1
|
Sun YY, Xia Y, Zhi QN, Liu XY. Diagnosis and treatment of bipolar disorder in Phelan-McDermid syndrome: A case report and review of literature. World J Psychiatry 2025; 15:101948. [PMID: 39974483 PMCID: PMC11758057 DOI: 10.5498/wjp.v15.i2.101948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 11/23/2024] [Accepted: 12/25/2024] [Indexed: 01/14/2025] Open
Abstract
BACKGROUND Phelan-McDermid syndrome (PMS) is a rare genetic disorder characterized by intellectual disability, delayed language development, autism spectrum disorders, motor tone abnormalities, and a high risk of psychiatric symptoms, including bipolar disorder. CASE SUMMARY This report presented an 18-year clinical history of a 36-year-old woman with PMS, marked by intellectual disabilities, social withdrawal, and stereotyped behaviors. Diagnosed with bipolar disorder at the age of 18 years old, she encountered significant treatment challenges, including severe adverse reactions to antipsychotic medications in 2022, which led to speech and functional regression. Through rehabilitation and comprehensive therapy, her condition gradually improved. In 2024, after further treatment, her symptoms stabilized, highlighting the complexities and successes of long-term management. CONCLUSION Effective management of PMS requires a thorough clinical history, genetic testing, and long-term supportive care.
Collapse
Affiliation(s)
- Yu-Yong Sun
- Department of Psychiatry, Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou 310000, Zhejiang Province, China
| | - Yong Xia
- Department of Psychiatry, Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou 310000, Zhejiang Province, China
| | - Qian-Na Zhi
- Department of Psychiatry, Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou 310000, Zhejiang Province, China
| | - Xiao-Yan Liu
- Department of Psychiatry, Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou 310000, Zhejiang Province, China
| |
Collapse
|
2
|
Kim YE, Kim M, Kim S, Lee R, Ujihara Y, Marquez-Wilkins EM, Jiang YH, Yang E, Kim H, Lee C, Park C, Kim IH. Endothelial SHANK3 regulates tight junctions in the neonatal mouse blood-brain barrier through β-Catenin signaling. Nat Commun 2025; 16:1407. [PMID: 39915488 PMCID: PMC11802743 DOI: 10.1038/s41467-025-56720-1] [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: 03/06/2024] [Accepted: 01/29/2025] [Indexed: 02/09/2025] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disability condition arising from a combination of genetic and environmental factors. Despite the blood-brain barrier (BBB) serving as a crucial gatekeeper, conveying environmental influences into the brain parenchyma, the contributions of BBB in ASD pathogenesis remain largely uncharted. Here we report that SHANK3, an ASD-risk gene, expresses in the BBB-forming brain endothelial cells (BECs) and regulates tight junctional (TJ) integrity essential for BBB's barrier function. Endothelium-specific Shank3 (eShank3) knockout (KO) neonatal mice exhibit male-specific BBB-hyperpermeability, reduced neuronal excitability, and impaired ultra-sonic communications. Although BBB permeability is restored during adult age, the male mutant mice display reduced neuronal excitability and impaired sociability. Further analysis reveals that the BBB-hyperpermeability is attributed to the β-Catenin imbalance triggered by eShank3-KO. These findings highlight a pathogenic mechanism stemming from the ASD-risk Shank3, emphasizing the significance of neonatal BECs in the BBB as a potential therapeutic target for ASD.
Collapse
Affiliation(s)
- Yong-Eun Kim
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN, USA
- Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Minseong Kim
- Department of Molecular and Cellular Physiology, Louisiana State University Health Science Center, Shreveport, LA, USA
| | - Sunwhi Kim
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN, USA
- Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Raham Lee
- Department of Molecular and Cellular Physiology, Louisiana State University Health Science Center, Shreveport, LA, USA
| | - Yusuke Ujihara
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN, USA
| | | | - Yong-Hui Jiang
- Department of Genetics, Pediatrics and Neuroscience, Yale University School of Medicine, New Haven, CT, USA
| | - Esther Yang
- Department of Anatomy, College of Medicine, Korea University, Seoul, South Korea
| | - Hyun Kim
- Department of Anatomy, College of Medicine, Korea University, Seoul, South Korea
| | - Changhoon Lee
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Changwon Park
- Department of Molecular and Cellular Physiology, Louisiana State University Health Science Center, Shreveport, LA, USA.
| | - Il Hwan Kim
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN, USA.
- Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN, USA.
| |
Collapse
|
3
|
Asta L, Ricciardello A, Cucinotta F, Turriziani L, Boncoddo M, Bellomo F, Angelini J, Gnazzo M, Scandolo G, Pisanò G, Pelagatti F, Chehbani F, Camia M, Persico AM. Clinical, developmental and serotonemia phenotyping of a sample of 70 Italian patients with Phelan-McDermid Syndrome. J Neurodev Disord 2024; 16:57. [PMID: 39363263 PMCID: PMC11451156 DOI: 10.1186/s11689-024-09572-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 09/18/2024] [Indexed: 10/05/2024] Open
Abstract
BACKGROUND Phelan-McDermid syndrome (PMS) is caused by monoallelic loss or inactivation at the SHANK3 gene, located in human chr 22q13.33, and is often associated with Autism Spectrum Disorder (ASD). OBJECTIVES To assess the clinical and developmental phenotype in a novel sample of PMS patients, including for the first time auxometric trajectories and serotonin blood levels. METHODS 70 Italian PMS patients were clinically characterized by parental report, direct medical observation, and a thorough medical and psychodiagnostic protocol. Serotonin levels were measured in platelet-rich plasma by HPLC. RESULTS Our sample includes 59 (84.3%) cases with chr. 22q13 terminal deletion, 5 (7.1%) disruptive SHANK3 mutations, and 6 (8.6%) ring chromosome 22. Intellectual disability was present in 69 (98.6%) cases, motor coordination disorder in 65 (92.9%), ASD in 20 (28.6%), and lifetime bipolar disorder in 12 (17.1%). Prenatal and postnatal complications were frequent (22.9%-48.6%). Expressive and receptive language were absent in 49 (70.0%) and 19 (27.1%) cases, respectively. Decreased pain sensitivity was reported in 56 (80.0%), hyperactivity in 49 (80.3%), abnormal sleep in 45 (64.3%), congenital dysmorphisms in 35 (58.3%), chronic stool abnormalities and especially constipation in 29 (41.4%). Parents reported noticing behavioral abnormalities during early childhood immediately after an infective episode in 34 (48.6%) patients. Brain MRI anomalies were observed in 53 (79.1%), EEG abnormalities in 16 (23.5%), kidney and upper urinary tract malformations in 18 (28.1%). Two novel phenotypes emerged: (a) a subgroup of 12/44 (27.3%) PMS patients displays smaller head size at enrollment (mean age 11.8 yrs) compared to their first year of neonatal life, documenting a deceleration of head growth (p < 0.001); (b) serotonin blood levels are significantly lower in 21 PMS patients compared to their 21 unaffected siblings (P < 0.05), and to 432 idiopathic ASD cases (p < 0.001). CONCLUSIONS We replicate and extend the description of many phenotypic characteristics present in PMS, and report two novel features: (1) growth trajectories are variable and head growth appears to slow down during childhood in some PMS patients; (2) serotonin blood levels are decreased in PMS, and not increased as frequently occurs in ASD. Further investigations of these novel features are under way.
Collapse
Affiliation(s)
- Lisa Asta
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Arianna Ricciardello
- Cantonal Psychiatric Clinic, Cantonal Socio-Psychiatric Organization (O.S.C.), Repubblica e Cantone Ticino, Mendrisio, Switzerland
| | | | - Laura Turriziani
- Center for Autism "Dopo Di Noi", Barcellona Pozzo Di Gotto (Messina), Italy
| | - Maria Boncoddo
- Institute for Biomedical Research and Innovation (I.R.I.B.), National Research Council of Italy (C.N.R.), Messina, Italy
| | - Fabiana Bellomo
- Child Neuropsychiatry Unit, "G. Martino" University Hospital, Messina, Italy
| | - Jessica Angelini
- Residency Program in Child & Adolescent Neuropsychiatry, University of Modena and Reggio Emilia, Modena, Italy
| | - Martina Gnazzo
- Residency Program in Child & Adolescent Neuropsychiatry, University of Modena and Reggio Emilia, Modena, Italy
| | - Giulia Scandolo
- Residency Program in Child & Adolescent Neuropsychiatry, University of Modena and Reggio Emilia, Modena, Italy
| | - Giulia Pisanò
- Residency Program in Child & Adolescent Neuropsychiatry, University of Modena and Reggio Emilia, Modena, Italy
| | - Francesco Pelagatti
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Fethia Chehbani
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Michela Camia
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Antonio M Persico
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy.
- Child & Adolescent Neuropsychiatry Program, Modena University Hospital, Modena, Italy.
| |
Collapse
|
4
|
Yin R, Wack M, Hassen-Khodja C, McDuffie MT, Bliss G, Horn EJ, Kothari C, McLarney B, Davis R, Hanson K, O'Boyle M, Betancur C, Avillach P. Phenome-wide profiling identifies genotype-phenotype associations in Phelan-McDermid syndrome using family-sourced data from an international registry. Mol Autism 2024; 15:40. [PMID: 39350236 PMCID: PMC11443936 DOI: 10.1186/s13229-024-00619-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 08/29/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND Phelan-McDermid syndrome (PMS) is a rare neurodevelopmental disorder caused by 22q13 deletions that include the SHANK3 gene or pathogenic sequence variants in SHANK3. It is characterized by global developmental delay, intellectual disability, speech impairment, autism spectrum disorder, and hypotonia; other variable features include epilepsy, brain and renal malformations, and mild dysmorphic features. Here, we conducted genotype-phenotype correlation analyses using the PMS International Registry, a family-driven registry that compiles clinical data in the form of family-reported outcomes and family-sourced genetic test results. METHODS Data from the registry were harmonized and integrated into the i2b2/tranSMART clinical and genomics data warehouse. We gathered information from 401 individuals with 22q13 deletions including SHANK3 (n = 350, ranging in size from 10 kb to 9.1 Mb) or pathogenic or likely pathogenic SHANK3 sequence variants (n = 51), and used regression models with deletion size as a potential predictor of clinical outcomes for 328 phenotypes. RESULTS Our results showed that increased deletion size was significantly associated with delay in gross and fine motor acquisitions, a spectrum of conditions related to poor muscle tone, renal malformations, mild dysmorphic features (e.g., large fleshy hands, sacral dimple, dysplastic toenails, supernumerary teeth), lymphedema, congenital heart defects, and more frequent neuroimaging abnormalities and infections. These findings indicate that genes upstream of SHANK3 also contribute to some of the manifestations of PMS in individuals with larger deletions. We also showed that self-help skills, verbal ability and a range of psychiatric diagnoses (e.g., autism, ADHD, anxiety disorder) were more common among individuals with smaller deletions and SHANK3 variants. LIMITATIONS Some participants were tested with targeted 22q microarrays rather than genome-wide arrays, and karyotypes were unavailable in many cases, thus precluding the analysis of the effect of other copy number variants or chromosomal rearrangements on the phenotype. CONCLUSIONS This is the largest reported case series of individuals with PMS. Overall, we demonstrate the feasibility of using data from a family-sourced registry to conduct genotype-phenotype analyses in rare genetic disorders. We replicate and strengthen previous findings, and reveal novel associations between larger 22q13 deletions and congenital heart defects, neuroimaging abnormalities and recurrent infections.
Collapse
Affiliation(s)
- Rui Yin
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, 02115, USA
- Department of Health Outcomes and Biomedical Informatics, University of Florida, Gainesville, FL, USA
| | - Maxime Wack
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, 02115, USA
| | - Claire Hassen-Khodja
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, 02115, USA
| | - Michael T McDuffie
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, 02115, USA
| | | | | | - Cartik Kothari
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, 02115, USA
| | | | - Rebecca Davis
- Phelan-McDermid Syndrome Foundation, Osprey, FL, 34229, USA
| | - Kristen Hanson
- Phelan-McDermid Syndrome Foundation, Osprey, FL, 34229, USA
| | - Megan O'Boyle
- Phelan-McDermid Syndrome Foundation, Osprey, FL, 34229, USA
| | - Catalina Betancur
- INSERM, CNRS, Neuroscience Paris Seine, Institut de Biologie Paris Seine, Sorbonne Université, 75005, Paris, France.
| | - Paul Avillach
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
5
|
Bui HTP, Huy Do D, Ly HTT, Tran KT, Le HTT, Nguyen KT, Pham LTD, Le HD, Le VS, Mukhopadhyay A, Nguyen LT. De novo copy number variations in candidate genomic regions in patients of severe autism spectrum disorder in Vietnam. PLoS One 2024; 19:e0290936. [PMID: 38451970 PMCID: PMC10919600 DOI: 10.1371/journal.pone.0290936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 12/09/2023] [Indexed: 03/09/2024] Open
Abstract
Autism spectrum disorder (ASD) is a developmental disorder with a prevalence of around 1% children worldwide and characterized by patient behaviour (communication, social interaction, and personal development). Data on the efficacy of diagnostic tests using copy number variations (CNVs) in candidate genes in ASD is currently around 10% but it is overrepresented by patients of Caucasian background. We report here that the diagnostic success of de novo candidate CNVs in Vietnamese ASD patients is around 6%. We recruited one hundred trios (both parents and a child) where the child was clinically diagnosed with ASD while the parents were not affected. We performed genetic screening to exclude RETT syndrome and Fragile X syndrome and performed genome-wide DNA microarray (aCGH) on all probands and their parents to analyse for de novo CNVs. We detected 1708 non-redundant CNVs in 100 patients and 118 (7%) of them were de novo. Using the filter for known CNVs from the Simons Foundation Autism Research Initiative (SFARI) database, we identified six CNVs (one gain and five loss CNVs) in six patients (3 males and 3 females). Notably, 3 of our patients had a deletion involving the SHANK3 gene-which is the highest compared to previous reports. This is the first report of candidate CNVs in ASD patients from Vietnam and provides the framework for building a CNV based test as the first tier screening for clinical management.
Collapse
Affiliation(s)
- Hoa Thi Phuong Bui
- High Technology Center, Vinmec Health Care System, Hai Ba Trung, Ha Noi, Vietnam
- Translational Medicine Laboratory, Biomedical Research Centre, University of Salford, Salford, United Kingdom
| | - Duong Huy Do
- High Technology Center, Vinmec Health Care System, Hai Ba Trung, Ha Noi, Vietnam
| | - Ha Thi Thanh Ly
- High Technology Center, Vinmec Health Care System, Hai Ba Trung, Ha Noi, Vietnam
| | - Kien Trung Tran
- Vinmec Research Institute of Stem Cell and Gene Technology, Vinmec Health Care System, Hai Ba Trung, Ha Noi, Vietnam
| | - Huong Thi Thanh Le
- Vinmec Research Institute of Stem Cell and Gene Technology, Vinmec Health Care System, Hai Ba Trung, Ha Noi, Vietnam
| | - Kien Trung Nguyen
- Vinmec Research Institute of Stem Cell and Gene Technology, Vinmec Health Care System, Hai Ba Trung, Ha Noi, Vietnam
| | - Linh Thi Dieu Pham
- Vinmec Research Institute of Stem Cell and Gene Technology, Vinmec Health Care System, Hai Ba Trung, Ha Noi, Vietnam
| | - Hau Duc Le
- Big Data Institute, Vinmec Health Care System, Hai Ba Trung, Ha Noi, Vietnam
| | - Vinh Sy Le
- Vinmec Research Institute of Stem Cell and Gene Technology, Vinmec Health Care System, Hai Ba Trung, Ha Noi, Vietnam
- University of Engineering and Technology, Vietnam National University Hanoi, Cau Giay, Hanoi, Vietnam
| | - Arijit Mukhopadhyay
- Translational Medicine Laboratory, Biomedical Research Centre, University of Salford, Salford, United Kingdom
| | - Liem Thanh Nguyen
- Vinmec Research Institute of Stem Cell and Gene Technology, Vinmec Health Care System, Hai Ba Trung, Ha Noi, Vietnam
| |
Collapse
|
6
|
Juan CX, Mao Y, Han X, Qian HY, Chu KK. EGR1 Regulates SHANK3 Transcription at Different Stages of Brain Development. Neuroscience 2024; 540:27-37. [PMID: 38218401 DOI: 10.1016/j.neuroscience.2024.01.006] [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: 06/11/2023] [Revised: 01/04/2024] [Accepted: 01/09/2024] [Indexed: 01/15/2024]
Abstract
The expression levels of SHANK3 are associated with autism spectrum disorder (ASD). The dynamic changes in SHANK3 expression during different stages of brain development may impact the progression of ASD. However, no studies or detailed analyses exploring the upstream mechanisms that regulate SHANK3 expression have been reported. In this study, we employed immunofluorescence to examine the expression of SHANK3 in brain organoids at various stages. Our results revealed elevated levels of SHANK3 expression in brain-like organoids at Day 60. Additionally, we utilized bioinformatics software to predict and analyze the SHANK3 gene's transcription start site. Through the dual luciferase reporter gene technique, we identified core transcription elements within the SHANK3 promoter. Site-directed mutations were used to identify specific transcription sites of SHANK3. To determine the physical binding of potential transcription factors to the SHANK3 promoter, we employed electrophoretic mobility shift assay (EMSA) and chromatin immunoprecipitation (ChIP). Our findings demonstrated that the transcription factor EGR1 regulates SHANK3 expression by binding to the transcription site of the SHANK3 promoter. Although this study did not investigate the pathological phenotypes of human brain organoids or animal model brains with EGR1 deficiency, which could potentially substantiate the findings observed for SHANK3 mutants, our findings provide valuable insights into the relationship between the transcription factor, EGR1, and SHANK3. This study contributes to the molecular understanding of ASD and offers potential foundations for precise targeted therapy.
Collapse
Affiliation(s)
- Chen-Xia Juan
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210004, China; Child Mental Health Research Center, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing 210029, China
| | - Yan Mao
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210004, China
| | - Xiao Han
- Institute for Stem Cell and Neural Regeneration, School of Pharmacy, State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Hua-Ying Qian
- Child Mental Health Research Center, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing 210029, China
| | - Kang-Kang Chu
- Child Mental Health Research Center, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing 210029, China.
| |
Collapse
|
7
|
Kinzig CG, Zakusilo G, Takai KK, Myler LR, de Lange T. ATR blocks telomerase from converting DNA breaks into telomeres. Science 2024; 383:763-770. [PMID: 38359122 PMCID: PMC11267623 DOI: 10.1126/science.adg3224] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 12/13/2023] [Indexed: 02/17/2024]
Abstract
Telomerase, the enzyme that maintains telomeres at natural chromosome ends, should be repressed at double-strand breaks (DSBs), where neotelomere formation can cause terminal truncations. We developed an assay to detect neotelomere formation at Cas9- or I-SceI-induced DSBs in human cells. Telomerase added telomeric repeats to DSBs, leading to interstitial telomeric repeat insertions or the formation of functional neotelomeres accompanied by terminal deletions. The threat that telomerase poses to genome integrity was minimized by ataxia telangiectasia and Rad3-related (ATR) kinase signaling, which inhibited telomerase at resected DSBs. In addition to acting at resected DSBs, telomerase used the extruded strand in the Cas9 enzyme-product complex as a primer for neotelomere formation. We propose that although neotelomere formation is detrimental in normal human cells, it may allow cancer cells to escape from breakage-fusion-bridge cycles.
Collapse
Affiliation(s)
- Charles G. Kinzig
- Laboratory for Cell Biology and Genetics, The Rockefeller University, New York, NY 10065, USA
- Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD/PhD Program, New York, NY 10065, USA
| | - George Zakusilo
- Laboratory for Cell Biology and Genetics, The Rockefeller University, New York, NY 10065, USA
- Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD/PhD Program, New York, NY 10065, USA
| | - Kaori K. Takai
- Laboratory for Cell Biology and Genetics, The Rockefeller University, New York, NY 10065, USA
| | - Logan R. Myler
- Laboratory for Cell Biology and Genetics, The Rockefeller University, New York, NY 10065, USA
| | - Titia de Lange
- Laboratory for Cell Biology and Genetics, The Rockefeller University, New York, NY 10065, USA
| |
Collapse
|
8
|
Shah S, Sarasua SM, Boccuto L, Dean BC, Wang L. Brain Gene Co-Expression Network Analysis Identifies 22q13 Region Genes Associated with Autism, Intellectual Disability, Seizures, Language Impairment, and Hypotonia. Genes (Basel) 2023; 14:1998. [PMID: 38002941 PMCID: PMC10671420 DOI: 10.3390/genes14111998] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 10/22/2023] [Accepted: 10/24/2023] [Indexed: 11/26/2023] Open
Abstract
Phelan-McDermid syndrome (PMS) is a rare genetic neurodevelopmental disorder caused by 22q13 region deletions or SHANK3 gene variants. Deletions vary in size and can affect other genes in addition to SHANK3. PMS is characterized by autism spectrum disorder (ASD), intellectual disability (ID), developmental delays, seizures, speech delay, hypotonia, and minor dysmorphic features. It is challenging to determine individual gene contributions due to variability in deletion sizes and clinical features. We implemented a genomic data mining approach for identifying and prioritizing the candidate genes in the 22q13 region for five phenotypes: ASD, ID, seizures, language impairment, and hypotonia. Weighted gene co-expression networks were constructed using the BrainSpan transcriptome dataset of a human brain. Bioinformatic analyses of the co-expression modules allowed us to select specific candidate genes, including EP300, TCF20, RBX1, XPNPEP3, PMM1, SCO2, BRD1, and SHANK3, for the common neurological phenotypes of PMS. The findings help understand the disease mechanisms and may provide novel therapeutic targets for the precise treatment of PMS.
Collapse
Affiliation(s)
- Snehal Shah
- Healthcare Genetics and Genomics, School of Nursing, Clemson University, Clemson, SC 29634, USA; (S.S.); (L.B.)
- Department of Genetics and Biochemistry, Clemson University, Clemson, SC 29634, USA
| | - Sara M. Sarasua
- Healthcare Genetics and Genomics, School of Nursing, Clemson University, Clemson, SC 29634, USA; (S.S.); (L.B.)
| | - Luigi Boccuto
- Healthcare Genetics and Genomics, School of Nursing, Clemson University, Clemson, SC 29634, USA; (S.S.); (L.B.)
| | - Brian C. Dean
- School of Computing, Clemson University, Clemson, SC 29634, USA
| | - Liangjiang Wang
- Department of Genetics and Biochemistry, Clemson University, Clemson, SC 29634, USA
- Center for Human Genetics, Clemson University, Greenwood, SC 29646, USA
| |
Collapse
|
9
|
Srivastava S, Sahin M, Buxbaum JD, Berry-Kravis E, Soorya LV, Thurm A, Bernstein JA, Asante-Otoo A, Bennett WE, Betancur C, Brickhouse TH, Passos Bueno MR, Chopra M, Christensen CK, Cully JL, Dies K, Friedman K, Gummere B, Holder JL, Jimenez-Gomez A, Kerins CA, Khan O, Kohlenberg T, Lacro RV, Levy LA, Levy T, Linnehan D, Loth E, Moshiree B, Neumeyer A, Paul SM, Phelan K, Persico A, Rapaport R, Rogers C, Saland J, Sethuram S, Shapiro J, Tarr PI, White KM, Wickstrom J, Williams KM, Winrow D, Wishart B, Kolevzon A. Updated consensus guidelines on the management of Phelan-McDermid syndrome. Am J Med Genet A 2023; 191:2015-2044. [PMID: 37392087 PMCID: PMC10524678 DOI: 10.1002/ajmg.a.63312] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 05/04/2023] [Accepted: 05/18/2023] [Indexed: 07/02/2023]
Abstract
Phelan-McDermid syndrome (PMS) is a genetic condition caused by SHANK3 haploinsufficiency and characterized by a wide range of neurodevelopmental and systemic manifestations. The first practice parameters for assessment and monitoring in individuals with PMS were published in 2014; recently, knowledge about PMS has grown significantly based on data from longitudinal phenotyping studies and large-scale genotype-phenotype investigations. The objective of these updated clinical management guidelines was to: (1) reflect the latest in knowledge in PMS and (2) provide guidance for clinicians, researchers, and the general community. A taskforce was established with clinical experts in PMS and representatives from the parent community. Experts joined subgroups based on their areas of specialty, including genetics, neurology, neurodevelopment, gastroenterology, primary care, physiatry, nephrology, endocrinology, cardiology, gynecology, and dentistry. Taskforce members convened regularly between 2021 and 2022 and produced specialty-specific guidelines based on iterative feedback and discussion. Taskforce leaders then established consensus within their respective specialty group and harmonized the guidelines. The knowledge gained over the past decade allows for improved guidelines to assess and monitor individuals with PMS. Since there is limited evidence specific to PMS, intervention mostly follows general guidelines for treating individuals with developmental disorders. Significant evidence has been amassed to guide the management of comorbid neuropsychiatric conditions in PMS, albeit mainly from caregiver report and the experience of clinical experts. These updated consensus guidelines on the management of PMS represent an advance for the field and will improve care in the community. Several areas for future research are also highlighted and will contribute to subsequent updates with more refined and specific recommendations as new knowledge accumulates.
Collapse
Affiliation(s)
- Siddharth Srivastava
- Department of Neurology, Boston Children’s Hospital, Boston, MA, USA
- Rosamund Stone Zander Translational Neuroscience Center, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Mustafa Sahin
- Department of Neurology, Boston Children’s Hospital, Boston, MA, USA
- Rosamund Stone Zander Translational Neuroscience Center, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Joseph D. Buxbaum
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Elizabeth Berry-Kravis
- Department of Pediatrics, Rush University Medical Center, Chicago, IL, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
- Department of Biochemistry, Rush University Medical Center, Chicago, IL, USA
| | | | - Audrey Thurm
- Neurodevelopmental and Behavioral Phenotyping Service, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | | | - Afua Asante-Otoo
- Rehabilitation Medicine Department, NIH Clinical Center, Bethesda, MD, USA
| | - William E. Bennett
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Catalina Betancur
- Sorbonne Université, INSERM, CNRS, Neuroscience Paris Seine, Institut de Biologie Paris Seine, Paris, France
| | - Tegwyn H. Brickhouse
- Department of Dental Public Health & Policy, School of Dentistry, Virginia Commonwealth University, Richmond, VA, USA
| | - Maria Rita Passos Bueno
- Departamento de Genética e Biologia Evolutiva, Centro de Estudos do Genoma Humano e Células-tronco, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Maya Chopra
- Department of Neurology, Boston Children’s Hospital, Boston, MA, USA
- Rosamund Stone Zander Translational Neuroscience Center, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Celanie K. Christensen
- Division of Developmental Medicine, Department of Pediatrics, Riley Children’s Health, Indianapolis, IN, USA
- Division of Child Neurology, Department of Neurology, Riley Children’s Health, Indianapolis, IN, USA
| | - Jennifer L. Cully
- Department of Pediatrics, College of Medicine and Division of Dentistry and Orthodontics, University of Cincinnati, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Kira Dies
- Department of Neurology, Boston Children’s Hospital, Boston, MA, USA
- Rosamund Stone Zander Translational Neuroscience Center, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Kate Friedman
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - J. Lloyd Holder
- Department of Pediatrics-Neurology, Baylor College of Medicine, Houston, TX, USA
| | | | - Carolyn A. Kerins
- Department of Pediatric Dentistry, School of Dentistry, Texas A&M University, Dallas, TX, USA
| | - Omar Khan
- National Institute of Neurological Disease and Stroke, Bethesda, MD, USA
| | | | - Ronald V. Lacro
- Department of Cardiology, Boston Children’s Hospital, Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | | | - Tess Levy
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Eva Loth
- Kings College London, London, UK
| | - Baharak Moshiree
- Department of Medicine, Wake Forest/Atrium Health, Charlotte, NC, USA
| | - Ann Neumeyer
- Lurie Center for Autism, Massachusetts General Hospital, Lexington MA, USA, Harvard Medical School, Boston, MA USA
| | - Scott M. Paul
- Rehabilitation Medicine Department, NIH Clinical Center, Bethesda, MD, USA
| | - Katy Phelan
- Genetics Laboratory, Florida Cancer Specialists and Research Institute, Fort Myers, FL, USA
| | - Antonio Persico
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Robert Rapaport
- Department of Pediatrics, Kravis Children’s Hospital, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Jeffrey Saland
- Department of Pediatrics, Kravis Children’s Hospital, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Swathi Sethuram
- Department of Pediatrics, Massachusetts General Hospital, Boston, MA, USA
| | | | - Phillip I. Tarr
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Kerry M. White
- Division of Developmental Medicine, Department of Pediatrics, Riley Children’s Health, Indianapolis, IN, USA
- Department of Medical and Molecular Genetics, Riley Children’s Health, Indianapolis, IN, USA
| | - Jordan Wickstrom
- Sinai Rehabilitation Center, Lifebridge Health, Baltimore, MD, USA
| | - Kent M. Williams
- Department of Pediatrics, The Ohio State University School of Medicine, Columbus, OH, USA
| | | | | | - Alexander Kolevzon
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pediatrics, Kravis Children’s Hospital, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
10
|
Ngo K, Gittens TH, Gonzalez DI, Hatmaker EA, Plotkin S, Engle M, Friedman GA, Goldin M, Hoerr RE, Eichman BF, Rokas A, Benton ML, Friedman KL. A comprehensive map of hotspots of de novo telomere addition in Saccharomyces cerevisiae. Genetics 2023; 224:iyad076. [PMID: 37119805 PMCID: PMC10474931 DOI: 10.1093/genetics/iyad076] [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: 03/20/2023] [Revised: 04/15/2023] [Accepted: 04/18/2023] [Indexed: 05/01/2023] Open
Abstract
Telomere healing occurs when telomerase, normally restricted to chromosome ends, acts upon a double-strand break to create a new, functional telomere. De novo telomere addition (dnTA) on the centromere-proximal side of a break truncates the chromosome but, by blocking resection, may allow the cell to survive an otherwise lethal event. We previously identified several sequences in the baker's yeast, Saccharomyces cerevisiae, that act as hotspots of dnTA [termed Sites of Repair-associated Telomere Addition (SiRTAs)], but the distribution and functional relevance of SiRTAs is unclear. Here, we describe a high-throughput sequencing method to measure the frequency and location of telomere addition within sequences of interest. Combining this methodology with a computational algorithm that identifies SiRTA sequence motifs, we generate the first comprehensive map of telomere-addition hotspots in yeast. Putative SiRTAs are strongly enriched in subtelomeric regions where they may facilitate formation of a new telomere following catastrophic telomere loss. In contrast, outside of subtelomeres, the distribution and orientation of SiRTAs appears random. Since truncating the chromosome at most SiRTAs would be lethal, this observation argues against selection for these sequences as sites of telomere addition per se. We find, however, that sequences predicted to function as SiRTAs are significantly more prevalent across the genome than expected by chance. Sequences identified by the algorithm bind the telomeric protein Cdc13, raising the possibility that association of Cdc13 with single-stranded regions generated during the response to DNA damage may facilitate DNA repair more generally.
Collapse
Affiliation(s)
- Katrina Ngo
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37232 USA
| | - Tristen H Gittens
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37232 USA
| | - David I Gonzalez
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37232 USA
| | - E Anne Hatmaker
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37232 USA
- Evolutionary Studies Initiative, Vanderbilt University, Nashville, TN 37232 USA
| | - Simcha Plotkin
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37232 USA
| | - Mason Engle
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37232 USA
| | - Geofrey A Friedman
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37232 USA
| | - Melissa Goldin
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37232 USA
| | - Remington E Hoerr
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37232 USA
| | - Brandt F Eichman
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37232 USA
- Department of Biochemistry, Vanderbilt University, Nashville, TN 37232 USA
| | - Antonis Rokas
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37232 USA
- Evolutionary Studies Initiative, Vanderbilt University, Nashville, TN 37232 USA
| | | | - Katherine L Friedman
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37232 USA
| |
Collapse
|
11
|
Koza SA, Tabet AC, Bonaglia MC, Andres S, Anderlid BM, Aten E, Stiefsohn D. Consensus recommendations on counselling in Phelan-McDermid syndrome, with special attention to recurrence risk and to ring chromosome 22. Eur J Med Genet 2023; 66:104773. [PMID: 37120077 DOI: 10.1016/j.ejmg.2023.104773] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 04/10/2023] [Accepted: 04/26/2023] [Indexed: 05/01/2023]
Abstract
This paper focuses on genetic counselling in Phelan-McDermid syndrome (PMS), a rare neurodevelopmental disorder caused by a deletion 22q13.3 or a pathogenic variant in SHANK3. It is one of a series of papers written by the European PMS consortium as a consensus guideline. We reviewed the available literature based on pre-set questions to formulate recommendations on counselling, diagnostic work-up and surveillance for tumours related to ring chromosome 22. All recommendations were approved by the consortium, which consists of professionals and patient representatives, using a voting procedure. PMS can only rarely be diagnosed based solely on clinical features and requires confirmation via genetic testing. In most cases, the family will be referred to a clinical geneticist for counselling after the genetic diagnosis has been made. Family members will be investigated and, if indicated, the chance of recurrence discussed with them. Most individuals with PMS have a de novo deletion or a pathogenic variant of SHANK3. The 22q13.3 deletion can be a simple deletion, a ring chromosome 22, or the result of a parental balanced chromosomal anomaly, influencing the risk of recurrence. Individuals with a ring chromosome 22 have an increased risk of NF2-related schwannomatosis (formerly neurofibromatosis type 2) and atypical teratoid rhabdoid tumours, which are associated with the tumour-suppressor genes NF2 and SMARCB1, respectively, and both genes are located on chromosome 22. The prevalence of PMS due to a ring chromosome 22 is estimated to be 10-20%. The risk of developing a tumour in an individual with a ring chromosome 22 can be calculated as 2-4%. However, those individuals who do develop tumours often have multiple. We recommend referring all individuals with PMS and their parents to a clinical geneticist or a comparably experienced medical specialist for genetic counselling, further genetic testing, follow-up and discussion of prenatal diagnostic testing in subsequent pregnancies. We also recommend karyotyping to diagnose or exclude a ring chromosome 22 in individuals with a deletion 22q13.3 detected by molecular tests. If a ring chromosome 22 is found, we recommend discussing personalised follow-up for NF2-related tumours and specifically cerebral imaging between the age of 14 and 16 years.
Collapse
Affiliation(s)
- Sylvia A Koza
- University of Groningen, University Medical Centre Groningen, Department of Genetics, Groningen, the Netherlands
| | - Anne C Tabet
- Cytogenetic Unit, Genetic Department, Robert Debré Hospital, Human Genetic and Cognitive Function, Pasteur Institute, Paris, France
| | - Maria C Bonaglia
- Cytogenetics Laboratory, Scientific Institute, IRCCS Eugenio Medea, Bosisio Parini, Lecco, Italy
| | | | - Britt-Marie Anderlid
- Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden
| | - Emmelien Aten
- Leiden University Medical Center, Department of Clinical Genetics, Leiden, the Netherlands
| | | |
Collapse
|
12
|
Ngo K, Gittens TH, Gonzalez DI, Hatmaker EA, Plotkin S, Engle M, Friedman GA, Goldin M, Hoerr RE, Eichman BF, Rokas A, Benton ML, Friedman KL. A comprehensive map of hotspots of de novo telomere addition in Saccharomyces cerevisiae. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.20.533556. [PMID: 36993206 PMCID: PMC10055226 DOI: 10.1101/2023.03.20.533556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Telomere healing occurs when telomerase, normally restricted to chromosome ends, acts upon a double-strand break to create a new, functional telomere. De novo telomere addition on the centromere-proximal side of a break truncates the chromosome but, by blocking resection, may allow the cell to survive an otherwise lethal event. We previously identified several sequences in the baker’s yeast, Saccharomyces cerevisiae , that act as hotspots of de novo telomere addition (termed Sites of Repair-associated Telomere Addition or SiRTAs), but the distribution and functional relevance of SiRTAs is unclear. Here, we describe a high-throughput sequencing method to measure the frequency and location of telomere addition within sequences of interest. Combining this methodology with a computational algorithm that identifies SiRTA sequence motifs, we generate the first comprehensive map of telomere-addition hotspots in yeast. Putative SiRTAs are strongly enriched in subtelomeric regions where they may facilitate formation of a new telomere following catastrophic telomere loss. In contrast, outside of subtelomeres, the distribution and orientation of SiRTAs appears random. Since truncating the chromosome at most SiRTAs would be lethal, this observation argues against selection for these sequences as sites of telomere addition per se. We find, however, that sequences predicted to function as SiRTAs are significantly more prevalent across the genome than expected by chance. Sequences identified by the algorithm bind the telomeric protein Cdc13, raising the possibility that association of Cdc13 with single-stranded regions generated during the response to DNA damage may facilitate DNA repair more generally.
Collapse
Affiliation(s)
- Katrina Ngo
- Department of Biological Sciences, Vanderbilt University
| | | | | | - E. Anne Hatmaker
- Department of Biological Sciences, Vanderbilt University
- Evolutionary Studies Initiative, Vanderbilt University
| | - Simcha Plotkin
- Department of Biological Sciences, Vanderbilt University
| | - Mason Engle
- Department of Biological Sciences, Vanderbilt University
| | | | - Melissa Goldin
- Department of Biological Sciences, Vanderbilt University
| | | | - Brandt F. Eichman
- Department of Biological Sciences, Vanderbilt University
- Department of Biochemistry, Vanderbilt University
| | - Antonis Rokas
- Department of Biological Sciences, Vanderbilt University
- Evolutionary Studies Initiative, Vanderbilt University
| | | | | |
Collapse
|
13
|
Jesse S, Kuhlmann L, Hildebrand LS, Magelssen H, Schmaus M, Timmermann B, Andres S, Fietkau R, Distel LV. Increased Radiation Sensitivity in Patients with Phelan-McDermid Syndrome. Cells 2023; 12:cells12050820. [PMID: 36899955 PMCID: PMC10000830 DOI: 10.3390/cells12050820] [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: 02/16/2023] [Revised: 03/03/2023] [Accepted: 03/03/2023] [Indexed: 03/09/2023] Open
Abstract
Phelan-McDermid syndrome is an inherited global developmental disorder commonly associated with autism spectrum disorder. Due to a significantly increased radiosensitivity, measured before the start of radiotherapy of a rhabdoid tumor in a child with Phelan-McDermid syndrome, the question arose whether other patients with this syndrome also have increased radiosensitivity. For this purpose, the radiation sensitivity of blood lymphocytes after irradiation with 2Gray was examined using the G0 three-color fluorescence in situ hybridization assay in a cohort of 20 patients with Phelan-McDermid syndrome from blood samples. The results were compared to healthy volunteers, breast cancer patients and rectal cancer patients. Independent of age and gender, all but two patients with Phelan-McDermid syndrome showed significantly increased radiosensitivity, with an average of 0.653 breaks per metaphase. These results correlated neither with the individual genetic findings nor with the individual clinical course, nor with the respective clinical severity of the disease. In our pilot study, we saw a significantly increased radiosensitivity in lymphocytes from patients with Phelan-McDermid syndrome, so pronounced that a dose reduction would be recommended if radiotherapy had to be performed. Ultimately, the question arises as to the interpretation of these data. There does not appear to be an increased risk of tumors in these patients, since tumors are rare overall. The question, therefore, arose as to whether our results could possibly be the basis for processes, such as aging/preaging, or, in this context, neurodegeneration. There are no data on this so far, but this issue should be pursued in further fundamentally based studies in order to better understand the pathophysiology of the syndrome.
Collapse
Affiliation(s)
- Sarah Jesse
- Department of Neurology, Ulm University, 89081 Ulm, Germany
| | - Lukas Kuhlmann
- Department of Radiation Biology, Erlangen University, 91054 Erlangen, Germany
| | - Laura S. Hildebrand
- Department of Radiation Biology, Erlangen University, 91054 Erlangen, Germany
| | - Henriette Magelssen
- Department of Oncology, Oslo University Hospital (The Norwegian Radium Hospital), 0424 Oslo, Norway
| | - Martina Schmaus
- Department of Radiotherapy and Radiation Oncology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Beate Timmermann
- Clinic for Particle Therapy at WPE, University Hospital Essen, 45147 Essen, Germany
| | | | - Rainer Fietkau
- Department of Radiation Biology, Erlangen University, 91054 Erlangen, Germany
| | - Luitpold V. Distel
- Department of Radiation Biology, Erlangen University, 91054 Erlangen, Germany
- Correspondence:
| |
Collapse
|
14
|
Vitrac A, Leblond CS, Rolland T, Cliquet F, Mathieu A, Maruani A, Delorme R, Schön M, Grabrucker AM, van Ravenswaaij-Arts C, Phelan K, Tabet AC, Bourgeron T. Dissecting the 22q13 region to explore the genetic and phenotypic diversity of patients with Phelan-McDermid syndrome. Eur J Med Genet 2023; 66:104732. [PMID: 36822569 DOI: 10.1016/j.ejmg.2023.104732] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 02/14/2023] [Accepted: 02/19/2023] [Indexed: 02/25/2023]
Abstract
SHANK3-related Phelan-McDermid syndrome (PMS) is caused by a loss of the distal part of chromosome 22, including SHANK3, or by a pathological SHANK3 variant. There is an important genetic and phenotypic diversity among patients who can present with developmental delay, language impairments, autism, epilepsy, and other symptoms. SHANK3, encoding a synaptic scaffolding protein, is deleted in the majority of patients with PMS and is considered a major gene involved in the neurological impairments of the patients. However, differences in deletion size can influence clinical features, and in some rare cases, deletions at the 22q13 locus in individuals with SHANK3-unrelated PMS do not encompass SHANK3. These individuals with SHANK3-unrelated PMS still display a PMS-like phenotype. This suggests the participation of other 22q13 genes in the pathogenesis of PMS. Here, we review the biological function and potential implication in PMS symptoms of 110 genes located in the 22q13 region, focusing on 35 genes with evidence for association with neurodevelopmental disorders, including 13 genes for epilepsy and 11 genes for microcephaly and/or macrocephaly. Our review is restricted to the 22q13 region, but future large-scale studies using whole genome sequencing and deep-phenotyping are warranted to develop predictive models of clinical trajectories and to target specific medical and educational care for each individual with PMS.
Collapse
Affiliation(s)
- Aline Vitrac
- Génétique Humaine et Fonctions Cognitives, Institut Pasteur, UMR3571 CNRS, Université de Paris Cité, IUF, 75015, Paris, France.
| | - Claire S Leblond
- Génétique Humaine et Fonctions Cognitives, Institut Pasteur, UMR3571 CNRS, Université de Paris Cité, IUF, 75015, Paris, France
| | - Thomas Rolland
- Génétique Humaine et Fonctions Cognitives, Institut Pasteur, UMR3571 CNRS, Université de Paris Cité, IUF, 75015, Paris, France
| | - Freddy Cliquet
- Génétique Humaine et Fonctions Cognitives, Institut Pasteur, UMR3571 CNRS, Université de Paris Cité, IUF, 75015, Paris, France
| | - Alexandre Mathieu
- Génétique Humaine et Fonctions Cognitives, Institut Pasteur, UMR3571 CNRS, Université de Paris Cité, IUF, 75015, Paris, France
| | - Anna Maruani
- Department of Child and Adolescent Psychiatry, Hôpital Robert Debré, APHP, Paris, France
| | - Richard Delorme
- Department of Child and Adolescent Psychiatry, Hôpital Robert Debré, APHP, Paris, France
| | - Michael Schön
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany
| | - Andreas M Grabrucker
- Bernal Institute, University of Limerick, Limerick, Ireland; Dept. of Biological Sciences, University of Limerick, Limerick, Ireland; Health Research Institute HRI, University of Limerick, Limerick, Ireland
| | - Conny van Ravenswaaij-Arts
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, Netherlands
| | - Katy Phelan
- Genetics Laboratory, Florida Cancer Specialists & Research Institute, Fort Myers, FL, 33916, USA
| | | | - Thomas Bourgeron
- Génétique Humaine et Fonctions Cognitives, Institut Pasteur, UMR3571 CNRS, Université de Paris Cité, IUF, 75015, Paris, France.
| |
Collapse
|
15
|
Sarasua SM, DeLuca JM, Rogers C, Phelan K, Rennert L, Powder KE, Weisensee K, Boccuto L. Head Size in Phelan-McDermid Syndrome: A Literature Review and Pooled Analysis of 198 Patients Identifies Candidate Genes on 22q13. Genes (Basel) 2023; 14:540. [PMID: 36980813 PMCID: PMC10048319 DOI: 10.3390/genes14030540] [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: 12/19/2022] [Revised: 02/11/2023] [Accepted: 02/12/2023] [Indexed: 02/24/2023] Open
Abstract
Phelan-McDermid syndrome (PMS) is a multisystem disorder that is associated with deletions of the 22q13 genomic region or pathogenic variants in the SHANK3 gene. Notable features include developmental issues, absent or delayed speech, neonatal hypotonia, seizures, autism or autistic traits, gastrointestinal problems, renal abnormalities, dolichocephaly, and both macro- and microcephaly. Assessment of the genetic factors that are responsible for abnormal head size in PMS has been hampered by small sample sizes as well as a lack of attention to these features. Therefore, this study was conducted to investigate the relationship between head size and genes on chromosome 22q13. A review of the literature was conducted to identify published cases of 22q13 deletions with information on head size to conduct a pooled association analysis. Across 56 studies, we identified 198 cases of PMS with defined deletion sizes and head size information. A total of 33 subjects (17%) had macrocephaly, 26 (13%) had microcephaly, and 139 (70%) were normocephalic. Individuals with macrocephaly had significantly larger genomic deletions than those with microcephaly or normocephaly (p < 0.0001). A genomic region on 22q13.31 was found to be significantly associated with macrocephaly with CELSR1, GRAMD4, and TBCD122 suggested as candidate genes. Investigation of these genes will aid the understanding of head and brain development.
Collapse
Affiliation(s)
- Sara M. Sarasua
- Healthcare Genetics and Genomics Program, Clemson University School of Nursing, Clemson, SC 29634, USA
| | - Jane M. DeLuca
- Healthcare Genetics and Genomics Program, Clemson University School of Nursing, Clemson, SC 29634, USA
| | | | - Katy Phelan
- Florida Cancer Specialists & Research Institute, Fort Myers, FL 33908, USA
| | - Lior Rennert
- Department of Public Health Sciences, Clemson University, Clemson, SC 29634, USA
| | - Kara E. Powder
- Department of Biological Sciences, Clemson University, Clemson, SC 29634, USA
| | - Katherine Weisensee
- Department of Sociology, Anthropology and Criminal Justice, Clemson University, Clemson, SC 29634, USA
| | - Luigi Boccuto
- Healthcare Genetics and Genomics Program, Clemson University School of Nursing, Clemson, SC 29634, USA
| |
Collapse
|
16
|
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
|
17
|
Breen MS, Fan X, Levy T, Pollak RM, Collins B, Osman A, Tocheva AS, Sahin M, Berry-Kravis E, Soorya L, Thurm A, Powell CM, Bernstein JA, Kolevzon A, Buxbaum JD. Large 22q13.3 deletions perturb peripheral transcriptomic and metabolomic profiles in Phelan-McDermid syndrome. HGG ADVANCES 2023; 4:100145. [PMID: 36276299 PMCID: PMC9579712 DOI: 10.1016/j.xhgg.2022.100145] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 09/23/2022] [Indexed: 11/25/2022] Open
Abstract
Phelan-McDermid syndrome (PMS) is a rare neurodevelopmental disorder caused at least in part by haploinsufficiency of the SHANK3 gene, due to sequence variants in SHANK3 or subtelomeric 22q13.3 deletions. Phenotypic differences have been reported between PMS participants carrying small "class I" mutations and large "class II" mutations; however, the molecular perturbations underlying these divergent phenotypes remain obscure. Using peripheral blood transcriptome and serum metabolome profiling, we examined the molecular perturbations in the peripheral circulation associated with a full spectrum of PMS genotypes spanning class I (n = 37) and class II mutations (n = 39). Transcriptomic data revealed 52 genes with blood expression profiles that tightly scale with 22q.13.3 deletion size. Furthermore, we uncover 208 underexpressed genes in PMS participants with class II mutations, which were unchanged in class I mutations. These genes were not linked to 22q13.3 and were strongly enriched for glycosphingolipid metabolism, NCAM1 interactions, and cytotoxic natural killer (NK) immune cell signatures. In silico predictions estimated a reduction in CD56+ CD16- NK cell proportions in class II mutations, which was validated by mass cytometry time of flight. Global metabolomics profiling identified 24 metabolites that were significantly altered in PMS participants with class II mutations and confirmed a general reduction in sphingolipid metabolism. Collectively, these results provide new evidence linking PMS participants carrying class II mutations with decreased expression of cytotoxic cell signatures, reduced relative proportions of NK cells, and lower sphingolipid metabolism. These findings highlight alternative avenues for therapeutic development and offer new mechanistic insights supporting genotype-to-phenotype associations in PMS.
Collapse
Affiliation(s)
- Michael S Breen
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Xuanjia Fan
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Tess Levy
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Rebecca M Pollak
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Brett Collins
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Aya Osman
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anna S Tocheva
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mustafa Sahin
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.,Rosamund Stone Zander Translational Neuroscience Center and F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Elizabeth Berry-Kravis
- Department of Pediatrics, Rush University Medical Center, Chicago, IL, USA.,Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Latha Soorya
- Department of Psychiatry, Rush University Medical Center, Chicago, IL, USA
| | - Audrey Thurm
- Neurodevelopmental and Behavioral Phenotyping Service, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Craig M Powell
- Department of Neurobiology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL, USA.,Civitan International Research Center, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL, USA
| | - Jonathan A Bernstein
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Alexander Kolevzon
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Joseph D Buxbaum
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | |
Collapse
|
18
|
I DV, Proskokova TN. [Phelan-McDermid syndrome associated with a novel heterozygous mutation in the SHANK3 gene]. Zh Nevrol Psikhiatr Im S S Korsakova 2023; 123:124-128. [PMID: 37655421 DOI: 10.17116/jnevro2023123081124] [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] [Indexed: 09/02/2023]
Abstract
Phelan-McDermid syndrome (PMS) is a hereditary disorder associated with microdeletions of chromosome 22q13 or point mutations in SHANK3, characterized by mental and speech delays, intellectual disability, epilepsy and autism spectrum disorder. We describe a case PMS associated with a heterozygous mutation c.2486delC (p.Pro829fs) in SHANK3. The diagnostic pathway of a female patient with PMS took more than 7 years; the reason for treatment was the onset of epileptic seizures and impaired speech development. The existence of different types of rearrangements and genomic variations can explain the high clinical variability observed in individuals with PMS. Only molecular diagnosis can accurately diagnose individuals with PMS for follow-up and medical genetic counselling of families.
Collapse
Affiliation(s)
- D V I
- Khabarovsk Center for the Development of Psychology and Childhood «Psylogia», Khabarovsk, Russia
- Far-East State Medical University, Khabarovsk, Russia
| | | |
Collapse
|
19
|
Wan L, Liu D, Xiao WB, Zhang BX, Yan XX, Luo ZH, Xiao B. Association of SHANK Family with Neuropsychiatric Disorders: An Update on Genetic and Animal Model Discoveries. Cell Mol Neurobiol 2022; 42:1623-1643. [PMID: 33595806 PMCID: PMC11421742 DOI: 10.1007/s10571-021-01054-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 02/02/2021] [Indexed: 12/14/2022]
Abstract
The Shank family proteins are enriched at the postsynaptic density (PSD) of excitatory glutamatergic synapses. They serve as synaptic scaffolding proteins and appear to play a critical role in the formation, maintenance and functioning of synapse. Increasing evidence from genetic association and animal model studies indicates a connection of SHANK genes defects with the development of neuropsychiatric disorders. In this review, we first update the current understanding of the SHANK family genes and their encoded protein products. We then denote the literature relating their alterations to the risk of neuropsychiatric diseases. We further review evidence from animal models that provided molecular insights into the biological as well as pathogenic roles of Shank proteins in synapses, and the potential relationship to the development of abnormal neurobehavioral phenotypes.
Collapse
Affiliation(s)
- Lily Wan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Du Liu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Taikang Tongji Hospital, Wuhan, 430050, Hubei, China
| | - Wen-Biao Xiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Bo-Xin Zhang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Xiao-Xin Yan
- Department of Anatomy and Neurobiology, Central South University, Changsha, 410013, Hunan, China
| | - Zhao-Hui Luo
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| | - Bo Xiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
20
|
Serrada-Tejeda S, Martínez-Piédrola RM, Máximo-Bocanegra N, Sánchez-Herrera-Baeza P, Pérez-de-Heredia-Torres M. Descriptive Analysis of Adaptive Behavior in Phelan–McDermid Syndrome and Autism Spectrum Disorder. Front Neurosci 2022; 16:893003. [PMID: 35864987 PMCID: PMC9295709 DOI: 10.3389/fnins.2022.893003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 06/07/2022] [Indexed: 12/02/2022] Open
Abstract
Introduction The variety in symptomatology and clinical presentation of individuals diagnosed with Phelan-McDermid Syndrome (PMS) can delay medical diagnosis, so identifying specific neurobehavioral variables and facilitating differential diagnosis with patients with idiopathic Autism Spectrum Disorder (ASD) can guide early detection. Methods A descriptive analysis of the level of adaptive behavior in 50 patients diagnosed with PMS was performed (SHANK3deletion: N = 44; SHANKmutation: N = 6). Subsequently, a comparative analysis was performed with 28 children aged between 4 years and 6 years and 11 months (SHANK3deletion = 14; ASD = 14). Differences between the two groups were evaluated and Bonferroni correction was applied for multiple comparisons. Results Differences were identified in the variables of communication (z = −2.715, p = 0.007), Self-Direction (z = −2.199, p = 0.028) and social participation (z = −3.190, p = 0.001), with better adaptive behavior skills being observed in participants with a SHANK3mutation. Better adaptive skills in the sample of participants with ASD, were found and statistically significant differences were identified in the variables of academic skills (z = −3.084, p = 0.002), use of community resources (z = −1.889, p = 0.050) and health and safety (z = −2.90, p = 0.004). Conclusion Participants with SHANK3mutation show better communication and social participation skills than those with a diagnosis of SHANK3deletion. The observed differences between ASD and individuals with PMS reflect deficits in practical and conceptual adaptive skills that may limit and hinder daily adaptive functioning.
Collapse
|
21
|
Yamashita H, Arakawa Y, Terada Y, Takeuchi Y, Mineharu Y, Sumiyoshi S, Tokunaga S, Nakajima K, Kawabata N, Tanaka K, Tanji M, Umeda K, Minamiguchi S, Ogawa S, Haga H, Takita J, Miyamoto S. Whole-genome sequencing analysis of an atypical teratoid/rhabdoid tumor in a patient with Phelan-McDermid syndrome: a case report and systematic review. Brain Tumor Pathol 2022; 39:232-239. [PMID: 35750988 DOI: 10.1007/s10014-022-00440-7] [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: 03/29/2022] [Accepted: 05/31/2022] [Indexed: 11/24/2022]
Abstract
Atypical teratoid/rhabdoid tumor (AT/RT) is a rare pediatric brain tumor with abnormalities in SMARCB1 located in 22q11.2. We report a case of AT/RT associated with Phelan-McDermid syndrome (PMS) characterized by congenital developmental disorder, mental retardation, and ring chromosome 22 with 22q13.3-qter depletion, for which we performed whole-genome sequencing (WGS). A 4-year-old girl with a developmental disability was referred to our hospital due to dysphoria. Brain magnetic resonance imaging showed a 5-cm well-demarcated mass that extended bilaterally in the frontal lobes. G-banding was performed preoperatively due to a history of developmental retardation. Ring chromosome 22 and deletion of 22q13.3-qter were observed, and she was diagnosed with PMS. She underwent gross total resection of the tumor, and the pathological diagnosis was AT/RT. WGS showed somatic SMARCB1 mutation (p.R201X) and somatic loss of the entire chromosome 22 in the tumor, but not in the blood sample. WGS confirmed previously unreported BRCA2 mutations, 6q loss, and 14q acquisition during tumor progression, but no other significant findings associated with tumor progression. The present case is discussed with reference to a systematic review of previous reports of AT/RT associated with PMS. PMS patients with ring chromosome 22 should be carefully followed up for AT/RT occurrence.
Collapse
Affiliation(s)
- Haruki Yamashita
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, 54 Kawahara-cho Shogoin Sakyo-ku, Kyoto, 606-8507, Japan
| | - Yoshiki Arakawa
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, 54 Kawahara-cho Shogoin Sakyo-ku, Kyoto, 606-8507, Japan.
| | - Yukinori Terada
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, 54 Kawahara-cho Shogoin Sakyo-ku, Kyoto, 606-8507, Japan
| | - Yasuhide Takeuchi
- Department of Diagnostic Pathology, Kyoto University Graduate School of Medicine, Kyoto, 606-8507, Japan
| | - Yohei Mineharu
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, 54 Kawahara-cho Shogoin Sakyo-ku, Kyoto, 606-8507, Japan
| | - Sosuke Sumiyoshi
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, 54 Kawahara-cho Shogoin Sakyo-ku, Kyoto, 606-8507, Japan
| | - Shinya Tokunaga
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, 54 Kawahara-cho Shogoin Sakyo-ku, Kyoto, 606-8507, Japan
| | - Kohei Nakajima
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, 54 Kawahara-cho Shogoin Sakyo-ku, Kyoto, 606-8507, Japan
| | - Naoko Kawabata
- Department of Pediatrics, Kyoto University Graduate School of Medicine, Kyoto, 606-8507, Japan
| | - Kuniaki Tanaka
- Department of Pediatrics, Kyoto University Graduate School of Medicine, Kyoto, 606-8507, Japan
| | - Masahiro Tanji
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, 54 Kawahara-cho Shogoin Sakyo-ku, Kyoto, 606-8507, Japan
| | - Katsutsugu Umeda
- Department of Pediatrics, Kyoto University Graduate School of Medicine, Kyoto, 606-8507, Japan
| | - Sachiko Minamiguchi
- Department of Diagnostic Pathology, Kyoto University Graduate School of Medicine, Kyoto, 606-8507, Japan
| | - Seishi Ogawa
- Department of Pathology and Tumor Biology, Kyoto University, Kyoto, 606-8507, Japan
| | - Hironori Haga
- Department of Diagnostic Pathology, Kyoto University Graduate School of Medicine, Kyoto, 606-8507, Japan
| | - Junko Takita
- Department of Pediatrics, Kyoto University Graduate School of Medicine, Kyoto, 606-8507, Japan
| | - Susumu Miyamoto
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, 54 Kawahara-cho Shogoin Sakyo-ku, Kyoto, 606-8507, Japan
| |
Collapse
|
22
|
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
|
23
|
Zuffardi O, Fichera M, Bonaglia MC. The embryo battle against adverse genomes: Are de novo terminal deletions the rescue of unfavorable zygotic imbalances? Eur J Med Genet 2022; 65:104532. [PMID: 35724817 DOI: 10.1016/j.ejmg.2022.104532] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 04/02/2022] [Accepted: 05/21/2022] [Indexed: 11/03/2022]
Abstract
De novo distal deletions are structural variants considered to be already present in the zygote. However, investigations especially in the prenatal setting have documented that they are often in mosaic with cell lines in which the same deleted chromosome shows different types of aberrations such as: 1) neutral copy variants with loss of heterozygosity that replace the deleted region with equivalent portions of the homologous chromosome and create distal uniparental disomy (UPD); 2) derivative chromosomes where the deleted one ends with the distal region of another chromosome or has the shape of a ring; 3) U-type mirror dicentric or inv-dup del rearrangements. Unstable dicentrics had already been entailed as causative of terminal deletions even when no trace of the reciprocal inv-dup del had been detected. To clarify the mechanism of origin of distal deletions, we examined PubMed using as keywords: complex/mosaic chromosomal deletions, distal UPD, U-type dicentrics, inv-dup del chromosomes, excluding the recurrent inv-dup del(8p)s which are known to originate by NAHR at the maternal meiosis. The literature has shown that U-type dicentrics leading to nearly complete trisomy and therefore incompatible with zygotic survival underlie many types of de novo unbalanced rearrangements, including terminal deletions. In the early embryo, the position of the postzygotic breaks of the dicentric, the different ways of acquiring telomeres by the broken portions and the selection of the most favorable cell lines in the different tissues determine the prevalence of one or the other rearrangement. Multiple lines with simple terminal deletions, inv-dup dels, unbalanced translocations and segmental UPDs can coexist in various mosaic combinations although it is rare to identify them all in the blood. Regarding the origin of the dicentric, among the 30 cases of non-recurrent inv-dup del with sufficient genotyping information, paternal origin was markedly prevalent with consistently identical polymorphisms within the duplication region, regardless of parental origin. The non-random parental origin made any postzygotic origin unlikely and suggested the occurrence of these dicentrics mainly in spermatogenesis. This study strengthens the evidence that non-recurrent de novo structural rearrangements are often secondary to the rescue of a zygotic genome incompatible with embryo survival.
Collapse
Affiliation(s)
- Orsetta Zuffardi
- Department of Molecular Medicine, University of Pavia, Pavia, Italy.
| | - Marco Fichera
- Department of Biomedical and Biotechnological Sciences, Medical Genetics, University of Catania, Catania, Italy; Oasi Research Institute-IRCCS, Troina, Italy.
| | - Maria Clara Bonaglia
- Cytogenetics Laboratory, Scientific Institute, IRCCS Eugenio Medea, Bosisio Parini, Lecco, Italy.
| |
Collapse
|
24
|
State of the Science for Kidney Disorders in Phelan-McDermid Syndrome: UPK3A, FBLN1, WNT7B, and CELSR1 as Candidate Genes. Genes (Basel) 2022; 13:genes13061042. [PMID: 35741804 PMCID: PMC9223119 DOI: 10.3390/genes13061042] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 05/30/2022] [Accepted: 06/02/2022] [Indexed: 01/27/2023] Open
Abstract
Phelan-McDermid syndrome (PMS) is a neurodevelopmental disorder caused by chromosomal rearrangements affecting the 22q13.3 region or by SHANK3 pathogenic variants. The scientific literature suggests that up to 40% of individuals with PMS have kidney disorders, yet little research has been conducted on the renal system to assess candidate genes attributed to these disorders. Therefore, we first conducted a systematic review of the literature to identify kidney disorders in PMS and then pooled the data to create a cohort of individuals to identify candidate genes for renal disorders in PMS. We found 7 types of renal disorders reported: renal cysts, renal hypoplasia or agenesis, hydronephrosis, vesicoureteral reflux, kidney dysplasia, horseshoe kidneys, and pyelectasis. Association analysis from the pooled data from 152 individuals with PMS across 22 articles identified three genomic regions spanning chromosomal bands 22q13.31, 22q13.32, and 22q13.33, significantly associated with kidney disorders. We propose UPK3A, FBLN1, WNT7B, and CELSR1, located from 4.5 Mb to 5.5 Mb from the telomere, as candidate genes. Our findings support the hypothesis that genes included in this region may play a role in the pathogenesis of kidney disorders in PMS.
Collapse
|
25
|
Nevado J, García-Miñaúr S, Palomares-Bralo M, Vallespín E, Guillén-Navarro E, Rosell J, Bel-Fenellós C, Mori MÁ, Milá M, del Campo M, Barrúz P, Santos-Simarro F, Obregón G, Orellana C, Pachajoa H, Tenorio JA, Galán E, Cigudosa JC, Moresco A, Saleme C, Castillo S, Gabau E, Pérez-Jurado L, Barcia A, Martín MS, Mansilla E, Vallcorba I, García-Murillo P, Cammarata-Scalisi F, Gonçalves Pereira N, Blanco-Lago R, Serrano M, Ortigoza-Escobar JD, Gener B, Seidel VA, Tirado P, Lapunzina P. Variability in Phelan-McDermid Syndrome in a Cohort of 210 Individuals. Front Genet 2022; 13:652454. [PMID: 35495150 PMCID: PMC9044489 DOI: 10.3389/fgene.2022.652454] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 02/16/2022] [Indexed: 12/18/2022] Open
Abstract
Phelan-McDermid syndrome (PMS, OMIM# 606232) results from either different rearrangements at the distal region of the long arm of chromosome 22 (22q13.3) or pathogenic sequence variants in the SHANK3 gene. SHANK3 codes for a structural protein that plays a central role in the formation of the postsynaptic terminals and the maintenance of synaptic structures. Clinically, patients with PMS often present with global developmental delay, absent or severely delayed speech, neonatal hypotonia, minor dysmorphic features, and autism spectrum disorders (ASD), among other findings. Here, we describe a cohort of 210 patients with genetically confirmed PMS. We observed multiple variant types, including a significant number of small deletions (<0.5 Mb, 64/189) and SHANK3 sequence variants (21 cases). We also detected multiple types of rearrangements among microdeletion cases, including a significant number with post-zygotic mosaicism (9.0%, 17/189), ring chromosome 22 (10.6%, 20/189), unbalanced translocations (de novo or inherited, 6.4%), and additional rearrangements at 22q13 (6.3%, 12/189) as well as other copy number variations in other chromosomes, unrelated to 22q deletions (14.8%, 28/189). We compared the clinical and genetic characteristics among patients with different sizes of deletions and with SHANK3 variants. Our findings suggest that SHANK3 plays an important role in this syndrome but is probably not uniquely responsible for all the spectrum features in PMS. We emphasize that only an adequate combination of different molecular and cytogenetic approaches allows an accurate genetic diagnosis in PMS patients. Thus, a diagnostic algorithm is proposed.
Collapse
Affiliation(s)
- Julián Nevado
- Instituto de Genética Médica y Molecular (INGEMM)-IdiPAZ, Hospital Universitario La Paz, Madrid, Spain
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain
- ITHACA-European Reference Network, Hospital La Paz, Madrid, Spain
| | - Sixto García-Miñaúr
- Instituto de Genética Médica y Molecular (INGEMM)-IdiPAZ, Hospital Universitario La Paz, Madrid, Spain
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain
- ITHACA-European Reference Network, Hospital La Paz, Madrid, Spain
| | - María Palomares-Bralo
- Instituto de Genética Médica y Molecular (INGEMM)-IdiPAZ, Hospital Universitario La Paz, Madrid, Spain
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain
- ITHACA-European Reference Network, Hospital La Paz, Madrid, Spain
| | - Elena Vallespín
- Instituto de Genética Médica y Molecular (INGEMM)-IdiPAZ, Hospital Universitario La Paz, Madrid, Spain
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain
- ITHACA-European Reference Network, Hospital La Paz, Madrid, Spain
| | | | | | - Cristina Bel-Fenellós
- Departamento de Investigación y Psicología en Educación, Facultad de Educación, UCM, Madrid, Spain
- CEE Estudio-3, Afanias, Madrid, Spain
| | - María Ángeles Mori
- Instituto de Genética Médica y Molecular (INGEMM)-IdiPAZ, Hospital Universitario La Paz, Madrid, Spain
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain
- ITHACA-European Reference Network, Hospital La Paz, Madrid, Spain
| | | | | | - Pilar Barrúz
- Instituto de Genética Médica y Molecular (INGEMM)-IdiPAZ, Hospital Universitario La Paz, Madrid, Spain
| | - Fernando Santos-Simarro
- Instituto de Genética Médica y Molecular (INGEMM)-IdiPAZ, Hospital Universitario La Paz, Madrid, Spain
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain
- ITHACA-European Reference Network, Hospital La Paz, Madrid, Spain
| | | | | | | | - Jair Antonio Tenorio
- Instituto de Genética Médica y Molecular (INGEMM)-IdiPAZ, Hospital Universitario La Paz, Madrid, Spain
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain
- ITHACA-European Reference Network, Hospital La Paz, Madrid, Spain
| | - Enrique Galán
- Hospital Materno-Infantil Infanta Cristina, Badajoz, Spain
| | | | | | - César Saleme
- Maternity Nuestra Señora de la Merced, Tucumán, Argentina
| | - Silvia Castillo
- Sección Genética, Hospital Clínico Universidad de Chile, Santiago, Chile
- Clínica Alemana, Santiago, Chile
| | | | - Luis Pérez-Jurado
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain
- Servicio de Genética, Instituto de Investigaciones Médicas Hospital del Mar (IMIM)/Universitat Pompeu Fabra, Barcelona, Spain
| | - Ana Barcia
- Hospital Universitario Virgen del Rocío, Sevilla, Spain
| | - Maria Soledad Martín
- Instituto de Genética Médica y Molecular (INGEMM)-IdiPAZ, Hospital Universitario La Paz, Madrid, Spain
| | - Elena Mansilla
- Instituto de Genética Médica y Molecular (INGEMM)-IdiPAZ, Hospital Universitario La Paz, Madrid, Spain
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain
- ITHACA-European Reference Network, Hospital La Paz, Madrid, Spain
| | - Isabel Vallcorba
- Instituto de Genética Médica y Molecular (INGEMM)-IdiPAZ, Hospital Universitario La Paz, Madrid, Spain
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain
- ITHACA-European Reference Network, Hospital La Paz, Madrid, Spain
| | | | | | | | - Raquel Blanco-Lago
- Servicio de Neuropediatría, Hospital Universitario Central de Asturias, Oviedo (Asturias), Spain
| | - Mercedes Serrano
- Unidad de Neuropediatría, Hospital San Joan de Deu, Barcelona, Spain
| | | | | | | | - Pilar Tirado
- Servicio de Neuropediatría, Hospital Universitario La Paz, Madrid, Spain
| | - Pablo Lapunzina
- Instituto de Genética Médica y Molecular (INGEMM)-IdiPAZ, Hospital Universitario La Paz, Madrid, Spain
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain
- ITHACA-European Reference Network, Hospital La Paz, Madrid, Spain
| |
Collapse
|
26
|
Kolevzon A, Breen MS, Siper PM, Halpern D, Frank Y, Rieger H, Weismann J, Trelles MP, Lerman B, Rapaport R, Buxbaum JD. Clinical trial of insulin-like growth factor-1 in Phelan-McDermid syndrome. Mol Autism 2022; 13:17. [PMID: 35395866 PMCID: PMC8994375 DOI: 10.1186/s13229-022-00493-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 03/01/2022] [Indexed: 11/24/2022] Open
Abstract
Background Phelan-McDermid syndrome (PMS) is caused by haploinsufficiency of the SHANK3 gene and is characterized by global developmental delays and autism spectrum disorder (ASD). Based on several converging lines of preclinical and clinical evidence supporting the use of insulin-like growth factor-1 (IGF-1) in PMS, this study aims to follow-up a previous pilot study with IGF-1 to further evaluate this novel therapeutic for core symptoms of ASD in children with PMS. Methods Ten children aged 5–9 with PMS were enrolled. Participants were randomized to receive IGF-1 or placebo (saline) using a 12-week, double-blind, crossover design. Efficacy was assessed using the primary outcome of the Aberrant Behavior Checklist—Social Withdrawal (ABC-SW) subscale as well as secondary outcome measures reflecting core symptoms of ASD. To increase power and sample size, we jointly analyzed the effect of IGF-1 reported here together with results from our previous controlled trail of IGF-1 in children with PMS (combined N = 19). Results Results on the ABC-SW did not reach statistical significance, however significant improvements in sensory reactivity symptoms were observed. In our pooled analyses, IGF-1 treatment also led to significant improvements in repetitive behaviors and hyperactivity. There were no other statistically significant effects seen across other clinical outcome measures. IGF-1 was well tolerated and there were no serious adverse events. Limitations The small sample size and expectancy bias due to relying on parent reported outcome measures may contribute to limitations in interpreting results. Conclusion IGF-1 is efficacious in improving sensory reactivity symptoms, repetitive behaviors, and hyperactivity in children with PMS. Trial registration NCT01525901. Supplementary Information The online version contains supplementary material available at 10.1186/s13229-022-00493-7.
Collapse
Affiliation(s)
- A Kolevzon
- Seaver Autism Center for Research and Treatment, New York, NY, USA. .,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA. .,Mindich Child Health Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA. .,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA. .,Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA. .,Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1230, New York, NY, 10029, USA.
| | - M S Breen
- Seaver Autism Center for Research and Treatment, New York, NY, USA.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Mindich Child Health Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1230, New York, NY, 10029, USA
| | - P M Siper
- Seaver Autism Center for Research and Treatment, New York, NY, USA.,Mindich Child Health Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1230, New York, NY, 10029, USA
| | - D Halpern
- Seaver Autism Center for Research and Treatment, New York, NY, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1230, New York, NY, 10029, USA
| | - Y Frank
- Seaver Autism Center for Research and Treatment, New York, NY, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1230, New York, NY, 10029, USA
| | - H Rieger
- Seaver Autism Center for Research and Treatment, New York, NY, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1230, New York, NY, 10029, USA
| | - J Weismann
- Seaver Autism Center for Research and Treatment, New York, NY, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1230, New York, NY, 10029, USA
| | - M P Trelles
- Seaver Autism Center for Research and Treatment, New York, NY, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1230, New York, NY, 10029, USA
| | - B Lerman
- Seaver Autism Center for Research and Treatment, New York, NY, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1230, New York, NY, 10029, USA
| | - R Rapaport
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Endocrinology and Diabetes, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1230, New York, NY, 10029, USA
| | - J D Buxbaum
- Seaver Autism Center for Research and Treatment, New York, NY, USA.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Mindich Child Health Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1230, New York, NY, 10029, USA
| |
Collapse
|
27
|
Mapelli L, Soda T, D’Angelo E, Prestori F. The Cerebellar Involvement in Autism Spectrum Disorders: From the Social Brain to Mouse Models. Int J Mol Sci 2022; 23:ijms23073894. [PMID: 35409253 PMCID: PMC8998980 DOI: 10.3390/ijms23073894] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 02/04/2023] Open
Abstract
Autism spectrum disorders (ASD) are pervasive neurodevelopmental disorders that include a variety of forms and clinical phenotypes. This heterogeneity complicates the clinical and experimental approaches to ASD etiology and pathophysiology. To date, a unifying theory of these diseases is still missing. Nevertheless, the intense work of researchers and clinicians in the last decades has identified some ASD hallmarks and the primary brain areas involved. Not surprisingly, the areas that are part of the so-called “social brain”, and those strictly connected to them, were found to be crucial, such as the prefrontal cortex, amygdala, hippocampus, limbic system, and dopaminergic pathways. With the recent acknowledgment of the cerebellar contribution to cognitive functions and the social brain, its involvement in ASD has become unmistakable, though its extent is still to be elucidated. In most cases, significant advances were made possible by recent technological developments in structural/functional assessment of the human brain and by using mouse models of ASD. Mouse models are an invaluable tool to get insights into the molecular and cellular counterparts of the disease, acting on the specific genetic background generating ASD-like phenotype. Given the multifaceted nature of ASD and related studies, it is often difficult to navigate the literature and limit the huge content to specific questions. This review fulfills the need for an organized, clear, and state-of-the-art perspective on cerebellar involvement in ASD, from its connections to the social brain areas (which are the primary sites of ASD impairments) to the use of monogenic mouse models.
Collapse
Affiliation(s)
- Lisa Mapelli
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy; (T.S.); (E.D.)
- Correspondence: (L.M.); (F.P.)
| | - Teresa Soda
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy; (T.S.); (E.D.)
| | - Egidio D’Angelo
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy; (T.S.); (E.D.)
- Brain Connectivity Center, IRCCS Mondino Foundation, 27100 Pavia, Italy
| | - Francesca Prestori
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy; (T.S.); (E.D.)
- Correspondence: (L.M.); (F.P.)
| |
Collapse
|
28
|
Gao L, Zhao J, Ardiel EL, Hall Q, Nurrish S, Kaplan JM. Shank promotes action potential repolarization by recruiting BK channels to calcium microdomains. eLife 2022; 11:75140. [PMID: 35266450 PMCID: PMC8937234 DOI: 10.7554/elife.75140] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 03/09/2022] [Indexed: 11/13/2022] Open
Abstract
Mutations altering the scaffolding protein Shank are linked to several psychiatric disorders, and to synaptic and behavioral defects in mice. Among its many binding partners, Shank directly binds CaV1 voltage activated calcium channels. Here we show that the C. elegans SHN-1/Shank promotes CaV1 coupling to calcium activated potassium channels. Mutations inactivating SHN-1, and those preventing SHN-1 binding to EGL-19/CaV1 all increase action potential durations in body muscles. Action potential repolarization is mediated by two classes of potassium channels: SHK-1/KCNA and SLO-1 and SLO-2 BK channels. BK channels are calcium-dependent, and their activation requires tight coupling to EGL-19/CaV1 channels. SHN-1's effects on AP duration are mediated by changes in BK channels. In shn-1 mutants, SLO-2 currents and channel clustering are significantly decreased in both body muscles and neurons. Finally, increased and decreased shn-1 gene copy number produce similar changes in AP width and SLO-2 current. Collectively, these results suggest that an important function of Shank is to promote microdomain coupling of BK with CaV1.
Collapse
Affiliation(s)
- Luna Gao
- Department of Molecular Biology, Massachusetts General Hospital, Boston, United States
| | - Jian Zhao
- Department of Molecular Biology, Massachusetts General Hospital, Boston, United States
| | - Evan L Ardiel
- Department of Molecular Biology, Massachusetts General Hospital, Boston, United States
| | - Qi Hall
- Department of Molecular Biology, Massachusetts General Hospital, Boston, United States
| | - Stephen Nurrish
- Department of Molecular Biology, Massachusetts General Hospital, Boston, United States
| | - Joshua M Kaplan
- Department of Molecular Biology, Massachusetts General Hospital, Boston, United States
| |
Collapse
|
29
|
Rysstad AL, Kildahl AN, Skavhaug JO, Dønnum MS, Helverschou SB. Case study: organizing outpatient pharmacological treatment of bipolar disorder in autism, intellectual disability and Phelan-McDermid syndrome (22q13.3 deletion syndrome). INTERNATIONAL JOURNAL OF DEVELOPMENTAL DISABILITIES 2022; 68:378-387. [PMID: 35603006 PMCID: PMC9122368 DOI: 10.1080/20473869.2020.1756113] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Phelan-McDermid syndrome (PHMDS)/22q13.3 deletion syndrome is a rare genetic disorder associated with autism spectrum disorder (ASD), intellectual disability (ID), and bipolar disorder. While numerous cases have been reported describing successful pharmacological treatment of bipolar disorder in PHMDS, there is currently little guidance available on how to organize and execute such treatment. The aim of the current case study was to explore how pharmacological treatment of bipolar disorder in PHMDS may be organized and evaluated in an outpatient setting. Through a complex process of try and fail, including systematic evaluation of any change to the intervention and never implementing more than one change at the time, the patient gradually improved, regaining his communicative and adaptive skills. Four years passed from referral to this result was achieved. Organizing assessment and treatment as a collaborative effort involving specialized mental health professionals, professional caregivers and the patient's family proved feasible. Many of the challenges present in assessment of psychiatric disorder in individuals with ASD and ID are likely to be present also in evaluation of treatment effects, particularly in disorders where symptoms occur in phases. The approach described in the current paper may contribute to reducing the impact of these challenges.
Collapse
Affiliation(s)
- Anne Langseth Rysstad
- Section for Intellectual Disabilities and Autism, Vestre Viken Hospital Trust, Asker, Norway
- Correspondence to: Anne Langseth Rysstad Section for Intellectual Disabilities and Autism, Seksjon utviklingshemming og autisme – Blakstad psyk. avd., Vestre Viken HF, Postboks 800, Asker, 3004Drammen.
| | - Arvid Nikolai Kildahl
- Regional Section Mental Health, Intellectual Disabilities/Autism, Oslo University Hospital, Oslo, Norway
- NevSom Norwegian Centre of Expertise for Neurodevelopmental Disorders and Hypersomnias, Oslo University Hospital, Oslo, Norway
| | - Jon Olav Skavhaug
- Section for Intellectual Disabilities and Autism, Vestre Viken Hospital Trust, Asker, Norway
| | - Monica Stolen Dønnum
- Section for Intellectual Disabilities and Autism, Vestre Viken Hospital Trust, Asker, Norway
| | - Sissel Berge Helverschou
- NevSom Norwegian Centre of Expertise for Neurodevelopmental Disorders and Hypersomnias, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
30
|
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: 4] [Impact Index Per Article: 1.3] [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
|
31
|
Postsynaptic autism spectrum disorder genes and synaptic dysfunction. Neurobiol Dis 2021; 162:105564. [PMID: 34838666 DOI: 10.1016/j.nbd.2021.105564] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 11/17/2021] [Accepted: 11/23/2021] [Indexed: 12/20/2022] Open
Abstract
This review provides an overview of the synaptic dysfunction of neuronal circuits and the ensuing behavioral alterations caused by mutations in autism spectrum disorder (ASD)-linked genes directly or indirectly affecting the postsynaptic neuronal compartment. There are plenty of ASD risk genes, that may be broadly grouped into those involved in gene expression regulation (epigenetic regulation and transcription) and genes regulating synaptic activity (neural communication and neurotransmission). Notably, the effects mediated by ASD-associated genes can vary extensively depending on the developmental time and/or subcellular site of expression. Therefore, in order to gain a better understanding of the mechanisms of disruptions in postsynaptic function, an effort to better model ASD in experimental animals is required to improve standardization and increase reproducibility within and among studies. Such an effort holds promise to provide deeper insight into the development of these disorders and to improve the translational value of preclinical studies.
Collapse
|
32
|
Pandey S, Hajikazemi M, Zacheja T, Schalbetter S, Baxter J, Guryev V, Hofmann A, Heermann DW, Juranek SA, Paeschke K. Telomerase subunit Est2 marks internal sites that are prone to accumulate DNA damage. BMC Biol 2021; 19:247. [PMID: 34801008 PMCID: PMC8605574 DOI: 10.1186/s12915-021-01167-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 10/15/2021] [Indexed: 12/14/2022] Open
Abstract
Background The main function of telomerase is at the telomeres but under adverse conditions telomerase can bind to internal regions causing deleterious effects as observed in cancer cells. Results By mapping the global occupancy of the catalytic subunit of telomerase (Est2) in the budding yeast Saccharomyces cerevisiae, we reveal that it binds to multiple guanine-rich genomic loci, which we termed “non-telomeric binding sites” (NTBS). We characterize Est2 binding to NTBS. Contrary to telomeres, Est2 binds to NTBS in G1 and G2 phase independently of Est1 and Est3. The absence of Est1 and Est3 renders telomerase inactive at NTBS. However, upon global DNA damage, Est1 and Est3 join Est2 at NTBS and telomere addition can be observed indicating that Est2 occupancy marks NTBS regions as particular risks for genome stability. Conclusions Our results provide a novel model of telomerase regulation in the cell cycle using internal regions as “parking spots” of Est2 but marking them as hotspots for telomere addition. Supplementary Information The online version contains supplementary material available at 10.1186/s12915-021-01167-1.
Collapse
Affiliation(s)
- Satyaprakash Pandey
- University of Groningen, University Medical Center Groningen, European Research Institute for the Biology of Ageing, 9713 AV, Groningen, Netherlands
| | - Mona Hajikazemi
- Clinic of Internal Medicine III, Oncology, Hematology, Rheumatology and Clinical Immunology, University Hospital Bonn, Bonn, Germany
| | - Theresa Zacheja
- Clinic of Internal Medicine III, Oncology, Hematology, Rheumatology and Clinical Immunology, University Hospital Bonn, Bonn, Germany
| | | | - Jonathan Baxter
- Department of Life Science, University of Sussex, Brighton, UK
| | - Victor Guryev
- University of Groningen, University Medical Center Groningen, European Research Institute for the Biology of Ageing, 9713 AV, Groningen, Netherlands
| | - Andreas Hofmann
- Institute for Theoretical Physics, University of Heidelberg, Philosophenweg 12, 69120, Heidelberg, Germany
| | - Dieter W Heermann
- Institute for Theoretical Physics, University of Heidelberg, Philosophenweg 12, 69120, Heidelberg, Germany
| | - Stefan A Juranek
- Clinic of Internal Medicine III, Oncology, Hematology, Rheumatology and Clinical Immunology, University Hospital Bonn, Bonn, Germany.
| | - Katrin Paeschke
- University of Groningen, University Medical Center Groningen, European Research Institute for the Biology of Ageing, 9713 AV, Groningen, Netherlands. .,Clinic of Internal Medicine III, Oncology, Hematology, Rheumatology and Clinical Immunology, University Hospital Bonn, Bonn, Germany.
| |
Collapse
|
33
|
Li D, Liu C, Huang Z, Li H, Xu Q, Zhou B, Hu C, Zhang Y, Wang Y, Nie J, Qiao Z, Yin D, Xu X. Common and Distinct Disruptions of Cortical Surface Morphology Between Autism Spectrum Disorder Children With and Without SHANK3 Deficiency. Front Neurosci 2021; 15:751364. [PMID: 34776852 PMCID: PMC8581670 DOI: 10.3389/fnins.2021.751364] [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: 08/01/2021] [Accepted: 09/29/2021] [Indexed: 11/13/2022] Open
Abstract
SH3 and Multiple Ankyrin Repeat Domains 3 (SHANK3)-caused autism spectrum disorder (ASD) may present a unique opportunity to clarify the heterogeneous neuropathological mechanisms of ASD. However, the specificity and commonality of disrupted large-scale brain organization in SHANK3-deficient children remain largely unknown. The present study combined genetic tests, neurobehavioral evaluations, and magnetic resonance imaging, aiming to explore the disruptions of both local and networked cortical structural organization in ASD children with and without SHANK3 deficiency. Multiple surface morphological parameters such as cortical thickness (CT) and sulcus depth were estimated, and the graph theory was adopted to characterize the topological properties of structural covariance networks (SCNs). Finally, a correlation analysis between the alterations in brain morphological features and the neurobehavioral evaluations was performed. Compared with typically developed children, increased CT and reduced nodal degree were found in both ASD children with and without SHANK3 defects mainly in the lateral temporal cortex, prefrontal cortex (PFC), temporo-parietal junction (TPJ), superior temporal gyrus (STG), and limbic/paralimbic regions. Besides commonality, our findings showed some distinct abnormalities in ASD children with SHANK3 defects compared to those without. Locally, more changes in the STG and orbitofrontal cortex were exhibited in ASD children with SHANK3 defects, while more changes in the TPJ and inferior parietal lobe (IPL) in those without SHANK3 defects were observed. For the SCNs, a trend toward regular network topology was observed in ASD children with SHANK3 defects, but not in those without. In addition, ASD children with SHANK3 defects showed more alterations of nodal degrees in the anterior and posterior cingulate cortices and right insular, while there were more disruptions in the sensorimotor areas and the left insular and dorsomedial PFC in ASD without SHANK3 defects. Our findings indicate dissociable disruptions of local and networked brain morphological features in ASD children with and without SHANK3 deficiency. Moreover, this monogenic study may provide a valuable path for parsing the heterogeneity of brain disturbances in ASD.
Collapse
Affiliation(s)
- Dongyun Li
- Department of Child Health Care, Children's Hospital of Fudan University, Shanghai, China
| | - Chunxue Liu
- Department of Child Health Care, Children's Hospital of Fudan University, Shanghai, China
| | - Ziyi Huang
- Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education), School of Psychology and Cognitive Science, Affiliated Mental Health Center, East China Normal University, Shanghai, China.,School of Psychology, South China Normal University, Guangzhou, China
| | - Huiping Li
- Department of Child Health Care, Children's Hospital of Fudan University, Shanghai, China
| | - Qiong Xu
- Department of Child Health Care, Children's Hospital of Fudan University, Shanghai, China
| | - Bingrui Zhou
- Department of Child Health Care, Children's Hospital of Fudan University, Shanghai, China
| | - Chunchun Hu
- Department of Child Health Care, Children's Hospital of Fudan University, Shanghai, China
| | - Ying Zhang
- Department of Child Health Care, Children's Hospital of Fudan University, Shanghai, China
| | - Yi Wang
- Department of Child Health Care, Children's Hospital of Fudan University, Shanghai, China
| | - Jingxin Nie
- School of Psychology, South China Normal University, Guangzhou, China
| | - Zhongwei Qiao
- Department of Radiology, Children's Hospital of Fudan University, Shanghai, China
| | - Dazhi Yin
- Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education), School of Psychology and Cognitive Science, Affiliated Mental Health Center, East China Normal University, Shanghai, China
| | - Xiu Xu
- Department of Child Health Care, Children's Hospital of Fudan University, Shanghai, China
| |
Collapse
|
34
|
Crocco M, Panciroli M, Milanaccio C, Morerio C, Verrico A, Garrè ML, Di Iorgi N, Capra V. Case Report: The Emerging Role of Ring Chromosome 22 in Phelan-McDermid Syndrome With Atypical Teratoid/Rhabdoid Tumor: The First Child Treated With Growth Hormone. Front Neurol 2021; 12:741062. [PMID: 34777208 PMCID: PMC8585933 DOI: 10.3389/fneur.2021.741062] [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: 07/14/2021] [Accepted: 10/04/2021] [Indexed: 01/05/2023] Open
Abstract
Atypical teratoid/rhabdoid tumors (AT/RTs) in the rhabdoid tumor predisposition syndromes are most often caused by germline mutations of the SMARCB1 gene located in chromosome 22q11.2. Although rarely, it can also result from the constitutional ring chromosome 22 (r22): during mitosis the ring chromosome may lead to an increased rate of somatic mutations, resulting in rhabdoid tumor predispositions when the tumor-suppressor gene SMARCB1 is involved. Individuals with r22 may present similar features as those with Phelan-McDermid syndrome (PMDS) due to 22q13.3 deletion, including the SHANK3 gene. Despite several reports on AT/RT in children with r22 and/or PMDS have been published, the role of constitutional r22 as new oncogenic mechanism for AT/RT is still under investigation. There is not a lot of data available on therapeutic and prognostic implications of r22 in AT/RT and PMDS. Herein, we present the first case of a child with constitutional r22, PMDS and AT/RT of the brain, who is a long term survivor and is been treated with growth hormone. We also describe an unexpected adverse reaction to midazolam.
Collapse
Affiliation(s)
- Marco Crocco
- Neuro-Oncology Unit, Istituto di Ricovero e Cura a Carattere Scientifico Giannina Gaslini Institute, Genoa, Italy.,Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Child and Maternal Health, University of Genova, Genoa, Italy
| | - Marta Panciroli
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Child and Maternal Health, University of Genova, Genoa, Italy
| | - Claudia Milanaccio
- Neuro-Oncology Unit, Istituto di Ricovero e Cura a Carattere Scientifico Giannina Gaslini Institute, Genoa, Italy
| | - Cristina Morerio
- Laboratory of Human Genetics, Istituto di Ricovero e Cura a Carattere Scientifico Giannina Gaslini Institute, Genoa, Italy
| | - Antonio Verrico
- Neuro-Oncology Unit, Istituto di Ricovero e Cura a Carattere Scientifico Giannina Gaslini Institute, Genoa, Italy
| | - Maria Luisa Garrè
- Neuro-Oncology Unit, Istituto di Ricovero e Cura a Carattere Scientifico Giannina Gaslini Institute, Genoa, Italy
| | - Natascia Di Iorgi
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Child and Maternal Health, University of Genova, Genoa, Italy.,Department of Pediatrics, Istituto di Ricovero e Cura a Carattere Scientifico Giannina Gaslini Institute, Genoa, Italy
| | - Valeria Capra
- Medical Genetics Unit, Istituto di Ricovero e Cura a Carattere Scientifico Giannina Gaslini Institute, Genoa, Italy
| |
Collapse
|
35
|
Delling JP, Boeckers TM. Comparison of SHANK3 deficiency in animal models: phenotypes, treatment strategies, and translational implications. J Neurodev Disord 2021; 13:55. [PMID: 34784886 PMCID: PMC8594088 DOI: 10.1186/s11689-021-09397-8] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 09/27/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Autism spectrum disorder (ASD) is a neurodevelopmental condition, which is characterized by clinical heterogeneity and high heritability. Core symptoms of ASD include deficits in social communication and interaction, as well as restricted, repetitive patterns of behavior, interests, or activities. Many genes have been identified that are associated with an increased risk for ASD. Proteins encoded by these ASD risk genes are often involved in processes related to fetal brain development, chromatin modification and regulation of gene expression in general, as well as the structural and functional integrity of synapses. Genes of the SH3 and multiple ankyrin repeat domains (SHANK) family encode crucial scaffolding proteins (SHANK1-3) of excitatory synapses and other macromolecular complexes. SHANK gene mutations are highly associated with ASD and more specifically the Phelan-McDermid syndrome (PMDS), which is caused by heterozygous 22q13.3-deletion resulting in SHANK3-haploinsufficiency, or by SHANK3 missense variants. SHANK3 deficiency and potential treatment options have been extensively studied in animal models, especially in mice, but also in rats and non-human primates. However, few of the proposed therapeutic strategies have translated into clinical practice yet. MAIN TEXT This review summarizes the literature concerning SHANK3-deficient animal models. In particular, the structural, behavioral, and neurological abnormalities are described and compared, providing a broad and comprehensive overview. Additionally, the underlying pathophysiologies and possible treatments that have been investigated in these models are discussed and evaluated with respect to their effect on ASD- or PMDS-associated phenotypes. CONCLUSIONS Animal models of SHANK3 deficiency generated by various genetic strategies, which determine the composition of the residual SHANK3-isoforms and affected cell types, show phenotypes resembling ASD and PMDS. The phenotypic heterogeneity across multiple models and studies resembles the variation of clinical severity in human ASD and PMDS patients. Multiple therapeutic strategies have been proposed and tested in animal models, which might lead to translational implications for human patients with ASD and/or PMDS. Future studies should explore the effects of new therapeutic approaches that target genetic haploinsufficiency, like CRISPR-mediated activation of promotors.
Collapse
Affiliation(s)
- Jan Philipp Delling
- Institute for Anatomy and Cell Biology, Ulm University, Albert-Einstein-Allee 11, Ulm, 89081, Germany.
| | - Tobias M Boeckers
- Institute for Anatomy and Cell Biology, Ulm University, Albert-Einstein-Allee 11, Ulm, 89081, Germany. .,Ulm Site, DZNE, Ulm, Germany.
| |
Collapse
|
36
|
Jain L, Oberman LM, Beamer L, Cascio L, May M, Srikanth S, Skinner C, Jones K, Allen B, Rogers C, Phelan K, Kaufmann WE, DuPont B, Sarasua SM, Boccuto L. Genetic and metabolic profiling of individuals with Phelan-McDermid syndrome presenting with seizures. Clin Genet 2021; 101:87-100. [PMID: 34664257 DOI: 10.1111/cge.14074] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 09/23/2021] [Accepted: 10/13/2021] [Indexed: 12/25/2022]
Abstract
Phelan-McDermid syndrome (PMS) (OMIM*606232) is a rare genetic disorder characterized by intellectual disability, autistic features, speech delay, minor dysmorphia, and seizures. This study was conducted to investigate the prevalence of seizures and the association with genetic and metabolic features since there has been little research related to seizures in PMS. For 57 individuals, seizure data was collected from caregiver interviews, genetic data from existing cytogenetic records and Sanger sequencing for nine 22q13 genes, and metabolic profiling from the Phenotype Mammalian MicroArray (PM-M) developed by Biolog. Results showed that 46% of individuals had seizures with the most common type being absence and grand-mal seizures. Seizures were most prevalent in individuals with pathogenic SHANK3 mutations (70%), those with deletion sizes >4 Mb (16%), and those with deletion sizes <4 Mb (71%) suggesting involvement of genes in addition to SHANK3. Additionally, a 3 Mb genomic region on 22q13.31 containing the gene TBC1D22A, was found to be significantly associated with seizure prevalence. A distinct metabolic profile was identified for individuals with PMS with seizures and suggested among other features a disrupted utilization of main energy sources using Biolog plates. The results of this study will be helpful for clinicians and families in anticipating seizures in these children and for researchers to identify candidate genes for the seizure phenotype.
Collapse
Affiliation(s)
- Lavanya Jain
- Greenwood Genetic Center, Greenwood, South Carolina, USA.,School of Nursing, Healthcare Genetics Program, Clemson University, Clemson, South Carolina, USA
| | - Lindsay M Oberman
- Henry M Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA.,Center for Neuroscience and Regenerative Medicine, Bethesda, Maryland, USA
| | - Laura Beamer
- Greenwood Genetic Center, Greenwood, South Carolina, USA
| | - Lauren Cascio
- Greenwood Genetic Center, Greenwood, South Carolina, USA
| | - Melanie May
- Greenwood Genetic Center, Greenwood, South Carolina, USA
| | | | - Cindy Skinner
- Greenwood Genetic Center, Greenwood, South Carolina, USA
| | - Kelly Jones
- Greenwood Genetic Center, Greenwood, South Carolina, USA
| | - Bridgette Allen
- School of Nursing, Healthcare Genetics Program, Clemson University, Clemson, South Carolina, USA
| | - Curtis Rogers
- Greenwood Genetic Center, Greenwood, South Carolina, USA
| | - Katy Phelan
- Genetics Laboratory, Florida Cancer Specialists and Research Institute, Fort Myers, Florida, USA
| | - Walter E Kaufmann
- Greenwood Genetic Center, Greenwood, South Carolina, USA.,Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia, USA.,Department of Neurology, Boston Children's Hospital, Boston, Massachusetts, USA.,Anavex Life Sciences Corp, New York, New York, USA
| | - Barbara DuPont
- Greenwood Genetic Center, Greenwood, South Carolina, USA
| | - Sara M Sarasua
- School of Nursing, Healthcare Genetics Program, Clemson University, Clemson, South Carolina, USA
| | - Luigi Boccuto
- Greenwood Genetic Center, Greenwood, South Carolina, USA.,School of Nursing, Healthcare Genetics Program, Clemson University, Clemson, South Carolina, USA.,Clemson University School of Health Research, Clemson, South Carolina, USA
| |
Collapse
|
37
|
Fastman J, Foss-Feig J, Frank Y, Halpern D, Harony-Nicolas H, Layton C, Sandin S, Siper P, Tang L, Trelles P, Zweifach J, Buxbaum JD, Kolevzon A. A randomized controlled trial of intranasal oxytocin in Phelan-McDermid syndrome. Mol Autism 2021; 12:62. [PMID: 34593045 PMCID: PMC8482590 DOI: 10.1186/s13229-021-00459-1] [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: 02/22/2021] [Accepted: 07/19/2021] [Indexed: 12/20/2022] Open
Abstract
Background Phelan-McDermid syndrome (PMS) is a rare neurodevelopmental disorder caused by haploinsufficiency of the SHANK3 gene and characterized by global developmental delays, deficits in speech and motor function, and autism spectrum disorder (ASD). Monogenic causes of ASD such as PMS are well suited to investigations with novel therapeutics, as interventions can be targeted based on established genetic etiology. While preclinical studies have demonstrated that the neuropeptide oxytocin can reverse electrophysiological, attentional, and social recognition memory deficits in Shank3-deficient rats, there have been no trials in individuals with PMS. The purpose of this study is to assess the efficacy and safety of intranasal oxytocin as a treatment for the core symptoms of ASD in a cohort of children with PMS. Methods Eighteen children aged 5–17 with PMS were enrolled. Participants were randomized to receive intranasal oxytocin or placebo (intranasal saline) and underwent treatment during a 12-week double-blind, parallel group phase, followed by a 12-week open-label extension phase during which all participants received oxytocin. Efficacy was assessed using the primary outcome of the Aberrant Behavior Checklist-Social Withdrawal (ABC-SW) subscale as well as a number of secondary outcome measures related to the core symptoms of ASD. Safety was monitored throughout the study period. Results There was no statistically significant improvement with oxytocin as compared to placebo on the ABC-SW (Mann–Whitney U = 50, p = 0.055), or on any secondary outcome measures, during either the double-blind or open-label phases. Oxytocin was generally well tolerated, and there were no serious adverse events.
Limitations The small sample size, potential challenges with drug administration, and expectancy bias due to relying on parent reported outcome measures may all contribute to limitations in interpreting results. Conclusion Our results suggest that intranasal oxytocin is not efficacious in improving the core symptoms of ASD in children with PMS. Trial registration NCT02710084. Supplementary Information The online version contains supplementary material available at 10.1186/s13229-021-00459-1.
Collapse
Affiliation(s)
- J Fastman
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - J Foss-Feig
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1230, New York, NY, 10029, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Y Frank
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1230, New York, NY, 10029, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - D Halpern
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - H Harony-Nicolas
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - C Layton
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1230, New York, NY, 10029, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - S Sandin
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1230, New York, NY, 10029, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - P Siper
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1230, New York, NY, 10029, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - L Tang
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1230, New York, NY, 10029, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - P Trelles
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1230, New York, NY, 10029, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - J Zweifach
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1230, New York, NY, 10029, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - J D Buxbaum
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1230, New York, NY, 10029, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - A Kolevzon
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1230, New York, NY, 10029, USA. .,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA. .,Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
38
|
Liu C, Wang Y, Deng J, Lin J, Hu C, Li Q, Xu X. Social Deficits and Repetitive Behaviors Are Improved by Early Postnatal Low-Dose VPA Intervention in a Novel shank3-Deficient Zebrafish Model. Front Neurosci 2021; 15:682054. [PMID: 34566559 PMCID: PMC8462462 DOI: 10.3389/fnins.2021.682054] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 08/11/2021] [Indexed: 12/27/2022] Open
Abstract
Mutations of the SHANK3 gene are found in some autism spectrum disorder (ASD) patients, and animal models harboring SHANK3 mutations exhibit a variety of ASD-like behaviors, presenting a unique opportunity to explore the underlying neuropathological mechanisms and potential pharmacological treatments. The histone deacetylase (HDAC) valproic acid (VPA) has demonstrated neuroprotective and neuroregenerative properties, suggesting possible therapeutic utility for ASD. Therefore, SHANK3-associated ASD-like symptoms present a convenient model to evaluate the potential benefits, therapeutic window, and optimal dose of VPA. We constructed a novel shank3-deficient (shank3ab–/–) zebrafish model through CRISPR/Cas9 editing and conducted comprehensive morphological and neurobehavioral evaluations, including of core ASD-like behaviors, as well as molecular analyses of synaptic proteins expression levels. Furthermore, different VPA doses and treatment durations were examined for effects on ASD-like phenotypes. Compared to wild types (WTs), shank3ab–/– zebrafish exhibited greater developmental mortality, more frequent abnormal tail bending, pervasive developmental delay, impaired social preference, repetitive swimming behaviors, and generally reduced locomotor activity. The expression levels of synaptic proteins were also dramatically reduced in shank3ab–/– zebrafish. These ASD-like behaviors were attenuated by low-dose (5 μM) VPA administered from 4 to 8 days post-fertilization (dpf), and the effects persisted to adulthood. In addition, the observed underexpression of grm5, encoding glutamate metabotropic receptor 5, was significantly improved in VPA-treated shank3ab–/– zebrafish. We report for the first time that low-dose VPA administered after neural tube closure has lasting beneficial effects on the social deficits and repetitive behavioral patterns in shank3-deficient ASD model zebrafish. These findings provide a promising strategy for ASD clinical drug development.
Collapse
Affiliation(s)
- Chunxue Liu
- Department of Child Health Care, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Yi Wang
- Department of Child Health Care, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Jingxin Deng
- Department of Child Health Care, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Jia Lin
- Center for Translational Medicine, Institute of Pediatrics, Shanghai Key Laboratory of Birth Defects Prevention and Control, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Chunchun Hu
- Department of Child Health Care, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Qiang Li
- Center for Translational Medicine, Institute of Pediatrics, Shanghai Key Laboratory of Birth Defects Prevention and Control, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Xiu Xu
- Department of Child Health Care, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| |
Collapse
|
39
|
Wan L, Ai JQ, Yang C, Jiang J, Zhang QL, Luo ZH, Huang RJ, Tu T, Pan A, Tu E, Manavis J, Xiao B, Yan XX. Expression of the Excitatory Postsynaptic Scaffolding Protein, Shank3, in Human Brain: Effect of Age and Alzheimer's Disease. Front Aging Neurosci 2021; 13:717263. [PMID: 34504419 PMCID: PMC8421777 DOI: 10.3389/fnagi.2021.717263] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 07/26/2021] [Indexed: 12/14/2022] Open
Abstract
Shank3 is a postsynaptic scaffolding protein of excitatory synapses. Mutations or variations of SHANK3 are associated with various psychiatric and neurological disorders. We set to determine its normal expression pattern in the human brain, and its change, if any, with age and Alzheimer’s disease (AD)-type β-amyloid (Aβ) and Tau pathogenesis. In general, Shank3 immunoreactivity (IR) exhibited largely a neuropil pattern with differential laminar/regional distribution across brain regions. In youth and adults, subsets of pyramidal/multipolar neurons in the cerebrum, striatum, and thalamus showed moderate IR, while some large-sized neurons in the brainstem and the granule cells in the cerebellar cortex exhibited light IR. In double immunofluorescence, Shank3 IR occurred at the sublemmal regions in neuronal somata and large dendrites, apposing to synaptophysin-labeled presynaptic terminals. In aged cases, immunolabeled neuronal somata were reduced, with disrupted neuropil labeling seen in the molecular layer of the dentate gyrus in AD cases. In immunoblot, levels of Shank3 protein were positively correlated with that of the postsynaptic density protein 95 (PSD95) among different brain regions. Levels of Shank3, PSD95, and synaptophysin immunoblotted in the prefrontal, precentral, and cerebellar cortical lysates were reduced in the aged and AD relative to youth and adult groups. Taken together, the differential Shank3 expression among brain structures/regions indicates the varied local density of the excitatory synapses. The enriched Shank3 expression in the forebrain subregions appears inconsistent with a role of this protein in the modulation of high cognitive functions. The decline of its expression in aged and AD brains may relate to the degeneration of excitatory synapses.
Collapse
Affiliation(s)
- Lily Wan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Jia-Qi Ai
- Department of Anatomy and Neurobiology, Central South University Xiangya School of Medicine, Changsha, China
| | - Chen Yang
- Department of Anatomy and Neurobiology, Central South University Xiangya School of Medicine, Changsha, China
| | - Juan Jiang
- Department of Anatomy and Neurobiology, Central South University Xiangya School of Medicine, Changsha, China
| | - Qi-Lei Zhang
- Department of Anatomy and Neurobiology, Central South University Xiangya School of Medicine, Changsha, China
| | - Zhao-Hui Luo
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Rou-Jie Huang
- Medical Doctor Program, Xiangya School of Medicine, Central South University, Changsha, China
| | - Tian Tu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Aihua Pan
- Department of Anatomy and Neurobiology, Central South University Xiangya School of Medicine, Changsha, China
| | - Ewen Tu
- Department of Neurology, Brain Hospital of Hunan Province, Changsha, China
| | - Jim Manavis
- Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia
| | - Bo Xiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Xiao-Xin Yan
- Department of Anatomy and Neurobiology, Central South University Xiangya School of Medicine, Changsha, China
| |
Collapse
|
40
|
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: 15] [Impact Index Per Article: 3.8] [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
|
41
|
Dyar B, Meaddough E, Sarasua SM, Rogers C, Phelan K, Boccuto L. Genetic Findings as the Potential Basis of Personalized Pharmacotherapy in Phelan-McDermid Syndrome. Genes (Basel) 2021; 12:1192. [PMID: 34440366 PMCID: PMC8392667 DOI: 10.3390/genes12081192] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 07/26/2021] [Accepted: 07/29/2021] [Indexed: 12/13/2022] Open
Abstract
Phelan-McDermid syndrome (PMS) is a genetic disorder often characterized by autism or autistic-like behavior. Most cases are associated with haploinsufficiency of the SHANK3 gene resulting from deletion of the gene at 22q13.3 or from a pathogenic variant in the gene. Treatment of PMS often targets SHANK3, yet deletion size varies from <50 kb to >9 Mb, potentially encompassing dozens of genes and disrupting regulatory elements altering gene expression, inferring the potential for multiple therapeutic targets. Repurposed drugs have been used in clinical trials investigating therapies for PMS: insulin-like growth factor 1 (IGF-1) for its effect on social and aberrant behaviors, intranasal insulin for improvements in cognitive and social ability, and lithium for reversing regression and stabilizing behavior. The pharmacogenomics of PMS is complicated by the CYP2D6 enzyme which metabolizes antidepressants and antipsychotics often used for treatment. The gene coding for CYP2D6 maps to 22q13.2 and is lost in individuals with deletions larger than 8 Mb. Because PMS has diverse neurological and medical symptoms, many concurrent medications may be prescribed, increasing the risk for adverse drug reactions. At present, there is no single best treatment for PMS. Approaches to therapy are necessarily complex and must target variable behavioral and physical symptoms of PMS.
Collapse
Affiliation(s)
- Brianna Dyar
- Healthcare Genetics Program, School of Nursing, Clemson University, Clemson, SC 29634, USA; (B.D.); (E.M.); (S.M.S.)
| | - Erika Meaddough
- Healthcare Genetics Program, School of Nursing, Clemson University, Clemson, SC 29634, USA; (B.D.); (E.M.); (S.M.S.)
| | - Sara M. Sarasua
- Healthcare Genetics Program, School of Nursing, Clemson University, Clemson, SC 29634, USA; (B.D.); (E.M.); (S.M.S.)
| | | | - Katy Phelan
- Florida Cancer Specialists & Research Institute, Fort Myers, FL 33905, USA;
| | - Luigi Boccuto
- Healthcare Genetics Program, School of Nursing, Clemson University, Clemson, SC 29634, USA; (B.D.); (E.M.); (S.M.S.)
| |
Collapse
|
42
|
Srikanth S, Jain L, Zepeda-Mendoza C, Cascio L, Jones K, Pauly R, DuPont B, Rogers C, Sarasua S, Phelan K, Morton C, Boccuto L. Position effects of 22q13 rearrangements on candidate genes in Phelan-McDermid syndrome. PLoS One 2021; 16:e0253859. [PMID: 34228749 PMCID: PMC8259982 DOI: 10.1371/journal.pone.0253859] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 06/15/2021] [Indexed: 11/19/2022] Open
Abstract
Phelan-McDermid syndrome (PMS) is a multi-system disorder characterized by significant variability in clinical presentation. The genetic etiology is also variable with differing sizes of deletions in the chromosome 22q13 region and types of genetic abnormalities (e.g., terminal or interstitial deletions, translocations, ring chromosomes, or SHANK3 variants). Position effects have been shown to affect gene expression and function and play a role in the clinical presentation of various genetic conditions. This study employed a topologically associating domain (TAD) analysis approach to investigate position effects of chromosomal rearrangements on selected candidate genes mapped to 22q13 in 81 individuals with PMS. Data collected were correlated with clinical information from these individuals and with expression and metabolic profiles of lymphoblastoid cells from selected cases. The data confirmed TAD predictions for genes encompassed in the deletions and the clinical and molecular data indicated clear differences among individuals with different 22q13 deletion sizes. The results of the study indicate a positive correlation between deletion size and phenotype severity in PMS and provide evidence of the contribution of other genes to the clinical variability in this developmental disorder by reduced gene expression and altered metabolomics.
Collapse
Affiliation(s)
- Sujata Srikanth
- Greenwood Genetic Center, Greenwood, SC, United States of America
| | - Lavanya Jain
- School of Nursing, Healthcare Genetics Program, Clemson University, Clemson, SC, United States of America
| | - Cinthya Zepeda-Mendoza
- Department of Obstetrics, Gynecology, and Reproductive Biology, Brigham and Women’s Hospital, Boston, MA, United States of America
- Harvard Medical School, Boston, MA, United States of America
| | - Lauren Cascio
- Greenwood Genetic Center, Greenwood, SC, United States of America
| | - Kelly Jones
- Greenwood Genetic Center, Greenwood, SC, United States of America
| | - Rini Pauly
- Greenwood Genetic Center, Greenwood, SC, United States of America
| | - Barb DuPont
- Greenwood Genetic Center, Greenwood, SC, United States of America
| | - Curtis Rogers
- Greenwood Genetic Center, Greenwood, SC, United States of America
| | - Sara Sarasua
- School of Nursing, Healthcare Genetics Program, Clemson University, Clemson, SC, United States of America
| | - Katy Phelan
- Genetics Laboratory, Florida Cancer Specialists and Research Institute, Fort Myers, FL, United States of America
| | - Cynthia Morton
- Department of Obstetrics, Gynecology, and Reproductive Biology, Brigham and Women’s Hospital, Boston, MA, United States of America
- Harvard Medical School, Boston, MA, United States of America
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, United States of America
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA, United States of America
- Division of Human Communication, Development and Hearing, School of Biological Sciences, Manchester Academic Health Science Center, Manchester, United Kingdom
| | - Luigi Boccuto
- Greenwood Genetic Center, Greenwood, SC, United States of America
- School of Nursing, College of Behavioral, Social and Health Sciences, Clemson University, Clemson, SC, United States of America
| |
Collapse
|
43
|
May HJ, Jeong J, Revah-Politi A, Cohen JS, Chassevent A, Baptista J, Baugh EH, Bier L, Bottani A, Carminho A Rodrigues MT, Conlon C, Fluss J, Guipponi M, Kim CA, Matsumoto N, Person R, Primiano M, Rankin J, Shinawi M, Smith-Hicks C, Telegrafi A, Toy S, Uchiyama Y, Aggarwal V, Goldstein DB, Roche KW, Anyane-Yeboa K. Truncating variants in the SHANK1 gene are associated with a spectrum of neurodevelopmental disorders. Genet Med 2021; 23:1912-1921. [PMID: 34113010 DOI: 10.1038/s41436-021-01222-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 05/07/2021] [Accepted: 05/10/2021] [Indexed: 11/09/2022] Open
Abstract
PURPOSE In this study, we aimed to characterize the clinical phenotype of a SHANK1-related disorder and define the functional consequences of SHANK1 truncating variants. METHODS Exome sequencing (ES) was performed for six individuals who presented with neurodevelopmental disorders. Individuals were ascertained with the use of GeneMatcher and Database of Chromosomal Imbalance and Phenotype in Humans Using Ensembl Resources (DECIPHER). We evaluated potential nonsense-mediated decay (NMD) of two variants by making knock-in cell lines of endogenous truncated SHANK1, and expressed the truncated SHANK1 complementary DNA (cDNA) in HEK293 cells and cultured hippocampal neurons to examine the proteins. RESULTS ES detected de novo truncating variants in SHANK1 in six individuals. Evaluation of NMD resulted in stable transcripts, and the truncated SHANK1 completely lost binding with Homer1, a linker protein that binds to the C-terminus of SHANK1. These variants may disrupt protein-protein networks in dendritic spines. Dispersed localization of the truncated SHANK1 variants within the spine and dendritic shaft was also observed when expressed in neurons, indicating impaired synaptic localization of truncated SHANK1. CONCLUSION This report expands the clinical spectrum of individuals with truncating SHANK1 variants and describes the impact these variants may have on the pathophysiology of neurodevelopmental disorders.
Collapse
Affiliation(s)
- Halie J May
- Institute for Genomic Medicine, Columbia University Irving Medical Center, New York, NY, USA.
| | - Jaehoon Jeong
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Anya Revah-Politi
- Institute for Genomic Medicine, Columbia University Irving Medical Center, New York, NY, USA.,Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Julie S Cohen
- Department of Neurology and Developmental Medicine, Kennedy Krieger Institute, Baltimore, MD, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Anna Chassevent
- Department of Neurology and Developmental Medicine, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Julia Baptista
- Exeter Genomics Laboratory, Royal Devon and Exeter NHS Foundation Trust, Exeter, UK.,Institute of Biomedical & Clinical Science, University of Exeter Medical School, Exeter, UK
| | - Evan H Baugh
- Institute for Genomic Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Louise Bier
- Institute for Genomic Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Armand Bottani
- Division of Genetic Medicine, University Hospitals of Geneva, Geneva, Switzerland
| | | | - Charles Conlon
- Department of Neurology and Developmental Medicine, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Joel Fluss
- Pediatric Neurology Unit, Pediatrics Subspecialties Service, Geneva Children's Hospital, Geneva, Switzerland
| | - Michel Guipponi
- Division of Genetic Medicine, University Hospitals of Geneva, Geneva, Switzerland
| | - Chong Ae Kim
- Genetics Unit, Instituto da Crianca, Hospital das Clinicas, Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Naomichi Matsumoto
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Richard Person
- Clinical Genomics Program, GeneDx, Gaithersburg, MD, USA
| | - Michelle Primiano
- Division of Clinical Genetics, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, USA
| | - Julia Rankin
- Department of Clinical Genetics, Royal Devon and Exeter NHS Foundation Trust, Exeter, UK
| | - Marwan Shinawi
- Department of Pediatrics, Division of Genetics and Genomic Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Constance Smith-Hicks
- Department of Neurology and Developmental Medicine, Kennedy Krieger Institute, Baltimore, MD, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Aida Telegrafi
- Clinical Genomics Program, GeneDx, Gaithersburg, MD, USA
| | - Samantha Toy
- Department of Pediatrics, Division of Genetics and Genomic Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Yuri Uchiyama
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan.,Department of Rare Disease Genomics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Vimla Aggarwal
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - David B Goldstein
- Institute for Genomic Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Katherine W Roche
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Kwame Anyane-Yeboa
- Institute for Genomic Medicine, Columbia University Irving Medical Center, New York, NY, USA. .,Division of Clinical Genetics, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
44
|
Ricciardello A, Tomaiuolo P, Persico AM. Genotype-phenotype correlation in Phelan-McDermid syndrome: A comprehensive review of chromosome 22q13 deleted genes. Am J Med Genet A 2021; 185:2211-2233. [PMID: 33949759 PMCID: PMC8251815 DOI: 10.1002/ajmg.a.62222] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 03/28/2021] [Accepted: 04/04/2021] [Indexed: 12/19/2022]
Abstract
Phelan‐McDermid syndrome (PMS, OMIM #606232), also known as chromosome 22q13 deletion syndrome, is a rare genetic disorder characterized by intellectual disability, hypotonia, delayed or absent speech, motor impairment, autism spectrum disorder, behavioral anomalies, and minor aspecific dysmorphic features. Haploinsufficiency of SHANK3, due to intragenic deletions or point mutations, is sufficient to cause many neurobehavioral features of PMS. However, several additional genes located within larger 22q13 deletions can contribute to the great interindividual variability observed in the PMS phenotype. This review summarizes the phenotypic contributions predicted for 213 genes distributed along the largest 22q13.2‐q13.33 terminal deletion detected in our sample of 63 PMS patients by array‐CGH analysis, spanning 9.08 Mb. Genes have been grouped into four categories: (1) genes causing human diseases with an autosomal dominant mechanism, or (2) with an autosomal recessive mechanism; (3) morphogenetically relevant genes, either involved in human diseases with additive co‐dominant, polygenic, and/or multifactorial mechanisms, or implicated in animal models but not yet documented in human pathology; (4) protein coding genes either functionally nonrelevant, with unknown function, or pathogenic through mechanisms other than haploinsufficiency; piRNAs, noncoding RNAs, miRNAs, novel transcripts and pseudogenes. Our aim is to understand genotype–phenotype correlations in PMS patients and to provide clinicians with a conceptual framework to promote evidence‐based genetic work‐ups, clinical assessments, and therapeutic interventions.
Collapse
Affiliation(s)
- Arianna Ricciardello
- Interdepartmental Program "Autism 0-90", "Gaetano Martino" University Hospital, University of Messina, Messina, Italy
| | - Pasquale Tomaiuolo
- Interdepartmental Program "Autism 0-90", "Gaetano Martino" University Hospital, University of Messina, Messina, Italy
| | - Antonio M Persico
- Interdepartmental Program "Autism 0-90", "Gaetano Martino" University Hospital, University of Messina, Messina, Italy
| |
Collapse
|
45
|
Stern S, Hacohen N, Meiner V, Yagel S, Zenvirt S, Shkedi-Rafid S, Macarov M, Valsky DV, Porat S, Yanai N, Frumkin A, Daum H. Universal chromosomal microarray analysis reveals high proportion of copy-number variants in low-risk pregnancies. ULTRASOUND IN OBSTETRICS & GYNECOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY OF ULTRASOUND IN OBSTETRICS AND GYNECOLOGY 2021; 57:813-820. [PMID: 32202684 DOI: 10.1002/uog.22026] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 03/12/2020] [Accepted: 03/16/2020] [Indexed: 06/10/2023]
Abstract
OBJECTIVES To evaluate the yield and utility of the routine use of chromosomal microarray analysis (CMA) for prenatal genetic diagnosis in a large cohort of pregnancies with normal ultrasound (US) at the time of genetic testing, compared with pregnancies with abnormal US findings. METHODS We reviewed all prenatal CMA results in our center between November 2013 and December 2018. The prevalence of different CMA results in pregnancies with normal US at the time of genetic testing ('low-risk pregnancies'), was compared with that in pregnancies with abnormal US findings ('high-risk pregnancies'). Medical records were searched in order to evaluate subsequent US follow-up and the outcome of pregnancies with a clinically relevant copy-number variant (CNV), i.e. a pathogenic or likely pathogenic CNV or a susceptibility locus for disease with > 10% penetrance, related to early-onset disease in the low-risk group. RESULTS In a cohort of 6431 low-risk pregnancies that underwent CMA, the prevalence of a clinically significant CNV related to early-onset disease was 1.1% (72/6431), which was significantly lower than the prevalence in high-risk pregnancies (4.9% (65/1326)). Of the low-risk pregnancies, 0.4% (27/6431) had a pathogenic or likely pathogenic CNV, and another 0.7% (45/6431) had a susceptibility locus with more than 10% penetrance. Follow-up of the low-risk pregnancies with a clinically significant early-onset CNV revealed that 31.9% (23/72) were terminated, while outcome data were missing in 26.4% (19/72). In 16.7% (12/72) of low-risk pregnancies, an US abnormality was discovered later on in gestation, after genetic testing had been performed. CONCLUSION Although the background risk of identifying a clinically significant early-onset abnormal CMA result in pregnancies with a low a-priori risk is lower than that observed in high-risk pregnancies, the risk is substantial and should be conveyed to all pregnant women. © 2020 International Society of Ultrasound in Obstetrics and Gynecology.
Collapse
Affiliation(s)
- S Stern
- Department of Obstetrics and Gynecology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - N Hacohen
- Department of Genetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - V Meiner
- Department of Genetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - S Yagel
- Department of Obstetrics and Gynecology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - S Zenvirt
- Department of Genetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - S Shkedi-Rafid
- Department of Genetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - M Macarov
- Department of Genetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - D V Valsky
- Department of Obstetrics and Gynecology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - S Porat
- Department of Obstetrics and Gynecology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - N Yanai
- Department of Obstetrics and Gynecology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - A Frumkin
- Department of Genetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - H Daum
- Department of Genetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
46
|
Hoerr RE, Ngo K, Friedman KL. When the Ends Justify the Means: Regulation of Telomere Addition at Double-Strand Breaks in Yeast. Front Cell Dev Biol 2021; 9:655377. [PMID: 33816507 PMCID: PMC8012806 DOI: 10.3389/fcell.2021.655377] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 02/15/2021] [Indexed: 11/23/2022] Open
Abstract
Telomeres, repetitive sequences located at the ends of most eukaryotic chromosomes, provide a mechanism to replenish terminal sequences lost during DNA replication, limit nucleolytic resection, and protect chromosome ends from engaging in double-strand break (DSB) repair. The ribonucleoprotein telomerase contains an RNA subunit that serves as the template for the synthesis of telomeric DNA. While telomere elongation is typically primed by a 3′ overhang at existing chromosome ends, telomerase can act upon internal non-telomeric sequences. Such de novo telomere addition can be programmed (for example, during chromosome fragmentation in ciliated protozoa) or can occur spontaneously in response to a chromosome break. Telomerase action at a DSB can interfere with conservative mechanisms of DNA repair and results in loss of distal sequences but may prevent additional nucleolytic resection and/or chromosome rearrangement through formation of a functional telomere (termed “chromosome healing”). Here, we review studies of spontaneous and induced DSBs in the yeast Saccharomyces cerevisiae that shed light on mechanisms that negatively regulate de novo telomere addition, in particular how the cell prevents telomerase action at DSBs while facilitating elongation of critically short telomeres. Much of our understanding comes from the use of perfect artificial telomeric tracts to “seed” de novo telomere addition. However, endogenous sequences that are enriched in thymine and guanine nucleotides on one strand (TG-rich) but do not perfectly match the telomere consensus sequence can also stimulate unusually high frequencies of telomere formation following a DSB. These observations suggest that some internal sites may fully or partially escape mechanisms that normally negatively regulate de novo telomere addition.
Collapse
Affiliation(s)
- Remington E Hoerr
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, United States
| | - Katrina Ngo
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, United States
| | - Katherine L Friedman
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, United States
| |
Collapse
|
47
|
Vogels A, Droogmans G, Vergaelen E, Van Buggenhout G, Swillen A. Recent developments in Phelan-McDermid syndrome research: an update on cognitive development, communication and psychiatric disorders. Curr Opin Psychiatry 2021; 34:118-122. [PMID: 33278153 DOI: 10.1097/yco.0000000000000672] [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] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW The purpose of this review is to summarize the literature on cognitive development, communication, behavioral or psychiatric aspects in Phelan-McDermid syndrome (PMS) and to discuss the clinical implications and recommendations of these summarized findings. RECENT FINDINGS PMS is often associated with severe communication impairments, behavioral or psychiatric problems and regression. These challenges may adversely affect and impair the quality of life of the individual with PMS and his family. SUMMARY Individuals with PMS experience intellectual disability, communication and behavioral/psychiatric challenges, such as catatonia, bipolar disorder and regression across the lifespan. Providing appropriate guidance and support to them and their families demands a better understanding of these challenges.
Collapse
Affiliation(s)
- Annick Vogels
- Center for Human Genetics, University Hospital Gasthuisberg
- Department of Human Genetics
| | | | - Elfi Vergaelen
- University Psychiatric Center, Mind Body Research Group, KU Leuven (University of Leuven), Leuven, Belgium
| | - Griet Van Buggenhout
- Center for Human Genetics, University Hospital Gasthuisberg
- Department of Human Genetics
| | - Ann Swillen
- Center for Human Genetics, University Hospital Gasthuisberg
- Department of Human Genetics
| |
Collapse
|
48
|
Schenkel LC, Aref-Eshghi E, Rooney K, Kerkhof J, Levy MA, McConkey H, Rogers RC, Phelan K, Sarasua SM, Jain L, Pauly R, Boccuto L, DuPont B, Cappuccio G, Brunetti-Pierri N, Schwartz CE, Sadikovic B. DNA methylation epi-signature is associated with two molecularly and phenotypically distinct clinical subtypes of Phelan-McDermid syndrome. Clin Epigenetics 2021; 13:2. [PMID: 33407854 PMCID: PMC7789817 DOI: 10.1186/s13148-020-00990-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 12/09/2020] [Indexed: 12/31/2022] Open
Abstract
Background Phelan-McDermid syndrome is characterized by a range of neurodevelopmental phenotypes with incomplete penetrance and variable expressivity. It is caused by a variable size and breakpoint microdeletions in the distal long arm of chromosome 22, referred to as 22q13.3 deletion syndrome, including the SHANK3 gene. Genetic defects in a growing number of neurodevelopmental genes have been shown to cause genome-wide disruptions in epigenomic profiles referred to as epi-signatures in affected individuals. Results In this study we assessed genome-wide DNA methylation profiles in a cohort of 22 individuals with Phelan-McDermid syndrome, including 11 individuals with large (2 to 5.8 Mb) 22q13.3 deletions, 10 with small deletions (< 1 Mb) or intragenic variants in SHANK3 and one mosaic case. We describe a novel genome-wide DNA methylation epi-signature in a subset of individuals with Phelan-McDermid syndrome. Conclusion We identified the critical region including the BRD1 gene as responsible for the Phelan-McDermid syndrome epi-signature. Metabolomic profiles of individuals with the DNA methylation epi-signature showed significantly different metabolomic profiles indicating evidence of two molecularly and phenotypically distinct clinical subtypes of Phelan-McDermid syndrome.
Collapse
Affiliation(s)
- L C Schenkel
- Molecular Genetics Laboratory, Molecular Diagnostics Division, London Health Sciences Centre, London, ON, N6A5W9, Canada.,Department of Pathology and Laboratory Medicine, Western University, London, ON, N6A3K7, Canada
| | - E Aref-Eshghi
- Molecular Genetics Laboratory, Molecular Diagnostics Division, London Health Sciences Centre, London, ON, N6A5W9, Canada
| | - K Rooney
- Molecular Genetics Laboratory, Molecular Diagnostics Division, London Health Sciences Centre, London, ON, N6A5W9, Canada
| | - J Kerkhof
- Molecular Genetics Laboratory, Molecular Diagnostics Division, London Health Sciences Centre, London, ON, N6A5W9, Canada
| | - M A Levy
- Molecular Genetics Laboratory, Molecular Diagnostics Division, London Health Sciences Centre, London, ON, N6A5W9, Canada
| | - H McConkey
- Molecular Genetics Laboratory, Molecular Diagnostics Division, London Health Sciences Centre, London, ON, N6A5W9, Canada
| | - R C Rogers
- Greenville Office, Greenwood Genetic Center, Greenville, SC, 29605, USA
| | - K Phelan
- Genetics Laboratory, Florida Cancer Specialists and Research Institute, Fort Myers, FL, 33816, USA
| | | | - L Jain
- Greenwood Genetic Center, Greenwood, SC, 29646, USA.,Clemson University, Clemson, SC, 29634, USA
| | - R Pauly
- Greenwood Genetic Center, Greenwood, SC, 29646, USA
| | - L Boccuto
- Greenwood Genetic Center, Greenwood, SC, 29646, USA.,Clemson University, Clemson, SC, 29634, USA
| | - B DuPont
- Greenwood Genetic Center, Greenwood, SC, 29646, USA
| | - G Cappuccio
- Department of Translational Medicine, University Federico II, 80131, Naples, NA, Italy.,Telethon Institute of Genetics and Medicine, Pozzuoli, NA, Italy
| | - N Brunetti-Pierri
- Department of Translational Medicine, University Federico II, 80131, Naples, NA, Italy.,Telethon Institute of Genetics and Medicine, Pozzuoli, NA, Italy
| | - C E Schwartz
- Greenwood Genetic Center, Greenwood, SC, 29646, USA.
| | - B Sadikovic
- Molecular Genetics Laboratory, Molecular Diagnostics Division, London Health Sciences Centre, London, ON, N6A5W9, Canada. .,Department of Pathology and Laboratory Medicine, Western University, London, ON, N6A3K7, Canada.
| |
Collapse
|
49
|
Mizban N, Vousooghi N, Mizban N. Association of SHANK3 Gene Polymorphism and Parkinson Disease in the North of Iran. Basic Clin Neurosci 2021; 12:57-62. [PMID: 33995927 PMCID: PMC8114862 DOI: 10.32598/bcn.12.1.255.4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Revised: 02/25/2018] [Accepted: 04/30/2018] [Indexed: 11/20/2022] Open
Abstract
INTRODUCTION Parkinson Disease (PD), the second most common chronic neurodegenerative disorder, is characterized by tremor, bradykinesia, rigidity, and postural instability. SHANK3 (SH3 and multiple ankyrin repeat domain 3) belongs to the extremely conserved ProSAP/Shank family of synaptic scaffolding proteins. Meanwhile, rs9616915 is a non-synonymous SNP (T>C) located in the exon 6 of the SHANK3 gene, which induces substitution of isoleucine to threonine and affects the function of the resulted protein. The present study aimed to evaluate whether rs9616915 polymorphism of SHANK3 is involved in the susceptibility to PD. METHODS The study subjects were 100 patients diagnosed with PD and 100 control volunteers. The obtained samples were evaluated by the polymerase chain reaction-restriction fragment length polymorphism method. RESULTS A significant association was found in genotype distribution between cases and controls. Individuals with TC genotype had increased risk of PD (P=0.035, OR=1.98, 95% CI=1.04 - 3.74). No significant difference was found in allele distribution (P=0.7). CONCLUSION The findings suggest that the SHANK3 rs9616915 polymorphism is associated with an increased risk of PD in the population. Further studies are needed to confirm the role of the SHANK3 gene in PD.
Collapse
Affiliation(s)
- Nahid Mizban
- Department of Biology, Faculty of Sciences, University of Guilan, Guilan, Iran
| | - Nasim Vousooghi
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nasrin Mizban
- Department of Biological Sciences, Faculty of Biological Sciences and Technologies, Shahid Beheshti University, Tehran, Iran
| |
Collapse
|
50
|
Liu C, Li D, Yang H, Li H, Xu Q, Zhou B, Hu C, Li C, Wang Y, Qiao Z, Jiang YH, Xu X. Altered striatum centered brain structures in SHANK3 deficient Chinese children with genotype and phenotype profiling. Prog Neurobiol 2020; 200:101985. [PMID: 33388374 PMCID: PMC8572121 DOI: 10.1016/j.pneurobio.2020.101985] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Revised: 12/15/2020] [Accepted: 12/27/2020] [Indexed: 12/01/2022]
Abstract
SHANK3 deficiency represents one of the most replicated monogenic risk factors for autism spectrum disorder (ASD) and SHANK3 caused ASD presents a unique opportunity to understand the underlying neuropathological mechanisms of ASD. In this study, genetic tests, comprehensive clinical and neurobehavioral evaluations, as well as multimodal structural MRI using voxel-based morphometry (VBM) and tract-based spatial statistics (TBSS) were conducted in SHANK3 group (N = 14 with SHANK3 defects), ASD controls (N = 26 with idiopathic ASD without SHANK3 defects) and typically developing (TD) controls (N = 32). Phenotypically, we reported several new features in Chinese SHANK3 deficient children including anteverted nares, sensory stimulation seeking, dental abnormalities and hematological problems. In SHANK3 group, VBM revealed decreased grey matter volumes mainly in dorsal striatum, amygdala, hippocampus and parahippocampal gyrus; TBSS demonstrated decreased fractional anisotropy in multiple tracts involving projection, association and commissural fibers, including middle cerebral peduncle, corpus callosum, superior longitudinal fasciculus, corona radiata, external and internal capsule, and posterior thalamic radiation, etc. We report that the disrupted striatum centered brain structures are associated with SHANK3 deficient children. Study of subjects with monogenic cause offer specific insights into the neuroimaging studies of ASD. The discovery may support a path for future functional connectivity studies to allow for more in-depth understandings of the abnormal neural circuits and the underlying neuropathological mechanisms for ASD.
Collapse
Affiliation(s)
- Chunxue Liu
- Department of Child Health Care, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai, China
| | - Dongyun Li
- Department of Child Health Care, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai, China
| | - Haowei Yang
- Department of Radiology, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai, China
| | - Huiping Li
- Department of Child Health Care, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai, China
| | - Qiong Xu
- Department of Child Health Care, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai, China
| | - Bingrui Zhou
- Department of Child Health Care, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai, China
| | - Chunchun Hu
- Department of Child Health Care, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai, China
| | - Chunyang Li
- Department of Child Health Care, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai, China
| | - Yi Wang
- Department of Child Health Care, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai, China
| | - Zhongwei Qiao
- Department of Radiology, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai, China.
| | - Yong-Hui Jiang
- Department of Genetics, Pediatrics and Neuroscience, Yale University School of Medicine, New Heaven CT 06520 USA.
| | - Xiu Xu
- Department of Child Health Care, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai, China.
| |
Collapse
|