50 results on '"Dobyns, William B."'
Search Results
2. Clinical and functional heterogeneity associated with the disruption of retinoic acid receptor beta
- Author
-
Caron, Véronique, Chassaing, Nicolas, Ragge, Nicola, Boschann, Felix, Ngu, Angelina My-Hoa, Meloche, Elisabeth, Chorfi, Sarah, Lakhani, Saquib A, Ji, Weizhen, Steiner, Laurie, Marcadier, Julien, Jansen, Philip R, van de Pol, Laura A, van Hagen, Johanna M, Russi, Alvaro Serrano, Le Guyader, Gwenaël, Nordenskjöld, Magnus, Nordgren, Ann, Anderlid, Britt-Marie, Plaisancié, Julie, Stoltenburg, Corinna, Horn, Denise, Drenckhahn, Anne, Hamdan, Fadi F, Lefebvre, Mathilde, Attie-Bitach, Tania, Forey, Peggy, Smirnov, Vasily, Ernould, Françoise, Jacquemont, Marie-Line, Grotto, Sarah, Alcantud, Alberto, Coret, Alicia, Ferrer-Avargues, Rosario, Srivastava, Siddharth, Vincent-Delorme, Catherine, Romoser, Shelby, Safina, Nicole, Saade, Dimah, Lupski, James R, Calame, Daniel G, Geneviève, David, Chatron, Nicolas, Schluth-Bolard, Caroline, Myers, Kenneth A, Dobyns, William B, Calvas, Patrick, Study, The DDD, Salmon, Caroline, Holt, Richard, Elmslie, Frances, Allaire, Marc, Prigozhin, Daniil M, Tremblay, André, and Michaud, Jacques L
- Subjects
Biological Sciences ,Genetics ,Clinical Research ,Pediatric ,2.1 Biological and endogenous factors ,Aetiology ,Humans ,Receptors ,Retinoic Acid ,Retinoids ,Microphthalmos ,DDD Study ,Dystonia ,Global developmental delay ,Microphthalmia ,Retinoic acid ,Retinoic acid receptor beta ,Clinical Sciences ,Genetics & Heredity - Abstract
PurposeDominant variants in the retinoic acid receptor beta (RARB) gene underlie a syndromic form of microphthalmia, known as MCOPS12, which is associated with other birth anomalies and global developmental delay with spasticity and/or dystonia. Here, we report 25 affected individuals with 17 novel pathogenic or likely pathogenic variants in RARB. This study aims to characterize the functional impact of these variants and describe the clinical spectrum of MCOPS12.MethodsWe used in vitro transcriptional assays and in silico structural analysis to assess the functional relevance of RARB variants in affecting the normal response to retinoids.ResultsWe found that all RARB variants tested in our assays exhibited either a gain-of-function or a loss-of-function activity. Loss-of-function variants disrupted RARB function through a dominant-negative effect, possibly by disrupting ligand binding and/or coactivators' recruitment. By reviewing clinical data from 52 affected individuals, we found that disruption of RARB is associated with a more variable phenotype than initially suspected, with the absence in some individuals of cardinal features of MCOPS12, such as developmental eye anomaly or motor impairment.ConclusionOur study indicates that pathogenic variants in RARB are functionally heterogeneous and associated with extensive clinical heterogeneity.
- Published
- 2023
3. TMEM161B modulates radial glial scaffolding in neocortical development
- Author
-
Wang, Lu, Heffner, Caleb, Vong, Keng loi, Barrows, Chelsea, Ha, Yoo-Jin, Lee, Sangmoon, Lara-Gonzalez, Pablo, Jhamb, Ishani, Van Der Meer, Dennis, Loughnan, Robert, Parker, Nadine, Sievert, David, Mittal, Swapnil, Issa, Mahmoud Y, Andreassen, Ole A, Dale, Anders, Dobyns, William B, Zaki, Maha S, Murray, Stephen A, and Gleeson, Joseph G
- Subjects
Biochemistry and Cell Biology ,Biomedical and Clinical Sciences ,Biological Sciences ,Genetics ,Clinical Research ,Intellectual and Developmental Disabilities (IDD) ,Brain Disorders ,Neurological ,Animals ,Humans ,Mice ,Ependymoglial Cells ,Mice ,Knockout ,Neocortex ,TMEM161B ,gyrification ,CDC42 ,knock-in crispant mice ,patient-derived brain organoids - Abstract
TMEM161B encodes an evolutionarily conserved widely expressed novel 8-pass transmembrane protein of unknown function in human. Here we identify TMEM161B homozygous hypomorphic missense variants in our recessive polymicrogyria (PMG) cohort. Patients carrying TMEM161B mutations exhibit striking neocortical PMG and intellectual disability. Tmem161b knockout mice fail to develop midline hemispheric cleavage, whereas knock-in of patient mutations and patient-derived brain organoids show defects in apical cell polarity and radial glial scaffolding. We found that TMEM161B modulates actin filopodia, functioning upstream of the Rho-GTPase CDC42. Our data link TMEM161B with human PMG, likely regulating radial glia apical polarity during neocortical development.
- Published
- 2023
4. Missense variants in DPYSL5 cause a neurodevelopmental disorder with corpus callosum agenesis and cerebellar abnormalities
- Author
-
Jeanne, Médéric, Demory, Hélène, Moutal, Aubin, Vuillaume, Marie-Laure, Blesson, Sophie, Thépault, Rose-Anne, Marouillat, Sylviane, Halewa, Judith, Maas, Saskia M, Motazacker, M Mahdi, Mancini, Grazia MS, van Slegtenhorst, Marjon A, Andreou, Avgi, Cox, Helene, Vogt, Julie, Laufman, Jason, Kostandyan, Natella, Babikyan, Davit, Hancarova, Miroslava, Bendova, Sarka, Sedlacek, Zdenek, Aldinger, Kimberly A, Sherr, Elliott H, Argilli, Emanuela, England, Eleina M, Audebert-Bellanger, Séverine, Bonneau, Dominique, Colin, Estelle, Denommé-Pichon, Anne-Sophie, Gilbert-Dussardier, Brigitte, Isidor, Bertrand, Küry, Sébastien, Odent, Sylvie, Redon, Richard, Khanna, Rajesh, Dobyns, William B, Bézieau, Stéphane, Honnorat, Jérôme, Lohkamp, Bernhard, Toutain, Annick, and Laumonnier, Frédéric
- Subjects
Biological Sciences ,Biomedical and Clinical Sciences ,Genetics ,Rare Diseases ,Neurosciences ,Brain Disorders ,Pediatric ,Intellectual and Developmental Disabilities (IDD) ,Mental Health ,Aetiology ,2.1 Biological and endogenous factors ,Mental health ,Neurological ,Adult ,Agenesis of Corpus Callosum ,Cerebellum ,Child ,Child ,Preschool ,Female ,Humans ,Hydrolases ,Intellectual Disability ,Male ,Microtubule-Associated Proteins ,Models ,Molecular ,Mutation ,Missense ,Neurodevelopmental Disorders ,Tubulin ,Young Adult ,DPYSL5 ,brain malformation ,corpus callosum agenesis ,de novo missense variants ,dendrite branching ,neurodevelopmental disorder ,primary neuronal cultures ,Medical and Health Sciences ,Genetics & Heredity ,Biological sciences ,Biomedical and clinical sciences ,Health sciences - Abstract
The collapsin response mediator protein (CRMP) family proteins are intracellular mediators of neurotrophic factors regulating neurite structure/spine formation and are essential for dendrite patterning and directional axonal pathfinding during brain developmental processes. Among this family, CRMP5/DPYSL5 plays a significant role in neuronal migration, axonal guidance, dendrite outgrowth, and synapse formation by interacting with microtubules. Here, we report the identification of missense mutations in DPYSL5 in nine individuals with brain malformations, including corpus callosum agenesis and/or posterior fossa abnormalities, associated with variable degrees of intellectual disability. A recurrent de novo p.Glu41Lys variant was found in eight unrelated patients, and a p.Gly47Arg variant was identified in one individual from the first family reported with Ritscher-Schinzel syndrome. Functional analyses of the two missense mutations revealed impaired dendritic outgrowth processes in young developing hippocampal primary neuronal cultures. We further demonstrated that these mutations, both located in the same loop on the surface of DPYSL5 monomers and oligomers, reduced the interaction of DPYSL5 with neuronal cytoskeleton-associated proteins MAP2 and βIII-tubulin. Our findings collectively indicate that the p.Glu41Lys and p.Gly47Arg variants impair DPYSL5 function on dendritic outgrowth regulation by preventing the formation of the ternary complex with MAP2 and βIII-tubulin, ultimately leading to abnormal brain development. This study adds DPYSL5 to the list of genes implicated in brain malformation and in neurodevelopmental disorders.
- Published
- 2021
5. A dyadic approach to the delineation of diagnostic entities in clinical genomics
- Author
-
Biesecker, Leslie G, Adam, Margaret P, Alkuraya, Fowzan S, Amemiya, Anne R, Bamshad, Michael J, Beck, Anita E, Bennett, James T, Bird, Lynne M, Carey, John C, Chung, Brian, Clark, Robin D, Cox, Timothy C, Curry, Cynthia, Dinulos, Mary Beth Palko, Dobyns, William B, Giampietro, Philip F, Girisha, Katta M, Glass, Ian A, Graham, John M, Gripp, Karen W, Haldeman-Englert, Chad R, Hall, Bryan D, Innes, A Micheil, Kalish, Jennifer M, Keppler-Noreuil, Kim M, Kosaki, Kenjiro, Kozel, Beth A, Mirzaa, Ghayda M, Mulvihill, John J, Nowaczyk, Malgorzata JM, Pagon, Roberta A, Retterer, Kyle, Rope, Alan F, Sanchez-Lara, Pedro A, Seaver, Laurie H, Shieh, Joseph T, Slavotinek, Anne M, Sobering, Andrew K, Stevens, Cathy A, Stevenson, David A, Tan, Tiong Yang, Tan, Wen-Hann, Tsai, Anne C, Weaver, David D, Williams, Marc S, Zackai, Elaine, and Zarate, Yuri A
- Subjects
Rare Diseases ,Genetics ,Cystic Fibrosis ,Cystic Fibrosis Transmembrane Conductance Regulator ,Genetic Diseases ,Inborn ,Genomics ,Genotype ,Humans ,Mutation ,Phenotype ,Biological Sciences ,Medical and Health Sciences ,Genetics & Heredity - Abstract
The delineation of disease entities is complex, yet recent advances in the molecular characterization of diseases provide opportunities to designate diseases in a biologically valid manner. Here, we have formalized an approach to the delineation of Mendelian genetic disorders that encompasses two distinct but inter-related concepts: (1) the gene that is mutated and (2) the phenotypic descriptor, preferably a recognizably distinct phenotype. We assert that only by a combinatorial or dyadic approach taking both of these attributes into account can a unitary, distinct genetic disorder be designated. We propose that all Mendelian disorders should be designated as "GENE-related phenotype descriptor" (e.g., "CFTR-related cystic fibrosis"). This approach to delineating and naming disorders reconciles the complexity of gene-to-phenotype relationships in a simple and clear manner yet communicates the complexity and nuance of these relationships.
- Published
- 2021
6. Reply to Hsueh YP et al.
- Author
-
Nambot, Sophie, Hevner, Robert F, and Dobyns, William B
- Subjects
Biological Sciences ,Biomedical and Clinical Sciences ,Clinical Sciences ,Genetics ,Autistic Disorder ,Humans ,Phenotype ,Genetics & Heredity ,Clinical sciences - Published
- 2020
7. De Novo Variants in WDR37 Are Associated with Epilepsy, Colobomas, Dysmorphism, Developmental Delay, Intellectual Disability, and Cerebellar Hypoplasia.
- Author
-
Kanca, Oguz, Andrews, Jonathan C, Lee, Pei-Tseng, Patel, Chirag, Braddock, Stephen R, Slavotinek, Anne M, Cohen, Julie S, Gubbels, Cynthia S, Aldinger, Kimberly A, Williams, Judy, Indaram, Maanasa, Fatemi, Ali, Yu, Timothy W, Agrawal, Pankaj B, Vezina, Gilbert, Simons, Cas, Crawford, Joanna, Lau, C Christopher, Undiagnosed Diseases Network, Chung, Wendy K, Markello, Thomas C, Dobyns, William B, Adams, David R, Gahl, William A, Wangler, Michael F, Yamamoto, Shinya, Bellen, Hugo J, and Malicdan, May Christine V
- Subjects
Undiagnosed Diseases Network ,Cerebellum ,Animals ,Humans ,Drosophila melanogaster ,Epilepsy ,Nervous System Malformations ,Coloboma ,Microfilament Proteins ,Developmental Disabilities ,Amino Acid Sequence ,Sequence Homology ,Phenotype ,Mutation ,Adult ,Child ,Infant ,Infant ,Newborn ,Female ,Male ,Young Adult ,Body Dysmorphic Disorders ,Intellectual Disability ,WD40 Repeats ,CG12333 ,Drosophila ,WD40 repeats ,WDR37 domains ,bang sensitivity ,wdr37 ,Genetics ,Pediatric ,Rare Diseases ,Congenital Structural Anomalies ,Neurodegenerative ,Neurosciences ,Brain Disorders ,Intellectual and Developmental Disabilities (IDD) ,Aetiology ,2.1 Biological and endogenous factors ,Neurological ,Biological Sciences ,Medical and Health Sciences ,Genetics & Heredity - Abstract
WD40 repeat-containing proteins form a large family of proteins present in all eukaryotes. Here, we identified five pediatric probands with de novo variants in WDR37, which encodes a member of the WD40 repeat protein family. Two probands shared one variant and the others have variants in nearby amino acids outside the WD40 repeats. The probands exhibited shared phenotypes of epilepsy, colobomas, facial dysmorphology reminiscent of CHARGE syndrome, developmental delay and intellectual disability, and cerebellar hypoplasia. The WDR37 protein is highly conserved in vertebrate and invertebrate model organisms and is currently not associated with a human disease. We generated a null allele of the single Drosophila ortholog to gain functional insights and replaced the coding region of the fly gene CG12333/wdr37 with GAL4. These flies are homozygous viable but display severe bang sensitivity, a phenotype associated with seizures in flies. Additionally, the mutant flies fall when climbing the walls of the vials, suggesting a defect in grip strength, and repeat the cycle of climbing and falling. Similar to wall clinging defect, mutant males often lose grip of the female abdomen during copulation. These phenotypes are rescued by using the GAL4 in the CG12333/wdr37 locus to drive the UAS-human reference WDR37 cDNA. The two variants found in three human subjects failed to rescue these phenotypes, suggesting that these alleles severely affect the function of this protein. Taken together, our data suggest that variants in WDR37 underlie a novel syndromic neurological disorder.
- Published
- 2019
8. SLC35A2‐CDG: Functional characterization, expanded molecular, clinical, and biochemical phenotypes of 30 unreported Individuals
- Author
-
Ng, Bobby G, Sosicka, Paulina, Agadi, Satish, Almannai, Mohammed, Bacino, Carlos A, Barone, Rita, Botto, Lorenzo D, Burton, Jennifer E, Carlston, Colleen, Chung, Brian Hon‐Yin, Cohen, Julie S, Coman, David, Dipple, Katrina M, Dorrani, Naghmeh, Dobyns, William B, Elias, Abdallah F, Epstein, Leon, Gahl, William A, Garozzo, Domenico, Hammer, Trine Bjørg, Haven, Jaclyn, Héron, Delphine, Herzog, Matthew, Hoganson, George E, Hunter, Jesse M, Jain, Mahim, Juusola, Jane, Lakhani, Shenela, Lee, Hane, Lee, Joy, Lewis, Katherine, Longo, Nicola, Lourenço, Charles Marques, Mak, Christopher CY, McKnight, Dianalee, Mendelsohn, Bryce A, Mignot, Cyril, Mirzaa, Ghayda, Mitchell, Wendy, Muhle, Hiltrud, Nelson, Stanley F, Olczak, Mariusz, Palmer, Christina GS, Partikian, Arthur, Patterson, Marc C, Pierson, Tyler M, Quinonez, Shane C, Regan, Brigid M, Ross, M Elizabeth, Sacoto, Maria J Guillen, Scaglia, Fernando, Scheffer, Ingrid E, Segal, Devorah, Singhal, Nilika Shah, Striano, Pasquale, Sturiale, Luisa, Symonds, Joseph D, Tang, Sha, Vilain, Eric, Willis, Mary, Wolfe, Lynne A, Yang, Hui, Yano, Shoji, Powis, Zöe, Suchy, Sharon F, Rosenfeld, Jill A, Edmondson, Andrew C, Grunewald, Stephanie, and Freeze, Hudson H
- Subjects
Clinical Research ,Pediatric ,Genetics ,Aetiology ,2.1 Biological and endogenous factors ,Animals ,Biopsy ,CHO Cells ,Cells ,Cultured ,Congenital Disorders of Glycosylation ,Cricetulus ,Female ,Humans ,Male ,Monosaccharide Transport Proteins ,Mutation ,Uridine Diphosphate Galactose ,congenital disorders of glycosylation ,glycoside ,nucleotide sugar transporter ,UDP-galactose ,Clinical Sciences ,Genetics & Heredity - Abstract
Pathogenic de novo variants in the X-linked gene SLC35A2 encoding the major Golgi-localized UDP-galactose transporter required for proper protein and lipid glycosylation cause a rare type of congenital disorder of glycosylation known as SLC35A2-congenital disorders of glycosylation (CDG; formerly CDG-IIm). To date, 29 unique de novo variants from 32 unrelated individuals have been described in the literature. The majority of affected individuals are primarily characterized by varying degrees of neurological impairments with or without skeletal abnormalities. Surprisingly, most affected individuals do not show abnormalities in serum transferrin N-glycosylation, a common biomarker for most types of CDG. Here we present data characterizing 30 individuals and add 26 new variants, the single largest study involving SLC35A2-CDG. The great majority of these individuals had normal transferrin glycosylation. In addition, expanding the molecular and clinical spectrum of this rare disorder, we developed a robust and reliable biochemical assay to assess SLC35A2-dependent UDP-galactose transport activity in primary fibroblasts. Finally, we show that transport activity is directly correlated to the ratio of wild-type to mutant alleles in fibroblasts from affected individuals.
- Published
- 2019
9. Mutations in MAST1 Cause Mega-Corpus-Callosum Syndrome with Cerebellar Hypoplasia and Cortical Malformations
- Author
-
Tripathy, Ratna, Leca, Ines, van Dijk, Tessa, Weiss, Janneke, van Bon, Bregje W, Sergaki, Maria Christina, Gstrein, Thomas, Breuss, Martin, Tian, Guoling, Bahi-Buisson, Nadia, Paciorkowski, Alexander R, Pagnamenta, Alistair T, Wenninger-Weinzierl, Andrea, Martinez-Reza, Maria Fernanda, Landler, Lukas, Lise, Stefano, Taylor, Jenny C, Terrone, Gaetano, Vitiello, Giuseppina, Del Giudice, Ennio, Brunetti-Pierri, Nicola, D’Amico, Alessandra, Reymond, Alexandre, Voisin, Norine, Bernstein, Jonathan A, Farrelly, Ellyn, Kini, Usha, Leonard, Thomas A, Valence, Stéphanie, Burglen, Lydie, Armstrong, Linlea, Hiatt, Susan M, Cooper, Gregory M, Aldinger, Kimberly A, Dobyns, William B, Mirzaa, Ghayda, Pierson, Tyler Mark, Baas, Frank, Chelly, Jamel, Cowan, Nicholas J, and Keays, David Anthony
- Subjects
Biomedical and Clinical Sciences ,Neurosciences ,Rare Diseases ,Pediatric ,Genetics ,Brain Disorders ,Intellectual and Developmental Disabilities (IDD) ,Congenital Structural Anomalies ,2.1 Biological and endogenous factors ,Aetiology ,Neurological ,Agenesis of Corpus Callosum ,Animals ,Animals ,Newborn ,Apoptosis ,Brain ,Cells ,Cultured ,Cerebellum ,Child ,Developmental Disabilities ,Disease Models ,Animal ,Embryo ,Mammalian ,Female ,Gene Expression Regulation ,Developmental ,Humans ,Male ,Malformations of Cortical Development ,Mice ,Mice ,Inbred C57BL ,Mice ,Knockout ,Microtubule-Associated Proteins ,Mutation ,Nerve Tissue Proteins ,Nervous System Malformations ,PAX6 Transcription Factor ,MAST1 ,cerebellar hypoplasia ,corpus callosum ,microdeletion ,microtubules ,Psychology ,Cognitive Sciences ,Neurology & Neurosurgery ,Biological psychology - Abstract
Corpus callosum malformations are associated with a broad range of neurodevelopmental diseases. We report that de novo mutations in MAST1 cause mega-corpus-callosum syndrome with cerebellar hypoplasia and cortical malformations (MCC-CH-CM) in the absence of megalencephaly. We show that MAST1 is a microtubule-associated protein that is predominantly expressed in post-mitotic neurons and is present in both dendritic and axonal compartments. We further show that Mast1 null animals are phenotypically normal, whereas the deletion of a single amino acid (L278del) recapitulates the distinct neurological phenotype observed in patients. In animals harboring Mast1 microdeletions, we find that the PI3K/AKT3/mTOR pathway is unperturbed, whereas Mast2 and Mast3 levels are diminished, indicative of a dominant-negative mode of action. Finally, we report that de novo MAST1 substitutions are present in patients with autism and microcephaly, raising the prospect that mutations in this gene give rise to a spectrum of neurodevelopmental diseases.
- Published
- 2018
10. NFIB Haploinsufficiency Is Associated with Intellectual Disability and Macrocephaly
- Author
-
Schanze, Ina, Bunt, Jens, Lim, Jonathan WC, Schanze, Denny, Dean, Ryan J, Alders, Marielle, Blanchet, Patricia, Attié-Bitach, Tania, Berland, Siren, Boogert, Steven, Boppudi, Sangamitra, Bridges, Caitlin J, Cho, Megan T, Dobyns, William B, Donnai, Dian, Douglas, Jessica, Earl, Dawn L, Edwards, Timothy J, Faivre, Laurence, Fregeau, Brieana, Genevieve, David, Gérard, Marion, Gatinois, Vincent, Holder-Espinasse, Muriel, Huth, Samuel F, Izumi, Kosuke, Kerr, Bronwyn, Lacaze, Elodie, Lakeman, Phillis, Mahida, Sonal, Mirzaa, Ghayda M, Morgan, Sian M, Nowak, Catherine, Peeters, Hilde, Petit, Florence, Pilz, Daniela T, Puechberty, Jacques, Reinstein, Eyal, Rivière, Jean-Baptiste, Santani, Avni B, Schneider, Anouck, Sherr, Elliott H, Smith-Hicks, Constance, Wieland, Ilse, Zackai, Elaine, Zhao, Xiaonan, Gronostajski, Richard M, Zenker, Martin, and Richards, Linda J
- Subjects
Biomedical and Clinical Sciences ,Mental Health ,Pediatric ,Intellectual and Developmental Disabilities (IDD) ,Neurosciences ,Rare Diseases ,Behavioral and Social Science ,Genetics ,Brain Disorders ,Aetiology ,2.1 Biological and endogenous factors ,Mental health ,Neurological ,Adolescent ,Adult ,Animals ,Cerebral Cortex ,Child ,Child ,Preschool ,Codon ,Nonsense ,Cohort Studies ,Corpus Callosum ,Female ,Haploinsufficiency ,Humans ,Intellectual Disability ,Male ,Megalencephaly ,Mice ,Mice ,Knockout ,NFI Transcription Factors ,Polymorphism ,Single Nucleotide ,Young Adult ,NFIB ,agenesis of the corpus callosum ,chromosome 9p22.3 ,chromosome 9p23 ,developmental delay ,haploinsufficiency ,intellectual disability ,macrocephaly ,megalencephaly ,nuclear factor I ,Biological Sciences ,Medical and Health Sciences ,Genetics & Heredity ,Biological sciences ,Biomedical and clinical sciences ,Health sciences - Abstract
The nuclear factor I (NFI) family of transcription factors play an important role in normal development of multiple organs. Three NFI family members are highly expressed in the brain, and deletions or sequence variants in two of these, NFIA and NFIX, have been associated with intellectual disability (ID) and brain malformations. NFIB, however, has not previously been implicated in human disease. Here, we present a cohort of 18 individuals with mild ID and behavioral issues who are haploinsufficient for NFIB. Ten individuals harbored overlapping microdeletions of the chromosomal 9p23-p22.2 region, ranging in size from 225 kb to 4.3 Mb. Five additional subjects had point sequence variations creating a premature termination codon, and three subjects harbored single-nucleotide variations resulting in an inactive protein as determined using an in vitro reporter assay. All individuals presented with additional variable neurodevelopmental phenotypes, including muscular hypotonia, motor and speech delay, attention deficit disorder, autism spectrum disorder, and behavioral abnormalities. While structural brain anomalies, including dysgenesis of corpus callosum, were variable, individuals most frequently presented with macrocephaly. To determine whether macrocephaly could be a functional consequence of NFIB disruption, we analyzed a cortex-specific Nfib conditional knockout mouse model, which is postnatally viable. Utilizing magnetic resonance imaging and histology, we demonstrate that Nfib conditional knockout mice have enlargement of the cerebral cortex but preservation of overall brain structure and interhemispheric connectivity. Based on our findings, we propose that haploinsufficiency of NFIB causes ID with macrocephaly.
- Published
- 2018
11. Primary brain calcification: an international study reporting novel variants and associated phenotypes.
- Author
-
Ramos, Eliana Marisa, Carecchio, Miryam, Lemos, Roberta, Ferreira, Joana, Legati, Andrea, Sears, Renee Louise, Hsu, Sandy Chan, Panteghini, Celeste, Magistrelli, Luca, Salsano, Ettore, Esposito, Silvia, Taroni, Franco, Richard, Anne-Claire, Tranchant, Christine, Anheim, Mathieu, Ayrignac, Xavier, Goizet, Cyril, Vidailhet, Marie, Maltete, David, Wallon, David, Frebourg, Thierry, Pimentel, Lylyan, Geschwind, Daniel H, Vanakker, Olivier, Galasko, Douglas, Fogel, Brent L, Innes, A Micheil, Ross, Alison, Dobyns, William B, Alcantara, Diana, O'Driscoll, Mark, Hannequin, Didier, Campion, Dominique, French PFBC study group, Oliveira, João R, Garavaglia, Barbara, Coppola, Giovanni, and Nicolas, Gaël
- Subjects
French PFBC study group ,Humans ,Brain Diseases ,Calcinosis ,Receptor ,Platelet-Derived Growth Factor beta ,Proto-Oncogene Proteins c-sis ,Receptors ,G-Protein-Coupled ,Receptors ,Virus ,Pedigree ,Heterozygote ,Phenotype ,Mutation ,Adolescent ,Adult ,Aged ,Aged ,80 and over ,Middle Aged ,Child ,Female ,Male ,Sodium-Phosphate Cotransporter Proteins ,Type III ,Genetic Variation ,Young Adult ,Cognitive Dysfunction ,Receptor ,Platelet-Derived Growth Factor beta ,Receptors ,G-Protein-Coupled ,Virus ,and over ,Sodium-Phosphate Cotransporter Proteins ,Type III ,Genetics ,Genetics & Heredity ,Clinical Sciences - Abstract
Primary familial brain calcification (PFBC) is a rare cerebral microvascular calcifying disorder with a wide spectrum of motor, cognitive, and neuropsychiatric symptoms. It is typically inherited as an autosomal-dominant trait with four causative genes identified so far: SLC20A2, PDGFRB, PDGFB, and XPR1. Our study aimed at screening the coding regions of these genes in a series of 177 unrelated probands that fulfilled the diagnostic criteria for primary brain calcification regardless of their family history. Sequence variants were classified as pathogenic, likely pathogenic, or of uncertain significance (VUS), based on the ACMG-AMP recommendations. We identified 45 probands (25.4%) carrying either pathogenic or likely pathogenic variants (n = 34, 19.2%) or VUS (n = 11, 6.2%). SLC20A2 provided the highest contribution (16.9%), followed by XPR1 and PDGFB (3.4% each), and PDGFRB (1.7%). A total of 81.5% of carriers were symptomatic and the most recurrent symptoms were parkinsonism, cognitive impairment, and psychiatric disturbances (52.3%, 40.9%, and 38.6% of symptomatic individuals, respectively), with a wide range of age at onset (from childhood to 81 years). While the pathogenic and likely pathogenic variants identified in this study can be used for genetic counseling, the VUS will require additional evidence, such as recurrence in unrelated patients, in order to be classified as pathogenic.
- Published
- 2018
12. Structural malformations of the brain, eye, and pituitary gland in PHACE syndrome
- Author
-
Steiner, Jack E, McCoy, Garrett N, Hess, Christopher P, Dobyns, William B, Metry, Denise W, Drolet, Beth A, Maheshwari, Mohit, and Siegel, Dawn H
- Subjects
Biomedical and Clinical Sciences ,Ophthalmology and Optometry ,Brain Disorders ,Congenital Structural Anomalies ,Orphan Drug ,Intellectual and Developmental Disabilities (IDD) ,Biomedical Imaging ,Neurosciences ,Clinical Research ,Pediatric ,Rare Diseases ,4.1 Discovery and preclinical testing of markers and technologies ,Detection ,screening and diagnosis ,Congenital ,Neurological ,Abnormalities ,Multiple ,Aortic Coarctation ,Brain ,Eye Abnormalities ,Female ,Humans ,Infant ,Infant ,Newborn ,Magnetic Resonance Imaging ,Male ,Neurocutaneous Syndromes ,Phenotype ,Pituitary Gland ,brain malformations ,eye abnormalities ,magnetic resonance imaging ,PHACE syndrome ,pituitary gland ,Genetics ,Clinical Sciences ,Clinical sciences - Abstract
PHACE syndrome is the association of segmental facial hemangiomas with congenital arterial, brain, cardiac, and ocular anomalies. Structural brain malformations affect 41-52% of PHACE patients and can be associated with focal neurologic deficits, developmental delays, and/or intellectual disability. To better characterize the spectrum of structural brain and other intracranial anomalies in PHACE syndrome, MRI scans of the head/neck were retrospectively reviewed in 55 patients from the PHACE Syndrome International Clinical Registry and Genetic Repository. All registry patients with a diagnosis of definite PHACE syndrome who had MRI scans of satisfactory quality were included. Of 55 patients, 34 (62%) demonstrated ≥1 non-vascular intracranial anomaly; structural brain malformations were present in 19 (35%). There was no difference in the prevalence of brain anomalies between genders. Brain anomalies were more likely in patients with S1 and/or S2 distribution of facial hemangioma. The most common structural brain defects were cerebellar hypoplasia (25%) and fourth ventricle abnormalities (13%). Dandy-Walker complex and malformations of cortical development were present in 9% and 7%, respectively. Extra-axial findings such as pituitary anomalies (18%) and intracranial hemangiomas (18%) were also observed. Six patients (11%) had anomalies of the globes or optic nerve/chiasm detectable on MRI. Brain malformations comprise a diverse group of structural developmental anomalies that are common in patients with PHACE syndrome. Along with brain malformations, numerous abnormalities of the pituitary, meninges, and globes were observed, highlighting the need for careful radiologic assessment of these structures in the neuroimaging workup for PHACE syndrome.
- Published
- 2018
13. Biallelic mutations in the 3′ exonuclease TOE1 cause pontocerebellar hypoplasia and uncover a role in snRNA processing
- Author
-
Lardelli, Rea M, Schaffer, Ashleigh E, Eggens, Veerle RC, Zaki, Maha S, Grainger, Stephanie, Sathe, Shashank, Van Nostrand, Eric L, Schlachetzki, Zinayida, Rosti, Basak, Akizu, Naiara, Scott, Eric, Silhavy, Jennifer L, Heckman, Laura Dean, Rosti, Rasim Ozgur, Dikoglu, Esra, Gregor, Anne, Guemez-Gamboa, Alicia, Musaev, Damir, Mande, Rohit, Widjaja, Ari, Shaw, Tim L, Markmiller, Sebastian, Marin-Valencia, Isaac, Davies, Justin H, de Meirleir, Linda, Kayserili, Hulya, Altunoglu, Umut, Freckmann, Mary Louise, Warwick, Linda, Chitayat, David, Blaser, Susan, Çağlayan, Ahmet Okay, Bilguvar, Kaya, Per, Huseyin, Fagerberg, Christina, Christesen, Henrik T, Kibaek, Maria, Aldinger, Kimberly A, Manchester, David, Matsumoto, Naomichi, Muramatsu, Kazuhiro, Saitsu, Hirotomo, Shiina, Masaaki, Ogata, Kazuhiro, Foulds, Nicola, Dobyns, William B, Chi, Neil C, Traver, David, Spaccini, Luigina, Bova, Stefania Maria, Gabriel, Stacey B, Gunel, Murat, Valente, Enza Maria, Nassogne, Marie-Cecile, Bennett, Eric J, Yeo, Gene W, Baas, Frank, Lykke-Andersen, Jens, and Gleeson, Joseph G
- Subjects
Biological Sciences ,Bioinformatics and Computational Biology ,Genetics ,Alleles ,Animals ,Cerebellar Diseases ,Exonucleases ,Female ,Humans ,Male ,Mice ,Mutation ,Neurodegenerative Diseases ,Nuclear Proteins ,RNA ,Messenger ,RNA ,Small Nuclear ,Spliceosomes ,Zebrafish ,Medical and Health Sciences ,Developmental Biology ,Agricultural biotechnology ,Bioinformatics and computational biology - Abstract
Deadenylases are best known for degrading the poly(A) tail during mRNA decay. The deadenylase family has expanded throughout evolution and, in mammals, consists of 12 Mg2+-dependent 3'-end RNases with substrate specificity that is mostly unknown. Pontocerebellar hypoplasia type 7 (PCH7) is a unique recessive syndrome characterized by neurodegeneration and ambiguous genitalia. We studied 12 human families with PCH7, uncovering biallelic, loss-of-function mutations in TOE1, which encodes an unconventional deadenylase. toe1-morphant zebrafish displayed midbrain and hindbrain degeneration, modeling PCH-like structural defects in vivo. Surprisingly, we found that TOE1 associated with small nuclear RNAs (snRNAs) incompletely processed spliceosomal. These pre-snRNAs contained 3' genome-encoded tails often followed by post-transcriptionally added adenosines. Human cells with reduced levels of TOE1 accumulated 3'-end-extended pre-snRNAs, and the immunoisolated TOE1 complex was sufficient for 3'-end maturation of snRNAs. Our findings identify the cause of a neurodegenerative syndrome linked to snRNA maturation and uncover a key factor involved in the processing of snRNA 3' ends.
- Published
- 2017
14. Astroglial-Mediated Remodeling of the Interhemispheric Midline Is Required for the Formation of the Corpus Callosum.
- Author
-
Gobius, Ilan, Morcom, Laura, Suárez, Rodrigo, Bunt, Jens, Bukshpun, Polina, Reardon, William, Dobyns, William B, Rubenstein, John LR, Barkovich, A James, Sherr, Elliott H, and Richards, Linda J
- Subjects
Corpus Callosum ,Astrocytes ,Axons ,Animals ,Humans ,Mice ,Transcription Factors ,Signal Transduction ,Cell Differentiation ,Organogenesis ,Phenotype ,Fibroblast Growth Factor 8 ,Cerebrum ,Agenesis of Corpus Callosum ,Fgf8 ,Nfia ,Nfib ,astrocyte ,callosal agenesis ,interhemispheric fissure ,Genetics ,Neurosciences ,Underpinning research ,1.1 Normal biological development and functioning ,Neurological ,Biochemistry and Cell Biology ,Medical Physiology - Abstract
The corpus callosum is the major axon tract that connects and integrates neural activity between the two cerebral hemispheres. Although ∼1:4,000 children are born with developmental absence of the corpus callosum, the primary etiology of this condition remains unknown. Here, we demonstrate that midline crossing of callosal axons is dependent upon the prior remodeling and degradation of the intervening interhemispheric fissure. This remodeling event is initiated by astroglia on either side of the interhemispheric fissure, which intercalate with one another and degrade the intervening leptomeninges. Callosal axons then preferentially extend over these specialized astroglial cells to cross the midline. A key regulatory step in interhemispheric remodeling is the differentiation of these astroglia from radial glia, which is initiated by Fgf8 signaling to downstream Nfi transcription factors. Crucially, our findings from human neuroimaging studies reveal that developmental defects in interhemispheric remodeling are likely to be a primary etiology underlying human callosal agenesis.
- Published
- 2016
15. A Recurrent Mosaic Mutation in SMO, Encoding the Hedgehog Signal Transducer Smoothened, Is the Major Cause of Curry-Jones Syndrome
- Author
-
Twigg, Stephen RF, Hufnagel, Robert B, Miller, Kerry A, Zhou, Yan, McGowan, Simon J, Taylor, John, Craft, Jude, Taylor, Jenny C, Santoro, Stephanie L, Huang, Taosheng, Hopkin, Robert J, Brady, Angela F, Clayton-Smith, Jill, Clericuzio, Carol L, Grange, Dorothy K, Groesser, Leopold, Hafner, Christian, Horn, Denise, Temple, I Karen, Dobyns, William B, Curry, Cynthia J, Jones, Marilyn C, and Wilkie, Andrew OM
- Subjects
Biological Sciences ,Biomedical and Clinical Sciences ,Clinical Sciences ,Brain Disorders ,Rare Diseases ,Brain Cancer ,Clinical Research ,Neurosciences ,Cancer ,Pediatric ,Genetics ,Child ,Preschool ,Craniofacial Abnormalities ,Female ,Humans ,Infant ,Infant ,Newborn ,Intestines ,Male ,Mutation ,Signal Transduction ,Skin Abnormalities ,Smoothened Receptor ,Syndactyly ,Medical and Health Sciences ,Genetics & Heredity ,Biological sciences ,Biomedical and clinical sciences ,Health sciences - Abstract
Curry-Jones syndrome (CJS) is a multisystem disorder characterized by patchy skin lesions, polysyndactyly, diverse cerebral malformations, unicoronal craniosynostosis, iris colobomas, microphthalmia, and intestinal malrotation with myofibromas or hamartomas. Cerebellar medulloblastoma has been described in a single affected individual; in another, biopsy of skin lesions showed features of trichoblastoma. The combination of asymmetric clinical features, patchy skin manifestations, and neoplastic association previously led to the suggestion that this could be a mosaic condition, possibly involving hedgehog (Hh) signaling. Here, we show that CJS is caused by recurrent somatic mosaicism for a nonsynonymous variant in SMO (c.1234C>T [p.Leu412Phe]), encoding smoothened (SMO), a G-protein-coupled receptor that transduces Hh signaling. We identified eight mutation-positive individuals (two of whom had not been reported previously) with highly similar phenotypes and demonstrated varying amounts of the mutant allele in different tissues. We present detailed findings from brain MRI in three mutation-positive individuals. Somatic SMO mutations that result in constitutive activation have been described in several tumors, including medulloblastoma, ameloblastoma, and basal cell carcinoma. Strikingly, the most common of these mutations is the identical nonsynonymous variant encoding p.Leu412Phe. Furthermore, this substitution has been shown to activate SMO in the absence of Hh signaling, providing an explanation for tumor development in CJS. This raises therapeutic possibilities for using recently generated Hh-pathway inhibitors. In summary, our work uncovers the major genetic cause of CJS and illustrates strategies for gene discovery in the context of low-level tissue-specific somatic mosaicism.
- Published
- 2016
16. Mandibulofacial Dysostosis with Microcephaly: Mutation and Database Update
- Author
-
Huang, Lijia, Vanstone, Megan R, Hartley, Taila, Osmond, Matthew, Barrowman, Nick, Allanson, Judith, Baker, Laura, Dabir, Tabib A, Dipple, Katrina M, Dobyns, William B, Estrella, Jane, Faghfoury, Hanna, Favaro, Francine P, Goel, Himanshu, Gregersen, Pernille A, Gripp, Karen W, Grix, Art, Guion‐Almeida, Maria‐Leine, Harr, Margaret H, Hudson, Cindy, Hunter, Alasdair GW, Johnson, John, Joss, Shelagh K, Kimball, Amy, Kini, Usha, Kline, Antonie D, Lauzon, Julie, Lildballe, Dorte L, López‐González, Vanesa, Martinezmoles, Johanna, Meldrum, Cliff, Mirzaa, Ghayda M, Morel, Chantal F, Morton, Jenny EV, Pyle, Louise C, Quintero‐Rivera, Fabiola, Richer, Julie, Scheuerle, Angela E, Schönewolf‐Greulich, Bitten, Shears, Deborah J, Silver, Josh, Smith, Amanda C, Temple, I Karen, Center, UCLA Clinical Genomics, de Kamp, Jiddeke M, Dijk, Fleur S, Vandersteen, Anthony M, White, Sue M, Zackai, Elaine H, Zou, Ruobing, Consortium, Care4Rare Canada, Bulman, Dennis E, Boycott, Kym M, and Lines, Matthew A
- Subjects
Pediatric ,Congenital Structural Anomalies ,Rare Diseases ,Genetics ,Prevention ,Human Genome ,Neurosciences ,Clinical Research ,Dental/Oral and Craniofacial Disease ,2.1 Biological and endogenous factors ,Aetiology ,Congenital ,Abnormalities ,Multiple ,Amino Acid Motifs ,Databases ,Genetic ,Gene Expression ,Haploinsufficiency ,Hearing Loss ,Humans ,Intellectual Disability ,Mandibulofacial Dysostosis ,Microcephaly ,Models ,Molecular ,Molecular Sequence Data ,Mutation ,Penetrance ,Peptide Elongation Factors ,Phenotype ,Protein Structure ,Secondary ,Protein Structure ,Tertiary ,RNA Splicing ,Ribonucleoprotein ,U5 Small Nuclear ,Spliceosomes ,EFTUD2 ,mandibulofacial dysostosis with microcephaly ,MFDM ,mandibulofacial dysostosis Guion-Almeida type ,mandibulofacial dysostosis ,microcephaly ,UCLA Clinical Genomics Center ,Care4Rare Canada Consortium ,Clinical Sciences ,Genetics & Heredity - Abstract
Mandibulofacial dysostosis with microcephaly (MFDM) is a multiple malformation syndrome comprising microcephaly, craniofacial anomalies, hearing loss, dysmorphic features, and, in some cases, esophageal atresia. Haploinsufficiency of a spliceosomal GTPase, U5-116 kDa/EFTUD2, is responsible. Here, we review the molecular basis of MFDM in the 69 individuals described to date, and report mutations in 38 new individuals, bringing the total number of reported individuals to 107 individuals from 94 kindreds. Pathogenic EFTUD2 variants comprise 76 distinct mutations and seven microdeletions. Among point mutations, missense substitutions are infrequent (14 out of 76; 18%) relative to stop-gain (29 out of 76; 38%), and splicing (33 out of 76; 43%) mutations. Where known, mutation origin was de novo in 48 out of 64 individuals (75%), dominantly inherited in 12 out of 64 (19%), and due to proven germline mosaicism in four out of 64 (6%). Highly penetrant clinical features include, microcephaly, first and second arch craniofacial malformations, and hearing loss; esophageal atresia is present in an estimated ∼27%. Microcephaly is virtually universal in childhood, with some adults exhibiting late "catch-up" growth and normocephaly at maturity. Occasionally reported anomalies, include vestibular and ossicular malformations, reduced mouth opening, atrophy of cerebral white matter, structural brain malformations, and epibulbar dermoid. All reported EFTUD2 mutations can be found in the EFTUD2 mutation database (http://databases.lovd.nl/shared/genes/EFTUD2).
- Published
- 2016
17. Malformations of cortical development and epilepsy.
- Author
-
Barkovich, A James, Dobyns, William B, and Guerrini, Renzo
- Subjects
Medical Biochemistry and Metabolomics ,Medical Microbiology ,Medical Physiology ,Biomedical and Clinical Sciences ,Neurosciences ,Genetics ,Neurodegenerative ,Brain Disorders ,Epilepsy ,Aetiology ,2.1 Biological and endogenous factors ,Detection ,screening and diagnosis ,4.1 Discovery and preclinical testing of markers and technologies ,Neurological ,Brain ,Diagnostic Imaging ,Humans ,Malformations of Cortical Development ,Medical biochemistry and metabolomics ,Medical microbiology ,Medical physiology - Abstract
Malformations of cortical development (MCDs) are an important cause of epilepsy and an extremely interesting group of disorders from the perspective of brain development and its perturbations. Many new MCDs have been described in recent years as a result of improvements in imaging, genetic testing, and understanding of the effects of mutations on the ability of their protein products to correctly function within the molecular pathways by which the brain functions. In this review, most of the major MCDs are reviewed from a clinical, embryological, and genetic perspective. The most recent literature regarding clinical diagnosis, mechanisms of development, and future paths of research are discussed.
- Published
- 2015
18. Mutations in CENPE define a novel kinetochore-centromeric mechanism for microcephalic primordial dwarfism
- Author
-
Mirzaa, Ghayda M, Vitre, Benjamin, Carpenter, Gillian, Abramowicz, Iga, Gleeson, Joseph G, Paciorkowski, Alex R, Cleveland, Don W, Dobyns, William B, and O’Driscoll, Mark
- Subjects
Genetics ,Underpinning research ,1.1 Normal biological development and functioning ,Adult ,Amino Acid Sequence ,Cell Cycle ,Cells ,Cultured ,Centromere ,Centrosome ,Child ,Child ,Preschool ,Chromosomal Proteins ,Non-Histone ,Chromosome Segregation ,Developmental Disabilities ,Dwarfism ,Female ,Fetal Growth Retardation ,Humans ,Kinetochores ,Lymphocytes ,Male ,Microcephaly ,Mitosis ,Molecular Sequence Data ,Mutation ,Osteochondrodysplasias ,Phosphorylation ,Protein Serine-Threonine Kinases ,Sequence Homology ,Amino Acid ,Complementary and Alternative Medicine ,Paediatrics and Reproductive Medicine ,Genetics & Heredity - Abstract
Defects in centrosome, centrosomal-associated and spindle-associated proteins are the most frequent cause of primary microcephaly (PM) and microcephalic primordial dwarfism (MPD) syndromes in humans. Mitotic progression and segregation defects, microtubule spindle abnormalities and impaired DNA damage-induced G2-M cell cycle checkpoint proficiency have been documented in cell lines from these patients. This suggests that impaired mitotic entry, progression and exit strongly contribute to PM and MPD. Considering the vast protein networks involved in coordinating this cell cycle stage, the list of potential target genes that could underlie novel developmental disorders is large. One such complex network, with a direct microtubule-mediated physical connection to the centrosome, is the kinetochore. This centromeric-associated structure nucleates microtubule attachments onto mitotic chromosomes. Here, we described novel compound heterozygous variants in CENPE in two siblings who exhibit a profound MPD associated with developmental delay, simplified gyri and other isolated abnormalities. CENPE encodes centromere-associated protein E (CENP-E), a core kinetochore component functioning to mediate chromosome congression initially of misaligned chromosomes and in subsequent spindle microtubule capture during mitosis. Firstly, we present a comprehensive clinical description of these patients. Then, using patient cells we document abnormalities in spindle microtubule organization, mitotic progression and segregation, before modeling the cellular pathogenicity of these variants in an independent cell system. Our cellular analysis shows that a pathogenic defect in CENP-E, a kinetochore-core protein, largely phenocopies PCNT-mutated microcephalic osteodysplastic primordial dwarfism-type II patient cells. PCNT encodes a centrosome-associated protein. These results highlight a common underlying pathomechanism. Our findings provide the first evidence for a kinetochore-based route to MPD in humans.
- Published
- 2014
19. The Developmental Brain Disorders Database (DBDB): A curated neurogenetics knowledge base with clinical and research applications
- Author
-
Mirzaa, Ghayda M, Millen, Kathleen J, Barkovich, A James, Dobyns, William B, and Paciorkowski, Alex R
- Subjects
Brain Disorders ,Genetics ,Genetic Testing ,Neurosciences ,Neurological ,Brain ,Brain Diseases ,Child ,Databases ,Genetic ,Developmental Disabilities ,Humans ,Internet ,Knowledge Bases ,developmental brain disorders ,database ,bioinformatics ,levels of evidence ,Clinical Sciences - Abstract
The number of single genes associated with neurodevelopmental disorders has increased dramatically over the past decade. The identification of causative genes for these disorders is important to clinical outcome as it allows for accurate assessment of prognosis, genetic counseling, delineation of natural history, inclusion in clinical trials, and in some cases determines therapy. Clinicians face the challenge of correctly identifying neurodevelopmental phenotypes, recognizing syndromes, and prioritizing the best candidate genes for testing. However, there is no central repository of definitions for many phenotypes, leading to errors of diagnosis. Additionally, there is no system of levels of evidence linking genes to phenotypes, making it difficult for clinicians to know which genes are most strongly associated with a given condition. We have developed the Developmental Brain Disorders Database (DBDB: https://www.dbdb.urmc.rochester.edu/home), a publicly available, online-curated repository of genes, phenotypes, and syndromes associated with neurodevelopmental disorders. DBDB contains the first referenced ontology of developmental brain phenotypes, and uses a novel system of levels of evidence for gene-phenotype associations. It is intended to assist clinicians in arriving at the correct diagnosis, select the most appropriate genetic test for that phenotype, and improve the care of patients with developmental brain disorders. For researchers interested in the discovery of novel genes for developmental brain disorders, DBDB provides a well-curated source of important genes against which research sequencing results can be compared. Finally, DBDB allows novel observations about the landscape of the neurogenetics knowledge base.
- Published
- 2014
20. Cerebellar and posterior fossa malformations in patients with autism‐associated chromosome 22q13 terminal deletion
- Author
-
Aldinger, Kimberly A, Kogan, Jillene, Kimonis, Virginia, Fernandez, Bridget, Horn, Denise, Klopocki, Eva, Chung, Brian, Toutain, Annick, Weksberg, Rosanna, Millen, Kathleen J, Barkovich, A James, and Dobyns, William B
- Subjects
Biological Sciences ,Biomedical and Clinical Sciences ,Genetics ,Intellectual and Developmental Disabilities (IDD) ,Clinical Research ,Rare Diseases ,Pediatric ,Neurosciences ,Brain Disorders ,Congenital Structural Anomalies ,Pediatric Research Initiative ,Aetiology ,2.1 Biological and endogenous factors ,Mental health ,Adaptor Proteins ,Signal Transducing ,Adolescent ,Autistic Disorder ,Cerebellum ,Child ,Child ,Preschool ,Chromosome Deletion ,Chromosome Disorders ,Chromosome Mapping ,Chromosomes ,Human ,Pair 22 ,Developmental Disabilities ,Genetic Association Studies ,Genetic Loci ,Genotype ,Humans ,Image Processing ,Computer-Assisted ,In Situ Hybridization ,Fluorescence ,Infant ,Mutation ,Nerve Tissue Proteins ,Oligonucleotide Array Sequence Analysis ,Phenotype ,Polymorphism ,Single Nucleotide ,cerebellum ,chromosome ,deletion ,SHANK3 ,Clinical Sciences ,Clinical sciences - Abstract
The 22q13.3 deletion causes a neurodevelopmental syndrome, also known as Phelan-McDermid syndrome (MIM #606232), characterized by developmental delay and severe delay or absence of expressive speech. Two patients with hemizygous chromosome 22q13.3 telomeric deletion were referred to us when brain-imaging studies revealed cerebellar vermis hypoplasia (CBVH). To determine whether developmental abnormalities of the cerebellum are a consistent feature of the 22q13.3 deletion syndrome, we examined brain-imaging studies for 10 unrelated subjects with 22q13 terminal deletion. In seven cases where the availability of DNA and array technology allowed, we mapped deletion boundaries using comparative intensity analysis with single nucleotide polymorphism (SNP) microarrays. Approximate deletion boundaries for three additional cases were derived from clinical or published molecular data. We also examined brain-imaging studies for a patient with an intragenic SHANK3 mutation. We report the first brain-imaging data showing that some patients with 22q13 deletions have severe posterior CBVH, and one individual with a SHANK3 mutation has a normal cerebellum. This genotype-phenotype study suggests that the 22q13 deletion phenotype includes abnormal posterior fossa structures that are unlikely to be attributed to SHANK3 disruption. Other genes in the region, including PLXNB2 and MAPK8IP2, display brain expression patterns and mouse mutant phenotypes critical for proper cerebellar development. Future studies of these genes may elucidate their relationship to 22q13.3 deletion phenotypes.
- Published
- 2013
21. CHMP1A encodes an essential regulator of BMI1-INK4A in cerebellar development
- Author
-
Mochida, Ganeshwaran H, Ganesh, Vijay S, de Michelena, Maria I, Dias, Hugo, Atabay, Kutay D, Kathrein, Katie L, Huang, Hsuan-Ting, Hill, R Sean, Felie, Jillian M, Rakiec, Daniel, Gleason, Danielle, Hill, Anthony D, Malik, Athar N, Barry, Brenda J, Partlow, Jennifer N, Tan, Wen-Hann, Glader, Laurie J, Barkovich, A James, Dobyns, William B, Zon, Leonard I, and Walsh, Christopher A
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Regenerative Medicine ,Rare Diseases ,Pediatric ,Stem Cell Research - Induced Pluripotent Stem Cell ,Stem Cell Research - Nonembryonic - Human ,Congenital Structural Anomalies ,Stem Cell Research - Induced Pluripotent Stem Cell - Human ,Stem Cell Research ,Stem Cell Research - Nonembryonic - Non-Human ,Animals ,Cell Proliferation ,Cerebellar Cortex ,Cyclin-Dependent Kinase Inhibitor p16 ,Endosomal Sorting Complexes Required for Transport ,Gene Expression Regulation ,Developmental ,Genetic Linkage ,HEK293 Cells ,Humans ,Mice ,Microcephaly ,Mitogen-Activated Protein Kinase 7 ,Mutation ,NIH 3T3 Cells ,Neural Stem Cells ,Neurons ,Polymorphism ,Single Nucleotide ,Vesicular Transport Proteins ,Zebrafish ,Medical and Health Sciences ,Developmental Biology ,Agricultural biotechnology ,Bioinformatics and computational biology ,Genetics - Abstract
Charged multivesicular body protein 1A (CHMP1A; also known as chromatin-modifying protein 1A) is a member of the ESCRT-III (endosomal sorting complex required for transport-III) complex but is also suggested to localize to the nuclear matrix and regulate chromatin structure. Here, we show that loss-of-function mutations in human CHMP1A cause reduced cerebellar size (pontocerebellar hypoplasia) and reduced cerebral cortical size (microcephaly). CHMP1A-mutant cells show impaired proliferation, with increased expression of INK4A, a negative regulator of stem cell proliferation. Chromatin immunoprecipitation suggests loss of the normal INK4A repression by BMI in these cells. Morpholino-based knockdown of zebrafish chmp1a resulted in brain defects resembling those seen after bmi1a and bmi1b knockdown, which were partially rescued by INK4A ortholog knockdown, further supporting links between CHMP1A and BMI1-mediated regulation of INK4A. Our results suggest that CHMP1A serves as a critical link between cytoplasmic signals and BMI1-mediated chromatin modifications that regulate proliferation of central nervous system progenitor cells.
- Published
- 2012
22. COL4A1 mutations cause ocular dysgenesis, neuronal localization defects, and myopathy in mice and Walker-Warburg syndrome in humans.
- Author
-
Labelle-Dumais, Cassandre, Dilworth, David J, Harrington, Emily P, de Leau, Michelle, Lyons, David, Kabaeva, Zhyldyz, Manzini, M Chiara, Dobyns, William B, Walsh, Christopher A, Michele, Daniel E, and Gould, Douglas B
- Subjects
Eye ,Neurons ,Animals ,Humans ,Mice ,Muscular Diseases ,Collagen Type IV ,Sequence Alignment ,Apoptosis ,Base Sequence ,Mutation ,Walker-Warburg Syndrome ,Developmental Biology ,Genetics - Abstract
Muscle-eye-brain disease (MEB) and Walker Warburg Syndrome (WWS) belong to a spectrum of autosomal recessive diseases characterized by ocular dysgenesis, neuronal migration defects, and congenital muscular dystrophy. Until now, the pathophysiology of MEB/WWS has been attributed to alteration in dystroglycan post-translational modification. Here, we provide evidence that mutations in a gene coding for a major basement membrane protein, collagen IV alpha 1 (COL4A1), are a novel cause of MEB/WWS. Using a combination of histological, molecular, and biochemical approaches, we show that heterozygous Col4a1 mutant mice have ocular dysgenesis, neuronal localization defects, and myopathy characteristic of MEB/WWS. Importantly, we identified putative heterozygous mutations in COL4A1 in two MEB/WWS patients. Both mutations occur within conserved amino acids of the triple-helix-forming domain of the protein, and at least one mutation interferes with secretion of the mutant proteins, resulting instead in intracellular accumulation. Expression and posttranslational modification of dystroglycan is unaltered in Col4a1 mutant mice indicating that COL4A1 mutations represent a distinct pathogenic mechanism underlying MEB/WWS. These findings implicate a novel gene and a novel mechanism in the etiology of MEB/WWS and expand the clinical spectrum of COL4A1-associated disorders.
- Published
- 2011
23. Association and Mutation Analyses of 16p11.2 Autism Candidate Genes
- Author
-
Kumar, Ravinesh A, Marshall, Christian R, Badner, Judith A, Babatz, Timothy D, Mukamel, Zohar, Aldinger, Kimberly A, Sudi, Jyotsna, Brune, Camille W, Goh, Gerald, KaraMohamed, Samer, Sutcliffe, James S, Cook, Edwin H, Geschwind, Daniel H, Dobyns, William B, Scherer, Stephen W, and Christian, Susan L
- Subjects
Neurosciences ,Clinical Research ,Biotechnology ,Pediatric ,Human Genome ,Brain Disorders ,Intellectual and Developmental Disabilities (IDD) ,Genetics ,Autism ,Mental Health ,2.1 Biological and endogenous factors ,Aetiology ,Mental health ,Neurological ,Animals ,Autistic Disorder ,Chromosomes ,Human ,Pair 16 ,DNA Mutational Analysis ,Embryo ,Mammalian ,Exons ,Family Health ,Genetic Predisposition to Disease ,Genetic Variation ,Humans ,Membrane Proteins ,Mice ,Promoter Regions ,Genetic ,General Science & Technology - Abstract
BackgroundAutism is a complex childhood neurodevelopmental disorder with a strong genetic basis. Microdeletion or duplication of a approximately 500-700-kb genomic rearrangement on 16p11.2 that contains 24 genes represents the second most frequent chromosomal disorder associated with autism. The role of common and rare 16p11.2 sequence variants in autism etiology is unknown.Methodology/principal findingsTo identify common 16p11.2 variants with a potential role in autism, we performed association studies using existing data generated from three microarray platforms: Affymetrix 5.0 (777 families), Illumina 550 K (943 families), and Affymetrix 500 K (60 families). No common variants were identified that were significantly associated with autism. To look for rare variants, we performed resequencing of coding and promoter regions for eight candidate genes selected based on their known expression patterns and functions. In total, we identified 26 novel variants in autism: 13 exonic (nine non-synonymous, three synonymous, and one untranslated region) and 13 promoter variants. We found a significant association between autism and a coding variant in the seizure-related gene SEZ6L2 (12/1106 autism vs. 3/1161 controls; p = 0.018). Sez6l2 expression in mouse embryos was restricted to the spinal cord and brain. SEZ6L2 expression in human fetal brain was highest in post-mitotic cortical layers, hippocampus, amygdala, and thalamus. Association analysis of SEZ6L2 in an independent sample set failed to replicate our initial findings.Conclusions/significanceWe have identified sequence variation in at least one candidate gene in 16p11.2 that may represent a novel genetic risk factor for autism. However, further studies are required to substantiate these preliminary findings.
- Published
- 2009
24. Polymicrogyria and deletion 22q11.2 syndrome: Window to the etiology of a common cortical malformation
- Author
-
Robin, Nathaniel H, Taylor, Clare J, McDonald‐McGinn, Donna M, Zackai, Elaine H, Bingham, Peter, Collins, Kevin J, Earl, Dawn, Gill, Deepak, Granata, Tiziana, Guerrini, Renzo, Katz, Naomi, Kimonis, Virginia, Lin, Jean‐Pierre, Lynch, David R, Mohammed, Shehla N, Massey, Roger F, McDonald, Marie, Rogers, R Curtis, Splitt, Miranda, Stevens, Cathy A, Tischkowitz, Marc D, Stoodley, Neil, Leventer, Richard J, Pilz, Daniela T, and Dobyns, William B
- Subjects
Biomedical and Clinical Sciences ,Clinical Sciences ,Intellectual and Developmental Disabilities (IDD) ,Clinical Research ,Neurosciences ,Pediatric ,Rare Diseases ,Brain Disorders ,Congenital Structural Anomalies ,Aetiology ,2.1 Biological and endogenous factors ,Congenital ,Cerebellum ,Cerebral Cortex ,Chromosome Deletion ,Chromosomes ,Human ,Pair 22 ,Female ,Humans ,Magnetic Resonance Imaging ,Male ,Syndrome ,Tomography ,X-Ray Computed ,Velopharyngeal Insufficiency ,Genetics ,Clinical sciences - Abstract
Several brain malformations have been described in rare patients with the deletion 22q11.2 syndrome (DEL22q11) including agenesis of the corpus callosum, pachygyria or polymicrogyria (PMG), cerebellar anomalies and meningomyelocele, with PMG reported most frequently. In view of our interest in the causes of PMG, we reviewed clinical data including brain-imaging studies on 21 patients with PMG associated with deletion 22q11.2 and another 11 from the literature. We found that the cortical malformation consists of perisylvian PMG of variable severity and frequent asymmetry with a striking predisposition for the right hemisphere (P = 0.008). This and other observations suggest that the PMG may be a sequela of abnormal embryonic vascular development rather than a primary brain malformation. We also noted mild cerebellar hypoplasia or mega-cisterna magna in 8 of 24 patients. Although this was not the focus of the present study, mild cerebellar anomalies are probably the most common brain malformation associated with DEL22q11.
- Published
- 2006
25. Reciprocal fusion transcripts of two novel Zn-finger genes in a female with absence of the corpus callosum, ocular colobomas and a balanced translocation between chromosomes 2p24 and 9q32
- Author
-
Ramocki, Melissa B, Dowling, James, Grinberg, Inessa, Kimonis, Virginia E, Cardoso, Carlos, Gross, Alyssa, Chung, June, Lese Martin, Christa, Ledbetter, David H, Dobyns, William B, and Millen, Kathleen J
- Subjects
Biological Sciences ,Biomedical and Clinical Sciences ,Genetics ,2.1 Biological and endogenous factors ,Aetiology ,Agenesis of Corpus Callosum ,Chromosome Mapping ,Chromosomes ,Human ,Pair 2 ,Chromosomes ,Human ,Pair 9 ,Coloboma ,Female ,Humans ,Sequence Analysis ,DNA ,Translocation ,Genetic ,Zinc Fingers ,corpus callosum agenesis ,Zn-finger ,translocation ,CNS development ,periventricular nodular ,heterotopia ,Clinical Sciences ,Genetics & Heredity ,Clinical sciences - Abstract
We have identified a female patient with a complex phenotype that includes complete agenesis of the corpus callosum, bilateral periventricular nodular heterotopia, and bilateral chorioretinal and iris colobomas. Karyotype analysis revealed an apparently balanced, reciprocal, de novo chromosome translocation t(2;9)(p24;q32). Physical mapping of the translocation breakpoint by fluorescence in situ hybridization and PCR analysis led to the identification of two novel, ubiquitously expressed, Zn-finger-encoding transcripts that are disrupted in this patient. Unexpectedly, the rearrangement produced in-frame reciprocal fusion transcripts, making genotype-phenotype correlation difficult.
- Published
- 2003
26. Linkage of a Human Brain Malformation, Familial Holoprosencephaly, to Chromosome 7 and Evidence for Genetic Heterogeneity
- Author
-
Muenke, Maximilian, Gurrieri, Fiorella, Bay, Carolyn, Yi, David H., Collins, Amanda L., Johnson, Virginia P., Schaefer, G. Bradley, Weik, LuAnn, Lubinsky, Mark S., Daack-Hirsch, Sandy, Moore, Cynthia A., Dobyns, William B., Murray, Jeffrey C., and Price, R. Arlen
- Published
- 1994
27. 66 A Recurrent Pattern of Posterior Vermis-Predominant Cerebellar Hypoplasia (Not Dandy-Walker) Occurring with Psychosis-Schizophrenia.
- Author
-
Leslie, Alison C., Dobyns, William B., and Ward, Mitchell P.
- Subjects
- *
SLEEP interruptions , *TEENAGE boys , *GENETIC disorders , *MEMORY disorders , *BRAIN imaging - Abstract
Objective: Schizophrenia (SCZ) is a neuropsychiatric disorder with strong genetic heritability and predicted genetic heterogeneity, but limited knowledge regarding the underlying genetic risk variants. Classification into phenotype-driven subgroups or endophenotypes is expected to facilitate genetic analysis. Here, we report a teen boy with chronic psychosis and cerebellar hypoplasia (CBLH) and analyze data on 16 reported individuals with SCZ or chronic psychosis not otherwise specified associated with cerebellar hypoplasia to look for shared features. Participants and Methods: We evaluated an 18-year-old boy with neurodevelopmental deficits from early childhood and onset of hallucinations and other features of SCZ at 10 years who had mild vermis-predominant CBLH on brain imaging. This prompted us to review prior reports of chronic psychosis or SCZ with cerebellar malformations using paired search terms including (1) cerebellar hypoplasia, Dandy-Walker malformation, Dandy-Walker variant, or mega-cisterna magna with (2) psychosis or SCZ. We found reports of 16 affected individuals from 13 reports. We reviewed clinical features focusing on demographic information, prenatal-perinatal history and neuropsychiatric and neurodevelopmental phenotypes, and independently reviewed brain imaging features. Results: All 17 individuals had classic psychiatric features of SCZ or chronic psychosis as well as shared neurodevelopmental features not previously highlighted including a downward shift in IQ of about 20 points, memory impairment, speech-language deficits, attention deficits and sleep disturbances. The brain imaging findings among these individuals consistently showed posterior vermis predominant CBLH with variable cerebellar hemisphere hypoplasia and enlarged posterior fossa (a.k.a. mega-cisterna magna). None had features of classic DWM. Conclusions: In 17 individuals with chronic psychosis or SCZ and cerebellar malformation, we found a high frequency of neurodevelopmental disorders, a consistent brain malformation consisting of posterior vermis-predominant (and usually symmetric) CBLH, and no evidence of prenatal risk factors. The consistent phenotype and lack of prenatal risk factors for CBLH leads us to hypothesize that psychosis or schizophrenia associated with vermis predominant CBLH comprises a homogeneous subgroup of individuals with chronic psychosis/schizophrenia that is likely to have an underlying genetic basis. No comprehensive targeted gene panel for CBLH has yet been defined, leading us to recommend trio-based exome sequencing for individuals who present with this combination of features. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
28. Erratum : Correction: Human mutations in integrator complex subunits link transcriptome integrity to brain development (PLoS genetics (2017) 13 5 (e1006809))
- Author
-
Oegema, Renske, Baillat, David, Schot, Rachel, van Unen, Leontine M., Brooks, Alice, Kia, Sima Kheradmand, Hoogeboom, A. Jeannette M., Xia, Zheng, Li, Wei, Cesaroni, Matteo, Lequin, Maarten H., van Slegtenhorst, Marjon, Dobyns, William B., de Coo, Irenaeus F.M., van den Berg, Debbie, Verheijen, Frans W., Kremer, Andreas, van der Spek, Peter J., Heijsman, Daphne, Wagner, Eric J., Fornerod, Maarten, and Mancini, Grazia M.S.
- Subjects
Cancer Research ,Published Erratum ,Genetics ,Genetics(clinical) ,Molecular Biology ,Ecology, Evolution, Behavior and Systematics - Abstract
[This corrects the article DOI: 10.1371/journal.pgen.1006809.].
- Published
- 2017
29. De novo CCND2 mutations leading to stabilization of cyclin D2 cause megalencephaly-polymicrogyria-polydactyly-hydrocephalus syndrome
- Author
-
Mirzaa, Ghayda M, Parry, David A, Fry, Andrew E, Giamanco, Kristin A, Schwartzentruber, Jeremy, Vanstone, Megan, Logan, Clare V., Roberts, Nicola, Johnson, Colin A., Singh, Shawn, Kholmanskikh, Stanislav S., Adams, Carissa, Hodge, Rebecca D., Hevner, Robert F., Bonthron, David T., Braun, Kees P J, Faivre, Laurence, Rivière, Jean-Baptiste, St-Onge, Judith, Gripp, Karen W., Mancini, Grazia M S, Pang, Ki, Sweeney, Elizabeth, van Esch, Hilde, Verbeek, Nienke, Wieczorek, Dagmar, Steinraths, Michelle, Majewski, Jacek, Boycott, Kim M., Pilz, Daniela T., Ross, Elizabeth, Dobyns, William B., Sheridan, Eamonn G., Friedman, J., Michaud, J., Bernier, F., Brudno, M., Fernandez, B., Knoppers, B., Samuels, M., Scherer, S., and Clinical Genetics
- Subjects
endocrine system ,Blotting, Western ,Molecular Sequence Data ,Mutant ,Medizin ,Biology ,medicine.disease_cause ,Article ,AKT3 ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Cyclin D2 ,GSK-3 ,Genetics ,medicine ,Animals ,Humans ,Abnormalities, Multiple ,Exome ,Megalencephaly ,PI3K/AKT/mTOR pathway ,030304 developmental biology ,0303 health sciences ,Mutation ,Base Sequence ,Sequence Analysis, DNA ,Syndrome ,Cell cycle ,medicine.disease ,Immunohistochemistry ,Molecular biology ,Malformations of Cortical Development ,Polydactyly ,Electroporation ,HEK293 Cells ,Bromodeoxyuridine ,Microscopy, Fluorescence ,Mutagenesis, Site-Directed ,Female ,030217 neurology & neurosurgery ,Hydrocephalus - Abstract
Activating mutations in genes encoding phosphatidylinositol 3-kinase (PI3K)-AKT pathway components cause megalencephaly-polymicrogyria-polydactyly-hydrocephalus syndrome (MPPH, OMIM 603387)(1-3). Here we report that individuals with MPPH lacking upstream PI3K-AKT pathway mutations carry de novo mutations in CCND2 (encoding cyclin D2) that are clustered around a residue that can be phosphorylated by glycogen synthase kinase 313 (GSK-3 beta)(4). Mutant CCND2 was resistant to proteasomal degradation in vitro compared to wild-type CCND2. The PI3K-AKT pathway modulates GSK-3 beta activity(4), and cells from individuals with PIK3CA, PIK3R2 or AKT3 mutations showed similar CCND2 accumulation. CCND2 was expressed at higher levels in brains of mouse embryos expressing activated AKT3. In utero electroporation of mutant CCND2 into embryonic mouse brains produced more proliferating transfected progenitors and a smaller fraction of progenitors exiting the cell cycle compared to cells electroporated with wild-type CCND2. These observations suggest that cyclin D2 stabilization, caused by CCND2 mutation or PI3K-AKT activation, is a unifying mechanism in PI3K-AKT-related megalencephaly syndromes.
- Published
- 2014
30. De novo and inherited private variants in MAP1B in periventricular nodular heterotopia.
- Author
-
Heinzen, Erin L., O'Neill, Adam C., Zhu, Xiaolin, Allen, Andrew S., Bahlo, Melanie, Chelly, Jamel, Dobyns, William B., Freytag, Saskia, Guerrini, Renzo, Leventer, Richard J., Poduri, Annapurna, Robertson, Stephen P., Walsh, Christopher A., Zhang, Mengqi, and null, null
- Subjects
EPILEPSY ,GENETICS of epilepsy ,NEURAL development ,EXOMES ,ALLELES ,NUCLEOTIDE sequence - Abstract
Periventricular nodular heterotopia (PVNH) is a malformation of cortical development commonly associated with epilepsy. We exome sequenced 202 individuals with sporadic PVNH to identify novel genetic risk loci. We first performed a trio-based analysis and identified 219 de novo variants. Although no novel genes were implicated in this initial analysis, PVNH cases were found overall to have a significant excess of nonsynonymous de novo variants in intolerant genes (p = 3.27x10
-7 ), suggesting a role for rare new alleles in genes yet to be associated with the condition. Using a gene-level collapsing analysis comparing cases and controls, we identified a genome-wide significant signal driven by four ultra-rare loss-of-function heterozygous variants in MAP1B, including one de novo variant. In at least one instance, the MAP1B variant was inherited from a parent with previously undiagnosed PVNH. The PVNH was frontally predominant and associated with perisylvian polymicrogyria. These results implicate MAP1B in PVNH. More broadly, our findings suggest that detrimental mutations likely arising in immediately preceding generations with incomplete penetrance may also be responsible for some apparently sporadic diseases. [ABSTRACT FROM AUTHOR]- Published
- 2018
- Full Text
- View/download PDF
31. A developmental and genetic classification for malformations of cortical development: update 2012
- Author
-
Barkovich, A James, Guerrini, Renzo, Kuzniecky, Ruben I, Jackson, Graeme D, and Dobyns, William B
- Subjects
Diagnostic Imaging ,History ,Intellectual and Developmental Disabilities (IDD) ,1.1 Normal biological development and functioning ,Developmental Disabilities ,DNA Mutational Analysis ,Nervous System Malformations ,Medical and Health Sciences ,Rare Diseases ,Clinical Research ,Underpinning research ,Cell Movement ,Diagnosis ,Genetics ,Humans ,Genetic Predisposition to Disease ,Genetic Testing ,microcephaly ,polymicrogyria ,Molecular Biology ,Pediatric ,Cerebral Cortex ,Epilepsy ,Neurology & Neurosurgery ,Psychology and Cognitive Sciences ,Neurosciences ,malformation of cortical development ,21st Century ,Brain Disorders ,Malformations of Cortical Development ,Neurological ,Differential ,Mutation ,Congenital Structural Anomalies ,cortical dysplasia - Abstract
Malformations of cerebral cortical development include a wide range of developmental disorders that are common causes of neurodevelopmental delay and epilepsy. In addition, study of these disorders contributes greatly to the understanding of normal brain development and its perturbations. The rapid recent evolution of molecular biology, genetics and imaging has resulted in an explosive increase in our knowledge of cerebral cortex development and in the number and types of malformations of cortical development that have been reported. These advances continue to modify our perception of these malformations. This review addresses recent changes in our perception of these disorders and proposes a modified classification based upon updates in our knowledge of cerebral cortical development.
- Published
- 2012
32. Human mutations in integrator complex subunits link transcriptome integrity to brain development.
- Author
-
Oegema, Renske, Baillat, David, Schot, Rachel, van Unen, Leontine M., Brooks, Alice, Kia, Sima Kheradmand, Hoogeboom, A. Jeannette M., Xia, Zheng, Li, Wei, Cesaroni, Matteo, Lequin, Maarten H., van Slegtenhorst, Marjon, Dobyns, William B., de Coo, Irenaeus F. M., Verheijen, Frans W., Kremer, Andreas, van der Spek, Peter J., Heijsman, Daphne, Wagner, Eric J., and Fornerod, Maarten
- Subjects
INTEGRITY ,NEURAL development ,RNA polymerases ,BRAIN diseases ,BRAIN tumors - Abstract
Integrator is an RNA polymerase II (RNAPII)-associated complex that was recently identified to have a broad role in both RNA processing and transcription regulation. Importantly, its role in human development and disease is so far largely unexplored. Here, we provide evidence that biallelic Integrator Complex Subunit 1 (INTS1) and Subunit 8 (INTS8) gene mutations are associated with rare recessive human neurodevelopmental syndromes. Three unrelated individuals of Dutch ancestry showed the same homozygous truncating INTS1 mutation. Three siblings harboured compound heterozygous INTS8 mutations. Shared features by these six individuals are severe neurodevelopmental delay and a distinctive appearance. The INTS8 family in addition presented with neuronal migration defects (periventricular nodular heterotopia). We show that the first INTS8 mutation, a nine base-pair deletion, leads to a protein that disrupts INT complex stability, while the second missense mutation introduces an alternative splice site leading to an unstable messenger. Cells from patients with INTS8 mutations show increased levels of unprocessed UsnRNA, compatible with the INT function in the 3’-end maturation of UsnRNA, and display significant disruptions in gene expression and RNA processing. Finally, the introduction of the INTS8 deletion mutation in P19 cells using genome editing alters gene expression throughout the course of retinoic acid-induced neural differentiation. Altogether, our results confirm the essential role of Integrator to transcriptome integrity and point to the requirement of the Integrator complex in human brain development. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
33. Description of a new oncogenic mechanism for atypical teratoid rhabdoid tumors in patients with ring chromosome 22.
- Author
-
Byers, Heather M., Adam, Margaret P., LaCroix, Amy, Leary, Sarah E. S., Cole, Bonnie, Dobyns, William B., and Mefford, Heather C.
- Abstract
Atypical teratoid rhabdoid tumors of the central nervous system are rare, highly malignant, embryonal tumors most often occurring in children under age 3 years. Most are due to a somatic change in tumor suppressor gene SMARCB1 followed by a second-hit, typically loss of heterozygosity, best detected on immunohistochemical staining. Despite the noteworthy genetic homogeneity of atypical teratoid rhabdoid tumors, relatively little is known about the oncogenic mechanisms that lead to biallelic inactivation of SMARCB1. Herein, we describe a patient with constitutional ring chromosome 22, Phelan-McDermid syndrome and atypical teratoid rhabdoid tumor of the brain. During mitosis, sister chromatids of a ring chromosome may form interlocking and dicentric rings, resulting in chromosomal loss, complex karyotypes, and ongoing somatic variation. We hypothesized that the inherent instability of the patient's ring chromosome could lead to mosaic monosomy chromosome 22, resulting in allelic inactivation of the tumor-suppressor gene SMARCB1 and AT/RT if a second-hit occurred. Utilizing high-density microarray technology to analyze peripheral blood and tumor tissue, we confirmed this oncogenic mechanism, previously undescribed in patients with atypical teratoid rhabdoid tumors. Our data demonstrate chromosomal loss as a consequence of ring chromosome instability serving as the first hit in oncogenesis. This rare but possibly under-recognized mechanism is important to note in children with ATRT and syndromic features. Further investigation is warranted to assess if this oncogenic mechanism has management and/or prognostic implications. © 2016 Wiley Periodicals, Inc. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
34. Infantile hydrocephalus: A review of epidemiology, classification and causes.
- Author
-
Tully, Hannah M. and Dobyns, William B.
- Subjects
- *
HYDROCEPHALUS in infants , *EPIDEMIOLOGY , *DILATATION & extraction abortion , *ENVIRONMENTAL impact analysis , *LACTATION consultants , *AGE groups - Abstract
Hydrocephalus is a common but complex condition caused by physical or functional obstruction of CSF flow that leads to progressive ventricular dilatation. Though hydrocephalus was recently estimated to affect 1.1 in 1000 infants, there have been few systematic assessments of the causes of hydrocephalus in this age group, which makes it a challenging condition to approach as a scientist or as a clinician. Here, we review contemporary literature on the epidemiology, classification and pathogenesis of infantile hydrocephalus. We describe the major environmental and genetic causes of hydrocephalus, with the goal of providing a framework to assess infants with hydrocephalus and guide future research. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
35. Malformations of cortical development: clinical features and genetic causes.
- Author
-
Guerrini, Renzo and Dobyns, William B
- Subjects
- *
CEREBRAL cortex abnormalities , *DEVELOPMENTAL delay , *EPILEPSY , *BRAIN diseases , *GENETIC testing , *CLINICAL medicine , *DIAGNOSIS methods , *GENETICS - Abstract
Summary: Malformations of cortical development are common causes of developmental delay and epilepsy. Some patients have early, severe neurological impairment, but others have epilepsy or unexpected deficits that are detectable only by screening. The rapid evolution of molecular biology, genetics, and imaging has resulted in a substantial increase in knowledge about the development of the cerebral cortex and the number and types of malformations reported. Genetic studies have identified several genes that might disrupt each of the main stages of cell proliferation and specification, neuronal migration, and late cortical organisation. Many of these malformations are caused by de-novo dominant or X-linked mutations occurring in sporadic cases. Genetic testing needs accurate assessment of imaging features, and familial distribution, if any, and can be straightforward in some disorders but requires a complex diagnostic algorithm in others. Because of substantial genotypic and phenotypic heterogeneity for most of these genes, a comprehensive analysis of clinical, imaging, and genetic data is needed to properly define these disorders. Exome sequencing and high-field MRI are rapidly modifying the classification of these disorders. [Copyright &y& Elsevier]
- Published
- 2014
- Full Text
- View/download PDF
36. Copy number variants and infantile spasms: evidence for abnormalities in ventral forebrain development and pathways of synaptic function.
- Author
-
Paciorkowski, Alex R, Thio, Liu Lin, Rosenfeld, Jill A, Gajecka, Marzena, Gurnett, Christina A, Kulkarni, Shashikant, Chung, Wendy K, Marsh, Eric D, Gentile, Mattia, Reggin, James D, Wheless, James W, Balasubramanian, Sandhya, Kumar, Ravinesh, Christian, Susan L, Marini, Carla, Guerrini, Renzo, Maltsev, Natalia, Shaffer, Lisa G, and Dobyns, William B
- Subjects
INFANTILE spasms ,PROSENCEPHALON abnormalities ,ETIOLOGY of diseases ,CHROMOSOME abnormalities ,BIOINFORMATICS ,GENETICS - Abstract
Infantile spasms (ISS) are an epilepsy disorder frequently associated with severe developmental outcome and have diverse genetic etiologies. We ascertained 11 subjects with ISS and novel copy number variants (CNVs) and combined these with a new cohort with deletion 1p36 and ISS, and additional published patients with ISS and other chromosomal abnormalities. Using bioinformatics tools, we analyzed the gene content of these CNVs for enrichment in pathways of pathogenesis. Several important findings emerged. First, the gene content was enriched for the gene regulatory network involved in ventral forebrain development. Second, genes in pathways of synaptic function were overrepresented, significantly those involved in synaptic vesicle transport. Evidence also suggested roles for GABAergic synapses and the postsynaptic density. Third, we confirm the association of ISS with duplication of 14q12 and maternally inherited duplication of 15q11q13, and report the association with duplication of 21q21. We also present a patient with ISS and deletion 7q11.3 not involving MAGI2. Finally, we provide evidence that ISS in deletion 1p36 may be associated with deletion of KLHL17 and expand the epilepsy phenotype in that syndrome to include early infantile epileptic encephalopathy. Several of the identified pathways share functional links, and abnormalities of forebrain synaptic growth and function may form a common biologic mechanism underlying both ISS and autism. This study demonstrates a novel approach to the study of gene content in subjects with ISS and copy number variation, and contributes further evidence to support specific pathways of pathogenesis. [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
37. A developmental and genetic classification for midbrain-hindbrain malformations.
- Author
-
Barkovich, A. James, Millen, Kathleen J., and Dobyns, William B.
- Subjects
BRAIN abnormalities ,MESENCEPHALON ,RHOMBENCEPHALON ,BRAIN imaging ,EMBRYOLOGY ,GENETICS ,ATROPHY - Abstract
Advances in neuroimaging, developmental biology and molecular genetics have increased the understanding of developmental disorders affecting the midbrain and hindbrain, both as isolated anomalies and as part of larger malformation syndromes. However, the understanding of these malformations and their relationships with other malformations, within the central nervous system and in the rest of the body, remains limited. A new classification system is proposed, based wherever possible, upon embryology and genetics. Proposed categories include: (i) malformations secondary to early anteroposterior and dorsoventral patterning defects, or to misspecification of mid-hindbrain germinal zones; (ii) malformations associated with later generalized developmental disorders that significantly affect the brainstem and cerebellum (and have a pathogenesis that is at least partly understood); (iii) localized brain malformations that significantly affect the brain stem and cerebellum (pathogenesis partly or largely understood, includes local proliferation, cell specification, migration and axonal guidance); and (iv) combined hypoplasia and atrophy of putative prenatal onset degenerative disorders. Pertinent embryology is discussed and the classification is justified. This classification will prove useful for both physicians who diagnose and treat patients with these disorders and for clinical scientists who wish to understand better the perturbations of developmental processes that produce them. Importantly, both the classification and its framework remain flexible enough to be easily modified when new embryologic processes are described or new malformations discovered. [ABSTRACT FROM PUBLISHER]
- Published
- 2009
- Full Text
- View/download PDF
38. Linkage to chromosome 2q36.1 in autosomal dominant Dandy-Walker malformation with occipital cephalocele and evidence for genetic heterogeneity.
- Author
-
Jalali, Ali, Aldinger, Kimberly A., Chary, Ajit, Mclone, David G., Bowman, Robin M., Luan Cong Le, Jardine, Phillip, Newbury-Ecob, Ruth, Mallick, Andrew, Jafari, Nadereh, Russell, Eric J., Curran, John, Nguyen, Pam, Ouahchi, Karim, Lee, Charles, Dobyns, William B., Millen, Kathleen J., Pina-Neto, Joao M., Kessler, John A., and Bassuk, Alexander G.
- Subjects
CHROMOSOMES ,GENETICS ,HEREDITY ,GENOMES ,GENES ,ETIOLOGY of diseases - Abstract
We previously reported a Vietnamese-American family with isolated autosomal dominant occipital cephalocele. Upon further neuroimaging studies, we have recharacterized this condition as autosomal dominant Dandy-Walker with occipital cephalocele (ADDWOC). A similar ADDWOC family from Brazil was also recently described. To determine the genetic etiology of ADDWOC, we performed genome-wide linkage analysis on members of the Vietnamese-American and Brazilian pedigrees. Linkage analysis of the Vietnamese-American family identified the ADDWOC causative locus on chromosome 2q36.1 with a multipoint parametric LOD score of 3.3, while haplotype analysis refined the locus to 1.1 Mb. Sequencing of the five known genes in this locus did not identify any protein-altering mutations. However, a terminal deletion of chromosome 2 in a patient with an isolated case of Dandy-Walker malformation also encompassed the 2q36.1 chromosomal region. The Brazilian pedigree did not show linkage to this 2q36.1 region. Taken together, these results demonstrate a locus for ADDWOC on 2q36.1 and also suggest locus heterogeneity for ADDWOC. [ABSTRACT FROM AUTHOR]
- Published
- 2008
- Full Text
- View/download PDF
39. Abnormal development of the human cerebral cortex: genetics, functional consequences and treatment options
- Author
-
Guerrini, Renzo, Dobyns, William B., and Barkovich, A. James
- Subjects
- *
NEUROLOGICAL disorders , *GENETICS , *CELL proliferation , *GENES , *HIGHER nervous activity , *DEVELOPMENTAL disabilities , *EPILEPSY - Abstract
Genetic studies have identified several of the genes associated with malformations of cortical development which might disrupt each of the main stages of cell proliferation and specification, neuronal migration and late cortical organization. The largest malformation groups, focal cortical dysplasia, heterotopia and polymicrogyria, express different perturbations of these stages and carry a variable propensity for lacking activation, preservation or reorganization of cortical function and for atypical cortical organization. Some patients have obvious neurological impairment, whereas others show unexpected deficits that are detectable only by screening. Drug-resistant epilepsy is frequent but might be amenable to surgical treatment. However, the epileptogenic zone might include remote cortical and subcortical regions. Completeness of resection, a key factor for successful surgery, might be difficult, especially in proximity to eloquent cortex. Surgical planning should be based on assessments of structural imaging and of the major functions relevant to the area in question in any such patient. [Copyright &y& Elsevier]
- Published
- 2008
- Full Text
- View/download PDF
40. Opinion: Genetic links between brain development and brain evolution.
- Author
-
Gilbert, Sandra L., Dobyns, William B., and Lahn, Bruce T.
- Subjects
- *
BRAIN , *GENETICS , *GENES , *BIOLOGICAL evolution , *CENTRAL nervous system - Abstract
The most defining biological attribute of Homo sapiens is its enormous brain size and accompanying cognitive prowess. How this was achieved by means of genetic changes over the course of human evolution has fascinated biologists and the general public alike. Recent studies have shown that genes controlling brain development — notably those implicated in microcephaly (a congenital defect that is characterized by severely reduced brain size) — are favoured targets of natural selection during human evolution. We propose that genes that regulate brain size during development, such as microcephaly genes, are chief contributors in driving the evolutionary enlargement of the human brain. Based on the synthesis of recent studies, we propose a general methodological template for the genetic analysis of human evolution. [ABSTRACT FROM AUTHOR]
- Published
- 2005
- Full Text
- View/download PDF
41. Human malformations of the midbrain and hindbrain: review and proposed classification scheme
- Author
-
Parisi, Melissa A. and Dobyns, William B.
- Subjects
- *
GENETICS , *ETIOLOGY of diseases , *GENETIC disorders , *FOSSA (Mammals) - Abstract
Although a great deal of interest in the genetics and etiology of cerebral, particularly forebrain, malformations has been generated in the past decade, relatively little is known about the basis of congenital malformations of the structures of the posterior fossa, namely the midbrain, cerebellum, pons, and medulla. In this review, we present a classification scheme for malformations of the midbrain and hindbrain based on their embryologic derivation, highlight four of the conditions associated with such abnormalities, and describe the genetics, prognosis, and recurrence risks for each. We describe several disorders in addition to Joubert syndrome with the distinctive radiologic sign known as the “molar tooth sign,” comprised of midbrain and hindbrain malformations. We discuss Dandy–Walker malformation, its classical definition, and the surprisingly good outcome in the absence of other brain malformations. We consider the heterogeneous entity of cerebellar vermis hypoplasia and describe the recently identified gene associated with an X-linked form of this condition. Finally, the pontocerebellar hypoplasias are discussed in the context of their generally progressive degenerative and severe course, and the differential diagnosis is emphasized. We anticipate that as imaging technologies improve, differentiation of the various disorders should aid in efforts to identify the causative genes. [Copyright &y& Elsevier]
- Published
- 2003
- Full Text
- View/download PDF
42. Mutation analysis of the DCX gene and genotype/phenotype correlation in subcortical band heterotopia.
- Author
-
Matsumoto, Naomichi, Leventer, Richard J, Kuc, Julie A, Mewborn, Stephanie K, Dudlicek, Laura L, Ramocki, Melissa B, Pilz, Daniela T, Mills, Patti L, Das, Soma, Ross, M Elizabeth, Ledbetter, David H, and Dobyns, William B
- Subjects
BRAIN abnormalities ,GENETIC mutation ,GENETICS - Abstract
Subcortical band heterotopia (SBH) comprises part of a spectrum of phenotypes associated with classical lissencephaly (LIS). LIS and SBH are caused by alterations in at least two genes: LIS1 (PAFAH1B1) at 17p13.3 and DCX (doublecortin) at Xq22.3-q23. DCX mutations predominantly cause LIS in hemizygous males and SBH in heterozygous females, and we have evaluated several families with LIS male and SBH female siblings. In this study, we performed detailed DCX mutation analysis and genotype-phenotype correlation in a large cohort with typical SBH. We screened 26 sporadic SBH females and 11 LIS/SBH families for DCX mutations by direct sequencing. We found 29 mutations in 22 sporadic patients and 11 pedigrees, including five deletions, four nonsense mutations, 19 missense mutations and one splice donor site mutation. The DCX mutation prevalence was 84.6% (22 of 26) in sporadic SBH patients and 100% (11 of 11) in SBH pedigrees. Maternal germline mosaicism was found in one family. Significant differences in genotype were found in relation to band thickness and familial vs sporadic status. [ABSTRACT FROM AUTHOR]
- Published
- 2001
- Full Text
- View/download PDF
43. Heterozygous deletion of the linked genes ZIC1 and ZIC4 is involved in Dandy-Walker malformation.
- Author
-
Grinberg, Inessa, Northrup, Hope, Ardinger, Holly, Prasad, Chitra, Dobyns, William B., and Millen, Kathleen J.
- Subjects
CEREBELLUM diseases ,GENETIC disorders ,GENETICS ,MOLECULAR genetics ,PHENOTYPES ,RODENTS - Abstract
Dandy-Walker malformation (DWM; OMIM#220200) is a common but poorly understood congenital cerebellar malformation in humans. Through physical mapping of 3q2 interstitial deletions in several individuals with DWM, we defined the first critical region associated with DWM, encompassing two adjacent Zinc finger in cerebellum genes, ZIC1 and ZIC4. Mice with a heterozygous deletion of these two linked genes have a phenotype that closely resembles DWM, providing a mouse model for this malformation. [ABSTRACT FROM AUTHOR]
- Published
- 2004
- Full Text
- View/download PDF
44. De novo and inherited private variants in MAP1B in periventricular nodular heterotopia
- Author
-
Heinzen, Erin L., O’Neill, Adam C., Zhu, Xiaolin, Allen, Andrew S., Bahlo, Melanie, Chelly, Jamel, Chen, Ming Hui, Dobyns, William B., Freytag, Saskia, Guerrini, Renzo, Leventer, Richard J., Poduri, Annapurna, Robertson, Stephen P., Walsh, Christopher A., Zhang, Mengqi, Epi4K Consortium, and Epilepsy Phenome/Genome Project
- Subjects
Epilepsy ,FOS: Biological sciences ,FOS: Clinical medicine ,Genetics ,Neurosciences ,Genomics ,3. Good health - Abstract
Periventricular nodular heterotopia (PVNH) is a malformation of cortical development commonly associated with epilepsy. We exome sequenced 202 individuals with sporadic PVNH to identify novel genetic risk loci. We first performed a trio-based analysis and identified 219 de novo variants. Although no novel genes were implicated in this initial analysis, PVNH cases were found overall to have a significant excess of nonsynonymous de novo variants in intolerant genes (p = 3.27x10-7), suggesting a role for rare new alleles in genes yet to be associated with the condition. Using a gene-level collapsing analysis comparing cases and controls, we identified a genome-wide significant signal driven by four ultra-rare loss-of-function heterozygous variants in MAP1B, including one de novo variant. In at least one instance, the MAP1B variant was inherited from a parent with previously undiagnosed PVNH. The PVNH was frontally predominant and associated with perisylvian polymicrogyria. These results implicate MAP1B in PVNH. More broadly, our findings suggest that detrimental mutations likely arising in immediately preceding generations with incomplete penetrance may also be responsible for some apparently sporadic diseases.
45. Pathological subtypes of polymicrogyria and brain development.
- Author
-
Judkins, Alexander Russell, Dobyns, William B., and Golden, Jeffrey A.
- Subjects
- *
BRAIN abnormalities , *NEUROLOGICAL disorders , *ETIOLOGY of diseases , *PATHOLOGY , *GENETICS - Abstract
The neuropathology of polymicrogyria (PMG) is controversial. It is generally agreed that PMG has multiple etiologies including destructive and genetic. The pathologic characteristics of PMG remain confusing. Two, 4, and 6 layer PMG have been described and attributed etiological significance. Accurate pathologic classification of PMG requires correlating with clinical, radiographic, and genetics. Seventeen cases of PMG were reviewed for: the number of laminae, the variability of neuropathologic features within cases, and other neuropathologic lesions. Cases included clear destructive lesions (eg. porencephaly), presumed genetic basis (eg. 22q13 deletion syndrome), and those with no known etiology. Cases included 4 layer, as well as 2 and 6 layer PMG. More than one histopathologic type of PMG was frequently observed in individual cases. Several findings were common, including heterotopia (nodular and periventricular). The findings in this series of PMG cases indicate that classification according to number of laminae is not useful. Future studies correlating pathology, radiology, genetics and clinical features are necessary and will likely provide insight into the heterogeneity observed in PMG. [ABSTRACT FROM AUTHOR]
- Published
- 2007
- Full Text
- View/download PDF
46. Somatic PDGFRB Activating Variants in Fusiform Cerebral Aneurysms.
- Author
-
Karasozen, Yigit, Osbun, Joshua W., Parada, Carolina Angelica, Busald, Tina, Tatman, Philip, Gonzalez-Cuyar, Luis F., Hale, Christopher J., Alcantara, Diana, O'Driscoll, Mark, Dobyns, William B., Murray, Mitzi, Kim, Louis J., Byers, Peter, Dorschner, Michael O., and Ferreira, Manuel
- Subjects
- *
INTRACRANIAL aneurysms , *PLATELET-derived growth factor receptors , *SOMATIC embryogenesis , *FISHER exact test - Abstract
The role of somatic genetic variants in the pathogenesis of intracranial-aneurysm formation is unknown. We identified a 23-year-old man with progressive, right-sided intracranial aneurysms, ipsilateral to an impressive cutaneous phenotype. The index individual underwent a series of genetic evaluations for known connective-tissue disorders, but the evaluations were unrevealing. Paired-sample exome sequencing between blood and fibroblasts derived from the diseased areas detected a single novel variant predicted to cause a p.Tyr562Cys (g.149505130T>C [GRCh37/hg19]; c.1685A>G) change within the platelet-derived growth factor receptor β gene (PDGFRB), a juxtamembrane-coding region. Variant-allele fractions ranged from 18.75% to 53.33% within histologically abnormal tissue, suggesting post-zygotic or somatic mosaicism. In an independent cohort of aneurysm specimens, we detected somatic-activating PDGFRB variants in the juxtamembrane domain or the kinase activation loop in 4/6 fusiform aneurysms (and 0/38 saccular aneurysms; Fisher's exact test, p < 0.001). PDGFRB -variant, but not wild-type, patient cells were found to have overactive auto-phosphorylation with downstream activation of ERK, SRC, and AKT. The expression of discovered variants demonstrated non-ligand-dependent auto-phosphorylation, responsive to the kinase inhibitor sunitinib. Somatic gain-of-function variants in PDGFRB are a novel mechanism in the pathophysiology of fusiform cerebral aneurysms and suggest a potential role for targeted therapy with kinase inhibitors. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
47. Mutation of FOXC1 and PITX2 induces cerebral small-vessel disease.
- Author
-
French, Curtis R., Seshadri, Sudha, Destefano, Anita L., Fornage, Myriam, Arnold, Corey R., Gage, Philip J., Skarie, Jonathan M., Dobyns, William B., Millen, Kathleen J., Ting Liu, Dietz, William, Tsutomu Kume, Hofker, Marten, Emery, Derek J., Childs, Sarah J., Waskiewicz, Andrew J., Lehmann, Ordan J., Liu, Ting, and Kume, Tsutomu
- Subjects
- *
CEREBRAL small vessel diseases , *STROKE , *GENETIC mutation , *GENOMES , *CARDIOVASCULAR diseases , *BRAIN diseases , *ANIMAL experimentation , *CEREBRAL hemorrhage , *CELLULAR signal transduction , *COMPARATIVE studies , *DISEASE susceptibility , *FISHES , *GENES , *GENETIC polymorphisms , *GENETICS , *RESEARCH methodology , *MEDICAL cooperation , *PLATELET-derived growth factor , *PROTEINS , *RESEARCH , *RESEARCH funding , *TRANSCRIPTION factors , *EVALUATION research , *SEQUENCE analysis - Abstract
Patients with cerebral small-vessel disease (CSVD) exhibit perturbed end-artery function and have an increased risk for stroke and age-related cognitive decline. Here, we used targeted genome-wide association (GWA) analysis and defined a CSVD locus adjacent to the forkhead transcription factor FOXC1. Moreover, we determined that the linked SNPs influence FOXC1 transcript levels and demonstrated that patients as young as 1 year of age with altered FOXC1 function exhibit CSVD. MRI analysis of patients with missense and nonsense mutations as well as FOXC1-encompassing segmental duplication and deletion revealed white matter hyperintensities, dilated perivascular spaces, and lacunar infarction. In a zebrafish model, overexpression or morpholino-induced suppression of foxc1 induced cerebral hemorrhage. Inhibition of foxc1 perturbed platelet-derived growth factor (Pdgf) signaling, impairing neural crest migration and the recruitment of mural cells, which are essential for vascular stability. GWA analysis also linked the FOXC1-interacting transcription factor PITX2 to CSVD, and both patients with PITX2 mutations and murine Pitx2-/- mutants displayed brain vascular phenotypes. Together, these results extend the genetic etiology of stroke and demonstrate an increasing developmental basis for human cerebrovascular disease. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
48. Mutations in the O-Mannosyltransferase Gene POMT1 Give Rise to the Severe Neuronal Migration Disorder Walker-Warburg Syndrome.
- Author
-
de Bernabe, Daniel Beltran-Valero, Currier, Sophie, Steinbrecher, Alice, Celli, Jacopo, van Beusekom, Ellen, van der Zwaag, Bert, Kayserili, Hulya, Merlini, Luciano, Chitayat, David, Dobyns, William B., Cormand, Bru, Lehesjoki, Ana-Elina, Cruces, Jesus, Voit, Thomas, Walsh, Christopher A., van Bokhoven, Hans, and Brunner, Hans G.
- Subjects
- *
SYNDROMES , *GENETIC mutation , *GENETICS - Abstract
Walker-Warburg syndrome (WWS) is an autosomal recessive developmental disorder characterized by congenital muscular dystrophy and complex brain and eye abnormalities. A similar combination of symptoms is presented by two other human diseases, muscle-eye-brain disease (MEB) and Fukuyama congenital muscular dystrophy (FCMD). Although the genes underlying FCMD (Fukutin) and MEB (POMGnT1) have been cloned, loci for WWS have remained elusive. The protein products of POMGnT1 and Fukutin have both been implicated in protein glycosylation. To unravel the genetic basis of WWS, we first performed a genomewide linkage analysis in 10 consanguineous families with WWS. The results indicated the existence of at least three WWS loci. Subsequently, we adopted a candidate-gene approach in combination with homozygosity mapping in 15 consanguineous families with WWS. Candidate genes were selected on the basis of the role of the FCMD and MEB genes. Since POMGnT1 encodes an O-mannoside N-acetylglucosaminyltransferase, we analyzed the possible implication of O-mannosyl glycan synthesis in WWS. Analysis of the locus for O-mannosyltransferase 1 (POMT1) revealed homozygosity in 5 of 15 families. Sequencing of the POMT1 gene revealed mutations in 6 of the 30 unrelated patients with WWS. Of the five mutations identified, two are nonsense mutations, two are frameshift mutations, and one is a missense mutation. Immunohistochemical analysis of muscle from patients with POMT1 mutations corroborated the Omannosylation defect, as judged by the absence of glycosylation of α-dystroglycan. The implication of O-mannosylation in MEB and WWS suggests new lines of study in understanding the molecular basis of neuronal migration. [ABSTRACT FROM AUTHOR]
- Published
- 2002
49. A Locus for Bilateral Perisylvian Polymicrogyria Maps to Xq28.
- Author
-
Villard, Laurent, Nguyen, Karine, Cardoso, Carlos, Martin, Christa Lese, Weiss, Ann M., Sifry-Platt, Mara, Grix, Arthur W., Graham Jr., John M., Winter, Robin M., Leventer, Richard J., and Dobyns, William B.
- Subjects
- *
BRAIN diseases , *GENES , *GENETICS - Abstract
Polymicrogyria (PMG) is one of a large group of human cortical malformations that collectively account for a significant percentage of patients with epilepsy, congenital neurological deficits, and intellectual disability. PMG is characterized by an excess of small gyri and abnormal cortical lamination. The most common distribution is bilateral, symmetrical, and maximal, in the region surrounding the sylvian fissures, and is known as bilateral perisylvian polymicrogyria (BPP). Most cases are sporadic, although several families have been observed with multiple affected members, usually following an X-linked inheritance pattern. Here we report the first genetic locus for BPP mapped by linkage analysis in five families. Linkage places the critical region for BPP at Xq28 (LOD score 3.08 in Xq28, distal to DXS8103 by multipoint analysis). We suggest that this region contains a gene that is necessary for correct neuronal organization and that the identification of this gene will both enhance our understanding of normal cortical development and accelerate the identification of other genes responsible for PMG. [ABSTRACT FROM AUTHOR]
- Published
- 2002
- Full Text
- View/download PDF
50. Mutations in MAST1 Cause Mega-Corpus-Callosum Syndrome with Cerebellar Hypoplasia and Cortical Malformations
- Author
-
William B. Dobyns, Maria Fernanda Martinez-Reza, Jamel Chelly, Tessa van Dijk, Lydie Burglen, Martin W. Breuss, Gregory M. Cooper, Stefano Lise, Nadia Bahi-Buisson, Norine Voisin, Usha Kini, Linlea Armstrong, Nicholas J. Cowan, Stéphanie Valence, Andrea Wenninger-Weinzierl, Thomas A. Leonard, Frank Baas, Lukas Landler, Ennio Del Giudice, Jonathan A. Bernstein, Ghayda Mirzaa, Guoling Tian, Kimberly A. Aldinger, Bregje W.M. van Bon, Alexandre Reymond, Tyler Mark Pierson, Giuseppina Vitiello, Ratna Tripathy, Thomas Gstrein, Gaetano Terrone, Alex R. Paciorkowski, Maria Christina Sergaki, Alessandra D'Amico, Susan M. Hiatt, Ines Leca, Janneke Weiss, Ellyn Farrelly, Alistair T. Pagnamenta, Jenny C. Taylor, Nicola Brunetti-Pierri, David A. Keays, Tripathy, Ratna, Leca, Ine, van Dijk, Tessa, Weiss, Janneke, van Bon, Bregje W, Sergaki, Maria Christina, Gstrein, Thoma, Breuss, Martin, Tian, Guoling, Bahi-Buisson, Nadia, Paciorkowski, Alexander R, Pagnamenta, Alistair T, Wenninger-Weinzierl, Andrea, Martinez-Reza, Maria Fernanda, Landler, Luka, DE LISE, Stefano, Taylor, Jenny C, Terrone, Gaetano, Vitiello, Giuseppina, Del Giudice, Ennio, Brunetti-Pierri, Nicola, D'Amico, Alessandra, Reymond, Alexandre, Voisin, Norine, Bernstein, Jonathan A, Farrelly, Ellyn, Kini, Usha, Leonard, Thomas A, Valence, Stéphanie, Burglen, Lydie, Armstrong, Linlea, Hiatt, Susan M, Cooper, Gregory M, Aldinger, Kimberly A, Dobyns, William B, Mirzaa, Ghayda, Pierson, Tyler Mark, Baas, Frank, Chelly, Jamel, Cowan, Nicholas J, and Keays, David Anthony
- Subjects
0301 basic medicine ,Male ,Microcephaly ,Pathology ,PAX6 Transcription Factor ,Developmental Disabilities ,Apoptosis ,Corpus callosum ,Inbred C57BL ,AKT3 ,corpus callosum ,Mice ,Cerebellum ,2.1 Biological and endogenous factors ,Psychology ,Developmental ,Megalencephaly ,Aetiology ,Child ,Cells, Cultured ,Mice, Knockout ,Pediatric ,Cultured ,General Neuroscience ,Gene Expression Regulation, Developmental ,Brain ,Phenotype ,Malformations of Cortical Development ,Embryo ,Neurological ,Female ,MAST1 ,Cognitive Sciences ,Microtubule-Associated Proteins ,Rare cancers Radboud Institute for Health Sciences [Radboudumc 9] ,medicine.medical_specialty ,cerebellar hypoplasia ,Cells ,Knockout ,Intellectual and Developmental Disabilities (IDD) ,Nerve Tissue Proteins ,Biology ,Nervous System Malformations ,Article ,microtubules ,03 medical and health sciences ,Rare Diseases ,All institutes and research themes of the Radboud University Medical Center ,medicine ,Genetics ,Animals ,Humans ,PI3K/AKT/mTOR pathway ,Neurology & Neurosurgery ,Animal ,Mammalian ,Neurosciences ,medicine.disease ,Embryo, Mammalian ,Newborn ,Brain Disorders ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,Animals, Newborn ,Gene Expression Regulation ,Disease Models ,Mutation ,Autism ,Congenital Structural Anomalies ,Cerebellar hypoplasia (non-human) ,Agenesis of Corpus Callosum ,microdeletion - Abstract
Corpus callosum malformations are associated with a broad range of neurodevelopmental diseases. We report that de novo mutations in MAST1 cause mega-corpus-callosum syndrome with cerebellar hypoplasia and cortical malformations (MCC-CH-CM) in the absence of megalencephaly. We show that MAST1 is a microtubule associated protein, that is predominantly expressed in post-mitotic neurons, and is present in both dendritic and axonal compartments. We further show that Mast1 null animals are phenotypically normal, whereas the deletion of a single amino acid (L278del) recapitulates the distinct neurological phenotype observed in patients. In animals harboring Mast1 microdeletions we find that the PI3K/AKT3/mTOR pathway is unperturbed, whereas Mast2 and Mast3 levels are diminished, indicative of a dominant negative mode of action. Finally, we report that de novo MAST1 substitutions are present in patients with autism and microcephaly, raising the prospect that mutations in this gene give rise to a spectrum of neurodevelopmental diseases.
- Published
- 2018
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.