1
|
Bangash MA, Cubuk C, Iseppon F, Haroun R, Garcia C, Luiz AP, Arcangeletti M, Gossage SJ, Santana-Varela S, Cox JJ, Lewis MJ, Wood JN, Zhao J. Analgesic targets identified in mouse sensory neuron somata and terminal pain translatomes. Cell Rep 2024; 43:114614. [PMID: 39163201 DOI: 10.1016/j.celrep.2024.114614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 06/07/2024] [Accepted: 07/24/2024] [Indexed: 08/22/2024] Open
Abstract
The relationship between transcription and protein expression is complex. We identified polysome-associated RNA transcripts in the somata and central terminals of mouse sensory neurons in control, painful (plus nerve growth factor), and pain-free conditions (Nav1.7-null mice). The majority (98%) of translated transcripts are shared between male and female mice in both the somata and terminals. Some transcripts are highly enriched in the somata or terminals. Changes in the translatome in painful and pain-free conditions include novel and known regulators of pain pathways. Antisense knockdown of selected somatic and terminal polysome-associated transcripts that correlate with pain states diminished pain behavior. Terminal-enriched transcripts included those encoding synaptic proteins (e.g., synaptotagmin), non-coding RNAs, transcription factors (e.g., Znf431), proteins associated with transsynaptic trafficking (HoxC9), GABA-generating enzymes (Gad1 and Gad2), and neuropeptides (Penk). Thus, central terminal translation may well be a significant regulatory locus for peripheral input from sensory neurons.
Collapse
Affiliation(s)
- M Ali Bangash
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London WC1E 6BT, UK
| | - Cankut Cubuk
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Federico Iseppon
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London WC1E 6BT, UK
| | - Rayan Haroun
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London WC1E 6BT, UK
| | - Chloe Garcia
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London WC1E 6BT, UK
| | - Ana P Luiz
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London WC1E 6BT, UK
| | - Manuel Arcangeletti
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London WC1E 6BT, UK
| | - Samuel J Gossage
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London WC1E 6BT, UK
| | - Sonia Santana-Varela
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London WC1E 6BT, UK
| | - James J Cox
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London WC1E 6BT, UK
| | - Myles J Lewis
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - John N Wood
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London WC1E 6BT, UK.
| | - Jing Zhao
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London WC1E 6BT, UK.
| |
Collapse
|
2
|
Miller SC, MacDonald CC, Kellogg MK, Karamysheva ZN, Karamyshev AL. Specialized Ribosomes in Health and Disease. Int J Mol Sci 2023; 24:ijms24076334. [PMID: 37047306 PMCID: PMC10093926 DOI: 10.3390/ijms24076334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/21/2023] [Accepted: 03/23/2023] [Indexed: 03/30/2023] Open
Abstract
Ribosomal heterogeneity exists within cells and between different cell types, at specific developmental stages, and occurs in response to environmental stimuli. Mounting evidence supports the existence of specialized ribosomes, or specific changes to the ribosome that regulate the translation of a specific group of transcripts. These alterations have been shown to affect the affinity of ribosomes for certain mRNAs or change the cotranslational folding of nascent polypeptides at the exit tunnel. The identification of specialized ribosomes requires evidence of the incorporation of different ribosomal proteins or of modifications to rRNA and/or protein that lead(s) to physiologically relevant changes in translation. In this review, we summarize ribosomal heterogeneity and specialization in mammals and discuss their relevance to several human diseases.
Collapse
Affiliation(s)
- Sarah C. Miller
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Clinton C. MacDonald
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Morgana K. Kellogg
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | | | - Andrey L. Karamyshev
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Correspondence: ; Tel.: +1-806-743-4102
| |
Collapse
|
3
|
Evaluation of Individuals with Non-Syndromic Global Developmental Delay and Intellectual Disability. CHILDREN 2023; 10:children10030414. [PMID: 36979972 PMCID: PMC10047567 DOI: 10.3390/children10030414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/11/2023] [Accepted: 02/16/2023] [Indexed: 02/24/2023]
Abstract
Global Developmental Delay (GDD) and Intellectual Disability (ID) are two of the most common presentations encountered by physicians taking care of children. GDD/ID is classified into non-syndromic GDD/ID, where GDD/ID is the sole evident clinical feature, or syndromic GDD/ID, where there are additional clinical features or co-morbidities present. Careful evaluation of children with GDD and ID, starting with detailed history followed by a thorough examination, remain the cornerstone for etiologic diagnosis. However, when initial history and examination fail to identify a probable underlying etiology, further genetic testing is warranted. In recent years, genetic testing has been shown to be the single most important diagnostic modality for clinicians evaluating children with non-syndromic GDD/ID. In this review, we discuss different genetic testing currently available, review common underlying copy-number variants and molecular pathways, explore the recent evidence and recommendations for genetic evaluation and discuss an approach to the diagnosis and management of children with non-syndromic GDD and ID.
Collapse
|
4
|
Sakka R, Abdelhedi F, Sellami H, Pichon B, Lajmi Y, Mnif M, Kebaili S, Derbel R, Kamoun H, Gdoura R, Delbaere A, Desir J, Abramowicz M, Vialard F, Dupont JM, Ammar-Keskes L. An unusual familial Xp22.12 microduplication including EIF1AX: A novel candidate dosage-sensitive gene for premature ovarian insufficiency. Eur J Med Genet 2022; 65:104613. [PMID: 36113757 DOI: 10.1016/j.ejmg.2022.104613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 07/22/2022] [Accepted: 09/09/2022] [Indexed: 11/19/2022]
Abstract
We report on the results of array-CGH and Whole exome sequencing (WES) studies carried out in a Tunisian family with 46,XX premature ovarian insufficiency (POI). This study has led to the identification of a familial Xp22.12 tandem duplication with a size of 559.4 kb, encompassing only three OMIM genes (RPS6KA3, SH3KBP1and EIF1AX), and a new heterozygous variant in SPIDR gene: NM_001080394.3:c.1845_1853delTATAATTGA (p.Ile616_Asp618del) segregating with POI. Increased mRNA expression levels were detected for SH3KBP1 and EIF1AX, while a normal transcript level for RPS6KA3 was detected in the three affected family members, explaining the absence of intellectual disability (ID). To the best of our knowledge, this is the first duplication involving the Xp22.12 region, reported in a family without ID, but rather with secondary amenorrhea (SA) and female infertility. As EIF1AX is a regulatory gene escaping X-inactivation, which has an extreme dosage sensitivity and highly expressed in the ovary, we suggest that this gene might be a candidate gene for ovarian function. Homozygous nonsense pathogenic variants of SPIDR gene have been reported in familial cases in POI. It has been suggested that chromosomal instability associated with SPIDR molecular defects supports the role of SPIDR protein in double-stranded DNA damage repair in vivo in humans and its causal role in POI. In this family, the variant (p.Ile616_Asp618del), present in a heterozygous state, is located in the domain that interacts with BLM and might disrupt the BLM binding ability of SPIDR protein. These findings strengthen the hypothesis that the additional effect of this variant could lead to POI in this family. Although the work represents the first evidence that EIF1AX duplication might be responsible for POI through its over-expression, further functional studies are needed to clarify and prove EIF1AX involvement in POI phenotype.
Collapse
Affiliation(s)
- Rim Sakka
- Human Molecular Genetics Laboratory, Faculty of Medicine of Sfax, University of Sfax, Tunisia; Center of Medical Genetics, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Fatma Abdelhedi
- Human Molecular Genetics Laboratory, Faculty of Medicine of Sfax, University of Sfax, Tunisia; Medical Genetics Department, Hedi Chaker Hospital, Sfax, Tunisia.
| | - Hanen Sellami
- Water Researches and Technologies Center (CERTE), University of Carthage, Tourist Road Soliman, Nabeul, Tunisia; Toxicology, Environmental Microbiology and Health Research Laboratory (LR17ES06), Faculty of Sciences of Sfax, University of Sfax, Tunisia
| | - Bruno Pichon
- Center of Medical Genetics, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Yosra Lajmi
- Cytogenetics Department, Cochin Hospital, Assistance Publique des Hôpitaux de Paris, Sorbonne Paris Cité, Paris Descartes University, Medical School, Paris, France
| | - Mouna Mnif
- Department of Endocrinology, Hedi Chaker Hospital, Sfax, Tunisia
| | - Sahbi Kebaili
- Department of Gynecology, HediChaker Hospital, Sfax, Tunisia
| | - Rihab Derbel
- Human Molecular Genetics Laboratory, Faculty of Medicine of Sfax, University of Sfax, Tunisia
| | - Hassen Kamoun
- Medical Genetics Department, Hedi Chaker Hospital, Sfax, Tunisia
| | - Radhouane Gdoura
- Toxicology, Environmental Microbiology and Health Research Laboratory (LR17ES06), Faculty of Sciences of Sfax, University of Sfax, Tunisia
| | - Anne Delbaere
- Fertility Clinic, Department of Gynecology and Obstetrics, Erasme Hospital, UniversitéLibre de Bruxelles, Brussels, Belgium
| | - Julie Desir
- Center of Medical Genetics, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Marc Abramowicz
- Center of Medical Genetics, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - François Vialard
- Genetics Department, CHI Poissy St Germain-en-Laye, F-78300, Poissy, France; RHuMA Team, UMR-BREED, INRAE-UVSQ-ENVA, UFR-SVS, F-78180, Montigny le Bretonneux, France
| | - Jean-Michel Dupont
- Cytogenetics Department, Cochin Hospital, Assistance Publique des Hôpitaux de Paris, Sorbonne Paris Cité, Paris Descartes University, Medical School, Paris, France
| | - Leila Ammar-Keskes
- Human Molecular Genetics Laboratory, Faculty of Medicine of Sfax, University of Sfax, Tunisia
| |
Collapse
|
5
|
Identification of a New Mutation in RSK2, the Gene for Coffin-Lowry Syndrome (CLS), in Two Related Patients with Mild and Atypical Phenotypes. Brain Sci 2021; 11:brainsci11081105. [PMID: 34439726 PMCID: PMC8394959 DOI: 10.3390/brainsci11081105] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 08/14/2021] [Accepted: 08/17/2021] [Indexed: 11/16/2022] Open
Abstract
Background: Coffin–Lowry syndrome (CLS) is a syndromic form of X-linked intellectual disability, in which specific associated facial, hand, and skeletal abnormalities are diagnostic features. Methods: In the present study, an unreported missense genetic variant of the ribosomal S6 kinase 2 (RSK2) gene has been identified, by next-generation sequencing, in two related males with two different phenotypes of intellectual disability (ID) and peculiar facial dysmorphisms. We performed functional studies on this variant and another one, already reported in the literature, involving the same amino acid residue but, to date, without an efficient characterization. Results: Our study demonstrated that the two variants involving residue 189 significantly impaired its kinase activity. Conclusions: We detected a loss-of-function RSK2 mutation with loss in kinase activity in a three-generation family with an X-linked ID.
Collapse
|
6
|
Uliana V, Bonatti F, Zanatta V, Mozzoni P, Martorana D, Percesepe A. Spectrum of X-linked intellectual disabilities and psychiatric symptoms in a family harbouring a Xp22.12 microduplication encompassing the RPS6KA3 gene. J Genet 2019. [DOI: 10.1007/s12041-019-1055-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
7
|
Rawashdeh O, Jilg A, Maronde E, Fahrenkrug J, Stehle JH. Period1gates the circadian modulation of memory-relevant signaling in mouse hippocampus by regulating the nuclear shuttling of the CREB kinase pP90RSK. J Neurochem 2016; 138:731-45. [DOI: 10.1111/jnc.13689] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 05/24/2016] [Accepted: 05/25/2016] [Indexed: 12/15/2022]
Affiliation(s)
- Oliver Rawashdeh
- Institute of Cellular and Molecular Anatomy; Dr. Senckenbergische Anatomie; Goethe-University; Frankfurt Germany
- School of Biomedical Sciences; University of Queensland; St Lucia Qld Australia
| | - Antje Jilg
- Institute of Cellular and Molecular Anatomy; Dr. Senckenbergische Anatomie; Goethe-University; Frankfurt Germany
| | - Erik Maronde
- Institute of Cellular and Molecular Anatomy; Dr. Senckenbergische Anatomie; Goethe-University; Frankfurt Germany
| | - Jan Fahrenkrug
- Department of Clinical Chemistry; Bispebjerg Hospital, Faculty of Health and Medical Sciences; University of Copenhagen; Copenhagen Denmark
| | - Jörg H. Stehle
- Institute of Cellular and Molecular Anatomy; Dr. Senckenbergische Anatomie; Goethe-University; Frankfurt Germany
| |
Collapse
|
8
|
Sandoval-Hernández AG, Hernández HG, Restrepo A, Muñoz JI, Bayon GF, Fernández AF, Fraga MF, Cardona-Gómez GP, Arboleda H, Arboleda GH. Liver X Receptor Agonist Modifies the DNA Methylation Profile of Synapse and Neurogenesis-Related Genes in the Triple Transgenic Mouse Model of Alzheimer's Disease. J Mol Neurosci 2015; 58:243-53. [PMID: 26553261 DOI: 10.1007/s12031-015-0665-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 10/08/2015] [Indexed: 12/22/2022]
Abstract
The liver X receptor agonist, GW3965, improves cognition in Alzheimer's disease (AD) mouse models. Here, we determined if short-term GW3965 treatment induces changes in the DNA methylation state of the hippocampus, which are associated with cognitive improvement. Twenty-four-month-old triple-transgenic AD (3xTg-AD) mice were treated with GW3965 (50 mg/kg/day for 6 days). DNA methylation state was examined by modified bisulfite conversion and hybridization on Illumina Infinium Methylation BeadChip 450 k arrays. The Morris water maze was used for behavioral analysis. Our results show in addition to improvement in cognition methylation changes in 39 of 13,715 interrogated probes in treated 3xTg-AD mice compared with untreated 3xTg-AD mice. These changes in methylation probes include 29 gene loci. Importantly, changes in methylation status were mainly from synapse-related genes (SYP, SYN1, and DLG3) and neurogenesis-associated genes (HMGB3 and RBBP7). Thus, our results indicate that liver X receptors (LXR) agonist treatment induces rapid changes in DNA methylation, particularly in loci associated with genes involved in neurogenesis and synaptic function. Our results suggest a new potential mechanism to explain the beneficial effect of GW3965.
Collapse
Affiliation(s)
- A G Sandoval-Hernández
- Grupo de Muerte Celular, Instituto de Genética, Universidad Nacional de Colombia, Bogotá, Colombia
| | - H G Hernández
- Grupo de Neurociencias, Universidad Nacional, Bogotá, Colombia
| | - A Restrepo
- Grupo de Muerte Celular, Instituto de Genética, Universidad Nacional de Colombia, Bogotá, Colombia
| | - J I Muñoz
- Área de Neurobiología Celular y Molecular, Grupo de Neurociencias de Antioquia, Universidad de Antioquia, Medellín, Colombia
| | - G F Bayon
- Cancer Epigenetics Laboratory, Institute of Oncology of Asturias (IUOPA), Hospital Universitario Central de Asturias (HUCA), Universidad de Oviedo, Oviedo, Spain
| | - A F Fernández
- Cancer Epigenetics Laboratory, Institute of Oncology of Asturias (IUOPA), Hospital Universitario Central de Asturias (HUCA), Universidad de Oviedo, Oviedo, Spain
| | - M F Fraga
- Cancer Epigenetics Laboratory, Institute of Oncology of Asturias (IUOPA), Hospital Universitario Central de Asturias (HUCA), Universidad de Oviedo, Oviedo, Spain
| | - G P Cardona-Gómez
- Área de Neurobiología Celular y Molecular, Grupo de Neurociencias de Antioquia, Universidad de Antioquia, Medellín, Colombia
| | - H Arboleda
- Grupo de Neurociencias, Universidad Nacional, Bogotá, Colombia
| | - Gonzalo H Arboleda
- Grupo de Muerte Celular, Instituto de Genética, Universidad Nacional de Colombia, Bogotá, Colombia.
- Departamento de Patología, Facultad de Medicina, Universidad Nacional de Colombia, Bogotá, Colombia.
| |
Collapse
|
9
|
625 kb microduplication at Xp22.12 including RPS6KA3 in a child with mild intellectual disability. J Hum Genet 2015; 60:777-80. [PMID: 26354035 DOI: 10.1038/jhg.2015.106] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Revised: 07/09/2015] [Accepted: 07/13/2015] [Indexed: 11/09/2022]
Abstract
Here, we report on a patient with a 625 kb duplication in Xp22.12, detected by array comparative genomic hybridization (CGH). The duplicated region contains only one gene, RPS6KA3, that results in partial duplication. The same duplication was present in his mother and his maternal uncle. This partial duplication inhibits the RPS6KA3 expression, mimicking the effect of loss-of-function mutations associated with Coffin-Lowry syndrome (CLS). The phenotype of the patient here presented is not fully evocative of this syndrome because he does not present some of the facial, digital and skeletal abnormalities that are considered the main diagnostic features of CLS. This case is one of the few examples where RPS6KA3 mutations are associated with a non-specific X-linked mental retardation.
Collapse
|
10
|
Matsumoto A, Kuwajima M, Miyake K, Kojima K, Nakashima N, Jimbo EF, Kubota T, Momoi MY, Yamagata T. An Xp22.12 microduplication including RPS6KA3 identified in a family with variably affected intellectual and behavioral disabilities. J Hum Genet 2013; 58:755-7. [DOI: 10.1038/jhg.2013.88] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Revised: 07/28/2013] [Accepted: 07/31/2013] [Indexed: 11/09/2022]
|
11
|
Maystadt I, Destree A, Benoit V, Aeby A, Lederer D, Moortgat S, Jurkiewicz D, Krajewska-Walasek M, Hanauer A, Thomas GM. RSK2mutation co-segregates with X-linked intellectual disability and attenuated Coffin-Lowry phenotype in a three-generation family. Clin Genet 2013; 85:96-9. [DOI: 10.1111/cge.12122] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2012] [Revised: 02/06/2013] [Accepted: 02/12/2013] [Indexed: 11/29/2022]
Affiliation(s)
| | | | - V Benoit
- Département de Biologie Moléculaire; Institut de Pathologie et de Génétique; Gosselies Belgium
| | - A Aeby
- Département de Neurologie Pédiatrique; Université Libre de Bruxelles-Hôpital Erasme; Brussels Belgium
| | | | | | - D Jurkiewicz
- Department of Medical Genetics; The Children's Memorial Health Institute; Warszaw Poland
| | - M Krajewska-Walasek
- Department of Medical Genetics; The Children's Memorial Health Institute; Warszaw Poland
| | - A Hanauer
- Translational Medicine & Neurogenetics; IGBMC; Illkirch France
| | - GM Thomas
- Shriners Hospital Pediatric Research Center; Temple University Medical School; Philadelphia PA USA
| |
Collapse
|
12
|
Abstract
Ten percent of cases of intellectual deficiency in boys are caused by genes located on the X chromosome. X-linked mental retardation (XLMR) includes more than 200 syndromes and 80 genes identified to date. The fragile X syndrome is the most frequent syndrome, due to a dynamic mutation with a CGG triplet amplification. Mental retardation is virtually always present. Phonological and syntactic impairments are often combined with pragmatic language impairment and visuospatial reasoning difficulties. A minority fulfill the criteria for autism. In girls, the clinical expression of the complete mutation varies according to the X chromosome inactivation profile. Several XLMR occur as severe early onset encephalopathies: Lowe oculocerebrorenal syndrome, ATR-X syndrome (alpha thalassemia/mental retardation X-linked), Allan-Herdon-Dudley syndrome (MCT8 gene). Two genes, ARX (X-LAG; Partington syndrome) and MECP2 (Rett syndrome in females; mild MR with spastic diplegia/psychotic problems in males) are associated with various phenotypes, according to the mutation involved. Oligophrenine 1 (OPHN-1) gene mutations lead to vermal dysplasia. PQBP1 gene mutations (Renpenning syndrome) are responsible for moderate to severe mental deficiency, microcephaly, and small stature. Although some forms of XLMR are not very specific and the phenotype for each given gene is somewhat heterogeneous, a clinical diagnostic strategy is emerging.
Collapse
Affiliation(s)
- Vincent des Portes
- Reference Center for Fragile X and other X-linked Intellectual Disabilities and Department of Pediatric Neurology, Hôpital Femme Mère Enfant, CHU de Lyon, Lyon, France.
| |
Collapse
|
13
|
O’Roak BJ, Vives L, Fu W, Egertson JD, Stanaway IB, Phelps IG, Carvill G, Kumar A, Lee C, Ankenman K, Munson J, Hiatt JB, Turner EH, Levy R, O’Day DR, Krumm N, Coe BP, Martin BK, Borenstein E, Nickerson DA, Mefford HC, Doherty D, Akey JM, Bernier R, Eichler EE, Shendure J. Multiplex targeted sequencing identifies recurrently mutated genes in autism spectrum disorders. Science 2012; 338:1619-22. [PMID: 23160955 PMCID: PMC3528801 DOI: 10.1126/science.1227764] [Citation(s) in RCA: 949] [Impact Index Per Article: 79.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Exome sequencing studies of autism spectrum disorders (ASDs) have identified many de novo mutations but few recurrently disrupted genes. We therefore developed a modified molecular inversion probe method enabling ultra-low-cost candidate gene resequencing in very large cohorts. To demonstrate the power of this approach, we captured and sequenced 44 candidate genes in 2446 ASD probands. We discovered 27 de novo events in 16 genes, 59% of which are predicted to truncate proteins or disrupt splicing. We estimate that recurrent disruptive mutations in six genes-CHD8, DYRK1A, GRIN2B, TBR1, PTEN, and TBL1XR1-may contribute to 1% of sporadic ASDs. Our data support associations between specific genes and reciprocal subphenotypes (CHD8-macrocephaly and DYRK1A-microcephaly) and replicate the importance of a β-catenin-chromatin-remodeling network to ASD etiology.
Collapse
Affiliation(s)
- Brian J. O’Roak
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Laura Vives
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Wenqing Fu
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Jarrett D. Egertson
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Ian B. Stanaway
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Ian G. Phelps
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA 98195, USA
- Seattle Children’s Hospital, Seattle, WA 98105, USA
| | - Gemma Carvill
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA 98195, USA
- Seattle Children’s Hospital, Seattle, WA 98105, USA
| | - Akash Kumar
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Choli Lee
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Katy Ankenman
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA 98195, USA
| | - Jeff Munson
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA 98195, USA
| | - Joseph B. Hiatt
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Emily H. Turner
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Roie Levy
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Diana R. O’Day
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Niklas Krumm
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Bradley P. Coe
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Beth K. Martin
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Elhanan Borenstein
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA 98195, USA
- Department of Computer Science and Engineering, University of Washington, Seattle, WA 98195, USA
- Santa Fe Institute, Santa Fe, NM 87501, USA
| | - Deborah A. Nickerson
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Heather C. Mefford
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA 98195, USA
- Seattle Children’s Hospital, Seattle, WA 98105, USA
| | - Dan Doherty
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA 98195, USA
- Seattle Children’s Hospital, Seattle, WA 98105, USA
| | - Joshua M. Akey
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Raphael Bernier
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA 98195, USA
| | - Evan E. Eichler
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA 98195, USA
- Howard Hughes Medical Institute, Seattle, WA 98195, USA
| | - Jay Shendure
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA 98195, USA
| |
Collapse
|
14
|
van Loo KMJ, Martens GJM. Genetic and environmental factors in complex neurodevelopmental disorders. Curr Genomics 2011; 8:429-44. [PMID: 19412416 PMCID: PMC2647153 DOI: 10.2174/138920207783591717] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2007] [Revised: 11/07/2007] [Accepted: 11/09/2007] [Indexed: 12/14/2022] Open
Abstract
Complex neurodevelopmental disorders, such as schizophrenia, autism, attention deficit (hyperactivity) disorder, (manic) depressive illness and addiction, are thought to result from an interaction between genetic and environmental factors. Association studies on candidate genes and genome-wide linkage analyses have identified many susceptibility chromosomal regions and genes, but considerable efforts to replicate association have been surprisingly often disappointing. Here, we summarize the current knowledge of the genetic contribution to complex neurodevelopmental disorders, focusing on the findings from association and linkage studies. Furthermore, the contribution of the interaction of the genetic with environmental and epigenetic factors to the aetiology of complex neurodevelopmental disorders as well as suggestions for future research are discussed.
Collapse
Affiliation(s)
- K M J van Loo
- Department of Molecular Animal Physiology, Donders Institute for Neuroscience, Nijmegen Center for Molecular Life Sciences (NCMLS), Faculty of Science, Radboud University Nijmegen, Geert Grooteplein Zuid 28, 6525 GA Nijmegen, The Netherlands
| | | |
Collapse
|
15
|
Gräff J, Kim D, Dobbin MM, Tsai LH. Epigenetic regulation of gene expression in physiological and pathological brain processes. Physiol Rev 2011; 91:603-49. [PMID: 21527733 DOI: 10.1152/physrev.00012.2010] [Citation(s) in RCA: 245] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Over the past decade, it has become increasingly obvious that epigenetic mechanisms are an integral part of a multitude of brain functions that range from the development of the nervous system over basic neuronal functions to higher order cognitive processes. At the same time, a substantial body of evidence has surfaced indicating that several neurodevelopmental, neurodegenerative, and neuropsychiatric disorders are in part caused by aberrant epigenetic modifications. Because of their inherent plasticity, such pathological epigenetic modifications are readily amenable to pharmacological interventions and have thus raised justified hopes that the epigenetic machinery provides a powerful new platform for therapeutic approaches against these diseases. In this review, we give a detailed overview of the implication of epigenetic mechanisms in both physiological and pathological brain processes and summarize the state-of-the-art of "epigenetic medicine" where applicable. Despite, or because of, these new and exciting findings, it is becoming apparent that the epigenetic machinery in the brain is highly complex and intertwined, which underscores the need for more refined studies to disentangle brain-region and cell-type specific epigenetic codes in a given environmental condition. Clearly, the brain contains an epigenetic "hotspot" with a unique potential to not only better understand its most complex functions, but also to treat its most vicious diseases.
Collapse
Affiliation(s)
- Johannes Gräff
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts 02139, USA
| | | | | | | |
Collapse
|
16
|
Abstract
Suicide completion rates are significantly higher in males than females in most societies. Although gender differences in suicide rates have been partially explained by environmental and behavioral factors, it is possible that genetic factors, through differential expression between genders, may also help explain gender moderation of suicide risk. This study investigated X-linked genes in suicide completers using a two-step strategy. We first took advantage of the genetic structure of the French-Canadian population and genotyped 722 unrelated French-Canadian male subjects, of whom 333 were suicide completers and 389 were non-suicide controls, using a panel of 37 microsatellite markers spanning the entire X chromosome. Nine haplotype windows and several individual markers were associated with suicide. Significant results aggregated primarily in two regions, one in the long arm and another in the short arm of chromosome X, limited by markers DXS8051 and DXS8102, and DXS1001 and DXS8106, respectively. The second stage of the study investigated differential brain expression of genes mapping to associated regions in Brodmann areas 8/9, 11, 44 and 46, in an independent sample of suicide completers and controls. Six genes within these regions, Rho GTPase-activating protein 6, adaptor-related protein complex 1 sigma 2 subunit, glycoprotein M6B, ribosomal protein S6 kinase 90 kDa polypeptide 3, spermidine/spermine N(1)-acetyltransferase 1 and THO complex 2, were found to be differentially expressed in suicide completers.
Collapse
|
17
|
Cossée M, Faivre L, Philippe C, Hichri H, de Saint-Martin A, Laugel V, Bahi-Buisson N, Lemaitre JF, Leheup B, Delobel B, Demeer B, Poirier K, Biancalana V, Pinoit JM, Julia S, Chelly J, Devys D, Mandel JL. ARX polyalanine expansions are highly implicated in familial cases of mental retardation with infantile epilepsy and/or hand dystonia. Am J Med Genet A 2010; 155A:98-105. [DOI: 10.1002/ajmg.a.33785] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
18
|
Abstract
Intellectual disability (ID) is the leading socio-economic problem of health care, but compared to autism and schizophrenia, it has received very little public attention. Important risk factors for ID are malnutrition, cultural deprivation, poor health care, and parental consanguinity. In the Western world, fetal alcohol exposure is the most common preventable cause. Most severe forms of ID have genetic causes. Cytogenetically detectable and submicroscopic chromosomal rearrangements account for approximately 25% of all cases. X-linked gene defects are responsible in 10-12% of males with ID; to date, 91 of these defects have been identified. In contrast, autosomal gene defects have been largely disregarded, but due to coordinated efforts and the advent of next-generation DNA sequencing, this is about to change. As shown for Fra(X) syndrome, this renewed focus on autosomal gene defects will pave the way for molecular diagnosis and prevention, shed more light on the pathogenesis of ID, and reveal new opportunities for therapy.
Collapse
|
19
|
Kaufman L, Ayub M, Vincent JB. The genetic basis of non-syndromic intellectual disability: a review. J Neurodev Disord 2010; 2:182-209. [PMID: 21124998 PMCID: PMC2974911 DOI: 10.1007/s11689-010-9055-2] [Citation(s) in RCA: 172] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2010] [Accepted: 06/25/2010] [Indexed: 11/06/2022] Open
Abstract
Intellectual disability (ID), also referred to as mental retardation (MR), is frequently the result of genetic mutation. Where ID is present together with additional clinical symptoms or physical anomalies, there is often sufficient information available for the diagnosing physician to identify a known syndrome, which may then educe the identification of the causative defect. However, where co-morbid features are absent, narrowing down a specific gene can only be done by ‘brute force’ using the latest molecular genetic techniques. Here we attempt to provide a systematic review of genetic causes of cases of ID where no other symptoms or co-morbid features are present, or non-syndromic ID. We attempt to summarize commonalities between the genes and the molecular pathways of their encoded proteins. Since ID is a common feature of autism, and conversely autistic features are frequently present in individuals with ID, we also look at possible overlaps in genetic etiology with non-syndromic ID.
Collapse
|
20
|
Pereira PM, Schneider A, Pannetier S, Heron D, Hanauer A. Coffin-Lowry syndrome. Eur J Hum Genet 2009; 18:627-33. [PMID: 19888300 DOI: 10.1038/ejhg.2009.189] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Coffin-Lowry syndrome (CLS) is a syndromic form of X-linked mental retardation, which is characterized in male patients by psychomotor and growth retardation and various skeletal anomalies. Typical facial changes and specific clinical and radiological signs in the hand are useful aids in the diagnosis. CLS is caused by mutations in the RPS6KA3 gene located at Xp22.2, which encodes RSK2, a growth-factor-regulated protein kinase. RPS6KA3 mutations are extremely heterogeneous and lead to loss of phosphotransferase activity in the RSK2 kinase, most often because of premature termination of translation.
Collapse
|
21
|
Gécz J, Shoubridge C, Corbett M. The genetic landscape of intellectual disability arising from chromosome X. Trends Genet 2009; 25:308-16. [PMID: 19556021 DOI: 10.1016/j.tig.2009.05.002] [Citation(s) in RCA: 140] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2009] [Revised: 05/14/2009] [Accepted: 05/14/2009] [Indexed: 01/07/2023]
Abstract
X-linked mental retardation (XLMR) or intellectual disability (ID) is a common, clinically complex and genetically heterogeneous disease arising from many mutations along the X chromosome. It affects between 1/600-1/1000 males and a substantial number of females. Research during the past decade has identified >90 different XLMR genes, affecting a wide range of cellular processes. Many more genes remain uncharacterized, especially for the non-syndromic XLMR forms. Currently, approximately 11% of X-chromosome genes are implicated in XLMR; however, apart from a few notable exceptions, most contribute individually to <0.1% of the total landscape, which arguably remains only about half complete. There remain many hills to climb and valleys to cross before the ID landscape is fully triangulated.
Collapse
Affiliation(s)
- Jozef Gécz
- Molecular Pathology, SA Pathology at Women's and Children's Hospital, North Adelaide, SA 5006, Australia
| | | | | |
Collapse
|
22
|
Mulley JC. Forty Years From Markers to Genes. Twin Res Hum Genet 2008; 11:368-83. [DOI: 10.1375/twin.11.4.368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
AbstractThere have been incredible advances made in human genetics over the past 40 years. I have set out in the next few pages to describe just some of these changes and to illustrate how they unfolded through my own experiences.
Collapse
|
23
|
Abidi FE, Miano MG, Murray JC, Schwartz CE. A novel mutation in the PHF8 gene is associated with X-linked mental retardation with cleft lip/cleft palate. Clin Genet 2007; 72:19-22. [PMID: 17594395 PMCID: PMC2570350 DOI: 10.1111/j.1399-0004.2007.00817.x] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Recently, two truncating mutations in the PHF8 (plant homeodomain finger protein 8) gene have been found to cause X-linked mental retardation associated with cleft lip/cleft palate (CL/P). One of the truncating mutations was found in the original family with Siderius-Hamel CL/P syndrome where only two of the three affected individuals had mental retardation (MR) with CL/P and one individual had mild MR. The second mutation was present in a family with four affected men, three of whom had MR and CL/P, while the fourth individual had mild MR without clefting. Here, we report a novel nonsense mutation (p.K177X) in a male patient who has MR associated with CL/P. The mutation results in a truncated PHF8 protein lacking the Jumonji-like C terminus domain and five nuclear localization signals. Our finding further supports the hypothesis that the PHF8 protein may play an important role in cognitive function and midline formation.
Collapse
Affiliation(s)
- F E Abidi
- Center for Molecular Studies, J C Self Research Institute, Greenwood Genetic Center, Greenwood, SC 29646, USA.
| | | | | | | |
Collapse
|
24
|
Kaphzan H, Doron G, Rosenblum K. Co-application of NMDA and dopamine-induced rapid translation of RSK2 in the mature hippocampus. J Neurochem 2007; 103:388-99. [PMID: 17645456 DOI: 10.1111/j.1471-4159.2007.04774.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Ribosomal S6 kinase2 (RSK2) is known to take part in several signal transduction cascades including Mitogen Activated Protein Kinase/Extracellular Regulated Kinase (MAPK/ERK). Following our recent observation that ERK can serve as a coincidence detector for fast and slow neurotransmission in the hippocampus, we analyzed the status of RSK2 phosphorylation subsequent to application of NMDA, dopamine, or both to preparations of mature hippocampal slices in Sprague-Dawley rats. RSK2 was indeed phosphorylated; however, in addition, the amount of RSK2 protein (60%) was induced within 10 min following stimulation. Moreover, the induced expression of RSK2 could be detected in both the cell body layer and the dendrites of hippocampal CA1 cells. Pharmacological analysis showed that RSK2 induction was MAPK ERK Kinase (MEK)-ERK independent, but mammalian Target of Rapamycin (mTOR) and translation dependent. We suggest that the fast kinetics of RSK2 translation that follows physiological stimulations, together with recent observations that its over-expression is vital for the attenuation of major signal transduction cascades, indicate an expanded physiological function of RSK2 in neurons, and sheds new light on the role of RSK2 in the Coffin-Lowry syndrome.
Collapse
Affiliation(s)
- Hanoch Kaphzan
- Department of Neurobiology and Ethology, Faculty of Science, Haifa University, Haifa, Israel
| | | | | |
Collapse
|
25
|
Micheli V, Sestini S, Parri V, Fichera M, Romano C, Ariani F, Longo I, Mari F, Bruttini M, Renieri A, Meloni I. RSK2 enzymatic assay as a second level diagnostic tool in Coffin-Lowry syndrome. Clin Chim Acta 2007; 384:35-40. [PMID: 17586481 DOI: 10.1016/j.cca.2007.05.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2007] [Revised: 04/17/2007] [Accepted: 05/14/2007] [Indexed: 10/23/2022]
Abstract
BACKGROUND Coffin-Lowry syndrome is a semi-dominant condition characterized by severe psychomotor retardation with facial, hand and skeletal malformations resulting from mutations in RSK2 gene, encoding for a serine/threonine kinase. More than 100 different mutations have been identified to date; however, about 50% of clinically diagnosed patients apparently do not have mutations. In order to exclude that these patients have RSK2 mutations missed by standard mutation detection techniques, a rapid and sensitive assay allowing evaluation of RSK2 activity was needed. METHODS RSK2 capacity to phosphorylate a synthetic CREB-peptide in basal and PMA-stimulated conditions was evaluated in lymphoblasts from 3 patients with RSK2 mutations and normal controls. RESULTS Patients RSK2 activity is normal in nonstimulated conditions but fails to grow following stimulation. The evaluation of the stimulated/non-stimulated activity ratio demonstrated a statistically significant impairment in patients. CONCLUSIONS We have set up an assay which allows the identification of even partial alterations of RSK2 activity and seems to give good results also in females with a balanced X-chromosome inactivation and thus with a presumably normal enzymatic activity in about 50% of cells. Moreover, our data seem to confirm previous reports of a potential direct correlation between the level of RSK2 activity and the severity of cognitive impairment.
Collapse
Affiliation(s)
- Vanna Micheli
- Biological Chemistry, Department of Molecular Biology, University of Siena, Siena, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Laumonnier F, Cuthbert PC, Grant SGN. The role of neuronal complexes in human X-linked brain diseases. Am J Hum Genet 2007; 80:205-20. [PMID: 17236127 PMCID: PMC1785339 DOI: 10.1086/511441] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2006] [Accepted: 11/28/2006] [Indexed: 01/28/2023] Open
Abstract
Beyond finding individual genes that are involved in medical disorders, an important challenge is the integration of sets of disease genes with the complexities of basic biological processes. We examine this issue by focusing on neuronal multiprotein complexes and their components encoded on the human X chromosome. Multiprotein signaling complexes in the postsynaptic terminal of central nervous system synapses are essential for the induction of neuronal plasticity and cognitive processes in animals. The prototype complex is the N-methyl-D-aspartate receptor complex/membrane-associated guanylate kinase-associated signaling complex (NRC/MASC) comprising 185 proteins and embedded within the postsynaptic density (PSD), which is a set of complexes totaling approximately 1,100 proteins. It is striking that 86% (6 of 7) of X-linked NRC/MASC genes and 49% (19 of 39) of X-chromosomal PSD genes are already known to be involved in human psychiatric disorders. Moreover, of the 69 known proteins mutated in X-linked mental retardation, 19 (28%) encode postsynaptic proteins. The high incidence of involvement in cognitive disorders is also found in mouse mutants and indicates that the complexes are functioning as integrated entities or molecular machines and that disruption of different components impairs their overall role in cognitive processes. We also noticed that NRC/MASC genes appear to be more strongly associated with mental retardation and autism spectrum disorders. We propose that systematic studies of PSD and NRC/MASC genes in mice and humans will give a high yield of novel genes important for human disease and new mechanistic insights into higher cognitive functions.
Collapse
Affiliation(s)
- Frédéric Laumonnier
- Genes to Cognition Programme, Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridge, UK
| | | | | |
Collapse
|
27
|
Tarpey PS, Raymond FL, O'Meara S, Edkins S, Teague J, Butler A, Dicks E, Stevens C, Tofts C, Avis T, Barthorpe S, Buck G, Cole J, Gray K, Halliday K, Harrison R, Hills K, Jenkinson A, Jones D, Menzies A, Mironenko T, Perry J, Raine K, Richardson D, Shepherd R, Small A, Varian J, West S, Widaa S, Mallya U, Moon J, Luo Y, Holder S, Smithson SF, Hurst JA, Clayton-Smith J, Kerr B, Boyle J, Shaw M, Vandeleur L, Rodriguez J, Slaugh R, Easton DF, Wooster R, Bobrow M, Srivastava AK, Stevenson RE, Schwartz CE, Turner G, Gecz J, Futreal PA, Stratton MR, Partington M. Mutations in CUL4B, which encodes a ubiquitin E3 ligase subunit, cause an X-linked mental retardation syndrome associated with aggressive outbursts, seizures, relative macrocephaly, central obesity, hypogonadism, pes cavus, and tremor. Am J Hum Genet 2007; 80:345-52. [PMID: 17236139 PMCID: PMC1785336 DOI: 10.1086/511134] [Citation(s) in RCA: 180] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2006] [Accepted: 11/20/2006] [Indexed: 12/23/2022] Open
Abstract
We have identified three truncating, two splice-site, and three missense variants at conserved amino acids in the CUL4B gene on Xq24 in 8 of 250 families with X-linked mental retardation (XLMR). During affected subjects' adolescence, a syndrome emerged with delayed puberty, hypogonadism, relative macrocephaly, moderate short stature, central obesity, unprovoked aggressive outbursts, fine intention tremor, pes cavus, and abnormalities of the toes. This syndrome was first described by Cazebas et al., in a family that was included in our study and that carried a CUL4B missense variant. CUL4B is a ubiquitin E3 ligase subunit implicated in the regulation of several biological processes, and CUL4B is the first XLMR gene that encodes an E3 ubiquitin ligase. The relatively high frequency of CUL4B mutations in this series indicates that it is one of the most commonly mutated genes underlying XLMR and suggests that its introduction into clinical diagnostics should be a high priority.
Collapse
Affiliation(s)
- Patrick S Tarpey
- Cancer Genome Project, Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Field M, Tarpey P, Boyle J, Edkins S, Goodship J, Luo Y, Moon J, Teague J, Stratton MR, Futreal PA, Wooster R, Raymond FL, Turner G. Mutations in the RSK2(RPS6KA3) gene cause Coffin-Lowry syndrome and nonsyndromic X-linked mental retardation. Clin Genet 2007; 70:509-15. [PMID: 17100996 PMCID: PMC2714973 DOI: 10.1111/j.1399-0004.2006.00723.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
We describe three families with X-linked mental retardation, two with a deletion of a single amino acid and one with a missense mutation in the proximal domain of the RSK2(RPS6KA3) (ribosomal protein S6 kinase, 90 kDa, polypeptide 3) protein similar to mutations found in Coffin-Lowry syndrome (CLS). In two families, the clinical diagnosis had been nonsyndromic X-linked mental retardation. In the third family, although CLS had been suspected, the clinical features were atypical and the degree of intellectual disability much less than expected. These families show that strict reliance on classical clinical criteria for mutation testing may result in a missed diagnosis. A less targeted screening approach to mutation testing is advocated.
Collapse
Affiliation(s)
- M Field
- The NSW GOLD Service, Hunter Genetics, Newcastle, Australia.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Abstract
Genetic abnormalities frequently give rise to a mental retardation phenotype. Recent advances in resolution of comparative genomic hybridization and genomic sequence annotation has identified new syndromes at chromosome 3q29 and 9q34. The finding of a significant number of copy number polymorphisms in the genome in the normal population, means that assigning pathogenicity to deletions and duplications in patients with mental retardation can be difficult but has been identified for duplications of MECP2 and L1CAM. Novel autosomal genes that cause mental retardation have been identified recently including CC2D1A identified by homozygosity mapping. Several new genes and pathways have been identified in the field of X-linked mental retardation but many more still await identification. Analysis of families where only a single male is affected reveals that the chance of this being due to a single X-linked gene abnormality is significantly less than would be expected if the excess of males in the population is entirely due to X-linked disease. Recent identification of novel X-linked mental retardation genes has identified components of the post-synaptic density and multiple zinc finger transcription factors as disease causing suggesting new mechanisms of disease causation. The first therapeutic treatments of animal models of mental retardation have been reported, a Drosophila model of Fragile X syndrome has been treated with lithium or metabotropic glutamate receptor (mGluR) antagonists and a mouse model of NF1 has been treated with the HMG-CoA reductase inhibitor lavastatin, which improves the learning and memory skills in these models.
Collapse
Affiliation(s)
- F Lucy Raymond
- Department of Medical Genetics, Cambridge Institute of Medical Research, University of Cambridge, Addenbrookes Hospital, Cambridge CB2 2XY, UK.
| | | |
Collapse
|
30
|
Bahi-Buisson N, Chelly J, des Portes V. [Update on the genetics of X-linked mental retardation]. Rev Neurol (Paris) 2006; 162:952-63. [PMID: 17028563 DOI: 10.1016/s0035-3787(06)75105-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Mutations in X-linked genes are likely to account for the observation that more males than females are affected with mental retardation. Causative mutations have been identified in both syndromic XLMR and in the genetically heterogeneous non-syndromic forms of XLMR, without a clear clinical phenotype other than cognitive deficit. Progress in genome analysis and the establishment of large collaborations between clinical and molecular research teams, especially the European XLMR consortium, have led to the identification of 20 non-syndromic XLMR genes and 25 syndromic XLMR genes. Given the extensive heterogeneity of non syndromic XLMR, different strategies are used for the identification of new genes: linkage analysis, studies of balanced chromosomal rearrangements (X-autosome translocations, microdeletions) and candidate genes strategies by mutation screening in regions of the X chromosome known to be involved in neuronal development and function. Delineating the monogenic causes of XLMR and their molecular and cellular consequences will provide insight into the mechanisms that are required for normal development of cognitive function in humans. Non syndromic XLMR proteins include 5 distinct classes: transmembrane receptors, small GTPases effectors or regulators, enzymes and translational regulators.
Collapse
Affiliation(s)
- N Bahi-Buisson
- Département de Pédiatrie, Service de Neuropédiatrie et Maladies Métaboliques, Hôpital Necker, and Université René Descartes, Paris, France.
| | | | | |
Collapse
|
31
|
Kleefstra T, Smidt M, Banning MJG, Oudakker AR, Van Esch H, de Brouwer APM, Nillesen W, Sistermans EA, Hamel BCJ, de Bruijn D, Fryns JP, Yntema HG, Brunner HG, de Vries BBA, van Bokhoven H. Disruption of the gene Euchromatin Histone Methyl Transferase1 (Eu-HMTase1) is associated with the 9q34 subtelomeric deletion syndrome. J Med Genet 2006; 42:299-306. [PMID: 15805155 PMCID: PMC1736026 DOI: 10.1136/jmg.2004.028464] [Citation(s) in RCA: 129] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
BACKGROUND A new syndrome has been recognised following thorough analysis of patients with a terminal submicroscopic subtelomeric deletion of chromosome 9q. These have in common severe mental retardation, hypotonia, brachycephaly, flat face with hypertelorism, synophrys, anteverted nares, thickened lower lip, carp mouth with macroglossia, and conotruncal heart defects. The minimum critical region responsible for this 9q subtelomeric deletion syndrome (9q-) is approximately 1.2 Mb and encompasses at least 14 genes. OBJECTIVE To characterise the breakpoints of a de novo balanced translocation t(X;9)(p11.23;q34.3) in a mentally retarded female patient with clinical features similar to the 9q- syndrome. RESULTS Sequence analysis of the break points showed that the translocation was fully balanced and only one gene on chromosome 9 was disrupted--Euchromatin Histone Methyl Transferase1 (Eu-HMTase1)--encoding a histone H3 lysine 9 methyltransferase (H3-K9 HMTase). This indicates that haploinsufficiency of Eu-HMTase1 is responsible for the 9q submicroscopic subtelomeric deletion syndrome. This observation was further supported by the spatio-temporal expression of the gene. Using tissue in situ hybridisation studies in mouse embryos and adult brain, Eu-HMTase1 was shown to be expressed in the developing nervous system and in specific peripheral tissues. While expression is selectively downregulated in adult brain, substantial expression is retained in the olfactory bulb, anterior/ventral lateral ventricular wall, and hippocampus and weakly in the piriform cortex. CONCLUSIONS The expression pattern of this gene suggests a role in the CNS development and function, which is in line with the severe mental retardation and behaviour problems in patients who lack one copy of the gene.
Collapse
Affiliation(s)
- T Kleefstra
- Department of Human Genetics, University Medical Centre St Radboud, Nijmegen, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Delaunoy JP, Dubos A, Marques Pereira P, Hanauer A. Identification of novel mutations in the RSK2 gene (RPS6KA3) in patients with Coffin-Lowry syndrome. Clin Genet 2006; 70:161-6. [PMID: 16879200 DOI: 10.1111/j.1399-0004.2006.00660.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The Coffin-Lowry syndrome (CLS) is a rare X-linked semidominant syndrome characterized by severe psychomotor retardation, facial dysmorphism, digit abnormalities and progressive skeletal deformations. CLS is caused by mutations in a gene located in Xp22.2, RPS6KA3. This gene encodes for a growth factor-regulated serine/threonine protein kinase, RSK2 (ribosomal S6 kinase 2), acting in the Ras-mitogen-activated protein kinase signaling pathway. Mutations in the RPS6KA3 gene are extremely heterogeneous and lead to premature termination of translation and/or to loss of phosphotransferase activity of the RSK2 protein. Screening for RSK2 mutations is essential in most cases to confirm the diagnosis as well as for genetic counseling. Here we present 44 novel mutations in RSK2 causing CLS. The overall number of CLS mutations reported now is 128. Thirty-three percent of mutations are missense mutations, 15% nonsense mutations, 20% splicing errors and 29% short deletion or insertion events. Only four large deletions have so far been found. They are distributed throughout the RPS6KA3 gene, and the majority has been found in a single family. This study further confirms the high rate of new mutations at the RSK2 locus. It is important to consider the possibility of mosaicism when providing genetic counseling in CLS families.
Collapse
Affiliation(s)
- J P Delaunoy
- Laboratoire de Diagnostic Génétique, Faculté de Médecine et CHRU, Strasbourg, France
| | | | | | | |
Collapse
|
33
|
Ropers HH. X-linked mental retardation: many genes for a complex disorder. Curr Opin Genet Dev 2006; 16:260-9. [PMID: 16647850 DOI: 10.1016/j.gde.2006.04.017] [Citation(s) in RCA: 126] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2006] [Accepted: 04/18/2006] [Indexed: 11/26/2022]
Abstract
X-linked mental retardation (XLMR) is a common cause of moderate to severe intellectual disability in males. XLMR is very heterogeneous, and about two-thirds of patients have clinically indistinguishable non-syndromic (NS-XLMR) forms, which has greatly hampered their molecular elucidation. A few years ago, international consortia overcame this impasse by collecting DNA and cell lines from large cohorts of XLMR families, thereby paving the way for the systematic study of the molecular causes of XLMR. Mutations in known genes might already account for 50% of the families with NS-XLMR, and various genes have been pinpointed that seem to be of particular diagnostic importance. Eventually, even therapy of XLMR might become possible, as suggested by the unexpected plasticity of the neuronal wiring in the brain, and the recent successful drug treatment of a fly model for fragile X syndrome.
Collapse
Affiliation(s)
- Hans-Hilger Ropers
- Max-Planck-Institute for Molecular Genetics, Ihnestrasse 73, D-14195 Berlin, Germany.
| |
Collapse
|
34
|
Falco M, Romano C, Alberti A, Greco D, Scuderi C, Avola E, Failla P, Belli S, Tolmie JL, Amata S, Fichera M. Identification of Novel Mutations in Patients with Coffin–Lowry Syndrome by a Denaturing HPLC-Based Assay. Clin Chem 2005; 51:2356-8. [PMID: 16306095 DOI: 10.1373/clinchem.2005.056036] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Michele Falco
- Laboratorio di Diagnosi Genetica, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Oasi Maria SS, Troina, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
Mental retardation is more common in males than females in the population, assumed to be due to mutations on the X chromosome. The prevalence of the 24 genes identified to date is low and less common than expansions in FMR1, which cause Fragile X syndrome. Systematic screening of all other X linked genes in X linked families with mental retardation is currently not feasible in a clinical setting. The phenotypes of genes causing syndromic and non-syndromic mental retardation (NLGN3, NLGN4, RPS6KA3(RSK2), OPHN1, ATRX, SLC6A8, ARX, SYN1, AGTR2, MECP2, PQBP1, SMCX, and SLC16A2) are first discussed, as these may be the focus of more targeted mutation analysis. Secondly, the relative prevalence of genes causing only non-syndromic mental retardation (IL1RAPL1, TM4SF2, ZNF41, FTSJ1, DLG3, FACL4, PAK3, ARHGEF6, FMR2, and GDI) is summarised. Thirdly, the problem of recurrence risk where a molecular genetics diagnosis has not been made and what proportion of the male excess of mental retardation is due to monogenic disorders of the X chromosome are discussed.
Collapse
Affiliation(s)
- F L Raymond
- Cambridge Institute of Medical Research, Department of Medical Genetics, University of Cambridge, Addenbrooke's Hospital, Cambridge, CB2 2XY, UK.
| |
Collapse
|
36
|
Cantagrel V, Lossi AM, Boulanger S, Depetris D, Mattei MG, Gecz J, Schwartz CE, Van Maldergem L, Villard L. Disruption of a new X linked gene highly expressed in brain in a family with two mentally retarded males. J Med Genet 2005; 41:736-42. [PMID: 15466006 PMCID: PMC1735597 DOI: 10.1136/jmg.2004.021626] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Mental retardation (MR) affects 2-3% of the human population and some of these cases are genetically determined. Although several genes responsible for MR have been identified, many cases have still not been explained. METHODS We have identified a pericentric inversion of the X chromosome inv(X)(p22.3;q13.2) segregating in a family where two male carriers have severe MR while female carriers are not affected. RESULTS The molecular characterisation of this inversion led us to identify two new genes which are disrupted by the breakpoints: KIAA2022 in Xq13.2 and P2RY8 in Xp22.3. These genes were not previously fully characterised in humans. KIAA2022 encodes a protein which lacks significant homology to any other known protein and is highly expressed in the brain. P2RY8 is a member of the purine nucleotide G-protein coupled receptor gene family. It is located in the pseudo-autosomal region of the X chromosome and is not expressed in brain. CONCLUSIONS Because the haploinsufficiency of P2RY8 in carrier mothers does not have a phenotypic consequence, we propose that the severe MR of the affected males in this family is due to the absence of the KIAA2022 gene product. However, screening 20 probands from X linked MR families did not reveal mutations in KIAA2022. Nonetheless, the high expression of this gene in fetal brain and in the adult cerebral cortex could be consistent with a role in brain development and/or cognitive function.
Collapse
Affiliation(s)
- V Cantagrel
- Inserm U491, Faculté de Médecine de La Timone, 27, Bd. Jean Moulin, 13385 Marseille Cedex 5, France
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Mandel JL, Chelly J. Monogenic X-linked mental retardation: is it as frequent as currently estimated? The paradox of the ARX (Aristaless X) mutations. Eur J Hum Genet 2005; 12:689-93. [PMID: 15319782 DOI: 10.1038/sj.ejhg.5201247] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Mental retardation affects 30 to 50% more males than females, and X-linked mental retardation (XLMR) is thought to account for the major part of this sex bias. Nonsyndromic XLMR is very heterogeneous, with more than 15 genes identified to date, each of them accounting for a very small proportion of nonsyndromic families. The Aristaless X (ARX) gene is an exception since it was found mutated in 11 of 136 such families, with a highly recurrent mutation (dup24) leading to an expansion of a polyalanine tract in the protein. The rather high frequency of dup24 reported in families with clear X-linked MR (6.6%) contrasts with the very low prevalence of this mutation observed in sporadic male MR (0.13%). We conclude that monogenic XLMR has much lower prevalence in male MR (< 10%) than the 23% that would be required to account for a 30% male excess of mental retardation.
Collapse
Affiliation(s)
- Jean-Louis Mandel
- Institut de Génétique et Biologie Moléculaire et Cellulaire (IGBMC) (CNRS/INSERM/Université Louis Pasteur, Collège de France), 67404 Illkirch/CU Strasbourg, France.
| | | |
Collapse
|
38
|
Ramser J, Abidi FE, Burckle CA, Lenski C, Toriello H, Wen G, Lubs HA, Engert S, Stevenson RE, Meindl A, Schwartz CE, Nguyen G. A unique exonic splice enhancer mutation in a family with X-linked mental retardation and epilepsy points to a novel role of the renin receptor. Hum Mol Genet 2005; 14:1019-27. [PMID: 15746149 DOI: 10.1093/hmg/ddi094] [Citation(s) in RCA: 142] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The renin-angiotensin system (RAS) is essential for blood pressure control and water-electrolyte balance. Until the discovery of the renin receptor, renin was believed to be mainly a circulating enzyme with a unique function, the cleavage of angiotensinogen. We report a unique mutation in the renin receptor gene (ATP6AP2) present in patients with X-linked mental retardation and epilepsy (OMIM no. 300423), but absent in 1200 control X-chromosomes. A silent mutation (c.321C>T, p.D107D) residing in a putative exonic splicing enhancer site resulted in inefficient inclusion of exon 4 in 50% of renin receptor mRNA, as demonstrated by quantitative RT-PCR. Analysis of membrane associated-receptor molecular forms showed the presence of full-length and truncated proteins in the patient. Functional analysis demonstrated that the mutated receptor could bind renin and increase renin catalytic activity, similar to the wild-type receptor, but resulted in a modest and reproducible impairment of ERK1/2 activation. Thus, our findings confirm the importance of the RAS in cognitive processes and indicate a novel specific role for the renin receptor in cognitive functions and brain development.
Collapse
Affiliation(s)
- Juliane Ramser
- 1Institute of Human Genetics, Ludwig-Maximilians-University, Munich 80336, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Kleefstra T, Franken CE, Arens YHJM, Ramakers GJA, Yntema HG, Sistermans EA, Hulsmans CFCH, Nillesen WN, van Bokhoven H, de Vries BBA, Hamel BCJ. Genotype-phenotype studies in three families with mutations in the polyglutamine-binding protein 1 gene (PQBP1). Clin Genet 2005; 66:318-26. [PMID: 15355434 DOI: 10.1111/j.1399-0004.2004.00308.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Recently, the polyglutamine-binding protein 1 (PQBP1) gene was found to be mutated in five of 29 families studied with X-linked mental retardation (XLMR) linked to Xp. The reported mutations include duplications or deletions of AG dinucleotides in the fourth coding exon that resulted in shifts of the open reading frame. Three of the five families with mutations in this newly identified XLMR gene have been reported previously. We characterized the phenotypic and neuropsychological features in the two unpublished families with aberrations in PQBP1 and in a family reported 10 years ago. In total, seven patients diagnosed with aberrations in this gene were examined, including a newly identified patient at 18 months of age. Additionally, the features were compared to those reported in the literature of three other families, comprising MRXS3 (Sutherland-Haan syndrome) MRX55 and MRXS8 (Renpenning syndrome). Characteristics seen in these patients are microcephaly, lean body habitus, short stature, striking facial appearance with long narrow faces, upward slant of the eyes, malar hypoplasia, prognathism, high-arched palate and nasal speech. In addition, small testes and midline defects as anal atresia or imperforate anus, clefting of palate and/or uvula, iris coloboma and Tetralogy of Fallot are seen in several patients. These observations contribute to the phenotypic knowledge of patients with PQBP1 mutations and make this XLMR syndrome well recognizable to clinicians.
Collapse
Affiliation(s)
- T Kleefstra
- Department of Human Genetics, University Medical Center, Nijmegen, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Jensen LR, Amende M, Gurok U, Moser B, Gimmel V, Tzschach A, Janecke AR, Tariverdian G, Chelly J, Fryns JP, Van Esch H, Kleefstra T, Hamel B, Moraine C, Gécz J, Turner G, Reinhardt R, Kalscheuer VM, Ropers HH, Lenzner S. Mutations in the JARID1C gene, which is involved in transcriptional regulation and chromatin remodeling, cause X-linked mental retardation. Am J Hum Genet 2005; 76:227-36. [PMID: 15586325 PMCID: PMC1196368 DOI: 10.1086/427563] [Citation(s) in RCA: 281] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2004] [Accepted: 11/17/2004] [Indexed: 11/03/2022] Open
Abstract
In families with nonsyndromic X-linked mental retardation (NS-XLMR), >30% of mutations seem to cluster on proximal Xp and in the pericentric region. In a systematic screen of brain-expressed genes from this region in 210 families with XLMR, we identified seven different mutations in JARID1C, including one frameshift mutation and two nonsense mutations that introduce premature stop codons, as well as four missense mutations that alter evolutionarily conserved amino acids. In two of these families, expression studies revealed the almost complete absence of the mutated JARID1C transcript, suggesting that the phenotype in these families results from functional loss of the JARID1C protein. JARID1C (Jumonji AT-rich interactive domain 1C), formerly known as "SMCX," is highly similar to the Y-chromosomal gene JARID1D/SMCY, which encodes the H-Y antigen. The JARID1C protein belongs to the highly conserved ARID protein family. It contains several DNA-binding motifs that link it to transcriptional regulation and chromatin remodeling, processes that are defective in various other forms of mental retardation. Our results suggest that JARID1C mutations are a relatively common cause of XLMR and that this gene might play an important role in human brain function.
Collapse
Affiliation(s)
- Lars Riff Jensen
- Max Planck Institute for Molecular Genetics, Berlin; Department of Medical Biology and Human Genetics, Innsbruck Medical University, Innsbruck; Institute for Human Genetics, University Heidelberg, Heidelberg; Institut Cochin de Génétique Moleculaire, CNRS/INSERM, CHU Cochin, Paris; Center for Human Genetics, Clinical Genetics Unit, Leuven, Belgium; Department of Human Genetics, University Medical Centre, Nijmegen, The Netherlands; Services de Génétique-INSERM U316, CHU Bretonneau, Tours, France; Women’s and Children’s Hospital and the University of Adelaide, Adelaide; and Genetics of Learning Disability (GOLD) Service, Hunter Genetics, University of Newcastle, New South Wales, Australia
| | - Marion Amende
- Max Planck Institute for Molecular Genetics, Berlin; Department of Medical Biology and Human Genetics, Innsbruck Medical University, Innsbruck; Institute for Human Genetics, University Heidelberg, Heidelberg; Institut Cochin de Génétique Moleculaire, CNRS/INSERM, CHU Cochin, Paris; Center for Human Genetics, Clinical Genetics Unit, Leuven, Belgium; Department of Human Genetics, University Medical Centre, Nijmegen, The Netherlands; Services de Génétique-INSERM U316, CHU Bretonneau, Tours, France; Women’s and Children’s Hospital and the University of Adelaide, Adelaide; and Genetics of Learning Disability (GOLD) Service, Hunter Genetics, University of Newcastle, New South Wales, Australia
| | - Ulf Gurok
- Max Planck Institute for Molecular Genetics, Berlin; Department of Medical Biology and Human Genetics, Innsbruck Medical University, Innsbruck; Institute for Human Genetics, University Heidelberg, Heidelberg; Institut Cochin de Génétique Moleculaire, CNRS/INSERM, CHU Cochin, Paris; Center for Human Genetics, Clinical Genetics Unit, Leuven, Belgium; Department of Human Genetics, University Medical Centre, Nijmegen, The Netherlands; Services de Génétique-INSERM U316, CHU Bretonneau, Tours, France; Women’s and Children’s Hospital and the University of Adelaide, Adelaide; and Genetics of Learning Disability (GOLD) Service, Hunter Genetics, University of Newcastle, New South Wales, Australia
| | - Bettina Moser
- Max Planck Institute for Molecular Genetics, Berlin; Department of Medical Biology and Human Genetics, Innsbruck Medical University, Innsbruck; Institute for Human Genetics, University Heidelberg, Heidelberg; Institut Cochin de Génétique Moleculaire, CNRS/INSERM, CHU Cochin, Paris; Center for Human Genetics, Clinical Genetics Unit, Leuven, Belgium; Department of Human Genetics, University Medical Centre, Nijmegen, The Netherlands; Services de Génétique-INSERM U316, CHU Bretonneau, Tours, France; Women’s and Children’s Hospital and the University of Adelaide, Adelaide; and Genetics of Learning Disability (GOLD) Service, Hunter Genetics, University of Newcastle, New South Wales, Australia
| | - Verena Gimmel
- Max Planck Institute for Molecular Genetics, Berlin; Department of Medical Biology and Human Genetics, Innsbruck Medical University, Innsbruck; Institute for Human Genetics, University Heidelberg, Heidelberg; Institut Cochin de Génétique Moleculaire, CNRS/INSERM, CHU Cochin, Paris; Center for Human Genetics, Clinical Genetics Unit, Leuven, Belgium; Department of Human Genetics, University Medical Centre, Nijmegen, The Netherlands; Services de Génétique-INSERM U316, CHU Bretonneau, Tours, France; Women’s and Children’s Hospital and the University of Adelaide, Adelaide; and Genetics of Learning Disability (GOLD) Service, Hunter Genetics, University of Newcastle, New South Wales, Australia
| | - Andreas Tzschach
- Max Planck Institute for Molecular Genetics, Berlin; Department of Medical Biology and Human Genetics, Innsbruck Medical University, Innsbruck; Institute for Human Genetics, University Heidelberg, Heidelberg; Institut Cochin de Génétique Moleculaire, CNRS/INSERM, CHU Cochin, Paris; Center for Human Genetics, Clinical Genetics Unit, Leuven, Belgium; Department of Human Genetics, University Medical Centre, Nijmegen, The Netherlands; Services de Génétique-INSERM U316, CHU Bretonneau, Tours, France; Women’s and Children’s Hospital and the University of Adelaide, Adelaide; and Genetics of Learning Disability (GOLD) Service, Hunter Genetics, University of Newcastle, New South Wales, Australia
| | - Andreas R. Janecke
- Max Planck Institute for Molecular Genetics, Berlin; Department of Medical Biology and Human Genetics, Innsbruck Medical University, Innsbruck; Institute for Human Genetics, University Heidelberg, Heidelberg; Institut Cochin de Génétique Moleculaire, CNRS/INSERM, CHU Cochin, Paris; Center for Human Genetics, Clinical Genetics Unit, Leuven, Belgium; Department of Human Genetics, University Medical Centre, Nijmegen, The Netherlands; Services de Génétique-INSERM U316, CHU Bretonneau, Tours, France; Women’s and Children’s Hospital and the University of Adelaide, Adelaide; and Genetics of Learning Disability (GOLD) Service, Hunter Genetics, University of Newcastle, New South Wales, Australia
| | - Gholamali Tariverdian
- Max Planck Institute for Molecular Genetics, Berlin; Department of Medical Biology and Human Genetics, Innsbruck Medical University, Innsbruck; Institute for Human Genetics, University Heidelberg, Heidelberg; Institut Cochin de Génétique Moleculaire, CNRS/INSERM, CHU Cochin, Paris; Center for Human Genetics, Clinical Genetics Unit, Leuven, Belgium; Department of Human Genetics, University Medical Centre, Nijmegen, The Netherlands; Services de Génétique-INSERM U316, CHU Bretonneau, Tours, France; Women’s and Children’s Hospital and the University of Adelaide, Adelaide; and Genetics of Learning Disability (GOLD) Service, Hunter Genetics, University of Newcastle, New South Wales, Australia
| | - Jamel Chelly
- Max Planck Institute for Molecular Genetics, Berlin; Department of Medical Biology and Human Genetics, Innsbruck Medical University, Innsbruck; Institute for Human Genetics, University Heidelberg, Heidelberg; Institut Cochin de Génétique Moleculaire, CNRS/INSERM, CHU Cochin, Paris; Center for Human Genetics, Clinical Genetics Unit, Leuven, Belgium; Department of Human Genetics, University Medical Centre, Nijmegen, The Netherlands; Services de Génétique-INSERM U316, CHU Bretonneau, Tours, France; Women’s and Children’s Hospital and the University of Adelaide, Adelaide; and Genetics of Learning Disability (GOLD) Service, Hunter Genetics, University of Newcastle, New South Wales, Australia
| | - Jean-Pierre Fryns
- Max Planck Institute for Molecular Genetics, Berlin; Department of Medical Biology and Human Genetics, Innsbruck Medical University, Innsbruck; Institute for Human Genetics, University Heidelberg, Heidelberg; Institut Cochin de Génétique Moleculaire, CNRS/INSERM, CHU Cochin, Paris; Center for Human Genetics, Clinical Genetics Unit, Leuven, Belgium; Department of Human Genetics, University Medical Centre, Nijmegen, The Netherlands; Services de Génétique-INSERM U316, CHU Bretonneau, Tours, France; Women’s and Children’s Hospital and the University of Adelaide, Adelaide; and Genetics of Learning Disability (GOLD) Service, Hunter Genetics, University of Newcastle, New South Wales, Australia
| | - Hilde Van Esch
- Max Planck Institute for Molecular Genetics, Berlin; Department of Medical Biology and Human Genetics, Innsbruck Medical University, Innsbruck; Institute for Human Genetics, University Heidelberg, Heidelberg; Institut Cochin de Génétique Moleculaire, CNRS/INSERM, CHU Cochin, Paris; Center for Human Genetics, Clinical Genetics Unit, Leuven, Belgium; Department of Human Genetics, University Medical Centre, Nijmegen, The Netherlands; Services de Génétique-INSERM U316, CHU Bretonneau, Tours, France; Women’s and Children’s Hospital and the University of Adelaide, Adelaide; and Genetics of Learning Disability (GOLD) Service, Hunter Genetics, University of Newcastle, New South Wales, Australia
| | - Tjitske Kleefstra
- Max Planck Institute for Molecular Genetics, Berlin; Department of Medical Biology and Human Genetics, Innsbruck Medical University, Innsbruck; Institute for Human Genetics, University Heidelberg, Heidelberg; Institut Cochin de Génétique Moleculaire, CNRS/INSERM, CHU Cochin, Paris; Center for Human Genetics, Clinical Genetics Unit, Leuven, Belgium; Department of Human Genetics, University Medical Centre, Nijmegen, The Netherlands; Services de Génétique-INSERM U316, CHU Bretonneau, Tours, France; Women’s and Children’s Hospital and the University of Adelaide, Adelaide; and Genetics of Learning Disability (GOLD) Service, Hunter Genetics, University of Newcastle, New South Wales, Australia
| | - Ben Hamel
- Max Planck Institute for Molecular Genetics, Berlin; Department of Medical Biology and Human Genetics, Innsbruck Medical University, Innsbruck; Institute for Human Genetics, University Heidelberg, Heidelberg; Institut Cochin de Génétique Moleculaire, CNRS/INSERM, CHU Cochin, Paris; Center for Human Genetics, Clinical Genetics Unit, Leuven, Belgium; Department of Human Genetics, University Medical Centre, Nijmegen, The Netherlands; Services de Génétique-INSERM U316, CHU Bretonneau, Tours, France; Women’s and Children’s Hospital and the University of Adelaide, Adelaide; and Genetics of Learning Disability (GOLD) Service, Hunter Genetics, University of Newcastle, New South Wales, Australia
| | - Claude Moraine
- Max Planck Institute for Molecular Genetics, Berlin; Department of Medical Biology and Human Genetics, Innsbruck Medical University, Innsbruck; Institute for Human Genetics, University Heidelberg, Heidelberg; Institut Cochin de Génétique Moleculaire, CNRS/INSERM, CHU Cochin, Paris; Center for Human Genetics, Clinical Genetics Unit, Leuven, Belgium; Department of Human Genetics, University Medical Centre, Nijmegen, The Netherlands; Services de Génétique-INSERM U316, CHU Bretonneau, Tours, France; Women’s and Children’s Hospital and the University of Adelaide, Adelaide; and Genetics of Learning Disability (GOLD) Service, Hunter Genetics, University of Newcastle, New South Wales, Australia
| | - Jozef Gécz
- Max Planck Institute for Molecular Genetics, Berlin; Department of Medical Biology and Human Genetics, Innsbruck Medical University, Innsbruck; Institute for Human Genetics, University Heidelberg, Heidelberg; Institut Cochin de Génétique Moleculaire, CNRS/INSERM, CHU Cochin, Paris; Center for Human Genetics, Clinical Genetics Unit, Leuven, Belgium; Department of Human Genetics, University Medical Centre, Nijmegen, The Netherlands; Services de Génétique-INSERM U316, CHU Bretonneau, Tours, France; Women’s and Children’s Hospital and the University of Adelaide, Adelaide; and Genetics of Learning Disability (GOLD) Service, Hunter Genetics, University of Newcastle, New South Wales, Australia
| | - Gillian Turner
- Max Planck Institute for Molecular Genetics, Berlin; Department of Medical Biology and Human Genetics, Innsbruck Medical University, Innsbruck; Institute for Human Genetics, University Heidelberg, Heidelberg; Institut Cochin de Génétique Moleculaire, CNRS/INSERM, CHU Cochin, Paris; Center for Human Genetics, Clinical Genetics Unit, Leuven, Belgium; Department of Human Genetics, University Medical Centre, Nijmegen, The Netherlands; Services de Génétique-INSERM U316, CHU Bretonneau, Tours, France; Women’s and Children’s Hospital and the University of Adelaide, Adelaide; and Genetics of Learning Disability (GOLD) Service, Hunter Genetics, University of Newcastle, New South Wales, Australia
| | - Richard Reinhardt
- Max Planck Institute for Molecular Genetics, Berlin; Department of Medical Biology and Human Genetics, Innsbruck Medical University, Innsbruck; Institute for Human Genetics, University Heidelberg, Heidelberg; Institut Cochin de Génétique Moleculaire, CNRS/INSERM, CHU Cochin, Paris; Center for Human Genetics, Clinical Genetics Unit, Leuven, Belgium; Department of Human Genetics, University Medical Centre, Nijmegen, The Netherlands; Services de Génétique-INSERM U316, CHU Bretonneau, Tours, France; Women’s and Children’s Hospital and the University of Adelaide, Adelaide; and Genetics of Learning Disability (GOLD) Service, Hunter Genetics, University of Newcastle, New South Wales, Australia
| | - Vera M. Kalscheuer
- Max Planck Institute for Molecular Genetics, Berlin; Department of Medical Biology and Human Genetics, Innsbruck Medical University, Innsbruck; Institute for Human Genetics, University Heidelberg, Heidelberg; Institut Cochin de Génétique Moleculaire, CNRS/INSERM, CHU Cochin, Paris; Center for Human Genetics, Clinical Genetics Unit, Leuven, Belgium; Department of Human Genetics, University Medical Centre, Nijmegen, The Netherlands; Services de Génétique-INSERM U316, CHU Bretonneau, Tours, France; Women’s and Children’s Hospital and the University of Adelaide, Adelaide; and Genetics of Learning Disability (GOLD) Service, Hunter Genetics, University of Newcastle, New South Wales, Australia
| | - Hans-Hilger Ropers
- Max Planck Institute for Molecular Genetics, Berlin; Department of Medical Biology and Human Genetics, Innsbruck Medical University, Innsbruck; Institute for Human Genetics, University Heidelberg, Heidelberg; Institut Cochin de Génétique Moleculaire, CNRS/INSERM, CHU Cochin, Paris; Center for Human Genetics, Clinical Genetics Unit, Leuven, Belgium; Department of Human Genetics, University Medical Centre, Nijmegen, The Netherlands; Services de Génétique-INSERM U316, CHU Bretonneau, Tours, France; Women’s and Children’s Hospital and the University of Adelaide, Adelaide; and Genetics of Learning Disability (GOLD) Service, Hunter Genetics, University of Newcastle, New South Wales, Australia
| | - Steffen Lenzner
- Max Planck Institute for Molecular Genetics, Berlin; Department of Medical Biology and Human Genetics, Innsbruck Medical University, Innsbruck; Institute for Human Genetics, University Heidelberg, Heidelberg; Institut Cochin de Génétique Moleculaire, CNRS/INSERM, CHU Cochin, Paris; Center for Human Genetics, Clinical Genetics Unit, Leuven, Belgium; Department of Human Genetics, University Medical Centre, Nijmegen, The Netherlands; Services de Génétique-INSERM U316, CHU Bretonneau, Tours, France; Women’s and Children’s Hospital and the University of Adelaide, Adelaide; and Genetics of Learning Disability (GOLD) Service, Hunter Genetics, University of Newcastle, New South Wales, Australia
| |
Collapse
|
41
|
Abstract
Genetic factors have an important role in the aetiology of mental retardation. However, their contribution is often underestimated because in developed countries, severely affected patients are mainly sporadic cases and familial cases are rare. X-chromosomal mental retardation is the exception to this rule, and this is one of the reasons why research into the genetic and molecular causes of mental retardation has focused almost entirely on the X-chromosome. Here, we review the remarkable recent progress in this field, its promise for understanding neural function, learning and memory, and the implications of this research for health care.
Collapse
Affiliation(s)
- H-Hilger Ropers
- Max-Planck-Institute for Molecular Genetics, Ihnestrasse 73, D-14195 Berlin, Germany.
| | | |
Collapse
|
42
|
Renieri A, Pescucci C, Longo I, Ariani F, Mari F, Meloni I. Non-syndromic X-linked mental retardation: From a molecular to a clinical point of view. J Cell Physiol 2005; 204:8-20. [PMID: 15690397 DOI: 10.1002/jcp.20296] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
This review focuses on the 19 identified genes involved in X-linked "non-syndromic" mental retardation (MR) and defines the signaling pathways in which they are involved, focusing on emerging common mechanisms. The majority of proteins are involved in three distinct pathways: (1) Rho GTPases pathway modulating neuronal differentiation and synaptic plasticity; (2) Rab GTPases pathway regulating synaptic vesicle cycling; (3) gene expression regulation. The function of four proteins (ACSL4, AT2, SLC6A8, and SAP102) could not be reconciled to a common pathway. From a clinical point of view, the review discusses whether some common dysmorphic features can be identified even in non-syndromic MR patients and whether it is correct to maintain the distinction between "non-syndromic" and "syndromic" MR.
Collapse
Affiliation(s)
- A Renieri
- Medical Genetics, Department of Molecular Biology, University of Siena, Siena, Italy.
| | | | | | | | | | | |
Collapse
|
43
|
Silverman E, Frödin M, Gammeltoft S, Maller JL. Activation of p90 Rsk1 is sufficient for differentiation of PC12 cells. Mol Cell Biol 2004; 24:10573-83. [PMID: 15572664 PMCID: PMC533971 DOI: 10.1128/mcb.24.24.10573-10583.2004] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We investigated the role of Rsk proteins in the nerve growth factor (NGF) signaling pathway in PC12 cells. When rat Rsk1 or murine Rsk2 proteins were transiently expressed, NGF treatment (100 ng/ml for 3 days) caused three- and fivefold increases in Rsk1 and Rsk2 activities, respectively. Increased activation of both wild-type Rsk proteins could be achieved by coexpression of a constitutively active (CA) mitogen-activated protein kinase (MAPK) kinase, MEK1-DD, which is known to cause differentiation of PC12 cells even in the absence of NGF. Rsk1 and Rsk2 mutated in the PDK1-binding site were not activated by either NGF or MEK1-DD. Expression of constitutively active Rsk1 or Rsk2 in PC12 cells resulted in highly active proteins whose levels of activity did not change either with NGF treatment or after coexpression with MEK1-DD. Rsk2-CA expression had no detectable effect on the cells. However, expression of Rsk1-CA led to differentiation of PC12 cells even in the absence of NGF, as evidenced by neurite outgrowth. Differentiation was not observed with a nonactive Rsk1-CA that was mutated in the PDK1-binding site. Expression of Rsk1-CA did not lead to activation of the endogenous MAPK pathway, indicating that Rsk1 is sufficient to induce neurite outgrowth and is the only target of MAPK required for this effect. Collectively, our data demonstrate a key role for Rsk1 in the differentiation process of PC12 cells.
Collapse
Affiliation(s)
- Eran Silverman
- Howard Hughes Medical Institute and Department of Pharmacology, University of Colorado School of Medicine, 4200 E. 9th Ave., Campus Box C236, Denver, CO 80262, USA.
| | | | | | | |
Collapse
|
44
|
Abstract
Affecting 1-3% of the population, mental retardation (MR) poses significant challenges for clinicians and scientists. Understanding the biology of MR is complicated by the extraordinary heterogeneity of genetic MR disorders. Detailed analyses of >1000 Online Mendelian Inheritance in Man (OMIM) database entries and literature searches through September 2003 revealed 282 molecularly identified MR genes. We estimate that hundreds more MR genes remain to be identified. A novel test, in which we distributed unmapped MR disorders proportionately across the autosomes, failed to eliminate the well-known X-chromosome overrepresentation of MR genes and candidate genes. This evidence argues against ascertainment bias as the main cause of the skewed distribution. On the basis of a synthesis of clinical and laboratory data, we developed a biological functions classification scheme for MR genes. Metabolic pathways, signaling pathways, and transcription are the most common functions, but numerous other aspects of neuronal and glial biology are controlled by MR genes as well. Using protein sequence and domain-organization comparisons, we found a striking conservation of MR genes and genetic pathways across the approximately 700 million years that separate Homo sapiens and Drosophila melanogaster. Eighty-seven percent have one or more fruit fly homologs and 76% have at least one candidate functional ortholog. We propose that D. melanogaster can be used in a systematic manner to study MR and possibly to develop bioassays for therapeutic drug discovery. We selected 42 Drosophila orthologs as most likely to reveal molecular and cellular mechanisms of nervous system development or plasticity relevant to MR.
Collapse
Affiliation(s)
- Jennifer K Inlow
- Arizona Research Laboratories Division of Neurobiology, University of Arizona, Tucson 85721-0077, USA
| | | |
Collapse
|
45
|
Tarpey P, Parnau J, Blow M, Woffendin H, Bignell G, Cox C, Cox J, Davies H, Edkins S, Holden S, Korny A, Mallya U, Moon J, O’Meara S, Parker A, Stephens P, Stevens C, Teague J, Donnelly A, Mangelsdorf M, Mulley J, Partington M, Turner G, Stevenson R, Schwartz C, Young I, Easton D, Bobrow M, Futreal PA, Stratton MR, Gecz J, Wooster R, Raymond FL. Mutations in the DLG3 gene cause nonsyndromic X-linked mental retardation. Am J Hum Genet 2004; 75:318-24. [PMID: 15185169 PMCID: PMC1216066 DOI: 10.1086/422703] [Citation(s) in RCA: 127] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2004] [Accepted: 05/11/2004] [Indexed: 11/03/2022] Open
Abstract
We have identified truncating mutations in the human DLG3 (neuroendocrine dlg) gene in 4 of 329 families with moderate to severe X-linked mental retardation. DLG3 encodes synapse-associated protein 102 (SAP102), a member of the membrane-associated guanylate kinase protein family. Neuronal SAP102 is expressed during early brain development and is localized to the postsynaptic density of excitatory synapses. It is composed of three amino-terminal PDZ domains, an src homology domain, and a carboxyl-terminal guanylate kinase domain. The PDZ domains interact directly with the NR2 subunits of the NMDA glutamate receptor and with other proteins responsible for NMDA receptor localization, immobilization, and signaling. The mutations identified in this study all introduce premature stop codons within or before the third PDZ domain, and it is likely that this impairs the ability of SAP102 to interact with the NMDA receptor and/or other proteins involved in downstream NMDA receptor signaling pathways. NMDA receptors have been implicated in the induction of certain forms of synaptic plasticity, such as long-term potentiation and long-term depression, and these changes in synaptic efficacy have been proposed as neural mechanisms underlying memory and learning. The disruption of NMDA receptor targeting or signaling, as a result of the loss of SAP102, may lead to altered synaptic plasticity and may explain the intellectual impairment observed in individuals with DLG3 mutations.
Collapse
Affiliation(s)
- Patrick Tarpey
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom; Cambridge Institute of Medical Research, Department of Medical Genetics, University of Cambridge, and Cancer Research UK, Genetic Epidemiology Unit, Strangeways Research Laboratory, University of Cambridge, Cambridge, United Kingdom; Women’s and Children’s Hospital and University of Adelaide, Adelaide, Australia; Hunter Genetics and University of Newcastle, Waratah, Australia; Greenwood Genetics Center, Greenwood, SC; and Department of Medical Genetics, Leicester Royal Infirmary, Leicester, United Kingdom
| | - Josep Parnau
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom; Cambridge Institute of Medical Research, Department of Medical Genetics, University of Cambridge, and Cancer Research UK, Genetic Epidemiology Unit, Strangeways Research Laboratory, University of Cambridge, Cambridge, United Kingdom; Women’s and Children’s Hospital and University of Adelaide, Adelaide, Australia; Hunter Genetics and University of Newcastle, Waratah, Australia; Greenwood Genetics Center, Greenwood, SC; and Department of Medical Genetics, Leicester Royal Infirmary, Leicester, United Kingdom
| | - Matthew Blow
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom; Cambridge Institute of Medical Research, Department of Medical Genetics, University of Cambridge, and Cancer Research UK, Genetic Epidemiology Unit, Strangeways Research Laboratory, University of Cambridge, Cambridge, United Kingdom; Women’s and Children’s Hospital and University of Adelaide, Adelaide, Australia; Hunter Genetics and University of Newcastle, Waratah, Australia; Greenwood Genetics Center, Greenwood, SC; and Department of Medical Genetics, Leicester Royal Infirmary, Leicester, United Kingdom
| | - Hayley Woffendin
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom; Cambridge Institute of Medical Research, Department of Medical Genetics, University of Cambridge, and Cancer Research UK, Genetic Epidemiology Unit, Strangeways Research Laboratory, University of Cambridge, Cambridge, United Kingdom; Women’s and Children’s Hospital and University of Adelaide, Adelaide, Australia; Hunter Genetics and University of Newcastle, Waratah, Australia; Greenwood Genetics Center, Greenwood, SC; and Department of Medical Genetics, Leicester Royal Infirmary, Leicester, United Kingdom
| | - Graham Bignell
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom; Cambridge Institute of Medical Research, Department of Medical Genetics, University of Cambridge, and Cancer Research UK, Genetic Epidemiology Unit, Strangeways Research Laboratory, University of Cambridge, Cambridge, United Kingdom; Women’s and Children’s Hospital and University of Adelaide, Adelaide, Australia; Hunter Genetics and University of Newcastle, Waratah, Australia; Greenwood Genetics Center, Greenwood, SC; and Department of Medical Genetics, Leicester Royal Infirmary, Leicester, United Kingdom
| | - Charles Cox
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom; Cambridge Institute of Medical Research, Department of Medical Genetics, University of Cambridge, and Cancer Research UK, Genetic Epidemiology Unit, Strangeways Research Laboratory, University of Cambridge, Cambridge, United Kingdom; Women’s and Children’s Hospital and University of Adelaide, Adelaide, Australia; Hunter Genetics and University of Newcastle, Waratah, Australia; Greenwood Genetics Center, Greenwood, SC; and Department of Medical Genetics, Leicester Royal Infirmary, Leicester, United Kingdom
| | - James Cox
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom; Cambridge Institute of Medical Research, Department of Medical Genetics, University of Cambridge, and Cancer Research UK, Genetic Epidemiology Unit, Strangeways Research Laboratory, University of Cambridge, Cambridge, United Kingdom; Women’s and Children’s Hospital and University of Adelaide, Adelaide, Australia; Hunter Genetics and University of Newcastle, Waratah, Australia; Greenwood Genetics Center, Greenwood, SC; and Department of Medical Genetics, Leicester Royal Infirmary, Leicester, United Kingdom
| | - Helen Davies
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom; Cambridge Institute of Medical Research, Department of Medical Genetics, University of Cambridge, and Cancer Research UK, Genetic Epidemiology Unit, Strangeways Research Laboratory, University of Cambridge, Cambridge, United Kingdom; Women’s and Children’s Hospital and University of Adelaide, Adelaide, Australia; Hunter Genetics and University of Newcastle, Waratah, Australia; Greenwood Genetics Center, Greenwood, SC; and Department of Medical Genetics, Leicester Royal Infirmary, Leicester, United Kingdom
| | - Sarah Edkins
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom; Cambridge Institute of Medical Research, Department of Medical Genetics, University of Cambridge, and Cancer Research UK, Genetic Epidemiology Unit, Strangeways Research Laboratory, University of Cambridge, Cambridge, United Kingdom; Women’s and Children’s Hospital and University of Adelaide, Adelaide, Australia; Hunter Genetics and University of Newcastle, Waratah, Australia; Greenwood Genetics Center, Greenwood, SC; and Department of Medical Genetics, Leicester Royal Infirmary, Leicester, United Kingdom
| | - Simon Holden
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom; Cambridge Institute of Medical Research, Department of Medical Genetics, University of Cambridge, and Cancer Research UK, Genetic Epidemiology Unit, Strangeways Research Laboratory, University of Cambridge, Cambridge, United Kingdom; Women’s and Children’s Hospital and University of Adelaide, Adelaide, Australia; Hunter Genetics and University of Newcastle, Waratah, Australia; Greenwood Genetics Center, Greenwood, SC; and Department of Medical Genetics, Leicester Royal Infirmary, Leicester, United Kingdom
| | - Angelique Korny
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom; Cambridge Institute of Medical Research, Department of Medical Genetics, University of Cambridge, and Cancer Research UK, Genetic Epidemiology Unit, Strangeways Research Laboratory, University of Cambridge, Cambridge, United Kingdom; Women’s and Children’s Hospital and University of Adelaide, Adelaide, Australia; Hunter Genetics and University of Newcastle, Waratah, Australia; Greenwood Genetics Center, Greenwood, SC; and Department of Medical Genetics, Leicester Royal Infirmary, Leicester, United Kingdom
| | - Uma Mallya
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom; Cambridge Institute of Medical Research, Department of Medical Genetics, University of Cambridge, and Cancer Research UK, Genetic Epidemiology Unit, Strangeways Research Laboratory, University of Cambridge, Cambridge, United Kingdom; Women’s and Children’s Hospital and University of Adelaide, Adelaide, Australia; Hunter Genetics and University of Newcastle, Waratah, Australia; Greenwood Genetics Center, Greenwood, SC; and Department of Medical Genetics, Leicester Royal Infirmary, Leicester, United Kingdom
| | - Jenny Moon
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom; Cambridge Institute of Medical Research, Department of Medical Genetics, University of Cambridge, and Cancer Research UK, Genetic Epidemiology Unit, Strangeways Research Laboratory, University of Cambridge, Cambridge, United Kingdom; Women’s and Children’s Hospital and University of Adelaide, Adelaide, Australia; Hunter Genetics and University of Newcastle, Waratah, Australia; Greenwood Genetics Center, Greenwood, SC; and Department of Medical Genetics, Leicester Royal Infirmary, Leicester, United Kingdom
| | - Sarah O’Meara
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom; Cambridge Institute of Medical Research, Department of Medical Genetics, University of Cambridge, and Cancer Research UK, Genetic Epidemiology Unit, Strangeways Research Laboratory, University of Cambridge, Cambridge, United Kingdom; Women’s and Children’s Hospital and University of Adelaide, Adelaide, Australia; Hunter Genetics and University of Newcastle, Waratah, Australia; Greenwood Genetics Center, Greenwood, SC; and Department of Medical Genetics, Leicester Royal Infirmary, Leicester, United Kingdom
| | - Adrian Parker
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom; Cambridge Institute of Medical Research, Department of Medical Genetics, University of Cambridge, and Cancer Research UK, Genetic Epidemiology Unit, Strangeways Research Laboratory, University of Cambridge, Cambridge, United Kingdom; Women’s and Children’s Hospital and University of Adelaide, Adelaide, Australia; Hunter Genetics and University of Newcastle, Waratah, Australia; Greenwood Genetics Center, Greenwood, SC; and Department of Medical Genetics, Leicester Royal Infirmary, Leicester, United Kingdom
| | - Philip Stephens
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom; Cambridge Institute of Medical Research, Department of Medical Genetics, University of Cambridge, and Cancer Research UK, Genetic Epidemiology Unit, Strangeways Research Laboratory, University of Cambridge, Cambridge, United Kingdom; Women’s and Children’s Hospital and University of Adelaide, Adelaide, Australia; Hunter Genetics and University of Newcastle, Waratah, Australia; Greenwood Genetics Center, Greenwood, SC; and Department of Medical Genetics, Leicester Royal Infirmary, Leicester, United Kingdom
| | - Claire Stevens
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom; Cambridge Institute of Medical Research, Department of Medical Genetics, University of Cambridge, and Cancer Research UK, Genetic Epidemiology Unit, Strangeways Research Laboratory, University of Cambridge, Cambridge, United Kingdom; Women’s and Children’s Hospital and University of Adelaide, Adelaide, Australia; Hunter Genetics and University of Newcastle, Waratah, Australia; Greenwood Genetics Center, Greenwood, SC; and Department of Medical Genetics, Leicester Royal Infirmary, Leicester, United Kingdom
| | - Jon Teague
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom; Cambridge Institute of Medical Research, Department of Medical Genetics, University of Cambridge, and Cancer Research UK, Genetic Epidemiology Unit, Strangeways Research Laboratory, University of Cambridge, Cambridge, United Kingdom; Women’s and Children’s Hospital and University of Adelaide, Adelaide, Australia; Hunter Genetics and University of Newcastle, Waratah, Australia; Greenwood Genetics Center, Greenwood, SC; and Department of Medical Genetics, Leicester Royal Infirmary, Leicester, United Kingdom
| | - Andrew Donnelly
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom; Cambridge Institute of Medical Research, Department of Medical Genetics, University of Cambridge, and Cancer Research UK, Genetic Epidemiology Unit, Strangeways Research Laboratory, University of Cambridge, Cambridge, United Kingdom; Women’s and Children’s Hospital and University of Adelaide, Adelaide, Australia; Hunter Genetics and University of Newcastle, Waratah, Australia; Greenwood Genetics Center, Greenwood, SC; and Department of Medical Genetics, Leicester Royal Infirmary, Leicester, United Kingdom
| | - Marie Mangelsdorf
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom; Cambridge Institute of Medical Research, Department of Medical Genetics, University of Cambridge, and Cancer Research UK, Genetic Epidemiology Unit, Strangeways Research Laboratory, University of Cambridge, Cambridge, United Kingdom; Women’s and Children’s Hospital and University of Adelaide, Adelaide, Australia; Hunter Genetics and University of Newcastle, Waratah, Australia; Greenwood Genetics Center, Greenwood, SC; and Department of Medical Genetics, Leicester Royal Infirmary, Leicester, United Kingdom
| | - John Mulley
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom; Cambridge Institute of Medical Research, Department of Medical Genetics, University of Cambridge, and Cancer Research UK, Genetic Epidemiology Unit, Strangeways Research Laboratory, University of Cambridge, Cambridge, United Kingdom; Women’s and Children’s Hospital and University of Adelaide, Adelaide, Australia; Hunter Genetics and University of Newcastle, Waratah, Australia; Greenwood Genetics Center, Greenwood, SC; and Department of Medical Genetics, Leicester Royal Infirmary, Leicester, United Kingdom
| | - Michael Partington
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom; Cambridge Institute of Medical Research, Department of Medical Genetics, University of Cambridge, and Cancer Research UK, Genetic Epidemiology Unit, Strangeways Research Laboratory, University of Cambridge, Cambridge, United Kingdom; Women’s and Children’s Hospital and University of Adelaide, Adelaide, Australia; Hunter Genetics and University of Newcastle, Waratah, Australia; Greenwood Genetics Center, Greenwood, SC; and Department of Medical Genetics, Leicester Royal Infirmary, Leicester, United Kingdom
| | - Gillian Turner
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom; Cambridge Institute of Medical Research, Department of Medical Genetics, University of Cambridge, and Cancer Research UK, Genetic Epidemiology Unit, Strangeways Research Laboratory, University of Cambridge, Cambridge, United Kingdom; Women’s and Children’s Hospital and University of Adelaide, Adelaide, Australia; Hunter Genetics and University of Newcastle, Waratah, Australia; Greenwood Genetics Center, Greenwood, SC; and Department of Medical Genetics, Leicester Royal Infirmary, Leicester, United Kingdom
| | - Roger Stevenson
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom; Cambridge Institute of Medical Research, Department of Medical Genetics, University of Cambridge, and Cancer Research UK, Genetic Epidemiology Unit, Strangeways Research Laboratory, University of Cambridge, Cambridge, United Kingdom; Women’s and Children’s Hospital and University of Adelaide, Adelaide, Australia; Hunter Genetics and University of Newcastle, Waratah, Australia; Greenwood Genetics Center, Greenwood, SC; and Department of Medical Genetics, Leicester Royal Infirmary, Leicester, United Kingdom
| | - Charles Schwartz
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom; Cambridge Institute of Medical Research, Department of Medical Genetics, University of Cambridge, and Cancer Research UK, Genetic Epidemiology Unit, Strangeways Research Laboratory, University of Cambridge, Cambridge, United Kingdom; Women’s and Children’s Hospital and University of Adelaide, Adelaide, Australia; Hunter Genetics and University of Newcastle, Waratah, Australia; Greenwood Genetics Center, Greenwood, SC; and Department of Medical Genetics, Leicester Royal Infirmary, Leicester, United Kingdom
| | - Ian Young
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom; Cambridge Institute of Medical Research, Department of Medical Genetics, University of Cambridge, and Cancer Research UK, Genetic Epidemiology Unit, Strangeways Research Laboratory, University of Cambridge, Cambridge, United Kingdom; Women’s and Children’s Hospital and University of Adelaide, Adelaide, Australia; Hunter Genetics and University of Newcastle, Waratah, Australia; Greenwood Genetics Center, Greenwood, SC; and Department of Medical Genetics, Leicester Royal Infirmary, Leicester, United Kingdom
| | - Douglas Easton
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom; Cambridge Institute of Medical Research, Department of Medical Genetics, University of Cambridge, and Cancer Research UK, Genetic Epidemiology Unit, Strangeways Research Laboratory, University of Cambridge, Cambridge, United Kingdom; Women’s and Children’s Hospital and University of Adelaide, Adelaide, Australia; Hunter Genetics and University of Newcastle, Waratah, Australia; Greenwood Genetics Center, Greenwood, SC; and Department of Medical Genetics, Leicester Royal Infirmary, Leicester, United Kingdom
| | - Martin Bobrow
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom; Cambridge Institute of Medical Research, Department of Medical Genetics, University of Cambridge, and Cancer Research UK, Genetic Epidemiology Unit, Strangeways Research Laboratory, University of Cambridge, Cambridge, United Kingdom; Women’s and Children’s Hospital and University of Adelaide, Adelaide, Australia; Hunter Genetics and University of Newcastle, Waratah, Australia; Greenwood Genetics Center, Greenwood, SC; and Department of Medical Genetics, Leicester Royal Infirmary, Leicester, United Kingdom
| | - P. Andrew Futreal
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom; Cambridge Institute of Medical Research, Department of Medical Genetics, University of Cambridge, and Cancer Research UK, Genetic Epidemiology Unit, Strangeways Research Laboratory, University of Cambridge, Cambridge, United Kingdom; Women’s and Children’s Hospital and University of Adelaide, Adelaide, Australia; Hunter Genetics and University of Newcastle, Waratah, Australia; Greenwood Genetics Center, Greenwood, SC; and Department of Medical Genetics, Leicester Royal Infirmary, Leicester, United Kingdom
| | - Michael R. Stratton
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom; Cambridge Institute of Medical Research, Department of Medical Genetics, University of Cambridge, and Cancer Research UK, Genetic Epidemiology Unit, Strangeways Research Laboratory, University of Cambridge, Cambridge, United Kingdom; Women’s and Children’s Hospital and University of Adelaide, Adelaide, Australia; Hunter Genetics and University of Newcastle, Waratah, Australia; Greenwood Genetics Center, Greenwood, SC; and Department of Medical Genetics, Leicester Royal Infirmary, Leicester, United Kingdom
| | - Jozef Gecz
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom; Cambridge Institute of Medical Research, Department of Medical Genetics, University of Cambridge, and Cancer Research UK, Genetic Epidemiology Unit, Strangeways Research Laboratory, University of Cambridge, Cambridge, United Kingdom; Women’s and Children’s Hospital and University of Adelaide, Adelaide, Australia; Hunter Genetics and University of Newcastle, Waratah, Australia; Greenwood Genetics Center, Greenwood, SC; and Department of Medical Genetics, Leicester Royal Infirmary, Leicester, United Kingdom
| | - Richard Wooster
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom; Cambridge Institute of Medical Research, Department of Medical Genetics, University of Cambridge, and Cancer Research UK, Genetic Epidemiology Unit, Strangeways Research Laboratory, University of Cambridge, Cambridge, United Kingdom; Women’s and Children’s Hospital and University of Adelaide, Adelaide, Australia; Hunter Genetics and University of Newcastle, Waratah, Australia; Greenwood Genetics Center, Greenwood, SC; and Department of Medical Genetics, Leicester Royal Infirmary, Leicester, United Kingdom
| | - F. Lucy Raymond
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom; Cambridge Institute of Medical Research, Department of Medical Genetics, University of Cambridge, and Cancer Research UK, Genetic Epidemiology Unit, Strangeways Research Laboratory, University of Cambridge, Cambridge, United Kingdom; Women’s and Children’s Hospital and University of Adelaide, Adelaide, Australia; Hunter Genetics and University of Newcastle, Waratah, Australia; Greenwood Genetics Center, Greenwood, SC; and Department of Medical Genetics, Leicester Royal Infirmary, Leicester, United Kingdom
| |
Collapse
|
46
|
Martínez-Garay I, Ballesta MJ, Oltra S, Orellana C, Palomeque A, Moltó MD, Prieto F, Martínez F. Intronic L1 insertion and F268S, novel mutations in RPS6KA3 (RSK2) causing Coffin-Lowry syndrome. Clin Genet 2004; 64:491-6. [PMID: 14986828 DOI: 10.1046/j.1399-0004.2003.00166.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Two novel mutations of the ribosomal S6 kinase 2 gene (also known as RSK2) have been identified in two unrelated patients with Coffin-Lowry syndrome. The first mutation consists of a de novo insertion of a 5'-truncated LINE-1 element at position -8 of intron 3, which leads to a skipping of exon 4, leading to a shift of the reading frame and a premature stop codon. The L1 fragment (2800 bp) showed a rearrangement with a small deletion, a partial inversion of the ORF 2, flanked by short direct repeats which duplicate the acceptor splice site. However, cDNA analysis of the patient shows that both sites are apparently not functional. The second family showed the nucleotide change 803T>C in exon 10, resulting in the F268S mutation. This mutation was detected in two monozygotic twin patients and in their mother, who was mildly affected. The patients fulfill the clinical criteria of the syndrome, and therefore the mutation provides further support for the importance of phenylalanine at position 268, which is highly conserved in the protein kinase domain of many serine-threonine protein kinases.
Collapse
Affiliation(s)
- I Martínez-Garay
- Departamento de Genética, Facultad de Ciencias Biológicas, Universidad de Valencia, Valencia, Spain
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Laumonnier F, Bonnet-Brilhault F, Gomot M, Blanc R, David A, Moizard MP, Raynaud M, Ronce N, Lemonnier E, Calvas P, Laudier B, Chelly J, Fryns JP, Ropers HH, Hamel BCJ, Andres C, Barthélémy C, Moraine C, Briault S. X-linked mental retardation and autism are associated with a mutation in the NLGN4 gene, a member of the neuroligin family. Am J Hum Genet 2004; 74:552-7. [PMID: 14963808 PMCID: PMC1182268 DOI: 10.1086/382137] [Citation(s) in RCA: 535] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2003] [Accepted: 12/11/2003] [Indexed: 12/15/2022] Open
Abstract
A large French family including members affected by nonspecific X-linked mental retardation, with or without autism or pervasive developmental disorder in affected male patients, has been found to have a 2-base-pair deletion in the Neuroligin 4 gene (NLGN4) located at Xp22.33. This mutation leads to a premature stop codon in the middle of the sequence of the normal protein and is thought to suppress the transmembrane domain and sequences important for the dimerization of neuroligins that are required for proper cell-cell interaction through binding to beta-neurexins. As the neuroligins are mostly enriched at excitatory synapses, these results suggest that a defect in synaptogenesis may lead to deficits in cognitive development and communication processes. The fact that the deletion was present in both autistic and nonautistic mentally retarded males suggests that the NLGN4 gene is not only involved in autism, as previously described, but also in mental retardation, indicating that some types of autistic disorder and mental retardation may have common genetic origins.
Collapse
Affiliation(s)
- Frédéric Laumonnier
- INSERM U619—Génétique de l’Autisme et de la Déficience Mentale and INSERM U619—Service Explorations Fonctionnelles et Neurophysiologie en Pédopsychiatrie, CHU Bretonneau, Tours, France; Service de Génétique Médicale, Institut de Biologie, CHU Hôtel Dieu, Nantes, France; Centre Inter Régional d’Etude et de Ressource sur l’Autisme, Brest, France; Service de Génétique, CHU Hôpital Purpan, Toulouse, France; INSERM U129—ICGM, CHU Cochin, Paris; Center for Human Genetics, Leuven, Belgium; Max Planck Institute for Molecular Genetics, Berlin; and Department of Human Genetics, University Hospital, Nijmegen, The Netherlands
| | - Frédérique Bonnet-Brilhault
- INSERM U619—Génétique de l’Autisme et de la Déficience Mentale and INSERM U619—Service Explorations Fonctionnelles et Neurophysiologie en Pédopsychiatrie, CHU Bretonneau, Tours, France; Service de Génétique Médicale, Institut de Biologie, CHU Hôtel Dieu, Nantes, France; Centre Inter Régional d’Etude et de Ressource sur l’Autisme, Brest, France; Service de Génétique, CHU Hôpital Purpan, Toulouse, France; INSERM U129—ICGM, CHU Cochin, Paris; Center for Human Genetics, Leuven, Belgium; Max Planck Institute for Molecular Genetics, Berlin; and Department of Human Genetics, University Hospital, Nijmegen, The Netherlands
| | - Marie Gomot
- INSERM U619—Génétique de l’Autisme et de la Déficience Mentale and INSERM U619—Service Explorations Fonctionnelles et Neurophysiologie en Pédopsychiatrie, CHU Bretonneau, Tours, France; Service de Génétique Médicale, Institut de Biologie, CHU Hôtel Dieu, Nantes, France; Centre Inter Régional d’Etude et de Ressource sur l’Autisme, Brest, France; Service de Génétique, CHU Hôpital Purpan, Toulouse, France; INSERM U129—ICGM, CHU Cochin, Paris; Center for Human Genetics, Leuven, Belgium; Max Planck Institute for Molecular Genetics, Berlin; and Department of Human Genetics, University Hospital, Nijmegen, The Netherlands
| | - Romuald Blanc
- INSERM U619—Génétique de l’Autisme et de la Déficience Mentale and INSERM U619—Service Explorations Fonctionnelles et Neurophysiologie en Pédopsychiatrie, CHU Bretonneau, Tours, France; Service de Génétique Médicale, Institut de Biologie, CHU Hôtel Dieu, Nantes, France; Centre Inter Régional d’Etude et de Ressource sur l’Autisme, Brest, France; Service de Génétique, CHU Hôpital Purpan, Toulouse, France; INSERM U129—ICGM, CHU Cochin, Paris; Center for Human Genetics, Leuven, Belgium; Max Planck Institute for Molecular Genetics, Berlin; and Department of Human Genetics, University Hospital, Nijmegen, The Netherlands
| | - Albert David
- INSERM U619—Génétique de l’Autisme et de la Déficience Mentale and INSERM U619—Service Explorations Fonctionnelles et Neurophysiologie en Pédopsychiatrie, CHU Bretonneau, Tours, France; Service de Génétique Médicale, Institut de Biologie, CHU Hôtel Dieu, Nantes, France; Centre Inter Régional d’Etude et de Ressource sur l’Autisme, Brest, France; Service de Génétique, CHU Hôpital Purpan, Toulouse, France; INSERM U129—ICGM, CHU Cochin, Paris; Center for Human Genetics, Leuven, Belgium; Max Planck Institute for Molecular Genetics, Berlin; and Department of Human Genetics, University Hospital, Nijmegen, The Netherlands
| | - Marie-Pierre Moizard
- INSERM U619—Génétique de l’Autisme et de la Déficience Mentale and INSERM U619—Service Explorations Fonctionnelles et Neurophysiologie en Pédopsychiatrie, CHU Bretonneau, Tours, France; Service de Génétique Médicale, Institut de Biologie, CHU Hôtel Dieu, Nantes, France; Centre Inter Régional d’Etude et de Ressource sur l’Autisme, Brest, France; Service de Génétique, CHU Hôpital Purpan, Toulouse, France; INSERM U129—ICGM, CHU Cochin, Paris; Center for Human Genetics, Leuven, Belgium; Max Planck Institute for Molecular Genetics, Berlin; and Department of Human Genetics, University Hospital, Nijmegen, The Netherlands
| | - Martine Raynaud
- INSERM U619—Génétique de l’Autisme et de la Déficience Mentale and INSERM U619—Service Explorations Fonctionnelles et Neurophysiologie en Pédopsychiatrie, CHU Bretonneau, Tours, France; Service de Génétique Médicale, Institut de Biologie, CHU Hôtel Dieu, Nantes, France; Centre Inter Régional d’Etude et de Ressource sur l’Autisme, Brest, France; Service de Génétique, CHU Hôpital Purpan, Toulouse, France; INSERM U129—ICGM, CHU Cochin, Paris; Center for Human Genetics, Leuven, Belgium; Max Planck Institute for Molecular Genetics, Berlin; and Department of Human Genetics, University Hospital, Nijmegen, The Netherlands
| | - Nathalie Ronce
- INSERM U619—Génétique de l’Autisme et de la Déficience Mentale and INSERM U619—Service Explorations Fonctionnelles et Neurophysiologie en Pédopsychiatrie, CHU Bretonneau, Tours, France; Service de Génétique Médicale, Institut de Biologie, CHU Hôtel Dieu, Nantes, France; Centre Inter Régional d’Etude et de Ressource sur l’Autisme, Brest, France; Service de Génétique, CHU Hôpital Purpan, Toulouse, France; INSERM U129—ICGM, CHU Cochin, Paris; Center for Human Genetics, Leuven, Belgium; Max Planck Institute for Molecular Genetics, Berlin; and Department of Human Genetics, University Hospital, Nijmegen, The Netherlands
| | - Eric Lemonnier
- INSERM U619—Génétique de l’Autisme et de la Déficience Mentale and INSERM U619—Service Explorations Fonctionnelles et Neurophysiologie en Pédopsychiatrie, CHU Bretonneau, Tours, France; Service de Génétique Médicale, Institut de Biologie, CHU Hôtel Dieu, Nantes, France; Centre Inter Régional d’Etude et de Ressource sur l’Autisme, Brest, France; Service de Génétique, CHU Hôpital Purpan, Toulouse, France; INSERM U129—ICGM, CHU Cochin, Paris; Center for Human Genetics, Leuven, Belgium; Max Planck Institute for Molecular Genetics, Berlin; and Department of Human Genetics, University Hospital, Nijmegen, The Netherlands
| | - Patrick Calvas
- INSERM U619—Génétique de l’Autisme et de la Déficience Mentale and INSERM U619—Service Explorations Fonctionnelles et Neurophysiologie en Pédopsychiatrie, CHU Bretonneau, Tours, France; Service de Génétique Médicale, Institut de Biologie, CHU Hôtel Dieu, Nantes, France; Centre Inter Régional d’Etude et de Ressource sur l’Autisme, Brest, France; Service de Génétique, CHU Hôpital Purpan, Toulouse, France; INSERM U129—ICGM, CHU Cochin, Paris; Center for Human Genetics, Leuven, Belgium; Max Planck Institute for Molecular Genetics, Berlin; and Department of Human Genetics, University Hospital, Nijmegen, The Netherlands
| | - Béatrice Laudier
- INSERM U619—Génétique de l’Autisme et de la Déficience Mentale and INSERM U619—Service Explorations Fonctionnelles et Neurophysiologie en Pédopsychiatrie, CHU Bretonneau, Tours, France; Service de Génétique Médicale, Institut de Biologie, CHU Hôtel Dieu, Nantes, France; Centre Inter Régional d’Etude et de Ressource sur l’Autisme, Brest, France; Service de Génétique, CHU Hôpital Purpan, Toulouse, France; INSERM U129—ICGM, CHU Cochin, Paris; Center for Human Genetics, Leuven, Belgium; Max Planck Institute for Molecular Genetics, Berlin; and Department of Human Genetics, University Hospital, Nijmegen, The Netherlands
| | - Jamel Chelly
- INSERM U619—Génétique de l’Autisme et de la Déficience Mentale and INSERM U619—Service Explorations Fonctionnelles et Neurophysiologie en Pédopsychiatrie, CHU Bretonneau, Tours, France; Service de Génétique Médicale, Institut de Biologie, CHU Hôtel Dieu, Nantes, France; Centre Inter Régional d’Etude et de Ressource sur l’Autisme, Brest, France; Service de Génétique, CHU Hôpital Purpan, Toulouse, France; INSERM U129—ICGM, CHU Cochin, Paris; Center for Human Genetics, Leuven, Belgium; Max Planck Institute for Molecular Genetics, Berlin; and Department of Human Genetics, University Hospital, Nijmegen, The Netherlands
| | - Jean-Pierre Fryns
- INSERM U619—Génétique de l’Autisme et de la Déficience Mentale and INSERM U619—Service Explorations Fonctionnelles et Neurophysiologie en Pédopsychiatrie, CHU Bretonneau, Tours, France; Service de Génétique Médicale, Institut de Biologie, CHU Hôtel Dieu, Nantes, France; Centre Inter Régional d’Etude et de Ressource sur l’Autisme, Brest, France; Service de Génétique, CHU Hôpital Purpan, Toulouse, France; INSERM U129—ICGM, CHU Cochin, Paris; Center for Human Genetics, Leuven, Belgium; Max Planck Institute for Molecular Genetics, Berlin; and Department of Human Genetics, University Hospital, Nijmegen, The Netherlands
| | - Hans-Hilger Ropers
- INSERM U619—Génétique de l’Autisme et de la Déficience Mentale and INSERM U619—Service Explorations Fonctionnelles et Neurophysiologie en Pédopsychiatrie, CHU Bretonneau, Tours, France; Service de Génétique Médicale, Institut de Biologie, CHU Hôtel Dieu, Nantes, France; Centre Inter Régional d’Etude et de Ressource sur l’Autisme, Brest, France; Service de Génétique, CHU Hôpital Purpan, Toulouse, France; INSERM U129—ICGM, CHU Cochin, Paris; Center for Human Genetics, Leuven, Belgium; Max Planck Institute for Molecular Genetics, Berlin; and Department of Human Genetics, University Hospital, Nijmegen, The Netherlands
| | - Ben C. J. Hamel
- INSERM U619—Génétique de l’Autisme et de la Déficience Mentale and INSERM U619—Service Explorations Fonctionnelles et Neurophysiologie en Pédopsychiatrie, CHU Bretonneau, Tours, France; Service de Génétique Médicale, Institut de Biologie, CHU Hôtel Dieu, Nantes, France; Centre Inter Régional d’Etude et de Ressource sur l’Autisme, Brest, France; Service de Génétique, CHU Hôpital Purpan, Toulouse, France; INSERM U129—ICGM, CHU Cochin, Paris; Center for Human Genetics, Leuven, Belgium; Max Planck Institute for Molecular Genetics, Berlin; and Department of Human Genetics, University Hospital, Nijmegen, The Netherlands
| | - Christian Andres
- INSERM U619—Génétique de l’Autisme et de la Déficience Mentale and INSERM U619—Service Explorations Fonctionnelles et Neurophysiologie en Pédopsychiatrie, CHU Bretonneau, Tours, France; Service de Génétique Médicale, Institut de Biologie, CHU Hôtel Dieu, Nantes, France; Centre Inter Régional d’Etude et de Ressource sur l’Autisme, Brest, France; Service de Génétique, CHU Hôpital Purpan, Toulouse, France; INSERM U129—ICGM, CHU Cochin, Paris; Center for Human Genetics, Leuven, Belgium; Max Planck Institute for Molecular Genetics, Berlin; and Department of Human Genetics, University Hospital, Nijmegen, The Netherlands
| | - Catherine Barthélémy
- INSERM U619—Génétique de l’Autisme et de la Déficience Mentale and INSERM U619—Service Explorations Fonctionnelles et Neurophysiologie en Pédopsychiatrie, CHU Bretonneau, Tours, France; Service de Génétique Médicale, Institut de Biologie, CHU Hôtel Dieu, Nantes, France; Centre Inter Régional d’Etude et de Ressource sur l’Autisme, Brest, France; Service de Génétique, CHU Hôpital Purpan, Toulouse, France; INSERM U129—ICGM, CHU Cochin, Paris; Center for Human Genetics, Leuven, Belgium; Max Planck Institute for Molecular Genetics, Berlin; and Department of Human Genetics, University Hospital, Nijmegen, The Netherlands
| | - Claude Moraine
- INSERM U619—Génétique de l’Autisme et de la Déficience Mentale and INSERM U619—Service Explorations Fonctionnelles et Neurophysiologie en Pédopsychiatrie, CHU Bretonneau, Tours, France; Service de Génétique Médicale, Institut de Biologie, CHU Hôtel Dieu, Nantes, France; Centre Inter Régional d’Etude et de Ressource sur l’Autisme, Brest, France; Service de Génétique, CHU Hôpital Purpan, Toulouse, France; INSERM U129—ICGM, CHU Cochin, Paris; Center for Human Genetics, Leuven, Belgium; Max Planck Institute for Molecular Genetics, Berlin; and Department of Human Genetics, University Hospital, Nijmegen, The Netherlands
| | - Sylvain Briault
- INSERM U619—Génétique de l’Autisme et de la Déficience Mentale and INSERM U619—Service Explorations Fonctionnelles et Neurophysiologie en Pédopsychiatrie, CHU Bretonneau, Tours, France; Service de Génétique Médicale, Institut de Biologie, CHU Hôtel Dieu, Nantes, France; Centre Inter Régional d’Etude et de Ressource sur l’Autisme, Brest, France; Service de Génétique, CHU Hôpital Purpan, Toulouse, France; INSERM U129—ICGM, CHU Cochin, Paris; Center for Human Genetics, Leuven, Belgium; Max Planck Institute for Molecular Genetics, Berlin; and Department of Human Genetics, University Hospital, Nijmegen, The Netherlands
| |
Collapse
|
48
|
Abstract
Abstract
Affecting 1-3% of the population, mental retardation (MR) poses significant challenges for clinicians and scientists. Understanding the biology of MR is complicated by the extraordinary heterogeneity of genetic MR disorders. Detailed analyses of >1000 Online Mendelian Inheritance in Man (OMIM) database entries and literature searches through September 2003 revealed 282 molecularly identified MR genes. We estimate that hundreds more MR genes remain to be identified. A novel test, in which we distributed unmapped MR disorders proportionately across the autosomes, failed to eliminate the well-known X-chromosome overrepresentation of MR genes and candidate genes. This evidence argues against ascertainment bias as the main cause of the skewed distribution. On the basis of a synthesis of clinical and laboratory data, we developed a biological functions classification scheme for MR genes. Metabolic pathways, signaling pathways, and transcription are the most common functions, but numerous other aspects of neuronal and glial biology are controlled by MR genes as well. Using protein sequence and domain-organization comparisons, we found a striking conservation of MR genes and genetic pathways across the ∼700 million years that separate Homo sapiens and Drosophila melanogaster. Eighty-seven percent have one or more fruit fly homologs and 76% have at least one candidate functional ortholog. We propose that D. melanogaster can be used in a systematic manner to study MR and possibly to develop bioassays for therapeutic drug discovery. We selected 42 Drosophila orthologs as most likely to reveal molecular and cellular mechanisms of nervous system development or plasticity relevant to MR.
Collapse
Affiliation(s)
- Jennifer K Inlow
- Arizona Research Laboratories Division of Neurobiology, University of Arizona, Tucson, Arizona 85721-0077
| | - Linda L Restifo
- Arizona Research Laboratories Division of Neurobiology, University of Arizona, Tucson, Arizona 85721-0077
- Department of Neurology, University of Arizona, Tucson, Arizona 85721-0077
- Genetics Graduate Interdisciplinary Program, University of Arizona, Tucson, Arizona 85721-0077
| |
Collapse
|
49
|
Stevenson RE, Schwartz CE. Clinical and molecular contributions to the understanding of X-linked mental retardation. Cytogenet Genome Res 2004; 99:265-75. [PMID: 12900574 DOI: 10.1159/000071603] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2002] [Accepted: 02/06/2003] [Indexed: 11/19/2022] Open
Abstract
X-linked mental retardation (XLMR) was first recognized in the 1940s, long before any human genes had been mapped. It is now estimated that XLMR has a prevalence of 2.6 cases per 1,000 population, accounting for over 10% of all cases of mental retardation. It is likely that over 150 genes are associated with XLMR. Fragile X syndrome, the most common form of XLMR, has a prevalence of about 1 in 4,000 males. Clinically, XLMR exists in syndromic (mental retardation with other somatic, neurological, behavioral, or metabolic findings) and nonsyndromic (mental retardation without other distinguishing features) forms. However, recent findings have caused this distinction to become blurred as mutations in some genes have been found in both syndromic and nonsyndromic XLMR. Progress in XLMR gene identification has allowed some insight into various pathways and cellular activities involved in developing cognitive functions. The genes involve signaling pathways, transcription factors, cytoskeletal organization, cell adhesion and migration, and maintenance of the cell membrane potential.
Collapse
Affiliation(s)
- R E Stevenson
- J.C. Self Research Institute, Greenwood Genetic Center, Greenwood, SC 29646, USA.
| | | |
Collapse
|
50
|
Echeverria V, Ducatenzeiler A, Dowd E, Jänne J, Grant SM, Szyf M, Wandosell F, Avila J, Grimm H, Dunnett SB, Hartmann T, Alhonen L, Cuello AC. Altered mitogen-activated protein kinase signaling, tau hyperphosphorylation and mild spatial learning dysfunction in transgenic rats expressing the β-amyloid peptide intracellularly in hippocampal and cortical neurons. Neuroscience 2004; 129:583-92. [PMID: 15541880 DOI: 10.1016/j.neuroscience.2004.07.036] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/13/2004] [Indexed: 11/16/2022]
Abstract
The pathological significance of intracellular Abeta accumulation in vivo is not yet fully understood. To address this, we have studied transgenic rats expressing Alzheimer's-related transgenes that accumulate Abeta intraneuronally in the cerebral and hippocampal cortices but do not develop extracellular amyloid plaques. In these rats, the presence of intraneuronal Abeta is sufficient to provoke up-regulation of the phosphorylated form of extracellular-regulated kinase (ERK) 2 and its enzymatic activity in the hippocampus while no changes were observed in the activity or phosphorylation status of other putative tau kinases such as p38, glycogen synthase kinase 3, and cycline-dependent kinase 5. The increase in active phospho-ERK2 was accompanied by increased levels of tau phosphorylation at S396 and S404 ERK2 sites and a decrease in the phosphorylation of the CREB kinase p90RSK. In a water maze paradigm, male transgenic rats displayed a mild spatial learning deficit relative to control littermates. Our results suggest that in the absence of plaques, intraneuronal accumulation of Abeta peptide correlates with the initial steps in the tau-phosphorylation cascade, alterations in ERK2 signaling and impairment of higher CNS functions in male rats.
Collapse
Affiliation(s)
- V Echeverria
- Department of Pharmacology, McGill University, 3655 Promenade Sir-William-Osler, Montreal, Quebec, Canada H3G 1Y6
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|