in discrimination learning in the Shank3B model of PMS and ASD, suggesting that this mouse model is a useful preclinical tool for studying neurobiological mechanisms behind cognitive impairments in PMS and ASD. The current findings are the starting point for our future research in which we will investigate multiple domains of cognition and explore pharmacological interventions.
Collapse
Affiliation(s)
- Nycole A Copping
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA 95817, United States
| | - Elizabeth L Berg
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA 95817, United States
| | - Gillian M Foley
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA 95817, United States
| | - Melanie D Schaffler
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA 95817, United States
| | - Beth L Onaga
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA 95817, United States
| | - Nathalie Buscher
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA 95817, United States
| | - Jill L Silverman
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA 95817, United States
| | - Mu Yang
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA 95817, United States.
| |
Collapse
|
30
|
Yi F, Danko T, Botelho SC, Patzke C, Pak C, Wernig M, Südhof TC. Autism-associated SHANK3 haploinsufficiency causes Ih channelopathy in human neurons. Science 2016; 352:aaf2669. [PMID: 26966193 DOI: 10.1126/science.aaf2669] [Citation(s) in RCA: 213] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 02/26/2016] [Indexed: 12/13/2022]
Abstract
Heterozygous SHANK3 mutations are associated with idiopathic autism and Phelan-McDermid syndrome. SHANK3 is a ubiquitously expressed scaffolding protein that is enriched in postsynaptic excitatory synapses. Here, we used engineered conditional mutations in human neurons and found that heterozygous and homozygous SHANK3 mutations severely and specifically impaired hyperpolarization-activated cation (Ih) channels. SHANK3 mutations caused alterations in neuronal morphology and synaptic connectivity; chronic pharmacological blockage of Ih channels reproduced these phenotypes, suggesting that they may be secondary to Ih-channel impairment. Moreover, mouse Shank3-deficient neurons also exhibited severe decreases in Ih currents. SHANK3 protein interacted with hyperpolarization-activated cyclic nucleotide-gated channel proteins (HCN proteins) that form Ih channels, indicating that SHANK3 functions to organize HCN channels. Our data suggest that SHANK3 mutations predispose to autism, at least partially, by inducing an Ih channelopathy that may be amenable to pharmacological intervention.
Collapse
Affiliation(s)
- Fei Yi
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA
| | - Tamas Danko
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA. Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA. Department of Pathology, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA
| | - Salome Calado Botelho
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA
| | - Christopher Patzke
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA
| | - ChangHui Pak
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA
| | - Marius Wernig
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA. Department of Pathology, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA
| | - Thomas C Südhof
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA. Howard Hughes Medical Institute, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA.
| |
Collapse
|
31
|
Guirado R, La Terra D, Bourguignon M, Carceller H, Umemori J, Sipilä P, Nacher J, Castrén E. Effects of PSA Removal from NCAM on the Critical Period Plasticity Triggered by the Antidepressant Fluoxetine in the Visual Cortex. Front Cell Neurosci 2016; 10:22. [PMID: 26903807 PMCID: PMC4743432 DOI: 10.3389/fncel.2016.00022] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 01/22/2016] [Indexed: 11/13/2022] Open
Abstract
Neuronal plasticity peaks during critical periods of postnatal development and is reduced towards adulthood. Recent data suggests that windows of juvenile-like plasticity can be triggered in the adult brain by antidepressant drugs such as Fluoxetine. Although the exact mechanisms of how Fluoxetine promotes such plasticity remains unknown, several studies indicate that inhibitory circuits play an important role. The polysialylated form of the neural cell adhesion molecules (PSA-NCAM) has been suggested to mediate the effects of Fluoxetine and it is expressed in the adult brain by mature interneurons. Moreover, the enzymatic removal of PSA by neuroaminidase-N not only affects the structure of interneurons but also has been shown to play a role in the onset of critical periods during development. We have here used ocular dominance plasticity in the mouse visual cortex as a model to investigate whether removal of PSA might influence the Fluoxetine-induced plasticity. We demonstrate that PSA removal in the adult visual cortex alters neither the baseline ocular dominance, nor the fluoxetine-induced shift in the ocular dominance. We also show that both chronic Fluoxetine treatment and PSA removal independently increase the basal FosB expression in parvalbumin (PV) interneurons in the primary visual cortex. Therefore, our data suggest that although PSA-NCAM regulates inhibitory circuitry, it is not required for the reactivation of juvenile-like plasticity triggered by Fluoxetine.
Collapse
Affiliation(s)
- Ramon Guirado
- Neuroscience Center, University of Helsinki Helsinki, Finland
| | - Danilo La Terra
- Neuroscience Center, University of Helsinki Helsinki, Finland
| | - Mathieu Bourguignon
- Department of Neuroscience and Biomedical Engineering, Aalto UniversityHelsinki, Finland; Basque Center on Cognition, Brain and LanguageDonostia, Spain
| | - Hector Carceller
- Departamento de Biologia Celular, Spanish National Network for Research in Mental Health, CIBERSAM, Fundación Investigación Hospital Clínico de Valencia, INCLIVA, Universitat de Valencia Valencia, Spain
| | - Juzoh Umemori
- Neuroscience Center, University of Helsinki Helsinki, Finland
| | - Pia Sipilä
- Neuroscience Center, University of HelsinkiHelsinki, Finland; Max Planck Institute for NeurobiologyMartinsried, Germany
| | - Juan Nacher
- Departamento de Biologia Celular, Spanish National Network for Research in Mental Health, CIBERSAM, Fundación Investigación Hospital Clínico de Valencia, INCLIVA, Universitat de Valencia Valencia, Spain
| | - Eero Castrén
- Neuroscience Center, University of Helsinki Helsinki, Finland
| |
Collapse
|
32
|
Kazdoba TM, Leach PT, Crawley JN. Behavioral phenotypes of genetic mouse models of autism. GENES, BRAIN, AND BEHAVIOR 2016; 15:7-26. [PMID: 26403076 PMCID: PMC4775274 DOI: 10.1111/gbb.12256] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Revised: 08/27/2015] [Accepted: 09/18/2015] [Indexed: 12/11/2022]
Abstract
More than a hundred de novo single gene mutations and copy-number variants have been implicated in autism, each occurring in a small subset of cases. Mutant mouse models with syntenic mutations offer research tools to gain an understanding of the role of each gene in modulating biological and behavioral phenotypes relevant to autism. Knockout, knockin and transgenic mice incorporating risk gene mutations detected in autism spectrum disorder and comorbid neurodevelopmental disorders are now widely available. At present, autism spectrum disorder is diagnosed solely by behavioral criteria. We developed a constellation of mouse behavioral assays designed to maximize face validity to the types of social deficits and repetitive behaviors that are central to an autism diagnosis. Mouse behavioral assays for associated symptoms of autism, which include cognitive inflexibility, anxiety, hyperactivity, and unusual reactivity to sensory stimuli, are frequently included in the phenotypic analyses. Over the past 10 years, we and many other laboratories around the world have employed these and additional behavioral tests to phenotype a large number of mutant mouse models of autism. In this review, we highlight mouse models with mutations in genes that have been identified as risk genes for autism, which work through synaptic mechanisms and through the mTOR signaling pathway. Robust, replicated autism-relevant behavioral outcomes in a genetic mouse model lend credence to a causal role for specific gene contributions and downstream biological mechanisms in the etiology of autism.
Collapse
Affiliation(s)
- T. M. Kazdoba
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA, USA
| | - P. T. Leach
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA, USA
| | - J. N. Crawley
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA, USA
| |
Collapse
|
33
|
Harony-Nicolas H, De Rubeis S, Kolevzon A, Buxbaum JD. Phelan McDermid Syndrome: From Genetic Discoveries to Animal Models and Treatment. J Child Neurol 2015; 30:1861-70. [PMID: 26350728 PMCID: PMC5321557 DOI: 10.1177/0883073815600872] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 07/15/2015] [Indexed: 01/16/2023]
Abstract
Phelan-McDermid syndrome or 22q13.3 deletion syndrome is a rare neurodevelopmental disorder characterized by generalized developmental delay, intellectual disability, absent or delayed speech, seizures, autism spectrum disorder, neonatal hypotonia, physical dysmorphic features, and recurrent medical comorbidities. Individuals with Phelan-McDermid syndrome have terminal deletions of the chromosomal region 22q13.3 encompassing SHANK3, a gene encoding a structural component of excitatory synapses indispensable for proper synaptogenesis and neuronal physiology, or point mutations within the gene. Here, we review the clinical aspects of the syndrome and the genetic findings shedding light onto the underlying etiology. We also provide an overview on the evidence from genetic studies and mouse models that supports SHANK3 haploinsufficiency as a major contributor of the neurobehavioral manifestations of Phelan-McDermid syndrome. Finally, we discuss how all these discoveries are uncovering the pathophysiology of Phelan-McDermid syndrome and are being translated into clinical trials for novel therapeutics ameliorating the core symptoms of the disorder.
Collapse
Affiliation(s)
- Hala Harony-Nicolas
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Silvia De Rubeis
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alexander Kolevzon
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Joseph D Buxbaum
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
34
|
McCammon JM, Sive H. Addressing the Genetics of Human Mental Health Disorders in Model Organisms. Annu Rev Genomics Hum Genet 2015; 16:173-97. [DOI: 10.1146/annurev-genom-090314-050048] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Jasmine M. McCammon
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts 02142;
| | - Hazel Sive
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts 02142;
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| |
Collapse
|
35
|
Sungur AÖ, Vörckel KJ, Schwarting RKW, Wöhr M. Repetitive behaviors in the Shank1 knockout mouse model for autism spectrum disorder: developmental aspects and effects of social context. J Neurosci Methods 2014; 234:92-100. [PMID: 24820912 DOI: 10.1016/j.jneumeth.2014.05.003] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Revised: 04/22/2014] [Accepted: 05/01/2014] [Indexed: 01/16/2023]
Abstract
BACKGROUND Autism spectrum disorder (ASD) is characterized by persistent deficits in social behavior and communication, together with restricted and repetitive patterns of behavior. Several ASD candidate genes have been identified, including the SHANK gene family with its three family members SHANK1, SHANK2, and SHANK3. METHODS Typically, repetitive behavior in mouse models for ASD is assessed by measuring self-grooming behavior. The first aim of the current study was to assess repetitive behaviors in Shank1(-/-) null mutant, Shank1(+/-) heterozygous, and Shank1(+/+) wildtype littermate control mice by means of a comprehensive approach, including the assessment of self-grooming, digging behavior, and marble burying. The second aim was to establish a test paradigm that allows for assessing the effects of social context on the occurrence of repetitive behaviors in a genotype-dependent manner. To this aim, repetitive behaviors were repeatedly tested on three consecutive days in distinct social contexts, namely in presence or absence of social odors. RESULTS Shank1(+/-) heterozygous and to a lesser extent Shank1(-/-) null mutant mice displayed slightly elevated levels of self-grooming behavior as adults, but not as juveniles, with genotype differences being most prominent in the social context. In contrast to elevated self-grooming behavior, marble burying was strongly reduced in adult Shank1(+/-) heterozygous and Shank1(-/-) null mutant mice across social contexts, as compared to adult Shank1(+/+) wildtype littermate controls. CONCLUSION The opposite effects of the Shank1 deletion on the two types of repetitive behaviors are in line with a number of studies on repetitive behaviors in other genetic Shank models.
Collapse
Affiliation(s)
- A Özge Sungur
- Behavioral Neuroscience, Experimental and Biological Psychology, Philipps-University of Marburg, Gutenbergstr. 18, D-35032 Marburg, Germany
| | - Karl J Vörckel
- Behavioral Neuroscience, Experimental and Biological Psychology, Philipps-University of Marburg, Gutenbergstr. 18, D-35032 Marburg, Germany
| | - Rainer K W Schwarting
- Behavioral Neuroscience, Experimental and Biological Psychology, Philipps-University of Marburg, Gutenbergstr. 18, D-35032 Marburg, Germany
| | - Markus Wöhr
- Behavioral Neuroscience, Experimental and Biological Psychology, Philipps-University of Marburg, Gutenbergstr. 18, D-35032 Marburg, Germany.
| |
Collapse
|