1
|
Ghosh S, Dutta M, Mukherjee D, Raychaudhuri D, Bandyopadhyay SN. Syndromic Association of Depigmentation With Congenital Hearing Loss: A Review of Three Children With Auditory Pigmentary Disorders. EAR, NOSE & THROAT JOURNAL 2024:1455613241306049. [PMID: 39641445 DOI: 10.1177/01455613241306049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024] Open
Abstract
Background: Congenital depigmentation may be associated with congenital sensorineural hearing loss leading to non-development of verbal speech. Objective: To illustrate the clinical features and work-up of 3 children diagnosed with auditory pigmentary disorders (APDs). Methodology: Case series with a review of the literature. Results: The APDs presented here in the 3 children include Waardenburg syndrome type 1, Clouston syndrome, and Waardenburg syndrome type 4 (Waardenburg-Shah syndrome). The characteristic clinical features, audiologic tests, imaging, and the necessary genetic tests carried out subsequently were noted and evaluated. All the children were male and were aged 2 years, 1 year, and 14 months, respectively. All of them had hearing loss and non-development of verbal speech and had some form of oculocutaneous depigmentation. The challenges in the diagnosis, the work-up, and the close differentials were discussed, and the relevant literature was reviewed. Conclusions: The APDs connect congenital depigmentation with prelingual hearing loss through various syndromic disorders. These disorders are not commonly encountered in routine clinical practice; therefore, their proper knowledge is essential for early diagnosis of congenital hearing loss and timely initiation of auditory and speech rehabilitation. This case series deals with a detailed illustration of a few syndromes of the APDs and highlights their clinical presentation and genetic background.
Collapse
Affiliation(s)
- Shabari Ghosh
- Department of Otorhinolaryngology and Head-Neck Surgery, Medical College and Hospital, Kolkata, West Bengal, India
| | - Mainak Dutta
- Department of Otorhinolaryngology and Head-Neck Surgery, All India Institute of Medical Sciences, Kalyani, Nadia, West Bengal, India
| | - Diptanshu Mukherjee
- Department of Otorhinolaryngology and Head-Neck Surgery, Medical College and Hospital, Kolkata, West Bengal, India
| | - Dibyendu Raychaudhuri
- Department of Pediatric Medicine, Medical College and Hospital, Kolkata, West Bengal, India
| | - Saumendra Nath Bandyopadhyay
- Department of Otorhinolaryngology and Head-Neck Surgery, Medical College and Hospital, Kolkata, West Bengal, India
| |
Collapse
|
2
|
Kaufmann WE, Luu S, Budimirovic DB. Drug Treatments for Neurodevelopmental Disorders: Targeting Signaling Pathways and Homeostasis. Curr Neurol Neurosci Rep 2024; 25:7. [PMID: 39641900 DOI: 10.1007/s11910-024-01394-3] [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] [Accepted: 10/08/2024] [Indexed: 12/07/2024]
Abstract
PURPOSE OF THE REVIEW Preclinical and clinical evidence support the notion that neurodevelopmental disorders (NDDs) are synaptic disorders, characterized by excitatory-inhibitory imbalance. Despite this, NDD drug development programs targeting glutamate or gamma-aminobutyric acid (GABA) receptors have been largely unsuccessful. Nonetheless, recent drug trials in Rett syndrome (RTT), fragile X syndrome (FXS), and other NDDs targeting other mechanisms have met their endpoints. The purpose of this review is to identify the basis of these successful studies. RECENT FINDINGS Despite increasing evidence of disruption in synaptic homeostasis, most genetic variants associated with NDDs implicate proteins involved in cell regulation and not in neurotransmission. Metabolic processes, in particular mitochondrial function, appear to play a role in NDD pathophysiology. NDDs are also characterized by distinctive cell signaling abnormalities, which link cellular and synaptic homeostasis. Recent successful trials in NDDs, including those of trofinetide, the first drug specifically approved for one of these disorders (i.e., RTT), implicate the targeting of downstream processes (i.e., signaling pathways) rather than neurotransmitter receptors. Recent positive drug studies in NDDs and their underlying mechanisms, in conjunction with new knowledge on the pathophysiology of these disorders, support the concept that targeting signaling and cellular and synaptic homeostasis may be a preferred approach for ameliorating synaptic abnormalities in many NDDs.
Collapse
Affiliation(s)
- Walter E Kaufmann
- Boston Children's Hospital, Boston, MA, 02115, USA.
- Emory University School of Medicine, Atlanta, GA, 30322, USA.
| | - Skylar Luu
- Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Dejan B Budimirovic
- Kennedy Krieger Institute and Department of Psychiatry & Behavioral Sciences-Child Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
3
|
Kshetri R, Beavers JO, Hyde R, Ewa R, Schwertman A, Porcayo S, Richardson BD. Behavioral decline in Shank3 Δex4-22 mice during early adulthood parallels cerebellar granule cell glutamatergic synaptic changes. Mol Autism 2024; 15:52. [PMID: 39633421 PMCID: PMC11616285 DOI: 10.1186/s13229-024-00628-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 10/23/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND SHANK3, a gene encoding a synaptic scaffolding protein, is implicated in autism spectrum disorder (ASD) and is disrupted in Phelan-McDermid syndrome (PMS). Despite evidence of regression or worsening of ASD-like symptoms in individuals with PMS, the underlying mechanisms remain unclear. Although Shank3 is highly expressed in the cerebellar cortical granule cells, its role in cerebellar function and contribution to behavioral deficits in ASD models are unknown. This study investigates behavioral changes and cerebellar synaptic alterations in Shank3Δex4-22 mice at two developmental stages. METHODS Shank3Δex4-22 wildtype, heterozygous, and homozygous knockout mice lacking exons 4-22 (all functional isoforms) were subjected to a behavioral battery in both juvenile (5-7 weeks old) and adult (3-5 months old) mouse cohorts of both sexes. Immunostaining was used to show the expression of Shank3 in the cerebellar cortex. Spontaneous excitatory postsynaptic currents (sEPSCs) from cerebellar granule cells (CGCs) were recorded by whole-cell patch-clamp electrophysiology. RESULTS Deletion of Shank3 caused deficits in motor function, heightened anxiety, and repetitive behaviors. These genotype-dependent behavioral alterations were more prominent in adult mice than in juveniles. Reduced social preference was only identified in adult Shank3Δex4-22 knockout male mice, while self-grooming was uniquely elevated in males across both age groups. Heterozygous mice showed little to no changes in behavioral phenotypes in most behavioral tests. Immunofluorescence staining indicated the presence of Shank3 predominantly in the dendrite-containing rosette-like structures in CGCs, colocalizing with presynaptic markers of glutamatergic mossy fiber. Electrophysiological findings identified a parallel relationship between the age-related exacerbation of behavioral impairments and the enhancement of sEPSC amplitude in CGCs. LIMITATIONS Other behavioral tests of muscle strength (grip strength test), memory (Barnes/water maze), and communication (ultrasonic vocalization), were not performed. Further study is necessary to elucidate how Shank3 modulates synaptic function at the mossy fiber-granule cell synapse in the cerebellum and whether these changes shape the behavioral phenotype. CONCLUSIONS Our findings reveal an age-related exacerbation of behavioral impairments in Shank3Δex4-22 mutant mice. These results suggest that Shank3 may alter the function of glutamatergic receptors at the mossy fiber-cerebellar granule cell synapse as a potential mechanism causing cerebellar disruption in ASD.
Collapse
Affiliation(s)
- Rajaram Kshetri
- Department of Pharmacology, Southern Illinois University - School of Medicine, Springfield, IL, 62702, USA
| | - James O Beavers
- Department of Pharmacology, Southern Illinois University - School of Medicine, Springfield, IL, 62702, USA
| | - Romana Hyde
- Department of Biological Sciences, University of Idaho, Moscow, ID, 83844, USA
| | - Roseline Ewa
- Department of Pharmacology, Southern Illinois University - School of Medicine, Springfield, IL, 62702, USA
| | - Amber Schwertman
- Department of Pharmacology, Southern Illinois University - School of Medicine, Springfield, IL, 62702, USA
| | - Sarahi Porcayo
- Department of Pharmacology, Southern Illinois University - School of Medicine, Springfield, IL, 62702, USA
| | - Ben D Richardson
- Department of Pharmacology, Southern Illinois University - School of Medicine, Springfield, IL, 62702, USA.
| |
Collapse
|
4
|
Lu XY, Li MQ, Li YT, Yao JY, Zhang LX, Zeng ZH, Yu-Liu, Chen ZR, Li CQ, Zhou XF, Li F. Oral edaravone ameliorates behavioral deficits and pathologies in a valproic acid-induced rat model of autism spectrum disorder. Neuropharmacology 2024; 258:110089. [PMID: 39033904 DOI: 10.1016/j.neuropharm.2024.110089] [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: 05/07/2024] [Revised: 07/15/2024] [Accepted: 07/18/2024] [Indexed: 07/23/2024]
Abstract
Autism spectrum disorder (ASD) is neurodevelopmental disorder with a high incidence rate, characterized by social deficits and repetitive behaviors. There is currently no effective management available to treat the core symptoms of ASD; however, oxidative stress has been implicated in its pathogenesis. Edaravone (EDA), a free-radical scavenger, is used to treat amyotrophic lateral sclerosis (ALS) and acute ischemic stroke (AIS). Here, we hypothesized that an oral formula of EDA may have therapeutic efficacy in the treatment of core ASD symptoms. A rat model of autism was established by prenatal exposure to valproic acid (VPA), and the offsprings were orally treated with EDA at low (3 mg/kg), medium (10 mg/kg), and high (30 mg/kg) doses once daily for 28 days starting from postnatal day 25 (PND25). Oral EDA administration alleviated the core symptoms in VPA rats in a dose-dependent manner, including repetitive stereotypical behaviors and impaired social interaction. Furthermore, oral administration of EDA significantly reduced oxidative stress in a dose-dependent manner, as evidenced by a reduction in oxidative stress markers and an increase in antioxidants in the blood and brain. In addition, oral EDA significantly attenuated downstream pathologies, including synaptic and mitochondrial damage in the brain. Proteomic analysis further revealed that EDA corrected the imbalance in brain oxidative reduction and mitochondrial proteins induced by prenatal VPA administration. Overall, these findings demonstrate that oral EDA has therapeutic potential for ASD by targeting the oxidative stress pathway of disease pathogenesis and paves the way towards clinical studies.
Collapse
Affiliation(s)
- Xiao-Yu Lu
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha 410013, Hunan Province, China
| | - Meng-Qing Li
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha 410013, Hunan Province, China
| | | | - Jia-Yu Yao
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha 410013, Hunan Province, China
| | - Lin-Xuan Zhang
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha 410013, Hunan Province, China
| | - Ze-Hao Zeng
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha 410013, Hunan Province, China
| | - Yu-Liu
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha 410013, Hunan Province, China
| | - Zhao-Rong Chen
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha 410013, Hunan Province, China
| | - Chang-Qi Li
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha 410013, Hunan Province, China
| | - Xin-Fu Zhou
- Suzhou Auzone Biotechnology, Suzhou, China; Health and Biomedical Innovation, Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia.
| | - Fang Li
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha 410013, Hunan Province, China.
| |
Collapse
|
5
|
Ko TH, Kim Y, Jin C, Yu B, Lee M, Luong PK, Trinh TN, Yang Y, Kang H, Zhang Y, Ma R, Yoo K, Choi J, Kim JY, Woo SH, Han K, Choi JI. Shank3 Overexpression Leads to Cardiac Dysfunction in Mice by Disrupting Calcium Homeostasis in Cardiomyocytes. Korean Circ J 2024; 55:55.e7. [PMID: 39506183 DOI: 10.4070/kcj.2024.0179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/30/2024] [Accepted: 09/09/2024] [Indexed: 11/08/2024] Open
Abstract
BACKGROUND AND OBJECTIVES SH3 and multiple ankyrin repeat domains 3 (Shank3) proteins play crucial roles as neuronal postsynaptic scaffolds. Alongside neuropsychiatric symptoms, individuals with SHANK3 mutations often exhibit symptoms related to dysfunctions in other organs, including the heart. However, detailed insights into the cardiac functions of Shank3 remain limited. This study aimed to characterize the cardiac phenotypes of Shank3-overexpressing transgenic mice and explore the underlying mechanisms. METHODS Cardiac histological analysis, electrocardiogram and echocardiogram recordings were conducted on Shank3-overexpressing transgenic mice. Electrophysiological properties, including action potentials and L-type Ca²⁺ channel (LTCC) currents, were measured in isolated cardiomyocytes. Ca²⁺ homeostasis was assessed by analyzing cytosolic Ca²⁺ transients and sarcoplasmic reticulum Ca²⁺ contents. Depolarization-induced cell shortening was examined in cardiomyocytes. Immunoprecipitation followed by mass spectrometry-based identification was employed to identify proteins in the cardiac Shank3 interactome. Western blot and immunocytochemical analyses were conducted to identify changes in protein expression in Shank3-overexpressing transgenic cardiomyocytes. RESULTS The hearts of Shank3-overexpressing transgenic mice displayed reduced weight and increased fibrosis. In vivo, sudden cardiac death, arrhythmia, and contractility impairments were identified. Shank3-overexpressing transgenic cardiomyocytes showed prolonged action potential duration and increased LTCC current density. Cytosolic Ca²⁺ transients were increased with prolonged decay time, while sarcoplasmic reticulum Ca²⁺ contents remained normal. Cell shortening was augmented in Shank3-overexpressing transgenic cardiomyocytes. The cardiac Shank3 interactome comprised 78 proteins with various functions. Troponin I levels were down-regulated in Shank3-overexpressing transgenic cardiomyocytes. CONCLUSIONS This study revealed cardiac dysfunction in Shank3-overexpressing transgenic mice, potentially attributed to changes in Ca²⁺ homeostasis and contraction, with a notable reduction in troponin I.
Collapse
Affiliation(s)
- Tae Hee Ko
- Division of Cardiology, Department of Internal Medicine, Korea University College of Medicine and Korea University Anam Hospital, Seoul, Korea
| | - Yoonhee Kim
- Department of Neuroscience, Korea University College of Medicine, Seoul, Korea
| | - Chunmei Jin
- Department of Neuroscience, Korea University College of Medicine, Seoul, Korea
| | - Byeongil Yu
- Division of Cardiology, Department of Internal Medicine, Korea University College of Medicine and Korea University Anam Hospital, Seoul, Korea
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Korea
| | - Minju Lee
- Division of Cardiology, Department of Internal Medicine, Korea University College of Medicine and Korea University Anam Hospital, Seoul, Korea
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Korea
| | - Phuong Kim Luong
- Laboratory of Pathophysiology, Chungnam National University College of Pharmacy, Daejeon, Korea
| | - Tran Nguyet Trinh
- Laboratory of Pathophysiology, Chungnam National University College of Pharmacy, Daejeon, Korea
| | - Yeji Yang
- Digital Omics Research Center, Korea Basic Science Institute (KBSI), Ochang, Korea
| | - Hyojin Kang
- Division of National Supercomputing, Korea Institute of Science and Technology Information (KISTI), Daejeon, Korea
| | - Yinhua Zhang
- Department of Neuroscience, Korea University College of Medicine, Seoul, Korea
| | - Ruiying Ma
- Department of Neuroscience, Korea University College of Medicine, Seoul, Korea
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Korea
| | - Kwangmin Yoo
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Korea
- Department of Biomedical Informatics, Korea University College of Medicine, Seoul, Korea
| | - Jungmin Choi
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Korea
- Department of Biomedical Informatics, Korea University College of Medicine, Seoul, Korea
| | - Jin Young Kim
- Digital Omics Research Center, Korea Basic Science Institute (KBSI), Ochang, Korea
| | - Sun-Hee Woo
- Laboratory of Pathophysiology, Chungnam National University College of Pharmacy, Daejeon, Korea
| | - Kihoon Han
- Department of Neuroscience, Korea University College of Medicine, Seoul, Korea
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Korea.
| | - Jong-Il Choi
- Division of Cardiology, Department of Internal Medicine, Korea University College of Medicine and Korea University Anam Hospital, Seoul, Korea
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Korea.
| |
Collapse
|
6
|
Mansoorshahi S, Yetman AT, Bissell MM, Kim YY, Michelena HI, De Backer J, Mosquera LM, Hui DS, Caffarelli A, Andreassi MG, Foffa I, Guo D, Citro R, De Marco M, Tretter JT, Morris SA, Body SC, Chong JX, Bamshad MJ, Milewicz DM, Prakash SK. Whole-exome sequencing uncovers the genetic complexity of bicuspid aortic valve in families with early-onset complications. Am J Hum Genet 2024; 111:2219-2231. [PMID: 39226896 PMCID: PMC11480851 DOI: 10.1016/j.ajhg.2024.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 08/05/2024] [Accepted: 08/06/2024] [Indexed: 09/05/2024] Open
Abstract
Bicuspid aortic valve (BAV) is the most common congenital heart lesion with an estimated population prevalence of 1%. We hypothesize that specific gene variants predispose to early-onset complications of BAV (EBAV). We analyzed whole-exome sequences (WESs) to identify rare coding variants that contribute to BAV disease in 215 EBAV-affected families. Predicted damaging variants in candidate genes with moderate or strong supportive evidence to cause developmental cardiac phenotypes were present in 107 EBAV-affected families (50% of total), including genes that cause BAV (9%) or heritable thoracic aortic disease (HTAD, 19%). After appropriate filtration, we also identified 129 variants in 54 candidate genes that are associated with autosomal-dominant congenital heart phenotypes, including recurrent deleterious variation of FBN2, MYH6, channelopathy genes, and type 1 and 5 collagen genes. These findings confirm our hypothesis that unique rare genetic variants drive early-onset presentations of BAV disease.
Collapse
Affiliation(s)
- Sara Mansoorshahi
- Department of Internal Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Anji T Yetman
- Children's Hospital and Medical Center, University of Nebraska, Omaha, NE, USA
| | - Malenka M Bissell
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Yuli Y Kim
- Division of Cardiovascular Medicine, The Hospital of the University of Pennsylvania, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | | | - Julie De Backer
- Department of Cardiology and Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
| | - Laura Muiño Mosquera
- Department of Cardiology and Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
| | - Dawn S Hui
- Department of Cardiothoracic Surgery, University of Texas Health Science Center, San Antonio, TX, USA
| | - Anthony Caffarelli
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Maria G Andreassi
- Consiglio Nazionale delle Richerche (CNR), Instituto di Fisiologia Clinica, Pisa, Italy
| | - Ilenia Foffa
- Consiglio Nazionale delle Richerche (CNR), Instituto di Fisiologia Clinica, Pisa, Italy
| | - Dongchuan Guo
- Department of Internal Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Rodolfo Citro
- Cardiothoracic and Vascular Department, University Hospital "San Giovanni di Dio e Ruggi d'Aragona," Salerno, Italy
| | - Margot De Marco
- Department of Medicine, Surgery and Dentistry Schola Medica Salernitana, University of Salerno, Baronissi, Italy
| | | | - Shaine A Morris
- Department of Pediatrics, Texas Children's Hospital and Baylor College of Medicine, Houston, TX, USA
| | - Simon C Body
- Department of Anesthesiology, Boston University School of Medicine, Boston, MA, USA
| | - Jessica X Chong
- Department of Pediatrics, University of Washington, Seattle, WA, USA
| | - Michael J Bamshad
- Department of Pediatrics, University of Washington, Seattle, WA, USA
| | - Dianna M Milewicz
- Department of Internal Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Siddharth K Prakash
- Department of Internal Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA.
| |
Collapse
|
7
|
Qin L, Wang H, Ning W, Cui M, Wang Q. New advances in the diagnosis and treatment of autism spectrum disorders. Eur J Med Res 2024; 29:322. [PMID: 38858682 PMCID: PMC11163702 DOI: 10.1186/s40001-024-01916-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 06/01/2024] [Indexed: 06/12/2024] Open
Abstract
Autism spectrum disorders (ASD) are a group of neurodevelopmental disorders that affect individuals' social interactions, communication skills, and behavioral patterns, with significant individual differences and complex etiology. This article reviews the definition and characteristics of ASD, epidemiological profile, early research and diagnostic history, etiological studies, advances in diagnostic methods, therapeutic approaches and intervention strategies, social and educational integration, and future research directions. The highly heritable nature of ASD, the role of environmental factors, genetic-environmental interactions, and the need for individualized, integrated, and technology-driven treatment strategies are emphasized. Also discussed is the interaction of social policy with ASD research and the outlook for future research and treatment, including the promise of precision medicine and emerging biotechnology applications. The paper points out that despite the remarkable progress that has been made, there are still many challenges to the comprehensive understanding and effective treatment of ASD, and interdisciplinary and cross-cultural research and global collaboration are needed to further deepen the understanding of ASD and improve the quality of life of patients.
Collapse
Affiliation(s)
- Lei Qin
- Department of Rehabilitation, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong, China
| | - Haijiao Wang
- Department of Intensive Care Medicine, Feicheng People's Hospital, Taian, Shandong, China
| | - Wenjing Ning
- Department of Rehabilitation, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong, China
| | - Mengmeng Cui
- Department of Rehabilitation, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong, China.
| | - Qian Wang
- Department of Central Laboratory, The Affiliated Taian City Central Hospital of Qingdao University, Taian, China.
| |
Collapse
|
8
|
Genovese AC, Butler MG. Behavioral and Psychiatric Disorders in Syndromic Autism. Brain Sci 2024; 14:343. [PMID: 38671997 PMCID: PMC11048128 DOI: 10.3390/brainsci14040343] [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: 02/23/2024] [Revised: 03/25/2024] [Accepted: 03/28/2024] [Indexed: 04/28/2024] Open
Abstract
Syndromic autism refers to autism spectrum disorder diagnosed in the context of a known genetic syndrome. The specific manifestations of any one of these syndromic autisms are related to a clinically defined genetic syndrome that can be traced to certain genes and variants, genetic deletions, or duplications at the chromosome level. The genetic mutations or defects in single genes associated with these genetic disorders result in a significant elevation of risk for developing autism relative to the general population and are related to recurrence with inheritance patterns. Additionally, these syndromes are associated with typical behavioral characteristics or phenotypes as well as an increased risk for specific behavioral or psychiatric disorders and clinical findings. Knowledge of these associations helps guide clinicians in identifying potentially treatable conditions that can help to improve the lives of affected patients and their families.
Collapse
Affiliation(s)
- Ann C. Genovese
- Department of Psychiatry and Behavioral Sciences, University of Kansas Medical Center, Kansas City, KS 66160, USA;
| | | |
Collapse
|
9
|
Prem S, Dev B, Peng C, Mehta M, Alibutud R, Connacher RJ, St Thomas M, Zhou X, Matteson P, Xing J, Millonig JH, DiCicco-Bloom E. Dysregulation of mTOR signaling mediates common neurite and migration defects in both idiopathic and 16p11.2 deletion autism neural precursor cells. eLife 2024; 13:e82809. [PMID: 38525876 PMCID: PMC11003747 DOI: 10.7554/elife.82809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 03/04/2024] [Indexed: 03/26/2024] Open
Abstract
Autism spectrum disorder (ASD) is defined by common behavioral characteristics, raising the possibility of shared pathogenic mechanisms. Yet, vast clinical and etiological heterogeneity suggests personalized phenotypes. Surprisingly, our iPSC studies find that six individuals from two distinct ASD subtypes, idiopathic and 16p11.2 deletion, have common reductions in neural precursor cell (NPC) neurite outgrowth and migration even though whole genome sequencing demonstrates no genetic overlap between the datasets. To identify signaling differences that may contribute to these developmental defects, an unbiased phospho-(p)-proteome screen was performed. Surprisingly despite the genetic heterogeneity, hundreds of shared p-peptides were identified between autism subtypes including the mTOR pathway. mTOR signaling alterations were confirmed in all NPCs across both ASD subtypes, and mTOR modulation rescued ASD phenotypes and reproduced autism NPC-associated phenotypes in control NPCs. Thus, our studies demonstrate that genetically distinct ASD subtypes have common defects in neurite outgrowth and migration which are driven by the shared pathogenic mechanism of mTOR signaling dysregulation.
Collapse
Affiliation(s)
- Smrithi Prem
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical SchoolPiscatawayUnited States
- Graduate Program in Neuroscience, Rutgers UniversityPiscatawayUnited States
| | - Bharati Dev
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical SchoolPiscatawayUnited States
| | - Cynthia Peng
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical SchoolPiscatawayUnited States
| | - Monal Mehta
- Graduate Program in Neuroscience, Rutgers UniversityPiscatawayUnited States
- Center for Advanced Biotechnology and Medicine, Rutgers UniversityPiscatawayUnited States
| | - Rohan Alibutud
- Department of Genetics, Rutgers UniversityPiscatawayUnited States
| | - Robert J Connacher
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical SchoolPiscatawayUnited States
- Graduate Program in Neuroscience, Rutgers UniversityPiscatawayUnited States
| | - Madeline St Thomas
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical SchoolPiscatawayUnited States
- Graduate Program in Neuroscience, Rutgers UniversityPiscatawayUnited States
| | - Xiaofeng Zhou
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical SchoolPiscatawayUnited States
| | - Paul Matteson
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical SchoolPiscatawayUnited States
- Center for Advanced Biotechnology and Medicine, Rutgers UniversityPiscatawayUnited States
| | - Jinchuan Xing
- Department of Genetics, Rutgers UniversityPiscatawayUnited States
| | - James H Millonig
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical SchoolPiscatawayUnited States
- Center for Advanced Biotechnology and Medicine, Rutgers UniversityPiscatawayUnited States
| | - Emanuel DiCicco-Bloom
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical SchoolPiscatawayUnited States
- Department of Pediatrics, Rutgers Robert Wood Johnson Medical SchoolNew BrunswickUnited States
| |
Collapse
|
10
|
Tolezano GC, Bastos GC, da Costa SS, Freire BL, Homma TK, Honjo RS, Yamamoto GL, Passos-Bueno MR, Koiffmann CP, Kim CA, Vianna-Morgante AM, de Lima Jorge AA, Bertola DR, Rosenberg C, Krepischi ACV. Burden of Rare Copy Number Variants in Microcephaly: A Brazilian Cohort of 185 Microcephalic Patients and Review of the Literature. J Autism Dev Disord 2024; 54:1181-1212. [PMID: 36502452 DOI: 10.1007/s10803-022-05853-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/26/2022] [Indexed: 12/14/2022]
Abstract
Microcephaly presents heterogeneous genetic etiology linked to several neurodevelopmental disorders (NDD). Copy number variants (CNVs) are a causal mechanism of microcephaly whose investigation is a crucial step for unraveling its molecular basis. Our purpose was to investigate the burden of rare CNVs in microcephalic individuals and to review genes and CNV syndromes associated with microcephaly. We performed chromosomal microarray analysis (CMA) in 185 Brazilian patients with microcephaly and evaluated microcephalic patients carrying < 200 kb CNVs documented in the DECIPHER database. Additionally, we reviewed known genes and CNV syndromes causally linked to microcephaly through the PubMed, OMIM, DECIPHER, and ClinGen databases. Rare clinically relevant CNVs were detected in 39 out of the 185 Brazilian patients investigated by CMA (21%). In 31 among the 60 DECIPHER patients carrying < 200 kb CNVs, at least one known microcephaly gene was observed. Overall, four gene sets implicated in microcephaly were disclosed: known microcephaly genes; genes with supporting evidence of association with microcephaly; known macrocephaly genes; and novel candidates, including OTUD7A, BBC3, CNTN6, and NAA15. In the review, we compiled 957 known microcephaly genes and 58 genomic CNV loci, comprising 13 duplications and 50 deletions, which have already been associated with clinical findings including microcephaly. We reviewed genes and CNV syndromes previously associated with microcephaly, reinforced the high CMA diagnostic yield for this condition, pinpointed novel candidate loci linked to microcephaly deserving further evaluation, and provided a useful resource for future research on the field of neurodevelopment.
Collapse
Affiliation(s)
- Giovanna Cantini Tolezano
- Department of Genetics and Evolutionary Biology, Human Genome and Stem-Cell Research Center, Institute of Biosciences, University of São Paulo, 106 Rua do Matão, São Paulo, SP, 05508-090, Brazil
| | - Giovanna Civitate Bastos
- Department of Genetics and Evolutionary Biology, Human Genome and Stem-Cell Research Center, Institute of Biosciences, University of São Paulo, 106 Rua do Matão, São Paulo, SP, 05508-090, Brazil
| | - Silvia Souza da Costa
- Department of Genetics and Evolutionary Biology, Human Genome and Stem-Cell Research Center, Institute of Biosciences, University of São Paulo, 106 Rua do Matão, São Paulo, SP, 05508-090, Brazil
| | - Bruna Lucheze Freire
- Unidade de Endocrinologia Genética (LIM25), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, 455 Avenida Doutor Arnaldo, São Paulo, SP, 01246-903, Brazil
| | - Thais Kataoka Homma
- Unidade de Endocrinologia Genética (LIM25), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, 455 Avenida Doutor Arnaldo, São Paulo, SP, 01246-903, Brazil
| | - Rachel Sayuri Honjo
- Unidade de Genética do Instituto da Criança, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, 647 Avenida Doutor Enéas Carvalho de Aguiar, São Paulo, SP, 05403-900, Brazil
| | - Guilherme Lopes Yamamoto
- Department of Genetics and Evolutionary Biology, Human Genome and Stem-Cell Research Center, Institute of Biosciences, University of São Paulo, 106 Rua do Matão, São Paulo, SP, 05508-090, Brazil
- Unidade de Genética do Instituto da Criança, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, 647 Avenida Doutor Enéas Carvalho de Aguiar, São Paulo, SP, 05403-900, Brazil
| | - Maria Rita Passos-Bueno
- Department of Genetics and Evolutionary Biology, Human Genome and Stem-Cell Research Center, Institute of Biosciences, University of São Paulo, 106 Rua do Matão, São Paulo, SP, 05508-090, Brazil
| | - Celia Priszkulnik Koiffmann
- Department of Genetics and Evolutionary Biology, Human Genome and Stem-Cell Research Center, Institute of Biosciences, University of São Paulo, 106 Rua do Matão, São Paulo, SP, 05508-090, Brazil
| | - Chong Ae Kim
- Unidade de Genética do Instituto da Criança, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, 647 Avenida Doutor Enéas Carvalho de Aguiar, São Paulo, SP, 05403-900, Brazil
| | - Angela Maria Vianna-Morgante
- Department of Genetics and Evolutionary Biology, Human Genome and Stem-Cell Research Center, Institute of Biosciences, University of São Paulo, 106 Rua do Matão, São Paulo, SP, 05508-090, Brazil
| | - Alexander Augusto de Lima Jorge
- Unidade de Endocrinologia Genética (LIM25), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, 455 Avenida Doutor Arnaldo, São Paulo, SP, 01246-903, Brazil
| | - Débora Romeo Bertola
- Department of Genetics and Evolutionary Biology, Human Genome and Stem-Cell Research Center, Institute of Biosciences, University of São Paulo, 106 Rua do Matão, São Paulo, SP, 05508-090, Brazil
- Unidade de Genética do Instituto da Criança, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, 647 Avenida Doutor Enéas Carvalho de Aguiar, São Paulo, SP, 05403-900, Brazil
| | - Carla Rosenberg
- Department of Genetics and Evolutionary Biology, Human Genome and Stem-Cell Research Center, Institute of Biosciences, University of São Paulo, 106 Rua do Matão, São Paulo, SP, 05508-090, Brazil
| | - Ana Cristina Victorino Krepischi
- Department of Genetics and Evolutionary Biology, Human Genome and Stem-Cell Research Center, Institute of Biosciences, University of São Paulo, 106 Rua do Matão, São Paulo, SP, 05508-090, Brazil.
- Institute of Biosciences, University of São Paulo, 277 Rua do Matão, São Paulo, SP, 05508-090, Brazil.
| |
Collapse
|
11
|
Jimenez-Gomez A, Nguyen MX, Gill JS. Understanding the role of AMPA receptors in autism: insights from circuit and synapse dysfunction. Front Psychiatry 2024; 15:1304300. [PMID: 38352654 PMCID: PMC10861716 DOI: 10.3389/fpsyt.2024.1304300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 01/08/2024] [Indexed: 02/16/2024] Open
Abstract
Autism spectrum disorders represent a diverse etiological spectrum that converge on a syndrome characterized by discrepant deficits in developmental domains often highlighted by concerns in socialization, sensory integration, and autonomic functioning. Importantly, the incidence and prevalence of autism spectrum disorders have seen sharp increases since the syndrome was first described in the 1940s. The wide etiological spectrum and rising number of individuals being diagnosed with the condition lend urgency to capturing a more nuanced understanding of the pathogenic mechanisms underlying the autism spectrum disorders. The current review seeks to understand how the disruption of AMPA receptor (AMPAr)-mediated neurotransmission in the cerebro-cerebellar circuit, particularly in genetic autism related to SHANK3 or SYNGAP1 protein dysfunction function and autism associated with in utero exposure to the anti-seizure medications valproic acid and topiramate, may contribute to the disease presentation. Initially, a discussion contextualizing AMPAr signaling in the cerebro-cerebellar circuitry and microstructural circuit considerations is offered. Subsequently, a detailed review of the literature implicating mutations or deletions of SHANK3 and SYNGAP1 in disrupted AMPAr signaling reveals how bidirectional pathogenic modulation of this key circuit may contribute to autism. Finally, how pharmacological exposure may interact with this pathway, via increased risk of autism diagnosis with valproic acid and topiramate exposure and potential treatment of autism using AMPAr modulator perampanel, is discussed. Through the lens of the review, we will offer speculation on how neuromodulation may be used as a rational adjunct to therapy. Together, the present review seeks to synthesize the disparate considerations of circuit understanding, genetic etiology, and pharmacological modulation to understand the mechanistic interaction of this important and complex disorder.
Collapse
Affiliation(s)
- Andres Jimenez-Gomez
- Neurodevelopmental Disabilities Program, Department of Neurology, Joe DiMaggio Children’s Hospital, Hollywood, FL, United States
| | - Megan X. Nguyen
- Department of Pediatrics, Division of Neurology & Developmental Neurosciences, Baylor College of Medicine, Houston, TX, United States
- Jan & Dan Duncan Neurologic Research Institute, Texas Children’s Hospital, Houston, TX, United States
| | - Jason S. Gill
- Department of Pediatrics, Division of Neurology & Developmental Neurosciences, Baylor College of Medicine, Houston, TX, United States
- Jan & Dan Duncan Neurologic Research Institute, Texas Children’s Hospital, Houston, TX, United States
| |
Collapse
|
12
|
Kereszturi É. Diversity and Classification of Genetic Variations in Autism Spectrum Disorder. Int J Mol Sci 2023; 24:16768. [PMID: 38069091 PMCID: PMC10706722 DOI: 10.3390/ijms242316768] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 11/19/2023] [Accepted: 11/25/2023] [Indexed: 12/18/2023] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental condition with symptoms that affect the whole personality and all aspects of life. Although there is a high degree of heterogeneity in both its etiology and its characteristic behavioral patterns, the disorder is well-captured along the autistic triad. Currently, ASD status can be confirmed following an assessment of behavioral features, but there is a growing emphasis on conceptualizing autism as a spectrum, which allows for establishing a diagnosis based on the level of support need, free of discrete categories. Since ASD has a high genetic predominance, the number of genetic variations identified in the background of the condition is increasing exponentially as genetic testing methods are rapidly evolving. However, due to the huge amount of data to be analyzed, grouping the different DNA variations is still challenging. Therefore, in the present review, a multidimensional classification scheme was developed to accommodate most of the currently known genetic variants associated with autism. Genetic variations have been grouped according to six criteria (extent, time of onset, information content, frequency, number of genes involved, inheritance pattern), which are themselves not discrete categories, but form a coherent continuum in line with the autism spectrum approach.
Collapse
Affiliation(s)
- Éva Kereszturi
- Department of Molecular Biology, Semmelweis University, H-1085 Budapest, Hungary
| |
Collapse
|
13
|
Vorstman JAS, Scherer SW. Contemplating syndromic autism. Genet Med 2023; 25:100919. [PMID: 37330697 DOI: 10.1016/j.gim.2023.100919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 06/07/2023] [Accepted: 06/08/2023] [Indexed: 06/19/2023] Open
Affiliation(s)
- Jacob A S Vorstman
- Department of Psychiatry, Hospital for Sick Children University of Toronto, Toronto, ON, Canada; Program in Genetics and Genome Biology, Hospital for Sick Children, Toronto, ON, Canada; The Centre for Applied Genomics, Hospital for Sick Children, Toronto, ON, Canada.
| | - Stephen W Scherer
- Program in Genetics and Genome Biology, Hospital for Sick Children, Toronto, ON, Canada; The Centre for Applied Genomics, Hospital for Sick Children, Toronto, ON, Canada; McLaughlin Centre and Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
14
|
Kozol RA, Dallman JE. Drugs prescribed for Phelan-McDermid syndrome differentially impact sensory behaviors in shank3 zebrafish models. F1000Res 2023; 12:84. [PMID: 37868296 PMCID: PMC10589628 DOI: 10.12688/f1000research.127830.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/07/2023] [Indexed: 10/24/2023] Open
Abstract
Background: Altered sensory processing is a pervasive symptom in individuals with Autism Spectrum Disorders (ASD); people with Phelan McDermid syndrome (PMS), in particular, show reduced responses to sensory stimuli. PMS is caused by deletions of the terminal end of chromosome 22 or point mutations in Shank3. People with PMS can present with an array of symptoms including ASD, epilepsy, gastrointestinal distress, and reduced responses to sensory stimuli. People with PMS are often medicated to manage behaviors like aggression and/or self-harm and/or epilepsy, and it remains unclear how these medications might impact perception/sensory processing. Here we test this using zebrafish mutant shank3ab PMS models that likewise show reduced sensory responses in a visual motor response (VMR) assay, in which increased locomotion is triggered by light to dark transitions. Methods: We screened three medications, risperidone, lithium chloride (LiCl), and carbamazepine (CBZ), prescribed to people with PMS and one drug, 2-methyl-6-(phenylethynyl) pyridine (MPEP) tested in rodent models of PMS, for their effects on a sensory-induced behavior in two zebrafish PMS models with frameshift mutations in either the N- or C- termini. To test how pharmacological treatments affect the VMR, we exposed larvae to selected drugs for 24 hours and then quantified their locomotion during four ten-minute cycles of lights on-to-off stimuli. Results: We found that risperidone normalized the VMR in shank3 models. LiCl and CBZ had no effect on the VMR in any of the three genotypes. MPEP reduced the VMR in wildtype (WT) to levels seen in shank3 models but caused no changes in either shank3 model. Finally, shank3 mutants showed resistance to the seizure-inducing drug pentylenetetrazol (PTZ), at a dosage that results in hyperactive swimming in WT zebrafish. Conclusions: Our work shows that the effects of drugs on sensory processing are varied in ways that can be highly genotype- and drug-dependent.
Collapse
Affiliation(s)
- Robert A. Kozol
- Charles E. Schmidt College of Science, Florida Atlantic University, Jupiter, Fl., USA
| | - Julia E. Dallman
- Department of Biology, University of Miami, Coral Gables, FL, 33146, USA
| |
Collapse
|
15
|
Rogers M, Motola S, Bechichi Y, Cluzeau C, Terray T, Berent A, Panagoulias J, Duis J, Eggenspieler D, Servais L. Qualitative Insights into Key Angelman Syndrome Motor Related Concepts Reported by Caregivers-A Thematic Analysis of Semi-Structured Interviews. CHILDREN (BASEL, SWITZERLAND) 2023; 10:1462. [PMID: 37761423 PMCID: PMC10529730 DOI: 10.3390/children10091462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/17/2023] [Accepted: 08/23/2023] [Indexed: 09/29/2023]
Abstract
Previous patient-centered concept models of Angelman syndrome (AS) are integral in developing our understanding of the symptoms and impact of this condition with a holistic perspective and have highlighted the importance of motor function. We aimed to develop the motor and movement aspects of the concept models, to support research regarding motor-related digital outcomes aligned with patients' and caregivers' perspectives. We conducted a qualitative analysis of semi-structured interviews of 24 caregivers to explore AS motor-related features, factors influencing them and their impact on patients and caregivers.The most impacted motor features were gait, walking and stair-climbing. Half of caregivers ranked motor symptoms as one of the most burdensome symptoms of AS. Caregivers frequently reported physical therapy, motivation, medical management and age as factors influencing motor function in AS and reported that impaired motor function affected both patients and caregivers. Measures of lower-limb motor function were identified as relevant to monitor drug effectiveness in AS. Caregivers discussed expected benefits of a digital outcome and potential issues with wearable technology in the context of AS. We propose a new motor function patient-centered concept model, providing insights for the development of relevant, motor-related, digital outcomes in AS.
Collapse
Affiliation(s)
- Miranda Rogers
- Department of Paediatrics, MDUK Oxford Neuromuscular Centre & NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford OX1 2JD, UK;
- Sysnav Co., 27200 Vernon, France; (S.M.); (Y.B.); (C.C.); (T.T.); (D.E.)
| | - Stéphane Motola
- Sysnav Co., 27200 Vernon, France; (S.M.); (Y.B.); (C.C.); (T.T.); (D.E.)
| | - Yacine Bechichi
- Sysnav Co., 27200 Vernon, France; (S.M.); (Y.B.); (C.C.); (T.T.); (D.E.)
| | - Céline Cluzeau
- Sysnav Co., 27200 Vernon, France; (S.M.); (Y.B.); (C.C.); (T.T.); (D.E.)
| | - Tanguy Terray
- Sysnav Co., 27200 Vernon, France; (S.M.); (Y.B.); (C.C.); (T.T.); (D.E.)
| | - Allyson Berent
- Foundation for Angelman Syndrome Therapeutics (FAST), P.O. Box 40307, Austin, TX 78704, USA; (A.B.); (J.P.)
| | - Jennifer Panagoulias
- Foundation for Angelman Syndrome Therapeutics (FAST), P.O. Box 40307, Austin, TX 78704, USA; (A.B.); (J.P.)
| | - Jessica Duis
- Section of Genetics and Inherited Metabolic Disease, Department of Pediatrics, Children’s Hospital Colorado, University of Colorado Anschutz Campus, Aurora, CO 80045, USA;
| | | | - Laurent Servais
- Department of Paediatrics, MDUK Oxford Neuromuscular Centre & NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford OX1 2JD, UK;
- Department of Paediatrics, Neuromuscular Reference Center, University Hospital Liège, University of Liège, 4000 Liège, Belgium
| |
Collapse
|
16
|
Ranjbar R, Zamanzadeh Z, Ahadi AM. Effects of Venlafaxine on the Size of Brain and Expression of SHANK3, TUBB5 and DDC Genes in BALB/c Mice. PSYCHOPHARMACOLOGY BULLETIN 2023; 53:22-34. [PMID: 37601086 PMCID: PMC10434312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 08/22/2023]
Abstract
Objectives A growing body of evidence has recently suggested that taking venlafaxine during pregnancy may be linked to increased risk of certain congenital defects. The study aimed to address the effects of venlafaxine use during pregnancy on the development of the brain in mice. Experimental design Fourteen female BALB/c mice were randomly divided into two equally-sized groups: venlafaxine-treated and control. After mating, pregnant mice of venlafaxine-treated group were orally received the venlafaxine 35 mg/kg/day throughout pregnancy, while pregnant control mice did not receive any treatment. All pups were killed on postnatal day 21 and brain images were quantified using ImageJ software. The mRNA expression levels of SHANK3, TUBB5 and DDC of genes in pups' brain tissue samples were evaluated using quantitative real-time PCR method. Principal observations The mean brain size of pups was significantly smaller in the venlafaxine-treated group than in the control group. Results showed that the mRNA expression levels of SHANK3 and TUBB5 was significantly downregulated in venlafaxine-treated mice compared to control group. Expression of DDC gene didn't showed significant differences between two groups. Conclusions These results provide evidence that use of venlafaxine during pregnancy may affect the brain development in mice and altered the expression of SHANK3 and TUBB5 genes in brain tissue.
Collapse
Affiliation(s)
- Ramesh Ranjbar
- Ranjbar, PhD candidate, Department of Genetics, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Zahra Zamanzadeh
- Zamanzadeh, PhD, Department of Genetics, Faculty of Biological Sciences and Technology, Shahid Ashrafi Esfahani University, Isfahan, Iran
| | - Ali Mohammad Ahadi
- Ahadi, PhD, Department of Genetics, Faculty of Basic Sciences, Shahrekord University, Shahrekord, Iran
| |
Collapse
|
17
|
Bian WJ, González OC, de Lecea L. Adolescent sleep defects and dopaminergic hyperactivity in mice with a schizophrenia-linked Shank3 mutation. Sleep 2023; 46:zsad131. [PMID: 37144901 PMCID: PMC10334736 DOI: 10.1093/sleep/zsad131] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 03/03/2023] [Indexed: 05/06/2023] Open
Abstract
Shank3 is a shared risk gene for autism spectrum disorders and schizophrenia. Sleep defects have been characterized for autism models with Shank3 mutations; however, evidence has been lacking for the potential sleep defects caused by Shank3 mutation associated with schizophrenia and how early in development these defects may occur. Here we characterized the sleep architecture of adolescent mice carrying a schizophrenia-linked, R1117X mutation in Shank3. We further employed GRABDA dopamine sensor and fiber photometry to record dopamine release in the nucleus accumbens during sleep/wake states. Our results show that homozygous mutant R1117X mice have significantly reduced sleep in the dark phase during adolescence, altered electroencephalogram power, especially during the rapid-eye-movement sleep, and dopamine hyperactivity during sleep but not during wakefulness. Further analyses suggest that these adolescent defects in sleep architecture and dopaminergic neuromodulation tightly correlate with the social novelty preference later in adulthood and predict adult social performance during same-sex social interactions. Our results provide novel insights into the sleep phenotypes in mouse models of schizophrenia and the potential use of developmental sleep as a predictive metric for adult social symptoms. Together with recent studies in other Shank3 models, our work underscores the idea that Shank3-involved circuit disruptions may be one of the shared pathologies in certain types of schizophrenia and autism. Future research is needed to establish the causal relationship among adolescent sleep defects, dopaminergic dysregulation, and adult behavioral changes in Shank3 mutation animals and other models.
Collapse
Affiliation(s)
- Wen-Jie Bian
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Oscar C González
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Luis de Lecea
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| |
Collapse
|
18
|
Duis J, Skinner A, Carson R, Gouelle A, Annoussamy M, Silverman JL, Apkon S, Servais L, Carollo J. Quantitative measures of motor development in Angelman syndrome. Am J Med Genet A 2023; 191:1711-1721. [PMID: 37019838 PMCID: PMC11068498 DOI: 10.1002/ajmg.a.63192] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/10/2023] [Accepted: 03/10/2023] [Indexed: 04/07/2023]
Abstract
Angelman Syndrome is a rare neurodevelopmental disorder characterized by developmental delay, lack of speech, seizures, intellectual disability, characteristic behavior, and movement disorders. Clinical gait analysis provides the opportunity for movement quantification to investigate an observed maladaptive change in gait pattern and offers an objective outcome of change. Pressure-sensor-based technology, inertial and activity monitoring, and instrumented gait analysis (IGA) were employed to define motor abnormalities in Angelman syndrome. Temporal-spatial gait parameters of persons with Angelman Syndrome (pwAS) show deficiencies in gait performance through walking speed, step length, step width, and walk ratio. pwAS walk with reduced step lengths, increased step width, and greater variability. Three-dimensional motion kinematics showed increased anterior pelvic tilt, hip flexion, and knee flexion. PwAS have a walk ratio more than two standard deviations below controls. Dynamic electromyography showed prolonged activation of knee extensors, which was associated with a decreased range of motion and the presence of hip flexion contractures. Use of multiple gait tracking modalities revealed that pwAS exhibit a change in gait pattern to a flexed knee gait pattern. Cross-sectional studies of individuals with AS show a regression toward this maladaptive gait pattern over development in pwAS ages 4-11. PwAS unexpectedly did not have spasticity associated with change in gait pattern. Multiple quantitative measures of motor patterning may offer early biomarkers of gait decline consistent with critical periods of intervention, insight into appropriate management strategies, objective primary outcomes, and early indicators of adverse events.
Collapse
Affiliation(s)
- Jessica Duis
- Center for Gait & Movement Analysis (CGMA), Children’s Hospital Colorado, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Section of Genetics and Inherited Metabolic Disease, Department of Pediatrics, Children’s Hospital Colorado, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Section of Pediatrics, Special Care Clinic, Department of Pediatrics, Children’s Hospital Colorado, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Austin Skinner
- Center for Gait & Movement Analysis (CGMA), Children’s Hospital Colorado, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Robert Carson
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Arnaud Gouelle
- Université de Reims Champagne-Ardenne, PSMS (Performance, Santé, Métrologie, Société), Reims, France
- Gait and Balance Academy, ProtoKinetics, Havertown, Pennsylvania, USA
| | | | - Jill L. Silverman
- MIND Institute and Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, California, USA
| | - Susan Apkon
- Department of Physical Medicine & Rehabilitation, Children’s Hospital Colorado, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Laurent Servais
- Department of Paediatrics, Oxford University, Oxford, UK
- Division of Child Neurology, Centre de References des Maladies Neuromusculaires, Department of Pediatrics, University Hospital Liege & University of Liege, Liege, Belgium
| | - James Carollo
- Center for Gait & Movement Analysis (CGMA), Children’s Hospital Colorado, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Department of Physical Medicine & Rehabilitation, Children’s Hospital Colorado, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
19
|
Maurer JJ, Choi A, An I, Sathi N, Chung S. Sleep disturbances in autism spectrum disorder: Animal models, neural mechanisms, and therapeutics. Neurobiol Sleep Circadian Rhythms 2023; 14:100095. [PMID: 37188242 PMCID: PMC10176270 DOI: 10.1016/j.nbscr.2023.100095] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 03/16/2023] [Accepted: 04/08/2023] [Indexed: 05/17/2023] Open
Abstract
Sleep is crucial for brain development. Sleep disturbances are prevalent in children with autism spectrum disorder (ASD). Strikingly, these sleep problems are positively correlated with the severity of ASD core symptoms such as deficits in social skills and stereotypic behavior, indicating that sleep problems and the behavioral characteristics of ASD may be related. In this review, we will discuss sleep disturbances in children with ASD and highlight mouse models to study sleep disturbances and behavioral phenotypes in ASD. In addition, we will review neuromodulators controlling sleep and wakefulness and how these neuromodulatory systems are disrupted in animal models and patients with ASD. Lastly, we will address how the therapeutic interventions for patients with ASD improve various aspects of sleep. Together, gaining mechanistic insights into the neural mechanisms underlying sleep disturbances in children with ASD will help us to develop better therapeutic interventions.
Collapse
|
20
|
Cope EC, Wang SH, Waters RC, Gore IR, Vasquez B, Laham BJ, Gould E. Activation of the CA2-ventral CA1 pathway reverses social discrimination dysfunction in Shank3B knockout mice. Nat Commun 2023; 14:1750. [PMID: 36991001 PMCID: PMC10060401 DOI: 10.1038/s41467-023-37248-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 03/07/2023] [Indexed: 03/30/2023] Open
Abstract
Mutation or deletion of the SHANK3 gene, which encodes a synaptic scaffolding protein, is linked to autism spectrum disorder and Phelan-McDermid syndrome, conditions associated with social memory impairments. Shank3B knockout mice also exhibit social memory deficits. The CA2 region of the hippocampus integrates numerous inputs and sends a major output to the ventral CA1 (vCA1). Despite finding few differences in excitatory afferents to the CA2 in Shank3B knockout mice, we found that activation of CA2 neurons as well as the CA2-vCA1 pathway restored social recognition function to wildtype levels. vCA1 neuronal oscillations have been linked to social memory, but we observed no differences in these measures between wildtype and Shank3B knockout mice. However, activation of the CA2 enhanced vCA1 theta power in Shank3B knockout mice, concurrent with behavioral improvements. These findings suggest that stimulating adult circuitry in a mouse model with neurodevelopmental impairments can invoke latent social memory function.
Collapse
Affiliation(s)
- Elise C Cope
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, 08544, USA
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
| | - Samantha H Wang
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, 08544, USA
| | - Renée C Waters
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, 08544, USA
| | - Isha R Gore
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, 08544, USA
| | - Betsy Vasquez
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, 08544, USA
| | - Blake J Laham
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, 08544, USA
| | - Elizabeth Gould
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, 08544, USA.
| |
Collapse
|
21
|
Moffitt BA, Sarasua SM, Ivankovic D, Ward LD, Valentine K, Bennett WE, Rogers C, Phelan K, Boccuto L. Stratification of a Phelan-McDermid Syndrome Population Based on Their Response to Human Growth Hormone and Insulin-like Growth Factor. Genes (Basel) 2023; 14:490. [PMID: 36833418 PMCID: PMC9956088 DOI: 10.3390/genes14020490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 02/11/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023] Open
Abstract
Phelan-McDermid syndrome (PMS), caused by pathogenic variants in the SHANK3 gene or 22q13 deletions, is characterized by intellectual disability, autistic features, developmental delays, and neonatal hypotonia. Insulin-like growth factor 1 (IGF-1) and human growth hormone (hGH) have been shown to reverse neurobehavioral deficits in PMS. We assessed the metabolic profiling of 48 individuals with PMS and 50 controls and determined subpopulations by taking the top and bottom 25% of responders to hGH and IGF-1. A distinct metabolic profile for individuals with PMS showed a reduced ability to metabolize major energy sources and a higher metabolism of alternative energy sources. The analysis of the metabolic response to hGH or IGF-1 highlighted a major overlap between both high and low responders, validating the model and suggesting that the two growth factors share many target pathways. When we investigated the effect of hGH and IGF-1 on the metabolism of glucose, the correlation between the high-responder subgroups showed less similarity, whereas the low-responders were still relatively similar. Classification of individuals with PMS into subgroups based on responses to a compound can allow an investigation into pathogenic mechanisms, the identification of molecular biomarkers, an exploration of in vitro responses to candidate drugs, and eventually the selection of better candidates for clinical trials.
Collapse
Affiliation(s)
- Bridgette A. Moffitt
- School of Nursing, Healthcare Genetics Program, Clemson University, Clemson, SC 29634, USA
| | - Sara M. Sarasua
- School of Nursing, Healthcare Genetics Program, Clemson University, Clemson, SC 29634, USA
| | - Diana Ivankovic
- School of Nursing, Healthcare Genetics Program, Clemson University, Clemson, SC 29634, USA
| | - Linda D. Ward
- School of Nursing, Healthcare Genetics Program, Clemson University, Clemson, SC 29634, USA
| | - Kathleen Valentine
- School of Nursing, Healthcare Genetics Program, Clemson University, Clemson, SC 29634, USA
| | - William E. Bennett
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Indiana University, Riley Hospital for Children, Indianapolis, IN 46202, USA
| | | | - Katy Phelan
- Genetics Laboratory, Florida Cancer Specialists & Research Institute, Fort Myers, FL 33916, USA
| | - Luigi Boccuto
- School of Nursing, Healthcare Genetics Program, Clemson University, Clemson, SC 29634, USA
| |
Collapse
|
22
|
Huang M, Qi Q, Xu T. Targeting Shank3 deficiency and paresthesia in autism spectrum disorder: A brief review. Front Mol Neurosci 2023; 16:1128974. [PMID: 36846568 PMCID: PMC9948097 DOI: 10.3389/fnmol.2023.1128974] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 01/18/2023] [Indexed: 02/11/2023] Open
Abstract
Autism spectrum disorder (ASD) includes a group of multifactorial neurodevelopmental disorders characterized by impaired social communication, social interaction, and repetitive behaviors. Several studies have shown an association between cases of ASD and mutations in the genes of SH3 and multiple ankyrin repeat domain protein 3 (SHANK3). These genes encode many cell adhesion molecules, scaffold proteins, and proteins involved in synaptic transcription, protein synthesis, and degradation. They have a profound impact on all aspects of synaptic transmission and plasticity, including synapse formation and degeneration, suggesting that the pathogenesis of ASD may be partially attributable to synaptic dysfunction. In this review, we summarize the mechanism of synapses related to Shank3 in ASD. We also discuss the molecular, cellular, and functional studies of experimental models of ASD and current autism treatment methods targeting related proteins.
Collapse
Affiliation(s)
- Min Huang
- Department of Anesthesiology, Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China,Department of Anesthesiology, Suzhou Hospital of Anhui Medical University, Suzhou, China
| | - Qi Qi
- Department of Anesthesiology, Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China,Department of Anesthesiology, Suzhou Hospital of Anhui Medical University, Suzhou, China
| | - Tao Xu
- Department of Anesthesiology, Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China,Department of Anesthesiology, Suzhou Hospital of Anhui Medical University, Suzhou, China,*Correspondence: Tao Xu,
| |
Collapse
|
23
|
Haploinsufficiency of Shank3 increases the orientation selectivity of V1 neurons. Sci Rep 2022; 12:22230. [PMID: 36564435 PMCID: PMC9789112 DOI: 10.1038/s41598-022-26402-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 12/14/2022] [Indexed: 12/24/2022] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder whose hallmarks are social deficits, language impairment, repetitive behaviors, and sensory alterations. It has been reported that patients with ASD show differential activity in cortical regions, for instance, increased neuronal activity in visual processing brain areas and atypical visual perception compared with healthy subjects. The causes of these alterations remain unclear, although many studies demonstrate that ASD has a strong genetic correlation. An example is Phelan-McDermid syndrome, caused by a deletion of the Shank3 gene in one allele of chromosome 22. However, the neuronal consequences relating to the haploinsufficiency of Shank3 in the brain remain unknown. Given that sensory abnormalities are often present along with the core symptoms of ASD, our goal was to study the tuning properties of the primary visual cortex to orientation and direction in awake, head-fixed Shank3+/- mice. We recorded neural activity in vivo in response to visual gratings in the primary visual cortex from a mouse model of ASD (Shank3+/- mice) using the genetically encoded calcium indicator GCaMP6f, imaged with a two-photon microscope through a cranial window. We found that Shank3+/- mice showed a higher proportion of neurons responsive to drifting gratings stimuli than wild-type mice. Shank3+/- mice also show increased responses to some specific stimuli. Furthermore, analyzing the distributions of neurons for the tuning width, we found that Shank3+/- mice have narrower tuning widths, which was corroborated by analyzing the orientation selectivity. Regarding this, Shank3+/- mice have a higher proportion of selective neurons, specifically neurons showing increased selectivity to orientation but not direction. Thus, the haploinsufficiency of Shank3 modified the neuronal response of the primary visual cortex.
Collapse
|
24
|
Bouquier N, Sakkaki S, Raynaud F, Hemonnot-Girard AL, Seube V, Compan V, Bertaso F, Perroy J, Moutin E. The Shank3 Venus/Venus knock in mouse enables isoform-specific functional studies of Shank3a. Front Neurosci 2022; 16:1081010. [PMID: 36570823 PMCID: PMC9773256 DOI: 10.3389/fnins.2022.1081010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 11/23/2022] [Indexed: 12/14/2022] Open
Abstract
Background Shank3 is a scaffolding protein essential for the organization and function of the glutamatergic postsynapse. Monogenic mutations in SHANK3 gene are among the leading genetic causes of Autism Spectrum Disorders (ASD). The multiplicity of Shank3 isoforms seems to generate as much functional diversity and yet, there are no tools to study endogenous Shank3 proteins in an isoform-specific manner. Methods In this study, we created a novel transgenic mouse line, the Shank3Venus/Venus knock in mouse, which allows to monitor the endogenous expression of the major Shank3 isoform in the brain, the full-length Shank3a isoform. Results We show that the endogenous Venus-Shank3a protein is localized in spines and is mainly expressed in the striatum, hippocampus and cortex of the developing and adult brain. We show that Shank3Venus/+ and Shank3Venus/Venus mice have no behavioral deficiency. We further crossed Shank3Venus/Venus mice with Shank3ΔC/ΔC mice, a model of ASD, to track the Venus-tagged wild-type copy of Shank3a in physiological (Shank3Venus/+) and pathological (Shank3Venus/ΔC) conditions. We report a developmental delay in brain expression of the Venus-Shank3a isoform in Shank3Venus/ΔC mice, compared to Shank3Venus/+ control mice. Conclusion Altogether, our results show that the Shank3Venus/Venus mouse line is a powerful tool to study endogenous Shank3a expression, in physiological conditions and in ASD.
Collapse
Affiliation(s)
- Nathalie Bouquier
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Sophie Sakkaki
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Fabrice Raynaud
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France,PhyMedExp, Univ Montpellier, INSERM, CNRS, CHU de Montpellier, Montpellier, France
| | | | - Vincent Seube
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Vincent Compan
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Federica Bertaso
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Julie Perroy
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France,*Correspondence: Julie Perroy,
| | - Enora Moutin
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France,Enora Moutin,
| |
Collapse
|
25
|
Groves Kuhnle C, Grimes M, Suárez Casanova VM, Turrigiano GG, Van Hooser SD. Juvenile Shank3 KO Mice Adopt Distinct Hunting Strategies during Prey Capture Learning. eNeuro 2022; 9:ENEURO.0230-22.2022. [PMID: 36446569 PMCID: PMC9768843 DOI: 10.1523/eneuro.0230-22.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 10/11/2022] [Accepted: 10/23/2022] [Indexed: 12/02/2022] Open
Abstract
Mice are opportunistic omnivores that readily learn to hunt and eat insects such as crickets. The details of how mice learn these behaviors and how these behaviors may differ in strains with altered neuroplasticity are unclear. We quantified the behavior of juvenile wild-type (WT) and Shank3 knock-out (KO) mice as they learned to hunt crickets during the critical period for ocular dominance plasticity. This stage involves heightened cortical plasticity including homeostatic synaptic scaling, which requires Shank3, a glutamatergic synaptic protein that, when mutated, produces Phelan-McDermid syndrome and is often comorbid with autism spectrum disorder (ASD). Both strains showed interest in examining live and dead crickets and learned to hunt. Shank3 knock-out mice took longer to become proficient, and, after 5 d, did not achieve the efficiency of wild-type mice in either time-to-capture or distance-to-capture. Shank3 knock-out mice also exhibited different characteristics when pursuing crickets that could not be explained by a simple motor deficit. Although both genotypes moved at the same average speed when approaching a cricket, Shank3 KO mice paused more often, did not begin final accelerations toward crickets as early, and did not close the distance gap to the cricket as quickly as wild-type mice. These differences in Shank3 KO mice are reminiscent of some behavioral characteristics of individuals with ASD as they perform complex tasks, such as slower action initiation and completion. This paradigm will be useful for exploring the neural circuit mechanisms that underlie these learning and performance differences in monogenic ASD rodent models.
Collapse
Affiliation(s)
| | - Micaela Grimes
- Department of Biology, Brandeis University, Waltham, MA 02453
| | | | | | | |
Collapse
|
26
|
Lord JS, Gay SM, Harper KM, Nikolova VD, Smith KM, Moy SS, Diering GH. Early life sleep disruption potentiates lasting sex-specific changes in behavior in genetically vulnerable Shank3 heterozygous autism model mice. Mol Autism 2022; 13:35. [PMID: 36038911 PMCID: PMC9425965 DOI: 10.1186/s13229-022-00514-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 08/11/2022] [Indexed: 12/24/2022] Open
Abstract
Background Patients with autism spectrum disorder (ASD) experience high rates of sleep disruption beginning early in life; however, the developmental consequences of this disruption are not understood. We examined sleep behavior and the consequences of sleep disruption in developing mice bearing C-terminal truncation mutation in the high-confidence ASD risk gene SHANK3 (Shank3ΔC). We hypothesized that sleep disruption may be an early sign of developmental divergence, and that clinically relevant Shank3WT/ΔC mice may be at increased risk of lasting deleterious outcomes following early life sleep disruption. Methods We recorded sleep behavior in developing Shank3ΔC/ΔC, Shank3WT/ΔC, and wild-type siblings of both sexes using a noninvasive home-cage monitoring system. Separately, litters of Shank3WT/ΔC and wild-type littermates were exposed to automated mechanical sleep disruption for 7 days prior to weaning (early life sleep disruption: ELSD) or post-adolescence (PASD) or undisturbed control (CON) conditions. All groups underwent standard behavioral testing as adults. Results Male and female Shank3ΔC/ΔC mice slept significantly less than wild-type and Shank3WT/ΔC siblings shortly after weaning, with increasing sleep fragmentation in adolescence, indicating that sleep disruption has a developmental onset in this ASD model. ELSD treatment interacted with genetic vulnerability in Shank3WT/ΔC mice, resulting in lasting, sex-specific changes in behavior, whereas wild-type siblings were largely resilient to these effects. Male ELSD Shank3WT/ΔC subjects demonstrated significant changes in sociability, sensory processing, and locomotion, while female ELSD Shank3WT/ΔC subjects had a significant reduction in risk aversion. CON Shank3WT/ΔC mice, PASD mice, and all wild-type mice demonstrated typical behavioral responses in most tests. Limitations This study tested the interaction between developmental sleep disruption and genetic vulnerability using a single ASD mouse model: Shank3ΔC (deletion of exon 21). The broader implications of this work should be supported by additional studies using ASD model mice with distinct genetic vulnerabilities. Conclusion Our study shows that sleep disruption during sensitive periods of early life interacts with underlying genetic vulnerability to drive lasting and sex-specific changes in behavior. As individuals progress through maturation, they gain resilience to the lasting effects of sleep disruption. This work highlights developmental sleep disruption as an important vulnerability in ASD susceptibility. Supplementary Information The online version contains supplementary material available at 10.1186/s13229-022-00514-5.
Collapse
Affiliation(s)
- Julia S Lord
- Department of Cell Biology and Physiology and the Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Sean M Gay
- Department of Cell Biology and Physiology and the Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Kathryn M Harper
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Viktoriya D Nikolova
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kirsten M Smith
- Department of Cell Biology and Physiology and the Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Sheryl S Moy
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA. .,Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Graham H Diering
- Department of Cell Biology and Physiology and the Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA. .,Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
27
|
Nehme R, Pietiläinen O, Artomov M, Tegtmeyer M, Valakh V, Lehtonen L, Bell C, Singh T, Trehan A, Sherwood J, Manning D, Peirent E, Malik R, Guss EJ, Hawes D, Beccard A, Bara AM, Hazelbaker DZ, Zuccaro E, Genovese G, Loboda AA, Neumann A, Lilliehook C, Kuismin O, Hamalainen E, Kurki M, Hultman CM, Kähler AK, Paulo JA, Ganna A, Madison J, Cohen B, McPhie D, Adolfsson R, Perlis R, Dolmetsch R, Farhi S, McCarroll S, Hyman S, Neale B, Barrett LE, Harper W, Palotie A, Daly M, Eggan K. The 22q11.2 region regulates presynaptic gene-products linked to schizophrenia. Nat Commun 2022; 13:3690. [PMID: 35760976 PMCID: PMC9237031 DOI: 10.1038/s41467-022-31436-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 06/08/2022] [Indexed: 12/30/2022] Open
Abstract
It is unclear how the 22q11.2 deletion predisposes to psychiatric disease. To study this, we generated induced pluripotent stem cells from deletion carriers and controls and utilized CRISPR/Cas9 to introduce the heterozygous deletion into a control cell line. Here, we show that upon differentiation into neural progenitor cells, the deletion acted in trans to alter the abundance of transcripts associated with risk for neurodevelopmental disorders including autism. In excitatory neurons, altered transcripts encoded presynaptic factors and were associated with genetic risk for schizophrenia, including common and rare variants. To understand how the deletion contributed to these changes, we defined the minimal protein-protein interaction network that best explains gene expression alterations. We found that many genes in 22q11.2 interact in presynaptic, proteasome, and JUN/FOS transcriptional pathways. Our findings suggest that the 22q11.2 deletion impacts genes that may converge with psychiatric risk loci to influence disease manifestation in each deletion carrier.
Collapse
Affiliation(s)
- Ralda Nehme
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA.
- Department of Stem Cell and Regenerative Biology, and the Harvard Institute for Stem Cell Biology, Harvard University, Cambridge, MA, 02138, USA.
| | - Olli Pietiläinen
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA.
- Department of Stem Cell and Regenerative Biology, and the Harvard Institute for Stem Cell Biology, Harvard University, Cambridge, MA, 02138, USA.
| | - Mykyta Artomov
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Matthew Tegtmeyer
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
- Department of Stem Cell and Regenerative Biology, and the Harvard Institute for Stem Cell Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Vera Valakh
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
- Department of Stem Cell and Regenerative Biology, and the Harvard Institute for Stem Cell Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Leevi Lehtonen
- Institute for Molecular Medicine Finland, University of Helsinki, FI-00014, Helsinki, Finland
| | - Christina Bell
- Department of Cell Biology, Blavatnik Institute of Harvard Medical School, Boston, MA, USA
| | - Tarjinder Singh
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Aditi Trehan
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
- Department of Stem Cell and Regenerative Biology, and the Harvard Institute for Stem Cell Biology, Harvard University, Cambridge, MA, 02138, USA
| | - John Sherwood
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
- Department of Stem Cell and Regenerative Biology, and the Harvard Institute for Stem Cell Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Danielle Manning
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Emily Peirent
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
- Department of Stem Cell and Regenerative Biology, and the Harvard Institute for Stem Cell Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Rhea Malik
- Department of Stem Cell and Regenerative Biology, and the Harvard Institute for Stem Cell Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Ellen J Guss
- Department of Stem Cell and Regenerative Biology, and the Harvard Institute for Stem Cell Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Derek Hawes
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
- Department of Stem Cell and Regenerative Biology, and the Harvard Institute for Stem Cell Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Amanda Beccard
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Anne M Bara
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
- Department of Stem Cell and Regenerative Biology, and the Harvard Institute for Stem Cell Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Dane Z Hazelbaker
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Emanuela Zuccaro
- Department of Stem Cell and Regenerative Biology, and the Harvard Institute for Stem Cell Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Giulio Genovese
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Alexander A Loboda
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA
- ITMO University, St. Petersburg, Russia
- Almazov National Medical Research Centre, Saint-Petersburg, Russia
| | - Anna Neumann
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Christina Lilliehook
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Outi Kuismin
- Psychiatric & Neurodevelopmental Genetics Unit, Massachusetts General Hospital, Boston, MA, 02114, USA
- PEDEGO Research Unit, University of Oulu, FI-90014, Oulu, Finland
- Medical Research Center, Oulu University Hospital, FI-90014, Oulu, Finland
- Department of Clinical Genetics, Oulu University Hospital, 90220, Oulu, Finland
| | - Eija Hamalainen
- Institute for Molecular Medicine Finland, University of Helsinki, FI-00014, Helsinki, Finland
| | - Mitja Kurki
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
- Institute for Molecular Medicine Finland, University of Helsinki, FI-00014, Helsinki, Finland
- Psychiatric & Neurodevelopmental Genetics Unit, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Christina M Hultman
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, SE-171 77, Stockholm, Sweden
| | - Anna K Kähler
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, SE-171 77, Stockholm, Sweden
| | - Joao A Paulo
- Department of Cell Biology, Blavatnik Institute of Harvard Medical School, Boston, MA, USA
| | - Andrea Ganna
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Jon Madison
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Bruce Cohen
- Department of Psychiatry, McLean Hospital, Belmont, MA, 02478, USA
| | - Donna McPhie
- Department of Psychiatry, McLean Hospital, Belmont, MA, 02478, USA
| | - Rolf Adolfsson
- Umea University, Faculty of Medicine, Department of Clinical Sciences, Psychiatry, 901 85, Umea, Sweden
| | - Roy Perlis
- Psychiatry Dept., Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Ricardo Dolmetsch
- Novartis Institutes for Biomedical Research, Novartis, Cambridge, MA, 02139, USA
| | - Samouil Farhi
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Steven McCarroll
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Steven Hyman
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
- Department of Stem Cell and Regenerative Biology, and the Harvard Institute for Stem Cell Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Ben Neale
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Lindy E Barrett
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
- Department of Stem Cell and Regenerative Biology, and the Harvard Institute for Stem Cell Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Wade Harper
- Department of Cell Biology, Blavatnik Institute of Harvard Medical School, Boston, MA, USA
| | - Aarno Palotie
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
- Institute for Molecular Medicine Finland, University of Helsinki, FI-00014, Helsinki, Finland
- Psychiatric & Neurodevelopmental Genetics Unit, Massachusetts General Hospital, Boston, MA, 02114, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Mark Daly
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA
- Institute for Molecular Medicine Finland, University of Helsinki, FI-00014, Helsinki, Finland
- Psychiatric & Neurodevelopmental Genetics Unit, Massachusetts General Hospital, Boston, MA, 02114, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Kevin Eggan
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA.
- Department of Stem Cell and Regenerative Biology, and the Harvard Institute for Stem Cell Biology, Harvard University, Cambridge, MA, 02138, USA.
- BioMarin Pharmaceutical, San Rafael, CA, 94901, USA.
| |
Collapse
|
28
|
Malara M, Lutz AK, Incearap B, Bauer HF, Cursano S, Volbracht K, Lerner JJ, Pandey R, Delling JP, Ioannidis V, Arévalo AP, von Bernhardi JE, Schön M, Bockmann J, Dimou L, Boeckers TM. SHANK3 deficiency leads to myelin defects in the central and peripheral nervous system. Cell Mol Life Sci 2022; 79:371. [PMID: 35726031 PMCID: PMC9209365 DOI: 10.1007/s00018-022-04400-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 05/11/2022] [Accepted: 05/25/2022] [Indexed: 01/04/2023]
Abstract
Mutations or deletions of the SHANK3 gene are causative for Phelan–McDermid syndrome (PMDS), a syndromic form of autism spectrum disorders (ASDs). We analyzed Shank3Δ11(−/−) mice and organoids from PMDS individuals to study effects on myelin. SHANK3 was found to be expressed in oligodendrocytes and Schwann cells, and MRI analysis of Shank3Δ11(−/−) mice revealed a reduced volume of the corpus callosum as seen in PMDS patients. Myelin proteins including myelin basic protein showed significant temporal and regional differences with lower levels in the CNS but increased amounts in the PNS of Shank3Δ11(−/−) animals. Node, as well as paranode, lengths were increased and ultrastructural analysis revealed region-specific alterations of the myelin sheaths. In PMDS hiPSC-derived cerebral organoids we observed an altered number and delayed maturation of myelinating cells. These findings provide evidence that, in addition to a synaptic deregulation, impairment of myelin might profoundly contribute to the clinical manifestation of SHANK3 deficiency.
Collapse
Affiliation(s)
- Mariagiovanna Malara
- Institute for Anatomy and Cell Biology, Ulm University, Albert-Einstein Allee 11, 89081, Ulm, Germany
- International Graduate School in Molecular Medicine, IGradU, 89081, Ulm, Germany
| | - Anne-Kathrin Lutz
- Institute for Anatomy and Cell Biology, Ulm University, Albert-Einstein Allee 11, 89081, Ulm, Germany
| | - Berra Incearap
- Institute for Anatomy and Cell Biology, Ulm University, Albert-Einstein Allee 11, 89081, Ulm, Germany
- International Graduate School in Molecular Medicine, IGradU, 89081, Ulm, Germany
| | - Helen Friedericke Bauer
- Institute for Anatomy and Cell Biology, Ulm University, Albert-Einstein Allee 11, 89081, Ulm, Germany
- International Graduate School in Molecular Medicine, IGradU, 89081, Ulm, Germany
| | - Silvia Cursano
- Institute for Anatomy and Cell Biology, Ulm University, Albert-Einstein Allee 11, 89081, Ulm, Germany
| | - Katrin Volbracht
- Molecular and Translational Neuroscience, Department of Neurology, Ulm University, 89081, Ulm, Germany
| | - Joanna Janina Lerner
- Institute for Anatomy and Cell Biology, Ulm University, Albert-Einstein Allee 11, 89081, Ulm, Germany
- International Graduate School in Molecular Medicine, IGradU, 89081, Ulm, Germany
| | - Rakshita Pandey
- Institute for Anatomy and Cell Biology, Ulm University, Albert-Einstein Allee 11, 89081, Ulm, Germany
| | - Jan Philipp Delling
- Institute for Anatomy and Cell Biology, Ulm University, Albert-Einstein Allee 11, 89081, Ulm, Germany
| | - Valentin Ioannidis
- Institute for Anatomy and Cell Biology, Ulm University, Albert-Einstein Allee 11, 89081, Ulm, Germany
| | - Andrea Pérez Arévalo
- Institute for Anatomy and Cell Biology, Ulm University, Albert-Einstein Allee 11, 89081, Ulm, Germany
| | | | - Michael Schön
- Institute for Anatomy and Cell Biology, Ulm University, Albert-Einstein Allee 11, 89081, Ulm, Germany
| | - Jürgen Bockmann
- Institute for Anatomy and Cell Biology, Ulm University, Albert-Einstein Allee 11, 89081, Ulm, Germany
| | - Leda Dimou
- Molecular and Translational Neuroscience, Department of Neurology, Ulm University, 89081, Ulm, Germany
| | - Tobias M Boeckers
- Institute for Anatomy and Cell Biology, Ulm University, Albert-Einstein Allee 11, 89081, Ulm, Germany.
- DZNE, Ulm Site, 89081, Ulm, Germany.
| |
Collapse
|
29
|
Neklyudova A, Smirnov K, Rebreikina A, Martynova O, Sysoeva O. Electrophysiological and Behavioral Evidence for Hyper- and Hyposensitivity in Rare Genetic Syndromes Associated with Autism. Genes (Basel) 2022; 13:671. [PMID: 35456477 PMCID: PMC9027402 DOI: 10.3390/genes13040671] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/29/2022] [Accepted: 04/05/2022] [Indexed: 01/27/2023] Open
Abstract
Our study reviewed abnormalities in spontaneous, as well as event-related, brain activity in syndromes with a known genetic underpinning that are associated with autistic symptomatology. Based on behavioral and neurophysiological evidence, we tentatively subdivided the syndromes on primarily hyper-sensitive (Fragile X, Angelman) and hypo-sensitive (Phelan-McDermid, Rett, Tuberous Sclerosis, Neurofibromatosis 1), pointing to the way of segregation of heterogeneous idiopathic ASD, that includes both hyper-sensitive and hypo-sensitive individuals. This segmentation links abnormalities in different genes, such as FMR1, UBE3A, GABRB3, GABRA5, GABRG3, SHANK3, MECP2, TSC1, TSC2, and NF1, that are causative to the above-mentioned syndromes and associated with synaptic transmission and cell growth, as well as with translational and transcriptional regulation and with sensory sensitivity. Excitation/inhibition imbalance related to GABAergic signaling, and the interplay of tonic and phasic inhibition in different brain regions might underlie this relationship. However, more research is needed. As most genetic syndromes are very rare, future investigations in this field will benefit from multi-site collaboration with a common protocol for electrophysiological and event-related potential (EEG/ERP) research that should include an investigation into all modalities and stages of sensory processing, as well as potential biomarkers of GABAergic signaling (such as 40-Hz ASSR).
Collapse
Affiliation(s)
- Anastasia Neklyudova
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Science, 117485 Moscow, Russia; (A.N.); (K.S.); (A.R.); (O.M.)
| | - Kirill Smirnov
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Science, 117485 Moscow, Russia; (A.N.); (K.S.); (A.R.); (O.M.)
| | - Anna Rebreikina
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Science, 117485 Moscow, Russia; (A.N.); (K.S.); (A.R.); (O.M.)
- Sirius Center for Cognitive Research, Sirius University of Science and Technology, 354340 Sochi, Russia
| | - Olga Martynova
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Science, 117485 Moscow, Russia; (A.N.); (K.S.); (A.R.); (O.M.)
| | - Olga Sysoeva
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Science, 117485 Moscow, Russia; (A.N.); (K.S.); (A.R.); (O.M.)
- Sirius Center for Cognitive Research, Sirius University of Science and Technology, 354340 Sochi, Russia
| |
Collapse
|
30
|
Xu Y, Wang Y, Tong G, Li L, Cheng J, Zhang L, Xu Q, Wang L, Zhang P. Expression of SH3 and Multiple Ankyrin Repeat Domains Protein 3 in Mouse Retina. Front Cell Neurosci 2022; 16:795668. [PMID: 35401120 PMCID: PMC8990853 DOI: 10.3389/fncel.2022.795668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 02/23/2022] [Indexed: 11/13/2022] Open
Abstract
Synapse-associated gene mutations of SH3 and multiple ankyrin repeat domains protein 3 (SHANK3) may lead to autism spectrum disorder (ASD). In some ASD cases, patients may also have vision disorders. However, the effects of SHANK3 in the retina are barely mentioned in the literature. In this study, we used wild-type mice to systematically map the distribution of SHANK3 expression in entire mouse retinas. Using Western blot analysis and immunofluorescence double labeling, we identified a large number of prominent cells expressing high levels of SHANK3 in the inner retina, in particular, the ganglion cell layer (GCL) and inner nucleus layer. The inner plexiform layer and outer nucleus layer were also exhibited positive SHANK3 signals. In the inner layer, GABAergic amacrine cells (ACs) labeled by glutamate decarboxylase were colocalized with SHANK3-positive cells. Dopaminergic ACs (labeled by tyrosine hydroxylase) and cholinergic ACs (labeled by choline acetyltransferase) were also found to contain SHANK3-positive signals. Additionally, most GCs (labeled by Brn3a) were also found to be SHANK3 positive. In the outer retina, bipolar cells (labeled by homeobox protein ChX10) and horizontal cells (labeled by calbindin) were SHANK3 positive. In the outer nucleus layers, the somata of blue cones (labeled by S-opsin) were weekly co-labeled with SHANK3. The inner segments of blue cones and the outer segments of red/green cones (labeled by L/M-opsin) were partially colocalized with SHANK3 and the outer segments of rods (labeled by Rho4D2) were partially SHANK3 positive too. Moreover, SHANK3-positive labeling was also observed in Müller cells (labeled by cellular retinaldehyde-binding protein). These wide expression patterns indicate that SHANK3 may play an important role in the visual signaling pathway.
Collapse
Affiliation(s)
- Yue Xu
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Ya’nan Wang
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Guang’an Tong
- Department of Neurology, The Affiliated Hospital of the Neurology Institute, Anhui University of Chinese Medicine, Hefei, China
| | - Lin Li
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Juan Cheng
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Lesha Zhang
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Qi Xu
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Liecheng Wang
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- *Correspondence: Liecheng Wang,
| | - Pingping Zhang
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- Pingping Zhang,
| |
Collapse
|
31
|
Cammarata-Scalisi F, Callea M, Martinelli D, Willoughby CE, Tadich AC, Araya Castillo M, Lacruz-Rengel MA, Medina M, Grimaldi P, Bertini E, Nevado J. Clinical and Genetic Aspects of Phelan-McDermid Syndrome: An Interdisciplinary Approach to Management. Genes (Basel) 2022; 13:504. [PMID: 35328058 PMCID: PMC8955098 DOI: 10.3390/genes13030504] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 03/08/2022] [Accepted: 03/10/2022] [Indexed: 02/04/2023] Open
Abstract
Phelan-McDermid syndrome (PMS) is a rare, heterogeneous, and complex neurodevelopmental disorder. It is generally caused by a heterozygous microdeletion of contiguous genes located in the distal portion of the long arm of chromosome 22, including the SHANK3 gene. Sequence variants of SHANK3, including frameshift, nonsense mutations, small indels and splice site mutations also result in PMS. Furthermore, haploinsufficiency in SHANK3 has been suggested as the main cause of PMS. SHANK3 is also associated with intellectual disability, autism spectrum disorder and schizophrenia. The phenotype of PMS is variable, and lacks a distinctive phenotypic characteristic, so the clinical diagnosis should be confirmed by genetic analysis. PMS is a multi-system disorder, and clinical care must encompass various specialties and therapists. The role of risperidone, intranasal insulin, insulin growth factor 1, and oxytocin as potential therapeutic options in PMS will be discussed in this review. The diagnosis of PMS is important to provide an appropriate clinical evaluation, treatment, and genetic counseling.
Collapse
Affiliation(s)
- Francisco Cammarata-Scalisi
- Pediatric Service, Regional of Antofagasta Hospital, Antofagasta 1240835, Chile; (F.C.-S.); (A.C.T.); (M.M.)
| | - Michele Callea
- Pediatric Dentistry and Special Dental Care Unit, Meyer Children’s University Hospital, 50139 Florence, Italy
| | - Diego Martinelli
- Unit of Metabolism, Bambino Gesù Children’s Research Hospital IRCCS, 00165 Rome, Italy
| | - Colin Eric Willoughby
- Genomic Medicine, Biomedical Sciences Research Institute, Ulster University, Coleraine Campus, Coleraine BT52 1SA, Northern Ireland, UK;
| | - Antonio Cárdenas Tadich
- Pediatric Service, Regional of Antofagasta Hospital, Antofagasta 1240835, Chile; (F.C.-S.); (A.C.T.); (M.M.)
| | | | | | - Marco Medina
- Pediatric Service, Regional of Antofagasta Hospital, Antofagasta 1240835, Chile; (F.C.-S.); (A.C.T.); (M.M.)
| | - Piercesare Grimaldi
- Department of Public Health and Pediatric Sciences, University of Torino, 10126 Torino, Italy;
| | - Enrico Bertini
- Unit of Neuromuscular and Neurodegenerative Disorders, Laboratory of Molecular Medicine, Department of Neurosciences, Bambino Gesu’ Children’s Research Hospital IRCCS, 00165 Rome, Italy;
| | - Julián Nevado
- Instituto de Genética Médica y Molecular (INGEMM), Instituto de Investigación del Hospital Universitario La Paz (IdIPaz), 28046 Madrid, Spain; or
- Centro de Investigación Biomédica en RED de Enfermedades Raras (CIBERER), 28046 Madrid, Spain
- ITHACA, European Reference Network on Rare Congenital Malformations and Rare Intellectual Disability, Hospital La Paz, 28046 Madrid, Spain
| |
Collapse
|
32
|
Schmidt JK, Jones KM, Van Vleck T, Emborg ME. Modeling genetic diseases in nonhuman primates through embryonic and germline modification: Considerations and challenges. Sci Transl Med 2022; 14:eabf4879. [PMID: 35235338 PMCID: PMC9373237 DOI: 10.1126/scitranslmed.abf4879] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Genetic modification of the embryo or germ line of nonhuman primates is envisioned as a method to develop improved models of human disease, yet the promise of such animal models remains unfulfilled. Here, we discuss current methods and their limitations for producing nonhuman primate genetic models that faithfully genocopy and phenocopy human disease. We reflect on how to ethically maximize the translational relevance of such models in the search for new therapeutic strategies to treat human disease.
Collapse
Affiliation(s)
- Jenna K. Schmidt
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Kathryn M. Jones
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Trevor Van Vleck
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Marina E. Emborg
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
33
|
Li T, Xie R, Zhao J, Xu H, Cui Y, Sun C, Wang C, Liu Y. Effectiveness of Recombinant Human Growth Hormone Therapy for Children With Phelan-McDermid Syndrome: An Open-Label, Cross-Over, Preliminary Study. Front Psychiatry 2022; 13:763565. [PMID: 35250656 PMCID: PMC8888442 DOI: 10.3389/fpsyt.2022.763565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 01/17/2022] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Phelan-McDermid syndrome (PMS), also known as the 22q13. 3 deletion syndrome, is a rare neurodevelopmental syndrome with approximately 2,800 patients reported worldwide. Previous pilot study demonstrated that IGF-1 could significantly improve in both social impairment and restrictive behaviors of the patients. However, most of the patients in the developing countries like China cannot afford the high cost of using IGF-1. Our research team speculated that rhGH might serve as a low-cost and more accessible treatment for PMS. Therefore, the purpose of this open-label, cross-over, pilot study was to further investigate the safety and efficiency of rhGH in patients with PMS. METHODS A total of six children with PMS were enrolled in in this open-label, cross-over, pilot study. The children were randomly divided into two different groups. Group A received placebo followed by rhGH, while group B was treated with rhGH first. Neuropsychological and behavior assessments of the patients were performed before the stage I of study and 3 months after the intervention of stage I. After a 4-week period of washout, these assessments were conducted again before the stage II of study and 3 months after the intervention of stage II. Serum insulin-like growth factor-1 (IGF-1) and insulin-like growth factor binding-protein (IGFBP)-3 were also evaluated monthly during the intervention phases of the pilot study. RESULTS Compared with the placebo, rhGH treatment significantly decreased subscale scores of GDS (P < 0.0085) and trended to improve the total scores of GDS (P < 0.05), while the total scores and subscale scores of SC-ABC significantly decreased (P < 0.0085) following 3-months rhGH treatment. The similar results were also observed in comparison with baseline. Compared with the baseline, the level of serum IGF-1 and IGFBP-3 increased significantly (P < 0.05) following 3-months rhGH treatment, while the placebo group had no significant impact on serum IGF-1 and IGFBP-3 (P > 0.05). One child developed skin allergy the day after the first rhGH treatment, which were resolved later. CONCLUSIONS In summary, this pilot study involving six PMS children patients reveals that rhGH has a positive treatment effect on PMS. These results encourage the undertaking of a large, randomized placebo-controlled trial to conclusively prove rhGH efficacy and tolerability in PMS, thereby promoting it as a low-cost, more accessible treatment for PMS, as compared to IGF-1.
Collapse
Affiliation(s)
- TianXiao Li
- Affiliated Hospital of JiangNan University, Wuxi, China
| | - Ruijin Xie
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Jinling Zhao
- Affiliated Hospital of JiangNan University, Wuxi, China
| | - Hua Xu
- Affiliated Hospital of JiangNan University, Wuxi, China
| | - Ying Cui
- Affiliated Hospital of JiangNan University, Wuxi, China
| | - Chenyu Sun
- AMITA Health Saint Joseph Hospital Chicago, Chicago, IL, United States
| | - Chunhong Wang
- Affiliated Hospital of JiangNan University, Wuxi, China
| | - Yueying Liu
- Affiliated Hospital of JiangNan University, Wuxi, China
| |
Collapse
|
34
|
Landlust AM, Visser L, Flapper BCT, Ruiter SAJ, Zwanenburg RJ, van Ravenswaaij-Arts CMA, van Balkom IDC. Understanding Behavior in Phelan-McDermid Syndrome. Front Psychiatry 2022; 13:836807. [PMID: 35693963 PMCID: PMC9178081 DOI: 10.3389/fpsyt.2022.836807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 04/27/2022] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Phelan-McDermid syndrome (PMS) or 22q13.3 deletion syndrome is a rare genetic disorder characterized by developmental delay, hypotonia and severely delayed speech. Behavioral difficulties are often reported in PMS, although knowledge of behavioral profiles and the interpretation of reported behavior remains limited. Understanding the meaning of behavior requires considering the context as well as other domains of functioning, for example the individual's level of cognitive, social and emotional development. Combining structured direct in-person neurodevelopmental assessments with contextual assessments to enable meaningful interpretations of reported behavior on functional dimensions across multiple units of analysis, as proposed by the RDoc framework, is essential. METHODS In this article we present a structured multidisciplinary method of assessment through direct in-person neurodevelopmental assessments and assessment of contextual factors. Our study sample includes data of 33 children with an average age of 6.2 years (range 1.1 to 15.7) with PMS, obtained through individual in-person assessments in combination with parent informed questionnaires. We assessed developmental age using the Bayley-III, adaptive behavior was assessed with the Vineland screener, social-emotional development with the ESSEON-R and behavior by using the CBCL. RESULTS Our results show a great deal of variability in phenotypic presentation with regard to behavior, symptom expression and symptom severity in individuals with PMS. The data on behavior is interpreted in the context of the individual's level of cognitive, adaptive development and the (genetic) context. Behavioral data showed high levels of withdrawn behavior and attention problems. More than half of the children showed borderline or clinical symptoms related to Autism Spectrum Disorder (ASD). CONCLUSIONS The interpretation of the meaning of certain behavior in PMS is often based on questionnaires and descriptions without taking the specific context of development into account. Combining questionnaires with direct in-person assessments measuring different domains of functioning should be considered a more accurate method to interpret the meaning of findings in order to understand behavior in rare genetic disorders associated with developmental delay such as PMS. Direct in-person assessment provides valuable and specific information relevant to understanding individual behavior and inform treatment as well as increase knowledge of the neurodevelopmental phenotype in individuals with PMS. More specific application of the proposed frameworks on behavior in PMS is desirable in making useful interpretations.
Collapse
Affiliation(s)
- Annemiek M Landlust
- Autism Team Northern-Netherlands, Jonx, Department of (Youth) Mental Health and Autism, Lentis Psychiatric Institute, Groningen, Netherlands.,Department of Genetics, University Medical Centre Groningen, Groningen, Netherlands
| | - Linda Visser
- Leibniz Institute for Research and Information in Education (DIPF), Frankfurt am Main, Germany.,Center for Research on Individual Development and Adaptive Education of Children at Risk, Frankfurt am Main, Germany
| | - Boudien C T Flapper
- Autism Team Northern-Netherlands, Jonx, Department of (Youth) Mental Health and Autism, Lentis Psychiatric Institute, Groningen, Netherlands.,Department of Genetics, University Medical Centre Groningen, Groningen, Netherlands.,Department of Paediatrics, University Medical Centre Groningen, Groningen, Netherlands
| | | | - Renée J Zwanenburg
- Department of Genetics, University Medical Centre Groningen, Groningen, Netherlands
| | - Conny M A van Ravenswaaij-Arts
- Autism Team Northern-Netherlands, Jonx, Department of (Youth) Mental Health and Autism, Lentis Psychiatric Institute, Groningen, Netherlands.,Department of Genetics, University Medical Centre Groningen, Groningen, Netherlands
| | - Ingrid D C van Balkom
- Autism Team Northern-Netherlands, Jonx, Department of (Youth) Mental Health and Autism, Lentis Psychiatric Institute, Groningen, Netherlands.,Department of Psychiatry, Rob Giel Research Centre, University Medical Center Groningen, Groningen, Netherlands
| |
Collapse
|
35
|
Meng Y, Xu D, Zhang W, Meng W, Lan X, Wang X, Li M, Zhang X, Zhao Y, Yang H, Zhang R, Zhen Z. Effect of Early Swimming on the Behavior and Striatal Transcriptome of the Shank3 Knockout Rat Model of Autism. Neuropsychiatr Dis Treat 2022; 18:681-694. [PMID: 35387206 PMCID: PMC8979754 DOI: 10.2147/ndt.s357338] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 03/17/2022] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Autism spectrum disorder (ASD) is a developmental disorder characterized by social behavior deficits and stereotyped behaviors in childhood that lacks satisfactory medical intervention. Early swimming intervention is a noninvasive method combining enriched environment and exercise, which has been proven to improve brain development in young children and to treat neurodevelopmental diseases. METHODS In this study, we tested the autism-like behavior of rats with deletions in exons 11-21 of the Shank3 gene and evaluated the effect of early swimming intervention (from postnatal day 8 to 60) on the behavior of this animal model of autism. In addition, the transcriptomes of the striatal tissues of wild-type, Shank3 knockout and Shank3 knockout swimming groups rats were analyzed. RESULTS Shank3 knockout rats exhibit core symptoms of autism, and early swimming improved the social and stereotyped behaviors in this autism rat model. Transcriptomics results revealed that compared to the wild-type group, 291 differentially expressed genes (DEGs) were identified in the striatum of the Shank3 knockout group. Compared to Shank3 knockout group, 534 DEGs were identified in the striatum of Shank3 knockout swimming group. The DEGs annotated by Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway shows that the impacts of Shank3 deletion were primarily reflected in synaptic structure, development, morphology, receptor function and signaling, and swimming primarily changed the terms related to the synapses in the striatum of Shank3 knockout rats, including the morphology, structure, composition, development and regulation of synapses. CONCLUSION Early swimming intervention can ameliorate behavioral abnormalities caused by Shank3 knockout, by a mechanism that may involve the process of striatal synaptic development and should be further investigated.
Collapse
Affiliation(s)
- Yunchen Meng
- College of P.E and Sports, Beijing Normal University, Beijing, People's Republic of China
| | - Dan Xu
- College of P.E and Sports, Beijing Normal University, Beijing, People's Republic of China
| | - Weinan Zhang
- College of P.E and Sports, Beijing Normal University, Beijing, People's Republic of China
| | - Wenshu Meng
- College of Life Sciences, Beijing Normal University, Beijing, People's Republic of China
| | - Xingyu Lan
- Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing, People's Republic of China
| | - Xiaoxi Wang
- Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing, People's Republic of China
| | - Mingjuan Li
- Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing, People's Republic of China
| | - Xiaoyan Zhang
- College of P.E and Sports, Beijing Normal University, Beijing, People's Republic of China
| | - Yu Zhao
- College of P.E and Sports, Beijing Normal University, Beijing, People's Republic of China
| | - Haodong Yang
- College of P.E and Sports, Beijing Normal University, Beijing, People's Republic of China
| | - Rong Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing, People's Republic of China.,Neuroscience Research Institute, Peking University, Beijing, People's Republic of China.,Key Laboratory for Neuroscience, Ministry of Education/National Health and Family Planning Commission, Peking University, Beijing, People's Republic of China.,Autism Research Center of Peking University Health Science Center, Beijing, People's Republic of China
| | - Zhiping Zhen
- College of P.E and Sports, Beijing Normal University, Beijing, People's Republic of China
| |
Collapse
|
36
|
Lutz AK, Pérez Arévalo A, Ioannidis V, Stirmlinger N, Demestre M, Delorme R, Bourgeron T, Boeckers TM. SHANK2 Mutations Result in Dysregulation of the ERK1/2 Pathway in Human Induced Pluripotent Stem Cells-Derived Neurons and Shank2(-/-) Mice. Front Mol Neurosci 2021; 14:773571. [PMID: 34899182 PMCID: PMC8662699 DOI: 10.3389/fnmol.2021.773571] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 10/07/2021] [Indexed: 12/28/2022] Open
Abstract
SHANK2 (ProSAP1) is a postsynaptic scaffolding protein of excitatory synapses in the central nervous system and implicated in the development of autism spectrum disorders (ASD). Patients with mutations in SHANK2 show autism-like behaviors, developmental delay, and intellectual disability. We generated human induced pluripotent stem cells (hiPSC) from a patient carrying a heterozygous deletion of SHANK2 and from the unaffected parents. In patient hiPSCs and derived neurons SHANK2 mRNA and protein expression was reduced. During neuronal maturation, a reduction in growth cone size and a transient increase in neuronal soma size were observed. Neuronal proliferation was increased, and apoptosis was decreased in young and mature neurons. Additionally, mature patient hiPSC-derived neurons showed dysregulated excitatory signaling and a decrease of a broad range of signaling molecules of the ERK-MAP kinase pathway. These findings could be confirmed in brain samples from Shank2(−/−) mice, which also showed decreased mGluR5 and phospho-ERK1/2 expression. Our study broadens the current knowledge of SHANK2-related ASD. We highlight the importance of excitatory-inhibitory balance and mGluR5 dysregulation with disturbed downstream ERK1/2 signaling in ASD, which provides possible future therapeutic strategies for SHANK2-related ASD.
Collapse
Affiliation(s)
- Anne-Kathrin Lutz
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany
| | | | | | | | - Maria Demestre
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany
| | | | - Thomas Bourgeron
- Génétique Humaine et Fonctions Cognitives, Institut Pasteur, Université Paris Diderot, Paris, France
| | - Tobias M Boeckers
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany.,Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Ulm Site, Ulm, Germany
| |
Collapse
|
37
|
Genetic determinants of autism spectrum disorders - a review. CURRENT PROBLEMS OF PSYCHIATRY 2021. [DOI: 10.2478/cpp-2021-0011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Abstract
Introduction: It is estimated that various types of abnormalities from the autistic spectrum disorder occur in up to 2% of the population. These include difficulties in maintaining relationships, communication, and repetitive behaviours. Literature describes them quite well, in contrast to the causes of these disorders, which include both environmental factors and a very long list of genetic aberrations.
Materials and methods: The papers available on the PubMed platform and other sources were reviewed to describe the most important genetic factors responsible for the development of autism spectrum disorders.
Results: There are many genes and their mutations associated with the prevalence of autism spectrum disorders in patients. One of the main factors is the SHANK gene family, with the type and degree of abnormality in patients depending on the damage to particular genes: SHANK1-SHANK3. Research also shows the potential of targeted symptom-relieving therapies in patients with SHANK3 mutations. A correlation with the occurrence of autism has also been demonstrated for genes responsible for calcium signaling - especially the group of IP3R calcium channels. Their calcium transmission is abnormal in the majority of patients with autism spectrum disorders. A number of mutations in the 7q region were discovered - including the AUTS2, GNAI1, RELN, KMT2E, BRAF genes - the occurrence of which is associated with the presence of symptoms of autism. Autism spectrum disorders occur in about 10% of patients suffering from monogenic syndromes such as fragile X chromosome syndrome, Timothy syndrome, tuberous sclerosis, Rett syndrome or hamartomatic tumor syndrome.
Conclusions: Research shows that many mutations can contribute to the development of autism spectrum disorders. Further studies are necessary to discover their therapeutic and diagnostic potential for autism.
Collapse
|
38
|
Benchoua A, Lasbareilles M, Tournois J. Contribution of Human Pluripotent Stem Cell-Based Models to Drug Discovery for Neurological Disorders. Cells 2021; 10:cells10123290. [PMID: 34943799 PMCID: PMC8699352 DOI: 10.3390/cells10123290] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/19/2021] [Accepted: 11/23/2021] [Indexed: 02/07/2023] Open
Abstract
One of the major obstacles to the identification of therapeutic interventions for central nervous system disorders has been the difficulty in studying the step-by-step progression of diseases in neuronal networks that are amenable to drug screening. Recent advances in the field of human pluripotent stem cell (PSC) biology offers the capability to create patient-specific human neurons with defined clinical profiles using reprogramming technology, which provides unprecedented opportunities for both the investigation of pathogenic mechanisms of brain disorders and the discovery of novel therapeutic strategies via drug screening. Many examples not only of the creation of human pluripotent stem cells as models of monogenic neurological disorders, but also of more challenging cases of complex multifactorial disorders now exist. Here, we review the state-of-the art brain cell types obtainable from PSCs and amenable to compound-screening formats. We then provide examples illustrating how these models contribute to the definition of new molecular or functional targets for drug discovery and to the design of novel pharmacological approaches for rare genetic disorders, as well as frequent neurodegenerative diseases and psychiatric disorders.
Collapse
Affiliation(s)
- Alexandra Benchoua
- Neuroplasticity and Therapeutics, CECS, I-STEM, AFM, 91100 Corbeil-Essonnes, France;
- High Throughput Screening Platform, CECS, I-STEM, AFM, 91100 Corbeil-Essonnes, France;
- Correspondence:
| | - Marie Lasbareilles
- Neuroplasticity and Therapeutics, CECS, I-STEM, AFM, 91100 Corbeil-Essonnes, France;
- UEVE UMR 861, I-STEM, AFM, 91100 Corbeil-Essonnes, France
| | - Johana Tournois
- High Throughput Screening Platform, CECS, I-STEM, AFM, 91100 Corbeil-Essonnes, France;
| |
Collapse
|
39
|
Frank Y. The Neurological Manifestations of Phelan-McDermid Syndrome. Pediatr Neurol 2021; 122:59-64. [PMID: 34325981 DOI: 10.1016/j.pediatrneurol.2021.06.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 06/04/2021] [Accepted: 06/09/2021] [Indexed: 11/17/2022]
Abstract
Phelan-McDermid syndrome (PMS) is a genetic disorder, caused by haploinsufficiency of the SHANK3 gene on chromosome 22q13.3. PMS is characterized by neurobehavioral symptoms and signs including intellectual disability, speech and language impairment, autism spectrum disorder (ASD), hypotonia, and other motor abnormalities. In the brain, SHANK3 is expressed in neurons, especially in the synapse, and encodes a master scaffolding protein that forms a key framework in the postsynaptic density of glutamatergic synapses. Mutations in SHANK3 have also been identified in individuals with ASD, intellectual deficiency (ID), and schizophrenia. Shank3 deficient mice have defects in basal glutamatergic synaptic transmission in the hippocampus, and in synaptic transmission plasticity, including deficits in long-term potentiation, and show behavioral deficits compatible with the clinical manifestations of PMS. The PMS phenotype varies between affected individuals, but ID and speech and language impairment are present in all cases. ASD is present in a great majority of these individuals. Neurological examination demonstrates hypotonia and abnormalities of motor coordination, visual motor coordination, and gait in the majority of affected individuals. Sleep disturbances and increased pain tolerance are frequent parental complaints. Seizures and epilepsy are common, affecting more than 40% of individuals. Brain magnetic resonance imaging abnormalities include corpus callosum hypoplasia, delayed myelination and white matter abnormalities, dilated ventricles, and arachnoid cysts. Recent advanced imaging anatomic studies including diffusion tensor imaging, point to abnormal brain connectivity. The natural history of the syndrome is not yet fully known, but some individuals with PMS have a later onset of psychiatric illnesses including bipolar disease, accompanied by functional and neurological regression. Individuals with the syndrome are treated symptomatically. Advances in understanding the pathophysiology of this syndrome and the generation of animal models have raised opportunities for a biological cure for PMS. A pilot clinical trial with insulin-like growth factor-1 (IGF-1) showed positive effects on some behavioral core symptoms.
Collapse
Affiliation(s)
- Yitzchak Frank
- Pediatric Neurologist, Seaver Autism Center for Research & Treatment, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York.
| |
Collapse
|
40
|
SHANK3 Genotype Mediates Speech and Language Phenotypes in a Nonclinical Population. AUTISM RESEARCH AND TREATMENT 2021; 2021:6634584. [PMID: 34188957 PMCID: PMC8195663 DOI: 10.1155/2021/6634584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 04/13/2021] [Accepted: 05/27/2021] [Indexed: 11/17/2022]
Abstract
Mutations affecting the synaptic-scaffold gene SHANK3 represent the most common genetic causes of autism with intellectual disability, accounting for about 1-2% of cases. Rare variants of this gene have also been associated with schizophrenia, and its deletion results in the autistic condition known as Phelan–McDermid syndrome. Despite the importance of SHANK3 as a paradigmatic gene mediating neurodevelopmental disorders, its psychological effects in nonclinical populations have yet to be studied. We genotyped the nonsynonymous, functional SHANK3 SNP rs9616915 in a large population of typical individuals scored for autism spectrum traits (the Autism Quotient, AQ) and schizotypy spectrum traits (the Schizotypal Personality Questionnaire, SPQ-BR). Males, but not females, showed significant genotypic effects for the SPQ-BR subscale associated with speech and language: Odd Speech. These findings, in conjunction with animal model studies showing vocalization and auditory effects of SHANK3 mutations, and studies indicating severe language alterations and speech-associated white matter tract abnormalities in Phelan–McDermid syndrome, suggest that SHANK3 differentially affects the development and expression of human language and speech. Imaging genetic and speech-language studies of typical individuals carrying different genotypes of rs9616915 should provide novel insights into the neurological and psychological bases of speech and language alterations among individuals with SHANK3 mutations and Phelan–McDermid syndrome.
Collapse
|
41
|
Schroeder KA, Witts BN, Traub MR. Opportunities for ABA intervention in Phelan-McDermid syndrome. INTERNATIONAL JOURNAL OF DEVELOPMENTAL DISABILITIES 2021; 68:984-989. [PMID: 36816984 PMCID: PMC9936992 DOI: 10.1080/20473869.2021.1895698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 02/18/2021] [Accepted: 02/18/2021] [Indexed: 06/18/2023]
Abstract
Phelan-McDermid syndrome (PMS), also called 22q13.3 deletion syndrome, is a rare genetic disorder affecting at least 2,000 people worldwide (Phelan-McDermid Syndrome Foundation, 2019, How rare is Phelan-McDermid?). PMS has many distinguishing characteristics and many medical specialties have been recommended to treat the clinical features. While many therapies, including behavioral therapy, have been speculated to be beneficial in treating PMS, there is little known regarding their effectiveness [Costales, J. L. and Kolevzon, A. 2015. Phelan-McDermid syndrome and SHANK3: Implications for treatment. Neurotherapeutics: The Journal of the American Society for Experimental Neurotherapeutics, 12, 620-630.]. Behavior analysis has the capability to help in many areas of treatment for PMS either directly through, for example, behavior treatment to address aggressive behavior, or through collaborating with other specialties treating PMS by combining, for example, behavioral principles in the alleviation of medical issues such as constipation. Currently, there is a role for the behavior analyst to expand our field and identify effective treatments for those with PMS while we wait for a cure. In this paper, we discuss how medical considerations may affect behavior interventions and make recommendations for the behavior analyst working with PMS.
Collapse
Affiliation(s)
- Kate A. Schroeder
- Community Psychology, Counseling, and Family, St. Cloud State University, St. Cloud, Minnesota, USA
| | - Benjamin N. Witts
- Community Psychology, Counseling, and Family, St. Cloud State University, St. Cloud, Minnesota, USA
| | - Michele R. Traub
- Community Psychology, Counseling, and Family, St. Cloud State University, St. Cloud, Minnesota, USA
| |
Collapse
|
42
|
Xie RJ, Li TX, Sun C, Cheng C, Zhao J, Xu H, Liu Y. A case report of Phelan-McDermid syndrome: preliminary results of the treatment with growth hormone therapy. Ital J Pediatr 2021; 47:49. [PMID: 33663540 PMCID: PMC7934562 DOI: 10.1186/s13052-021-01003-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 02/22/2021] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Phelan-McDermid syndrome (PMS), also known as 22q13.3 deletion syndrome, is a rare neurodevelopmental syndrome resulting from a deletion of the distal long arm of chromosome 22. CASE PRESENTATION We report a case of a 21 months old Chinese girl presenting with global developmental delay, regression of language skills, unable to understand a few words or walk independently, insomnia, and autism-like behaviors. Copy number variation (CNV) analysis showed a heterozygous loss of SHANK3 gene in the 22q13 region, consistent with a diagnosis of PMS. After treatment with recombinant human growth hormone (rhGH), this patient had an improvement in motor skills and social behaviors. No side effects from rhGH therapy were reported. CONCLUSIONS This is the first report of using rhGH to treat a Chinese girl diagnosed with PMS. We speculate rhGH could be a reasonable alternative choice for PMS treatment with similar clinical outcomes in comparison to insulin-like growth factor-1(IGF-1). However, further clinical trials are needed to confirm this hypothesis.
Collapse
Affiliation(s)
- Rui Jin Xie
- Affiliated Hospital of Jiangnan University, No. 1000, Hefeng Avenue, Wuxi, 214122, People's Republic of China
| | - Tian Xiao Li
- Affiliated Hospital of Jiangnan University, No. 1000, Hefeng Avenue, Wuxi, 214122, People's Republic of China
| | - Chenyu Sun
- AMITA Health Saint Joseph Hospital Chicago, 2900 N. Lake Shore Drive, Chicago, IL, 60657, USA
| | - Ce Cheng
- The University of Arizona College of Medicine at South Campus, 2800 E. Ajo Way, Tucson, AZ, 85718, USA
| | - Jinlin Zhao
- Affiliated Hospital of Jiangnan University, No. 1000, Hefeng Avenue, Wuxi, 214122, People's Republic of China
| | - Hua Xu
- Affiliated Hospital of Jiangnan University, No. 1000, Hefeng Avenue, Wuxi, 214122, People's Republic of China
| | - Yueying Liu
- Affiliated Hospital of Jiangnan University, No. 1000, Hefeng Avenue, Wuxi, 214122, People's Republic of China.
| |
Collapse
|
43
|
Neklyudova AK, Portnova GV, Rebreikina AB, Voinova VY, Vorsanova SG, Iourov IY, Sysoeva OV. 40-Hz Auditory Steady-State Response (ASSR) as a Biomarker of Genetic Defects in the SHANK3 Gene: A Case Report of 15-Year-Old Girl with a Rare Partial SHANK3 Duplication. Int J Mol Sci 2021; 22:ijms22041898. [PMID: 33673024 PMCID: PMC7917917 DOI: 10.3390/ijms22041898] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 01/26/2021] [Accepted: 02/09/2021] [Indexed: 12/02/2022] Open
Abstract
SHANK3 encodes a scaffold protein involved in postsynaptic receptor density in glutamatergic synapses, including those in the parvalbumin (PV)+ inhibitory neurons—the key players in the generation of sensory gamma oscillations, such as 40-Hz auditory steady-state response (ASSR). However, 40-Hz ASSR was not studied in relation to SHANK3 functioning. Here, we present a 15-year-old girl (SH01) with previously unreported duplication of the first seven exons of the SHANK3 gene (22q13.33). SH01’s electroencephalogram (EEG) during 40-Hz click trains of 500 ms duration binaurally presented with inter-trial intervals of 500–800 ms were compared with those from typically developing children (n = 32). SH01 was diagnosed with mild mental retardation and learning disabilities (F70.88), dysgraphia, dyslexia, and smaller vocabulary than typically developing (TD) peers. Her clinical phenotype resembled the phenotype of previously described patients with 22q13.33 microduplications (≈30 reported so far). SH01 had mild autistic symptoms but below the threshold for ASD diagnosis and microcephaly. No seizures or MRI abnormalities were reported. While SH01 had relatively preserved auditory event-related potential (ERP) with slightly attenuated P1, her 40-Hz ASSR was totally absent significantly deviating from TD’s ASSR. The absence of 40-Hz ASSR in patients with microduplication, which affected the SHANK3 gene, indicates deficient temporal resolution of the auditory system, which might underlie language problems and represent a neurophysiological biomarker of SHANK3 abnormalities.
Collapse
Affiliation(s)
- Anastasia K. Neklyudova
- Laboratory of Human Higher Nervous Activity, Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Science, 117485 Moscow, Russia; (A.K.N.); (G.V.P.); (A.B.R.)
| | - Galina V. Portnova
- Laboratory of Human Higher Nervous Activity, Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Science, 117485 Moscow, Russia; (A.K.N.); (G.V.P.); (A.B.R.)
| | - Anna B. Rebreikina
- Laboratory of Human Higher Nervous Activity, Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Science, 117485 Moscow, Russia; (A.K.N.); (G.V.P.); (A.B.R.)
| | - Victoria Yu Voinova
- Veltischev Research and Clinical Institute for Pediatrics of the Pirogov, Russian National Research Medical University, Ministry of Health of Russian Federation, 125412 Moscow, Russia; (V.Y.V.); (S.G.V.); (I.Y.I.)
- Mental Health Research Center, 117152 Moscow, Russia
| | - Svetlana G. Vorsanova
- Veltischev Research and Clinical Institute for Pediatrics of the Pirogov, Russian National Research Medical University, Ministry of Health of Russian Federation, 125412 Moscow, Russia; (V.Y.V.); (S.G.V.); (I.Y.I.)
- Mental Health Research Center, 117152 Moscow, Russia
| | - Ivan Y. Iourov
- Veltischev Research and Clinical Institute for Pediatrics of the Pirogov, Russian National Research Medical University, Ministry of Health of Russian Federation, 125412 Moscow, Russia; (V.Y.V.); (S.G.V.); (I.Y.I.)
- Mental Health Research Center, 117152 Moscow, Russia
| | - Olga V. Sysoeva
- Laboratory of Human Higher Nervous Activity, Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Science, 117485 Moscow, Russia; (A.K.N.); (G.V.P.); (A.B.R.)
- Correspondence:
| |
Collapse
|
44
|
Gandhi T, Lee CC. Neural Mechanisms Underlying Repetitive Behaviors in Rodent Models of Autism Spectrum Disorders. Front Cell Neurosci 2021; 14:592710. [PMID: 33519379 PMCID: PMC7840495 DOI: 10.3389/fncel.2020.592710] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [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
|
45
|
Sleep Duration in Mouse Models of Neurodevelopmental Disorders. Brain Sci 2020; 11:brainsci11010031. [PMID: 33396736 PMCID: PMC7824512 DOI: 10.3390/brainsci11010031] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 12/15/2020] [Accepted: 12/22/2020] [Indexed: 01/06/2023] Open
Abstract
Sleep abnormalities are common in patients with neurodevelopmental disorders, and it is thought that deficits in sleep may contribute to the unfolding of symptoms in these disorders. Appreciating sleep abnormalities in neurodevelopmental disorders could be important for designing a treatment for these disorders. We studied sleep duration in three mouse models by means of home-cage monitoring: Tsc2+/- (tuberous sclerosis complex), oxytocin receptor (Oxtr) knockout (KO) (autism spectrum disorders), and Shank3 e4-9 KO (Phelan-McDermid syndrome). We studied both male and female mice, and data were analyzed to examine effects of both genotype and sex. In general, we found that female mice slept less than males regardless of genotype or phase. We did not find any differences in sleep duration in either Tsc2+/- or Oxtr KO mice, compared to controls. In Shank3 e4-9 KO mice, we found a statistically significant genotype x phase interaction (p = 0.002) with a trend that Shank3e4-9 KO mice regardless of sex slept more than control mice in the active phase. Our results have implications for the management of patients with Phelan-McDermid syndrome.
Collapse
|
46
|
Gómez Taylor B, Moreno Sancho ML, Drehmer Rieger E, Carrera Juliá S, Nevado J, Sempere Ferre F. [Prevalence of the Phelan-McDermid Syndrome in Spain.]. Rev Esp Salud Publica 2020; 94:e202012121. [PMID: 33342995 PMCID: PMC11582991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 09/09/2020] [Indexed: 06/12/2023] Open
Abstract
OBJECTIVE Phelan-McDermid syndrome is a rare genetic condition caused by a deletion of the terminal end of chromosome 22 in the 13.3 region, as well as, by point mutations within SHANK3 gene. The aims of this research were to determine the prevalence of the disease in the Spanish population, to establish the geographical distribution of the syndrome among the different autonomous communities, to elucidate the age range that affects more patients, to study the disease-sex relationship, as well as the age at diagnosis. METHODS For the research, patients diagnosed with the disease for twelve years were recruited throughout the Spanish territory. The clinical patient information was obtained from the referral doctors using two standardized questionnaires completed with data from the medical reports and the interview with the parents. The molecular diagnosis of the disease was carried out using different formats of microarrays. Data were processed using Microsoft Excel and Statgraphics Centurion XVII. RESULTS Currently in Spain there are 201 people diagnosed with the disease. Currently in Spain there are 201 people diagnosed with the disease, its prevalence being 4x10-4/10,000 inhabitants. The community with the most diagnosed patients was Madrid and there were no significant differences in terms of sex and disease, the mean age at diagnosis was around 6.67 years. CONCLUSIONS The prevalence of the disease in Spain is very low, and it can be stated that it is very likely that there are more people with this syndrome in the population.
Collapse
Affiliation(s)
- Bárbara Gómez Taylor
- Facultad de Medicina y Ciencias de la Salud. Universidad Católica de Valencia San Vicente Mártir. Valencia. España
| | - Mari Luz Moreno Sancho
- Facultad de Medicina y Ciencias de la Salud. Universidad Católica de Valencia San Vicente Mártir. Valencia. España
| | - Eraci Drehmer Rieger
- Facultad de Medicina y Ciencias de la Salud. Universidad Católica de Valencia San Vicente Mártir. Valencia. España
| | - Sandra Carrera Juliá
- Facultad de Medicina y Ciencias de la Salud. Universidad Católica de Valencia San Vicente Mártir. Valencia. España
| | - Julián Nevado
- Instituto de Genética Médica y Molecular (INGEMM)-IdiPAZ. Hospital Universitario la Paz. Madrid. España
- CIBERER (Centro de Investigación Básica en Red de Enfermedades Raras). Instituto de Salud Carlos III. Madrid. España
| | - Francisca Sempere Ferre
- Facultad de Medicina y Ciencias de la Salud. Universidad Católica de Valencia San Vicente Mártir. Valencia. España
- Departamento de Estadística e Investigación Operativa Aplicadas y Calidad. Universidad Politécnica de Valencia. Valencia. España
| |
Collapse
|
47
|
Soleimani Zakeri NS, Pashazadeh S, MotieGhader H. Gene biomarker discovery at different stages of Alzheimer using gene co-expression network approach. Sci Rep 2020; 10:12210. [PMID: 32699331 PMCID: PMC7376049 DOI: 10.1038/s41598-020-69249-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 07/08/2020] [Indexed: 12/24/2022] Open
Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disorder. It is the most common type of dementia that has remained as an incurable disease in the world, which destroys the brain cells irreversibly. In this study, a systems biology approach was adopted to discover novel micro-RNA and gene-based biomarkers of the diagnosis of Alzheimer's disease. The gene expression data from three AD stages (Normal, Mild Cognitive Impairment, and Alzheimer) were used to reconstruct co-expression networks. After preprocessing and normalization, Weighted Gene Co-Expression Network Analysis (WGCNA) was used on a total of 329 samples, including 145 samples of Alzheimer stage, 80 samples of Mild Cognitive Impairment (MCI) stage, and 104 samples of the Normal stage. Next, three gene-miRNA bipartite networks were reconstructed by comparing the changes in module groups. Then, the functional enrichment analyses of extracted genes of three bipartite networks and miRNAs were done, respectively. Finally, a detailed analysis of the authentic studies was performed to discuss the obtained biomarkers. The outcomes addressed proposed novel genes, including MBOAT1, ARMC7, RABL2B, HNRNPUL1, LAMTOR1, PLAGL2, CREBRF, LCOR, and MRI1and novel miRNAs comprising miR-615-3p, miR-4722-5p, miR-4768-3p, miR-1827, miR-940 and miR-30b-3p which were related to AD. These biomarkers were proposed to be related to AD for the first time and should be examined in future clinical studies.
Collapse
Affiliation(s)
| | - Saeid Pashazadeh
- Faculty of Electrical and Computer Engineering, University of Tabriz, Tabriz, Iran.
| | - Habib MotieGhader
- Department of Computer Engineering, Gowgan Educational Center, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| |
Collapse
|
48
|
Early Restoration of Shank3 Expression in Shank3 Knock-Out Mice Prevents Core ASD-Like Behavioral Phenotypes. eNeuro 2020; 7:ENEURO.0332-19.2020. [PMID: 32327468 PMCID: PMC7294460 DOI: 10.1523/eneuro.0332-19.2020] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 04/02/2020] [Accepted: 04/09/2020] [Indexed: 12/14/2022] Open
Abstract
Several genes are associated with increased risk for autism spectrum disorder (ASD), neurodevelopmental disorders that present with repetitive movements and restricted interests along with deficits in social interaction/communication. While genetic alterations associated with ASD are present early in life, ASD-like behaviors are difficult to detect in early infancy. This raises the issue of whether reversal of an ASD-associated genetic alteration early in life can prevent the onset of ASD-like behaviors. Genetic alterations of SHANK3, a well-characterized gene encoding a postsynaptic scaffolding protein, are estimated to contribute to ∼0.5% of ASD and remain one of the more replicated and well-characterized genetic defects in ASD. Here, we investigate whether early genetic reversal of a Shank3 mutation can prevent the onset of ASD-like behaviors in a mouse model. Previously, we have demonstrated that mice deficient in Shank3 display a wide range of behavioral abnormalities such as repetitive grooming, social deficits, anxiety, and motor abnormalities. In this study, we replicate many of these behaviors in Shank3 mutant mice. With early genetic restoration of wild-type (WT) Shank3, we rescue behaviors including repetitive grooming and social, locomotor, and rearing deficits. Our findings support the idea that the underlying mechanisms involving ASD behaviors in mice deficient in Shank3 are susceptible to early genetic correction of Shank3 mutations.
Collapse
|
49
|
Li T, Li F, Lin J, Zhang Y, Zhang Q, Sun Y, Chen X, Xu M, Wang X, Li Q. Deletion of c16orf45 in zebrafish results in a low fertilization rate and increased thigmotaxis. Dev Psychobiol 2020; 62:1003-1010. [PMID: 32421859 DOI: 10.1002/dev.21984] [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: 12/03/2019] [Revised: 03/28/2020] [Accepted: 04/13/2020] [Indexed: 11/06/2022]
Abstract
c16orf45 is located at 16p13.11, an important locus related to neurodevelopmental diseases. Clinical studies have demonstrated that c16orf45 is associated with various neurodevelopmental diseases. To further elucidate the effect of c16orf45 on neural development, we constructed a zebrafish model with a stably inherited c16orf45 deletion via CRISPR/Cas9 technology. We found that deletion of c16orf45 significantly reduced the zebrafish fertilization rate, and both females and males showed reduced fertility. Meanwhile, the homozygous c16orf45 knockout zebrafish showed a developmental delay at 24 hr postfertilization (hpf). However, morphological changes were not apparent after 2 days postfertilization (dpf). Notably, the results of behavioral experiments revealed increased thigmotaxis in c16orf45- / - zebrafish at 2 months. In conclusion, these findings demonstrate that c16orf45 plays an important role in nervous system and reproductive system.
Collapse
Affiliation(s)
- Tingting Li
- Translational Medical Center for Developmental and Disease, Shanghai Key Laboratory of Birth Defect, Institute of Pediatrics, Children's Hospital of Fudan University, Shanghai, China
| | - Fei Li
- Translational Medical Center for Developmental and Disease, Shanghai Key Laboratory of Birth Defect, Institute of Pediatrics, Children's Hospital of Fudan University, Shanghai, China
| | - Jia Lin
- Translational Medical Center for Developmental and Disease, Shanghai Key Laboratory of Birth Defect, Institute of Pediatrics, Children's Hospital of Fudan University, Shanghai, China
| | - Yinglan Zhang
- Translational Medical Center for Developmental and Disease, Shanghai Key Laboratory of Birth Defect, Institute of Pediatrics, Children's Hospital of Fudan University, Shanghai, China
| | - Qi Zhang
- Translational Medical Center for Developmental and Disease, Shanghai Key Laboratory of Birth Defect, Institute of Pediatrics, Children's Hospital of Fudan University, Shanghai, China
| | - Yanhe Sun
- Translational Medical Center for Developmental and Disease, Shanghai Key Laboratory of Birth Defect, Institute of Pediatrics, Children's Hospital of Fudan University, Shanghai, China
| | - Xudong Chen
- Translational Medical Center for Developmental and Disease, Shanghai Key Laboratory of Birth Defect, Institute of Pediatrics, Children's Hospital of Fudan University, Shanghai, China
| | - Mingqing Xu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xu Wang
- Cancer Metabolism Laboratory, Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Qiang Li
- Translational Medical Center for Developmental and Disease, Shanghai Key Laboratory of Birth Defect, Institute of Pediatrics, Children's Hospital of Fudan University, Shanghai, China
| |
Collapse
|
50
|
Huang M, Pu S, Jiang W, Worley PF, Xu T. Deficiency of SHANK3 isoforms impairs thermal hyperalgesia and dysregulates the expression of postsynaptic proteins in the spinal cord. Neurosci Res 2020; 163:26-33. [PMID: 32147472 DOI: 10.1016/j.neures.2020.03.001] [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: 09/05/2019] [Revised: 12/27/2019] [Accepted: 03/03/2020] [Indexed: 10/24/2022]
Abstract
SHANK3 is one of the scaffolding proteins in the postsynaptic density (PSD). Pain perception and underlying mechanisms were investigated in Shank3 exon 21 deficient (Shank3△C) mice. Sixty-six mice were attributed according to their genotype to three groups: (1) wild-type (WT), (2) heterozygous Shank3△C/+, and (3) homozygous Shank3△C/△C. Complete Freund's adjuvant (CFA) was used to induce inflammatory pain, and thermal hyperalgesia was determined. CFA treatment reduced the thermal threshold in the WT group; groups expressing mutations of Shank3 (△C/+ and △C/△C) had higher thresholds after CFA administration compared to the WT group. Mice with Shank3 mutations (△C/+ or △C/△C) had a lower expression of GluN2A and IP3R proteins and a higher expression of mGluR5 protein in the PSD compared to WT mice without changes in GluN1, GluN2B, and Homer expression. The crosslinking of Homer-IP3R, but not Homer-mGluR5, was decreased in the total lysate. Deficit of Shank3 exon 21 may lead to impaired perception of thermal pain in mice under inflammatory conditions. This impairment may result from protein dysregulation in the PSD like downregulation of the GluN2A subunit, which may reduce NMDAR-mediated currents, and/or decreased crosslinking between Homer and IP3R, which may reduce the release of Ca2+ from intracellular stores.
Collapse
Affiliation(s)
- Min Huang
- Department of Anesthesiology, Affiliated Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai 200233, China
| | - Shaofeng Pu
- Pain Management Center, Affiliated Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai 200233, China
| | - Wei Jiang
- Department of Anesthesiology, Affiliated Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai 200233, China
| | - Paul F Worley
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Tao Xu
- Department of Anesthesiology, Affiliated Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai 200233, China; Department of Anesthesiology, Tongzhou People's Hospital, Nantong 226300, China.
| |
Collapse
|