1
|
Li R, Anzai M, Shibata A, Ito-Ishida A. Synaptic disturbance in neurodevelopmental disorders: Perspectives from fragile X and Rett syndromes. Brain Dev 2025; 47:104358. [PMID: 40228442 DOI: 10.1016/j.braindev.2025.104358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 03/18/2025] [Accepted: 03/25/2025] [Indexed: 04/16/2025]
Abstract
Neurodevelopmental disorders (NDDs) are often referred to as "synaptopathies" because many of their behavioral symptoms arise from impaired synaptic development and function. However, the mechanisms that connect synaptic dysfunction to neurological symptoms remain unclear, mainly due to the wide variety of genetic and environmental factors involved in these disorders. Fragile X syndrome and Rett syndrome, two extensively studied monogenic NDDs, provide a unique opportunity to explore these mechanisms at molecular, cellular, and synaptic levels. This review summarizes the current understanding of how synaptic alterations contribute to the neurological symptoms observed in fragile X and Rett syndromes. A comparison of findings from mouse models indicates that an imbalance in local and distal connectivity may serve as a common feature of both disorders.
Collapse
Affiliation(s)
- Ruixiang Li
- Laboratory for Brain Development and Disorders, RIKEN Center for Brain Science, Saitama, Japan; Graduate School of Brain Science, Doshisha University, Kyoto, Japan
| | - Mai Anzai
- Laboratory for Brain Development and Disorders, RIKEN Center for Brain Science, Saitama, Japan
| | - Akiko Shibata
- Laboratory for Brain Development and Disorders, RIKEN Center for Brain Science, Saitama, Japan
| | - Aya Ito-Ishida
- Laboratory for Brain Development and Disorders, RIKEN Center for Brain Science, Saitama, Japan.
| |
Collapse
|
2
|
Torkzaban B, Zhu Y, Lopez C, Alexander JM, Ma J, Sun Y, Maschhoff KR, Hu W, Jacob MH, Lin D, Mao HQ, Martin S, Coller J. Use of polyadenosine tail mimetics to enhance mRNA expression from genes associated with haploinsufficiency disorders. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102453. [PMID: 39967850 PMCID: PMC11834087 DOI: 10.1016/j.omtn.2025.102453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 01/10/2025] [Indexed: 02/20/2025]
Abstract
Polyadenosine (poly(A)) tails are nearly ubiquitous in human messenger RNA (mRNA) governing mRNA stability and translation. Crucially, the poly(A) tail regulates cytoplasmic gene expression by undergoing controlled removal upon exposure to the cytoplasm. Upon removal, mRNA ceases protein production and may subsequently be degraded or silenced. We have generated a therapeutic modality that tethers a poly(A) tail mimetic on the 3' end of specifically targeted mRNAs, thereby enhancing their expression beyond their normal utility. This technology, which we term mRNA boosters, lends itself to uses on haploinsufficiency disorders, where reduced gene expression manifests in a disease state. By polyadenylating short RNA sequences antisense to the 3' untranslated region (UTR) of specific mRNAs, we demonstrate that we can selectively and significantly enhance mRNA expression both in vitro and in vivo. We showcase the effectiveness of this technology on genes linked to autism spectrum disorders such as SYNGAP1, M E CP2, PURA, and CTNNB1, illustrating increased expression in both human cell cultures and animal models. These findings indicate that small poly(A) tail mimetics can substantially enhance mRNA expression, providing the potential to efficaciously treat haploinsufficiency disorders.
Collapse
Affiliation(s)
- Bahareh Torkzaban
- Department of Molecular Biology and Genetics, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Yining Zhu
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Christian Lopez
- Department of Molecular Biology and Genetics, Johns Hopkins University, Baltimore, MD 21205, USA
| | | | - Jingyao Ma
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Yongzhi Sun
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | | | - Wenqian Hu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| | - Michele H. Jacob
- Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, USA
| | - Dingchang Lin
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Hai-Quan Mao
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Sophie Martin
- Department of Molecular Biology and Genetics, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Jeff Coller
- Department of Molecular Biology and Genetics, Johns Hopkins University, Baltimore, MD 21205, USA
- RNA Innovation Center, Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
3
|
Tang B, Wu Z, Wang Q, Tang J. Neuronal Network Activation Induced by Forniceal Deep Brain Stimulation in Mice. Genes (Basel) 2025; 16:210. [PMID: 40004540 PMCID: PMC11855867 DOI: 10.3390/genes16020210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 01/26/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
Background: The fimbria-fornix is a nerve fiber bundle that connects various structures of the limbic system in the brain and plays a key role in cognition. It has become a major target of deep brain stimulation (DBS) to treat memory impairment in both dementia patients and animal models of neurological diseases. Previously, we have reported the beneficial memory effects of chronic forniceal DBS in mouse models of intellectual disability disorders. In Rett syndrome and CDKL5 deficiency disorder models, DBS strengthens hippocampal synaptic plasticity, reduces dentate inhibitory transmission or increases adult hippocampal neurogenesis that aids memory. However, the underlying neuronal circuitry mechanisms remain unknown. This study we explored the neural network circuits involved in forniceal DBS treatment. Methods: We used acute forniceal DBS-induced expression of c-Fos, an activity-dependent neuronal marker, to map the brain structures functionally connected to the fornix. We also evaluated the mouse behavior of locomotion, anxiety, and fear memory after acute forniceal DBS treatment. Results: Acute forniceal DBS induces robust activation of multiple structures in the limbic system. DBS-induced neuronal activation extends beyond hippocampal formation and includes brain structures not directly innervated by the fornix. Conclusions: Acute forniceal DBS activates multiple limbic structures associated with emotion and memory. The neural circuits revealed here help elucidate the neural network effect and pave the way for further research on the mechanism by which forniceal DBS induces benefits on cognitive impairments.
Collapse
Affiliation(s)
- Bin Tang
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX 77030, USA; (B.T.); (Z.W.); (Q.W.)
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Zhenyu Wu
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX 77030, USA; (B.T.); (Z.W.); (Q.W.)
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Qi Wang
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX 77030, USA; (B.T.); (Z.W.); (Q.W.)
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jianrong Tang
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX 77030, USA; (B.T.); (Z.W.); (Q.W.)
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
4
|
Samanta D, Haneef Z, Albert GW, Naik S, Reeders PC, Jain P, Abel TJ, Al-Ramadhani R, Ibrahim GM, Warren AEL. Neuromodulation strategies in developmental and epileptic encephalopathies. Epilepsy Behav 2024; 160:110067. [PMID: 39393142 DOI: 10.1016/j.yebeh.2024.110067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 09/19/2024] [Accepted: 09/28/2024] [Indexed: 10/13/2024]
Abstract
Developmental and epileptic encephalopathies (DEEs) are a group of childhood-onset epilepsy syndromes characterized by frequent seizures, severe cognitive and behavioral impairments, and poor long-term outcomes. These conditions are typically refractory to currently available medical therapies, prompting recent exploration of neuromodulation treatments such as deep brain stimulation (DBS) and responsive neurostimulation (RNS), which aim to modulate epileptic networks spanning cortical and subcortical regions. These advances have occurred alongside an improved understanding of syndrome-specific and interictal epileptiform discharge/seizure-specific brain networks. By targeting key nodes within these networks, DBS and RNS hold promise for influencing seizures and associated cognitive and behavioral comorbidities. Initial experiences with centromedian (CM) thalamic DBS for Lennox-Gastaut syndrome (LGS) have shown modest efficacy across multiple seizure types. Reports also indicate the application of DBS and RNS across various genetic and structural etiologies commonly associated with DEEs, with mixed success. Although DBS and RNS are increasingly used in LGS and other DEEs, their mixed efficacy highlights a knowledge gap in understanding why some patients with LGS do not respond and which neuromodulation approach is most effective for other DEEs. To address these issues, this review first discusses recent neuroimaging studies showing similarities and differences in the epileptic brain networks underlying various DEEs, revealing the common involvement of the thalamus and the default-mode network (DMN) across multiple DEEs. We then examine thalamic DBS for LGS to illustrate how such network insights may be used to optimize neuromodulation. Although network-based neuromodulation is still in its infancy, the LGS model may serve as a framework for other DEEs, where optimal treatment necessitates consideration of the underlying epileptic networks. Lastly, the review suggests future research directions, including individualized connectivity assessment and biomarker identification through collaborative efforts, which may enhance the therapeutic potential of neuromodulation for individuals living with DEEs.
Collapse
Affiliation(s)
- Debopam Samanta
- Division of Child Neurology, Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
| | - Zulfi Haneef
- Department of Neurology, Baylor College of Medicine, Houston, TX 77030, USA; Neurology Care Line, Michael E. DeBakey VA Medical Center, Houston, TX 77030, USA
| | - Gregory W Albert
- Department of Neurosurgery, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Sunil Naik
- Department of Pediatrics and Neurology, Penn State Health Milton S. Hershey Medical Center, Hershey, PA 17033, USA
| | - Puck C Reeders
- Brain Institute, Nicklaus Children's Hospital, Miami, FL, USA
| | - Puneet Jain
- Epilepsy Program, Division of Neurology, Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Taylor J Abel
- Departmen of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ruba Al-Ramadhani
- Division of Child Neurology, University of Pittsburgh, Department of Pediatrics, Pittsburgh, PA, USA
| | - George M Ibrahim
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada; Program in Neuroscience and Mental Health, The Hospital for Sick Children Research Institute, Toronto, ON, Canada; Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, ON, Canada; Division of Neurosurgery, The Hospital for Sick Children, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Aaron E L Warren
- Department of Neurosurgery, Mass General Brigham, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
5
|
Zhang KK, Matin R, Gorodetsky C, Ibrahim GM, Gouveia FV. Systematic review of rodent studies of deep brain stimulation for the treatment of neurological, developmental and neuropsychiatric disorders. Transl Psychiatry 2024; 14:186. [PMID: 38605027 PMCID: PMC11009311 DOI: 10.1038/s41398-023-02727-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 12/19/2023] [Accepted: 12/22/2023] [Indexed: 04/13/2024] Open
Abstract
Deep brain stimulation (DBS) modulates local and widespread connectivity in dysfunctional networks. Positive results are observed in several patient populations; however, the precise mechanisms underlying treatment remain unknown. Translational DBS studies aim to answer these questions and provide knowledge for advancing the field. Here, we systematically review the literature on DBS studies involving models of neurological, developmental and neuropsychiatric disorders to provide a synthesis of the current scientific landscape surrounding this topic. A systematic analysis of the literature was performed following PRISMA guidelines. 407 original articles were included. Data extraction focused on study characteristics, including stimulation protocol, behavioural outcomes, and mechanisms of action. The number of articles published increased over the years, including 16 rat models and 13 mouse models of transgenic or healthy animals exposed to external factors to induce symptoms. Most studies targeted telencephalic structures with varying stimulation settings. Positive behavioural outcomes were reported in 85.8% of the included studies. In models of psychiatric and neurodevelopmental disorders, DBS-induced effects were associated with changes in monoamines and neuronal activity along the mesocorticolimbic circuit. For movement disorders, DBS improves symptoms via modulation of the striatal dopaminergic system. In dementia and epilepsy models, changes to cellular and molecular aspects of the hippocampus were shown to underlie symptom improvement. Despite limitations in translating findings from preclinical to clinical settings, rodent studies have contributed substantially to our current knowledge of the pathophysiology of disease and DBS mechanisms. Direct inhibition/excitation of neural activity, whereby DBS modulates pathological oscillatory activity within brain networks, is among the major theories of its mechanism. However, there remain fundamental questions on mechanisms, optimal targets and parameters that need to be better understood to improve this therapy and provide more individualized treatment according to the patient's predominant symptoms.
Collapse
Affiliation(s)
- Kristina K Zhang
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Program in Neuroscience and Mental Health, The Hospital for Sick Children, Toronto, ON, Canada
| | - Rafi Matin
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Program in Neuroscience and Mental Health, The Hospital for Sick Children, Toronto, ON, Canada
| | | | - George M Ibrahim
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Program in Neuroscience and Mental Health, The Hospital for Sick Children, Toronto, ON, Canada
- Division of Neurosurgery, The Hospital for Sick Children, Toronto, ON, Canada
| | | |
Collapse
|
6
|
Severino L, Kim J, Nam MH, McHugh TJ. From synapses to circuits: What mouse models have taught us about how autism spectrum disorder impacts hippocampal function. Neurosci Biobehav Rev 2024; 158:105559. [PMID: 38246230 DOI: 10.1016/j.neubiorev.2024.105559] [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: 11/29/2023] [Revised: 01/16/2024] [Accepted: 01/17/2024] [Indexed: 01/23/2024]
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder that impacts a variety of cognitive and behavioral domains. While a genetic component of ASD has been well-established, none of the numerous syndromic genes identified in humans accounts for more than 1% of the clinical patients. Due to this large number of target genes, numerous mouse models of the disorder have been generated. However, the focus on distinct brain circuits, behavioral phenotypes and diverse experimental approaches has made it difficult to synthesize the overwhelming number of model animal studies into concrete throughlines that connect the data across levels of investigation. Here we chose to focus on one circuit, the hippocampus, and one hypothesis, a shift in excitatory/inhibitory balance, to examine, from the level of the tripartite synapse up to the level of in vivo circuit activity, the key commonalities across disparate models that can illustrate a path towards a better mechanistic understanding of ASD's impact on hippocampal circuit function.
Collapse
Affiliation(s)
- Leandra Severino
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, South Korea; Division of Bio-Medical Science & Technology, KIST-School, University of Science and Technology, Seoul, South Korea
| | - Jinhyun Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, South Korea; Division of Bio-Medical Science & Technology, KIST-School, University of Science and Technology, Seoul, South Korea
| | - Min-Ho Nam
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, South Korea; Division of Bio-Medical Science & Technology, KIST-School, University of Science and Technology, Seoul, South Korea.
| | - Thomas J McHugh
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, South Korea; Laboratory for Circuit and Behavioral Physiology, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako-shi Saitama, Japan.
| |
Collapse
|
7
|
Sands TT, Gelinas JN. Epilepsy and Encephalopathy. Pediatr Neurol 2024; 150:24-31. [PMID: 37948790 DOI: 10.1016/j.pediatrneurol.2023.09.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 09/14/2023] [Accepted: 09/24/2023] [Indexed: 11/12/2023]
Abstract
BACKGROUND Epilepsy encompasses more than the predisposition to unprovoked seizures. In children, epileptic activity during (ictal) and between (interictal) seizures has the potential to disrupt normal brain development. The term "epileptic encephalopathy (EE)" refers to the concept that such abnormal activity may contribute to cognitive and behavioral impairments beyond that expected from the underlying cause of the epileptic activity. METHODS In this review, we survey the concept of EE across a diverse selection of syndromes to illustrate its broad applicability in pediatric epilepsy. We review experimental evidence that provides mechanistic insights into how epileptic activity has the potential to impact normal brain processes and the development of neural networks. We then discuss opportunities to improve developmental outcomes in epilepsy now and in the future. RESULTS Epileptic activity in the brain poses a threat to normal physiology and brain development. CONCLUSION Until we have treatments that reliably target and effectively treat the underlying causes of epilepsy, a major goal of management is to prevent epileptic activity from worsening developmental outcomes.
Collapse
Affiliation(s)
- Tristan T Sands
- Center for Translational Research in Neurodevelopmental Disease, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York; Departments of Neurology and Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York.
| | - Jennifer N Gelinas
- Center for Translational Research in Neurodevelopmental Disease, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York; Departments of Neurology and Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York
| |
Collapse
|
8
|
Lax E, Do Carmo S, Enuka Y, Sapozhnikov DM, Welikovitch LA, Mahmood N, Rabbani SA, Wang L, Britt JP, Hancock WW, Yarden Y, Szyf M. Methyl-CpG binding domain 2 (Mbd2) is an epigenetic regulator of autism-risk genes and cognition. Transl Psychiatry 2023; 13:259. [PMID: 37443311 PMCID: PMC10344909 DOI: 10.1038/s41398-023-02561-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 06/29/2023] [Accepted: 07/04/2023] [Indexed: 07/15/2023] Open
Abstract
The Methyl-CpG-Binding Domain Protein family has been implicated in neurodevelopmental disorders. The Methyl-CpG-binding domain 2 (Mbd2) binds methylated DNA and was shown to play an important role in cancer and immunity. Some evidence linked this protein to neurodevelopment. However, its exact role in neurodevelopment and brain function is mostly unknown. Here we show that Mbd2-deficiency in mice (Mbd2-/-) results in deficits in cognitive, social and emotional functions. Mbd2 binds regulatory DNA regions of neuronal genes in the hippocampus and loss of Mbd2 alters the expression of hundreds of genes with a robust down-regulation of neuronal gene pathways. Further, a genome-wide DNA methylation analysis found an altered DNA methylation pattern in regulatory DNA regions of neuronal genes in Mbd2-/- mice. Differentially expressed genes significantly overlap with gene-expression changes observed in brains of Autism Spectrum Disorder (ASD) individuals. Notably, downregulated genes are significantly enriched for human ortholog ASD risk genes. Observed hippocampal morphological abnormalities were similar to those found in individuals with ASD and ASD rodent models. Hippocampal Mbd2 knockdown partially recapitulates the behavioral phenotypes observed in Mbd2-/- mice. These findings suggest that Mbd2 is a novel epigenetic regulator of genes that are associated with ASD in humans. Mbd2 loss causes behavioral alterations that resemble those found in ASD individuals.
Collapse
Affiliation(s)
- Elad Lax
- Department of Molecular Biology, Ariel University, Ariel, Israel.
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada.
| | - Sonia Do Carmo
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Yehoshua Enuka
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Daniel M Sapozhnikov
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Lindsay A Welikovitch
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, 02129, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Niaz Mahmood
- Department of Medicine, McGill University Health Center, Montreal, QC, Canada
- Department of Biochemistry, McGill University, Montreal, QC, Canada
| | - Shafaat A Rabbani
- Department of Medicine, McGill University Health Center, Montreal, QC, Canada
| | - Liqing Wang
- Division of Transplant Immunology, Department of Pathology and Laboratory Medicine, and Biesecker Center for Pediatric Liver Diseases, Children's Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jonathan P Britt
- Department of Psychology, McGill University, Montreal, QC, Canada
| | - Wayne W Hancock
- Division of Transplant Immunology, Department of Pathology and Laboratory Medicine, and Biesecker Center for Pediatric Liver Diseases, Children's Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yosef Yarden
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Moshe Szyf
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| |
Collapse
|
9
|
Forrest MP, Dos Santos M, Piguel NH, Wang YZ, Hawkins NA, Bagchi VA, Dionisio LE, Yoon S, Simkin D, Martin-de-Saavedra MD, Gao R, Horan KE, George AL, LeDoux MS, Kearney JA, Savas JN, Penzes P. Rescue of neuropsychiatric phenotypes in a mouse model of 16p11.2 duplication syndrome by genetic correction of an epilepsy network hub. Nat Commun 2023; 14:825. [PMID: 36808153 PMCID: PMC9938216 DOI: 10.1038/s41467-023-36087-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 01/16/2023] [Indexed: 02/19/2023] Open
Abstract
Neuropsychiatric disorders (NPDs) are frequently co-morbid with epilepsy, but the biological basis of shared risk remains poorly understood. The 16p11.2 duplication is a copy number variant that confers risk for diverse NPDs including autism spectrum disorder, schizophrenia, intellectual disability and epilepsy. We used a mouse model of the 16p11.2 duplication (16p11.2dup/+) to uncover molecular and circuit properties associated with this broad phenotypic spectrum, and examined genes within the locus capable of phenotype reversal. Quantitative proteomics revealed alterations to synaptic networks and products of NPD risk genes. We identified an epilepsy-associated subnetwork that was dysregulated in 16p11.2dup/+ mice and altered in brain tissue from individuals with NPDs. Cortical circuits from 16p11.2dup/+ mice exhibited hypersynchronous activity and enhanced network glutamate release, which increased susceptibility to seizures. Using gene co-expression and interactome analysis, we show that PRRT2 is a major hub in the epilepsy subnetwork. Remarkably, correcting Prrt2 copy number rescued aberrant circuit properties, seizure susceptibility and social deficits in 16p11.2dup/+ mice. We show that proteomics and network biology can identify important disease hubs in multigenic disorders, and reveal mechanisms relevant to the complex symptomatology of 16p11.2 duplication carriers.
Collapse
Affiliation(s)
- Marc P Forrest
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
- Center for Autism and Neurodevelopment, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Marc Dos Santos
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
- Center for Autism and Neurodevelopment, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Nicolas H Piguel
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
- Center for Autism and Neurodevelopment, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Yi-Zhi Wang
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Nicole A Hawkins
- Department of Pharmacology Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Vikram A Bagchi
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
- Center for Autism and Neurodevelopment, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Leonardo E Dionisio
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
- Center for Autism and Neurodevelopment, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Sehyoun Yoon
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
- Center for Autism and Neurodevelopment, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Dina Simkin
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
- Department of Pharmacology Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Maria Dolores Martin-de-Saavedra
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
- Center for Autism and Neurodevelopment, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
- Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad Complutense, 28040, Madrid, Spain
| | - Ruoqi Gao
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
- Center for Autism and Neurodevelopment, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Katherine E Horan
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
- Center for Autism and Neurodevelopment, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Alfred L George
- Center for Autism and Neurodevelopment, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
- Department of Pharmacology Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Mark S LeDoux
- Department of Psychology, University of Memphis, Memphis, TN, 38152, USA
- Veracity Neuroscience LLC, Memphis, TN, 38157, USA
| | - Jennifer A Kearney
- Center for Autism and Neurodevelopment, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
- Department of Pharmacology Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Jeffrey N Savas
- Center for Autism and Neurodevelopment, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Peter Penzes
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA.
- Center for Autism and Neurodevelopment, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA.
| |
Collapse
|
10
|
Loss of Rai1 enhances hippocampal excitability and epileptogenesis in mouse models of Smith-Magenis syndrome. Proc Natl Acad Sci U S A 2022; 119:e2210122119. [PMID: 36256819 PMCID: PMC9618093 DOI: 10.1073/pnas.2210122119] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Smith–Magenis syndrome (SMS) is a neurodevelopmental disorder associated with autism and epileptic seizures. SMS is caused by losing one copy of the gene encoding retinoic acid induced 1 (RAI1), a ubiquitously expressed transcriptional regulator. To pinpoint brain regions and cell types contributing to neuronal hyperexcitability in SMS, we combined electrophysiology and three-dimensional imaging of Fos expression in the intact mouse brain. We found that Rai1-deficient hippocampal dentate gyrus granule cells (dGCs) show increased intrinsic excitability and enhanced glutamatergic synaptic transmission. Our findings indicate that Rai1 safeguards the hippocampal network from hyperexcitability and could help explain abnormal brain activity in SMS. Hyperexcitability of brain circuits is a common feature of autism spectrum disorders (ASDs). Genetic deletion of a chromatin-binding protein, retinoic acid induced 1 (RAI1), causes Smith–Magenis syndrome (SMS). SMS is a syndromic ASD associated with intellectual disability, autistic features, maladaptive behaviors, overt seizures, and abnormal electroencephalogram (EEG) patterns. The molecular and neural mechanisms underlying abnormal brain activity in SMS remain unclear. Here we show that panneural Rai1 deletions in mice result in increased seizure susceptibility and prolonged hippocampal seizure duration in vivo and increased dentate gyrus population spikes ex vivo. Brain-wide mapping of neuronal activity pinpointed selective cell types within the limbic system, including the hippocampal dentate gyrus granule cells (dGCs) that are hyperactivated by chemoconvulsant administration or sensory experience in Rai1-deficient brains. Deletion of Rai1 from glutamatergic neurons, but not from gamma-aminobutyric acidergic (GABAergic) neurons, was responsible for increased seizure susceptibility. Deleting Rai1 from the Emx1Cre-lineage glutamatergic neurons resulted in abnormal dGC properties, including increased excitatory synaptic transmission and increased intrinsic excitability. Our work uncovers the mechanism of neuronal hyperexcitability in SMS by identifying Rai1 as a negative regulator of dGC intrinsic and synaptic excitability.
Collapse
|
11
|
Yue Y, Ash RT, Boyle N, Kinter A, Li Y, Zeng C, Lu H. MeCP2 deficiency impairs motor cortical circuit flexibility associated with motor learning. Mol Brain 2022; 15:76. [PMID: 36064580 PMCID: PMC9446698 DOI: 10.1186/s13041-022-00965-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 08/29/2022] [Indexed: 11/19/2022] Open
Abstract
Loss of function mutations in the X-linked gene encoding methyl-CpG binding protein 2 (MECP2) cause Rett syndrome (RTT), a postnatal neurological disorder. The loss of motor function is an important clinical feature of RTT that manifests early during the course of the disease. RTT mouse models with mutations in the murine orthologous Mecp2 gene replicate many human phenotypes, including progressive motor impairments. However, relatively little is known about the changes in circuit function during the progression of motor deficit in this model. As the motor cortex is the key node in the motor system for the control of voluntary movement, we measured firing activity in populations of motor cortical neurons during locomotion on a motorized wheel-treadmill. Different populations of neurons intermingled in the motor cortex signal different aspects of the locomotor state of the animal. The proportion of running selective neurons whose activity positively correlates with locomotion speed gradually decreases with weekly training in wild-type mice, but not in Mecp2-null mice. The fraction of rest-selective neurons whose activity negatively correlates with locomotion speed does not change with training in wild-type mice, but is higher and increases with the progression of locomotion deficit in mutant mice. The synchronization of population activity that occurs in WT mice with training did not occur in Mecp2-null mice, a phenotype most clear during locomotion and observable across all functional cell types. Our results could represent circuit-level biomarkers for motor regression in Rett syndrome.
Collapse
Affiliation(s)
- Yuanlei Yue
- grid.253615.60000 0004 1936 9510Department of Pharmacology and Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037 USA
| | - Ryan T. Ash
- grid.168010.e0000000419368956Department of Psychiatry, Stanford University, Palo Alto, CA 94305 USA
| | - Natalie Boyle
- grid.253615.60000 0004 1936 9510Department of Pharmacology and Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037 USA
| | - Anna Kinter
- grid.253615.60000 0004 1936 9510Department of Pharmacology and Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037 USA
| | - Yipeng Li
- grid.253615.60000 0004 1936 9510Department of Pharmacology and Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037 USA
| | - Chen Zeng
- grid.253615.60000 0004 1936 9510Department of Physics, Columbian College of Arts and Sciences, The George Washington, University, Washington, DC 20037 USA
| | - Hui Lu
- Department of Pharmacology and Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, DC, 20037, USA.
| |
Collapse
|
12
|
Xu M, Qi S, Calhoun V, Dai J, Yu B, Zhang K, Pei M, Li C, Wei Y, Jiang R, Zhi D, Huang Z, Qiu Z, Liang Z, Sui J. Aberrant brain functional and structural developments in MECP2 duplication rats. Neurobiol Dis 2022; 173:105838. [PMID: 35985556 PMCID: PMC9631682 DOI: 10.1016/j.nbd.2022.105838] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/22/2022] [Accepted: 08/11/2022] [Indexed: 12/02/2022] Open
Abstract
Transgenic animal models with homologous etiology provide a promising way to pursue the neurobiological substrates of the behavioral deficits in autism spectrum disorder (ASD). Gain-of-function mutations of MECP2 cause MECP2 duplication syndrome, a severe neurological disorder with core symptoms of ASD. However, abnormal brain developments underlying the autistic-like behavioral deficits of MECP2 duplication syndrome are rarely investigated. To this end, a human MECP2 duplication (MECP2-DP) rat model was created by the bacterial artificial chromosome transgenic method. Functional and structural magnetic resonance imaging (MRI) with high-field were performed on 16 male MECP2-DP rats and 15 male wildtype rats at postnatal 28 days, 42 days, and 56 days old. Multimodal fusion analyses guided by locomotor-relevant metrics and social novelty time separately were applied to identify abnormal brain networks associated with diverse behavioral deficits induced by MECP2 duplication. Aberrant functional developments of a core network primarily composed of the dorsal medial prefrontal cortex (dmPFC) and retrosplenial cortex (RSP) were detected to associate with diverse behavioral phenotypes in MECP2-DP rats. Altered developments of gray matter volume were detected in the hippocampus and thalamus. We conclude that gain-of-function mutations of MECP2 induce aberrant functional activities in the default-mode-like network and aberrant volumetric changes in the brain, resulting in autistic-like behavioral deficits. Our results gain critical insights into the biomarker of MECP2 duplication syndrome and the neurobiological underpinnings of the behavioral deficits in ASD.
Collapse
Affiliation(s)
- Ming Xu
- Brainnetome Center and National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China; School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, 100190, China
| | - Shile Qi
- College of Computer Science and Technology, Nanjing University of Aeronautics and Astronautics, Nanjing 211106, China
| | - Vince Calhoun
- Tri-institutional Center for Translational Research in Neuroimaging and Data Science (TReNDS), Georgia Institute of Technology, Georgia State University, Emory University, Atlanta, GA 30303, USA
| | - Jiankun Dai
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, State Key Laboratory of Neuroscience, CAS Key Laboratory of Primate Neurobiology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Bin Yu
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, State Key Laboratory of Neuroscience, CAS Key Laboratory of Primate Neurobiology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Kaiwei Zhang
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, State Key Laboratory of Neuroscience, CAS Key Laboratory of Primate Neurobiology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Mengchao Pei
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, State Key Laboratory of Neuroscience, CAS Key Laboratory of Primate Neurobiology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Chenjian Li
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Peking University School of Life Sciences, Beijing 100871, China; PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, Beijing 100871, China
| | - Yusheng Wei
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Peking University School of Life Sciences, Beijing 100871, China; PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, Beijing 100871, China
| | - Rongtao Jiang
- Brainnetome Center and National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Dongmei Zhi
- Brainnetome Center and National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China; School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, 100190, China
| | - Zhimin Huang
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Peking University School of Life Sciences, Beijing 100871, China; PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, Beijing 100871, China
| | - Zilong Qiu
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, State Key Laboratory of Neuroscience, CAS Key Laboratory of Primate Neurobiology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Zhifeng Liang
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, State Key Laboratory of Neuroscience, CAS Key Laboratory of Primate Neurobiology, Chinese Academy of Sciences, Shanghai 200031, China.
| | - Jing Sui
- IDG/McGovern Institute for Brain Research, State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China.
| |
Collapse
|
13
|
Ash RT, Palagina G, Fernandez-Leon JA, Park J, Seilheimer R, Lee S, Sabharwal J, Reyes F, Wang J, Lu D, Sarfraz M, Froudarakis E, Tolias AS, Wu SM, Smirnakis SM. Increased Reliability of Visually-Evoked Activity in Area V1 of the MECP2-Duplication Mouse Model of Autism. J Neurosci 2022; 42:6469-6482. [PMID: 35831173 PMCID: PMC9398540 DOI: 10.1523/jneurosci.0654-22.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 05/15/2022] [Accepted: 06/02/2022] [Indexed: 11/21/2022] Open
Abstract
Atypical sensory processing is now thought to be a core feature of the autism spectrum. Influential theories have proposed that both increased and decreased neural response reliability within sensory systems could underlie altered sensory processing in autism. Here, we report evidence for abnormally increased reliability of visual-evoked responses in layer 2/3 neurons of adult male and female primary visual cortex in the MECP2-duplication syndrome animal model of autism. Increased response reliability was due in part to decreased response amplitude, decreased fluctuations in endogenous activity, and an abnormal decoupling of visual-evoked activity from endogenous activity. Similar to what was observed neuronally, the optokinetic reflex occurred more reliably at low contrasts in mutant mice compared with controls. Retinal responses did not explain our observations. These data suggest that the circuit mechanisms for combining sensory-evoked and endogenous signal and noise processes may be altered in this form of syndromic autism.SIGNIFICANCE STATEMENT Atypical sensory processing is now thought to be a core feature of the autism spectrum. Influential theories have proposed that both increased and decreased neural response reliability within sensory systems could underlie altered sensory processing in autism. Here, we report evidence for abnormally increased reliability of visual-evoked responses in primary visual cortex of the animal model for MECP2-duplication syndrome, a high-penetrance single-gene cause of autism. Visual-evoked activity was abnormally decoupled from endogenous activity in mutant mice, suggesting in line with the influential "hypo-priors" theory of autism that sensory priors embedded in endogenous activity may have less influence on perception in autism.
Collapse
Affiliation(s)
- Ryan T Ash
- Department of Psychiatry, Stanford University School of Medicine, Stanford, California 94305
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030
| | - Ganna Palagina
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Jose A Fernandez-Leon
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030
- Centro de Investigaciones en Física e Ingeniería del Centro de la Provincia de Buenos Aires and Instituto de Investigación en Tecnología Informática Avanzada, Exact Sciences Faculty-Universidad Nacional del Centro de la Provincia de Buenos Aires, Tandil, Argentina
| | - Jiyoung Park
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115
- Program in Structural and Computational Biology and Molecular Biophysics, Baylor College of Medicine, Houston, Texas 77030
| | - Rob Seilheimer
- Program in Structural and Computational Biology and Molecular Biophysics, Baylor College of Medicine, Houston, Texas 77030
- Department of Psychiatry, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Sangkyun Lee
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Jasdeep Sabharwal
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030
- Wilmer Eye Institute, Johns Hopkins University, Baltimore, Maryland 21205
| | - Fredy Reyes
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030
| | - Jing Wang
- Department of Ophthalmology, Baylor College of Medicine, Houston, Texas 77030
| | - Dylan Lu
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Muhammad Sarfraz
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Emmanouil Froudarakis
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030
- FORTH, Heraklion, Crete, Greece 70013
| | - Andreas S Tolias
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030
| | - Samuel M Wu
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030
- Department of Ophthalmology, Baylor College of Medicine, Houston, Texas 77030
| | - Stelios M Smirnakis
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030
| |
Collapse
|
14
|
Horvath PM, Piazza MK, Kavalali ET, Monteggia LM. MeCP2 loss-of-function dysregulates microRNAs regionally and disrupts excitatory/inhibitory synaptic transmission balance. Hippocampus 2022; 32:610-623. [PMID: 35851733 PMCID: PMC9344394 DOI: 10.1002/hipo.23455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 06/11/2022] [Accepted: 06/25/2022] [Indexed: 11/06/2022]
Abstract
Rett syndrome is a leading cause of intellectual disability in females primarily caused by loss of function mutations in the transcriptional regulator MeCP2. Loss of MeCP2 leads to a host of synaptic phenotypes that are believed to underlie Rett syndrome pathophysiology. Synaptic deficits vary by brain region upon MeCP2 loss, suggesting distinct molecular alterations leading to disparate synaptic outcomes. In this study, we examined the contribution of MeCP2's newly described role in miRNA regulation to regional molecular and synaptic impairments. Two miRNAs, miR-101a and miR-203, were identified and confirmed as upregulated in MeCP2 KO mice in the hippocampus and cortex, respectively. miR-101a overexpression in hippocampal cultures led to opposing effects at excitatory and inhibitory synapses and in spontaneous and evoked neurotransmission, revealing the potential for a single miRNA to broadly regulate synapse function in the hippocampus. These results highlight the importance of regional alterations in miRNA expression and the specific impact on synaptic function with potential implications for Rett syndrome.
Collapse
Affiliation(s)
- Patricia M. Horvath
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA,Department of Neuroscience, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Michelle K. Piazza
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee, USA,Neuroscience Program, Vanderbilt University, Nashville, Tennessee, USA
| | - Ege T. Kavalali
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA,Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee, USA
| | - Lisa M. Monteggia
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA,Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee, USA
| |
Collapse
|
15
|
Lu S, Chen Y, Wang Z. Advances in the pathogenesis of Rett syndrome using cell models. Animal Model Exp Med 2022; 5:532-541. [PMID: 35785421 PMCID: PMC9773312 DOI: 10.1002/ame2.12236] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 05/05/2022] [Indexed: 12/30/2022] Open
Abstract
Rett syndrome (RTT) is a progressive neurodevelopmental disorder that occurs mainly in girls with a range of typical symptoms of autism spectrum disorders. MeCP2 protein loss-of-function in neural lineage cells is the main cause of RTT pathogenicity. As it is still hard to understand the mechanism of RTT on the basis of only clinical patients or animal models, cell models cultured in vitro play indispensable roles. Here we reviewed the research progress in the pathogenesis of RTT at the cellular level, summarized the preclinical-research-related applications, and prospected potential future development.
Collapse
Affiliation(s)
- Sijia Lu
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational MedicineKunming University of Science and TechnologyKunmingChina,Yunnan Key Laboratory of Primate Biomedical ResearchKunmingChina
| | - Yongchang Chen
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational MedicineKunming University of Science and TechnologyKunmingChina,Yunnan Key Laboratory of Primate Biomedical ResearchKunmingChina
| | - Zhengbo Wang
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational MedicineKunming University of Science and TechnologyKunmingChina,Yunnan Key Laboratory of Primate Biomedical ResearchKunmingChina
| |
Collapse
|
16
|
Choi M, Ko SY, Seo JY, Kim DG, Lee H, Chung H, Son H. Autistic-like social deficits in hippocampal MeCP2 knockdown rat models are rescued by ketamine. BMB Rep 2022. [PMID: 35410641 PMCID: PMC9152577 DOI: 10.5483/bmbrep.2022.55.5.038] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Autism or autism spectrum disorder (ASD) is a behavioral syndrome characterized by persistent deficits in social interaction, and repetitive patterns of behavior, interests, or activities. The gene encoding Methyl-CpG binding protein 2 (MeCP2) is one of a few exceptional genes of established causal effect in ASD. Although genetically engineered mice studies may shed light on how MeCP2 loss affects synaptic activity patterns across the whole brain, such studies are not considered practical in ASD patients due to the overall level of impairment, and are technically challenging in mice. For the first time, we show that hippo-campal MeCP2 knockdown produces behavioral abnormalities associated with autism-like traits in rats, providing a new strategy to investigate the efficacy of therapeutics in ASD. Ketamine, an N-Methyl-D-aspartate (NMDA) blocker, has been proposed as a possible treatment for autism. Using the MeCP2 knockdown rats in conjunction with a rat model of valproic acid (VPA)-induced ASD, we examined gene expression and ASD behaviors upon ketamine treatment. We report that the core symptoms of autism in MeCP2 knockdown rats with social impairment recovered dramatically following a single treatment with ketamine.
Collapse
Affiliation(s)
- Miyeon Choi
- Hanyang Biomedical Research Institute, Hanyang University, Seoul 04763, Korea
| | - Seung Yeon Ko
- Hanyang Biomedical Research Institute, Hanyang University, Seoul 04763, Korea
| | - Jee Young Seo
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, Korea
| | - Do Gyeong Kim
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, Korea
| | - Huiju Lee
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, Korea
| | - Heekyoung Chung
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, Korea
| | - Hyeon Son
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, Korea
- Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul 04763, Korea
| |
Collapse
|
17
|
He L, Caudill MS, Jing J, Wang W, Sun Y, Tang J, Jiang X, Zoghbi HY. A weakened recurrent circuit in the hippocampus of Rett syndrome mice disrupts long-term memory representations. Neuron 2022; 110:1689-1699.e6. [PMID: 35290792 PMCID: PMC9930308 DOI: 10.1016/j.neuron.2022.02.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 12/30/2021] [Accepted: 02/16/2022] [Indexed: 02/02/2023]
Abstract
Successful recall of a contextual memory requires reactivating ensembles of hippocampal cells that were allocated during memory formation. Altering the ratio of excitation-to-inhibition (E/I) during memory retrieval can bias cell participation in an ensemble and hinder memory recall. In the case of Rett syndrome (RTT), a neurological disorder with severe learning and memory deficits, the E/I balance is altered, but the source of this imbalance is unknown. Using in vivo imaging during an associative memory task, we show that during long-term memory retrieval, RTT CA1 cells poorly distinguish mnemonic context and form larger ensembles than wild-type mouse cells. Simultaneous multiple whole-cell recordings revealed that mutant somatostatin expressing interneurons (SOM) are poorly recruited by CA1 pyramidal cells and are less active during long-term memory retrieval in vivo. Chemogenetic manipulation revealed that reduced SOM activity underlies poor long-term memory recall. Our findings reveal a disrupted recurrent CA1 circuit contributing to RTT memory impairment.
Collapse
Affiliation(s)
- Lingjie He
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA; Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX, USA
| | - Matthew S Caudill
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA; Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Junzhan Jing
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA; Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Wei Wang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA
| | - Yaling Sun
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA; Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX, USA
| | - Jianrong Tang
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA; Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Xiaolong Jiang
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA; Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Huda Y Zoghbi
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA; Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA; Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA; Department of Neurology, Baylor College of Medicine, Houston, TX, USA; Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
18
|
Davidian D, LeGro M, Barghouth PG, Rojas S, Ziman B, Maciel EI, Ardell D, Escobar AL, Oviedo NJ. Restoration of DNA integrity and cell cycle by electric stimulation in planarian tissues damaged by ionizing radiation. J Cell Sci 2022; 135:274829. [PMID: 35322853 PMCID: PMC9264365 DOI: 10.1242/jcs.259304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 03/05/2022] [Indexed: 10/18/2022] Open
Abstract
Exposure to high levels of ionizing γ-radiation leads to irreversible DNA damage and cell death. Here, we establish that exogenous application of electric stimulation enables cellular plasticity to reestablish stem cell activity in tissues damaged by ionizing radiation. We show that sub-threshold direct current stimulation (DCS) rapidly restores pluripotent stem cell populations previously eliminated by lethally γ-irradiated tissues of the planarian flatworm Schmidtea mediterranea. Our findings reveal that DCS enhances DNA repair, transcriptional activity, and cell cycle entry in post-mitotic cells. These responses involve rapid increases in cytosolic [Ca2+] through the activation of L-type Cav channels and intracellular Ca2+ stores leading to the activation of immediate early genes and ectopic expression of stem cell markers in postmitotic cells. Overall, we show the potential of electric current stimulation to reverse the damaging effects of high dose γ-radiation in adult tissues. Furthermore, our results provide mechanistic insights describing how electric stimulation effectively translates into molecular responses capable of regulating fundamental cellular functions without the need for genetic or pharmacological intervention.
Collapse
Affiliation(s)
- Devon Davidian
- Department of Molecular & Cell Biology, University of California, Merced, USA.,Quantitative and Systems Biology Graduate Program, University of California, Merced, USA
| | - Melanie LeGro
- Department of Molecular & Cell Biology, University of California, Merced, USA.,Quantitative and Systems Biology Graduate Program, University of California, Merced, USA
| | - Paul G Barghouth
- Department of Molecular & Cell Biology, University of California, Merced, USA.,Quantitative and Systems Biology Graduate Program, University of California, Merced, USA
| | - Salvador Rojas
- Department of Molecular & Cell Biology, University of California, Merced, USA.,Quantitative and Systems Biology Graduate Program, University of California, Merced, USA
| | - Benjamin Ziman
- Department of Molecular & Cell Biology, University of California, Merced, USA.,Quantitative and Systems Biology Graduate Program, University of California, Merced, USA
| | - Eli Isael Maciel
- Department of Molecular & Cell Biology, University of California, Merced, USA.,Quantitative and Systems Biology Graduate Program, University of California, Merced, USA
| | - David Ardell
- Department of Molecular & Cell Biology, University of California, Merced, USA.,Health Sciences Research Institute, University of California, Merced, USA
| | - Ariel L Escobar
- Department of Bioengineering, University of California, Merced, USA.,Health Sciences Research Institute, University of California, Merced, USA
| | - Néstor J Oviedo
- Department of Molecular & Cell Biology, University of California, Merced, USA.,Health Sciences Research Institute, University of California, Merced, USA
| |
Collapse
|
19
|
Li W. Excitation and Inhibition Imbalance in Rett Syndrome. Front Neurosci 2022; 16:825063. [PMID: 35250460 PMCID: PMC8894599 DOI: 10.3389/fnins.2022.825063] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 01/31/2022] [Indexed: 12/12/2022] Open
Abstract
A loss of the excitation/inhibition (E/I) balance in the neural circuit has emerged as a common neuropathological feature in many neurodevelopmental disorders. Rett syndrome (RTT), a prevalent neurodevelopmental disorder that affects 1:10,000-15,000 women globally, is caused by loss-of-function mutations in the Methyl-CpG-binding Protein-2 (Mecp2) gene. E/I imbalance is recognized as the leading cellular and synaptic hallmark that is fundamental to diverse RTT neurological symptoms, including stereotypic hand movements, impaired motor coordination, breathing irregularities, seizures, and learning/memory dysfunctions. E/I balance in RTT is not homogeneously altered but demonstrates brain region and cell type specificity instead. In this review, I elaborate on the current understanding of the loss of E/I balance in a range of brain areas at molecular and cellular levels. I further describe how the underlying cellular mechanisms contribute to the disturbance of the proper E/I ratio. Last, I discuss current pharmacologic innervations for RTT and their role in modifying the E/I balance.
Collapse
Affiliation(s)
- Wei Li
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
20
|
Astrocytic Gap Junctions Contribute to Aberrant Neuronal Synchronization in a Mouse Model of MeCP2 Duplication Syndrome. Neurosci Bull 2022; 38:591-606. [PMID: 35147909 DOI: 10.1007/s12264-022-00824-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 12/07/2021] [Indexed: 10/19/2022] Open
Abstract
Abnormal synchronous neuronal activity has been widely detected by brain imaging of autistic patients, but its underlying neural mechanism remains unclear. Compared with wild-type mice, our in vivo two-photon imaging showed that transgenic (Tg1) mice over-expressing human autism risk gene MeCP2 exhibited higher neuronal synchrony in the young but lower synchrony in the adult stage. Whole-cell recording of neuronal pairs in brain slices revealed that higher neuronal synchrony in young postnatal Tg1 mice was attributed mainly to more prevalent giant slow inward currents (SICs). Both in vivo and slice imaging further demonstrated more dynamic activity and higher synchrony in astrocytes from young Tg1 mice. Blocking astrocytic gap junctions markedly decreased the generation of SICs and overall cell synchrony in the Tg1 brain. Furthermore, the expression level of Cx43 protein and the coupling efficiency of astrocyte gap junctions remained unchanged in Tg1 mice. Thus, astrocytic gap junctions facilitate but do not act as a direct trigger for the abnormal neuronal synchrony in young Tg1 mice, revealing the potential role of the astrocyte network in the pathogenesis of MeCP2 duplication syndrome.
Collapse
|
21
|
Abstract
Deep brain stimulation (DBS) entails neurosurgery to implant electrodes in specific brain structures to modulate the behavior of a particular neural circuit. DBS is best known for treating advanced Parkinson disease and can potentially be applicable to other motor and even cognitive dysfunctions. Here, we describe a detailed protocol allowing for electrode preparation, surgical procedures, stimulation delivery, and field potential recordings in both anesthetized and behaving mice, and the benefit evaluation of DBS at the fimbria-fornix by using a fear conditioning test. For complete details on the use and execution of this protocol, please refer to Hao et al. (2015).
Collapse
Affiliation(s)
- Qi Wang
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA,Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX 77030, USA
| | - Bin Tang
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA,Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX 77030, USA
| | - Jianrong Tang
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA,Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX 77030, USA,Corresponding author
| |
Collapse
|
22
|
Contractor A, Ethell IM, Portera-Cailliau C. Cortical interneurons in autism. Nat Neurosci 2021; 24:1648-1659. [PMID: 34848882 PMCID: PMC9798607 DOI: 10.1038/s41593-021-00967-6] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 09/21/2021] [Indexed: 01/01/2023]
Abstract
The mechanistic underpinnings of autism remain a subject of debate and controversy. Why do individuals with autism share an overlapping set of atypical behaviors and symptoms, despite having different genetic and environmental risk factors? A major challenge in developing new therapies for autism has been the inability to identify convergent neural phenotypes that could explain the common set of symptoms that result in the diagnosis. Although no striking macroscopic neuropathological changes have been identified in autism, there is growing evidence that inhibitory interneurons (INs) play an important role in its neural basis. In this Review, we evaluate and interpret this evidence, focusing on recent findings showing reduced density and activity of the parvalbumin class of INs. We discuss the need for additional studies that investigate how genes and the environment interact to change the developmental trajectory of INs, permanently altering their numbers, connectivity and circuit engagement.
Collapse
Affiliation(s)
- Anis Contractor
- Department of Neuroscience Feinberg School of Medicine, Weinberg College of Arts and Sciences, Northwestern University, Chicago, IL, USA
- Department of Neurobiology, Weinberg College of Arts and Sciences, Northwestern University, Chicago, IL, USA
| | - Iryna M Ethell
- Division of Biomedical Sciences, UC Riverside School of Medicine, Riverside, CA, USA
| | - Carlos Portera-Cailliau
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.
- Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.
| |
Collapse
|
23
|
Hao S, Wang Q, Tang B, Wu Z, Yang T, Tang J. CDKL5 Deficiency Augments Inhibitory Input into the Dentate Gyrus That Can Be Reversed by Deep Brain Stimulation. J Neurosci 2021; 41:9031-9046. [PMID: 34544833 PMCID: PMC8549531 DOI: 10.1523/jneurosci.1010-21.2021] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 07/16/2021] [Accepted: 08/16/2021] [Indexed: 12/12/2022] Open
Abstract
Cognitive impairment is a core feature of cyclin-dependent kinase-like 5 (CDKL5) deficiency, a neurodevelopmental disorder characterized by early epileptic seizures, intellectual disability, and autistic behaviors. Although loss of CDKL5 affects a number of molecular pathways, very little has been discovered about the physiological effects of these changes on the neural circuitry. We therefore studied synaptic plasticity and local circuit activity in the dentate gyrus of both Cdkl5-/y and Cdkl5+/- mutant mice. We found that CDKL5 haploinsufficiency in both male and female mice impairs hippocampus-dependent learning and memory in multiple tasks. In vivo, loss of CDKL5 reduced LTP of the perforant path to the dentate gyrus and augmented feedforward inhibition in this pathway; ex vivo experiments confirmed that excitatory/inhibitory input into the dentate gyrus is skewed toward inhibition. Injecting the GABAergic antagonist gabazine into the dentate improved contextual fear memory in Cdkl5-/y mice. Finally, chronic forniceal deep brain stimulation rescued hippocampal memory deficits, restored synaptic plasticity, and relieved feedforward inhibition in Cdkl5+/- mice. These results indicate that CDKL5 is important for maintaining proper dentate excitatory/inhibitory balance, with consequences for hippocampal memory.SIGNIFICANCE STATEMENT Cognitive impairment is a core feature of cyclin-dependent kinase-like 5 (CDKL5) deficiency disorder. Although CDKL5 deficiency has been found to affect a number of molecular pathways, little is known about its physiological effects on the neural circuitry. We find that CDKL5 loss reduces hippocampal synaptic plasticity and augments feedforward inhibition in the perforant path to the dentate gyrus in vivo in Cdkl5 mutant mice. Chronic forniceal deep brain stimulation rescued hippocampal memory deficits, restored synaptic plasticity, and relieved feedforward inhibition in Cdkl5+/- mice, as it had previously done with Rett syndrome mice, suggesting that such stimulation may be useful for other neurodevelopmental disorders.
Collapse
Affiliation(s)
- Shuang Hao
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas 77030
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas 77030
| | - Qi Wang
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas 77030
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas 77030
| | - Bin Tang
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas 77030
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas 77030
| | - Zhenyu Wu
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas 77030
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas 77030
| | - Tingting Yang
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas 77030
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas 77030
- Department of Neurology, People's Hospital of Guizhou Province, Guiyang, 560000, China
| | - Jianrong Tang
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas 77030
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas 77030
| |
Collapse
|
24
|
Yue Y, Xu P, Liu Z, Sun X, Su J, Du H, Chen L, Ash RT, Smirnakis S, Simha R, Kusner L, Zeng C, Lu H. Motor training improves coordination and anxiety in symptomatic Mecp2-null mice despite impaired functional connectivity within the motor circuit. SCIENCE ADVANCES 2021; 7:eabf7467. [PMID: 34678068 PMCID: PMC8535852 DOI: 10.1126/sciadv.abf7467] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 09/01/2021] [Indexed: 05/03/2023]
Abstract
Rett syndrome (RTT) is a severe neurodevelopmental disorder caused by loss of function of the X-linked methyl-CpG–binding protein 2 (MECP2). Several case studies report that gross motor function can be improved in children with RTT through treadmill walking, but whether the MeCP2-deficient motor circuit can support actual motor learning remains unclear. We used two-photon calcium imaging to simultaneously observe layer (L) 2/3 and L5a excitatory neuronal activity in the motor cortex (M1) while mice adapted to changing speeds on a computerized running wheel. Despite circuit hypoactivity and weakened functional connectivity across L2/3 and L5a, the Mecp2-null circuit’s firing pattern evolved with improved performance over 2 weeks. Moreover, trained mice became less anxious and lived 20% longer than untrained mice. Because motor deficits and anxiety are core symptoms of RTT, which is not diagnosed until well after symptom onset, these results underscore the benefit of motor learning.
Collapse
Affiliation(s)
- Yuanlei Yue
- Department of Pharmacology and Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA
| | - Pan Xu
- Department of Pharmacology and Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA
| | - Zhichao Liu
- Department of Physics, Columbian College of Arts and Sciences, The George Washington University, Washington, DC 20037, USA
| | - Xiaoqian Sun
- Department of Computer Science, School of Engineering and Applied Science, The George Washington University, Washington, DC 20037, USA
| | - Juntao Su
- Department of Statistics, Columbian College of Arts and Sciences, The George Washington University, Washington, DC 20037, USA
| | - Hongfei Du
- Department of Statistics, Columbian College of Arts and Sciences, The George Washington University, Washington, DC 20037, USA
| | - Lingling Chen
- Department of Pharmacology and Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA
| | - Ryan T. Ash
- Department of Psychiatry, Stanford University, Palo Alto, CA 94305, USA
| | - Stelios Smirnakis
- Department of Neurology, Brigham and Women’s Hospital, Jamaica Plain VA Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Rahul Simha
- Department of Computer Science, School of Engineering and Applied Science, The George Washington University, Washington, DC 20037, USA
| | - Linda Kusner
- Department of Pharmacology and Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA
| | - Chen Zeng
- Department of Physics, Columbian College of Arts and Sciences, The George Washington University, Washington, DC 20037, USA
| | - Hui Lu
- Department of Pharmacology and Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA
| |
Collapse
|
25
|
Samarasinghe RA, Miranda OA, Buth JE, Mitchell S, Ferando I, Watanabe M, Allison TF, Kurdian A, Fotion NN, Gandal MJ, Golshani P, Plath K, Lowry WE, Parent JM, Mody I, Novitch BG. Identification of neural oscillations and epileptiform changes in human brain organoids. Nat Neurosci 2021; 24:1488-1500. [PMID: 34426698 PMCID: PMC9070733 DOI: 10.1038/s41593-021-00906-5] [Citation(s) in RCA: 147] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 07/08/2021] [Indexed: 02/06/2023]
Abstract
Brain organoids represent a powerful tool for studying human neurological diseases, particularly those that affect brain growth and structure. However, many diseases manifest with clear evidence of physiological and network abnormality in the absence of anatomical changes, raising the question of whether organoids possess sufficient neural network complexity to model these conditions. Here, we explore the network-level functions of brain organoids using calcium sensor imaging and extracellular recording approaches that together reveal the existence of complex network dynamics reminiscent of intact brain preparations. We demonstrate highly abnormal and epileptiform-like activity in organoids derived from induced pluripotent stem cells from individuals with Rett syndrome, accompanied by transcriptomic differences revealed by single-cell analyses. We also rescue key physiological activities with an unconventional neuroregulatory drug, pifithrin-α. Together, these findings provide an essential foundation for the utilization of brain organoids to study intact and disordered human brain network formation and illustrate their utility in therapeutic discovery.
Collapse
Affiliation(s)
- Ranmal A Samarasinghe
- Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA
- Intellectual Development and Disabilities Research Center, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Osvaldo A Miranda
- Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA
- Intellectual Development and Disabilities Research Center, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Jessie E Buth
- Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA
- Intellectual Development and Disabilities Research Center, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Simon Mitchell
- Institute for Quantitative and Computational Biosciences, Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
- Brighton and Sussex Medical School, Falmer, United Kingdom
| | - Isabella Ferando
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Momoko Watanabe
- Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA
- Intellectual Development and Disabilities Research Center, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Department of Anatomy & Neurobiology, Sue & Bill Gross Stem Cell Research Center, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Thomas F Allison
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Biological Chemistry, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Arinnae Kurdian
- Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA
- Intellectual Development and Disabilities Research Center, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- CIRM CSUN-UCLA Stem Cell Training Program, California State University, Northridge, CA, USA
| | - Namie N Fotion
- Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA
- Intellectual Development and Disabilities Research Center, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Michael J Gandal
- Intellectual Development and Disabilities Research Center, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Peyman Golshani
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Intellectual Development and Disabilities Research Center, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, USA
- West Los Angeles VA Medical Center, Los Angeles, CA, USA
| | - Kathrin Plath
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Biological Chemistry, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - William E Lowry
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jack M Parent
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, USA
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
- Ann Arbor VA Healthcare System, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Istvan Mody
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Bennett G Novitch
- Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA.
- Intellectual Development and Disabilities Research Center, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.
| |
Collapse
|
26
|
Ash RT, Buffington SA, Park J, Suter B, Costa-Mattioli M, Zoghbi HY, Smirnakis SM. Inhibition of Elevated Ras-MAPK Signaling Normalizes Enhanced Motor Learning and Excessive Clustered Dendritic Spine Stabilization in the MECP2-Duplication Syndrome Mouse Model of Autism. eNeuro 2021; 8:ENEURO.0056-21.2021. [PMID: 34021030 PMCID: PMC8260274 DOI: 10.1523/eneuro.0056-21.2021] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 04/06/2021] [Accepted: 04/07/2021] [Indexed: 12/26/2022] Open
Abstract
The inflexible repetitive behaviors and "insistence on sameness" seen in autism imply a defect in neural processes controlling the balance between stability and plasticity of synaptic connections in the brain. It has been proposed that abnormalities in the Ras-ERK/MAPK pathway, a key plasticity-related cell signaling pathway known to drive consolidation of clustered synaptic connections, underlie altered learning phenotypes in autism. However, a link between altered Ras-ERK signaling and clustered dendritic spine plasticity has yet to be explored in an autism animal model in vivo The formation and stabilization of dendritic spine clusters is abnormally increased in the MECP2-duplication syndrome mouse model of syndromic autism, suggesting that ERK signaling may be increased. Here, we show that the Ras-ERK pathway is indeed hyperactive following motor training in MECP2-duplication mouse motor cortex. Pharmacological inhibition of ERK signaling normalizes the excessive clustered spine stabilization and enhanced motor learning behavior in MECP2-duplication mice. We conclude that hyperactive ERK signaling may contribute to abnormal clustered dendritic spine consolidation and motor learning in this model of syndromic autism.
Collapse
Affiliation(s)
- Ryan Thomas Ash
- Department of Psychiatry and Behavioral Sciences, Stanford University, CA 94305
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
- Medical Scientist Training Program, Baylor College of Medicine, Houston, TX 77030
- Department of Neurology, Brigham and Women's Hospital and Jamaica Plain Veterans Administration Hospital, Harvard Medical School, Boston, MA 02115
| | - Shelly Alexandra Buffington
- Memory and Brain Research Center, Baylor College of Medicine, Houston, TX 77030
- Department of Neuroscience, Cell Biology, and Anatomy, University of Texas Medical Branch, Galveston, TX 77555
| | - Jiyoung Park
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
- Department of Neurology, Brigham and Women's Hospital and Jamaica Plain Veterans Administration Hospital, Harvard Medical School, Boston, MA 02115
| | - Bernhard Suter
- Department of Neurology, Brigham and Women's Hospital and Jamaica Plain Veterans Administration Hospital, Harvard Medical School, Boston, MA 02115
- Department of Pediatrics, Texas Children's Hospital and Baylor College of Medicine, Houston, TX 77030
| | - Mauro Costa-Mattioli
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
- Memory and Brain Research Center, Baylor College of Medicine, Houston, TX 77030
| | - Huda Yaya Zoghbi
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
- Department of Pediatrics, Texas Children's Hospital and Baylor College of Medicine, Houston, TX 77030
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030
- Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX 77030
| | - Stelios Manolis Smirnakis
- Department of Neurology, Brigham and Women's Hospital and Jamaica Plain Veterans Administration Hospital, Harvard Medical School, Boston, MA 02115
| |
Collapse
|
27
|
Reviewing Evidence for the Relationship of EEG Abnormalities and RTT Phenotype Paralleled by Insights from Animal Studies. Int J Mol Sci 2021; 22:ijms22105308. [PMID: 34069993 PMCID: PMC8157853 DOI: 10.3390/ijms22105308] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 05/09/2021] [Accepted: 05/12/2021] [Indexed: 12/29/2022] Open
Abstract
Rett syndrome (RTT) is a rare neurodevelopmental disorder that is usually caused by mutations of the MECP2 gene. Patients with RTT suffer from severe deficits in motor, perceptual and cognitive domains. Electroencephalogram (EEG) has provided useful information to clinicians and scientists, from the very first descriptions of RTT, and yet no reliable neurophysiological biomarkers related to the pathophysiology of the disorder or symptom severity have been identified to date. To identify consistently observed and potentially informative EEG characteristics of RTT pathophysiology, and ascertain areas most worthy of further systematic investigation, here we review the literature for EEG abnormalities reported in patients with RTT and in its disease models. While pointing to some promising potential EEG biomarkers of RTT, our review identify areas of need to realize the potential of EEG including (1) quantitative investigation of promising clinical-EEG observations in RTT, e.g., shift of mu rhythm frequency and EEG during sleep; (2) closer alignment of approaches between patients with RTT and its animal models to strengthen the translational significance of the work (e.g., EEG measurements and behavioral states); (3) establishment of large-scale consortium research, to provide adequate Ns to investigate age and genotype effects.
Collapse
|
28
|
Delayed motor learning in a 16p11.2 deletion mouse model of autism is rescued by locus coeruleus activation. Nat Neurosci 2021; 24:646-657. [PMID: 33753944 DOI: 10.1038/s41593-021-00815-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 02/05/2021] [Indexed: 01/30/2023]
Abstract
Children with autism spectrum disorder often exhibit delays in achieving motor developmental milestones such as crawling, walking and speech articulation. However, little is known about the neural mechanisms underlying motor-related deficits. Here, we reveal that mice with a syntenic deletion of the chromosome 16p11.2, a common copy number variation associated with autism spectrum disorder, also exhibit delayed motor learning without showing gross motor deficits. Using in vivo two-photon imaging in awake mice, we find that layer 2/3 excitatory neurons in the motor cortex of adult male 16p11.2-deletion mice show abnormally high activity during the initial phase of learning, and the process of learning-induced spine reorganization is prolonged. Pharmacogenetic activation of locus coeruleus noradrenergic neurons was sufficient to rescue the circuit deficits and the delayed motor learning in these mice. Our results unveil an unanticipated role of noradrenergic neuromodulation in improving the delayed motor learning in 16p11.2-deletion male mice.
Collapse
|
29
|
Presymptomatic training mitigates functional deficits in a mouse model of Rett syndrome. Nature 2021; 592:596-600. [PMID: 33762729 DOI: 10.1038/s41586-021-03369-7] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 02/17/2021] [Indexed: 01/26/2023]
Abstract
Mutations in the X-linked gene MECP2 cause Rett syndrome, a progressive neurological disorder in which children develop normally for the first one or two years of life before experiencing profound motor and cognitive decline1-3. At present there are no effective treatments for Rett syndrome, but we hypothesized that using the period of normal development to strengthen motor and memory skills might confer some benefit. Here we find, using a mouse model of Rett syndrome, that intensive training beginning in the presymptomatic period dramatically improves the performance of specific motor and memory tasks, and significantly delays the onset of symptoms. These benefits are not observed when the training begins after symptom onset. Markers of neuronal activity and chemogenetic manipulation reveal that task-specific neurons that are repeatedly activated during training develop more dendritic arbors and have better neurophysiological responses than those in untrained animals, thereby enhancing their functionality and delaying symptom onset. These results provide a rationale for genetic screening of newborns for Rett syndrome, as presymptomatic intervention might mitigate symptoms or delay their onset. Similar strategies should be studied for other childhood neurological disorders.
Collapse
|
30
|
Davidian D, Ziman B, Escobar AL, Oviedo NJ. Direct Current Electric Stimulation Alters the Frequency and the Distribution of Mitotic Cells in Planarians. Bioelectricity 2021; 3:77-91. [PMID: 34476379 DOI: 10.1089/bioe.2020.0026] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Background: The use of direct current electric stimulation (DCS) is an effective strategy to treat disease and enhance body functionality. Thus, treatment with DCS is an attractive biomedical alternative, but the molecular underpinnings remain mostly unknown. The lack of experimental models to dissect the effects of DCS from molecular to organismal levels is an important caveat. Here, we introduce the planarian flatworm Schmidtea mediterranea as a tractable organism for in vivo studies of DCS. We developed an experimental method that facilitates the application of direct current electrical stimulation to the whole planarian body (pDCS). Materials and Methods: Planarian immobilization was achieved by combining treatment with anesthesia, agar embedding, and low temperature via a dedicated thermoelectric cooling unit. Electric currents for pDCS were delivered using pulled glass microelectrodes. The electric potential was supplied through a constant voltage power supply. pDCS was administered up to six hours, and behavioral and molecular effects were measured by using video recordings, immunohistochemistry, and gene expression analysis. Results: The behavioral immobilization effects are reversible, and pDCS resulted in a redistribution of mitotic cells along the mediolateral axis of the planarian body. The pDCS effects were dependent on the polarity of the electric field, which led to either increase in reductions in mitotic densities associated with the time of pDCS. The changes in mitotic cells were consistent with apparent redistribution in gene expression of the stem cell marker smedwi-1. Conclusion: The immobilization technique presented in this work facilitates studies aimed at dissecting the effects of exogenous electric stimulation in the adult body. Treatment with DCS can be administered for varying times, and the consequences evaluated at different levels, including animal behavior, cellular and transcriptional changes. Indeed, treatment with pDCS can alter cellular and transcriptional parameters depending on the polarity of the electric field and duration of the exposure.
Collapse
Affiliation(s)
- Devon Davidian
- Department of Molecular & Cell Biology and University of California Merced, Merced, California, USA
| | - Benjamin Ziman
- Department of Molecular & Cell Biology and University of California Merced, Merced, California, USA
| | - Ariel L Escobar
- Department of Bioengineering, University of California Merced, Merced, California, USA
| | - Néstor J Oviedo
- Department of Molecular & Cell Biology and University of California Merced, Merced, California, USA
| |
Collapse
|
31
|
D'Mello SR. MECP2 and the Biology of MECP2 Duplication Syndrome. J Neurochem 2021; 159:29-60. [PMID: 33638179 DOI: 10.1111/jnc.15331] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/21/2021] [Accepted: 02/18/2021] [Indexed: 11/27/2022]
Abstract
MECP2 duplication syndrome (MDS), a rare X-linked genomic disorder affecting predominantly males, is caused by duplication of the chromosomal region containing the methyl CpG binding protein-2 (MECP2) gene, which encodes methyl-CpG-binding protein 2 (MECP2), a multi-functional protein required for proper brain development and maintenance of brain function during adulthood. Disease symptoms include severe motor and cognitive impairment, delayed or absent speech development, autistic features, seizures, ataxia, recurrent respiratory infections and shortened lifespan. The cellular and molecular mechanisms by which a relatively modest increase in MECP2 protein causes such severe disease symptoms are poorly understood and consequently there are no treatments available for this fatal disorder. This review summarizes what is known to date about the structure and complex regulation of MECP2 and its many functions in the developing and adult brain. Additionally, recent experimental findings on the cellular and molecular underpinnings of MDS based on cell culture and mouse models of the disorder are reviewed. The emerging picture from these studies is that MDS is a neurodegenerative disorder in which neurons die in specific parts of the central nervous system, including the cortex, hippocampus, cerebellum and spinal cord. Neuronal death likely results from astrocytic dysfunction, including a breakdown of glutamate homeostatic mechanisms. The role of elevations in the expression of glial acidic fibrillary protein (GFAP) in astrocytes and the microtubule-associated protein, Tau, in neurons to the pathogenesis of MDS is discussed. Lastly, potential therapeutic strategies to potentially treat MDS are discussed.
Collapse
|
32
|
Scott RC. Brains, complex systems and therapeutic opportunities in epilepsy. Seizure 2021; 90:155-159. [PMID: 33582003 DOI: 10.1016/j.seizure.2021.02.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Revised: 01/27/2021] [Accepted: 02/01/2021] [Indexed: 12/16/2022] Open
Abstract
The treatment of epilepsy remains extremely challenging for the thirty percent of people that do not become seizure free. This is despite the introduction of multiple new drugs over that last several decades, highlighting the need for new approaches to identifying novel therapeutic strategies. Conceptualizing the brain as a complex adaptive system and applying the tools that are used in addressing such systems provides an opportunity for expanding the space in which to search for new therapies. Epilepsy has long been considered a network disease at the level of whole brain connectivity, but the application of the concepts to gene and protein expression networks as well as to the dynamic behaviors of microcircuits has been underexplored. These levels of the brain complex adaptive system will be reviewed and a case made for the epilepsy community to embrace these concepts in order to reap to enormous potential rewards.
Collapse
Affiliation(s)
- Rod C Scott
- University of Vermont, 95 Carrigan Drive, Burlington, VT, 05405, United States; University of Vermont Medical Center, United States; Great Ormond Street Hospital for Children NHS Trust, United Kingdom.
| |
Collapse
|
33
|
Pires J, Nelissen R, Mansvelder HD, Meredith RM. Spontaneous synchronous network activity in the neonatal development of mPFC in mice. Dev Neurobiol 2021; 81:207-225. [PMID: 33453138 PMCID: PMC8048581 DOI: 10.1002/dneu.22811] [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] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 12/31/2020] [Accepted: 01/03/2021] [Indexed: 12/28/2022]
Abstract
Spontaneous Synchronous Network Activity (SSA) is a hallmark of neurodevelopment found in numerous central nervous system structures, including neocortex. SSA occurs during restricted developmental time‐windows, commonly referred to as critical periods in sensory neocortex. Although part of the neocortex, the critical period for SSA in the medial prefrontal cortex (mPFC) and the underlying mechanisms for generation and propagation are unknown. Using Ca2+ imaging and whole‐cell patch‐clamp in an acute mPFC slice mouse model, the development of spontaneous activity and SSA was investigated at cellular and network levels during the two first postnatal weeks. The data revealed that developing mPFC neuronal networks are spontaneously active and exhibit SSA in the first two postnatal weeks, with peak synchronous activity at postnatal days (P)8–9. Networks remain active but are desynchronized by the end of this 2‐week period. SSA was driven by excitatory ionotropic glutamatergic transmission with a small contribution of excitatory GABAergic transmission at early time points. The neurohormone oxytocin desynchronized SSA in the first postnatal week only without affecting concurrent spontaneous activity. By the end of the second postnatal week, inhibiting GABAA receptors restored SSA. These findings point to the emergence of GABAA receptor‐mediated inhibition as a major factor in the termination of SSA in mouse mPFC.
Collapse
Affiliation(s)
- Johny Pires
- Department of Integrative Neurophysiology, Center for Neurogenomics & Cognitive Research, Faculty of Science, Neuroscience Campus Amsterdam, VU University Amsterdam, Amsterdam, the Netherlands
| | - Rosalie Nelissen
- Department of Integrative Neurophysiology, Center for Neurogenomics & Cognitive Research, Faculty of Science, Neuroscience Campus Amsterdam, VU University Amsterdam, Amsterdam, the Netherlands
| | - Huibert D Mansvelder
- Department of Integrative Neurophysiology, Center for Neurogenomics & Cognitive Research, Faculty of Science, Neuroscience Campus Amsterdam, VU University Amsterdam, Amsterdam, the Netherlands
| | - Rhiannon M Meredith
- Department of Integrative Neurophysiology, Center for Neurogenomics & Cognitive Research, Faculty of Science, Neuroscience Campus Amsterdam, VU University Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
34
|
Achilly NP, He LJ, Kim OA, Ohmae S, Wojaczynski GJ, Lin T, Sillitoe RV, Medina JF, Zoghbi HY. Deleting Mecp2 from the cerebellum rather than its neuronal subtypes causes a delay in motor learning in mice. eLife 2021; 10:64833. [PMID: 33494858 PMCID: PMC7837679 DOI: 10.7554/elife.64833] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 01/13/2021] [Indexed: 12/28/2022] Open
Abstract
Rett syndrome is a devastating childhood neurological disorder caused by mutations in MECP2. Of the many symptoms, motor deterioration is a significant problem for patients. In mice, deleting Mecp2 from the cortex or basal ganglia causes motor dysfunction, hypoactivity, and tremor, which are abnormalities observed in patients. Little is known about the function of Mecp2 in the cerebellum, a brain region critical for motor function. Here we show that deleting Mecp2 from the cerebellum, but not from its neuronal subtypes, causes a delay in motor learning that is overcome by additional training. We observed irregular firing rates of Purkinje cells and altered heterochromatin architecture within the cerebellum of knockout mice. These findings demonstrate that the motor deficits present in Rett syndrome arise, in part, from cerebellar dysfunction. For Rett syndrome and other neurodevelopmental disorders, our results highlight the importance of understanding which brain regions contribute to disease phenotypes.
Collapse
Affiliation(s)
- Nathan P Achilly
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, United States.,Program in Developmental Biology, Baylor College of Medicine, Houston, United States.,Medical Scientist Training Program, Baylor College of Medicine, Houston, United States
| | - Ling-Jie He
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, United States.,Department of Human and Molecular Genetics, Baylor College of Medicine, Houston, United States.,Howard Hughes Medical Institute, Baylor College of Medicine, Houston, United States
| | - Olivia A Kim
- Department of Neuroscience, Baylor College of Medicine, Houston, United States
| | - Shogo Ohmae
- Department of Neuroscience, Baylor College of Medicine, Houston, United States
| | | | - Tao Lin
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, United States.,Department of Pathology and Immunology, Baylor College of Medicine, Houston, United States
| | - Roy V Sillitoe
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, United States.,Program in Developmental Biology, Baylor College of Medicine, Houston, United States.,Department of Neuroscience, Baylor College of Medicine, Houston, United States.,Department of Pathology and Immunology, Baylor College of Medicine, Houston, United States
| | - Javier F Medina
- Department of Neuroscience, Baylor College of Medicine, Houston, United States
| | - Huda Y Zoghbi
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, United States.,Program in Developmental Biology, Baylor College of Medicine, Houston, United States.,Howard Hughes Medical Institute, Baylor College of Medicine, Houston, United States.,Department of Neuroscience, Baylor College of Medicine, Houston, United States.,Department of Pathology and Immunology, Baylor College of Medicine, Houston, United States.,Department of Neurology, Baylor College of Medicine, Houston, United States.,Department of Pediatrics, Baylor College of Medicine, Houston, United States
| |
Collapse
|
35
|
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: 44] [Impact Index Per Article: 11.0] [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
|
36
|
Ash RT, Park J, Suter B, Zoghbi HY, Smirnakis SM. Excessive Formation and Stabilization of Dendritic Spine Clusters in the MECP2-Duplication Syndrome Mouse Model of Autism. eNeuro 2021; 8:ENEURO.0282-20.2020. [PMID: 33168618 PMCID: PMC7877475 DOI: 10.1523/eneuro.0282-20.2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/11/2020] [Accepted: 09/14/2020] [Indexed: 11/21/2022] Open
Abstract
Autism-associated genetic mutations may perturb the balance between stability and plasticity of synaptic connections in the brain. Here, we report an increase in the formation and stabilization of dendritic spines in the cerebral cortex of the mouse model of MECP2-duplication syndrome, a high-penetrance form of syndromic autism. Increased stabilization is mediated entirely by spines that form cooperatively in 10-μm clusters and is observable across multiple cortical areas both spontaneously and following motor training. Excessive stability of dendritic spine clusters could contribute to behavioral rigidity and other phenotypes in syndromic autism.
Collapse
Affiliation(s)
- Ryan Thomas Ash
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
- Medical Scientist Training Program, Baylor College of Medicine, Houston, TX 77030
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115
| | - Jiyoung Park
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115
| | - Bernhard Suter
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115
- Department of Pediatrics, Texas Children's Hospital and Baylor College of Medicine, Houston, TX 77030
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030
| | - Huda Yaya Zoghbi
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
- Department of Pediatrics, Texas Children's Hospital and Baylor College of Medicine, Houston, TX 77030
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030
- Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX 77030
| | - Stelios Manolis Smirnakis
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115
| |
Collapse
|
37
|
Disruption of NEUROD2 causes a neurodevelopmental syndrome with autistic features via cell-autonomous defects in forebrain glutamatergic neurons. Mol Psychiatry 2021; 26:6125-6148. [PMID: 34188164 PMCID: PMC8760061 DOI: 10.1038/s41380-021-01179-x] [Citation(s) in RCA: 14] [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: 08/20/2020] [Revised: 05/17/2021] [Accepted: 05/26/2021] [Indexed: 02/06/2023]
Abstract
While the transcription factor NEUROD2 has recently been associated with epilepsy, its precise role during nervous system development remains unclear. Using a multi-scale approach, we set out to understand how Neurod2 deletion affects the development of the cerebral cortex in mice. In Neurod2 KO embryos, cortical projection neurons over-migrated, thereby altering the final size and position of layers. In juvenile and adults, spine density and turnover were dysregulated in apical but not basal compartments in layer 5 neurons. Patch-clamp recordings in layer 5 neurons of juvenile mice revealed increased intrinsic excitability. Bulk RNA sequencing showed dysregulated expression of many genes associated with neuronal excitability and synaptic function, whose human orthologs were strongly associated with autism spectrum disorders (ASD). At the behavior level, Neurod2 KO mice displayed social interaction deficits, stereotypies, hyperactivity, and occasionally spontaneous seizures. Mice heterozygous for Neurod2 had similar defects, indicating that Neurod2 is haploinsufficient. Finally, specific deletion of Neurod2 in forebrain excitatory neurons recapitulated cellular and behavioral phenotypes found in constitutive KO mice, revealing the region-specific contribution of dysfunctional Neurod2 in symptoms. Informed by these neurobehavioral features in mouse mutants, we identified eleven patients from eight families with a neurodevelopmental disorder including intellectual disability and ASD associated with NEUROD2 pathogenic mutations. Our findings demonstrate crucial roles for Neurod2 in neocortical development, whose alterations can cause neurodevelopmental disorders including intellectual disability and ASD.
Collapse
|
38
|
Sandweiss AJ, Brandt VL, Zoghbi HY. Advances in understanding of Rett syndrome and MECP2 duplication syndrome: prospects for future therapies. Lancet Neurol 2020; 19:689-698. [PMID: 32702338 DOI: 10.1016/s1474-4422(20)30217-9] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Revised: 05/07/2020] [Accepted: 05/12/2020] [Indexed: 01/07/2023]
Abstract
The X-linked gene encoding MECP2 is involved in two severe and complex neurodevelopmental disorders. Loss of function of the MeCP2 protein underlies Rett syndrome, whereas duplications of the MECP2 locus cause MECP2 duplication syndrome. Research on the mechanisms by which MeCP2 exerts effects on gene expression in neurons, studies of animal models bearing different disease-causing mutations, and more in-depth observations of clinical presentations have clarified some issues even as they have raised further questions. Yet there is enough evidence so far to suggest possible approaches to therapy for these two diseases that could go beyond attempting to address specific signs and symptoms (of which there are many) and instead target the pathophysiology underlying MECP2 disorders. Further work could bring antisense oligonucleotides, deep brain stimulation, and gene therapy into the clinic within the next decade or so.
Collapse
Affiliation(s)
- Alexander J Sandweiss
- Department of Pediatrics, Section of Neurology and Developmental Neurosciences, Baylor College of Medicine, Houston, TX, USA; Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA
| | - Vicky L Brandt
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA
| | - Huda Y Zoghbi
- Department of Pediatrics, Section of Neurology and Developmental Neurosciences, Baylor College of Medicine, Houston, TX, USA; Howard Hughes Medical Institute, and Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA.
| |
Collapse
|
39
|
Javed S, Selliah T, Lee YJ, Huang WH. Dosage-sensitive genes in autism spectrum disorders: From neurobiology to therapy. Neurosci Biobehav Rev 2020; 118:538-567. [PMID: 32858083 DOI: 10.1016/j.neubiorev.2020.08.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 07/26/2020] [Accepted: 08/17/2020] [Indexed: 12/24/2022]
Abstract
Autism spectrum disorders (ASDs) are a group of heterogenous neurodevelopmental disorders affecting 1 in 59 children. Syndromic ASDs are commonly associated with chromosomal rearrangements or dosage imbalance involving a single gene. Many of these genes are dosage-sensitive and regulate transcription, protein homeostasis, and synaptic function in the brain. Despite vastly different molecular perturbations, syndromic ASDs share core symptoms including social dysfunction and repetitive behavior. However, each ASD subtype has a unique pathogenic mechanism and combination of comorbidities that require individual attention. We have learned a great deal about how these dosage-sensitive genes control brain development and behaviors from genetically-engineered mice. Here we describe the clinical features of eight monogenic neurodevelopmental disorders caused by dosage imbalance of four genes, as well as recent advances in using genetic mouse models to understand their pathogenic mechanisms and develop intervention strategies. We propose that applying newly developed quantitative molecular and neuroscience technologies will advance our understanding of the unique neurobiology of each disorder and enable the development of personalized therapy.
Collapse
Affiliation(s)
- Sehrish Javed
- Centre for Research in Neuroscience, Department of Neurology and Neurosurgery, The Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| | - Tharushan Selliah
- Centre for Research in Neuroscience, Department of Neurology and Neurosurgery, The Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| | - Yu-Ju Lee
- Centre for Research in Neuroscience, Department of Neurology and Neurosurgery, The Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| | - Wei-Hsiang Huang
- Centre for Research in Neuroscience, Department of Neurology and Neurosurgery, The Research Institute of the McGill University Health Centre, Montréal, Québec, Canada.
| |
Collapse
|
40
|
Fagiolini M, Patrizi A, LeBlanc J, Jin LW, Maezawa I, Sinnett S, Gray SJ, Molholm S, Foxe JJ, Johnston MV, Naidu S, Blue M, Hossain A, Kadam S, Zhao X, Chang Q, Zhou Z, Zoghbi H. Intellectual and Developmental Disabilities Research Centers: A Multidisciplinary Approach to Understand the Pathogenesis of Methyl-CpG Binding Protein 2-related Disorders. Neuroscience 2020; 445:190-206. [PMID: 32360592 DOI: 10.1016/j.neuroscience.2020.04.037] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 04/20/2020] [Accepted: 04/21/2020] [Indexed: 12/20/2022]
Abstract
Disruptions in the gene encoding methyl-CpG binding protein 2 (MECP2) underlie complex neurodevelopmental disorders including Rett Syndrome (RTT), MECP2 duplication disorder, intellectual disabilities, and autism. Significant progress has been made on the molecular and cellular basis of MECP2-related disorders providing a new framework for understanding how altered epigenetic landscape can derail the formation and refinement of neuronal circuits in early postnatal life and proper neurological function. This review will summarize selected major findings from the past years and particularly highlight the integrated and multidisciplinary work done at eight NIH-funded Intellectual and Developmental Disabilities Research Centers (IDDRC) across the US. Finally, we will outline a path forward with identification of reliable biomarkers and outcome measures, longitudinal preclinical and clinical studies, reproducibility of results across centers as a synergistic effort to decode and treat the pathogenesis of the complex MeCP2 disorders.
Collapse
Affiliation(s)
- Michela Fagiolini
- Children's Hospital Intellectual and Developmental Disabilities Research Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Annarita Patrizi
- Children's Hospital Intellectual and Developmental Disabilities Research Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jocelyn LeBlanc
- Children's Hospital Intellectual and Developmental Disabilities Research Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Lee-Way Jin
- UC Davis MIND Institute, University of California, Sacramento, CA, USA
| | - Izumi Maezawa
- UC Davis MIND Institute, University of California, Sacramento, CA, USA
| | - Sarah Sinnett
- UNC Intellectual and Developmental Disabilities Research Center, University of North Carolina, Gene Therapy Center and Dept. of Ophthalmology, Chapel Hill, NC, USA; Department of Pediatrics, UT Southwestern Medical Center, Dallas, TX, USA
| | - Steven J Gray
- UNC Intellectual and Developmental Disabilities Research Center, University of North Carolina, Gene Therapy Center and Dept. of Ophthalmology, Chapel Hill, NC, USA; Department of Pediatrics, UT Southwestern Medical Center, Dallas, TX, USA
| | - Sophie Molholm
- The Cognitive Neurophysiology Laboratory, Departments of Pediatrics, Neuroscience, and Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, NY, USA
| | - John J Foxe
- The Cognitive Neurophysiology Laboratory, Ernest J. Del Monte Institute for Neuroscience, Department of Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Michael V Johnston
- Kennedy Krieger Institute Intellectual and Developmental Disabilities Research Center/Hugo Moser Research Institute at Kennedy Krieger and Johns Hopkins School of Medicine, USA
| | - Sakkubai Naidu
- Kennedy Krieger Institute Intellectual and Developmental Disabilities Research Center/Hugo Moser Research Institute at Kennedy Krieger and Johns Hopkins School of Medicine, USA
| | - Mary Blue
- Kennedy Krieger Institute Intellectual and Developmental Disabilities Research Center/Hugo Moser Research Institute at Kennedy Krieger and Johns Hopkins School of Medicine, USA
| | - Ahamed Hossain
- Kennedy Krieger Institute Intellectual and Developmental Disabilities Research Center/Hugo Moser Research Institute at Kennedy Krieger and Johns Hopkins School of Medicine, USA
| | - Shilpa Kadam
- Kennedy Krieger Institute Intellectual and Developmental Disabilities Research Center/Hugo Moser Research Institute at Kennedy Krieger and Johns Hopkins School of Medicine, USA
| | - Xinyu Zhao
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Quiang Chang
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Zhaolan Zhou
- Department of Genetic, Epigenetic Institute, University of Pennsylvania Perelman School of Medicine, Intellectual and Developmental Disabilities Research Center, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Huda Zoghbi
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Program in Developmental Biology, Baylor College of Medicine, Houston, TX, USA; Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA; Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA; Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
41
|
MECP2 Duplication Causes Aberrant GABA Pathways, Circuits and Behaviors in Transgenic Monkeys: Neural Mappings to Patients with Autism. J Neurosci 2020; 40:3799-3814. [PMID: 32269107 DOI: 10.1523/jneurosci.2727-19.2020] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 03/15/2020] [Accepted: 03/16/2020] [Indexed: 12/21/2022] Open
Abstract
MECP2 gain-of-function and loss-of-function in genetically engineered monkeys recapitulates typical phenotypes in patients with autism, yet where MECP2 mutation affects the monkey brain and whether/how it relates to autism pathology remain unknown. Here we report a combination of gene-circuit-behavior analyses including MECP2 coexpression network, locomotive and cognitive behaviors, and EEG and fMRI findings in 5 MECP2 overexpressed monkeys (Macaca fascicularis; 3 females) and 20 wild-type monkeys (Macaca fascicularis; 11 females). Whole-genome expression analysis revealed MECP2 coexpressed genes significantly enriched in GABA-related signaling pathways, whereby reduced β-synchronization within fronto-parieto-occipital networks was associated with abnormal locomotive behaviors. Meanwhile, MECP2-induced hyperconnectivity in prefrontal and cingulate networks accounted for regressive deficits in reversal learning tasks. Furthermore, we stratified a cohort of 49 patients with autism and 72 healthy controls of 1112 subjects using functional connectivity patterns, and identified dysconnectivity profiles similar to those in monkeys. By establishing a circuit-based construct link between genetically defined models and stratified patients, these results pave new avenues to deconstruct clinical heterogeneity and advance accurate diagnosis in psychiatric disorders.SIGNIFICANCE STATEMENT Autism spectrum disorder (ASD) is a complex disorder with co-occurring symptoms caused by multiple genetic variations and brain circuit abnormalities. To dissect the gene-circuit-behavior causal chain underlying ASD, animal models are established by manipulating causative genes such as MECP2 However, it is unknown whether such models have captured any circuit-level pathology in ASD patients, as demonstrated by human brain imaging studies. Here, we use transgenic macaques to examine the causal effect of MECP2 overexpression on gene coexpression, brain circuits, and behaviors. For the first time, we demonstrate that the circuit abnormalities linked to MECP2 and autism-like traits in the monkeys can be mapped to a homogeneous ASD subgroup, thereby offering a new strategy to deconstruct clinical heterogeneity in ASD.
Collapse
|
42
|
Yu B, Yuan B, Dai JK, Cheng TL, Xia SN, He LJ, Yuan YT, Zhang YF, Xu HT, Xu FQ, Liang ZF, Qiu ZL. Reversal of Social Recognition Deficit in Adult Mice with MECP2 Duplication via Normalization of MeCP2 in the Medial Prefrontal Cortex. Neurosci Bull 2020; 36:570-584. [PMID: 32144612 DOI: 10.1007/s12264-020-00467-w] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 11/16/2019] [Indexed: 01/05/2023] Open
Abstract
Methyl-CpG binding protein 2 (MeCP2) is a basic nuclear protein involved in the regulation of gene expression and microRNA processing. Duplication of MECP2-containing genomic segments causes MECP2 duplication syndrome, a severe neurodevelopmental disorder characterized by intellectual disability, motor dysfunction, heightened anxiety, epilepsy, autistic phenotypes, and early death. Reversal of the abnormal phenotypes in adult mice with MECP2 duplication (MECP2-TG) by normalizing the MeCP2 levels across the whole brain has been demonstrated. However, whether different brain areas or neural circuits contribute to different aspects of the behavioral deficits is still unknown. Here, we found that MECP2-TG mice showed a significant social recognition deficit, and were prone to display aversive-like behaviors, including heightened anxiety-like behaviors and a fear generalization phenotype. In addition, reduced locomotor activity was observed in MECP2-TG mice. However, appetitive behaviors and learning and memory were comparable in MECP2-TG and wild-type mice. Functional magnetic resonance imaging illustrated that the differences between MECP2-TG and wild-type mice were mainly concentrated in brain areas regulating emotion and social behaviors. We used the CRISPR-Cas9 method to restore normal MeCP2 levels in the medial prefrontal cortex (mPFC) and bed nuclei of the stria terminalis (BST) of adult MECP2-TG mice, and found that normalization of MeCP2 levels in the mPFC but not in the BST reversed the social recognition deficit. These data indicate that the mPFC is responsible for the social recognition deficit in the transgenic mice, and provide new insight into potential therapies for MECP2 duplication syndrome.
Collapse
Affiliation(s)
- Bin Yu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Chinese Academy of Sciences Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Bo Yuan
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Chinese Academy of Sciences Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Jian-Kun Dai
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Chinese Academy of Sciences Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Tian-Lin Cheng
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Chinese Academy of Sciences Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Sheng-Nan Xia
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Chinese Academy of Sciences Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ling-Jie He
- Department of Molecular and Human Genetics, Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Yi-Ting Yuan
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Chinese Academy of Sciences Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yue-Fang Zhang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Chinese Academy of Sciences Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Hua-Tai Xu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Chinese Academy of Sciences Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Fu-Qiang Xu
- Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Zhi-Feng Liang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Chinese Academy of Sciences Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Zi-Long Qiu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Chinese Academy of Sciences Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China.
| |
Collapse
|
43
|
Ryu HH, Kim SY, Lee YS. Connecting the dots between SHP2 and glutamate receptors. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2020; 24:129-135. [PMID: 32140036 PMCID: PMC7043995 DOI: 10.4196/kjpp.2020.24.2.129] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 01/06/2020] [Accepted: 01/07/2020] [Indexed: 11/18/2022]
Abstract
SHP2 is an unusual protein phosphatase that functions as an activator for several signaling pathways, including the RAS pathway, while most other phosphatases suppress their downstream signaling cascades. The physiological and pathophysiological roles of SHP2 have been extensively studied in the field of cancer research. Mutations in the PTPN11 gene which encodes SHP2 are also highly associated with developmental disorders, such as Noonan syndrome (NS), and cognitive deficits including learning disabilities are common among NS patients. However, the molecular and cellular mechanism by which SHP2 is involved in cognitive functions is not well understood. Recent studies using SHP2 mutant mice or pharmacological inhibitors have shown that SHP2 plays critical role in learning and memory and synaptic plasticity. Here, we review the recent studies demonstrating that SHP2 is involved in synaptic plasticity, and learning and memory, by the regulation of the expression and/or function of glutamate receptors. We suggest that each cell type may have distinct paths connecting the dots between SHP2 and glutamate receptors, and these paths may also change with aging.
Collapse
Affiliation(s)
- Hyun-Hee Ryu
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea.,Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea.,Neuroscience Research Institute, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Sun Yong Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea.,Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Yong-Seok Lee
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea.,Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea.,Neuroscience Research Institute, Seoul National University College of Medicine, Seoul 03080, Korea
| |
Collapse
|
44
|
Forrest ME, Pinkard O, Martin S, Sweet TJ, Hanson G, Coller J. Codon and amino acid content are associated with mRNA stability in mammalian cells. PLoS One 2020; 15:e0228730. [PMID: 32053646 PMCID: PMC7018022 DOI: 10.1371/journal.pone.0228730] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 01/21/2020] [Indexed: 12/31/2022] Open
Abstract
Messenger RNA (mRNA) degradation plays a critical role in regulating transcript levels in the cell and is a major control point for modulating gene expression. In yeast and other model organisms, codon identity is a powerful determinant of transcript stability, contributing broadly to impact half-lives. General principles governing mRNA stability are poorly understood in mammalian systems. Importantly, however, the degradation machinery is highly conserved, thus it seems logical that mammalian transcript half-lives would also be strongly influenced by coding determinants. Herein we characterize the contribution of coding sequence towards mRNA decay in human and Chinese Hamster Ovary cells. In agreement with previous studies, we observed that synonymous codon usage impacts mRNA stability in mammalian cells. Surprisingly, however, we also observe that the amino acid content of a gene is an additional determinant correlating with transcript stability. The impact of codon and amino acid identity on mRNA decay appears to be associated with underlying tRNA and intracellular amino acid concentrations. Accordingly, genes of similar physiological function appear to coordinate their mRNA stabilities in part through codon and amino acid content. Together, these results raise the possibility that intracellular tRNA and amino acid levels interplay to mediate coupling between translational elongation and mRNA degradation rate in mammals.
Collapse
Affiliation(s)
- Megan E. Forrest
- Center for RNA Science and Therapeutics, Case Western Reserve University, Cleveland, Ohio, United States of America
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Otis Pinkard
- Center for RNA Science and Therapeutics, Case Western Reserve University, Cleveland, Ohio, United States of America
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Sophie Martin
- Center for RNA Science and Therapeutics, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Thomas J. Sweet
- Center for RNA Science and Therapeutics, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Gavin Hanson
- Center for RNA Science and Therapeutics, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Jeff Coller
- Center for RNA Science and Therapeutics, Case Western Reserve University, Cleveland, Ohio, United States of America
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, United States of America
- * E-mail:
| |
Collapse
|
45
|
Interneuron deficits in neurodevelopmental disorders: Implications for disease pathology and interneuron-based therapies. Eur J Paediatr Neurol 2020; 24:81-88. [PMID: 31870698 PMCID: PMC7152321 DOI: 10.1016/j.ejpn.2019.12.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 12/06/2019] [Indexed: 12/16/2022]
|
46
|
Poon CH, Chan YS, Fung ML, Lim LW. Memory and neuromodulation: A perspective of DNA methylation. Neurosci Biobehav Rev 2019; 111:57-68. [PMID: 31846654 DOI: 10.1016/j.neubiorev.2019.12.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 11/05/2019] [Accepted: 12/13/2019] [Indexed: 02/07/2023]
Abstract
Neuromodulation techniques have shown promising efficacy on memory function and understanding the epigenetic mechanisms contributing to these processes would shed light on the molecular outcomes essential for cognition. In this review, we highlight some epigenetic mechanisms underlying neuromodulation and regulatory effects of neuronal activity-induced DNA methylation on genes that are highly involved in memory formation. Next, we examine the evidence to support DNA methyltransferase 3a, methyl-CpG binding protein 2, and DNA demethylase as possible memory modulation targets. Finally, we report the recent developments in the field of neuromodulation and explore the potential of these techniques for future neuroepigenetic research.
Collapse
Affiliation(s)
- Chi Him Poon
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Ying-Shing Chan
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Man Lung Fung
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Lee Wei Lim
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
47
|
Lavery LA, Zoghbi HY. The distinct methylation landscape of maturing neurons and its role in Rett syndrome pathogenesis. Curr Opin Neurobiol 2019; 59:180-188. [PMID: 31542590 PMCID: PMC6892602 DOI: 10.1016/j.conb.2019.08.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 08/21/2019] [Indexed: 02/06/2023]
Abstract
Rett syndrome (RTT) is one of the most common causes of intellectual and developmental disabilities in girls, and is caused by mutations in the gene encoding methyl-CpG binding protein 2 (MECP2). Here we will review our current understanding of RTT, the landscape of pathogenic mutations and function of MeCP2, and culminate with recent advances elucidating the distinct DNA methylation landscape in the brain that may explain why disease symptoms are delayed and selective to the nervous system.
Collapse
Affiliation(s)
- Laura A Lavery
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Huda Y Zoghbi
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA; Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA; Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
48
|
Smith ES, Smith DR, Eyring C, Braileanu M, Smith-Connor KS, Ei Tan Y, Fowler AY, Hoffman GE, Johnston MV, Kannan S, Blue ME. Altered trajectories of neurodevelopment and behavior in mouse models of Rett syndrome. Neurobiol Learn Mem 2019; 165:106962. [PMID: 30502397 PMCID: PMC8040058 DOI: 10.1016/j.nlm.2018.11.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 10/17/2018] [Accepted: 11/16/2018] [Indexed: 12/12/2022]
Abstract
Rett Syndrome (RTT) is a genetic disorder that is caused by mutations in the x-linked gene coding for methyl-CpG-biding-protein 2 (MECP2) and that mainly affects females. Male and female transgenic mouse models of RTT have been studied extensively, and we have learned a great deal regarding RTT neuropathology and how MeCP2 deficiency may be influencing brain function and maturation. In this manuscript we review what is known concerning structural and coinciding functional and behavioral deficits in RTT and in mouse models of MeCP2 deficiency. We also introduce our own corroborating data regarding behavioral phenotype and morphological alterations in volume of the cortex and striatum and the density of neurons, aberrations in experience-dependent plasticity within the barrel cortex and the impact of MeCP2 loss on glial structure. We conclude that regional structural changes in genetic models of RTT show great similarity to the alterations in brain structure of patients with RTT. These region-specific modifications often coincide with phenotype onset and contribute to larger issues of circuit connectivity, progression, and severity. Although the alterations seen in mouse models of RTT appear to be primarily due to cell-autonomous effects, there are also non-cell autonomous mechanisms including those caused by MeCP2-deficient glia that negatively impact healthy neuronal function. Collectively, this body of work has provided a solid foundation on which to continue to build our understanding of the role of MeCP2 on neuronal and glial structure and function, its greater impact on neural development, and potential new therapeutic avenues.
Collapse
Affiliation(s)
- Elizabeth S Smith
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Dani R Smith
- Department of Psychological and Brain Sciences, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Charlotte Eyring
- The Hugo W. Moser Research Institute at Kennedy Krieger, Inc., Baltimore, MD 21205, USA
| | - Maria Braileanu
- Undergraduate Program in Neuroscience, The Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Karen S Smith-Connor
- The Hugo W. Moser Research Institute at Kennedy Krieger, Inc., Baltimore, MD 21205, USA
| | - Yew Ei Tan
- Perdana University Graduate School of Medicine, Kuala Lumpur, Malaysia
| | - Amanda Y Fowler
- Department of Biology, Morgan State University, Baltimore, MD 21251, USA
| | - Gloria E Hoffman
- Department of Biology, Morgan State University, Baltimore, MD 21251, USA
| | - Michael V Johnston
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; The Hugo W. Moser Research Institute at Kennedy Krieger, Inc., Baltimore, MD 21205, USA
| | - Sujatha Kannan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; The Hugo W. Moser Research Institute at Kennedy Krieger, Inc., Baltimore, MD 21205, USA
| | - Mary E Blue
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; The Hugo W. Moser Research Institute at Kennedy Krieger, Inc., Baltimore, MD 21205, USA.
| |
Collapse
|
49
|
Banerjee A, Miller MT, Li K, Sur M, Kaufmann WE. Towards a better diagnosis and treatment of Rett syndrome: a model synaptic disorder. Brain 2019; 142:239-248. [PMID: 30649225 DOI: 10.1093/brain/awy323] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 10/31/2018] [Indexed: 12/21/2022] Open
Abstract
With the recent 50th anniversary of the first publication on Rett syndrome, and the almost 20 years since the first report on the link between Rett syndrome and MECP2 mutations, it is important to reflect on the tremendous advances in our understanding and their implications for the diagnosis and treatment of this neurodevelopmental disorder. Rett syndrome features an interesting challenge for biologists and clinicians, as the disorder lies at the intersection of molecular mechanisms of epigenetic regulation and neurophysiological alterations in synapses and circuits that together contribute to severe pathophysiological endophenotypes. Genetic, clinical, and neurobiological evidences support the notion that Rett syndrome is primarily a synaptic disorder, and a disease model for both intellectual disability and autism spectrum disorder. This review examines major developments in both recent neurobiological and preclinical findings of Rett syndrome, and to what extent they are beginning to impact our understanding and management of the disorder. It also discusses potential applications of knowledge on synaptic plasticity abnormalities in Rett syndrome to its diagnosis and treatment.
Collapse
Affiliation(s)
- Abhishek Banerjee
- Laboratory of Neural Circuit Dynamics, Brain Research Institute, University of Zürich, Zürich, Switzerland
| | - Meghan T Miller
- Roche Pharma Research and Early Development, Roche Innovation Center, F. Hoffman-La Roche, Basel, Switzerland
| | - Keji Li
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge MA, USA
| | - Mriganka Sur
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge MA, USA
| | - Walter E Kaufmann
- Department of Human Genetics, Emory University School of Medicine, Atlanta GA, USA
| |
Collapse
|
50
|
Walkley SU, Abbeduto L, Batshaw ML, Bhattacharyya A, Bookheimer SY, Christian BT, Constantino JN, de Vellis J, Doherty DA, Nelson DL, Piven J, Poduri A, Pomeroy SL, Samaco RC, Zoghbi HY, Guralnick MJ. Intellectual and developmental disabilities research centers: Fifty years of scientific accomplishments. Ann Neurol 2019; 86:332-343. [PMID: 31206741 PMCID: PMC8320680 DOI: 10.1002/ana.25531] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 06/12/2019] [Accepted: 06/13/2019] [Indexed: 12/17/2022]
Abstract
Progress in addressing the origins of intellectual and developmental disabilities accelerated with the establishment 50 years ago of the Eunice Kennedy Shriver National Institute of Child Health and Human Development of the National Institutes of Health and associated Intellectual and Developmental Disabilities Research Centers. Investigators at these Centers have made seminal contributions to understanding human brain and behavioral development and defining mechanisms and treatments of disorders of the developing brain. ANN NEUROL 2019;86:332-343.
Collapse
Affiliation(s)
- Steven U Walkley
- Department of Neuroscience, Albert Einstein College of Medicine, Rose F. Kennedy Intellectual and Developmental Disabilities Research Center, Bronx, NY
| | - Leonard Abbeduto
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, University of California, Davis Memory Impairments and Neurological Disorders Institute, Sacramento, CA
| | - Mark L Batshaw
- Children's Research Institute, Children's National Medical Center, Washington, DC
| | - Anita Bhattacharyya
- Department of Cell and Regenerative Biology, Waisman Center, University of Wisconsin-Madison, Madison, WI
| | - Susan Y Bookheimer
- Department of Psychiatry and Biobehavioral Sciences, Intellectual and Developmental Research Center, University of California, Los Angeles School of Medicine, Los Angeles, CA
| | - Bradley T Christian
- Departments of Medical Physics and Psychiatry, Waisman Center, University of Wisconsin-Madison, Madison, WI
| | - John N Constantino
- Departments of Psychiatry and Pediatrics, Washington University School of Medicine, Washington University in St Louis Intellectual and Developmental Disabilities Research Center, St Louis, MO
| | - Jean de Vellis
- Department of Psychiatry and Biobehavioral Sciences, Intellectual and Developmental Research Center, University of California, Los Angeles School of Medicine, Los Angeles, CA
| | - Daniel A Doherty
- Department of Pediatrics, Center on Human Development and Disability, University of Washington, Seattle, WA
| | - David L Nelson
- Department of Molecular and Human Genetics, Baylor College of Medicine, Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Baylor College of Medicine Intellectual and Developmental Disabilities Research Center, Houston, TX
| | - Joseph Piven
- Carolina Institute for Developmental Disabilities, University of North Carolina, University of North Carolina Intellectual and Developmental Disabilities Research Center, Chapel Hill, NC
| | - Annapurna Poduri
- Department of Neurology, Harvard Medical School, Boston Children's Hospital and Harvard Medical School Intellectual and Developmental Disabilities Research Center, Boston, MA
| | - Scott L Pomeroy
- Department of Neurology, Harvard Medical School, Boston Children's Hospital and Harvard Medical School Intellectual and Developmental Disabilities Research Center, Boston, MA
| | - Rodney C Samaco
- Department of Molecular and Human Genetics, Baylor College of Medicine, Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Baylor College of Medicine Intellectual and Developmental Disabilities Research Center, Houston, TX
| | - Huda Y Zoghbi
- Department of Molecular and Human Genetics, Baylor College of Medicine, Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Baylor College of Medicine Intellectual and Developmental Disabilities Research Center, Houston, TX
| | - Michael J Guralnick
- Departments of Psychology and Pediatrics, Center on Human Development and Disability, University of Washington, Seattle, WA
| |
Collapse
|