1
|
Billes A, Pujalte M, Jedraszak G, Amsallem D, Boudry-Labis E, Boute O, Bouquillon S, Brischoux-Boucher E, Callier P, Coutton C, Denizet ALA, Dieterich K, Kuentz P, Lespinasse J, Mazel B, Morin G, Amram F, Pennamen P, Rio M, Piard J, Putoux A, Rama M, Roze-Guillaumey V, Schluth-Bolard C, Till M, Trouvé C, Vieville G, Rooryck C, Sanlaville D, Chatron N. Possible incomplete penetrance of Xq28 int22h-1/int22h-2 duplication. Clin Genet 2024; 106:234-246. [PMID: 38561231 DOI: 10.1111/cge.14525] [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: 12/03/2023] [Revised: 03/16/2024] [Accepted: 03/18/2024] [Indexed: 04/04/2024]
Abstract
Xq28 int22h-1/int22h-2 duplication is the result of non-allelic homologous recombination between int22h-1/int22h-2 repeats separated by 0.5 Mb. It is responsible for a syndromic form of intellectual disability (ID), with recurrent infections and atopic diseases. Minor defects, nonspecific facial dysmorphic features, and overweight have also been described. Half of female carriers have been reported with ID, whereas all reported evaluated born males present mild to moderate ID, suggesting complete penetrance. We collected data on 15 families from eight university hospitals. Among them, 40 patients, 21 females (one fetus), and 19 males (two fetuses), were carriers of typical or atypical Xq28 int22h-1/int22h-2 duplication. Twenty-one individuals were considered asymptomatic (16 females and 5 males), without significantly higher rate of recurrent infections, atopia, overweight, or facial dysmorphism. Approximately 67% live-born males and 23% live-born female carriers of the typical duplication did not have obvious signs of intellectual disability, suggesting previously undescribed incomplete penetrance or low expression in certain carriers. The possibility of a second-hit or modifying factors to this possible susceptibility locus is yet to be studied but a possible observational bias should be considered in assessing such challenging X-chromosome copy number gains. Additional segregation studies should help to quantify this newly described incomplete penetrance.
Collapse
Affiliation(s)
- Alexis Billes
- CHU Amiens Picardie, Service de Génétique Clinique, Amiens, France
- CHU Amiens Picardie, Laboratoire de Génétique Constitutionnelle, Amiens, France
| | - Mathilde Pujalte
- Service de Génétique, Groupement Hospitalier Est, Hospices Civils de Lyon, Bron, France
| | - Guillaume Jedraszak
- CHU Amiens Picardie, Laboratoire de Génétique Constitutionnelle, Amiens, France
- CHU Amiens Picardie, Département de génétique, UR4666 HEMATIM, CURS, Université Picardie Jules Verne, Amiens, France
| | - Daniel Amsallem
- Service de Neuropédiatrie, CHU de Besançon, Besançon, France
| | - Elise Boudry-Labis
- Institut de Génétique Médicale, Hôpital Jeanne de Flandre, Centre Hospitalier Universitaire de Lille, Lille, France
| | - Odile Boute
- Génétique Clinique, Centre Hospitalier Universitaire de Lille, Hôpital Jeanne de Flandre, Lille, France
| | - Sonia Bouquillon
- Institut de Génétique Médicale, Hôpital Jeanne de Flandre, Centre Hospitalier Universitaire de Lille, Lille, France
| | - Elise Brischoux-Boucher
- Centre de Génétique Humaine - CHU de Besançon, Université de Bourgogne-Franche-Comté, Besançon, France
| | - Patrick Callier
- Inserm UMR 1231 GAD Génétique des Anomalies du Développement, Université de Bourgogne, Dijon, France
- Unité Fonctionnelle Innovation diagnostique dans les maladies rares, laboratoire de génétique chromosomique et moléculaire, Plateau Technique de Biologie, CHU Dijon Bourgogne, Dijon, France
- Fédération Hospitalo-Universitaire Médecine Translationnelle et Anomalies du Développement (FHU TRANSLAD), CHU Dijon Bourgogne et Université de Bourgogne-Franche Comté, Dijon, France
| | - Charles Coutton
- Service de Génétique, Génomique, et Procréation, Centre Hospitalier Universitaire Grenoble Alpes, La Tronche, France
- INSERM 1209, CNRS UMR 5309, Institut pour l'Avancée des Biosciences (IAB), Université Grenoble Alpes, Grenoble, France
| | - Anne-Laude Avice Denizet
- Centre de Génétique Humaine - CHU de Besançon, Université de Bourgogne-Franche-Comté, Besançon, France
| | - Klaus Dieterich
- Université Grenoble Alpes, Inserm, U1216, CHU Grenoble Alpes, Medical Genetics, Grenoble Institute of Neurosciences, Grenoble, France
- CHU Grenoble, UM Génétique Chromosomique, Grenoble, France
| | - Paul Kuentz
- Oncobiologie Génétique Bioinformatique, PCBio, CHU de Besançon, Besançon, France
- UFR Des Sciences de Santé, INSERM-Université de Bourgogne UMR1231 GAD "Génétique des Anomalies du Développement", FHUTRANSLAD, Dijon, France
| | - James Lespinasse
- Centre Hospitalier de Chambéry, Service de Cytogénétique, Chambéry, France
| | - Benoît Mazel
- Centre de Génétique et Centre de Référence Anomalies du Développement et Syndromes Malformatifs, FHUTRANSLAD - CHU Dijon Bourgogne, Dijon, France
| | - Gilles Morin
- CHU Amiens Picardie, Service de Génétique Clinique, Amiens, France
| | - Florence Amram
- CHU Amiens Picardie, Service de Génétique Clinique, Amiens, France
| | - Perrine Pennamen
- CHU Bordeaux, Laboratoire de Génétique Biologique, Bordeaux, France
| | - Marlène Rio
- Université Paris Cité, Institut Imagine, Inserm U1163, Paris, France
- Service de Médecine Génomique des maladies rares, AP-HP, Centre Hôpital Necker-Enfants Malades, Paris, France
| | - Juliette Piard
- Centre de Génétique Humaine - CHU de Besançon, Université de Bourgogne-Franche-Comté, Besançon, France
- UMR 1231 GAD, Inserm, Université de Bourgogne Franche Comté, Dijon, France
| | - Audrey Putoux
- Hospices Civils de Lyon, Service de Génétique, Groupement Hospitalier Est, Bron, France
- Equipe GENDEV, Centre de Recherche en Neurosciences de Lyon, INSERM U1028 CNRS UMR5292, Université Claude Bernard Lyon 1, Lyon, France
| | - Mélanie Rama
- Institut de Génétique Médicale, Hôpital Jeanne de Flandre, Centre Hospitalier Universitaire de Lille, Lille, France
| | | | - Caroline Schluth-Bolard
- Service de Génétique, Groupement Hospitalier Est, Hospices Civils de Lyon, Bron, France
- Institute NeuroMyoGène, Laboratoire Physiopathologie et Génétique du Neurone et du Muscle, CNRS UMR 5261-INSERM U1315, Université de Lyon - Université Claude Bernard Lyon 1, Lyon, France
- Laboratoire de Diagnostic Génétique, Institut de Génétique Médicale d'Alsace, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Marianne Till
- Service de Génétique, Groupement Hospitalier Est, Hospices Civils de Lyon, Bron, France
| | - Chloé Trouvé
- Centre de Génétique Humaine - CHU de Besançon, Université de Bourgogne-Franche-Comté, Besançon, France
| | - Gaëlle Vieville
- Département de Génétique et Procréation, Hôpital Couple Enfant, CHU Grenoble-Alpes, Grenoble, France
| | - Caroline Rooryck
- CHU de Bordeaux, Service de Génétique Médicale, Bordeaux, France
- Univ. Bordeaux, Maladies Rares: Génétique et Métabolisme (MRGM), U 1211 INSERM, Bordeaux, France
| | - Damien Sanlaville
- Service de Génétique, Groupement Hospitalier Est, Hospices Civils de Lyon, Bron, France
- Institute NeuroMyoGène, Laboratoire Physiopathologie et Génétique du Neurone et du Muscle, CNRS UMR 5261-INSERM U1315, Université de Lyon - Université Claude Bernard Lyon 1, Lyon, France
| | - Nicolas Chatron
- Service de Génétique, Groupement Hospitalier Est, Hospices Civils de Lyon, Bron, France
- Institute NeuroMyoGène, Laboratoire Physiopathologie et Génétique du Neurone et du Muscle, CNRS UMR 5261-INSERM U1315, Université de Lyon - Université Claude Bernard Lyon 1, Lyon, France
| |
Collapse
|
2
|
Cabrera Zapata LE, Garcia-Segura LM, Cambiasso MJ, Arevalo MA. Genetics and Epigenetics of the X and Y Chromosomes in the Sexual Differentiation of the Brain. Int J Mol Sci 2022; 23:ijms232012288. [PMID: 36293143 PMCID: PMC9603441 DOI: 10.3390/ijms232012288] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/10/2022] [Accepted: 10/11/2022] [Indexed: 11/27/2022] Open
Abstract
For many decades to date, neuroendocrinologists have delved into the key contribution of gonadal hormones to the generation of sex differences in the developing brain and the expression of sex-specific physiological and behavioral phenotypes in adulthood. However, it was not until recent years that the role of sex chromosomes in the matter started to be seriously explored and unveiled beyond gonadal determination. Now we know that the divergent evolutionary process suffered by X and Y chromosomes has determined that they now encode mostly dissimilar genetic information and are subject to different epigenetic regulations, characteristics that together contribute to generate sex differences between XX and XY cells/individuals from the zygote throughout life. Here we will review and discuss relevant data showing how particular X- and Y-linked genes and epigenetic mechanisms controlling their expression and inheritance are involved, along with or independently of gonadal hormones, in the generation of sex differences in the brain.
Collapse
Affiliation(s)
- Lucas E. Cabrera Zapata
- Instituto de Investigación Médica Mercedes y Martín Ferreyra (INIMEC), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Córdoba, Córdoba 5016, Argentina
- Instituto Cajal (IC), Consejo Superior de Investigaciones Científicas (CSIC), 28002 Madrid, Spain
| | | | - María Julia Cambiasso
- Instituto de Investigación Médica Mercedes y Martín Ferreyra (INIMEC), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Córdoba, Córdoba 5016, Argentina
- Cátedra de Biología Celular, Facultad de Odontología, Universidad Nacional de Córdoba, Córdoba 5000, Argentina
- Correspondence: (M.J.C.); (M.A.A.)
| | - Maria Angeles Arevalo
- Instituto Cajal (IC), Consejo Superior de Investigaciones Científicas (CSIC), 28002 Madrid, Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Correspondence: (M.J.C.); (M.A.A.)
| |
Collapse
|
3
|
Cokyaman T, Silan F. Diagnostic Utility of Array Comparative Genomic Hybridization in Children with Neurological Diseases. Fetal Pediatr Pathol 2022; 41:68-76. [PMID: 32401632 DOI: 10.1080/15513815.2020.1764683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
INTRODUCTION We evaluated the contribution of array comparative genomic hybridization (aCGH) to the final diagnosis in children with neurocognitive disturbances or dysmorphic findings, but lacked a specific diagnosis. MATERIALS AND METHODS Medical files of pediatric patients with neurocognitive disturbances who underwent aCGH analysis were reviewed retrospectively. RESULTS Of 155 patients, 77 copy number variations were detected and 50% (39/77) were considered causative. The aCGH's final diagnostic rate was 25.1% (39/155). CONCLUSION With aCGH analysis, the diagnosis rate for patients with undiagnosed neurocognitive disturbances or dysmorphic syndrome may increase by 25-30%. If the phenotypic findings of the widely known neurocognitive disturbances cannot be identified during the initial clinical assessment, aCGH analysis may be beneficial.
Collapse
Affiliation(s)
- Turgay Cokyaman
- Pediatric Neurology, Faculty of Medicine, Çanakkale Onsekiz Mart University, Çanakkale, Turkey
| | - Fatma Silan
- Medical Genetics, Faculty of Medicine, Çanakkale Onsekiz Mart University, Canakkale, Turkey
| |
Collapse
|
4
|
Kessi M, Chen B, Peng J, Yan F, Yang L, Yin F. Calcium channelopathies and intellectual disability: a systematic review. Orphanet J Rare Dis 2021; 16:219. [PMID: 33985586 PMCID: PMC8120735 DOI: 10.1186/s13023-021-01850-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 05/04/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Calcium ions are involved in several human cellular processes including corticogenesis, transcription, and synaptogenesis. Nevertheless, the relationship between calcium channelopathies (CCs) and intellectual disability (ID)/global developmental delay (GDD) has been poorly investigated. We hypothesised that CCs play a major role in the development of ID/GDD and that both gain- and loss-of-function variants of calcium channel genes can induce ID/GDD. As a result, we performed a systematic review to investigate the contribution of CCs, potential mechanisms underlying their involvement in ID/GDD, advancements in cell and animal models, treatments, brain anomalies in patients with CCs, and the existing gaps in the knowledge. We performed a systematic search in PubMed, Embase, ClinVar, OMIM, ClinGen, Gene Reviews, DECIPHER and LOVD databases to search for articles/records published before March 2021. The following search strategies were employed: ID and calcium channel, mental retardation and calcium channel, GDD and calcium channel, developmental delay and calcium channel. MAIN BODY A total of 59 reports describing 159 cases were found in PubMed, Embase, ClinVar, and LOVD databases. Variations in ten calcium channel genes including CACNA1A, CACNA1C, CACNA1I, CACNA1H, CACNA1D, CACNA2D1, CACNA2D2, CACNA1E, CACNA1F, and CACNA1G were found to be associated with ID/GDD. Most variants exhibited gain-of-function effect. Severe to profound ID/GDD was observed more for the cases with gain-of-function variants as compared to those with loss-of-function. CACNA1E, CACNA1G, CACNA1F, CACNA2D2 and CACNA1A associated with more severe phenotype. Furthermore, 157 copy number variations (CNVs) spanning calcium genes were identified in DECIPHER database. The leading genes included CACNA1C, CACNA1A, and CACNA1E. Overall, the underlying mechanisms included gain- and/ or loss-of-function, alteration in kinetics (activation, inactivation) and dominant-negative effects of truncated forms of alpha1 subunits. Forty of the identified cases featured cerebellar atrophy. We identified only a few cell and animal studies that focused on the mechanisms of ID/GDD in relation to CCs. There is a scarcity of studies on treatment options for ID/GDD both in vivo and in vitro. CONCLUSION Our results suggest that CCs play a major role in ID/GDD. While both gain- and loss-of-function variants are associated with ID/GDD, the mechanisms underlying their involvement need further scrutiny.
Collapse
Affiliation(s)
- Miriam Kessi
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Hunan Intellectual and Developmental Disabilities Research Center, Changsha, Hunan, China
- Kilimanjaro Christian Medical University College, Moshi, Tanzania
- Mawenzi Regional Referral Hospital, Moshi, Tanzania
| | - Baiyu Chen
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Hunan Intellectual and Developmental Disabilities Research Center, Changsha, Hunan, China
| | - Jing Peng
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Hunan Intellectual and Developmental Disabilities Research Center, Changsha, Hunan, China
| | - Fangling Yan
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Hunan Intellectual and Developmental Disabilities Research Center, Changsha, Hunan, China
| | - Lifen Yang
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Hunan Intellectual and Developmental Disabilities Research Center, Changsha, Hunan, China
| | - Fei Yin
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- Hunan Intellectual and Developmental Disabilities Research Center, Changsha, Hunan, China.
| |
Collapse
|
5
|
D'Adamo P, Horvat A, Gurgone A, Mignogna ML, Bianchi V, Masetti M, Ripamonti M, Taverna S, Velebit J, Malnar M, Muhič M, Fink K, Bachi A, Restuccia U, Belloli S, Moresco RM, Mercalli A, Piemonti L, Potokar M, Bobnar ST, Kreft M, Chowdhury HH, Stenovec M, Vardjan N, Zorec R. Inhibiting glycolysis rescues memory impairment in an intellectual disability Gdi1-null mouse. Metabolism 2021; 116:154463. [PMID: 33309713 PMCID: PMC7871014 DOI: 10.1016/j.metabol.2020.154463] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 11/30/2020] [Accepted: 12/04/2020] [Indexed: 01/08/2023]
Abstract
OBJECTIVES GDI1 gene encodes for αGDI, a protein controlling the cycling of small GTPases, reputed to orchestrate vesicle trafficking. Mutations in human GDI1 are responsible for intellectual disability (ID). In mice with ablated Gdi1, a model of ID, impaired working and associative short-term memory was recorded. This cognitive phenotype worsens if the deletion of αGDI expression is restricted to neurons. However, whether astrocytes, key homeostasis providing neuroglial cells, supporting neurons via aerobic glycolysis, contribute to this cognitive impairment is unclear. METHODS We carried out proteomic analysis and monitored [18F]-fluoro-2-deoxy-d-glucose uptake into brain slices of Gdi1 knockout and wild type control mice. d-Glucose utilization at single astrocyte level was measured by the Förster Resonance Energy Transfer (FRET)-based measurements of cytosolic cyclic AMP, d-glucose and L-lactate, evoked by agonists selective for noradrenaline and L-lactate receptors. To test the role of astrocyte-resident processes in disease phenotype, we generated an inducible Gdi1 knockout mouse carrying the Gdi1 deletion only in adult astrocytes and conducted behavioural tests. RESULTS Proteomic analysis revealed significant changes in astrocyte-resident glycolytic enzymes. Imaging [18F]-fluoro-2-deoxy-d-glucose revealed an increased d-glucose uptake in Gdi1 knockout tissue versus wild type control mice, consistent with the facilitated d-glucose uptake determined by FRET measurements. In mice with Gdi1 deletion restricted to astrocytes, a selective and significant impairment in working memory was recorded, which was rescued by inhibiting glycolysis by 2-deoxy-d-glucose injection. CONCLUSIONS These results reveal a new astrocyte-based mechanism in neurodevelopmental disorders and open a novel therapeutic opportunity of targeting aerobic glycolysis, advocating a change in clinical practice.
Collapse
Affiliation(s)
- Patrizia D'Adamo
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy; University of Ljubljana, Faculty of Medicine, Institute of Pathophysiology, Laboratory of Neuroendocrinology - Molecular Cell Physiology, Ljubljana, Slovenia; Celica Biomedical, Laboratory for Cell Engineering, Ljubljana, Slovenia.
| | - Anemari Horvat
- University of Ljubljana, Faculty of Medicine, Institute of Pathophysiology, Laboratory of Neuroendocrinology - Molecular Cell Physiology, Ljubljana, Slovenia; Celica Biomedical, Laboratory for Cell Engineering, Ljubljana, Slovenia
| | - Antonia Gurgone
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | - Veronica Bianchi
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Michela Masetti
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Maddalena Ripamonti
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Stefano Taverna
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Jelena Velebit
- University of Ljubljana, Faculty of Medicine, Institute of Pathophysiology, Laboratory of Neuroendocrinology - Molecular Cell Physiology, Ljubljana, Slovenia; Celica Biomedical, Laboratory for Cell Engineering, Ljubljana, Slovenia
| | - Maja Malnar
- University of Ljubljana, Faculty of Medicine, Institute of Pathophysiology, Laboratory of Neuroendocrinology - Molecular Cell Physiology, Ljubljana, Slovenia
| | - Marko Muhič
- University of Ljubljana, Faculty of Medicine, Institute of Pathophysiology, Laboratory of Neuroendocrinology - Molecular Cell Physiology, Ljubljana, Slovenia
| | - Katja Fink
- University of Ljubljana, Faculty of Medicine, Institute of Pathophysiology, Laboratory of Neuroendocrinology - Molecular Cell Physiology, Ljubljana, Slovenia
| | - Angela Bachi
- IFOM, the FIRC Institute of Molecular Oncology, Milan, Italy
| | | | - Sara Belloli
- Institute of Bioimaging and Physiology, CNR, Segrate (MI), Italy; Experimental Imaging Center (EIC), San Raffaele Scientific Institute, Milan, Italy
| | - Rosa Maria Moresco
- Experimental Imaging Center (EIC), San Raffaele Scientific Institute, Milan, Italy; Medicine and Surgery Department, University of Milano-Bicocca, Monza (MB), Italy
| | - Alessia Mercalli
- Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milano, Italy
| | - Lorenzo Piemonti
- Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milano, Italy; Università Vita-Salute San Raffaele, Milano, Italy
| | - Maja Potokar
- University of Ljubljana, Faculty of Medicine, Institute of Pathophysiology, Laboratory of Neuroendocrinology - Molecular Cell Physiology, Ljubljana, Slovenia; Celica Biomedical, Laboratory for Cell Engineering, Ljubljana, Slovenia
| | - Saša Trkov Bobnar
- University of Ljubljana, Faculty of Medicine, Institute of Pathophysiology, Laboratory of Neuroendocrinology - Molecular Cell Physiology, Ljubljana, Slovenia; Celica Biomedical, Laboratory for Cell Engineering, Ljubljana, Slovenia
| | - Marko Kreft
- University of Ljubljana, Faculty of Medicine, Institute of Pathophysiology, Laboratory of Neuroendocrinology - Molecular Cell Physiology, Ljubljana, Slovenia; Celica Biomedical, Laboratory for Cell Engineering, Ljubljana, Slovenia; University of Ljubljana, Biotechnical Faculty, Department of Biology, Ljubljana, Slovenia
| | - Helena H Chowdhury
- University of Ljubljana, Faculty of Medicine, Institute of Pathophysiology, Laboratory of Neuroendocrinology - Molecular Cell Physiology, Ljubljana, Slovenia; Celica Biomedical, Laboratory for Cell Engineering, Ljubljana, Slovenia
| | - Matjaž Stenovec
- University of Ljubljana, Faculty of Medicine, Institute of Pathophysiology, Laboratory of Neuroendocrinology - Molecular Cell Physiology, Ljubljana, Slovenia; Celica Biomedical, Laboratory for Cell Engineering, Ljubljana, Slovenia
| | - Nina Vardjan
- University of Ljubljana, Faculty of Medicine, Institute of Pathophysiology, Laboratory of Neuroendocrinology - Molecular Cell Physiology, Ljubljana, Slovenia; Celica Biomedical, Laboratory for Cell Engineering, Ljubljana, Slovenia.
| | - Robert Zorec
- University of Ljubljana, Faculty of Medicine, Institute of Pathophysiology, Laboratory of Neuroendocrinology - Molecular Cell Physiology, Ljubljana, Slovenia; Celica Biomedical, Laboratory for Cell Engineering, Ljubljana, Slovenia.
| |
Collapse
|
6
|
Chiurazzi P, Kiani AK, Miertus J, Paolacci S, Barati S, Manara E, Stuppia L, Gurrieri F, Bertelli M. Genetic analysis of intellectual disability and autism. ACTA BIO-MEDICA : ATENEI PARMENSIS 2020; 91:e2020003. [PMID: 33170170 PMCID: PMC8023126 DOI: 10.23750/abm.v91i13-s.10684] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 10/23/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND AND AIM Intellectual disability (ID) and autism spectrum disorders (ASD) are neurodevelopmental conditions that often co-exist and affect children from birth, impacting on their cognition and adaptive behaviour. Social interaction and communication ability are also severely impaired in ASD. Almost 1-3% of the population is affected and it has been estimated that approximately 30% of intellectual disability and autism is caused by genetic factors. The aim of this review is to summarize monogenic conditions characterized by intellectual disability and/or autism for which the causative genes have been identified. METHODS AND RESULTS We identified monogenic ID/ASD conditions through PubMed and other NCBI databases. Many such genes are located on the X chromosome (>150 out of 900 X-linked protein-coding genes), but at least 2000 human genes are estimated to be involved in ID/ASD. We selected 174 genes (64 X-linked and 110 autosomal) for an NGS panel in order to screen patients with ID and/or ASD, after fragile X syndrome and significant Copy Number Variants have been excluded. CONCLUSIONS Accurate clinical and genetic diagnosis is required for precise treatment of these disorders, but due to their genetic heterogeneity, most cases remain undiagnosed. Next generation sequencing technologies have greatly enhanced the identification of new genes associated with intellectual disability and autism, ultimately leading to the development of better treatment options.
Collapse
Affiliation(s)
- Pietro Chiurazzi
- Istituto di Medicina Genomica, Università Cattolica del Sacro Cuore, Rome, Italy; Fondazione Policlinico Universitario "A. Gemelli" IRCCS, UOC Genetica Medica, Rome, Italy.
| | | | - Jan Miertus
- Génius n.o., Mestská Poliklinika, Trnava, Slovakia; MAGI´'S LAB, Rovereto (TN), Italy.
| | | | | | | | - Liborio Stuppia
- Dipartimento di Scienze Psicologiche della Salute e del Territorio, Università degli Studi "G. D'Annunzio", Chieti, Italy.
| | | | - Matteo Bertelli
- MAGI EUREGIO, Bolzano, Italy; MAGI'S LAB, Rovereto (TN), Italy; EBTNA-LAB, Rovereto (TN), Italy.
| |
Collapse
|
7
|
Krasnova NV, Chernova TA, Alekseeva IV, Gimalieva GG, Misyakova T, Sinitsyna LG. Clinical case of Bloch — Sulzberger syndrome. VESTNIK DERMATOLOGII I VENEROLOGII 2020. [DOI: 10.25208/vdv1117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Purpose.To present a clinical case of Bloch Sulzberger syndrome.
Material and methods. The examinations were performed to diagnose the disease: а visual examination of the skin, cytological analysis of the gallbladder fluid, general and biochemical blood tests, genetic research.
Results.During a visual examination of the skin, a differential diagnosis was made with infectious dermatitis, toxic-allergic dermatitis, epidermolysis bullosa and linear IgA-dependent dermatosis in children. Crucial in the diagnosis belonged to a genetic study, after which a deletion of exons 410 of the IKBKG gene was detected, which confirmed Bloch Sulzberger syndrome.
Conclusion.Newborns with vesicle-bullous rashes entering the neonatal pathology department and observed by neonatologists require a thorough examination, a mandatory consultation of a dermatologist inorder to determine further management tactics.
Collapse
|
8
|
Cisternas CD, Cabrera Zapata LE, Mir FR, Scerbo MJ, Arevalo MA, Garcia-Segura LM, Cambiasso MJ. Estradiol-dependent axogenesis and Ngn3 expression are determined by XY sex chromosome complement in hypothalamic neurons. Sci Rep 2020; 10:8223. [PMID: 32427857 PMCID: PMC7237695 DOI: 10.1038/s41598-020-65183-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 04/14/2020] [Indexed: 01/15/2023] Open
Abstract
Hypothalamic neurons show sex differences in neuritogenesis, female neurons have longer axons and higher levels of the neuritogenic factor neurogenin 3 (Ngn3) than male neurons in vitro. Moreover, the effect of 17-β-estradiol (E2) on axonal growth and Ngn3 expression is only found in male-derived neurons. To investigate whether sex chromosomes regulate these early sex differences in neuritogenesis by regulating the E2 effect on Ngn3, we evaluated the growth and differentiation of hypothalamic neurons derived from the “four core genotypes” mouse model, in which the factors of “gonadal sex” and “sex chromosome complement” are dissociated. We showed that sex differences in neurite outgrowth are determined by sex chromosome complement (XX > XY). Moreover, E2 increased the mRNA expression of Ngn3 and axonal length only in XY neurons. ERα/β expressions are regulated by sex chromosome complement; however, E2-effect on Ngn3 expression in XY neurons was only fully reproduced by PPT, a specific ligand of ERα, and prevented by MPP, a specific antagonist of ERα. Together our data indicate that sex chromosomes regulate early development of hypothalamic neurons by orchestrating not only sex differences in neuritogenesis, but also regulating the effect of E2 on Ngn3 expression through activation of ERα in hypothalamic neurons.
Collapse
Affiliation(s)
- Carla Daniela Cisternas
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina.,Departamento de Biología Bucal, Facultad de Odontología -Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Lucas Ezequiel Cabrera Zapata
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Franco Rafael Mir
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
| | - María Julia Scerbo
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
| | - María Angeles Arevalo
- Instituto Cajal, CSIC, Madrid, Spain.,Ciber de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Luis Miguel Garcia-Segura
- Instituto Cajal, CSIC, Madrid, Spain.,Ciber de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - María Julia Cambiasso
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina. .,Departamento de Biología Bucal, Facultad de Odontología -Universidad Nacional de Córdoba, Córdoba, Argentina.
| |
Collapse
|
9
|
Abstract
Mammalian sex chromosomes evolved from an ordinary pair of autosomes. The X chromosome is highly conserved, whereas the Y chromosome varies among species in size, structure, and gene content. Unlike autosomes that contain randomly mixed collections of genes, the sex chromosomes are enriched in testis-biased genes related to sexual development and reproduction, particularly in spermatogenesis and male fertility. This review focuses on how sex chromosome dosage compensation takes place and why meiotic sex chromosome inactivation occurs during spermatogenesis. Furthermore, the review also emphasizes how testis-biased genes are enriched on the sex chromosomes and their functions in male fertility. It is concluded that sex chromosomes are critical to sexual development and male fertility; however, our understanding of how sex chromosome genes direct sexual development and fertility has been hampered by the structural complexities of the sex chromosomes and by the multicopy nature of the testis gene families that also play a role in immunity, cancer development, and brain function.
Collapse
Affiliation(s)
- Wan-Sheng Liu
- Department of Animal Science, Center for Reproductive Biology and Health, College of Agricultural Sciences, The Pennsylvania State University, University Park, Pennsylvania 16802, USA
| |
Collapse
|
10
|
Zhu C, Cheng C, Wang Y, Muhammad W, Liu S, Zhu W, Shao B, Zhang Z, Yan X, He Q, Xu Z, Yu C, Qian X, Lu L, Zhang S, Zhang Y, Xiong W, Gao X, Xu Z, Chai R. Loss of ARHGEF6 Causes Hair Cell Stereocilia Deficits and Hearing Loss in Mice. Front Mol Neurosci 2018; 11:362. [PMID: 30333726 PMCID: PMC6176010 DOI: 10.3389/fnmol.2018.00362] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 09/13/2018] [Indexed: 11/13/2022] Open
Abstract
ARHGEF6 belongs to the family of guanine nucleotide exchange factors (GEFs) for Rho GTPases, and it specifically activates Rho GTPases CDC42 and RAC1. Arhgef6 is the X-linked intellectual disability gene also known as XLID46, and clinical features of patients carrying Arhgef6 mutations include intellectual disability and, in some cases, sensorineural hearing loss. Rho GTPases act as molecular switches in many cellular processes. Their activities are regulated by binding or hydrolysis of GTP, which is facilitated by GEFs and GTPase-activating proteins, respectively. RAC1 and CDC42 have been shown to play important roles in hair cell (HC) stereocilia development. However, the role of ARHGEF6 in inner ear development and hearing function has not yet been investigated. Here, we found that ARHGEF6 is expressed in mouse cochlear HCs, including the HC stereocilia. We established Arhgef6 knockdown mice using the clustered regularly interspaced short palindromic repeat-associated Cas9 nuclease (CRISPR-Cas9) genome editing technique. We showed that ARHGEF6 was indispensable for the maintenance of outer hair cell (OHC) stereocilia, and loss of ARHGEF6 in mice caused HC stereocilia deficits that eventually led to progressive HC loss and hearing loss. However, the loss of ARHGEF6 did not affect the synapse density and did not affect the mechanoelectrical transduction currents in OHCs at postnatal day 3. At the molecular level, the levels of active CDC42 and RAC1 were dramatically decreased in the Arhgef6 knockdown mice, suggesting that ARHGEF6 regulates stereocilia maintenance through RAC1/CDC42.
Collapse
Affiliation(s)
- Chengwen Zhu
- Department of Otolaryngology Head and Neck Surgery, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, China.,Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, China.,Research Institute of Otolaryngology, Nanjing, China
| | - Cheng Cheng
- Department of Otolaryngology Head and Neck Surgery, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, China.,Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, China.,Research Institute of Otolaryngology, Nanjing, China.,Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Yanfei Wang
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Jinan, China.,Shandong Provincial Collaborative Innovation Center of Cell Biology, Shandong Normal University, Jinan, China
| | - Waqas Muhammad
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, China.,Department of Biotechnology, Federal Urdu University of Arts, Science and Technology, Karachi, Pakistan
| | - Shuang Liu
- School of Life Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China
| | - Weijie Zhu
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, China
| | - Buwei Shao
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, China
| | - Zhong Zhang
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, China
| | - Xiaoqian Yan
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, China
| | - Qingqing He
- Department of Otolaryngology Head and Neck Surgery, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Zhengrong Xu
- Department of Otolaryngology Head and Neck Surgery, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Chenjie Yu
- Department of Otolaryngology Head and Neck Surgery, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Xiaoyun Qian
- Department of Otolaryngology Head and Neck Surgery, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Ling Lu
- Department of Otolaryngology Head and Neck Surgery, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Shasha Zhang
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, China.,Research Institute of Otolaryngology, Nanjing, China.,Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China.,Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, China
| | - Yuan Zhang
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, China
| | - Wei Xiong
- School of Life Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China
| | - Xia Gao
- Department of Otolaryngology Head and Neck Surgery, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, China.,Research Institute of Otolaryngology, Nanjing, China
| | - Zhigang Xu
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Jinan, China.,Shandong Provincial Collaborative Innovation Center of Cell Biology, Shandong Normal University, Jinan, China
| | - Renjie Chai
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast University, Nanjing, China.,Research Institute of Otolaryngology, Nanjing, China.,Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China.,Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
11
|
Neri G, Schwartz CE, Lubs HA, Stevenson RE. X-linked intellectual disability update 2017. Am J Med Genet A 2018; 176:1375-1388. [PMID: 29696803 DOI: 10.1002/ajmg.a.38710] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 02/23/2018] [Accepted: 03/23/2018] [Indexed: 12/28/2022]
Abstract
The X-chromosome comprises only about 5% of the human genome but accounts for about 15% of the genes currently known to be associated with intellectual disability. The early progress in identifying the X-linked intellectual disability (XLID)-associated genes through linkage analysis and candidate gene sequencing has been accelerated with the use of high-throughput technologies. In the 10 years since the last update, the number of genes associated with XLID has increased by 96% from 72 to 141 and duplications of all 141 XLID genes have been described, primarily through the application of high-resolution microarrays and next generation sequencing. The progress in identifying genetic and genomic alterations associated with XLID has not been matched with insights that improve the clinician's ability to form differential diagnoses, that bring into view the possibility of curative therapies for patients, or that inform scientists of the impact of the genetic alterations on cell organization and function.
Collapse
Affiliation(s)
- Giovanni Neri
- J.C. Self Research Institute of Human Genetics, Greenwood Genetic Center, Greenwood, South Carolina.,Istituto di Medicina Genomica, Università Cattolica del S. Cuore, Rome, Italy
| | - Charles E Schwartz
- J.C. Self Research Institute of Human Genetics, Greenwood Genetic Center, Greenwood, South Carolina
| | - Herbert A Lubs
- J.C. Self Research Institute of Human Genetics, Greenwood Genetic Center, Greenwood, South Carolina
| | - Roger E Stevenson
- J.C. Self Research Institute of Human Genetics, Greenwood Genetic Center, Greenwood, South Carolina
| |
Collapse
|
12
|
Vallianatos CN, Farrehi C, Friez MJ, Burmeister M, Keegan CE, Iwase S. Altered Gene-Regulatory Function of KDM5C by a Novel Mutation Associated With Autism and Intellectual Disability. Front Mol Neurosci 2018; 11:104. [PMID: 29670509 PMCID: PMC5893713 DOI: 10.3389/fnmol.2018.00104] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 03/15/2018] [Indexed: 01/03/2023] Open
Abstract
Intellectual disability (ID) affects up to 2% of the population world-wide and often coincides with other neurological conditions such as autism spectrum disorders. Mutations in KDM5C cause Mental Retardation, X-linked, Syndromic, Claes-Jensen type (MRXSCJ, OMIM #300534) and are one of the most common causes of X-linked ID. KDM5C encodes a histone demethylase for di- and tri-methylated histone H3 lysine 4 (H3K4me2/3), which are enriched in transcriptionally engaged promoter regions. KDM5C regulates gene transcription; however, it remains unknown whether removal of H3K4me is fully responsible for KDM5C-mediated gene regulation. Most mutations functionally tested to date result in reduced enzymatic activity of KDM5C, indicating loss of demethylase function as the primary mechanism underlying MRXSCJ. Here, we report a novel KDM5C mutation, R1115H, identified in an individual displaying MRXSCJ-like symptoms. The carrier mother's cells exhibited a highly skewed X-inactivation pattern. The KDM5C-R1115H substitution does not have an impact on enzymatic activity nor protein stability. However, when overexpressed in post-mitotic neurons, KDM5C-R1115H failed to fully suppress expression of target genes, while the mutant also affected expression of a distinct set of genes compared to KDM5C-wildtype. These results suggest that KDM5C may have non-enzymatic roles in gene regulation, and alteration of these roles contributes to MRXSCJ in this patient.
Collapse
Affiliation(s)
| | - Clara Farrehi
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, United States
| | - Michael J. Friez
- Diagnostic Laboratory, Greenwood Genetic Center, Greenwood, SC, United States
| | - Margit Burmeister
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, United States
- Molecular & Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI, United States
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, United States
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, United States
| | - Catherine E. Keegan
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, United States
- Division of Genetics, Department of Pediatrics, University of Michigan, Ann Arbor, MI, United States
| | - Shigeki Iwase
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
13
|
Unraveling unusual X-chromosome patterns during fragile-X syndrome genetic testing. Clin Chim Acta 2018; 476:167-172. [DOI: 10.1016/j.cca.2017.11.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 11/16/2017] [Accepted: 11/19/2017] [Indexed: 01/07/2023]
|
14
|
Ghosh S, Roy S, Pal P, Dutta A, Halder A. Cytogenetic analysis of patients with primary amenorrhea in Eastern India. J OBSTET GYNAECOL 2017; 38:270-275. [PMID: 29022424 DOI: 10.1080/01443615.2017.1353595] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Amenorrhea is the absence of menstruation in women of reproductive age. Previous reports suggest that chromosomal abnormality is the second most common cause of amenorrhoea. Early referral for cytogenetic evaluation is recommended for the identification of underlying chromosomal aberrations in amenorrhoea patients. This was an observational study which was conducted in Vivekananda Institute of Medical sciences, Kolkata, during January 2013-December 2015. This study aims to estimate the frequency and types of chromosomal abnormalities in primary amenorrhoea (PA) patients in Eastern India and correlate their hormonal profile with chromosomal reports. Clinical features of 150 patients were recorded with clinical expertise. Peripheral venous blood was taken following informed consent, followed by karyotyping for chromosomal analysis. Results revealed 76.1% of PA with normal female karyotype (46, XX) and 23.9% with different abnormal karyotypes. Among the abnormal karyotype constituents, 50% numerical abnormalities, most frequent being Turner syndrome, pure (n = 12, 8%) and mosaic (n = 5, 3.3%). Three cases (2%) showed male (XY) karyotype. The other cases showed X structural abnormalities. This study emphasises the need for cytogenetic analysis as integral part of the diagnostic protocol in case of PA for precise identification of chromosomal abnormalities and for appropriate management and counselling of these patients.
Collapse
Affiliation(s)
- Shanoli Ghosh
- a Department of Genetics , Vivekananda Institute of Medical Sciences , Kolkata , India
| | - Sanchita Roy
- b Department of Anatomy , Institute of Post-Graduation Medical Education and Research , Kolkata , India
| | - Pritha Pal
- a Department of Genetics , Vivekananda Institute of Medical Sciences , Kolkata , India
| | - Atreyee Dutta
- a Department of Genetics , Vivekananda Institute of Medical Sciences , Kolkata , India
| | - Ajanta Halder
- a Department of Genetics , Vivekananda Institute of Medical Sciences , Kolkata , India
| |
Collapse
|
15
|
Matsuo M, Awano H, Matsumoto M, Nagai M, Kawaguchi T, Zhang Z, Nishio H. Dystrophin Dp116: A yet to Be Investigated Product of the Duchenne Muscular Dystrophy Gene. Genes (Basel) 2017; 8:genes8100251. [PMID: 28974057 PMCID: PMC5664101 DOI: 10.3390/genes8100251] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 09/26/2017] [Indexed: 12/12/2022] Open
Abstract
The Duchenne muscular dystrophy (DMD) gene is one of the largest genes in the human genome. The gene exhibits a complex arrangement of seven alternative promoters, which drive the expression of three full length and four shorter isoforms. Dp116, the second smallest product of the DMD gene, is a Schwann cell-specific isoform encoded by a transcript corresponding to DMD exons 56–79, starting from a promoter/exon S1 within intron 55. The physiological roles of Dp116 are poorly understood, because of its extensive homology with other isoforms and its expression in specific tissues. This review summarizes studies on Dp116, focusing on clinical findings and alternative activation of the upstream translation initiation codon that is predicted to produce Dp118.
Collapse
Affiliation(s)
- Masafumi Matsuo
- Department of Physical Therapy, Faculty of Rehabilitation, Kobe Gakuin University, Kobe 651-2180, Japan.
| | - Hiroyuki Awano
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan.
| | - Masaaki Matsumoto
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan.
| | - Masashi Nagai
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan.
| | - Tatsuya Kawaguchi
- Biomedical Analysis and Pathology Research Group, Discovery Science and Technology Department, Daiichi Sankyo RD Novare Co., Tokyo 134-8630, Japan.
| | - Zhujun Zhang
- Department of Physical Therapy, Faculty of Rehabilitation, Kobe Gakuin University, Kobe 651-2180, Japan.
| | - Hisahide Nishio
- Department of Community Medicine and Social Healthcare Sciences, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan.
| |
Collapse
|
16
|
Leonenko G, Richards AL, Walters JT, Pocklington A, Chambert K, Al Eissa MM, Sharp SI, O'Brien NL, Curtis D, Bass NJ, McQuillin A, Hultman C, Moran JL, McCarroll SA, Sklar P, Neale BM, Holmans PA, Owen MJ, Sullivan PF, O'Donovan MC. Mutation intolerant genes and targets of FMRP are enriched for nonsynonymous alleles in schizophrenia. Am J Med Genet B Neuropsychiatr Genet 2017; 174:724-731. [PMID: 28719003 PMCID: PMC5669020 DOI: 10.1002/ajmg.b.32560] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 05/19/2017] [Indexed: 12/24/2022]
Abstract
Risk of schizophrenia is conferred by alleles occurring across the full spectrum of frequencies from common SNPs of weak effect through to ultra rare alleles, some of which may be moderately to highly penetrant. Previous studies have suggested that some of the risk of schizophrenia is attributable to uncommon alleles represented on Illumina exome arrays. Here, we present the largest study of exomic variation in schizophrenia to date, using samples from the United Kingdom and Sweden (10,011 schizophrenia cases and 13,791 controls). Single variants, genes, and gene sets were analyzed for association with schizophrenia. No single variant or gene reached genome-wide significance. Among candidate gene sets, we found significant enrichment for rare alleles (minor allele frequency [MAF] < 0.001) in genes intolerant of loss-of-function (LoF) variation and in genes whose messenger RNAs bind to fragile X mental retardation protein (FMRP). We further delineate the genetic architecture of schizophrenia by excluding a role for uncommon exomic variants (0.01 ≤ MAF ≥ 0.001) that confer a relatively large effect (odds ratio [OR] > 4). We also show risk alleles within this frequency range exist, but confer smaller effects and should be identified by larger studies.
Collapse
Affiliation(s)
- Ganna Leonenko
- Division of Psychological Medicine and Clinical NeurosciencesMRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University School of MedicineCardiffUK
| | - Alexander L. Richards
- Division of Psychological Medicine and Clinical NeurosciencesMRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University School of MedicineCardiffUK
| | - James T. Walters
- Division of Psychological Medicine and Clinical NeurosciencesMRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University School of MedicineCardiffUK
| | - Andrew Pocklington
- Division of Psychological Medicine and Clinical NeurosciencesMRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University School of MedicineCardiffUK
| | - Kimberly Chambert
- Stanley Center for Psychiatric ResearchBroad Institute of MIT and HarvardCambridgeMassachusetts
| | - Mariam M. Al Eissa
- Division of Psychiatry, Molecular Psychiatry LaboratoryUniversity College LondonLondonUK
| | - Sally I. Sharp
- Division of Psychiatry, Molecular Psychiatry LaboratoryUniversity College LondonLondonUK
| | - Niamh L. O'Brien
- Division of Psychiatry, Molecular Psychiatry LaboratoryUniversity College LondonLondonUK
| | | | - Nicholas J. Bass
- Division of Psychiatry, Molecular Psychiatry LaboratoryUniversity College LondonLondonUK
| | - Andrew McQuillin
- Division of Psychiatry, Molecular Psychiatry LaboratoryUniversity College LondonLondonUK
| | - Christina Hultman
- Department of Medical Epidemiology and BiostatisticsKarolinska InstituteStockholmSweden
| | - Jennifer L. Moran
- Stanley Center for Psychiatric ResearchBroad Institute of MIT and HarvardCambridgeMassachusetts
| | - Steven A. McCarroll
- Stanley Center for Psychiatric ResearchBroad Institute of MIT and HarvardCambridgeMassachusetts
- Program in Medical and Population GeneticsBroad Institute of MIT and HarvardCambridgeMassachusetts
- Department of GeneticsHarvard Medical SchoolBostonMassachusetts
| | - Pamela Sklar
- Icahn School of Medicine at Mount SinaiNew YorkNew York
| | - Benjamin M. Neale
- Stanley Center for Psychiatric ResearchBroad Institute of MIT and HarvardCambridgeMassachusetts
- Analytical and Translational Genetics UnitMassachusetts General HospitalBostonMassachusetts
| | - Peter A. Holmans
- Division of Psychological Medicine and Clinical NeurosciencesMRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University School of MedicineCardiffUK
| | - Michael J. Owen
- Division of Psychological Medicine and Clinical NeurosciencesMRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University School of MedicineCardiffUK
| | - Patrick F. Sullivan
- Department of Medical Epidemiology and BiostatisticsKarolinska InstituteStockholmSweden
- Departments of Genetics and PsychiatryUniversity of North CarolinaChapel HillNorth Carolina
| | - Michael C. O'Donovan
- Division of Psychological Medicine and Clinical NeurosciencesMRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University School of MedicineCardiffUK
| |
Collapse
|
17
|
Kaur Y, de Souza RJ, Gibson WT, Meyre D. A systematic review of genetic syndromes with obesity. Obes Rev 2017; 18:603-634. [PMID: 28346723 DOI: 10.1111/obr.12531] [Citation(s) in RCA: 113] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 02/01/2017] [Accepted: 02/02/2017] [Indexed: 11/29/2022]
Abstract
Syndromic monogenic obesity typically follows Mendelian patterns of inheritance and involves the co-presentation of other characteristics, such as mental retardation, dysmorphic features and organ-specific abnormalities. Previous reviews on obesity have reported 20 to 30 syndromes but no systematic review has yet been conducted on syndromic obesity. We searched seven databases using terms such as 'obesity', 'syndrome' and 'gene' to conduct a systematic review of literature on syndromic obesity. Our literature search identified 13,719 references. After abstract and full-text review, 119 relevant papers were eligible, and 42 papers were identified through additional searches. Our analysis of these 161 papers found that 79 obesity syndromes have been reported in literature. Of the 79 syndromes, 19 have been fully genetically elucidated, 11 have been partially elucidated, 27 have been mapped to a chromosomal region and for the remaining 22, neither the gene(s) nor the chromosomal location(s) have yet been identified. Interestingly, 54.4% of the syndromes have not been assigned a name, whereas 13.9% have more than one name. We report on organizational inconsistencies (e.g. naming discrepancies and syndrome classification) and provide suggestions for improvements. Overall, this review illustrates the need for increased clinical and genetic research on syndromes with obesity.
Collapse
Affiliation(s)
- Y Kaur
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Canada
| | - R J de Souza
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Canada
| | - W T Gibson
- Department of Medical Genetics, University of British Columbia, Vancouver, Canada.,British Columbia Children's Hospital Research Institute, Vancouver, Canada
| | - D Meyre
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Canada
| |
Collapse
|
18
|
Griesi-Oliveira K, Suzuki AM, Muotri AR. TRPC Channels and Mental Disorders. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 976:137-148. [PMID: 28508319 DOI: 10.1007/978-94-024-1088-4_12] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Transient receptor potential canonical (TRPC) channels mediate the influx of different types of cations through the cell membrane and are involved in many functions of the organism. Evidences of involvement of TRPC channels in neuronal development suggest that this family of proteins might play a role in certain neurological disorders. As reported, knockout mice for different TRPC channels show alterations in neuronal morphological and functional parameters, with behavioral abnormalities, such as in exploratory and social behaviors. Although mutations in TRPC channels could be related to mental/neurological disorders, there are only a few cases reported in literature, indicating that this correlation should be further explored. Nonetheless, other functional evidences support the implication of these channels in neurological diseases. In this chapter, we summarize the main findings relating TRPC channels to neurological disorders, such as autism spectrum disorders, bipolar disorder, and intellectual disability among others.
Collapse
Affiliation(s)
| | - Angela May Suzuki
- Department of Genetics and Evolutionary Biology, Bioscience Institute, University of São Paulo, São Paulo, SP, Brazil
| | - Alysson Renato Muotri
- Department of Pediatrics and Department of Cellular & Molecular Medicine, University of California San Diego School of Medicine, La Jolla, CA, USA. .,Rady Children's Hospital San Diego, San Diego, CA, USA. .,UCSD Stem Cell Program, Institute for Genomic Medicine, New York, NY, USA.
| |
Collapse
|
19
|
Abstract
Intellectual disability is the most common developmental disorder characterized by a congenital limitation in intellectual functioning and adaptive behavior. It often co-occurs with other mental conditions like attention deficit/hyperactivity disorder and autism spectrum disorder, and can be part of a malformation syndrome that affects other organs. Considering the heterogeneity of its causes (environmental and genetic), its frequency worldwide varies greatly. This review focuses on known genes underlying (syndromic and non-syndromic) intellectual disability, it provides a succinct analysis of their Gene Ontology, and it suggests the use of transcriptional profiling for the prioritization of candidate genes.
Collapse
Affiliation(s)
- Pietro Chiurazzi
- Institute of Genomic Medicine, Catholic University School of Medicine, Rome, Italy
| | - Filomena Pirozzi
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| |
Collapse
|
20
|
Oikonomakis V, Kosma K, Mitrakos A, Sofocleous C, Pervanidou P, Syrmou A, Pampanos A, Psoni S, Fryssira H, Kanavakis E, Kitsiou-Tzeli S, Tzetis M. Recurrent copy number variations as risk factors for autism spectrum disorders: analysis of the clinical implications. Clin Genet 2016; 89:708-18. [PMID: 26777411 DOI: 10.1111/cge.12740] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 01/08/2016] [Accepted: 01/13/2016] [Indexed: 12/31/2022]
Abstract
Chromosomal microarray analysis (CMA) is currently considered a first-tier diagnostic assay for the investigation of autism spectrum disorders (ASD), developmental delay and intellectual disability of unknown etiology. High-resolution arrays were utilized for the identification of copy number variations (CNVs) in 195 ASD patients of Greek origin (126 males, 69 females). CMA resulted in the detection of 65 CNVs, excluding the known polymorphic copy number polymorphisms also found in the Database of Genomic Variants, for 51/195 patients (26.1%). Parental DNA testing in 20/51 patients revealed that 17 CNVs were de novo, 6 paternal and 3 of maternal origin. The majority of the 65 CNVs were deletions (66.1%), of which 5 on the X-chromosome while the duplications, of which 7 on the X-chromosome, were rarer (22/65, 33.8%). Fifty-one CNVs from a total of 65, reported for our cohort of ASD patients, were of diagnostic significance and well described in the literature while 14 CNVs (8 losses, 6 gains) were characterized as variants of unknown significance and need further investigation. Among the 51 patients, 39 carried one CNV, 10 carried two CNVs and 2 carried three CNVs. The use of CMA, its clinical validity and utility was assessed.
Collapse
Affiliation(s)
- V Oikonomakis
- Department of Medical Genetics, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - K Kosma
- Department of Medical Genetics, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - A Mitrakos
- Department of Medical Genetics, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - C Sofocleous
- Department of Medical Genetics, Medical School, National and Kapodistrian University of Athens, Athens, Greece.,Research Institute for the Study of Genetic and Malignant Diseases in Childhood, "Aghia Sophia" Children's Hospital, Athens, Greece
| | - P Pervanidou
- 1st Department of Pediatrics, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - A Syrmou
- Department of Medical Genetics, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - A Pampanos
- Department of Medical Genetics, Medical School, National and Kapodistrian University of Athens, Athens, Greece.,Department of Genetics, "Alexandra" University Maternal Hospital, Athens, Greece
| | - S Psoni
- Department of Medical Genetics, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - H Fryssira
- Department of Medical Genetics, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - E Kanavakis
- Department of Medical Genetics, Medical School, National and Kapodistrian University of Athens, Athens, Greece.,Research Institute for the Study of Genetic and Malignant Diseases in Childhood, "Aghia Sophia" Children's Hospital, Athens, Greece
| | - S Kitsiou-Tzeli
- Department of Medical Genetics, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - M Tzetis
- Department of Medical Genetics, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
21
|
Liu F, Guo H, Ou M, Hou X, Sun G, Gong W, Jing H, Tan Q, Xue W, Dai Y, Sui W. ARHGAP4 mutated in a Chinese intellectually challenged family. Gene 2015; 578:205-9. [PMID: 26707211 DOI: 10.1016/j.gene.2015.12.035] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Revised: 12/05/2015] [Accepted: 12/11/2015] [Indexed: 10/22/2022]
Abstract
OBJECTIVE Mental retardation is characterized by lower intelligence compared to the average intelligence of persons the same age. These patients have low adaptive capacity acquired by society. The genetic factors of causing MR include monogenic disease, chromosome structural aberration, and chromosome number aberration and so on. We explored the cause of a Chinese family suffering from mental retardation. METHODS We used karyotyping technology to determine the karyotype of the proband, and we used FISH to verify the result of the karyotyping. We used whole-exome sequencing to identify the disease-causing gene and used Sanger sequencing to verify the result of whole-exome sequencing to assess the family's gene expression. RESULTS The G-banding of the karyotype revealed that the patient's karyotype is 46, XY. FISH revealed that the patient does not have a trisomy syndrome. The karyotype of the proband is normal. Using whole-exome sequencing, we identified 108,767 variants in the exome gene of the patient, including 101,787 SNPs and 6980 InDels. Combining clinical information and bioinformatics analysis, including databases filtering and SIFT analysis, we found ARHGAP4 in X chromosome was candidate MR disease-causing gene. PCR and Sanger sequencing results were consistent with whole-exome sequencing. ARHGAP4 (T491M) mutation was present in the genome of the proband and his mother is a carrier, while his father, sister, and brother do not carry this mutation. CONCLUSION According to clinical information, whole-exome sequencing results and Sanger verification results, ARHGAP4 (T491M) mutation may be disease-causing gene of the MR patient. The relation between ARHGAP4 mutation and MR clinical characteristic is needed to be illuminated with participation of more MR patients.
Collapse
Affiliation(s)
- Fuhua Liu
- Nephrology Department of Guilin, 181 St Hospital, Guangxi Key Laboratory of Metabolic Diseases Research, 541002 Guilin, Guangxi, China; College of Life Science, Guangxi Normal University, 541004 Guilin, Guangxi, China
| | - Hui Guo
- Clinical Medical Research Center, the Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, 518020, Shenzhen, Guangdong, China
| | - Minglin Ou
- Nephrology Department of Guilin, 181 St Hospital, Guangxi Key Laboratory of Metabolic Diseases Research, 541002 Guilin, Guangxi, China
| | - Xianliang Hou
- Nephrology Department of Guilin, 181 St Hospital, Guangxi Key Laboratory of Metabolic Diseases Research, 541002 Guilin, Guangxi, China
| | - Guoping Sun
- Lab. Center, Shenzhen Pingshan People's Hospital, Shenzhen, Guangdong 518118, China
| | - Weiwei Gong
- Nephrology Department of Guilin, 181 St Hospital, Guangxi Key Laboratory of Metabolic Diseases Research, 541002 Guilin, Guangxi, China
| | - Huanyun Jing
- Nephrology Department of Guilin, 181 St Hospital, Guangxi Key Laboratory of Metabolic Diseases Research, 541002 Guilin, Guangxi, China
| | - Qiupei Tan
- Nephrology Department of Guilin, 181 St Hospital, Guangxi Key Laboratory of Metabolic Diseases Research, 541002 Guilin, Guangxi, China
| | - Wen Xue
- Nephrology Department of Guilin, 181 St Hospital, Guangxi Key Laboratory of Metabolic Diseases Research, 541002 Guilin, Guangxi, China
| | - Yong Dai
- Clinical Medical Research Center, the Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, 518020, Shenzhen, Guangdong, China.
| | - Weiguo Sui
- Nephrology Department of Guilin, 181 St Hospital, Guangxi Key Laboratory of Metabolic Diseases Research, 541002 Guilin, Guangxi, China.
| |
Collapse
|
22
|
Jin Z, Yu L, Geng J, Wang J, Jin X, Huang H. A novel 47.2Mb duplication on chromosomal bands Xq21.1–25 associated with mental retardation. Gene 2015; 567:98-102. [DOI: 10.1016/j.gene.2015.04.083] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 04/20/2015] [Accepted: 04/22/2015] [Indexed: 11/24/2022]
|
23
|
Craig IW, Haworth CMA, Plomin R. Commentary on "A Role for the X Chromosome in Sex Differences in Variability in General Intelligence?" (Johnson et al., 2009). PERSPECTIVES ON PSYCHOLOGICAL SCIENCE 2015; 4:615-21. [PMID: 26161737 DOI: 10.1111/j.1745-6924.2009.01170.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Johnson et al.'s (2009) article highlights the role of X-chromosomal genes in general intelligence and draws attention to their potential role in explaining the observed greater variance for this trait in males and their excess at both extremes of the distribution. We note that this would result from a simple additive effect of X-linked intelligence genes and also discuss the potentially important contribution of recessive deleterious loci. The buffering effect of heterozygosity in females will be partly constrained by the skewing of X-inactivation patterns increasing the variance of females beyond what is expected. Furthermore, escape of some X-linked genes from in-activation may also be relevant to male-female variance comparisons. We also comment on the difficulty of establishing the extent to which the X chromosome is enriched for intelligence genes and point out that their estimates of the proportion of genes influencing general intelligence that might be located on the X chromosome rely on some doubtful premises, especially concerning the likely equivalence of X-linked gene action in males and females. Finally, we discuss the increasingly compelling evidence for the accumulation of genes on the X chromosome that have selective benefit to males, including those implicated infertility and some manifestations of intelligence.
Collapse
Affiliation(s)
- Ian W Craig
- Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, London, United Kingdom
| | - Claire M A Haworth
- Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, London, United Kingdom
| | - Robert Plomin
- Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, London, United Kingdom
| |
Collapse
|
24
|
Farrell MS, Werge T, Sklar P, Owen MJ, Ophoff RA, O'Donovan MC, Corvin A, Cichon S, Sullivan PF. Evaluating historical candidate genes for schizophrenia. Mol Psychiatry 2015; 20:555-62. [PMID: 25754081 PMCID: PMC4414705 DOI: 10.1038/mp.2015.16] [Citation(s) in RCA: 212] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Revised: 11/14/2014] [Accepted: 01/05/2015] [Indexed: 12/19/2022]
Abstract
Prior to the genome-wide association era, candidate gene studies were a major approach in schizophrenia genetics. In this invited review, we consider the current status of 25 historical candidate genes for schizophrenia (for example, COMT, DISC1, DTNBP1 and NRG1). The initial study for 24 of these genes explicitly evaluated common variant hypotheses about schizophrenia. Our evaluation included a meta-analysis of the candidate gene literature, incorporation of the results of the largest genomic study yet published for schizophrenia, ratings from informed researchers who have published on these genes, and ratings from 24 schizophrenia geneticists. On the basis of current empirical evidence and mostly consensual assessments of informed opinion, it appears that the historical candidate gene literature did not yield clear insights into the genetic basis of schizophrenia. A likely reason why historical candidate gene studies did not achieve their primary aims is inadequate statistical power. However, the considerable efforts embodied in these early studies unquestionably set the stage for current successes in genomic approaches to schizophrenia.
Collapse
Affiliation(s)
- M S Farrell
- Center for Psychiatric Genomics, Department of Genetics, Genomic Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - T Werge
- 1] Institute of Biological Psychiatry, MHC Sct. Hans, Mental Health Services Copenhagen, Denmark [2] Department of Clinical Medicine, University of Copenhagen, Copenhagen, Aarhus, Denmark [3] The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
| | - P Sklar
- 1] Division of Psychiatric Genomics, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA [2] Institute for Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA [3] Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - M J Owen
- 1] MRC Centre for Neuropsychiatric Genetics and Genomics, Institute of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK [2] National Centre for Mental Health, Cardiff University, Cardiff, UK
| | - R A Ophoff
- 1] Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA [2] Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA, USA [3] Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, The Netherlands
| | - M C O'Donovan
- 1] MRC Centre for Neuropsychiatric Genetics and Genomics, Institute of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK [2] National Centre for Mental Health, Cardiff University, Cardiff, UK
| | - A Corvin
- Neuropsychiatric Genetics Research Group, Department of Psychiatry, Trinity College Dublin, Ireland
| | - S Cichon
- 1] Division of Medical Genetics, Department of Biomedicine, University Basel, Basel, Switzerland [2] Institute of Human Genetics, University of Bonn, Bonn, Germany [3] Department of Genomics, Life and Brain Center, Bonn, Germany
| | - P F Sullivan
- 1] Center for Psychiatric Genomics, Department of Genetics, Genomic Medicine, University of North Carolina, Chapel Hill, NC, USA [2] Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden [3] Department of Psychiatry, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
25
|
Moeschler JB, Shevell M. Comprehensive evaluation of the child with intellectual disability or global developmental delays. Pediatrics 2014; 134:e903-18. [PMID: 25157020 PMCID: PMC9923626 DOI: 10.1542/peds.2014-1839] [Citation(s) in RCA: 343] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Global developmental delay and intellectual disability are relatively common pediatric conditions. This report describes the recommended clinical genetics diagnostic approach. The report is based on a review of published reports, most consisting of medium to large case series of diagnostic tests used, and the proportion of those that led to a diagnosis in such patients. Chromosome microarray is designated as a first-line test and replaces the standard karyotype and fluorescent in situ hybridization subtelomere tests for the child with intellectual disability of unknown etiology. Fragile X testing remains an important first-line test. The importance of considering testing for inborn errors of metabolism in this population is supported by a recent systematic review of the literature and several case series recently published. The role of brain MRI remains important in certain patients. There is also a discussion of the emerging literature on the use of whole-exome sequencing as a diagnostic test in this population. Finally, the importance of intentional comanagement among families, the medical home, and the clinical genetics specialty clinic is discussed.
Collapse
|
26
|
Szatkiewicz JP, O'Dushlaine C, Chen G, Chambert K, Moran JL, Neale BM, Fromer M, Ruderfer D, Akterin S, Bergen SE, Kähler A, Magnusson PKE, Kim Y, Crowley JJ, Rees E, Kirov G, O'Donovan MC, Owen MJ, Walters J, Scolnick E, Sklar P, Purcell S, Hultman CM, McCarroll SA, Sullivan PF. Copy number variation in schizophrenia in Sweden. Mol Psychiatry 2014; 19:762-73. [PMID: 24776740 PMCID: PMC4271733 DOI: 10.1038/mp.2014.40] [Citation(s) in RCA: 207] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Revised: 02/25/2014] [Accepted: 03/20/2014] [Indexed: 12/13/2022]
Abstract
Schizophrenia (SCZ) is a highly heritable neuropsychiatric disorder of complex genetic etiology. Previous genome-wide surveys have revealed a greater burden of large, rare copy number variations (CNVs) in SCZ cases and identified multiple rare recurrent CNVs that increase risk of SCZ although with incomplete penetrance and pleiotropic effects. Identification of additional recurrent CNVs and biological pathways enriched for SCZ CNVs requires greater sample sizes. We conducted a genome-wide survey for CNVs associated with SCZ using a Swedish national sample (4719 cases and 5917 controls). High-confidence CNV calls were generated using genotyping array intensity data, and their effect on risk of SCZ was measured. Our data confirm increased burden of large, rare CNVs in SCZ cases as well as significant associations for recurrent 16p11.2 duplications, 22q11.2 deletions and 3q29 deletions. We report a novel association for 17q12 duplications (odds ratio=4.16, P=0.018), previously associated with autism and mental retardation but not SCZ. Intriguingly, gene set association analyses implicate biological pathways previously associated with SCZ through common variation and exome sequencing (calcium channel signaling and binding partners of the fragile X mental retardation protein). We found significantly increased burden of the largest CNVs (>500 kb) in genes present in the postsynaptic density, in genomic regions implicated via SCZ genome-wide association studies and in gene products localized to mitochondria and cytoplasm. Our findings suggest that multiple lines of genomic inquiry--genome-wide screens for CNVs, common variation and exonic variation--are converging on similar sets of pathways and/or genes.
Collapse
Affiliation(s)
- J P Szatkiewicz
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - C O'Dushlaine
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - G Chen
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - K Chambert
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - J L Moran
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - B M Neale
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - M Fromer
- Department of Psychiatry, Mount Sinai School of Medicine, New York, NY, USA
| | - D Ruderfer
- Department of Psychiatry, Mount Sinai School of Medicine, New York, NY, USA
| | - S Akterin
- Department of Medical Epidemiology, Karolinska Institutet, Stockholm, Sweden
| | - S E Bergen
- 1] Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA [2] Department of Medical Epidemiology, Karolinska Institutet, Stockholm, Sweden
| | - A Kähler
- Department of Medical Epidemiology, Karolinska Institutet, Stockholm, Sweden
| | - P K E Magnusson
- Department of Medical Epidemiology, Karolinska Institutet, Stockholm, Sweden
| | - Y Kim
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - J J Crowley
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - E Rees
- MRC Centre for Neuropsychiatric Genetics and Genomics, Institute of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | - G Kirov
- MRC Centre for Neuropsychiatric Genetics and Genomics, Institute of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | - M C O'Donovan
- MRC Centre for Neuropsychiatric Genetics and Genomics, Institute of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | - M J Owen
- MRC Centre for Neuropsychiatric Genetics and Genomics, Institute of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | - J Walters
- MRC Centre for Neuropsychiatric Genetics and Genomics, Institute of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | - E Scolnick
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - P Sklar
- Department of Psychiatry, Mount Sinai School of Medicine, New York, NY, USA
| | - S Purcell
- 1] Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA [2] Department of Psychiatry, Mount Sinai School of Medicine, New York, NY, USA
| | - C M Hultman
- Department of Medical Epidemiology, Karolinska Institutet, Stockholm, Sweden
| | - S A McCarroll
- 1] Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA [2] Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - P F Sullivan
- 1] Department of Genetics, University of North Carolina, Chapel Hill, NC, USA [2] Department of Medical Epidemiology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
27
|
Mignon-Ravix C, Cacciagli P, Choucair N, Popovici C, Missirian C, Milh M, Mégarbané A, Busa T, Julia S, Girard N, Badens C, Sigaudy S, Philip N, Villard L. Intragenic rearrangements in X-linked intellectual deficiency: results of a-CGH in a series of 54 patients and identification of TRPC5 and KLHL15 as potential XLID genes. Am J Med Genet A 2014; 164A:1991-7. [PMID: 24817631 DOI: 10.1002/ajmg.a.36602] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Accepted: 04/03/2014] [Indexed: 01/24/2023]
Abstract
High-resolution array comparative genomic hybridization (a-CGH) enables the detection of intragenic rearrangements, such as single exon deletion or duplication. This approach can lead to the identification of new disease genes. We report on the analysis of 54 male patients presenting with intellectual deficiency (ID) and a family history suggesting X-linked (XL) inheritance or maternal skewed X-chromosome inactivation (XCI), using a home-made X-chromosome-specific microarray covering the whole human X-chromosome at high resolution. The majority of patients had whole genome array-CGH prior to the selection and we did not include large rearrangements such as MECP2 and FMR1 duplications. We identified four rearrangements considered as causative or potentially pathogenic, corresponding to a detection rate of 8%. Two CNVs affected known XLID genes and were therefore considered as causative (IL1RAPL1 and OPHN1 intragenic deletions). Two new CNVs were considered as potentially pathogenic as they affected interesting candidates for ID. The first CNV is a deletion of the first exon of the TRPC5 gene, encoding a cation channel implicated in dendrite growth and patterning, in a child presenting with ID and an autism spectrum disorder (ASD). The second CNV is a partial deletion of KLHL15, in a patient with severe ID, epilepsy, and anomalies of cortical development. In both cases, in spite of strong arguments for clinical relevance, we were not able at this stage to confirm pathogenicity of the mutations, and the causality of the variants identified in XLID remains to be confirmed.
Collapse
Affiliation(s)
- Cécile Mignon-Ravix
- Inserm, UMR_S 910, Marseille, France; Aix Marseille Université, GMGF, Marseille, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Wright FA, Sullivan PF, Brooks AI, Zou F, Sun W, Xia K, Madar V, Jansen R, Chung W, Zhou YH, Abdellaoui A, Batista S, Butler C, Chen G, Chen TH, D'Ambrosio D, Gallins P, Ha MJ, Hottenga JJ, Huang S, Kattenberg M, Kochar J, Middeldorp CM, Qu A, Shabalin A, Tischfield J, Todd L, Tzeng JY, van Grootheest G, Vink JM, Wang Q, Wang W, Wang W, Willemsen G, Smit JH, de Geus EJ, Yin Z, Penninx BWJH, Boomsma DI. Heritability and genomics of gene expression in peripheral blood. Nat Genet 2014; 46:430-7. [PMID: 24728292 PMCID: PMC4012342 DOI: 10.1038/ng.2951] [Citation(s) in RCA: 257] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2012] [Accepted: 03/14/2014] [Indexed: 12/14/2022]
Abstract
We assessed gene expression profiles in 2,752 twins, using a classic twin design to quantify expression heritability and quantitative trait loci (eQTLs) in peripheral blood. The most highly heritable genes (∼777) were grouped into distinct expression clusters, enriched in gene-poor regions, associated with specific gene function or ontology classes, and strongly associated with disease designation. The design enabled a comparison of twin-based heritability to estimates based on dizygotic identity-by-descent sharing and distant genetic relatedness. Consideration of sampling variation suggests that previous heritability estimates have been upwardly biased. Genotyping of 2,494 twins enabled powerful identification of eQTLs, which we further examined in a replication set of 1,895 unrelated subjects. A large number of non-redundant local eQTLs (6,756) met replication criteria, whereas a relatively small number of distant eQTLs (165) met quality control and replication standards. Our results provide a new resource toward understanding the genetic control of transcription.
Collapse
Affiliation(s)
- Fred A Wright
- 1] Bioinformatics Research Center, North Carolina State University, Raleigh, North Carolina, USA. [2] Department of Statistics, North Carolina State University, Raleigh, North Carolina, USA. [3] Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, USA. [4]
| | - Patrick F Sullivan
- 1] Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA. [2]
| | - Andrew I Brooks
- Department of Genetics, Rutgers University, New Brunswick, New Jersey, USA
| | - Fei Zou
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Wei Sun
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Kai Xia
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Vered Madar
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Rick Jansen
- Department of Psychiatry, VU Medical Center, Amsterdam, The Netherlands
| | - Wonil Chung
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Yi-Hui Zhou
- 1] Bioinformatics Research Center, North Carolina State University, Raleigh, North Carolina, USA. [2] Department of Statistics, North Carolina State University, Raleigh, North Carolina, USA
| | - Abdel Abdellaoui
- Department of Biological Psychology, VU University, Amsterdam, The Netherlands
| | - Sandra Batista
- Department of Computer Science, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Casey Butler
- Department of Computer Science, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Guanhua Chen
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Ting-Huei Chen
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - David D'Ambrosio
- Environmental and Occupational Health Sciences Institute, Rutgers University, New Brunswick, New Jersey, USA
| | - Paul Gallins
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Min Jin Ha
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Jouke Jan Hottenga
- Department of Biological Psychology, VU University, Amsterdam, The Netherlands
| | - Shunping Huang
- Department of Computer Science, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Mathijs Kattenberg
- Department of Biological Psychology, VU University, Amsterdam, The Netherlands
| | - Jaspreet Kochar
- Environmental and Occupational Health Sciences Institute, Rutgers University, New Brunswick, New Jersey, USA
| | | | - Ani Qu
- Environmental and Occupational Health Sciences Institute, Rutgers University, New Brunswick, New Jersey, USA
| | - Andrey Shabalin
- Department of Pharmacotherapy and Outcomes Science, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Jay Tischfield
- Department of Genetics, Rutgers University, New Brunswick, New Jersey, USA
| | - Laura Todd
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Jung-Ying Tzeng
- 1] Bioinformatics Research Center, North Carolina State University, Raleigh, North Carolina, USA. [2] Department of Statistics, North Carolina State University, Raleigh, North Carolina, USA
| | | | - Jacqueline M Vink
- Department of Biological Psychology, VU University, Amsterdam, The Netherlands
| | - Qi Wang
- Environmental and Occupational Health Sciences Institute, Rutgers University, New Brunswick, New Jersey, USA
| | - Wei Wang
- Department of Computer Science, University of California, Los Angeles, Los Angeles, California, USA
| | - Weibo Wang
- Department of Computer Science, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Gonneke Willemsen
- Department of Biological Psychology, VU University, Amsterdam, The Netherlands
| | - Johannes H Smit
- Department of Psychiatry, VU Medical Center, Amsterdam, The Netherlands
| | - Eco J de Geus
- Department of Biological Psychology, VU University, Amsterdam, The Netherlands
| | - Zhaoyu Yin
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | | | - Dorret I Boomsma
- Department of Biological Psychology, VU University, Amsterdam, The Netherlands
| |
Collapse
|
29
|
Neurodevelopmental and neuropsychiatric disorders represent an interconnected molecular system. Mol Psychiatry 2014; 19:294-301. [PMID: 23439483 DOI: 10.1038/mp.2013.16] [Citation(s) in RCA: 147] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Revised: 12/14/2012] [Accepted: 01/02/2013] [Indexed: 12/18/2022]
Abstract
Many putative genetic factors that confer risk to neurodevelopmental disorders such as autism spectrum disorders (ASDs) and X-linked intellectual disability (XLID), and to neuropsychiatric disorders including attention deficit hyperactivity disorder (ADHD) and schizophrenia (SZ) have been identified in individuals from diverse human populations. Although there is significant aetiological heterogeneity within and between these conditions, recent data show that genetic factors contribute to their comorbidity. Many studies have identified candidate gene associations for these mental health disorders, albeit this is often done in a piecemeal fashion with little regard to the inherent molecular complexity. Here, we sought to abstract relationships from our knowledge of systems level biology to help understand the unique and common genetic drivers of these conditions. We undertook a global and systematic approach to build and integrate available data in gene networks associated with ASDs, XLID, ADHD and SZ. Complex network concepts and computational methods were used to investigate whether candidate genes associated with these conditions were related through mechanisms of gene regulation, functional protein-protein interactions, transcription factor (TF) and microRNA (miRNA) binding sites. Although our analyses show that genetic variations associated with the four disorders can occur in the same molecular pathways and functional domains, including synaptic transmission, there are patterns of variation that define significant differences between disorders. Of particular interest is DNA variations located in intergenic regions that comprise regulatory sites for TFs or miRNA. Our approach provides a hypothetical framework, which will help discovery and analysis of candidate genes associated with neurodevelopmental and neuropsychiatric disorders.
Collapse
|
30
|
D'Adamo P, Masetti M, Bianchi V, Morè L, Mignogna ML, Giannandrea M, Gatti S. RAB GTPases and RAB-interacting proteins and their role in the control of cognitive functions. Neurosci Biobehav Rev 2014; 46 Pt 2:302-14. [PMID: 24412241 DOI: 10.1016/j.neubiorev.2013.12.009] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Revised: 11/15/2013] [Accepted: 12/16/2013] [Indexed: 12/18/2022]
Abstract
A RAS-related class of small monomeric G proteins, the RAB GTPases, is emerging as of key biological importance in compartment specific directional control of vesicles formation, transport and fusion. Thanks to human genetic observation and to the consequent dedicated biochemical work, substantial progress has been made on the understanding of the role played by RAB GTPases and their effector proteins on neuronal development and the shaping of cognitive functions. This review is highlighting these initial elements to broaden the current scope of research on developmental cognitive deficits and take the point of view of RAB GTPases control on membrane transport in neurons and astrocytes.
Collapse
Affiliation(s)
- Patrizia D'Adamo
- Dulbecco Telethon Institute at San Raffaele Scientific Institute, Division of Neuroscience, via Olgettina 58, 20132 Milan, Italy; Vita-Salute San Raffaele University, via Olgettina 58, 20132 Milan, Italy.
| | - Michela Masetti
- Dulbecco Telethon Institute at San Raffaele Scientific Institute, Division of Neuroscience, via Olgettina 58, 20132 Milan, Italy
| | - Veronica Bianchi
- Dulbecco Telethon Institute at San Raffaele Scientific Institute, Division of Neuroscience, via Olgettina 58, 20132 Milan, Italy; Vita-Salute San Raffaele University, via Olgettina 58, 20132 Milan, Italy
| | - Lorenzo Morè
- Dulbecco Telethon Institute at San Raffaele Scientific Institute, Division of Neuroscience, via Olgettina 58, 20132 Milan, Italy
| | - Maria Lidia Mignogna
- Dulbecco Telethon Institute at San Raffaele Scientific Institute, Division of Neuroscience, via Olgettina 58, 20132 Milan, Italy; Vita-Salute San Raffaele University, via Olgettina 58, 20132 Milan, Italy
| | - Maila Giannandrea
- Dulbecco Telethon Institute at San Raffaele Scientific Institute, Division of Neuroscience, via Olgettina 58, 20132 Milan, Italy; F. Hoffmann-La Roche AG, pRED Pharma Research & Early Development, DTA Neuroscience Grenzacherstrasse 124, Basel CH4070, Switzerland
| | - Silvia Gatti
- F. Hoffmann-La Roche AG, pRED Pharma Research & Early Development, DTA Neuroscience Grenzacherstrasse 124, Basel CH4070, Switzerland
| |
Collapse
|
31
|
Sarasua SM, Dwivedi A, Boccuto L, Chen CF, Sharp JL, Rollins JD, Collins JS, Rogers RC, Phelan K, DuPont BR. 22q13.2q13.32 genomic regions associated with severity of speech delay, developmental delay, and physical features in Phelan-McDermid syndrome. Genet Med 2013; 16:318-28. [PMID: 24136618 DOI: 10.1038/gim.2013.144] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Accepted: 08/07/2013] [Indexed: 12/18/2022] Open
Abstract
PURPOSE Phelan-McDermid syndrome is a developmental disability syndrome with varying deletions of 22q13 and varying clinical severity. We tested the hypothesis that, in addition to loss of the telomeric gene SHANK3, specific genomic regions within 22q13 are associated with important clinical features. METHODS We used a customized oligo array comparative genomic hybridization of 22q12.3-terminus to obtain deletion breakpoints in a cohort of 70 patients with terminal 22q13 deletions. We used association and receiver operating characteristic statistical methods in a novel manner and also incorporated protein interaction networks to identify 22q13 genomic locations and genes associated with clinical features. RESULTS Specific genomic regions and candidate genes within 22q13.2q13.32 were associated with severity of speech/language delay, neonatal hypotonia, delayed age at walking, hair-pulling behaviors, male genital anomalies, dysplastic toenails, large/fleshy hands, macrocephaly, short and tall stature, facial asymmetry, and atypical reflexes. We also found regions suggestive of a negative association with autism spectrum disorders. CONCLUSION This work advances the field of research beyond the observation of a correlation between deletion size and phenotype and identifies candidate 22q13 loci, and in some cases specific genes, associated with singular clinical features observed in Phelan-McDermid syndrome. Our statistical approach may be useful in genotype-phenotype analyses for other microdeletion or microduplication syndromes.
Collapse
Affiliation(s)
- Sara M Sarasua
- Greenwood Genetic Center, Greenwood, South Carolina, USA
| | - Alka Dwivedi
- Greenwood Genetic Center, Greenwood, South Carolina, USA
| | - Luigi Boccuto
- Greenwood Genetic Center, Greenwood, South Carolina, USA
| | - Chin-Fu Chen
- Greenwood Genetic Center, Greenwood, South Carolina, USA
| | - Julia L Sharp
- Medical University of South Carolina, Charleston, South Carolina, USA
| | | | - Julianne S Collins
- 1] Greenwood Genetic Center, Greenwood, South Carolina, USA [2] Deceased
| | | | - Katy Phelan
- Hayward Genetics Center, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | | |
Collapse
|
32
|
Ripke S, O'Dushlaine C, Chambert K, Moran JL, Kähler AK, Akterin S, Bergen SE, Collins AL, Crowley JJ, Fromer M, Kim Y, Lee SH, Magnusson PKE, Sanchez N, Stahl EA, Williams S, Wray NR, Xia K, Bettella F, Borglum AD, Bulik-Sullivan BK, Cormican P, Craddock N, de Leeuw C, Durmishi N, Gill M, Golimbet V, Hamshere ML, Holmans P, Hougaard DM, Kendler KS, Lin K, Morris DW, Mors O, Mortensen PB, Neale BM, O'Neill FA, Owen MJ, Milovancevic MP, Posthuma D, Powell J, Richards AL, Riley BP, Ruderfer D, Rujescu D, Sigurdsson E, Silagadze T, Smit AB, Stefansson H, Steinberg S, Suvisaari J, Tosato S, Verhage M, Walters JT, Levinson DF, Gejman PV, Kendler KS, Laurent C, Mowry BJ, O'Donovan MC, Owen MJ, Pulver AE, Riley BP, Schwab SG, Wildenauer DB, Dudbridge F, Holmans P, Shi J, Albus M, Alexander M, Campion D, Cohen D, Dikeos D, Duan J, Eichhammer P, Godard S, Hansen M, Lerer FB, Liang KY, Maier W, Mallet J, Nertney DA, Nestadt G, Norton N, O'Neill FA, Papadimitriou GN, Ribble R, Sanders AR, Silverman JM, Walsh D, Williams NM, Wormley B, Arranz MJ, Bakker S, Bender S, Bramon E, Collier D, Crespo-Facorro B, Hall J, Iyegbe C, et alRipke S, O'Dushlaine C, Chambert K, Moran JL, Kähler AK, Akterin S, Bergen SE, Collins AL, Crowley JJ, Fromer M, Kim Y, Lee SH, Magnusson PKE, Sanchez N, Stahl EA, Williams S, Wray NR, Xia K, Bettella F, Borglum AD, Bulik-Sullivan BK, Cormican P, Craddock N, de Leeuw C, Durmishi N, Gill M, Golimbet V, Hamshere ML, Holmans P, Hougaard DM, Kendler KS, Lin K, Morris DW, Mors O, Mortensen PB, Neale BM, O'Neill FA, Owen MJ, Milovancevic MP, Posthuma D, Powell J, Richards AL, Riley BP, Ruderfer D, Rujescu D, Sigurdsson E, Silagadze T, Smit AB, Stefansson H, Steinberg S, Suvisaari J, Tosato S, Verhage M, Walters JT, Levinson DF, Gejman PV, Kendler KS, Laurent C, Mowry BJ, O'Donovan MC, Owen MJ, Pulver AE, Riley BP, Schwab SG, Wildenauer DB, Dudbridge F, Holmans P, Shi J, Albus M, Alexander M, Campion D, Cohen D, Dikeos D, Duan J, Eichhammer P, Godard S, Hansen M, Lerer FB, Liang KY, Maier W, Mallet J, Nertney DA, Nestadt G, Norton N, O'Neill FA, Papadimitriou GN, Ribble R, Sanders AR, Silverman JM, Walsh D, Williams NM, Wormley B, Arranz MJ, Bakker S, Bender S, Bramon E, Collier D, Crespo-Facorro B, Hall J, Iyegbe C, Jablensky A, Kahn RS, Kalaydjieva L, Lawrie S, Lewis CM, Lin K, Linszen DH, Mata I, McIntosh A, Murray RM, Ophoff RA, Powell J, Rujescu D, Van Os J, Walshe M, Weisbrod M, Wiersma D, Donnelly P, Barroso I, Blackwell JM, Bramon E, Brown MA, Casas JP, Corvin AP, Deloukas P, Duncanson A, Jankowski J, Markus HS, Mathew CG, Palmer CNA, Plomin R, Rautanen A, Sawcer SJ, Trembath RC, Viswanathan AC, Wood NW, Spencer CCA, Band G, Bellenguez C, Freeman C, Hellenthal G, Giannoulatou E, Pirinen M, Pearson RD, Strange A, Su Z, Vukcevic D, Donnelly P, Langford C, Hunt SE, Edkins S, Gwilliam R, Blackburn H, Bumpstead SJ, Dronov S, Gillman M, Gray E, Hammond N, Jayakumar A, McCann OT, Liddle J, Potter SC, Ravindrarajah R, Ricketts M, Tashakkori-Ghanbaria A, Waller MJ, Weston P, Widaa S, Whittaker P, Barroso I, Deloukas P, Mathew CG, Blackwell JM, Brown MA, Corvin AP, McCarthy MI, Spencer CCA, Bramon E, Corvin AP, O'Donovan MC, Stefansson K, Scolnick E, Purcell S, McCarroll SA, Sklar P, Hultman CM, Sullivan PF. Genome-wide association analysis identifies 13 new risk loci for schizophrenia. Nat Genet 2013; 45:1150-9. [PMID: 23974872 PMCID: PMC3827979 DOI: 10.1038/ng.2742] [Show More Authors] [Citation(s) in RCA: 1176] [Impact Index Per Article: 98.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2012] [Accepted: 08/01/2013] [Indexed: 12/11/2022]
Abstract
Schizophrenia is an idiopathic mental disorder with a heritable component and a substantial public health impact. We conducted a multi-stage genome-wide association study (GWAS) for schizophrenia beginning with a Swedish national sample (5,001 cases and 6,243 controls) followed by meta-analysis with previous schizophrenia GWAS (8,832 cases and 12,067 controls) and finally by replication of SNPs in 168 genomic regions in independent samples (7,413 cases, 19,762 controls and 581 parent-offspring trios). We identified 22 loci associated at genome-wide significance; 13 of these are new, and 1 was previously implicated in bipolar disorder. Examination of candidate genes at these loci suggests the involvement of neuronal calcium signaling. We estimate that 8,300 independent, mostly common SNPs (95% credible interval of 6,300-10,200 SNPs) contribute to risk for schizophrenia and that these collectively account for at least 32% of the variance in liability. Common genetic variation has an important role in the etiology of schizophrenia, and larger studies will allow more detailed understanding of this disorder.
Collapse
Affiliation(s)
- Stephan Ripke
- 1] Analytical and Translational Genetics Unit, Massachusetts General Hospital, Boston, Massachusetts, USA. [2] Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA. [3]
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Ikeuchi Y, de la Torre-Ubieta L, Matsuda T, Steen H, Okazawa H, Bonni A. The XLID protein PQBP1 and the GTPase Dynamin 2 define a signaling link that orchestrates ciliary morphogenesis in postmitotic neurons. Cell Rep 2013; 4:879-89. [PMID: 23994472 DOI: 10.1016/j.celrep.2013.07.042] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Revised: 07/17/2013] [Accepted: 07/26/2013] [Indexed: 10/26/2022] Open
Abstract
Intellectual disability (ID) is a prevalent developmental disorder of cognition that remains incurable. Here, we report that knockdown of the X-linked ID (XLID) protein polyglutamine-binding protein 1 (PQBP1) in neurons profoundly impairs the morphogenesis of the primary cilium, including in the mouse cerebral cortex in vivo. PQBP1 is localized at the base of the neuronal cilium, and targeting its WW effector domain to the cilium stimulates ciliary morphogenesis. We also find that PQBP1 interacts with Dynamin 2 and thereby inhibits its GTPase activity. Accordingly, Dynamin 2 knockdown in neurons stimulates ciliogenesis and suppresses the PQBP1 knockdown-induced ciliary phenotype. Strikingly, a mutation of the PQBP1 WW domain that causes XLID disrupts its ability to interact with and inhibit Dynamin 2 and to induce neuronal ciliogenesis. These findings define PQBP1 and Dynamin 2 as components of a signaling pathway that orchestrates neuronal ciliary morphogenesis in the brain.
Collapse
Affiliation(s)
- Yoshiho Ikeuchi
- Department of Anatomy and Neurobiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | |
Collapse
|
34
|
Jorge P, Oliveira B, Marques I, Santos R. Development and validation of a multiplex-PCR assay for X-linked intellectual disability. BMC MEDICAL GENETICS 2013; 14:80. [PMID: 23914978 PMCID: PMC3751858 DOI: 10.1186/1471-2350-14-80] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Accepted: 06/07/2013] [Indexed: 12/03/2022]
Abstract
Background X-linked intellectual disability is a common cause of inherited cognitive deficit affecting mostly males. There are several genetic causes implicated in this condition, which has hampered the establishment of an accurate diagnosis. We developed a multiplex-PCR assay for the mutational hotspot regions of the FMR1, AFF2 and ARX genes. Methods The multiplex-PCR was validated in a cohort of 100 males selected to include known alleles for the FMR1 repetitive region: five full mutations (250–650 CGGs), ten premutations (70–165 CGGs) and eighty-five in the normal range (19–42 CGGs). Sequencing or Southern blotting was used to confirm the results, depending on the allele class. In this cohort, with the exception of one sample showing an AFF2 intermediate-sized allele, all other samples were normal (8–34 CCGs). No ARX variant was found besides the c.429_452dup. The validated assay was applied to 5000 samples (64.4% males and 35.6% females). Results The normal-allelic range of both FMR1 and AFF2 genes as well as the nature of ARX variants identified was similar in both genders. The rate of homozygosity observed in female samples, 27.5% for FMR1 and 17.8% for AFF2 alleles, is comparable to that published by others. Two FMR1 premutations were identified, in a male (58 CGGs) and a female case [(CGG)47/(CGG)61], as well as several FMR1 or AFF2 intermediate-sized alleles. One AFF2 premutation (68 CCGs) and two putative full expansions were picked up in male subjects, which seems relevant considering the rarity of reported AFF2 mutations found in the absence of a family history. Conclusions We developed a robust multiplex-PCR that can be used to screen the mutational hotspot regions of FMR1, AFF2 and ARX genes. Moreover, this strategy led to the identification of variants in all three genes, representing not only an improvement in allele-sizing but also in achieving a differential diagnosis. Although the distinction between females who are truly homozygous and those with a second pre- or full mutation sized allele, as well as a definitive diagnosis, requires a specific downstream technique, the use of this multiplex-PCR for initial screening is a cost-effective approach which widens the scope of detection.
Collapse
Affiliation(s)
- Paula Jorge
- Centro de Genética Médica Dr, Jacinto Magalhães, CHP, Praça Pedro Nunes 88, 4099-028, Porto, Portugal.
| | | | | | | |
Collapse
|
35
|
Maris AF, Barbato IT, Trott A, Montano MAE. Familial mental retardation: a review and practical classification. CIENCIA & SAUDE COLETIVA 2013. [DOI: 10.1590/s1413-81232013000600023] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Mental retardation (MR) is a definition which comprises a series of conditions whose common feature is an intellectual handicap that develops before the age of 18, afflicting 2-3% of the world's population. The classification of MR into different categories is determined by the extent of the handicap instead of its cause, which often remains unrecognized. Sometimes, MR runs in a family, characterizing familial MR, and those cases permit an in-depth look into the genetic causes and consequences of the problem. However, almost no work is available on the prevalence of familial MR among the registered MR cases, possibly because familial MR is a term with no clear definition. The scope of this work is to review the topic and discuss the implications of different genetic and environmental factors, which characterize particular categories of familial cases, suggesting a practical classification of familial MR, which is important for epidemiologic studies and also for counseling in the clinic. Some of the aspects are discussed under the perspective of a newly-developed country like Brazil.
Collapse
|
36
|
Isrie M, Kalscheuer VM, Holvoet M, Fieremans N, Van Esch H, Devriendt K. HUWE1 mutation explains phenotypic severity in a case of familial idiopathic intellectual disability. Eur J Med Genet 2013; 56:379-82. [PMID: 23721686 DOI: 10.1016/j.ejmg.2013.05.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2013] [Accepted: 05/08/2013] [Indexed: 01/12/2023]
Abstract
The advent of next-generation sequencing has proven to be a key force in the identification of new genes associated with intellectual disability. In this study, high-throughput sequencing of the coding regions of the X-chromosome led to the identification of a missense variant in the HUWE1 gene. The same variant has been reported before by Froyen et al. (2008). We compare the phenotypes and demonstrate that, in the present family, the HUWE1 mutation segregates with the more severe ID phenotypes of two out of three brothers. The third brother has a milder form of ID and does not carry the mutation.
Collapse
Affiliation(s)
- Mala Isrie
- Center for Human Genetics, University Hospitals Leuven, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | | | | | | | | | | |
Collapse
|
37
|
Seixas E, Barros M, Seabra MC, Barral DC. Rab and Arf proteins in genetic diseases. Traffic 2013; 14:871-85. [PMID: 23565987 DOI: 10.1111/tra.12072] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2012] [Revised: 04/03/2013] [Accepted: 04/08/2013] [Indexed: 01/29/2023]
Abstract
Rab and ADP-ribosylation factor (Arf) family proteins are master regulators of membrane trafficking and are involved in all steps of vesicular transport. These families of small guanine-nucleotide-binding (G) proteins are well suited to regulate membrane trafficking processes since their nucleotide state determines their conformation and the capacity to bind to a multitude of effectors, which mediate their functions. In recent years, several inherited diseases have been associated with mutations in genes encoding proteins belonging to these two families or in proteins that regulate their GTP-binding cycle. The genetic diseases that are caused by defects in Rabs, Arfs or their regulatory proteins are heterogeneous and display diverse symptoms. However, these diseases mainly affect two types of subcellular compartments, namely lysosome-related organelles and cilia. Also, several of these diseases affect the nervous system. Thus, the study of these diseases represents an opportunity to understand their etiology and the molecular mechanisms involved, as well as to develop novel therapeutic strategies.
Collapse
Affiliation(s)
- Elsa Seixas
- CEDOC, Faculdade de Ciências Médicas, FCM, Universidade Nova de Lisboa, 1169-056, Lisboa, Portugal
| | | | | | | |
Collapse
|
38
|
XLID CUL4B mutants are defective in promoting TSC2 degradation and positively regulating mTOR signaling in neocortical neurons. Biochim Biophys Acta Mol Basis Dis 2013; 1832:585-93. [DOI: 10.1016/j.bbadis.2013.01.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Revised: 12/17/2012] [Accepted: 01/08/2013] [Indexed: 01/06/2023]
|
39
|
Minić S, Trpinac D, Obradović M. Systematic review of central nervous system anomalies in incontinentia pigmenti. Orphanet J Rare Dis 2013; 8:25. [PMID: 23406512 PMCID: PMC3576363 DOI: 10.1186/1750-1172-8-25] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Accepted: 02/05/2013] [Indexed: 11/16/2022] Open
Abstract
The objective of this study was to present a systematic review of the central nervous system (CNS) types of anomalies and to consider the possibility to include CNS anomalies in Incontinentia pigmenti (IP) criteria. The analyzed literature data from 1,393 IP cases were from the period 1993–2012. CNS anomalies were diagnosed for 30.44% of the investigated IP patients. The total number of CNS types of anomalies per patient was 1.62. In the present study there was no significantly higher number of anomalies per patient in females than males. The most frequent CNS types of anomalies were seizures, motor impairment, mental retardation, and microcephaly. The most frequently registered CNS lesions found using brain imaging methods were brain infarcts or necrosis, brain atrophies, and corpus callosum lesions. IKBKG exon 4–10 deletion was present in 86.00% of genetically confirmed IP patients. The frequency of CNS anomalies, similar to the frequency of retinal anomalies in IP patients, concurrent with their severity, supports their recognition in the list of IP minor criteria.
Collapse
Affiliation(s)
- Snežana Minić
- School of Medicine, University of Belgrade, and Dermatovenerology Clinic, Clinical Center of Serbia, Belgrade, Serbia.
| | | | | |
Collapse
|
40
|
Grafodatskaya D, Chung BHY, Butcher DT, Turinsky AL, Goodman SJ, Choufani S, Chen YA, Lou Y, Zhao C, Rajendram R, Abidi FE, Skinner C, Stavropoulos J, Bondy CA, Hamilton J, Wodak S, Scherer SW, Schwartz CE, Weksberg R. Multilocus loss of DNA methylation in individuals with mutations in the histone H3 lysine 4 demethylase KDM5C. BMC Med Genomics 2013; 6:1. [PMID: 23356856 PMCID: PMC3573947 DOI: 10.1186/1755-8794-6-1] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Accepted: 01/14/2013] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND A number of neurodevelopmental syndromes are caused by mutations in genes encoding proteins that normally function in epigenetic regulation. Identification of epigenetic alterations occurring in these disorders could shed light on molecular pathways relevant to neurodevelopment. RESULTS Using a genome-wide approach, we identified genes with significant loss of DNA methylation in blood of males with intellectual disability and mutations in the X-linked KDM5C gene, encoding a histone H3 lysine 4 demethylase, in comparison to age/sex matched controls. Loss of DNA methylation in such individuals is consistent with known interactions between DNA methylation and H3 lysine 4 methylation. Further, loss of DNA methylation at the promoters of the three top candidate genes FBXL5, SCMH1, CACYBP was not observed in more than 900 population controls. We also found that DNA methylation at these three genes in blood correlated with dosage of KDM5C and its Y-linked homologue KDM5D. In addition, parallel sex-specific DNA methylation profiles in brain samples from control males and females were observed at FBXL5 and CACYBP. CONCLUSIONS We have, for the first time, identified epigenetic alterations in patient samples carrying a mutation in a gene involved in the regulation of histone modifications. These data support the concept that DNA methylation and H3 lysine 4 methylation are functionally interdependent. The data provide new insights into the molecular pathogenesis of intellectual disability. Further, our data suggest that some DNA methylation marks identified in blood can serve as biomarkers of epigenetic status in the brain.
Collapse
Affiliation(s)
- Daria Grafodatskaya
- Genetics and Genome Biology Program, Hospital for Sick Children, Toronto, ON, Canada
| | - Barian HY Chung
- Division of Clinical and Metabolic Genetics, Hospital for Sick Children, Toronto, ON, Canada
- Centre of Reproduction, Growth & Development, Department of Pediatrics & Adolescent Medicine, The University of Hong Kong, Hong Kong, Hong Kong
| | - Darci T Butcher
- Genetics and Genome Biology Program, Hospital for Sick Children, Toronto, ON, Canada
| | - Andrei L Turinsky
- Program in Molecular Structure and Function, Hospital for Sick Children, Toronto, ON, Canada
| | - Sarah J Goodman
- Centre of Reproduction, Growth & Development, Department of Pediatrics & Adolescent Medicine, The University of Hong Kong, Hong Kong, Hong Kong
| | - Sana Choufani
- Centre of Reproduction, Growth & Development, Department of Pediatrics & Adolescent Medicine, The University of Hong Kong, Hong Kong, Hong Kong
| | - Yi-An Chen
- Genetics and Genome Biology Program, Hospital for Sick Children, Toronto, ON, Canada
| | - Youliang Lou
- Genetics and Genome Biology Program, Hospital for Sick Children, Toronto, ON, Canada
| | - Chunhua Zhao
- Genetics and Genome Biology Program, Hospital for Sick Children, Toronto, ON, Canada
| | - Rageen Rajendram
- Genetics and Genome Biology Program, Hospital for Sick Children, Toronto, ON, Canada
| | - Fatima E Abidi
- J.C. Self Research Institute, Greenwood Genetic Center, Greenwood, SC, USA
| | - Cindy Skinner
- J.C. Self Research Institute, Greenwood Genetic Center, Greenwood, SC, USA
| | - James Stavropoulos
- Department of Pediatric Laboratory Medicine, Hospital for Sick Children, Toronto, ON, Canada
| | - Carolyn A Bondy
- Developmental Endocrinology Branch, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Jill Hamilton
- Division of Endocrinology, Department of Pediatrics, Hospital for Sick Children, Toronto, ON, Canada
- Department of Pediatrics, University of Toronto, Toronto, ON, Canada
| | - Shoshana Wodak
- Program in Molecular Structure and Function, Hospital for Sick Children, Toronto, ON, Canada
- Department of Molecular and Medical Genetics, University of Toronto, Toronto, ON, Canada
| | - Stephen W Scherer
- Department of Molecular and Medical Genetics, University of Toronto, Toronto, ON, Canada
- The Centre for Applied Genomics, Hospital for Sick Children, Toronto, ON, Canada
| | - Charles E Schwartz
- J.C. Self Research Institute, Greenwood Genetic Center, Greenwood, SC, USA
| | - Rosanna Weksberg
- Division of Clinical and Metabolic Genetics, Hospital for Sick Children, Toronto, ON, Canada
- Department of Pediatrics, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
41
|
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
|
42
|
Bedoyan JK, Schaibley VM, Peng W, Bai Y, Mondal K, Shetty AC, Durham M, Micucci JA, Dhiraaj A, Skidmore JM, Kaplan JB, Skinner C, Schwartz CE, Antonellis A, Zwick ME, Cavalcoli JD, Li JZ, Martin DM. Disruption of RAB40AL function leads to Martin--Probst syndrome, a rare X-linked multisystem neurodevelopmental human disorder. J Med Genet 2012; 49:332-40. [PMID: 22581972 PMCID: PMC3350147 DOI: 10.1136/jmedgenet-2011-100575] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
BACKGROUND AND AIM Martin--Probst syndrome (MPS) is a rare X-linked disorder characterised by deafness, cognitive impairment, short stature and distinct craniofacial dysmorphisms, among other features. The authors sought to identify the causative mutation for MPS. METHODS AND RESULTS Massively parallel sequencing in two affected, related male subjects with MPS identified a RAB40AL (also called RLGP) missense mutation (chrX:102,079,078-102,079,079AC→GA p.D59G; hg18). RAB40AL encodes a small Ras-like GTPase protein with one suppressor of cytokine signalling box. The p.D59G variant is located in a highly conserved region of the GTPase domain between β-2 and β-3 strands. Using RT-PCR, the authors show that RAB40AL is expressed in human fetal and adult brain and kidney, and adult lung, heart, liver and skeletal muscle. RAB40AL appears to be a primate innovation, with no orthologues found in mouse, Xenopus or zebrafish. Western analysis and fluorescence microscopy of GFP-tagged RAB40AL constructs from transiently transfected COS7 cells show that the D59G missense change renders RAB40AL unstable and disrupts its cytoplasmic localisation. CONCLUSIONS This is the first study to show that mutation of RAB40AL is associated with a human disorder. Identification of RAB40AL as the gene mutated in MPS allows for further investigations into the molecular mechanism(s) of RAB40AL and its roles in diverse processes such as cognition, hearing and skeletal development.
Collapse
|
43
|
MED12 mutations link intellectual disability syndromes with dysregulated GLI3-dependent Sonic Hedgehog signaling. Proc Natl Acad Sci U S A 2012; 109:19763-8. [PMID: 23091001 DOI: 10.1073/pnas.1121120109] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Recurrent missense mutations in the RNA polymerase II Mediator subunit MED12 are associated with X-linked intellectual disability (XLID) and multiple congenital anomalies, including craniofacial, musculoskeletal, and behavioral defects in humans with FG (or Opitz-Kaveggia) and Lujan syndromes. However, the molecular mechanism(s) underlying these phenotypes is poorly understood. Here we report that MED12 mutations R961W and N1007S causing FG and Lujan syndromes, respectively, disrupt a Mediator-imposed constraint on GLI3-dependent Sonic Hedgehog (SHH) signaling. We show that the FG/R961W and Lujan/N1007S mutations disrupt the gene-specific association of MED12 with a second Mediator subunit, CDK8, identified herein to be a suppressor of GLI3 transactivation activity. In FG/R961W and Lujan/N1007S patient-derived cells, we document enhanced SHH pathway activation and GLI3-target gene induction coincident with impaired recruitment of CDK8 onto promoters of GLI3-target genes, but not non-GLI3-target genes. Together, these findings suggest that dysregulated GLI3-dependent SHH signaling contributes to phenotypes of individuals with FG and Lujan syndromes and further reveal a basis for the gene-specific manifestation of pathogenic mutations in a global transcriptional coregulator.
Collapse
|
44
|
Mekhoubad S, Bock C, de Boer AS, Kiskinis E, Meissner A, Eggan K. Erosion of dosage compensation impacts human iPSC disease modeling. Cell Stem Cell 2012; 10:595-609. [PMID: 22560080 DOI: 10.1016/j.stem.2012.02.014] [Citation(s) in RCA: 264] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2011] [Revised: 01/07/2012] [Accepted: 02/06/2012] [Indexed: 11/16/2022]
Abstract
Although distinct human induced pluripotent stem cell (hiPSC) lines can display considerable epigenetic variation, it has been unclear whether such variability impacts their utility for disease modeling. Here, we show that although low-passage female hiPSCs retain the inactive X chromosome of the somatic cell they are derived from, over time in culture they undergo an "erosion" of X chromosome inactivation (XCI). This erosion of XCI is characterized by loss of XIST expression and foci of H3-K27-trimethylation, as well as transcriptional derepression of genes on the inactive X that cannot be reversed by either differentiation or further reprogramming. We specifically demonstrate that erosion of XCI has a significant impact on the use of female hiPSCs for modeling Lesch-Nyhan syndrome. However, our finding that most genes subject to XCI are derepressed by this erosion of XCI suggests that it should be a significant consideration when selecting hiPSC lines for modeling any disease.
Collapse
Affiliation(s)
- Shila Mekhoubad
- The Harvard Stem Cell Institute, Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | | | | | | | | | | |
Collapse
|
45
|
Genome-wide DNA methylation analysis in patients with familial ATR-X mental retardation syndrome. Epigenomics 2012. [DOI: 10.1017/cbo9780511777271.037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
46
|
Takano K, Liu D, Tarpey P, Gallant E, Lam A, Witham S, Alexov E, Chaubey A, Stevenson RE, Schwartz CE, Board PG, Dulhunty AF. An X-linked channelopathy with cardiomegaly due to a CLIC2 mutation enhancing ryanodine receptor channel activity. Hum Mol Genet 2012; 21:4497-507. [PMID: 22814392 DOI: 10.1093/hmg/dds292] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Chloride intracellular channel 2 (CLIC2) protein is a member of the glutathione transferase class of proteins. Its' only known function is the regulation of ryanodine receptor (RyR) intracellular Ca(2+) release channels. These RyR proteins play a major role in the regulation of Ca(2+) signaling in many cells. Utilizing exome capture and deep sequencing of genes on the X-chromosome, we have identified a mutation in CLIC2 (c.303C>G, p.H101Q) which is associated with X-linked intellectual disability (ID), atrial fibrillation, cardiomegaly, congestive heart failure (CHF), some somatic features and seizures. Functional studies of the H101Q variant indicated that it stimulated rather than inhibited the action of RyR channels, with channels remaining open for longer times and potentially amplifying Ca(2+) signals dependent on RyR channel activity. The overly active RyRs in cardiac and skeletal muscle cells and neuronal cells would result in abnormal cardiac function and trigger post-synaptic pathways and neurotransmitter release. The presence of both cardiomegaly and CHF in the two affected males and atrial fibrillation in one are consistent with abnormal RyR2 channel function. Since the dysfunction of RyR2 channels in the brain via 'leaky mutations' can result in mild developmental delay and seizures, our data also suggest a vital role for the CLIC2 protein in maintaining normal cognitive function via its interaction with RyRs in the brain. Therefore, our patients appear to suffer from a new channelopathy comprised of ID, seizures and cardiac problems because of enhanced Ca(2+) release through RyRs in neuronal cells and cardiac muscle cells.
Collapse
Affiliation(s)
- Kyoko Takano
- JC Self Research Institute, Greenwood Genetic Center, Greenwood, SC 29646, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Sullivan PF, Daly MJ, O'Donovan M. Genetic architectures of psychiatric disorders: the emerging picture and its implications. Nat Rev Genet 2012; 13:537-51. [PMID: 22777127 PMCID: PMC4110909 DOI: 10.1038/nrg3240] [Citation(s) in RCA: 842] [Impact Index Per Article: 64.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Psychiatric disorders are among the most intractable enigmas in medicine. In the past 5 years, there has been unprecedented progress on the genetics of many of these conditions. In this Review, we discuss the genetics of nine cardinal psychiatric disorders (namely, Alzheimer's disease, attention-deficit hyperactivity disorder, alcohol dependence, anorexia nervosa, autism spectrum disorder, bipolar disorder, major depressive disorder, nicotine dependence and schizophrenia). Empirical approaches have yielded new hypotheses about aetiology and now provide data on the often debated genetic architectures of these conditions, which have implications for future research strategies. Further study using a balanced portfolio of methods to assess multiple forms of genetic variation is likely to yield many additional new findings.
Collapse
Affiliation(s)
- Patrick F Sullivan
- Departments of Genetics and Psychiatry, CB# 7264, 5097 Genomic Medicine, University of North Carolina at Chapel Hill, North Carolina 27599-27264, USA.
| | | | | |
Collapse
|
48
|
Chen CY, Tsai MS, Lin CY, Yu IS, Chen YT, Lin SR, Juan LW, Chen YT, Hsu HM, Lee LJ, Lin SW. Rescue of the genetically engineered Cul4b mutant mouse as a potential model for human X-linked mental retardation. Hum Mol Genet 2012; 21:4270-85. [PMID: 22763239 DOI: 10.1093/hmg/dds261] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Mutation in CUL4B, which encodes a scaffold protein of the E3 ubiquitin ligase complex, has been found in patients with X-linked mental retardation (XLMR). However, early deletion of Cul4b in mice causes prenatal lethality, which has frustrated attempts to characterize the phenotypes in vivo. In this report, we successfully rescued Cul4b mutant mice by crossing female mice in which exons 4-5 of Cul4b were flanked by loxP sequences with Sox2-Cre male mice. In Cul4b-deficient (Cul4b(Δ)/Y) mice, no CUL4B protein was detected in any of the major organs, including the brain. In the hippocampus, the levels of CUL4A, CUL4B substrates (TOP1, β-catenin, cyclin E and WDR5) and neuronal markers (MAP2, tau-1, GAP-43, PSD95 and syn-1) were not sensitive to Cul4b deletion, whereas the number of parvalbumin (PV)-positive GABAergic interneurons was decreased in Cul4b(Δ)/Y mice, especially in the dentate gyrus (DG). Some dendritic features, including the complexity, diameter and spine density in the CA1 and DG hippocampal neurons, were also affected by Cul4b deletion. Together, the decrease in the number of PV-positive neurons and altered dendritic properties in Cul4b(Δ)/Y mice imply a reduction in inhibitory regulation and dendritic integration in the hippocampal neural circuit, which lead to increased epileptic susceptibility and spatial learning deficits. Our results identify Cul4b(Δ)/Y mice as a potential model for the non-syndromic model of XLMR that replicates the CUL4B-associated MR and is valuable for the development of a therapeutic strategy for treating MR.
Collapse
Affiliation(s)
- Chun-Yu Chen
- Department of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Alesi V, Bertoli M, Barrano G, Torres B, Pusceddu S, Pastorino M, Perria C, Nardone AM, Novelli A, Serra G. 335.4 kb microduplication in chromosome band Xp11.2p11.3 associated with developmental delay, growth retardation, autistic disorder and dysmorphic features. Gene 2012; 505:384-7. [PMID: 22634100 DOI: 10.1016/j.gene.2012.05.031] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Revised: 04/30/2012] [Accepted: 05/06/2012] [Indexed: 02/02/2023]
Abstract
About 10% of causative mutations for mental retardation in male patients involve X chromosome (X-linked mental retardation, XLMR). We describe a case of a 3-year-old boy presenting with developmental delay, autistic features and growth and speech delay. Array-CGH analysis detected a microduplication on the X chromosome (Xp11.2p11.3), spanning 335.4 kb and including 3 known genes (ZNF81, ZNF182 and SPACA5). Genome-wide association studies show that approximately 30% of mutations causing XLMR are located in Xp11.2p11.3, where few pathogenic genes have been identified to date (such as ZNF41, PQB1 and ZNF81). ZNF81 codifies a zinc finger protein and mutations (non-sense mutations, deletions and structural rearrangements) involving this gene have already been described in association with mental retardation. Larger duplications in the same region have also been observed in association with mental retardation, and, in one case, the over-expression of ZNF81 has also been verified by mRNA quantification. No duplications of the single gene have been identified. To our knowledge, the microduplication found in our patient is the smallest ever described in Xp11.2p11.3. This suggests that the over-expression of ZNF81 could have pathological effects.
Collapse
Affiliation(s)
- Viola Alesi
- S. Pietro Fatebenefratelli Hospital, UOSD Medical Genetics, Rome, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Zhao Y, Sun Y. CUL4B ubiquitin ligase in mouse development: a model for human X-linked mental retardation syndrome? Cell Res 2012; 22:1224-6. [PMID: 22584258 DOI: 10.1038/cr.2012.79] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
CUL4B, a member of the cullin-RING ubiquitin ligase family, is frequently mutated in X-linked mental retardation (XLMR) patients. The study by Liu et al. showed that Cul4b plays an essential developmental role in the extra-embryonic tissues, while it is dispensable in the embryo proper during mouse embryogenesis. Viable Cul4b-null mice provide the first animal model to study neuronal and behavioral deficiencies seen in human CUL4B XLMR patients.
Collapse
Affiliation(s)
- Yongchao Zhao
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, University of Michigan, 4424B MS-1, 1301 Catherine Street, Ann Arbor, MI 48109, USA
| | | |
Collapse
|