1
|
Serrano-Juárez CA, Prieto-Corona B, Rodríguez-Camacho M, Sandoval-Lira L, Villalva-Sánchez ÁF, Yáñez-Téllez MG, López MFR. Neuropsychological Genotype-Phenotype in Patients with Williams Syndrome with Atypical Deletions: A Systematic Review. Neuropsychol Rev 2023; 33:891-911. [PMID: 36520254 DOI: 10.1007/s11065-022-09571-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 11/04/2022] [Indexed: 12/16/2022]
Abstract
Williams syndrome (WS) is a neurodevelopmental disorder caused by a microdeletion in the q11.23 region of chromosome 7. Recent case series reports and clinical case studies have suggested that the cognitive, behavioral, emotional, and social profile in WS could depend on the genes involved in the deletion. The objective of this systematic review was to analyze and synthesize the variability of the cognitive and behavioral profile of WS with atypical deletion and its probable relationship with the affected genes. The medical subject headings searched were "Williams syndrome," "genotype," "phenotype," "cognitive profile," and "atypical deletion." The studies included were in English or Spanish, with children and adults, and published between January 2000 and October 2022. Twenty-three studies are reported. The characteristics of the participants, the genes involved, the neuropsychological domains and instruments, and the prevalence of the WS cognitive profile criteria were used for the genotype-phenotype analysis. The genes with a major impact on the cognitive profile of WS were (a) LIMK1 and those belonging to the GTF2I family, the former with a greater influence on visuospatial abilities; (b) GTF2IRD1 and GTF2I, which have an impact on intellectual capacity as well as on visuospatial and social skills; (c) FZD9, BAZ1B, STX1A, and CLIP2, which influence the cognitive profile if other genes are also effected; and (d) GTF2IRD2, which is related to the severity of the effect on visuospatial and social skills, producing a behavioral phenotype like that of the autism spectrum. The review revealed four neuropsychological phenotypes, depending on the genes involved, and established the need for more comprehensive study of the neuropsychological profile of these patients. Based on the results found, we propose a model for the investigation of and clinical approach to the WS neuropsychological phenotype.
Collapse
Affiliation(s)
- Carlos Alberto Serrano-Juárez
- Neuroscience Group. Laboratorio de Neurometría, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Av. De los Barrios #1, Col. Los Reyes Iztacala, Tlalnepantla, Estado de México, CP 54090, México
| | - Belén Prieto-Corona
- Neuroscience Group. Laboratorio de Neurometría, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Av. De los Barrios #1, Col. Los Reyes Iztacala, Tlalnepantla, Estado de México, CP 54090, México.
| | - Mario Rodríguez-Camacho
- Neuroscience Group. Laboratorio de Neurometría, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Av. De los Barrios #1, Col. Los Reyes Iztacala, Tlalnepantla, Estado de México, CP 54090, México
| | - Lucero Sandoval-Lira
- Neuroscience Group. Laboratorio de Neurometría, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Av. De los Barrios #1, Col. Los Reyes Iztacala, Tlalnepantla, Estado de México, CP 54090, México
| | - Ángel Fernando Villalva-Sánchez
- Neuroscience Group. Laboratorio de Neurometría, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Av. De los Barrios #1, Col. Los Reyes Iztacala, Tlalnepantla, Estado de México, CP 54090, México
| | - Ma Guillermina Yáñez-Téllez
- Neuroscience Group. Laboratorio de Neurometría, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Av. De los Barrios #1, Col. Los Reyes Iztacala, Tlalnepantla, Estado de México, CP 54090, México
| | | |
Collapse
|
2
|
Strong E, Mervis CB, Tam E, Morris CA, Klein-Tasman BP, Velleman SL, Osborne LR. DNA methylation profiles in individuals with rare, atypical 7q11.23 CNVs correlate with GTF2I and GTF2IRD1 copy number. NPJ Genom Med 2023; 8:25. [PMID: 37709781 PMCID: PMC10502022 DOI: 10.1038/s41525-023-00368-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 08/18/2023] [Indexed: 09/16/2023] Open
Abstract
Williams-Beuren syndrome (WBS) and 7q11.23 duplication syndrome (Dup7) are rare neurodevelopmental disorders caused by deletion and duplication of a 1.5 Mb region that includes at least five genes with a known role in epigenetic regulation. We have shown that CNV of this chromosome segment causes dose-dependent, genome-wide changes in DNA methylation, but the specific genes driving these changes are unknown. We measured genome-wide whole blood DNA methylation in six participants with atypical CNV of 7q11.23 (three with deletions and three with duplications) using the Illumina HumanMethylation450k array and compared their profiles with those from groups of individuals with classic WBS or classic Dup7 and with typically developing (TD) controls. Across the top 1000 most variable positions we found that only the atypical rearrangements that changed the copy number of GTF2IRD1 and/or GTF2I (coding for the TFII-IRD1 and TFII-I proteins) clustered with their respective syndromic cohorts. This finding was supported by results from hierarchical clustering across a selection of differentially methylated CpGs, in addition to pyrosequencing validation. These findings suggest that CNV of the GTF2I genes at the telomeric end of the 7q11.23 interval is a key contributor to the large changes in DNA methylation that are seen in blood DNA from our WBS and Dup7 cohorts, compared to TD controls. Our findings suggest that members of the TFII-I protein family are involved in epigenetic processes that alter DNA methylation on a genome-wide level.
Collapse
Affiliation(s)
- Emma Strong
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Division of Genome Diagnostics, Department of Pathology and Laboratory Medicine, BC Children's and Women's Hospital, Vancouver, BC, Canada
| | - Carolyn B Mervis
- Department of Psychological and Brain Sciences, University of Louisville, Louisville, KY, USA
| | - Elaine Tam
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Colleen A Morris
- Department of Pediatrics, Kirk Kerkorian School of Medicine at University of Nevada Las Vegas, Las Vegas, NV, USA
| | | | - Shelley L Velleman
- Department of Communication Sciences and Disorders, University of Vermont, Burlington, VT, USA
| | - Lucy R Osborne
- Departments of Medicine and Molecular Genetics, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
3
|
Carlotto BS, Deconte D, Diniz BL, Silva PRD, Zen PRG, Silva AAD. Fluorescence in situ hybridization (FISH) as an irreplaceable diagnostic tool for Williams-Beuren syndrome in developing countries: a literature review. REVISTA PAULISTA DE PEDIATRIA : ORGAO OFICIAL DA SOCIEDADE DE PEDIATRIA DE SAO PAULO 2023; 42:e2022125. [PMID: 37436242 DOI: 10.1590/1984-0462/2023/41/2022125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 11/14/2022] [Indexed: 07/13/2023]
Abstract
OBJECTIVE The aim of this study was to sum up and characterize all Williams-Beuren syndrome cases diagnosed by fluorescence in situ hybridization (FISH) since its implementation, as well as to discuss FISH as a cost-effective methodology in developing countries. DATA SOURCE From January 1986 to January 2022, articles were selected using the databases in PubMed (Medline) and SciELO. The following terms were used: Williams syndrome and In Situ Hybridization, Fluorescence. Inclusion criteria included Williams-Beuren syndrome cases diagnosed by FISH with a stratified phenotype of each patient. Only studies written in English, Spanish, and Portuguese were included. Studies with overlapping syndromes or genetic conditions were excluded. DATA SYNTHESIS After screening, 64 articles were included. A total of 205 individuals with Williams-Beuren syndrome diagnosed by FISH were included and further analyzed. Cardiovascular malformations were the most frequent finding (85.4%). Supravalvular aortic stenosis (62.4%) and pulmonary stenosis (30.7%) were the main cardiac alterations described. CONCLUSIONS Our literature review reinforces that cardiac features may be the key to early diagnosis in Williams-Beuren syndrome patients. In addition, FISH may be the best diagnostic tool for developing nations that have limited access to new technologic resources.
Collapse
Affiliation(s)
| | - Desirée Deconte
- Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, RS, Brazil
| | - Bruna Lixinski Diniz
- Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, RS, Brazil
| | | | - Paulo Ricardo Gazzola Zen
- Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, RS, Brazil
- Irmandade da Santa Casa de Misericórdia de Porto Alegre, Porto Alegre, RS, Brazil
| | | |
Collapse
|
4
|
Davenport CM, Teubner BJW, Han SB, Patton MH, Eom TY, Garic D, Lansdell BJ, Shirinifard A, Chang TC, Klein J, Pruett-Miller SM, Blundon JA, Zakharenko SS. Innate frequency-discrimination hyperacuity in Williams-Beuren syndrome mice. Cell 2022; 185:3877-3895.e21. [PMID: 36152627 PMCID: PMC9588278 DOI: 10.1016/j.cell.2022.08.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 06/14/2022] [Accepted: 08/24/2022] [Indexed: 01/26/2023]
Abstract
Williams-Beuren syndrome (WBS) is a rare disorder caused by hemizygous microdeletion of ∼27 contiguous genes. Despite neurodevelopmental and cognitive deficits, individuals with WBS have spared or enhanced musical and auditory abilities, potentially offering an insight into the genetic basis of auditory perception. Here, we report that the mouse models of WBS have innately enhanced frequency-discrimination acuity and improved frequency coding in the auditory cortex (ACx). Chemogenetic rescue showed frequency-discrimination hyperacuity is caused by hyperexcitable interneurons in the ACx. Haploinsufficiency of one WBS gene, Gtf2ird1, replicated WBS phenotypes by downregulating the neuropeptide receptor VIPR1. VIPR1 is reduced in the ACx of individuals with WBS and in the cerebral organoids derived from human induced pluripotent stem cells with the WBS microdeletion. Vipr1 deletion or overexpression in ACx interneurons mimicked or reversed, respectively, the cellular and behavioral phenotypes of WBS mice. Thus, the Gtf2ird1-Vipr1 mechanism in ACx interneurons may underlie the superior auditory acuity in WBS.
Collapse
Affiliation(s)
- Christopher M Davenport
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Brett J W Teubner
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Seung Baek Han
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Mary H Patton
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Tae-Yeon Eom
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Dusan Garic
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Benjamin J Lansdell
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Abbas Shirinifard
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Ti-Cheng Chang
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Jonathon Klein
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Shondra M Pruett-Miller
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Jay A Blundon
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Stanislav S Zakharenko
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| |
Collapse
|
5
|
Grad M, Nir A, Levy G, Trangle SS, Shapira G, Shomron N, Assaf Y, Barak B. Altered White Matter and microRNA Expression in a Murine Model Related to Williams Syndrome Suggests That miR-34b/c Affects Brain Development via Ptpru and Dcx Modulation. Cells 2022; 11:cells11010158. [PMID: 35011720 PMCID: PMC8750756 DOI: 10.3390/cells11010158] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/15/2021] [Accepted: 12/28/2021] [Indexed: 11/16/2022] Open
Abstract
Williams syndrome (WS) is a multisystem neurodevelopmental disorder caused by a de novo hemizygous deletion of ~26 genes from chromosome 7q11.23, among them the general transcription factor II-I (GTF2I). By studying a novel murine model for the hypersociability phenotype associated with WS, we previously revealed surprising aberrations in myelination and cell differentiation properties in the cortices of mutant mice compared to controls. These mutant mice had selective deletion of Gtf2i in the excitatory neurons of the forebrain. Here, we applied diffusion magnetic resonance imaging and fiber tracking, which showed a reduction in the number of streamlines in limbic outputs such as the fimbria/fornix fibers and the stria terminalis, as well as the corpus callosum of these mutant mice compared to controls. Furthermore, we utilized next-generation sequencing (NGS) analysis of cortical small RNAs' expression (RNA-Seq) levels to identify altered expression of microRNAs (miRNAs), including two from the miR-34 cluster, known to be involved in prominent processes in the developing nervous system. Luciferase reporter assay confirmed the direct binding of miR-34c-5p to the 3'UTR of PTPRU-a gene involved in neural development that was elevated in the cortices of mutant mice relative to controls. Moreover, we found an age-dependent variation in the expression levels of doublecortin (Dcx)-a verified miR-34 target. Thus, we demonstrate the substantial effect a single gene deletion can exert on miRNA regulation and brain structure, and advance our understanding and, hopefully, treatment of WS.
Collapse
Affiliation(s)
- Meitar Grad
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel; (M.G.); (A.N.); (G.L.); (N.S.); (Y.A.)
| | - Ariel Nir
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel; (M.G.); (A.N.); (G.L.); (N.S.); (Y.A.)
| | - Gilad Levy
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel; (M.G.); (A.N.); (G.L.); (N.S.); (Y.A.)
| | - Sari Schokoroy Trangle
- Faculty of Social Sciences, School of Psychological Sciences, Tel Aviv University, Tel Aviv 6997801, Israel;
| | - Guy Shapira
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel;
- Edmond J. Safra Center for Bioinformatics, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Noam Shomron
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel; (M.G.); (A.N.); (G.L.); (N.S.); (Y.A.)
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel;
- Edmond J. Safra Center for Bioinformatics, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Yaniv Assaf
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel; (M.G.); (A.N.); (G.L.); (N.S.); (Y.A.)
- Faculty of Life Sciences, School of Neurobiology, Biochemistry & Biophysics, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Boaz Barak
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel; (M.G.); (A.N.); (G.L.); (N.S.); (Y.A.)
- Faculty of Social Sciences, School of Psychological Sciences, Tel Aviv University, Tel Aviv 6997801, Israel;
- Correspondence:
| |
Collapse
|
6
|
Honjo RS, Monteleone VF, Aiello VD, Wagenfuhr J, Issa VS, Pomerantzeff PMA, Furusawa EA, Zanardo EA, Kulikowski LD, Bertola DR, Kim CA. Cardiovascular findings in Williams-Beuren Syndrome: Experience of a single center with 127 cases. Am J Med Genet A 2021; 188:676-682. [PMID: 34713566 DOI: 10.1002/ajmg.a.62542] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 09/10/2021] [Accepted: 09/15/2021] [Indexed: 11/12/2022]
Abstract
Williams-Beuren syndrome (WBS) is a rare, microdeletion syndrome characterized by facial dysmorphisms, intellectual disability, a friendly personality, cardiovascular and other abnormalities. Cardiovascular defects (CVD) are among the most prevalent characteristics in WBS, being supravalvular aortic stenosis (SVAS) the most frequent, followed by peripheral pulmonary stenosis (PPS). A comprehensive retrospective review of medical records of 127 patients with molecular diagnosis of WBS, in a period of 20 years, was done to evaluate the incidence, the natural history of cardiovascular disease, and the need for surgical intervention, including heart transplantation (HT). A total of 94/127 patients presented with CVD. Of these 94 patients, 50% presented with SVAS and 22.3% needed heart surgery and/or cardiac catheterization including one that required HT due to severe SVAS-related heart failure at 19 years of age. The patient died in the postoperative period due to infectious complications. Cardiovascular problems are the major cause of sudden death in patients with WBS, who have a significantly higher mortality risk associated with surgical interventions. There is a higher risk for anesthesia-related adverse events and for major adverse cardiac events following surgery. End-stage heart failure due to myocardial ischemia has been described in WBS patients and it is important to consider that HT can become their only viable option. To our knowledge, the case mentioned here is the first HT reported in an adolescent with WBS. HT can be a viable therapeutic option in WBS patients with adequate evaluation, planning, and a multidisciplinary team to provide the required perioperative care and follow-up.
Collapse
Affiliation(s)
- Rachel Sayuri Honjo
- Genetics Unit, Instituto da Criança do Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Vanessa Figueiredo Monteleone
- Genetics Unit, Instituto da Criança do Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Vera Demarchi Aiello
- Pathology Laboratory, Instituto do Coração do Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Jaqueline Wagenfuhr
- Pediatric Cardiology Unit, Instituto da Criança do Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo, Brazil
| | | | - Pablo Maria Alberto Pomerantzeff
- Heart Transplantation Unit, Instituto do Coraçao do Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Erika Arai Furusawa
- Pediatric Nephrology Unit, Instituto da Criança do Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Evelin Aline Zanardo
- Cytogenomic Laboratory, LIM 03, Pathology Department, Faculdade de Medicina HC-FMUSP, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Leslie Domenici Kulikowski
- Cytogenomic Laboratory, LIM 03, Pathology Department, Faculdade de Medicina HC-FMUSP, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Debora Romeo Bertola
- Genetics Unit, Instituto da Criança do Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Chong Ae Kim
- Genetics Unit, Instituto da Criança do Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
7
|
Kopp ND, Nygaard KR, Liu Y, McCullough KB, Maloney SE, Gabel HW, Dougherty JD. Functions of Gtf2i and Gtf2ird1 in the developing brain: transcription, DNA binding and long-term behavioral consequences. Hum Mol Genet 2021; 29:1498-1519. [PMID: 32313931 DOI: 10.1093/hmg/ddaa070] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 02/19/2020] [Accepted: 04/13/2020] [Indexed: 12/13/2022] Open
Abstract
Gtf2ird1 and Gtf2i are two transcription factors (TFs) among the 28 genes deleted in Williams syndrome, and prior mouse models of each TF show behavioral phenotypes. Here we identify their genomic binding sites in the developing brain and test for additive effects of their mutation on transcription and behavior. GTF2IRD1 binding targets were enriched for transcriptional and chromatin regulators and mediators of ubiquitination. GTF2I targets were enriched for signal transduction proteins, including regulators of phosphorylation and WNT. Both TFs are highly enriched at promoters, strongly overlap CTCF binding and topological associating domain boundaries and moderately overlap each other, suggesting epistatic effects. Shared TF targets are enriched for reactive oxygen species-responsive genes, synaptic proteins and transcription regulators such as chromatin modifiers, including a significant number of highly constrained genes and known ASD genes. We next used single and double mutants to test whether mutating both TFs will modify transcriptional and behavioral phenotypes of single Gtf2ird1 mutants, though with the caveat that our Gtf2ird1 mutants, like others previously reported, do produce low levels of a truncated protein product. Despite little difference in DNA binding and transcriptome-wide expression, homozygous Gtf2ird1 mutation caused balance, marble burying and conditioned fear phenotypes. However, mutating Gtf2i in addition to Gtf2ird1 did not further modify transcriptomic or most behavioral phenotypes, suggesting Gtf2ird1 mutation alone was sufficient for the observed phenotypes.
Collapse
Affiliation(s)
- Nathan D Kopp
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA.,Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Kayla R Nygaard
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA.,Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Yating Liu
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA.,Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Katherine B McCullough
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA.,Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Susan E Maloney
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA.,Intellectual and Developmental Disabilities Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Harrison W Gabel
- Department of Neuroscience, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Joseph D Dougherty
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA.,Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA.,Intellectual and Developmental Disabilities Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
8
|
Kozel BA, Barak B, Ae Kim C, Mervis CB, Osborne LR, Porter M, Pober BR. Williams syndrome. Nat Rev Dis Primers 2021; 7:42. [PMID: 34140529 PMCID: PMC9437774 DOI: 10.1038/s41572-021-00276-z] [Citation(s) in RCA: 102] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/13/2021] [Indexed: 11/09/2022]
Abstract
Williams syndrome (WS) is a relatively rare microdeletion disorder that occurs in as many as 1:7,500 individuals. WS arises due to the mispairing of low-copy DNA repetitive elements at meiosis. The deletion size is similar across most individuals with WS and leads to the loss of one copy of 25-27 genes on chromosome 7q11.23. The resulting unique disorder affects multiple systems, with cardinal features including but not limited to cardiovascular disease (characteristically stenosis of the great arteries and most notably supravalvar aortic stenosis), a distinctive craniofacial appearance, and a specific cognitive and behavioural profile that includes intellectual disability and hypersociability. Genotype-phenotype evidence is strongest for ELN, the gene encoding elastin, which is responsible for the vascular and connective tissue features of WS, and for the transcription factor genes GTF2I and GTF2IRD1, which are known to affect intellectual ability, social functioning and anxiety. Mounting evidence also ascribes phenotypic consequences to the deletion of BAZ1B, LIMK1, STX1A and MLXIPL, but more work is needed to understand the mechanism by which these deletions contribute to clinical outcomes. The age of diagnosis has fallen in regions of the world where technological advances, such as chromosomal microarray, enable clinicians to make the diagnosis of WS without formally suspecting it, allowing earlier intervention by medical and developmental specialists. Phenotypic variability is considerable for all cardinal features of WS but the specific sources of this variability remain unknown. Further investigation to identify the factors responsible for these differences may lead to mechanism-based rather than symptom-based therapies and should therefore be a high research priority.
Collapse
Affiliation(s)
- Beth A. Kozel
- Translational Vascular Medicine Branch, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, USA
| | - Boaz Barak
- The Sagol School of Neuroscience and The School of Psychological Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Chong Ae Kim
- Department of Pediatrics, Universidade de São Paulo, São Paulo, Brazil
| | - Carolyn B. Mervis
- Department of Psychological and Brain Sciences, University of Louisville, Louisville, USA
| | - Lucy R. Osborne
- Department of Medicine, University of Toronto, Ontario, Canada
| | - Melanie Porter
- Department of Psychology, Macquarie University, Sydney, Australia
| | - Barbara R. Pober
- Department of Pediatrics, Massachusetts General Hospital and Harvard Medical School, Boston, USA
| |
Collapse
|
9
|
Spatiotemporal 7q11.23 protein network analysis implicates the role of DNA repair pathway during human brain development. Sci Rep 2021; 11:8246. [PMID: 33859276 PMCID: PMC8050238 DOI: 10.1038/s41598-021-87632-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 03/25/2021] [Indexed: 01/10/2023] Open
Abstract
Recurrent deletions and duplications of chromosome 7q11.23 copy number variants (CNVs) are associated with several psychiatric disorders. Although phenotypic abnormalities have been observed in patients, causal genes responsible for CNV-associated diagnoses and traits are still poorly understood. Furthermore, the targeted human brain regions, developmental stages, protein networks, and signaling pathways, influenced by this CNV remain unclear. Previous works showed GTF2I involved in Williams-Beuren syndrome, but pathways affected by GTF2I are indistinct. We first constructed dynamic spatiotemporal networks of 7q11.23 genes by combining data from the brain developmental transcriptome with physical interactions of 7q11.23 proteins. Topological changes were observed in protein-protein interaction (PPI) networks throughout different stages of brain development. Early and late fetal periods of development in the cortex, striatum, hippocampus, and amygdale were observed as the vital periods and regions for 7q11.23 CNV proteins. CNV proteins and their partners are significantly enriched in DNA repair pathway. As a driver gene, GTF2I interacted with PRKDC and BRCA1 to involve in DNA repair pathway. The physical interaction between GTF2I with PRKDC was confirmed experimentally by the liquid chromatography-tandem mass spectrometry (LC-MS/MS). We identified that early and late fetal periods are crucial for 7q11.23 genes to affect brain development. Our results implicate that 7q11.23 CNV genes converge on the DNA repair pathway to contribute to the pathogenesis of psychiatric diseases.
Collapse
|
10
|
Kopp N, McCullough K, Maloney SE, Dougherty JD. Gtf2i and Gtf2ird1 mutation do not account for the full phenotypic effect of the Williams syndrome critical region in mouse models. Hum Mol Genet 2020; 28:3443-3465. [PMID: 31418010 DOI: 10.1093/hmg/ddz176] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 06/04/2019] [Accepted: 06/27/2019] [Indexed: 12/31/2022] Open
Abstract
Williams syndrome (WS) is a neurodevelopmental disorder caused by a 1.5-1.8 Mbp deletion on chromosome 7q11.23, affecting the copy number of 26-28 genes. Phenotypes of WS include cardiovascular problems, craniofacial dysmorphology, deficits in visual-spatial cognition and a characteristic hypersocial personality. There are still no genes in the region that have been consistently linked to the cognitive and behavioral phenotypes, although human studies and mouse models have led to the current hypothesis that the general transcription factor 2 I family of genes, GTF2I and GTF2IRD1, are responsible. Here we test the hypothesis that these two transcription factors are sufficient to reproduce the phenotypes that are caused by deletion of the WS critical region (WSCR). We compare a new mouse model with loss of function mutations in both Gtf2i and Gtf2ird1 to an established mouse model lacking the complete WSCR. We show that the complete deletion (CD) model has deficits across several behavioral domains including social communication, motor functioning and conditioned fear that are not explained by loss of function mutations in Gtf2i and Gtf2ird1. Furthermore, transcriptome profiling of the hippocampus shows changes in synaptic genes in the CD model that are not seen in the double mutants. Thus, we have thoroughly defined a set of molecular and behavioral consequences of complete WSCR deletion and shown that genes or combinations of genes beyond Gtf2i and Gtf2ird1 are necessary to produce these phenotypic effects.
Collapse
Affiliation(s)
- Nathan Kopp
- Department of Genetics.,Department of Psychiatry
| | | | - Susan E Maloney
- Department of Psychiatry.,Intellectual and Developmental Disabilities Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Joseph D Dougherty
- Department of Genetics.,Department of Psychiatry.,Intellectual and Developmental Disabilities Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
11
|
Lew CH, Groeniger KM, Hanson KL, Cuevas D, Greiner DMZ, Hrvoj-Mihic B, Bellugi U, Schumann CM, Semendeferi K. Serotonergic innervation of the amygdala is increased in autism spectrum disorder and decreased in Williams syndrome. Mol Autism 2020; 11:12. [PMID: 32024554 PMCID: PMC7003328 DOI: 10.1186/s13229-019-0302-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 12/04/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Williams syndrome (WS) and autism spectrum disorder (ASD) are neurodevelopmental disorders that demonstrate overlapping genetic associations, dichotomous sociobehavioral phenotypes, and dichotomous pathological differences in neuronal distribution in key social brain areas, including the prefrontal cortex and the amygdala. The serotonergic system is critical to many processes underlying neurodevelopment and is additionally an important neuromodulator associated with behavioral variation. The amygdala is heavily innervated by serotonergic projections, suggesting that the serotonergic system is a significant mediator of neuronal activity. Disruptions to the serotonergic system, and atypical structure and function of the amygdala, are implicated in both WS and ASD. METHODS We quantified the serotonergic axon density in the four major subdivisions of the amygdala in the postmortem brains of individuals diagnosed with ASD and WS and neurotypical (NT) brains. RESULTS We found opposing directions of change in serotonergic innervation in the two disorders, with ASD displaying an increase in serotonergic axons compared to NT and WS displaying a decrease. Significant differences (p < 0.05) were observed between WS and ASD data sets across multiple amygdala nuclei. LIMITATIONS This study is limited by the availability of human postmortem tissue. Small sample size is an unavoidable limitation of most postmortem human brain research and particularly postmortem research in rare disorders. CONCLUSIONS Differential alterations to serotonergic innervation of the amygdala may contribute to differences in sociobehavioral phenotype in WS and ASD. These findings will inform future work identifying targets for future therapeutics in these and other disorders characterized by atypical social behavior.
Collapse
Affiliation(s)
- C H Lew
- Department of Anthropology, University of California, San Diego, USA
| | - K M Groeniger
- Department of Anthropology, University of California, San Diego, USA
| | - K L Hanson
- Department of Anthropology, University of California, San Diego, USA
| | - D Cuevas
- Department of Biological Sciences, University of California, San Diego, USA
| | - D M Z Greiner
- Department of Biological Sciences, University of California, San Diego, USA
| | - B Hrvoj-Mihic
- Department of Anthropology, University of California, San Diego, USA
| | - U Bellugi
- Salk Institute for Biological Sciences, San Diego, USA
| | - C M Schumann
- Department of Psychiatry and Behavioral Sciences, University of California, Davis School of Medicine, the MIND Institute, Sacramento, USA
| | - K Semendeferi
- Department of Anthropology, University of California, San Diego, USA.
| |
Collapse
|
12
|
Wu Y, Lin X, Lim IY, Chen L, Teh AL, MacIsaac JL, Tan KH, Kobor MS, Chong YS, Gluckman PD, Karnani N. Analysis of two birth tissues provides new insights into the epigenetic landscape of neonates born preterm. Clin Epigenetics 2019; 11:26. [PMID: 30744680 PMCID: PMC6371604 DOI: 10.1186/s13148-018-0599-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 12/17/2018] [Indexed: 01/04/2023] Open
Abstract
Background Preterm birth (PTB), defined as child birth before completion of 37 weeks of gestation, is a major challenge in perinatal health care and can bear long-term medical and financial burden. Over a million children die each year due to PTB complications, and those who survive can face developmental delays. Unfortunately, our understanding of the molecular pathways associated with PTB remains limited. There is a growing body of evidence suggesting the role of DNA methylation (DNAm) in mediating the effects of PTB on future health outcomes. Thus, epigenome-wide association studies (EWAS), where DNAm sites are examined for associations with PTB, can help shed light on the biological mechanisms linking the two. Results In an Asian cohort of 1019 infants (68 preterm, 951 full term), we examined and compared the associations between PTB and genome-wide DNAm profiles using both cord tissue (n = 1019) and cord blood (n = 332) samples on Infinium HumanMethylation450 arrays. PTB was significantly associated (P < 5.8e−7) with DNAm at 296 CpGs (209 genes) in the cord blood. Over 95% of these CpGs were replicated in other PTB/gestational age EWAS conducted in (cord) blood. This replication was apparent even across populations of different ethnic origin (Asians, Caucasians, and African Americans). More than a third of these 296 CpGs were replicated in at least 4 independent studies, thereby identifying a robust set of PTB-linked epigenetic signatures in cord blood. Interrogation of cord tissue in addition to cord blood provided novel insights into the epigenetic status of the neonates born preterm. Overall, 994 CpGs (608 genes, P < 3.7e−7) associated with PTB in cord tissue, of which only 10 of these CpGs were identified in the analysis using cord blood. Genes from cord tissue showed enrichment of molecular pathways related to fetal growth and development, while those from cord blood showed enrichment of immune response pathways. A substantial number of PTB-associated CpGs from both the birth tissues were also associated with gestational age. Conclusions Our findings provide insights into the epigenetic landscape of neonates born preterm, and that its status is captured more comprehensively by interrogation of more than one neonatal tissue in tandem. Both these neonatal tissues are clinically relevant in their unique ways and require careful consideration in identification of biomarkers related to PTB and gestational age. Trial registration This birth cohort is a prospective observational study designed to study the developmental origins of health and disease, and was retrospectively registered on 1 July 2010 under the identifier NCT01174875. Electronic supplementary material The online version of this article (10.1186/s13148-018-0599-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yonghui Wu
- Singapore Institute for Clinical Sciences, A*STAR, 30 Medical Drive, Singapore, 117609, Singapore
| | - Xinyi Lin
- Singapore Institute for Clinical Sciences, A*STAR, 30 Medical Drive, Singapore, 117609, Singapore.,Duke-NUS Medical School, Singapore, Singapore
| | - Ives Yubin Lim
- Singapore Institute for Clinical Sciences, A*STAR, 30 Medical Drive, Singapore, 117609, Singapore
| | - Li Chen
- Singapore Institute for Clinical Sciences, A*STAR, 30 Medical Drive, Singapore, 117609, Singapore
| | - Ai Ling Teh
- Singapore Institute for Clinical Sciences, A*STAR, 30 Medical Drive, Singapore, 117609, Singapore
| | - Julia L MacIsaac
- Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, University of British Columbia, Vancouver, Canada
| | - Kok Hian Tan
- KK Women's and Children's Hospital, Singapore, Singapore
| | - Michael S Kobor
- Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, University of British Columbia, Vancouver, Canada
| | - Yap Seng Chong
- Singapore Institute for Clinical Sciences, A*STAR, 30 Medical Drive, Singapore, 117609, Singapore.,Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Peter D Gluckman
- Singapore Institute for Clinical Sciences, A*STAR, 30 Medical Drive, Singapore, 117609, Singapore.,Centre for Human Evolution, Adaptation and Disease, Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Neerja Karnani
- Singapore Institute for Clinical Sciences, A*STAR, 30 Medical Drive, Singapore, 117609, Singapore. .,Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
13
|
Kopp ND, Parrish PCR, Lugo M, Dougherty JD, Kozel BA. Exome sequencing of 85 Williams-Beuren syndrome cases rules out coding variation as a major contributor to remaining variance in social behavior. Mol Genet Genomic Med 2018; 6:749-765. [PMID: 30008175 PMCID: PMC6160704 DOI: 10.1002/mgg3.429] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 05/03/2018] [Accepted: 06/11/2018] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Large, multigenic deletions at chromosome 7q11.23 result in a highly penetrant constellation of physical and behavioral symptoms known as Williams-Beuren syndrome (WS). Of particular interest is the unusual social-cognitive profile evidenced by deficits in social cognition and communication reminiscent of autism spectrum disorders (ASD) that are juxtaposed with normal or even relatively enhanced social motivation. Interestingly, duplications in the same region also result in ASD-like phenotypes as well as social phobias. Thus, the region clearly regulates human social motivation and behavior, yet the relevant gene(s) have not been definitively identified. METHOD Here, we deeply phenotyped 85 individuals with WS and used exome sequencing to analyze common and rare variation for association with the remaining variance in social behavior as assessed by the Social Responsiveness Scale. RESULTS We replicated the previously reported unusual juxtaposition of behavioral symptoms in this new patient collection, but we did not find any new alleles of large effect in the targeted analysis of the remaining copy of genes in the Williams syndrome critical region. However, we report on two nominally significant SNPs in two genes that have been implicated in the cognitive and social phenotypes of Williams syndrome, BAZ1B and GTF2IRD1. Secondary discovery driven explorations focusing on known ASD genes and an exome wide scan do not highlight any variants of a large effect. CONCLUSIONS Whole exome sequencing of 85 individuals with WS did not support the hypothesis that there are variants of large effect within the remaining Williams syndrome critical region that contribute to the social phenotype. This deeply phenotyped and genotyped patient cohort with a defined mutation provides the opportunity for similar analyses focusing on noncoding variation and/or other phenotypic domains.
Collapse
Affiliation(s)
- Nathan D. Kopp
- Department of GeneticsWashington University School of MedicineSt. LouisMissouri
| | - Phoebe C. R. Parrish
- National Heart Lung and Blood InstituteNational Institutes of HealthBethesdaMaryland
| | - Michael Lugo
- National Heart Lung and Blood InstituteNational Institutes of HealthBethesdaMaryland
- Department of PediatricsWashington University School of MedicineSt. LouisMissouri
| | - Joseph D. Dougherty
- Department of GeneticsWashington University School of MedicineSt. LouisMissouri
- Department of PsychiatryWashington University School of MedicineSt. LouisMissouri
| | - Beth A. Kozel
- National Heart Lung and Blood InstituteNational Institutes of HealthBethesdaMaryland
- Department of PediatricsWashington University School of MedicineSt. LouisMissouri
| |
Collapse
|
14
|
Chailangkarn T, Noree C, Muotri AR. The contribution of GTF2I haploinsufficiency to Williams syndrome. Mol Cell Probes 2018; 40:45-51. [PMID: 29305905 DOI: 10.1016/j.mcp.2017.12.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Revised: 12/19/2017] [Accepted: 12/28/2017] [Indexed: 01/14/2023]
Abstract
Williams syndrome (WS) is a neurodevelopmental disorder involving hemideletion of as many as 26-28 genes, resulting in a constellation of unique physical, cognitive and behavior phenotypes. The haploinsufficiency effect of each gene has been studied and correlated with phenotype(s) using several models including WS subjects, animal models, and peripheral cell lines. However, links for most of the genes to WS phenotypes remains unclear. Among those genes, general transcription factor 2I (GTF2I) is of particular interest as its haploinsufficiency is possibly associated with hypersociability in WS. Here, we describe studies of atypical WS cases as well as mouse models focusing on GTF2I that support a role for this protein in the neurocognitive and behavioral profiles of WS individuals. We also review collective studies on diverse molecular functions of GTF2I that may provide mechanistic explanation for phenotypes recently reported in our relevant cellular model, namely WS induced pluripotent stem cell (iPSC)-derived neurons. Finally, in light of the progress in gene-manipulating approaches, we suggest their uses in revealing the neural functions of GTF2I in the context of WS.
Collapse
Affiliation(s)
- Thanathom Chailangkarn
- National Center for Genetic Engineering and Biotechnology (BIOTEC), Virology and Cell Technology Laboratory, Pathum Thani, 12120, Thailand.
| | - Chalongrat Noree
- Institute of Molecular Biosciences, Mahidol University, 25/25 Phuttamonthon 4 Road, Salaya, Phuttamonthon, Nakhon Pathom, 73170, Thailand
| | - Alysson R Muotri
- University of California San Diego, School of Medicine, UCSD Stem Cell Program, Department of Pediatrics/Rady Children's Hospital San Diego, La Jolla, CA 92037, USA; University of California San Diego, School of Medicine, Department of Cellular & Molecular Medicine, La Jolla, CA 92037, USA; Center for Academic Research and Training in Anthropogeny (CARTA), La Jolla, CA 92093, USA
| |
Collapse
|
15
|
Kim SJ, Ka S, Ha JW, Kim J, Yoo D, Kim K, Lee HK, Lim D, Cho S, Hanotte O, Mwai OA, Dessie T, Kemp S, Oh SJ, Kim H. Cattle genome-wide analysis reveals genetic signatures in trypanotolerant N'Dama. BMC Genomics 2017; 18:371. [PMID: 28499406 PMCID: PMC5427609 DOI: 10.1186/s12864-017-3742-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 04/27/2017] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Indigenous cattle in Africa have adapted to various local environments to acquire superior phenotypes that enhance their survival under harsh conditions. While many studies investigated the adaptation of overall African cattle, genetic characteristics of each breed have been poorly studied. RESULTS We performed the comparative genome-wide analysis to assess evidence for subspeciation within species at the genetic level in trypanotolerant N'Dama cattle. We analysed genetic variation patterns in N'Dama from the genomes of 101 cattle breeds including 48 samples of five indigenous African cattle breeds and 53 samples of various commercial breeds. Analysis of SNP variances between cattle breeds using wMI, XP-CLR, and XP-EHH detected genes containing N'Dama-specific genetic variants and their potential associations. Functional annotation analysis revealed that these genes are associated with ossification, neurological and immune system. Particularly, the genes involved in bone formation indicate that local adaptation of N'Dama may engage in skeletal growth as well as immune systems. CONCLUSIONS Our results imply that N'Dama might have acquired distinct genotypes associated with growth and regulation of regional diseases including trypanosomiasis. Moreover, this study offers significant insights into identifying genetic signatures for natural and artificial selection of diverse African cattle breeds.
Collapse
Affiliation(s)
- Soo-Jin Kim
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea.,C&K Genomics, Seoul National University Research Park, Seoul, 151-919, Republic of Korea
| | - Sojeong Ka
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jung-Woo Ha
- Clova, NAVER Corp., Seongnam, 13561, Republic of Korea
| | - Jaemin Kim
- C&K Genomics, Seoul National University Research Park, Seoul, 151-919, Republic of Korea
| | - DongAhn Yoo
- C&K Genomics, Seoul National University Research Park, Seoul, 151-919, Republic of Korea.,Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, 08826, Republic of Korea
| | - Kwondo Kim
- C&K Genomics, Seoul National University Research Park, Seoul, 151-919, Republic of Korea.,Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hak-Kyo Lee
- Department of Animal Biotechnology, Chonbuk National University, Jeonju, 66414, Republic of Korea
| | - Dajeong Lim
- Division of Animal Genomics and Bioinformatics, National Institute of Animal Science, RDA, Jeonju, 55365, Republic of Korea
| | - Seoae Cho
- C&K Genomics, Seoul National University Research Park, Seoul, 151-919, Republic of Korea
| | - Olivier Hanotte
- University of Nottingham, School of Life Sciences, Nottingham, NG7 2RD, UK.,International Livestock Research Institute, Addis Ababa, Ethiopia
| | - Okeyo Ally Mwai
- International Livestock Research Institute, Box 30709-00100, Nairobi, Kenya
| | - Tadelle Dessie
- International Livestock Research Institute, Addis Ababa, Ethiopia
| | - Stephen Kemp
- International Livestock Research Institute, Box 30709-00100, Nairobi, Kenya.,The Centre for Tropical Livestock Genetics and Health, The Roslin Institute, University of Edinburgh, Easter Bush Campus, Edinburgh, Scotland, UK
| | - Sung Jong Oh
- National Institute of Animal Science, RDA, Wanju, 55365, Republic of Korea.
| | - Heebal Kim
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea. .,C&K Genomics, Seoul National University Research Park, Seoul, 151-919, Republic of Korea. .,Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
16
|
Chailangkarn T, Muotri AR. Modeling Williams syndrome with induced pluripotent stem cells. NEUROGENESIS (AUSTIN, TEX.) 2017; 4:e1283187. [PMID: 28229087 PMCID: PMC5305168 DOI: 10.1080/23262133.2017.1283187] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 12/01/2016] [Accepted: 01/10/2017] [Indexed: 12/20/2022]
Abstract
The development of induced pluripotent stem cells (iPSCs) like never before has opened novel opportunity to study diseases in relevant cell types. In our recent study, Williams syndrome (WS), a rare genetic neurodevelopmental disorder, that is caused by hemizygous deletion of 25-28 genes on chromosome 7, is of interest because of its unique cognitive and social profiles. Little is known about haploinsufficiency effect of those deleted genes on molecular and cellular phenotypes at the neural level due to the lack of relevant human cellular model. Using the cellular reprogramming approach, we reported that WS iPSC-derived neural progenitor cells (NPCs) has increased apoptosis and therefore increased doubling time, which could be rescued by complementation of frizzled 9, one of the genes typically deleted in WS. Moreover, WS iPSC-derived CTIP2-positive pyramidal neurons exhibit morphologic alterations including longer total dendrites and increasing dendritic spine number. In addition, WS iPSC-derived neurons show an increase in calcium transient frequency and synchronized activity likely due to increased number of dendritic spines and synapses. Our work integrated cross-level data from genetics to behavior of WS individuals and revealed altered cellular phenotypes in WS human NPCs and neurons that could be validated in other model systems such as magnetic resonance imaging (MRI) in live subjects and postmortem brain tissues.
Collapse
Affiliation(s)
- Thanathom Chailangkarn
- University of California San Diego, School of Medicine, Department of Pediatrics/Rady Children's Hospital San Diego, Department of Cellular & Molecular Medicine, Stem Cell Program, Center for Academic Research and Training in Anthropogeny (CARTA), La Jolla, CA, USA
- National Center for Genetic Engineering and Biotechnology (BIOTEC), Virology and Cell Technology Laboratory, Pathum Thani, Thailand
| | - Alysson R. Muotri
- University of California San Diego, School of Medicine, Department of Pediatrics/Rady Children's Hospital San Diego, Department of Cellular & Molecular Medicine, Stem Cell Program, Center for Academic Research and Training in Anthropogeny (CARTA), La Jolla, CA, USA
| |
Collapse
|
17
|
Burggraaf R, Frens MA, Hooge ITC, van der Geest JN. Performance on tasks of visuospatial memory and ability: A cross-sectional study in 330 adolescents aged 11 to 20. APPLIED NEUROPSYCHOLOGY-CHILD 2017; 7:129-142. [PMID: 28075186 DOI: 10.1080/21622965.2016.1268960] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Cognitive functions mature at different points in time between birth and adulthood. Of these functions, visuospatial skills, such as spatial memory and part-to-whole organization, have often been tested in children and adults but have been less frequently evaluated during adolescence. We studied visuospatial memory and ability during this critical developmental period, as well as the correlation between these abilities, in a large group of 330 participants (aged 11 to 20 years, 55% male). To assess visuospatial memory, the participants were asked to memorize and reproduce sequences of random locations within a grid using a computer. Visuospatial ability was tested using a variation of the Design Organization Test (DOT). In this paper-and-pencil test, the participants had one minute to reproduce as many visual patterns as possible using a numerical code. On the memory task, compared with younger participants, older participants correctly reproduced more locations overall and longer sequences of locations, made fewer mistakes and needed less time to reproduce the sequences. In the visuospatial ability task, the number of correctly reproduced patterns increased with age. We show that both visuospatial memory and ability improve significantly throughout adolescence and that performance on both tasks is significantly correlated.
Collapse
Affiliation(s)
- Rudolf Burggraaf
- a Department of Neuroscience , Erasmus MC , Rotterdam , The Netherlands.,b Department of Experimental Psychology , Utrecht University , Utrecht , The Netherlands
| | - Maarten A Frens
- a Department of Neuroscience , Erasmus MC , Rotterdam , The Netherlands.,c Erasmus University College , Erasmus University , Rotterdam , The Netherlands
| | - Ignace T C Hooge
- b Department of Experimental Psychology , Utrecht University , Utrecht , The Netherlands
| | | |
Collapse
|
18
|
Barak B, Feng G. Neurobiology of social behavior abnormalities in autism and Williams syndrome. Nat Neurosci 2016; 19:647-655. [PMID: 29323671 PMCID: PMC4896837 DOI: 10.1038/nn.4276] [Citation(s) in RCA: 148] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2015] [Accepted: 02/22/2016] [Indexed: 12/14/2022]
Abstract
Social behavior is a basic behavior mediated by multiple brain regions and neural circuits, and is crucial for the survival and development of animals and humans. Two neuropsychiatric disorders that have prominent social behavior abnormalities are autism spectrum disorders (ASD), which is characterized mainly by hyposociability, and Williams syndrome (WS), whose subjects exhibit hypersociability. Here we review the unique properties of social behavior in ASD and WS, and discuss the major theories in social behavior in the context of these disorders. We conclude with a discussion of the research questions needing further exploration to enhance our understanding of social behavior abnormalities.
Collapse
Affiliation(s)
- Boaz Barak
- McGovern Institute for Brain Research, MIT, Cambridge, Massachusetts, USA
- Department of Brain &Cognitive Sciences, MIT, Cambridge, Massachusetts, USA
| | - Guoping Feng
- McGovern Institute for Brain Research, MIT, Cambridge, Massachusetts, USA
- Department of Brain &Cognitive Sciences, MIT, Cambridge, Massachusetts, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| |
Collapse
|
19
|
Dutra RL, Piazzon FB, Zanardo ÉA, Costa TVMM, Montenegro MM, Novo-Filho GM, Dias AT, Nascimento AM, Kim CA, Kulikowski LD. Rare genomic rearrangement in a boy with Williams-Beuren syndrome associated to XYY syndrome and intriguing behavior. Am J Med Genet A 2015; 167A:3197-203. [PMID: 26420477 DOI: 10.1002/ajmg.a.37360] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 08/17/2015] [Indexed: 12/27/2022]
Abstract
Williams-Beuren syndrome (WBS) is caused by a hemizygous contiguous gene microdeletion of 1.55-1.84 Mb at 7q11.23 region. Approximately, 28 genes have been shown to contribute to classical phenotype of SWB with presence of dysmorphic facial features, supravalvular aortic stenosis (SVAS), intellectual disability, and overfriendliness. With the use of Microarray-based comparative genomic hybridization and other molecular cytogenetic techniques, is possible define with more accuracy partial or atypical deletion and refine the genotype-phenotype correlation. Here, we report on a rare genomic structural rearrangement in a boy with atypical deletion in 7q11.23 and XYY syndrome with characteristic clinical signs, but not sufficient for the diagnosis of WBS. Cytogenetic analysis of G-banding showed a karyotype 47,XYY. Analysis of DNA with the technique of MLPA (Multiplex Ligation-dependent Probe Amplification) using kits a combination of kits (P064, P036, P070, and P029) identified an atypical deletion on 7q11.23. In addition, high resolution SNP Oligonucleotide Microarray Analysis (SNP-array) confirmed the alterations found by MLPA and revealed others pathogenic CNVs, in the chromosomes 7 and X. The present report demonstrates an association not yet described in literature, between Williams-Beuren syndrome and 47,XYY. The identification of atypical deletion in 7q11.23 concomitant to additional pathogenic CNVs in others genomic regions allows a better comprehension of clinical consequences of atypical genomic rearrangements.
Collapse
Affiliation(s)
- Roberta L Dutra
- Genetics Unit, Instituto da Criança, Faculdade de Medicina da Universidade de São Paulo (USP), São Paulo, Brazil.,Department of Pathology, Cytogenomics Lab - LIM03, Faculdade de Medicina da Universidade de São Paulo (USP), São Paulo, Brazil
| | - Flavia B Piazzon
- Department of Pathology, Cytogenomics Lab - LIM03, Faculdade de Medicina da Universidade de São Paulo (USP), São Paulo, Brazil
| | - Évelin A Zanardo
- Department of Pathology, Cytogenomics Lab - LIM03, Faculdade de Medicina da Universidade de São Paulo (USP), São Paulo, Brazil
| | | | - Marília M Montenegro
- Genetics Unit, Instituto da Criança, Faculdade de Medicina da Universidade de São Paulo (USP), São Paulo, Brazil.,Department of Pathology, Cytogenomics Lab - LIM03, Faculdade de Medicina da Universidade de São Paulo (USP), São Paulo, Brazil
| | - Gil M Novo-Filho
- Genetics Unit, Instituto da Criança, Faculdade de Medicina da Universidade de São Paulo (USP), São Paulo, Brazil.,Department of Pathology, Cytogenomics Lab - LIM03, Faculdade de Medicina da Universidade de São Paulo (USP), São Paulo, Brazil
| | - Alexandre T Dias
- Department of Pathology, Cytogenomics Lab - LIM03, Faculdade de Medicina da Universidade de São Paulo (USP), São Paulo, Brazil
| | - Amom M Nascimento
- Genetics Unit, Instituto da Criança, Faculdade de Medicina da Universidade de São Paulo (USP), São Paulo, Brazil.,Department of Pathology, Cytogenomics Lab - LIM03, Faculdade de Medicina da Universidade de São Paulo (USP), São Paulo, Brazil
| | - Chong Ae Kim
- Genetics Unit, Instituto da Criança, Faculdade de Medicina da Universidade de São Paulo (USP), São Paulo, Brazil
| | - Leslie D Kulikowski
- Department of Pathology, Cytogenomics Lab - LIM03, Faculdade de Medicina da Universidade de São Paulo (USP), São Paulo, Brazil.,Department of Collective Health - Human Reproduction and Genetics Center, Faculdade de Medicina do ABC, Santo André, São Paulo, Brazil
| |
Collapse
|
20
|
Sharma P, Gupta N, Chowdhury MR, Phadke SR, Sapra S, Halder A, Ghosh M, Kabra M. Williams-Beuren Syndrome: Experience of 43 Patients and a Report of an Atypical Case from a Tertiary Care Center in India. Cytogenet Genome Res 2015; 146:187-94. [PMID: 26352091 DOI: 10.1159/000439205] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/07/2015] [Indexed: 11/19/2022] Open
Abstract
Williams-Beuren syndrome (WBS) or Williams syndrome (OMIM 194050) is a multisystem disorder manifested by neurodevelopmental delay and is caused by a hemizygous deletion of ∼ 1.5-1.8 Mb in the 7q11.23 region. Clinical features include cardiovascular anomalies (mainly supravalvular aortic stenosis), peripheral pulmonary stenosis, distinctive facies, intellectual disability (usually mild), unique personality characteristics, and growth and endocrine abnormalities. Clinical diagnostic criteria are available for WBS; however, the mainstay of diagnosis is the detection of the contiguous gene deletion. Although FISH remains the most widely used laboratory test, the diagnosis can also be established by means of qPCR, MLPA, microsatellite marker analysis, and chromosomal microarray (CMA). We evaluated the utility of MLPA to detect deletion/duplication in the 7q11.23 region in 43 patients suspected to have WBS using MLPA kits for microdeletion syndromes. A hemizygous deletion in the 7q11.23 region was found in 41 (95.3%) patients using MLPA. One patient had an atypical deletion detected by CMA. During the initial period of this study, the results of 12 patients tested by MLPA were also confirmed by FISH. Compared to FISH and CMA, MLPA is a cheaper, high-throughput, less labor-intensive and less time-consuming technique for the diagnosis of WBS. Although CMA is expensive and labor-intensive, its effectiveness is demonstrated to detect an atypical deletion and to delineate the breakpoints.
Collapse
Affiliation(s)
- Pankaj Sharma
- Division of Genetics, Department of Pediatrics, All India Institute of Medical Sciences, New Delhi, India
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Borralleras C, Sahun I, Pérez-Jurado LA, Campuzano V. Intracisternal Gtf2i Gene Therapy Ameliorates Deficits in Cognition and Synaptic Plasticity of a Mouse Model of Williams-Beuren Syndrome. Mol Ther 2015. [PMID: 26216516 DOI: 10.1038/mt.2015.130] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Williams-Beuren syndrome (WBS) is a neurodevelopmental disorder caused by a heterozygous deletion of 26-28 genes at chromosome band 7q11.23. Haploinsufficiency at GTF2I has been shown to play a major role in the neurobehavioral phenotype. By characterizing the neuronal architecture in four animal models with intragenic, partial, and complete deletions of the WBS critical interval (ΔGtf2i(+/-), ΔGtf2i( -/-), PD, and CD), we clarify the involvement of Gtf2i in neurocognitive features. All mutant mice showed hypersociability, impaired motor learning and coordination, and altered anxiety-like behavior. Dendritic length was decreased in the CA1 of ΔGtf2i(+/-), ΔGtf2i ( -/-), and CD mice. Spine density was reduced, and spines were shorter in ΔGtf2i ( -/-), PD, and CD mice. Overexpression of Pik3r1 and downregulation of Bdnf were observed in ΔGtf2i(+/-), PD, and CD mice. Intracisternal Gtf2i-gene therapy in CD mice using adeno-associated virus resulted in increased mGtf2i expression and normalization of Bdnf levels, along with beneficial effects in motor coordination, sociability, and anxiety, despite no significant changes in neuronal architecture. Our findings further indicate that Gtf2i haploinsufficiency plays an important role in the neurodevelopmental and cognitive abnormalities of WBS and that it is possible to rescue part of this neurocognitive phenotype by restoring Gtf2i expression levels in specific brain areas.
Collapse
Affiliation(s)
- Cristina Borralleras
- Neurosciences Program, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Unitat de Genètica, Barcelona, Spain; Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Spain
| | - Ignasi Sahun
- PCB-PRBB Animal Facility Alliance, Barcelona, Spain
| | - Luis A Pérez-Jurado
- Neurosciences Program, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Unitat de Genètica, Barcelona, Spain; Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Spain
| | - Victoria Campuzano
- Neurosciences Program, Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Unitat de Genètica, Barcelona, Spain; Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Spain.
| |
Collapse
|
22
|
Tekendo-Ngongang C, Dahoun S, Nguefack S, Gimelli S, Sloan-Béna F, Wonkam A. Challenges in clinical diagnosis of williams-beuren syndrome in sub-saharan africans: case reports from cameroon. Mol Syndromol 2014; 5:287-92. [PMID: 25565928 DOI: 10.1159/000369421] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/11/2014] [Indexed: 12/25/2022] Open
Abstract
Williams-Beuren syndrome (WBS) is a rare neurodevelopmental condition caused by a recurrent chromosomal microdeletion involving about 28 contiguous genes at 7q11.23. Most patients display a specific congenital heart defect, characteristic facial features, a particular behavior, and intellectual disability. Cases from sub-Saharan Africa have been seldom reported. The present study describes 3 Cameroonian patients affected by WBS, aged 19 months, 13 and 14 years, in whom the diagnosis was confirmed by fluorescent in situ hybridization (FISH) and comparative genomic hybridization (CGH). The first patient presented with a congenital heart defect, the second and third with learning difficulties as well as developmental and behavioral issues. In the latter 2 cases, the facial phenotypes were similar to those of the unaffected population with the same ethnic background. However, the cardiovascular anomalies and friendly behavioral attitudes led to suspicion of WBS. FISH revealed the deletion of the WBS critical region in the first patient, and array-CGH detected a heterozygous ∼1.4-Mb deletion in the 7q11.23 region in the second and third patient. This preliminary report suggests that for sub-Saharan Africans clinical suspicion of WBS could be mostly based on behavioral phenotype and structural heart defects, and less on the classical facial dysmorphic signs.
Collapse
Affiliation(s)
- Cedrik Tekendo-Ngongang
- Faculty of Medicine and Biomedical Sciences, University of Yaoundé I, Yaoundé, Cameroon ; Division of Human Genetics, University of Cape Town, Cape Town, South Africa
| | - Sophie Dahoun
- Service of Medical Genetics, Geneva University Hospitals, Geneva, Switzerland
| | - Seraphin Nguefack
- Faculty of Medicine and Biomedical Sciences, University of Yaoundé I, Yaoundé, Cameroon
| | - Stefania Gimelli
- Service of Medical Genetics, Geneva University Hospitals, Geneva, Switzerland
| | | | - Ambroise Wonkam
- Faculty of Medicine and Biomedical Sciences, University of Yaoundé I, Yaoundé, Cameroon ; Division of Human Genetics, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
23
|
Euteneuer J, Carvalho CMB, Kulkarni S, Vineyard M, Grady RM, Lupski JR, Shinawi M. Molecular and phenotypic characterization of atypical Williams-Beuren syndrome. Clin Genet 2013; 86:487-91. [PMID: 24246242 DOI: 10.1111/cge.12305] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2013] [Revised: 10/08/2013] [Accepted: 10/16/2013] [Indexed: 11/29/2022]
Abstract
Williams-Beuren syndrome (WBS) is a multisystemic genomic disorder typically caused by a recurrent ˜1.5-1.8 Mb deletion on 7q11.23. Atypical deletions can provide important insight into the genotype-phenotype correlations. Here, we report the phenotypic and molecular characterization of a girl with a de novo 81.8 kb deletion in the WBS critical region, which involves the ELN and LIMK1 genes only. The patient presented at 2 months of age with extensive vascular abnormalities, mild facial dysmorphism and delays in her fine motor skills. We discuss potential molecular mechanisms and the role of ELN and LIMK1 in the different phenotypic features. We compare the findings in our patient with previously reported overlapping deletions. The phenotypic variability among these patients suggests that other factors are important in the phenotype and possibly include: position effects related to copy number variation size, variations in the non-deleted alleles, genetic modifiers elsewhere in the genome, or reduced penetrance for specific phenotypes.
Collapse
Affiliation(s)
- J Euteneuer
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | | | | | | | | | | | | |
Collapse
|
24
|
Smaller and larger deletions of the Williams Beuren syndrome region implicate genes involved in mild facial phenotype, epilepsy and autistic traits. Eur J Hum Genet 2013; 22:64-70. [PMID: 23756441 DOI: 10.1038/ejhg.2013.101] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Revised: 03/06/2013] [Accepted: 04/10/2013] [Indexed: 11/08/2022] Open
Abstract
Williams Beuren syndrome (WBS) is a multisystemic disorder caused by a hemizygous deletion of 1.5 Mb on chromosome 7q11.23 spanning 28 genes. A few patients with larger and smaller WBS deletion have been reported. They show clinical features that vary between isolated SVAS to the full spectrum of WBS phenotype, associated with epilepsy or autism spectrum behavior. Here we describe four patients with atypical WBS 7q11.23 deletions. Two carry ~3.5 Mb larger deletion towards the telomere that includes Huntingtin-interacting protein 1 (HIP1) and tyrosine 3-monooxygenase/tryptophan 5-monooxigenase activation protein gamma (YWHAG) genes. Other two carry a shorter deletion of ~1.2 Mb at centromeric side that excludes the distal WBS genes BAZ1B and FZD9. Along with previously reported cases, genotype-phenotype correlation in the patients described here further suggests that haploinsufficiency of HIP1 and YWHAG might cause the severe neurological and neuropsychological deficits including epilepsy and autistic traits, and that the preservation of BAZ1B and FZD9 genes may be related to mild facial features and moderate neuropsychological deficits. This report highlights the importance to characterize additional patients with 7q11.23 atypical deletions comparing neuropsychological and clinical features between these individuals to shed light on the pathogenic role of genes within and flanking the WBS region.
Collapse
|
25
|
Zafeiriou DI, Ververi A, Dafoulis V, Kalyva E, Vargiami E. Autism spectrum disorders: the quest for genetic syndromes. Am J Med Genet B Neuropsychiatr Genet 2013; 162B:327-66. [PMID: 23650212 DOI: 10.1002/ajmg.b.32152] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2011] [Accepted: 03/01/2013] [Indexed: 11/10/2022]
Abstract
Autism spectrum disorders (ASD) are a heterogeneous group of neurodevelopmental disabilities with various etiologies, but with a heritability estimate of more than 90%. Although the strong correlation between autism and genetic factors has been long established, the exact genetic background of ASD remains unclear. A number of genetic syndromes manifest ASD at higher than expected frequencies compared to the general population. These syndromes account for more than 10% of all ASD cases and include tuberous sclerosis, fragile X, Down, neurofibromatosis, Angelman, Prader-Willi, Williams, Duchenne, etc. Clinicians are increasingly required to recognize genetic disorders in individuals with ASD, in terms of providing proper care and prognosis to the patient, as well as genetic counseling to the family. Vice versa, it is equally essential to identify ASD in patients with genetic syndromes, in order to ensure correct management and appropriate educational placement. During investigation of genetic syndromes, a number of issues emerge: impact of intellectual disability in ASD diagnoses, identification of autistic subphenotypes and differences from idiopathic autism, validity of assessment tools designed for idiopathic autism, possible mechanisms for the association with ASD, etc. Findings from the study of genetic syndromes are incorporated into the ongoing research on autism etiology and pathogenesis; different syndromes converge upon common biological backgrounds (such as disrupted molecular pathways and brain circuitries), which probably account for their comorbidity with autism. This review paper critically examines the prevalence and characteristics of the main genetic syndromes, as well as the possible mechanisms for their association with ASD.
Collapse
|
26
|
Atypical deletion in Williams-Beuren syndrome critical region detected by MLPA in a patient with supravalvular aortic stenosis and learning difficulty. J Genet Genomics 2012; 39:571-4. [PMID: 23089367 DOI: 10.1016/j.jgg.2012.07.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2012] [Revised: 06/23/2012] [Accepted: 07/09/2012] [Indexed: 12/25/2022]
|
27
|
Vandeweyer G, Van der Aa N, Reyniers E, Kooy RF. The contribution of CLIP2 haploinsufficiency to the clinical manifestations of the Williams-Beuren syndrome. Am J Hum Genet 2012; 90:1071-8. [PMID: 22608712 DOI: 10.1016/j.ajhg.2012.04.020] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2011] [Revised: 03/16/2012] [Accepted: 04/10/2012] [Indexed: 11/16/2022] Open
Abstract
Williams-Beuren syndrome is a rare contiguous gene syndrome, characterized by intellectual disability, facial dysmorphisms, connective-tissue abnormalities, cardiac defects, structural brain abnormalities, and transient infantile hypercalcemia. Genes lying telomeric to RFC2, including CLIP2, GTF2I and GTF2IRD1, are currently thought to be the most likely major contributors to the typical Williams syndrome cognitive profile, characterized by a better-than-expected auditory rote-memory ability, a relative sparing of language capabilities, and a severe visual-spatial constructive impairment. Atypical deletions in the region have helped to establish genotype-phenotype correlations. So far, however, hardly any deletions affecting only a single gene in the disease region have been described. We present here two healthy siblings with a pure, hemizygous deletion of CLIP2. A putative role in the cognitive and behavioral abnormalities seen in Williams-Beuren patients has been suggested for this gene on the basis of observations in a knock-out mouse model. The presented siblings did not show any of the clinical features associated with the syndrome. Cognitive testing showed an average IQ for both and no indication of the Williams syndrome cognitive profile. This shows that CLIP2 haploinsufficiency by itself does not lead to the physical or cognitive characteristics of the Williams-Beuren syndrome, nor does it lead to the Williams syndrome cognitive profile. Although contribution of CLIP2 to the phenotype cannot be excluded when it is deleted in combination with other genes, our results support the hypothesis that GTF2IRD1 and GTF2I are the main genes causing the cognitive defects associated with Williams-Beuren syndrome.
Collapse
Affiliation(s)
- Geert Vandeweyer
- Department of Medical Genetics, University Hospital of Antwerp, University of Antwerp, Edegem, Belgium
| | | | | | | |
Collapse
|
28
|
Mutation of Gtf2ird1 from the Williams-Beuren syndrome critical region results in facial dysplasia, motor dysfunction, and altered vocalisations. Neurobiol Dis 2011; 45:913-22. [PMID: 22198572 DOI: 10.1016/j.nbd.2011.12.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2011] [Revised: 11/10/2011] [Accepted: 12/04/2011] [Indexed: 01/09/2023] Open
Abstract
Insufficiency of the transcriptional regulator GTF2IRD1 has become a strong potential explanation for some of the major characteristic features of the neurodevelopmental disorder Williams-Beuren syndrome (WBS). Genotype/phenotype correlations in humans indicate that the hemizygous loss of the GTF2IRD1 gene and an adjacent paralogue, GTF2I, play crucial roles in the neurocognitive and craniofacial aspects of the disease. In order to explore this genetic relationship in greater detail, we have generated a targeted Gtf2ird1 mutation in mice that blocks normal GTF2IRD1 protein production. Detailed analyses of homozygous null Gtf2ird1 mice have revealed a series of phenotypes that share some intriguing parallels with WBS. These include reduced body weight, a facial deformity resulting from localised epidermal hyperplasia, a motor coordination deficit, alterations in exploratory activity and, in response to specific stress-inducing stimuli; a novel audible vocalisation and increased serum corticosterone. Analysis of Gtf2ird1 expression patterns in the brain using a knock-in LacZ reporter and c-fos activity mapping illustrates the regions where these neurological abnormalities may originate. These data provide new mechanistic insight into the clinical genetic findings in WBS patients and indicate that insufficiency of GTF2IRD1 protein contributes to abnormalities of facial development, motor function and specific behavioural disorders that accompany this disease.
Collapse
|
29
|
Mason TBA, Arens R, Sharman J, Bintliff-Janisak B, Schultz B, Walters AS, Cater JR, Kaplan P, Pack AI. Sleep in children with Williams Syndrome. Sleep Med 2011; 12:892-7. [PMID: 21940205 PMCID: PMC3210863 DOI: 10.1016/j.sleep.2011.05.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2011] [Revised: 05/21/2011] [Accepted: 05/22/2011] [Indexed: 10/17/2022]
Abstract
OBJECTIVE To analyze sleep in children with Williams Syndrome (WS) compared to normal healthy controls in order to determine whether particular sleep features are characteristic of WS, and to explore associations between disturbed sleep and behavior. METHODS Thirty-five children with genetically-confirmed WS and 35 matched controls underwent overnight polysomnography and performance testing in the Sleep Center at the Children's Hospital of Philadelphia. Parents completed questionnaires regarding the subjects' sleep and behavior. RESULTS WS subjects had significantly different sleep than controls, with decreased sleep efficiency, increased respiratory-related arousals and increased slow wave sleep on overnight polysomnography. WS subjects were also noted to have more difficulty falling asleep, with greater restlessness and more arousals from sleep than controls. Fifty-two percent of WS subjects had features of attention deficit-hyperactivity disorder. CONCLUSION Children with WS had significantly different sleep than controls in our sample. These differences demonstrated in our study may reflect genetic influences on sleep.
Collapse
Affiliation(s)
- Thornton B A Mason
- Sleep Center, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Henrichsen CN, Csárdi G, Zabot MT, Fusco C, Bergmann S, Merla G, Reymond A. Using transcription modules to identify expression clusters perturbed in Williams-Beuren syndrome. PLoS Comput Biol 2011; 7:e1001054. [PMID: 21304579 PMCID: PMC3024257 DOI: 10.1371/journal.pcbi.1001054] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2010] [Accepted: 12/07/2010] [Indexed: 11/19/2022] Open
Abstract
The genetic dissection of the phenotypes associated with Williams-Beuren Syndrome (WBS) is advancing thanks to the study of individuals carrying typical or atypical structural rearrangements, as well as in vitro and animal studies. However, little is known about the global dysregulations caused by the WBS deletion. We profiled the transcriptomes of skin fibroblasts from WBS patients and compared them to matched controls. We identified 868 differentially expressed genes that were significantly enriched in extracellular matrix genes, major histocompatibility complex (MHC) genes, as well as genes in which the products localize to the postsynaptic membrane. We then used public expression datasets from human fibroblasts to establish transcription modules, sets of genes coexpressed in this cell type. We identified those sets in which the average gene expression was altered in WBS samples. Dysregulated modules are often interconnected and share multiple common genes, suggesting that intricate regulatory networks connected by a few central genes are disturbed in WBS. This modular approach increases the power to identify pathways dysregulated in WBS patients, thus providing a testable set of additional candidates for genes and their interactions that modulate the WBS phenotypes.
Collapse
Affiliation(s)
| | - Gábor Csárdi
- Department of Medical Genetics, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Marie-Thérèse Zabot
- Centre de Biotechnologie Cellulaire, Hospices Civils de Lyon, Groupement Hospitalier Est, Bron, France
| | - Carmela Fusco
- Laboratory of Medical Genetics, IRCCS- Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Sven Bergmann
- Department of Medical Genetics, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- * E-mail: (AR); (GM); (SB)
| | - Giuseppe Merla
- Laboratory of Medical Genetics, IRCCS- Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
- * E-mail: (AR); (GM); (SB)
| | - Alexandre Reymond
- The Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
- * E-mail: (AR); (GM); (SB)
| |
Collapse
|
31
|
Sakurai T, Dorr NP, Takahashi N, McInnes LA, Elder GA, Buxbaum JD. Haploinsufficiency of Gtf2i, a gene deleted in Williams Syndrome, leads to increases in social interactions. Autism Res 2010; 4:28-39. [DOI: 10.1002/aur.169] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2010] [Accepted: 10/12/2010] [Indexed: 11/10/2022]
|
32
|
Merla G, Brunetti-Pierri N, Micale L, Fusco C. Copy number variants at Williams–Beuren syndrome 7q11.23 region. Hum Genet 2010; 128:3-26. [DOI: 10.1007/s00439-010-0827-2] [Citation(s) in RCA: 112] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2010] [Accepted: 04/13/2010] [Indexed: 01/06/2023]
|
33
|
Lucena J, Pezzi S, Aso E, Valero MC, Carreiro C, Dubus P, Sampaio A, Segura M, Barthelemy I, Zindel MY, Sousa N, Barbero JL, Maldonado R, Pérez-Jurado LA, Campuzano V. Essential role of the N-terminal region of TFII-I in viability and behavior. BMC MEDICAL GENETICS 2010; 11:61. [PMID: 20403157 PMCID: PMC2865459 DOI: 10.1186/1471-2350-11-61] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2009] [Accepted: 04/19/2010] [Indexed: 01/02/2023]
Abstract
Background GTF2I codes for a general intrinsic transcription factor and calcium channel regulator TFII-I, with high and ubiquitous expression, and a strong candidate for involvement in the morphological and neuro-developmental anomalies of the Williams-Beuren syndrome (WBS). WBS is a genetic disorder due to a recurring deletion of about 1,55-1,83 Mb containing 25-28 genes in chromosome band 7q11.23 including GTF2I. Completed homozygous loss of either the Gtf2i or Gtf2ird1 function in mice provided additional evidence for the involvement of both genes in the craniofacial and cognitive phenotype. Unfortunately nothing is now about the behavioral characterization of heterozygous mice. Methods By gene targeting we have generated a mutant mice with a deletion of the first 140 amino-acids of TFII-I. mRNA and protein expression analysis were used to document the effect of the study deletion. We performed behavioral characterization of heterozygous mutant mice to document in vivo implications of TFII-I in the cognitive profile of WBS patients. Results Homozygous and heterozygous mutant mice exhibit craniofacial alterations, most clearly represented in homozygous condition. Behavioral test demonstrate that heterozygous mutant mice exhibit some neurobehavioral alterations and hyperacusis or odynacusis that could be associated with specific features of WBS phenotype. Homozygous mutant mice present highly compromised embryonic viability and fertility. Regarding cellular model, we documented a retarded growth in heterozygous MEFs respect to homozygous or wild-type MEFs. Conclusion Our data confirm that, although additive effects of haploinsufficiency at several genes may contribute to the full craniofacial or neurocognitive features of WBS, correct expression of GTF2I is one of the main players. In addition, these findings show that the deletion of the fist 140 amino-acids of TFII-I altered it correct function leading to a clear phenotype, at both levels, at the cellular model and at the in vivo model.
Collapse
Affiliation(s)
- Jaume Lucena
- Genetics Unit, de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Li HH, Roy M, Kuscuoglu U, Spencer CM, Halm B, Harrison KC, Bayle JH, Splendore A, Ding F, Meltzer LA, Wright E, Paylor R, Deisseroth K, Francke U. Induced chromosome deletions cause hypersociability and other features of Williams-Beuren syndrome in mice. EMBO Mol Med 2010; 1:50-65. [PMID: 20049703 PMCID: PMC3378107 DOI: 10.1002/emmm.200900003] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The neurodevelopmental disorder Williams-Beuren syndrome is caused by spontaneous approximately 1.5 Mb deletions comprising 25 genes on human chromosome 7q11.23. To functionally dissect the deletion and identify dosage-sensitive genes, we created two half-deletions of the conserved syntenic region on mouse chromosome 5G2. Proximal deletion (PD) mice lack Gtf2i to Limk1, distal deletion (DD) mice lack Limk1 to Fkbp6, and the double heterozygotes (D/P) model the complete human deletion. Gene transcript levels in brain are generally consistent with gene dosage. Increased sociability and acoustic startle response are associated with PD, and cognitive defects with DD. Both PD and D/P males are growth-retarded, while skulls are shortened and brains are smaller in DD and D/P. Lateral ventricle (LV) volumes are reduced, and neuronal cell density in the somatosensory cortex is increased, in PD and D/P. Motor skills are most impaired in D/P. Together, these partial deletion mice replicate crucial aspects of the human disorder and serve to identify genes and gene networks contributing to the neural substrates of complex behaviours and behavioural disorders.
Collapse
Affiliation(s)
- Hong Hua Li
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Ferrero GB, Howald C, Micale L, Biamino E, Augello B, Fusco C, Turturo MG, Forzano S, Reymond A, Merla G. An atypical 7q11.23 deletion in a normal IQ Williams-Beuren syndrome patient. Eur J Hum Genet 2010; 18:33-8. [PMID: 19568270 DOI: 10.1038/ejhg.2009.108] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Williams-Beuren syndrome (WBS; OMIM no. 194050) is a multisystemic neurodevelopmental disorder caused by a hemizygous deletion of 1.55 Mb on chromosome 7q11.23 spanning 28 genes. Haploinsufficiency of the ELN gene was shown to be responsible for supravalvular aortic stenosis and generalized arteriopathy, whereas LIMK1, CLIP2, GTF2IRD1 and GTF2I genes were suggested to be linked to the specific cognitive profile and craniofacial features. These insights for genotype-phenotype correlations came from the molecular and clinical analysis of patients with atypical deletions and mice models. Here we report a patient showing mild WBS physical phenotype and normal IQ, who carries a shorter 1 Mb atypical deletion. This rearrangement does not include the GTF2IRD1 and GTF2I genes and only partially the BAZ1B gene. Our results are consistent with the hypothesis that hemizygosity of the GTF2IRD1 and GTF2I genes might be involved in the facial dysmorphisms and in the specific motor and cognitive deficits observed in WBS patients.
Collapse
|
36
|
Palmer SJ, Santucci N, Widagdo J, Bontempo SJ, Taylor KM, Tay ESE, Hook J, Lemckert F, Gunning PW, Hardeman EC. Negative autoregulation of GTF2IRD1 in Williams-Beuren syndrome via a novel DNA binding mechanism. J Biol Chem 2010; 285:4715-24. [PMID: 20007321 PMCID: PMC2836076 DOI: 10.1074/jbc.m109.086660] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2009] [Revised: 12/07/2009] [Indexed: 11/06/2022] Open
Abstract
The GTF2IRD1 gene is of principal interest to the study of Williams-Beuren syndrome (WBS). This neurodevelopmental disorder results from the hemizygous deletion of a region of chromosome 7q11.23 containing 28 genes including GTF2IRD1. WBS is thought to be caused by haploinsufficiency of certain dosage-sensitive genes within the deleted region, and the feature of supravalvular aortic stenosis (SVAS) has been attributed to reduced elastin caused by deletion of ELN. Human genetic mapping data have implicated two related genes GTF2IRD1 and GTF2I in the cause of some the key features of WBS, including craniofacial dysmorphology, hypersociability, and visuospatial deficits. Mice with mutations of the Gtf2ird1 allele show evidence of craniofacial abnormalities and behavioral changes. Here we show the existence of a negative autoregulatory mechanism that controls the level of GTF2IRD1 transcription via direct binding of the GTF2IRD1 protein to a highly conserved region of the GTF2IRD1 promoter containing an array of three binding sites. The affinity for this protein-DNA interaction is critically dependent upon multiple interactions between separate domains of the protein and at least two of the DNA binding sites. This autoregulatory mechanism leads to dosage compensation of GTF2IRD1 transcription in WBS patients. The GTF2IRD1 promoter represents the first established in vivo gene target of the GTF2IRD1 protein, and we use it to model its DNA interaction capabilities.
Collapse
Affiliation(s)
- Stephen J Palmer
- Department of Anatomy, School of Medical Sciences, The University of New South Wales, Sydney 2052, Australia.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Affiliation(s)
- Barbara R Pober
- Center for Human Genetics, Massachusetts General Hospital, Boston, MA 02114, USA.
| |
Collapse
|
38
|
A genome-wide linkage and association scan reveals novel loci for autism. Nature 2009; 461:802-8. [PMID: 19812673 PMCID: PMC2772655 DOI: 10.1038/nature08490] [Citation(s) in RCA: 457] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2008] [Accepted: 09/08/2009] [Indexed: 12/12/2022]
Abstract
Although autism is a highly heritable neurodevelopmental disorder, attempts to identify specific susceptibility genes have thus far met with limited success. Genome-wide association studies using half a million or more markers, particularly those with very large sample sizes achieved through meta-analysis, have shown great success in mapping genes for other complex genetic traits. Consequently, we initiated a linkage and association mapping study using half a million genome-wide single nucleotide polymorphisms (SNPs) in a common set of 1,031 multiplex autism families (1,553 affected offspring). We identified regions of suggestive and significant linkage on chromosomes 6q27 and 20p13, respectively. Initial analysis did not yield genome-wide significant associations; however, genotyping of top hits in additional families revealed an SNP on chromosome 5p15 (between SEMA5A and TAS2R1) that was significantly associated with autism (P = 2 x 10(-7)). We also demonstrated that expression of SEMA5A is reduced in brains from autistic patients, further implicating SEMA5A as an autism susceptibility gene. The linkage regions reported here provide targets for rare variation screening whereas the discovery of a single novel association demonstrates the action of common variants.
Collapse
|
39
|
Krzeminska D, Steinfeld C, Cloez JL, Vibert M, Chery M, Menzies D, Gilbert-Dussardier B. Prenatal diagnosis of Williams syndrome based on ultrasound signs. Prenat Diagn 2009; 29:710-2. [PMID: 19340838 DOI: 10.1002/pd.2263] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
40
|
Jackowski AP, Rando K, Maria de Araújo C, Del Cole CG, Silva I, Tavares de Lacerda AL. Brain abnormalities in Williams syndrome: a review of structural and functional magnetic resonance imaging findings. Eur J Paediatr Neurol 2009; 13:305-16. [PMID: 18722146 DOI: 10.1016/j.ejpn.2008.07.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2008] [Revised: 07/02/2008] [Accepted: 07/09/2008] [Indexed: 11/27/2022]
Abstract
Williams syndrome (WS) is rare genetic form of mental retardation caused by a microdeletion on chromosome 7q11.23 that causes cognitive impairment and a variety of physical abnormalities. MRI studies of WS have demonstrated a series of brain abnormalities, including decreased brain size, with a relatively greater decrease in the volume of the cerebral white matter volume as compared to the cerebral gray matter. Moreover there is evidence that the posterior cerebrum is more affected in that persons with WS have a greater ratio of frontal to posterior regional volume. These findings are further supported by automated analyses that have shown reduced gray matter density in the superior parietal lobe areas. Functional MRI studies have demonstrated hypofunction immediately adjacent to, and anterior to, the intraparietal sulcus, a region in which structural brain differences had been identified. These anatomical and functional differences are consistent with the neuropsychological profile of WS - in particular, with evidence of dorsal stream visual processing deficits. To date, however, studies have always been performed in comparison to intellectually average controls. It is not clear, therefore, if findings are specific to the WS population or whether they represent a morphological disturbance characteristic of mental retardation, irrespective of genetic etiology. In this article, we reviewed recent advances underlying the structural and functional neural substrate of WS in Medical Literature Analysis and Retrieval System Online (MEDLINE; 1997-2007).
Collapse
Affiliation(s)
- Andrea Parolin Jackowski
- Laboratório Interdiciplinar de Neurociências Clínicas (LiNC), Universidade Federal de Sao Paulo (UNIFESP), São Paulo, São Paulo, Brazil.
| | | | | | | | | | | |
Collapse
|
41
|
Dai L, Bellugi U, Chen XN, Pulst-Korenberg AM, Järvinen-Pasley A, Tirosh-Wagner T, Eis PS, Graham J, Mills D, Searcy Y, Korenberg JR. Is it Williams syndrome? GTF2IRD1 implicated in visual-spatial construction and GTF2I in sociability revealed by high resolution arrays. Am J Med Genet A 2009; 149A:302-14. [PMID: 19205026 DOI: 10.1002/ajmg.a.32652] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Genetic contributions to human cognition and behavior are clear but difficult to define. Williams syndrome (WS) provides a unique model for relating single genes to visual-spatial cognition and social behavior. We defined a approximately 1.5 Mb region of approximately 25 genes deleted in >98% of typical WS and then rare small deletions, showing that visual-spatial construction (VSC) in WS was associated with the genes GTF2IRD1 and GTF2I. To distinguish the roles of GTF2IRD1 and GTF2I in VSC and social behavior, we utilized multiple genomic methods (custom high resolution oligonucleotide microarray, multicolor FISH and somatic cell hybrids analyzed by PCR) to identify individuals deleted for either gene but not both. We analyzed genetic, cognitive and social behavior in a unique individual with WS features (heart defects, small size, facies), but with an atypical deletion of a set of genes that includes GTF2IRD1, but not GTF2I. The centromeric breakpoint localized to the region 72.32-72.38 Mb and the telomeric breakpoint to 72.66 Mb, 10 kb downstream of GTF2IRD1. Cognitive testing (WPPSI-R, K-BIT, and PLS-3) demonstrated striking deficits in VSC (Block Design, Object Assembly) but overall performance 1.5-3 SD above WS means. We have now integrated the genetic, clinical and cognitive data with previous reports of social behavior in this subject. These results combine with previous data from small deletions to suggest the gene GTF2IRD1 is associated with WS facies and VSC, and that GTF2I may contribute to WS social behaviors including increased gaze and attention to strangers.
Collapse
Affiliation(s)
- L Dai
- The Center for Integrated Neuroscience and Human Behavior, The Brain Institute, University of Utah, Salt Lake City, Utah 84108, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Collette JC, Chen XN, Mills DL, Galaburda AM, Reiss AL, Bellugi U, Korenberg JR. William's syndrome: gene expression is related to parental origin and regional coordinate control. J Hum Genet 2009; 54:193-8. [PMID: 19282872 DOI: 10.1038/jhg.2009.5] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
William's syndrome (WS) features a spectrum of neurocognitive and behavioral abnormalities due to a rare 1.5 MB deletion that includes about 24-28 genes on chromosome band 7q11.23. Study of the expression of these genes from the single normal copy provides an opportunity to elucidate the genetic and epigenetic controls on these genes as well as their roles in both WS and normal brain development and function. We used quantitative RT-PCR to determine the transcriptional level of 14 WS gene markers in a cohort of 77 persons with WS and 48 normal controls. Results reported here: (1) show that the expression of the genes deleted in WS is decreased in some but not all cases, (2) demonstrate that the parental origin of the deletion contributes to the level of expression of GTF2I independently of age and gender and (3) indicate that the correlation of expression between GTF2I and some other genes in the WS region differs in WS subjects and normal controls, which in turn points toward a regulatory role for this gene. Interspecies comparisons suggest GTF2I may play a key role in normal brain development.
Collapse
Affiliation(s)
- Jeremy C Collette
- Division of Neurogenetics, Cedars-Sinai Medical Center and Departments of Human Genetics and Pediatrics, UCLA, Los Angeles, CA, USA
| | | | | | | | | | | | | |
Collapse
|
43
|
Le syndrome de Williams-Beuren : une approche pluridisciplinaire. Arch Pediatr 2009; 16:273-82. [DOI: 10.1016/j.arcped.2008.11.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2007] [Revised: 08/28/2008] [Accepted: 11/01/2008] [Indexed: 01/05/2023]
|
44
|
|
45
|
Identifying disease-causal genes using Semantic Web-based representation of integrated genomic and phenomic knowledge. J Biomed Inform 2008; 41:717-29. [DOI: 10.1016/j.jbi.2008.07.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2007] [Revised: 07/20/2008] [Accepted: 07/23/2008] [Indexed: 12/22/2022]
|
46
|
Identification of the TFII-I family target genes in the vertebrate genome. Proc Natl Acad Sci U S A 2008; 105:9006-10. [PMID: 18579769 DOI: 10.1073/pnas.0803051105] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
GTF2I and GTF2IRD1 encode members of the TFII-I transcription factor family and are prime candidates in the Williams syndrome, a complex neurodevelopmental disorder. Our previous expression microarray studies implicated TFII-I proteins in the regulation of a number of genes critical in various aspects of cell physiology. Here, we combined bioinformatics and microarray results to identify TFII-I downstream targets in the vertebrate genome. These results were validated by chromatin immunoprecipitation and siRNA analysis. The collected evidence revealed the complexity of TFII-I-mediated processes that involve distinct regulatory networks. Altogether, these results lead to a better understanding of specific molecular events, some of which may be responsible for the Williams syndrome phenotype.
Collapse
|
47
|
Hocking DR, Bradshaw JL, Rinehart NJ. Fronto-parietal and cerebellar contributions to motor dysfunction in Williams syndrome: A review and future directions. Neurosci Biobehav Rev 2008; 32:497-507. [DOI: 10.1016/j.neubiorev.2007.09.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2007] [Revised: 09/12/2007] [Accepted: 09/30/2007] [Indexed: 12/29/2022]
|
48
|
Williams syndrome: a genetic deletion disorder presenting clues to the biology of sociability and clinical challenges of hypersociability. CNS Spectr 2007; 12:903-7. [PMID: 18163035 DOI: 10.1017/s1092852900015686] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Williams syndrome is a neurodevelopmental disorder that results from the deletion of approximately 25-30 genes spanning about 1.5 megabases in the q11.23 region of chromosome 7. Patients with this syndrome present with a combination of a distinctive elfin-like facial appearance; growth retardation; mild mental retardation; an inconsistent cognitive profile that includes visuospatial impairments with good facial discrimination and relatively preserved expressive language skills; and cardiovascular abnormalities. In addition, a striking behavioral feature of the syndrome is the high sociability and empathy that these patients show for others. The study of patients with "partial" deletions of the chromosome band 7q11.23, mutated genes in this region and knockout mice with deletions of specific genes in the homologous G1-G2 region of mouse chromosome 5 are clarifying some genotype/phenotype relationships. Furthermore, genes located in this region that are prominently expressed have been implicated in brain development and function. The neuropsychological profile of patients with Williams syndrome is heterogeneous, highlights important dissociations between cognitive functions and suggests that the behavioral dimensions of sociability, empathy, engageability, and talkativeness may be independent of, or not easily explained by, the cognitive deficits. Williams syndrome has enormous heuristic value because its pathological feature of heightened "sociability" can be a "deficit" symptom of major complex neuropsychiatric disorders, such as schizophrenia and autism. Data consistent with a core inability of patients with Williams syndrome to inhibit social approach suggest that this disorder may afford an opportunity to study the biological basis of the "drive" toward socialization. From a research perspective, the syndrome lends itself to neurobiological studies of sociability as a dimension that varies independently of cognition (or at least many separable cognitive processes). Importantly, from a clinical perspective, the syndrome challenges us to administer strategic psychosocial interventions that take advantage of the opportunities that "pathological" sociability provide, while avoiding its threats. An illustrative example of an effective strategically planned psychosocial intervention for a patient with Williams syndrome is briefly presented.
Collapse
|
49
|
Jaworski J, Hoogenraad CC, Akhmanova A. Microtubule plus-end tracking proteins in differentiated mammalian cells. Int J Biochem Cell Biol 2007; 40:619-37. [PMID: 18023603 DOI: 10.1016/j.biocel.2007.10.015] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2007] [Revised: 09/16/2007] [Accepted: 10/11/2007] [Indexed: 11/16/2022]
Abstract
Differentiated mammalian cells are often characterized by highly specialized and polarized structure. Its formation and maintenance depends on cytoskeletal components, among which microtubules play an important role. The shape and dynamic properties of microtubule networks are controlled by multiple microtubule-associated factors. These include molecular motors and non-motor proteins, some of which accumulate specifically at the growing microtubule plus-ends (the so-called microtubule plus-end tracking proteins). Plus-end tracking proteins can contribute to the regulation of microtubule dynamics, mediate the cross-talk between microtubule ends, the actin cytoskeleton and the cell cortex, and participate in transport and positioning of structural and regulatory factors and membrane organelles. Malfunction of these proteins results in various human diseases including some forms of cancer, neurodevelopmental disorders and mental retardation. In this article we discuss recent data on microtubule dynamics and activities of microtubule plus-end binding proteins important for the physiology and pathology of differentiated mammalian cells such as neurons, polarized epithelia, muscle and sperm cells.
Collapse
Affiliation(s)
- Jacek Jaworski
- International Institute of Molecular and Cell Biology , Warsaw, Poland.
| | | | | |
Collapse
|
50
|
van Hagen JM, Eussen HJ, van Schooten R, van Der Geest JN, Lagers-van Haselen GC, Wouters CH, De Zeeuw CI, Gille JJ. Comparing Two Diagnostic Laboratory Tests for Williams Syndrome: Fluorescent In Situ Hybridization versus Multiplex Ligation-Dependent Probe Amplification. ACTA ACUST UNITED AC 2007; 11:321-7. [DOI: 10.1089/gte.2007.0007] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Johanna M. van Hagen
- Department of Clinical Genetics, VU University Medical Center, 1007 MB Amsterdam, The Netherlands
| | | | - Ron van Schooten
- Department of Clinical Genetics, VU University Medical Center, 1007 MB Amsterdam, The Netherlands
| | | | | | - Cokkie H. Wouters
- Department of Clinical Genetics, Erasmus MC, 3000 DR Rotterdam, The Netherlands
| | - Chris I. De Zeeuw
- Department of Neuroscience, Erasmus MC, 3000 CA Rotterdam, The Netherlands
| | - Johan J.P. Gille
- Department of Clinical Genetics, VU University Medical Center, 1007 MB Amsterdam, The Netherlands
| |
Collapse
|