1
|
Gins C, Guimiot F, Drunat S, Prévost C, Rosenblatt J, Capri Y, Letard P, Khung-Savatovsky S, Mahi Henni MA, Elalaoui SC, Alison M, Guilmin Crepon S, Gressens P, Verloes A, Basto R, El Ghouzzi V, Passemard S. Radial Microbrain (Micrencephaly) Is Caused by a Recurrent Variant in the RTTN Gene. Neurol Genet 2025; 11:e200221. [PMID: 40151166 PMCID: PMC11949245 DOI: 10.1212/nxg.0000000000200221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 10/22/2024] [Indexed: 03/29/2025]
Abstract
Background and Objectives Genetic primary microcephaly (PM) is a defect in early brain development leading to congenital microcephaly, mostly recessively inherited, and mild-to-moderate intellectual disability. PM has been largely elucidated, thanks to exome and genome sequencing. However, radial microbrain, the most severe form of genetic PM or micrencephaly described in the 1980s, which leads to early lethality or very severe intellectual handicap, remains without a molecular diagnosis. We sought to identify the cause of radial microbrain by analyzing the genotype of children/adults and fetuses with an extremely small brain. Methods We searched for individuals with the smallest head circumference among patients with a confirmed diagnosis of PM included in 2 French and European observational studies coordinated at the Robert Debré Children's Hospital in Paris. Their neurodevelopment and brain imaging were analyzed, as well as next-generation sequencing for a panel of microcephaly genes or exome sequencing. Neuropathologic and immunohistologic analyses of extremely severe microcephalic fetal brains and stage-matched controls were performed. A nonparametric test and Mann-Whitney post-test were used to compare the cortical thickness between groups. Results We identified 5 individuals (4 female patients, 7 years 10 months-19 years) with a particularly small brain among a series of 50, all suffering from a severe neurodevelopmental disorder with no ability to communicate verbally and, in 3 of them, no ability to walk. Genetic analysis revealed in all individuals the presence of the same homozygous variant c.2953A>G (p.R985G) in the RTTN gene (ROTATIN). The same variant was found in 2 fetuses whose neuropathologic evaluation showed a major reduction in the thickness of the ventricular zone and neuronal heterotopias. The cortical plate was reduced by 70% compared with controls, irrespective of the region considered. Immunostaining with vimentin showed a 50% loss of radial glial columns, characteristic of radial microbrain. Discussion Our data show that the homozygous c.2953A>G substitution in RTTN is a recurrent variant responsible for radial microbrain, the most severe form of primary microcephaly. Our combined neurologic, imaging, and histopathologic approaches provide a better understanding of the severity of this condition and its prognosis. Trial Registration Information ClinicalTrials.gov number: NCT01565005.
Collapse
Affiliation(s)
- Clarisse Gins
- Service de Neurologie Pédiatrique, DMU INOV-RDB, APHP, Hôpital Robert Debré, Paris, France
| | - Fabien Guimiot
- Université Paris Cité, Inserm UMR 1141, NeuroDiderot, Paris, France
- Département de Génétique, UF de fœtopathologie, DMU BIOGE'M, APHP, Hôpital Robert Debré, Paris, France
| | - Séverine Drunat
- Université Paris Cité, Inserm UMR 1141, NeuroDiderot, Paris, France
- Département de Génétique, UF de Génétique Moléculaire, DMU BIOGE'M, APHP, Hôpital Robert Debré, Paris, France
| | - Clemence Prévost
- Université Paris Cité, Inserm UMR 1141, NeuroDiderot, Paris, France
| | - Jonathan Rosenblatt
- Unité de Médecine Foetale, DMU Gynécologie Périnatalité, APHP, Hôpital Robert Debré, Paris, France
| | - Yline Capri
- Département de Génétique, UF de Génétique Clinique, DMU BIOGE'M, APHP, Hôpital Robert Debré, Paris, France
| | - Pascaline Letard
- Département de Génétique, UF de fœtopathologie, DMU BIOGE'M, APHP, Hôpital Robert Debré, Paris, France
| | - Suonavy Khung-Savatovsky
- Département de Génétique, UF de fœtopathologie, DMU BIOGE'M, APHP, Hôpital Robert Debré, Paris, France
| | | | | | - Marianne Alison
- Université Paris Cité, Inserm UMR 1141, NeuroDiderot, Paris, France
- Service de Radiologie pédiatrique, APHP, Hôpital Robert Debré, Paris, France
| | | | - Pierre Gressens
- Université Paris Cité, Inserm UMR 1141, NeuroDiderot, Paris, France
| | - Alain Verloes
- Université Paris Cité, Inserm UMR 1141, NeuroDiderot, Paris, France
- Département de Génétique, UF de Génétique Clinique, DMU BIOGE'M, APHP, Hôpital Robert Debré, Paris, France
| | - Renata Basto
- Biology of Centrosomes and Genetic instability, Institut Curie, PSL Research University, CNRS UMR 144, Paris, France
| | | | - Sandrine Passemard
- Service de Neurologie Pédiatrique, DMU INOV-RDB, APHP, Hôpital Robert Debré, Paris, France
- Université Paris Cité, Inserm UMR 1141, NeuroDiderot, Paris, France
| |
Collapse
|
2
|
Guguin J, Chen TY, Cuinat S, Besson A, Bertiaux E, Boutaud L, Ardito N, Imaz Murguiondo M, Cabet S, Hamel V, Thomas S, Pain B, Edery P, Putoux A, Tang TK, Mazoyer S, Delous M. A Taybi-Linder syndrome-related RTTN variant impedes neural rosette formation in human cortical organoids. PLoS Genet 2024; 20:e1011517. [PMID: 39680576 PMCID: PMC11684760 DOI: 10.1371/journal.pgen.1011517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 12/30/2024] [Accepted: 11/27/2024] [Indexed: 12/18/2024] Open
Abstract
Taybi-Linder syndrome (TALS) is a rare autosomal recessive disorder characterized by severe microcephaly with abnormal gyral pattern, severe growth retardation and bone abnormalities. It is caused by pathogenic variants in the RNU4ATAC gene. Its transcript, the small nuclear RNA U4atac, is involved in the excision of ~850 minor introns. Here, we report a patient presenting with TALS features but no pathogenic variants were found in RNU4ATAC, instead the homozygous RTTN c.2953A>G variant was detected by whole-exome sequencing. After deciphering the impact of the variant on the RTTN protein function at centrosome in engineered RTTN-depleted RPE1 cells and patient fibroblasts, we analysed neural stem cells (NSC) derived from CRISPR/Cas9-edited induced pluripotent stem cells and revealed major cell cycle and mitotic abnormalities, leading to aneuploidy, cell cycle arrest and cell death. In cortical organoids, we discovered an additional function of RTTN in the self-organisation of NSC into neural rosettes, by observing delayed apico-basal polarization of NSC. Altogether, these defects contributed to a marked delay of rosette formation in RTTN-mutated organoids, thus impeding their overall growth and shedding light on mechanisms leading to microcephaly.
Collapse
Affiliation(s)
- Justine Guguin
- Université Claude Bernard Lyon 1, CNRS, INSERM, Centre de Recherche en Neurosciences de Lyon CRNL U1028 UMR5292, GENDEV, Bron, France
| | - Ting-Yu Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Silvestre Cuinat
- Université Claude Bernard Lyon 1, CNRS, INSERM, Centre de Recherche en Neurosciences de Lyon CRNL U1028 UMR5292, GENDEV, Bron, France
| | - Alicia Besson
- Université Claude Bernard Lyon 1, CNRS, INSERM, Centre de Recherche en Neurosciences de Lyon CRNL U1028 UMR5292, GENDEV, Bron, France
| | - Eloïse Bertiaux
- University of Geneva, Molecular and Cellular biology department, Sciences faculty, Geneva, Switzerland
| | - Lucile Boutaud
- INSERM UMR 1163, Institut Imagine, Université Paris Cité, Paris, France
| | - Nolan Ardito
- Université Claude Bernard Lyon 1, CNRS, INSERM, Centre de Recherche en Neurosciences de Lyon CRNL U1028 UMR5292, GENDEV, Bron, France
| | | | - Sara Cabet
- Service d’imagerie Pédiatrique et Fœtale, Hôpital Femme Mère Enfant, Hospices Civils de Lyon, Bron, France
- CNRS, Inserm, Physiopathologie et Génétique du Neurone et du Muscle, Institut NeuroMyoGène, Université de Lyon, Lyon, France
| | - Virginie Hamel
- University of Geneva, Molecular and Cellular biology department, Sciences faculty, Geneva, Switzerland
| | - Sophie Thomas
- INSERM UMR 1163, Institut Imagine, Université Paris Cité, Paris, France
| | - Bertrand Pain
- University of Lyon, Université de Lyon 1, INSERM, INRAE, Stem Cell and Brain Research Institute, U1208, USC1361, Bron, France
| | - Patrick Edery
- Université Claude Bernard Lyon 1, CNRS, INSERM, Centre de Recherche en Neurosciences de Lyon CRNL U1028 UMR5292, GENDEV, Bron, France
- Unité de génétique clinique et Centre de référence labellisé des Anomalies du Développement Sud-Est, Département de génétique, Hospices Civils de Lyon, Bron, France
| | - Audrey Putoux
- Université Claude Bernard Lyon 1, CNRS, INSERM, Centre de Recherche en Neurosciences de Lyon CRNL U1028 UMR5292, GENDEV, Bron, France
- Unité de génétique clinique et Centre de référence labellisé des Anomalies du Développement Sud-Est, Département de génétique, Hospices Civils de Lyon, Bron, France
| | - Tang K. Tang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Sylvie Mazoyer
- Université Claude Bernard Lyon 1, CNRS, INSERM, Centre de Recherche en Neurosciences de Lyon CRNL U1028 UMR5292, GENDEV, Bron, France
| | - Marion Delous
- Université Claude Bernard Lyon 1, CNRS, INSERM, Centre de Recherche en Neurosciences de Lyon CRNL U1028 UMR5292, GENDEV, Bron, France
| |
Collapse
|
3
|
Li N, Xu Y, Chen H, Lin J, AlAbdi L, Bekheirnia MR, Li G, Gofin Y, Bekheirnia N, Faqeih E, Chen L, Chang G, Tang J, Yao R, Yu T, Wang X, Fu W, Fu Q, Shen Y, Alkuraya FS, Machol K, Wang J. Bi-allelic variants in CEP295 cause Seckel-like syndrome presenting with primary microcephaly, developmental delay, intellectual disability, short stature, craniofacial and digital abnormalities. EBioMedicine 2024; 99:104940. [PMID: 38154379 PMCID: PMC10784679 DOI: 10.1016/j.ebiom.2023.104940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 12/13/2023] [Accepted: 12/13/2023] [Indexed: 12/30/2023] Open
Abstract
BACKGROUND Pathogenic variants in the centrosome protein (CEP) family have been implicated in primary microcephaly, Seckel syndrome, and classical ciliopathies. However, most CEP genes remain unlinked to specific Mendelian genetic diseases in humans. We sought to explore the roles of CEP295 in human pathology. METHODS Whole-exome sequencing was performed to screen for pathogenic variants in patients with severe microcephaly. Patient-derived fibroblasts and CEP295-depleted U2OS and RPE1 cells were used to clarify the underlying pathomechanisms, including centriole/centrosome development, cell cycle and proliferation changes, and ciliogenesis. Complementary experiments using CEP295 mRNA were performed to determine the pathogenicity of the identified missense variant. FINDINGS Here, we report bi-allelic variants of CEP295 in four children from two unrelated families, characterized by severe primary microcephaly, short stature, developmental delay, intellectual disability, facial deformities, and abnormalities of fingers and toes, suggesting a Seckel-like syndrome. Mechanistically, depletion of CEP295 resulted in a decrease in the numbers of centrioles and centrosomes and triggered p53-dependent G1 cell cycle arrest. Moreover, loss of CEP295 causes extensive primary ciliary defects in both patient-derived fibroblasts and RPE1 cells. The results from complementary experiments revealed that the wild-type CEP295, but not the mutant protein, can correct the developmental defects of the centrosome/centriole and cilia in the patient-derived skin fibroblasts. INTERPRETATION This study reports CEP295 as a causative gene of the syndromic microcephaly phenotype in humans. Our study also demonstrates that defects in CEP295 result in primary ciliary defects. FUNDING A full list of funding bodies that contributed to this study can be found under "Acknowledgments."
Collapse
Affiliation(s)
- Niu Li
- Department of Reproductive Genetics, International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China; Shanghai Key Laboratory of Embryo Original Diseases, International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; Department of Medical Genetics and Molecular Diagnostic Laboratory, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China; Faculty of Medical Laboratory Science, College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yufei Xu
- Department of Medical Genetics and Molecular Diagnostic Laboratory, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Hongzhu Chen
- Department of Medical Genetics and Molecular Diagnostic Laboratory, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Jingqi Lin
- Shanghai Key Laboratory of Embryo Original Diseases, International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Lama AlAbdi
- Department of Zoology, College of Science, King Saud University, Riyadh, 11533, Saudi Arabia
| | - Mir Reza Bekheirnia
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA; Texas Children's Hospital, Houston, TX, 77030, USA; Renal Section, Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Guoqiang Li
- Department of Reproductive Genetics, International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China; Shanghai Key Laboratory of Embryo Original Diseases, International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Yoel Gofin
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA; Texas Children's Hospital, Houston, TX, 77030, USA
| | - Nasim Bekheirnia
- Texas Children's Hospital, Houston, TX, 77030, USA; Renal Section, Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Eissa Faqeih
- Department of Pediatric Subspecialties, Children's Hospital, King Fahad Medical City, Riyadh, 11533, Saudi Arabia
| | - Lina Chen
- Department of Medical Genetics and Molecular Diagnostic Laboratory, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Guoying Chang
- Department of Endocrinology and Metabolism, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Jie Tang
- Department of Medical Genetics and Molecular Diagnostic Laboratory, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Ruen Yao
- Department of Medical Genetics and Molecular Diagnostic Laboratory, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China; Faculty of Medical Laboratory Science, College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Tingting Yu
- Department of Medical Genetics and Molecular Diagnostic Laboratory, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China; Faculty of Medical Laboratory Science, College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xiumin Wang
- Department of Endocrinology and Metabolism, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Wei Fu
- Institute of Pediatric Translational Medicine, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Qihua Fu
- Institute of Pediatric Translational Medicine, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China; Shanghai Key Laboratory of Clinical Molecular Diagnostics for Pediatrics, Shanghai, 200127, China
| | - Yiping Shen
- Department of Medical Genetics and Molecular Diagnostic Laboratory, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Fowzan S Alkuraya
- Department of Translational Genomics, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, 11211, Saudi Arabia
| | - Keren Machol
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA; Texas Children's Hospital, Houston, TX, 77030, USA.
| | - Jian Wang
- Shanghai Key Laboratory of Embryo Original Diseases, International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; Department of Medical Genetics and Molecular Diagnostic Laboratory, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China; Faculty of Medical Laboratory Science, College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Shanghai Key Laboratory of Clinical Molecular Diagnostics for Pediatrics, Shanghai, 200127, China.
| |
Collapse
|
4
|
Romano F, Amadori E, Madia F, Severino M, Capra V, Rizzo R, Barone R, Corradi B, Maragliano L, Shams Nosrati MS, Falace A, Striano P, Zara F, Scala M. Case Report: Novel biallelic moderately damaging variants in RTTN in a patient with cerebellar dysplasia. Front Pediatr 2023; 11:1326552. [PMID: 38178912 PMCID: PMC10764497 DOI: 10.3389/fped.2023.1326552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 12/05/2023] [Indexed: 01/06/2024] Open
Abstract
Rotatin, encoded by the RTTN gene, is a centrosomal protein with multiple, emerging functions, including left-right specification, ciliogenesis, and neuronal migration. Recessive variants in RTTN are associated with a neurodevelopmental disorder with microcephaly and malformations of cortical development known as "Microcephaly, short stature, and polymicrogyria with seizures" (MSSP, MIM #614833). Affected individuals show a wide spectrum of clinical manifestations like intellectual disability, poor/absent speech, short stature, microcephaly, and congenital malformations. Here, we report a subject showing a distinctive neuroradiological phenotype and harboring novel biallelic variants in RTTN: the c.5500A>G, p.(Asn1834Asp), (dbSNP: rs200169343, ClinVar ID:1438510) and c.19A>G, p.(Ile7Val), (dbSNP: rs201165599, ClinVar ID:1905275) variants. In particular brain magnetic resonance imaging (MRI) showed a peculiar pattern, with cerebellar hypo-dysplasia, and multiple arachnoid cysts in the lateral cerebello-medullary cisterns, in addition to left Meckel cave. Thus, we compare his phenotypic features with current literature, speculating a possible role of newly identified RTTN variants in his clinical picture, and supporting a relevant variability in this emerging condition.
Collapse
Affiliation(s)
- Ferruccio Romano
- Genomics and Clinical Genetics Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Elisabetta Amadori
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, Università Degli Studi di Genova, Genoa, Italy
- Child Neuropsichiatry Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Francesca Madia
- Medical Genetics Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | | | - Valeria Capra
- Genomics and Clinical Genetics Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Renata Rizzo
- Child Neuropsychiatry Unit, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Rita Barone
- Child Neuropsychiatry Unit, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Beatrice Corradi
- Department of Experimental Medicine, University of Genova, Genova, Italy
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy
| | - Luca Maragliano
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genova, Italy
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona, Italy
| | | | - Antonio Falace
- Medical Genetics Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Pasquale Striano
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, Università Degli Studi di Genova, Genoa, Italy
- Pediatric Neurology and Muscular Diseases Unit, IRCCS, Genoa, Italy
| | - Federico Zara
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, Università Degli Studi di Genova, Genoa, Italy
- Medical Genetics Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Marcello Scala
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, Università Degli Studi di Genova, Genoa, Italy
- Medical Genetics Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| |
Collapse
|
5
|
Chun YW, Miyamoto M, Williams CH, Neitzel LR, Silver-Isenstadt M, Cadar AG, Fuller DT, Fong DC, Liu H, Lease R, Kim S, Katagiri M, Durbin MD, Wang KC, Feaster TK, Sheng CC, Neely MD, Sreenivasan U, Cortes-Gutierrez M, Finn AV, Schot R, Mancini GMS, Ament SA, Ess KC, Bowman AB, Han Z, Bichell DP, Su YR, Hong CC. Impaired Reorganization of Centrosome Structure Underlies Human Infantile Dilated Cardiomyopathy. Circulation 2023; 147:1291-1303. [PMID: 36970983 PMCID: PMC10133173 DOI: 10.1161/circulationaha.122.060985] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 02/22/2023] [Indexed: 03/29/2023]
Abstract
BACKGROUND During cardiomyocyte maturation, the centrosome, which functions as a microtubule organizing center in cardiomyocytes, undergoes dramatic structural reorganization where its components reorganize from being localized at the centriole to the nuclear envelope. This developmentally programmed process, referred to as centrosome reduction, has been previously associated with cell cycle exit. However, understanding of how this process influences cardiomyocyte cell biology, and whether its disruption results in human cardiac disease, remains unknown. We studied this phenomenon in an infant with a rare case of infantile dilated cardiomyopathy (iDCM) who presented with left ventricular ejection fraction of 18% and disrupted sarcomere and mitochondria structure. METHODS We performed an analysis beginning with an infant who presented with a rare case of iDCM. We derived induced pluripotent stem cells from the patient to model iDCM in vitro. We performed whole exome sequencing on the patient and his parents for causal gene analysis. CRISPR/Cas9-mediated gene knockout and correction in vitro were used to confirm whole exome sequencing results. Zebrafish and Drosophila models were used for in vivo validation of the causal gene. Matrigel mattress technology and single-cell RNA sequencing were used to characterize iDCM cardiomyocytes further. RESULTS Whole exome sequencing and CRISPR/Cas9 gene knockout/correction identified RTTN, the gene encoding the centrosomal protein RTTN (rotatin), as the causal gene underlying the patient's condition, representing the first time a centrosome defect has been implicated in a nonsyndromic dilated cardiomyopathy. Genetic knockdowns in zebrafish and Drosophila confirmed an evolutionarily conserved requirement of RTTN for cardiac structure and function. Single-cell RNA sequencing of iDCM cardiomyocytes showed impaired maturation of iDCM cardiomyocytes, which underlie the observed cardiomyocyte structural and functional deficits. We also observed persistent localization of the centrosome at the centriole, contrasting with expected programmed perinuclear reorganization, which led to subsequent global microtubule network defects. In addition, we identified a small molecule that restored centrosome reorganization and improved the structure and contractility of iDCM cardiomyocytes. CONCLUSIONS This study is the first to demonstrate a case of human disease caused by a defect in centrosome reduction. We also uncovered a novel role for RTTN in perinatal cardiac development and identified a potential therapeutic strategy for centrosome-related iDCM. Future study aimed at identifying variants in centrosome components may uncover additional contributors to human cardiac disease.
Collapse
Affiliation(s)
- Young Wook Chun
- Division of Cardiovascular Medicine, Department of Medicine, University of Maryland Medical Center, Baltimore, MD 21201
| | - Matthew Miyamoto
- Division of Cardiovascular Medicine, Department of Medicine, University of Maryland Medical Center, Baltimore, MD 21201
| | - Charles H. Williams
- Division of Cardiovascular Medicine, Department of Medicine, University of Maryland Medical Center, Baltimore, MD 21201
| | - Leif R. Neitzel
- Division of Cardiovascular Medicine, Department of Medicine, University of Maryland Medical Center, Baltimore, MD 21201
| | - Maya Silver-Isenstadt
- Division of Cardiovascular Medicine, Department of Medicine, University of Maryland Medical Center, Baltimore, MD 21201
| | - Adrian G. Cadar
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN 37201
| | - Daniela T. Fuller
- Division of Cardiovascular Medicine, Department of Medicine, University of Maryland Medical Center, Baltimore, MD 21201
| | - Daniel C. Fong
- Division of Cardiovascular Medicine, Department of Medicine, University of Maryland Medical Center, Baltimore, MD 21201
| | - Hanhan Liu
- Division of Cardiovascular Medicine, Department of Medicine, University of Maryland Medical Center, Baltimore, MD 21201
| | - Robert Lease
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Sungseek Kim
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN 37201
| | - Mikako Katagiri
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN 37201
| | - Matthew D. Durbin
- Division of Neonatology-Perinatology, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 26202
| | - Kuo-Chen Wang
- Division of Cardiovascular Medicine, Department of Medicine, University of Maryland Medical Center, Baltimore, MD 21201
| | - Tromondae K. Feaster
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN 37201
| | - Calvin C. Sheng
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN 37201
| | - M. Diana Neely
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37201
| | - Urmila Sreenivasan
- Division of Cardiovascular Medicine, Department of Medicine, University of Maryland Medical Center, Baltimore, MD 21201
| | - Marcia Cortes-Gutierrez
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Aloke V. Finn
- Division of Cardiovascular Medicine, Department of Medicine, University of Maryland Medical Center, Baltimore, MD 21201
| | - Rachel Schot
- Division of Neonatology-Perinatology, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 26202
| | - Grazia M. S. Mancini
- Department of Clinical Genetics, Erasmus University Medical Center (Erasmus MC), P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Seth A. Ament
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Kevin C. Ess
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN37201
| | - Aaron B. Bowman
- School of Health Sciences, Purdue University, West Lafayette, IN 47906
| | - Zhe Han
- Division of Cardiovascular Medicine, Department of Medicine, University of Maryland Medical Center, Baltimore, MD 21201
| | - David P. Bichell
- Department of Pediatric Cardiac Surgery, Vanderbilt University Medical Center, Nashville, TN 37201
| | - Yan Ru Su
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN 37201
| | - Charles C. Hong
- Division of Cardiovascular Medicine, Department of Medicine, University of Maryland Medical Center, Baltimore, MD 21201
| |
Collapse
|
6
|
Dawood M, Akay G, Mitani T, Marafi D, Fatih JM, Gezdirici A, Najmabadi H, Kahrizi K, Punetha J, Grochowski CM, Du H, Jolly A, Li H, Coban-Akdemir Z, Sedlazeck FJ, Hunter JV, Jhangiani SN, Muzny D, Pehlivan D, Posey JE, Carvalho CM, Gibbs RA, Lupski JR. A biallelic frameshift indel in PPP1R35 as a cause of primary microcephaly. Am J Med Genet A 2023; 191:794-804. [PMID: 36598158 PMCID: PMC9928800 DOI: 10.1002/ajmg.a.63080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 11/05/2022] [Accepted: 12/01/2022] [Indexed: 01/05/2023]
Abstract
Protein phosphatase 1 regulatory subunit 35 (PPP1R35) encodes a centrosomal protein required for recruiting microtubule-binding elongation machinery. Several proteins in this centriole biogenesis pathway correspond to established primary microcephaly (MCPH) genes, and multiple model organism studies hypothesize PPP1R35 as a candidate MCPH gene. Here, using exome sequencing (ES) and family-based rare variant analyses, we report a homozygous, frameshifting indel deleting the canonical stop codon in the last exon of PPP1R35 [Chr7: c.753_*3delGGAAGCGTAGACCinsCG (p.Trp251Cysfs*22)]; the variant allele maps in a 3.7 Mb block of absence of heterozygosity (AOH) in a proband with severe MCPH (-4.3 SD at birth, -6.1 SD by 42 months), pachygyria, and global developmental delay from a consanguineous Turkish kindred. Droplet digital PCR (ddPCR) confirmed mutant mRNA expression in fibroblasts. In silico prediction of the translation of mutant PPP1R35 is expected to be elongated by 18 amino acids before encountering a downstream stop codon. This complex indel allele is absent in public databases (ClinVar, gnomAD, ARIC, 1000 genomes) and our in-house database of 14,000+ exomes including 1800+ Turkish exomes supporting predicted pathogenicity. Comprehensive literature searches for PPP1R35 variants yielded two probands affected with severe microcephaly (-15 SD and -12 SD) with the same homozygous indel from a single, consanguineous, Iranian family from a cohort of 404 predominantly Iranian families. The lack of heterozygous cases in two large cohorts representative of the genetic background of these two families decreased our suspicion of a founder allele and supports the contention of a recurrent mutation. We propose two potential secondary structure mutagenesis models for the origin of this variant allele mediated by hairpin formation between complementary GC rich segments flanking the stop codon via secondary structure mutagenesis.
Collapse
Affiliation(s)
- Moez Dawood
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, 77030, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, 77030, USA
- Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Gulsen Akay
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, 77030, USA
| | - Tadahiro Mitani
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, 77030, USA
| | - Dana Marafi
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, 77030, USA
- Department of Pediatrics, Faculty of Medicine, Kuwait University, P.O. Box 24923, 13110 Safat, Kuwait
| | - Jawid M. Fatih
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, 77030, USA
| | - Alper Gezdirici
- Department of Medical Genetics, Basaksehir Cam and Sakura City Hospital, Istanbul 34480, Turkey
| | - Hossein Najmabadi
- Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Kimia Kahrizi
- Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Jaya Punetha
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, 77030, USA
| | | | - Haowei Du
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, 77030, USA
| | - Angad Jolly
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, 77030, USA
- Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, 77030, USA
| | - He Li
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, 77030, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, 77030, USA
| | - Zeynep Coban-Akdemir
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, 77030, USA
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Fritz J. Sedlazeck
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, 77030, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, 77030, USA
| | - Jill V. Hunter
- Department of Radiology, Baylor College of Medicine, Houston, Texas, 77030, USA
- E.B. Singleton Department of Pediatric Radiology, Texas Children’s Hospital, Houston, Texas, 77030, USA
- Section of Pediatric Neurology and Developmental Neuroscience, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, 77030, USA
| | - Shalini N. Jhangiani
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, 77030, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, 77030, USA
| | - Donna Muzny
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, 77030, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, 77030, USA
| | - Davut Pehlivan
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, 77030, USA
- Section of Pediatric Neurology and Developmental Neuroscience, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, 77030, USA
- Texas Children’s Hospital, Houston, Texas, 77030, USA
| | - Jennifer E. Posey
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, 77030, USA
| | - Claudia M.B. Carvalho
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, 77030, USA
- Pacific Northwest Research Institute, Seattle, WA, 98122, USA
| | - Richard A. Gibbs
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, 77030, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, 77030, USA
| | - James R. Lupski
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, 77030, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, 77030, USA
- Texas Children’s Hospital, Houston, Texas, 77030, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030
| |
Collapse
|
7
|
Analysis of recent shared ancestry in a familial cohort identifies coding and noncoding autism spectrum disorder variants. NPJ Genom Med 2022; 7:13. [PMID: 35190550 PMCID: PMC8861044 DOI: 10.1038/s41525-022-00284-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 01/21/2022] [Indexed: 12/02/2022] Open
Abstract
Autism spectrum disorder (ASD) is a collection of neurodevelopmental disorders characterized by deficits in social communication and restricted, repetitive patterns of behavior or interests. ASD is highly heritable, but genetically and phenotypically heterogeneous, reducing the power to identify causative genes. We performed whole genome sequencing (WGS) in an ASD cohort of 68 individuals from 22 families enriched for recent shared ancestry. We identified an average of 3.07 million variants per genome, of which an average of 112,512 were rare. We mapped runs of homozygosity (ROHs) in affected individuals and found an average genomic homozygosity of 9.65%, consistent with expectations for multiple generations of consanguineous unions. We identified potentially pathogenic rare exonic or splice site variants in 12 known (including KMT2C, SCN1A, SPTBN1, SYNE1, ZNF292) and 12 candidate (including CHD5, GRB10, PPP1R13B) ASD genes. Furthermore, we annotated noncoding variants in ROHs with brain-specific regulatory elements and identified putative disease-causing variants within brain-specific promoters and enhancers for 5 known ASD and neurodevelopmental disease genes (ACTG1, AUTS2, CTNND2, CNTNAP4, SPTBN4). We also identified copy number variants in two known ASD and neurodevelopmental disease loci in two affected individuals. In total we identified potentially etiological variants in known ASD or neurodevelopmental disease genes for ~61% (14/23) of affected individuals. We combined WGS with homozygosity mapping and regulatory element annotations to identify candidate ASD variants. Our analyses add to the growing number of ASD genes and variants and emphasize the importance of leveraging recent shared ancestry to map disease variants in complex neurodevelopmental disorders.
Collapse
|
8
|
Duerinckx S, Désir J, Perazzolo C, Badoer C, Jacquemin V, Soblet J, Maystadt I, Tunca Y, Blaumeiser B, Ceulemans B, Courtens W, Debray F, Destree A, Devriendt K, Jansen A, Keymolen K, Lederer D, Loeys B, Meuwissen M, Moortgat S, Mortier G, Nassogne M, Sekhara T, Van Coster R, Van Den Ende J, Van der Aa N, Van Esch H, Vanakker O, Verhelst H, Vilain C, Weckhuysen S, Passemard S, Verloes A, Aeby A, Deconinck N, Van Bogaert P, Pirson I, Abramowicz M. Phenotypes and genotypes in non-consanguineous and consanguineous primary microcephaly: High incidence of epilepsy. Mol Genet Genomic Med 2021; 9:e1768. [PMID: 34402213 PMCID: PMC8457702 DOI: 10.1002/mgg3.1768] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 05/06/2021] [Accepted: 07/03/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Primary microcephaly (PM) is defined as a significant reduction in occipitofrontal circumference (OFC) of prenatal onset. Clinical and genetic heterogeneity of PM represents a diagnostic challenge. METHODS We performed detailed phenotypic and genomic analyses in a large cohort (n = 169) of patients referred for PM and could establish a molecular diagnosis in 38 patients. RESULTS Pathogenic variants in ASPM and WDR62 were the most frequent causes in non-consanguineous patients in our cohort. In consanguineous patients, microarray and targeted gene panel analyses reached a diagnostic yield of 67%, which contrasts with a much lower rate in non-consanguineous patients (9%). Our series includes 11 novel pathogenic variants and we identify novel candidate genes including IGF2BP3 and DNAH2. We confirm the progression of microcephaly over time in affected children. Epilepsy was an important associated feature in our PM cohort, affecting 34% of patients with a molecular confirmation of the PM diagnosis, with various degrees of severity and seizure types. CONCLUSION Our findings will help to prioritize genomic investigations, accelerate molecular diagnoses, and improve the management of PM patients.
Collapse
Affiliation(s)
- Sarah Duerinckx
- Institut de Recherche Interdisciplinaire en Biologie Humaine et moléculaireUniversité Libre de BruxellesBrusselsBelgium
| | - Julie Désir
- Centre de Génétique HumaineInstitut de Pathologie et de GénétiqueGosseliesBelgium
| | - Camille Perazzolo
- Institut de Recherche Interdisciplinaire en Biologie Humaine et moléculaireUniversité Libre de BruxellesBrusselsBelgium
| | - Cindy Badoer
- Department of GeneticsHôpital ErasmeULB Center of Human GeneticsUniversité Libre de BruxellesBrusselsBelgium
| | - Valérie Jacquemin
- Institut de Recherche Interdisciplinaire en Biologie Humaine et moléculaireUniversité Libre de BruxellesBrusselsBelgium
| | - Julie Soblet
- Department of GeneticsHôpital ErasmeULB Center of Human GeneticsUniversité Libre de BruxellesBrusselsBelgium
- Hôpital Universitaire des Enfants Reine Fabiola (HUDERF)Université Libre de BruxellesBrusselsBelgium
| | - Isabelle Maystadt
- Centre de Génétique HumaineInstitut de Pathologie et de GénétiqueGosseliesBelgium
| | - Yusuf Tunca
- Department of Medical GeneticsGülhane Faculty of Medicine & Gülhane Training and Research HospitalUniversity of Health Sciences TurkeyAnkaraTurkey
| | | | | | | | | | - Anne Destree
- Centre de Génétique HumaineInstitut de Pathologie et de GénétiqueGosseliesBelgium
| | | | - Anna Jansen
- Universitair Ziekenhuis Brussel (UZ Brussel)Centrum Medische GeneticaUniversiteit Brussel (VUB)BrusselsBelgium
| | - Kathelijn Keymolen
- Universitair Ziekenhuis Brussel (UZ Brussel)Centrum Medische GeneticaUniversiteit Brussel (VUB)BrusselsBelgium
| | - Damien Lederer
- Centre de Génétique HumaineInstitut de Pathologie et de GénétiqueGosseliesBelgium
| | - Bart Loeys
- University and University Hospital of AntwerpAntwerpBelgium
| | | | - Stéphanie Moortgat
- Centre de Génétique HumaineInstitut de Pathologie et de GénétiqueGosseliesBelgium
| | - Geert Mortier
- University and University Hospital of AntwerpAntwerpBelgium
| | | | | | | | | | | | - Hilde Van Esch
- Center for Human GeneticsUniversity Hospitals LeuvenLeuvenBelgium
| | | | | | - Catheline Vilain
- Department of GeneticsHôpital ErasmeULB Center of Human GeneticsUniversité Libre de BruxellesBrusselsBelgium
- Hôpital Universitaire des Enfants Reine Fabiola (HUDERF)Université Libre de BruxellesBrusselsBelgium
| | | | | | - Alain Verloes
- Department of GeneticsAPHPRobert Debré University HospitalParisFrance
| | - Alec Aeby
- Hôpital Universitaire des Enfants Reine Fabiola (HUDERF)Université Libre de BruxellesBrusselsBelgium
| | - Nicolas Deconinck
- Hôpital Universitaire des Enfants Reine Fabiola (HUDERF)Université Libre de BruxellesBrusselsBelgium
| | | | - Isabelle Pirson
- Institut de Recherche Interdisciplinaire en Biologie Humaine et moléculaireUniversité Libre de BruxellesBrusselsBelgium
| | - Marc Abramowicz
- Institut de Recherche Interdisciplinaire en Biologie Humaine et moléculaireUniversité Libre de BruxellesBrusselsBelgium
- Department of Genetic Medicine and DevelopmentUniversity of GenevaGenèveSwitzerland
| |
Collapse
|
9
|
Human Microcephaly Protein RTTN Is Required for Proper Mitotic Progression and Correct Spindle Position. Cells 2021; 10:cells10061441. [PMID: 34207628 PMCID: PMC8229632 DOI: 10.3390/cells10061441] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/28/2021] [Accepted: 06/07/2021] [Indexed: 01/16/2023] Open
Abstract
Autosomal recessive primary microcephaly (MCPH) is a complex neurodevelopmental disorder characterized by a small brain size with mild to moderate intellectual disability. We previously demonstrated that human microcephaly RTTN played an important role in regulating centriole duplication during interphase, but the role of RTTN in mitosis is not fully understood. Here, we show that RTTN is required for normal mitotic progression and correct spindle position. The depletion of RTTN induces the dispersion of the pericentriolar protein γ-tubulin and multiple mitotic abnormalities, including monopolar, abnormal bipolar, and multipolar spindles. Importantly, the loss of RTTN altered NuMA/p150Glued congression to the spindle poles, perturbed NuMA cortical localization, and reduced the number and the length of astral microtubules. Together, our results provide a new insight into how RTTN functions in mitosis.
Collapse
|
10
|
Imran Naseer M, Abdulrahman Abdulkareem A, Yousef Muthaffar O, Chaudhary AG. Exome sequencing reveled a compound heterozygous mutations in RTTN gene causing developmental delay and primary microcephaly. Saudi J Biol Sci 2021; 28:2824-2829. [PMID: 34012324 PMCID: PMC8116967 DOI: 10.1016/j.sjbs.2021.02.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 01/26/2021] [Accepted: 02/01/2021] [Indexed: 11/27/2022] Open
Abstract
RTTN (Rotatin) (OMIM 614833) is a large centrosomal protein coding gene. RTTN mutations are responsible for syndromic forms of malformation of brain development, leading to polymicrogyria, microcephaly, primordial dwarfism, seizure along with many other malformations. In this study we have identified a compound heterozygous mutation in RTTN gene having NM_173630 c.5225A > G p.His1742Arg in exon 39 and NM_173630 c.6038G > T p.Cys2013Phe in exon 45 of a consanguineous Saudi family leading to brain malformation, seizure, developmental delay, dysmorphic feature and microcephaly. Whole exome sequencing (WES) techniques was used to identify the causative mutation in the affected members of the family. WES data analysis was done and obtained data were further confirmed by using Sanger sequencing analysis. Moreover, the mutation was ruled out in 100 healthy control from normal population. To the best of our knowledge the novel compound heterozygous mutation observed in this study is the first report from Saudi Arabia. The identified compound heterozygous mutation will further explain the role of RTTN gene in development of microcephaly and neurodevelopmental disorders.
Collapse
Affiliation(s)
- Muhammad Imran Naseer
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, 21589 Jeddah, Saudi Arabia
| | - Angham Abdulrahman Abdulkareem
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | | | - Adeel G Chaudhary
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, 21589 Jeddah, Saudi Arabia.,Center for Innovation in Personalized Medicine, King Abdulaziz University, 21589 Jeddah, Saudi Arabia
| |
Collapse
|
11
|
Liu S, Trupiano MX, Simon J, Guo J, Anton ES. The essential role of primary cilia in cerebral cortical development and disorders. Curr Top Dev Biol 2021; 142:99-146. [PMID: 33706927 DOI: 10.1016/bs.ctdb.2020.11.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Primary cilium, first described in the 19th century in different cell types and organisms by Alexander Ecker, Albert Kolliker, Aleksandr Kowalevsky, Paul Langerhans, and Karl Zimmermann (Ecker, 1844; Kolliker, 1854; Kowalevsky, 1867; Langerhans, 1876; Zimmermann, 1898), play an essential modulatory role in diverse aspects of nervous system development and function. The primary cilium, sometimes referred to as the cell's 'antennae', can receive wide ranging inputs from cellular milieu, including morphogens, growth factors, neuromodulators, and neurotransmitters. Its unique structural and functional organization bequeaths it the capacity to hyper-concentrate signaling machinery in a restricted cellular domain approximately one-thousandth the volume of cell soma. Thus enabling it to act as a signaling hub that integrates diverse developmental and homestatic information from cellular milieu to regulate the development and function of neural cells. Dysfunction of primary cilia contributes to the pathophysiology of several brain malformations, intellectual disabilities, epilepsy, and psychiatric disorders. This review focuses on the most essential contributions of primary cilia to cerebral cortical development and function, in the context of neurodevelopmental disorders and malformations. It highlights the recent progress made in identifying the mechanisms underlying primary cilia's role in cortical progenitors, neurons and glia, in health and disease. A future challenge will be to translate these insights and advances into effective clinical treatments for ciliopathies.
Collapse
Affiliation(s)
- Siling Liu
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC, United States
| | - Mia X Trupiano
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC, United States
| | - Jeremy Simon
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC, United States
| | - Jiami Guo
- Hotchkiss Brain Institute, Alberta Children's Hospital Research Institute, and the Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB, Canada
| | - E S Anton
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC, United States.
| |
Collapse
|
12
|
Cody JD. The Consequences of Abnormal Gene Dosage: Lessons from Chromosome 18. Trends Genet 2020; 36:764-776. [PMID: 32660784 DOI: 10.1016/j.tig.2020.06.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 06/17/2020] [Accepted: 06/18/2020] [Indexed: 12/18/2022]
Abstract
Accurate interpretation of genomic copy number variation (CNV) remains a challenge and has important consequences for both congenital and late-onset disease. Hemizygosity dosage characterization of the genes on chromosome 18 reveals a spectrum of outcomes ranging from no clinical effect, to risk factors for disease, to both low- and high-penetrance disease. These data are important for accurate and predictive clinical management. Additionally, the potential mechanisms of reduced penetrance due to dosage compensation are discussed as a key to understanding avenues for potential treatment. This review describes the chromosome 18 findings, and discusses the molecular mechanisms that allow haploinsufficiency, reduced penetrance, and dosage compensation.
Collapse
Affiliation(s)
- Jannine DeMars Cody
- Department of Pediatrics, University of Texas Health San Antonio, San Antonio, TX 78229, USA; Chromosome 18 Registry and Research Society, San Antonio, TX 78229, USA.
| |
Collapse
|
13
|
Lin YN, Lee YS, Li SK, Tang TK. Loss of CPAP in developing mouse brain and its functional implication for human primary microcephaly. J Cell Sci 2020; 133:jcs243592. [PMID: 32501282 DOI: 10.1242/jcs.243592] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 05/07/2020] [Indexed: 12/21/2022] Open
Abstract
Primary microcephaly (MCPH) is a neurodevelopmental disorder characterized by small brain size with mental retardation. CPAP (also known as CENPJ), a known microcephaly-associated gene, plays a key role in centriole biogenesis. Here, we generated a previously unreported conditional knockout allele in the mouse Cpap gene. Our results showed that conditional Cpap deletion in the central nervous system preferentially induces formation of monopolar spindles in radial glia progenitors (RGPs) at around embryonic day 14.5 and causes robust apoptosis that severely disrupts embryonic brains. Interestingly, microcephalic brains with reduced apoptosis are detected in conditional Cpap gene-deleted mice that lose only one allele of p53 (also known as Trp53), while simultaneous removal of p53 and Cpap rescues RGP death. Furthermore, Cpap deletion leads to cilia loss, RGP mislocalization, junctional integrity disruption, massive heterotopia and severe cerebellar hypoplasia. Together, these findings indicate that complete CPAP loss leads to severe and complex phenotypes in developing mouse brain, and provide new insights into the causes of MCPH.
Collapse
Affiliation(s)
- Yi-Nan Lin
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529 Taiwan
| | - Ying-Shan Lee
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529 Taiwan
| | - Shu-Kuei Li
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529 Taiwan
| | - Tang K Tang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529 Taiwan
| |
Collapse
|
14
|
Gabriel E, Ramani A, Altinisik N, Gopalakrishnan J. Human Brain Organoids to Decode Mechanisms of Microcephaly. Front Cell Neurosci 2020; 14:115. [PMID: 32457578 PMCID: PMC7225330 DOI: 10.3389/fncel.2020.00115] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 04/09/2020] [Indexed: 12/13/2022] Open
Abstract
Brain organoids are stem cell-based self-assembling 3D structures that recapitulate early events of human brain development. Recent improvements with patient-specific 3D brain organoids have begun to elucidate unprecedented details of the defective mechanisms that cause neurodevelopmental disorders of congenital and acquired microcephaly. In particular, brain organoids derived from primary microcephaly patients have uncovered mechanisms that deregulate neural stem cell proliferation, maintenance, and differentiation. Not only did brain organoids reveal unknown aspects of neurogenesis but also have illuminated surprising roles of cellular structures of centrosomes and primary cilia in regulating neurogenesis during brain development. Here, we discuss how brain organoids have started contributing to decoding the complexities of microcephaly, which are unlikely to be identified in the existing non-human models. Finally, we discuss the yet unresolved questions and challenges that can be addressed with the use of brain organoids as in vitro models of neurodevelopmental disorders.
Collapse
Affiliation(s)
- Elke Gabriel
- Laboratory for Centrosome and Cytoskeleton Biology, Institute für Humangenetik, Universitätsklinikum Düsseldorf, Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Anand Ramani
- Laboratory for Centrosome and Cytoskeleton Biology, Institute für Humangenetik, Universitätsklinikum Düsseldorf, Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Nazlican Altinisik
- Laboratory for Centrosome and Cytoskeleton Biology, Institute für Humangenetik, Universitätsklinikum Düsseldorf, Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Jay Gopalakrishnan
- Laboratory for Centrosome and Cytoskeleton Biology, Institute für Humangenetik, Universitätsklinikum Düsseldorf, Heinrich-Heine-Universität, Düsseldorf, Germany
| |
Collapse
|
15
|
Vandervore LV, Schot R, Kasteleijn E, Oegema R, Stouffs K, Gheldof A, Grochowska MM, van der Sterre MLT, van Unen LMA, Wilke M, Elfferich P, van der Spek PJ, Heijsman D, Grandone A, Demmers JAA, Dekkers DHW, Slotman JA, Kremers GJ, Schaaf GJ, Masius RG, van Essen AJ, Rump P, van Haeringen A, Peeters E, Altunoglu U, Kalayci T, Poot RA, Dobyns WB, Bahi-Buisson N, Verheijen FW, Jansen AC, Mancini GMS. Heterogeneous clinical phenotypes and cerebral malformations reflected by rotatin cellular dynamics. Brain 2019; 142:867-884. [PMID: 30879067 PMCID: PMC6439326 DOI: 10.1093/brain/awz045] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 11/26/2018] [Accepted: 01/07/2019] [Indexed: 12/16/2022] Open
Abstract
Recessive mutations in RTTN, encoding the protein rotatin, were originally identified as cause of polymicrogyria, a cortical malformation. With time, a wide variety of other brain malformations has been ascribed to RTTN mutations, including primary microcephaly. Rotatin is a centrosomal protein possibly involved in centriolar elongation and ciliogenesis. However, the function of rotatin in brain development is largely unknown and the molecular disease mechanism underlying cortical malformations has not yet been elucidated. We performed both clinical and cell biological studies, aimed at clarifying rotatin function and pathogenesis. Review of the 23 published and five unpublished clinical cases and genomic mutations, including the effect of novel deep intronic pathogenic mutations on RTTN transcripts, allowed us to extrapolate the core phenotype, consisting of intellectual disability, short stature, microcephaly, lissencephaly, periventricular heterotopia, polymicrogyria and other malformations. We show that the severity of the phenotype is related to residual function of the protein, not only the level of mRNA expression. Skin fibroblasts from eight affected individuals were studied by high resolution immunomicroscopy and flow cytometry, in parallel with in vitro expression of RTTN in HEK293T cells. We demonstrate that rotatin regulates different phases of the cell cycle and is mislocalized in affected individuals. Mutant cells showed consistent and severe mitotic failure with centrosome amplification and multipolar spindle formation, leading to aneuploidy and apoptosis, which could relate to depletion of neuronal progenitors often observed in microcephaly. We confirmed the role of rotatin in functional and structural maintenance of primary cilia and determined that the protein localized not only to the basal body, but also to the axoneme, proving the functional interconnectivity between ciliogenesis and cell cycle progression. Proteomics analysis of both native and exogenous rotatin uncovered that rotatin interacts with the neuronal (non-muscle) myosin heavy chain subunits, motors of nucleokinesis during neuronal migration, and in human induced pluripotent stem cell-derived bipolar mature neurons rotatin localizes at the centrosome in the leading edge. This illustrates the role of rotatin in neuronal migration. These different functions of rotatin explain why RTTN mutations can lead to heterogeneous cerebral malformations, both related to proliferation and migration defects.
Collapse
Affiliation(s)
- Laura V Vandervore
- Department of Clinical Genetics, Erasmus University Medical Center (Erasmus MC), CA Rotterdam, The Netherlands.,Neurogenetics Research Group, Research Cluster Reproduction, Genetics and Regenerative Medicine, Vrije Universiteit Brussel, Brussels, Belgium.,Center for Medical Genetics, UZ Brussel, Brussels, Belgium
| | - Rachel Schot
- Department of Clinical Genetics, Erasmus University Medical Center (Erasmus MC), CA Rotterdam, The Netherlands
| | - Esmee Kasteleijn
- Department of Clinical Genetics, Erasmus University Medical Center (Erasmus MC), CA Rotterdam, The Netherlands
| | - Renske Oegema
- Department of Clinical Genetics, Erasmus University Medical Center (Erasmus MC), CA Rotterdam, The Netherlands.,Department of Pathology, Clinical Bio-informatics, Erasmus University Medical Center (Erasmus MC), CA Rotterdam, The Netherlands
| | - Katrien Stouffs
- Neurogenetics Research Group, Research Cluster Reproduction, Genetics and Regenerative Medicine, Vrije Universiteit Brussel, Brussels, Belgium.,Center for Medical Genetics, UZ Brussel, Brussels, Belgium
| | - Alexander Gheldof
- Neurogenetics Research Group, Research Cluster Reproduction, Genetics and Regenerative Medicine, Vrije Universiteit Brussel, Brussels, Belgium.,Center for Medical Genetics, UZ Brussel, Brussels, Belgium
| | - Martyna M Grochowska
- Department of Clinical Genetics, Erasmus University Medical Center (Erasmus MC), CA Rotterdam, The Netherlands
| | - Marianne L T van der Sterre
- Department of Clinical Genetics, Erasmus University Medical Center (Erasmus MC), CA Rotterdam, The Netherlands
| | - Leontine M A van Unen
- Department of Clinical Genetics, Erasmus University Medical Center (Erasmus MC), CA Rotterdam, The Netherlands
| | - Martina Wilke
- Department of Clinical Genetics, Erasmus University Medical Center (Erasmus MC), CA Rotterdam, The Netherlands
| | - Peter Elfferich
- Department of Clinical Genetics, Erasmus University Medical Center (Erasmus MC), CA Rotterdam, The Netherlands
| | - Peter J van der Spek
- Dipartimento della Donna, del Bambino, di Chirurgia Generale e Specialistica, Seconda Università degli studi della Campania "L. Vanvitelli", Naples, Italy
| | - Daphne Heijsman
- Department of Clinical Genetics, Erasmus University Medical Center (Erasmus MC), CA Rotterdam, The Netherlands.,Dipartimento della Donna, del Bambino, di Chirurgia Generale e Specialistica, Seconda Università degli studi della Campania "L. Vanvitelli", Naples, Italy
| | - Anna Grandone
- Department of Molecular Genetics, Proteomics Center, Erasmus University Medical Center (Erasmus MC), CA Rotterdam, The Netherlands
| | - Jeroen A A Demmers
- Department of Pathology, Optical Imaging Center, Erasmus University Medical Center (Erasmus MC), CA Rotterdam, The Netherlands
| | - Dick H W Dekkers
- Department of Pathology, Optical Imaging Center, Erasmus University Medical Center (Erasmus MC), CA Rotterdam, The Netherlands
| | - Johan A Slotman
- Center for Lysosomal and Metabolic Diseases, Erasmus Medical Center (Erasmus MC), 3015 CN Rotterdam, The Netherlands
| | - Gert-Jan Kremers
- Center for Lysosomal and Metabolic Diseases, Erasmus Medical Center (Erasmus MC), 3015 CN Rotterdam, The Netherlands
| | - Gerben J Schaaf
- Department of Clinical Genetics, Erasmus University Medical Center (Erasmus MC), CA Rotterdam, The Netherlands.,Department of Genetics, University of Groningen, University Medical Center Groningen, RB, Groningen, The Netherlands
| | - Roy G Masius
- Department of Clinical Genetics, Erasmus University Medical Center (Erasmus MC), CA Rotterdam, The Netherlands
| | - Anton J van Essen
- Department of Clinical Genetics, LUMC, Leiden University Medical Center, Postzone K-5-R, Postbus 9600, RC Leiden, The Netherlands
| | - Patrick Rump
- Department of Clinical Genetics, LUMC, Leiden University Medical Center, Postzone K-5-R, Postbus 9600, RC Leiden, The Netherlands
| | - Arie van Haeringen
- Department of Pediatric Neurology, Juliana Hospital, Els Borst-Eilersplein 275, 2545 AA Den Haag, The Netherlands
| | - Els Peeters
- Department of Medical genetics, Istanbul Medical Faculty, Istanbul University, Topkapı Mahallesi, Turgut Özal Millet Cd, 34093 Fatih/İstanbul, Turkey
| | - Umut Altunoglu
- Department of Cell biology, Erasmus University Medical Center (Erasmus MC), CA Rotterdam, The Netherlands
| | - Tugba Kalayci
- Department of Cell biology, Erasmus University Medical Center (Erasmus MC), CA Rotterdam, The Netherlands
| | - Raymond A Poot
- Department of Pediatrics, University of Washington, Seattle, WA, USA
| | - William B Dobyns
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA.,Imagine Institute, INSERM UMR-1163, Laboratory Genetics and Embryology of Congenital Malformations, Paris Descartes University, Institut des Maladies Génétiques 24, Boulevard de Montparnasse, Paris, France
| | - Nadia Bahi-Buisson
- Pediatric Neurology Unit, Department of Pediatrics, UZ Brussel, Brussels, Belgium
| | - Frans W Verheijen
- Department of Clinical Genetics, Erasmus University Medical Center (Erasmus MC), CA Rotterdam, The Netherlands
| | - Anna C Jansen
- Neurogenetics Research Group, Research Cluster Reproduction, Genetics and Regenerative Medicine, Vrije Universiteit Brussel, Brussels, Belgium.,Center for Medical Genetics, UZ Brussel, Brussels, Belgium
| | - Grazia M S Mancini
- Department of Clinical Genetics, Erasmus University Medical Center (Erasmus MC), CA Rotterdam, The Netherlands
| |
Collapse
|
16
|
Zakaria M, Fatima A, Klar J, Wikström J, Abdullah U, Ali Z, Akram T, Tariq M, Ahmad H, Schuster J, Baig SM, Dahl N. Primary microcephaly, primordial dwarfism, and brachydactyly in adult cases with biallelic skipping of RTTN exon 42. Hum Mutat 2019; 40:899-903. [PMID: 30927481 DOI: 10.1002/humu.23755] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 02/27/2019] [Accepted: 03/24/2019] [Indexed: 11/11/2022]
Abstract
Biallelic and pathogenic variants in the RTTN gene, encoding the centrosomal protein Rotatin, are associated with variable degrees of neurodevelopmental abnormalities, microcephaly, and extracranial malformations. To date, no reported case has reached their third decade. Herein, we report on a consanguineous family with three adult members, age 43, 57, and 60 years respectively, with primary microcephaly, developmental delay, primordial dwarfism, and brachydactyly segregating a homozygous splice site variant NM_173630.3:c.5648-5T>A in RTTN. The variant RTTN allele results in a nonhypomorphic skipping of exon 42 and a frameshift [(NP_775901.3:p.Ala1883Glyfs*6)]. Brain MRI of one affected individual showed markedly reduced volume of cerebral lobes and enlarged sulci but without signs of neural migration defects. Our assessment of three adult cases with a biallelic RTTN variant shows that a predicted shortened Rotatin, lacking the C-terminal end, are associated with stationary clinical features into the seventh decade. Furthermore, our report adds brachydactyly to the phenotypic spectrum in this pleiotropic entity.
Collapse
Affiliation(s)
- Muhammad Zakaria
- Human Molecular Genetics Laboratory, National Institute for Biotechnology and Genetic Engineering (NIBGE), Faisalabad, Pakistan.,Department of Immunology, Genetics, and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden.,Centre for Human Genetics, Hazara University, Mansehra, Pakistan
| | - Ambrin Fatima
- Department of Immunology, Genetics, and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Joakim Klar
- Department of Immunology, Genetics, and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Johan Wikström
- Department of Radiology, Uppsala University, Uppsala, Sweden
| | - Uzma Abdullah
- Human Molecular Genetics Laboratory, National Institute for Biotechnology and Genetic Engineering (NIBGE), Faisalabad, Pakistan
| | - Zafar Ali
- Human Molecular Genetics Laboratory, National Institute for Biotechnology and Genetic Engineering (NIBGE), Faisalabad, Pakistan
| | - Talia Akram
- Human Molecular Genetics Laboratory, National Institute for Biotechnology and Genetic Engineering (NIBGE), Faisalabad, Pakistan
| | - Muhammad Tariq
- Human Molecular Genetics Laboratory, National Institute for Biotechnology and Genetic Engineering (NIBGE), Faisalabad, Pakistan
| | - Habib Ahmad
- Department of Botany, Islamia College University Peshawar, Peshawar, Khyber Pakhtunkhwa, Pakistan
| | - Jens Schuster
- Department of Immunology, Genetics, and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Shahid M Baig
- Human Molecular Genetics Laboratory, National Institute for Biotechnology and Genetic Engineering (NIBGE), Faisalabad, Pakistan
| | - Niklas Dahl
- Department of Immunology, Genetics, and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| |
Collapse
|
17
|
Image-guided phenotyping of ovariectomized mice: altered functional connectivity, cognition, myelination, and dopaminergic functionality. Neurobiol Aging 2019; 74:77-89. [DOI: 10.1016/j.neurobiolaging.2018.10.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 09/20/2018] [Accepted: 10/06/2018] [Indexed: 01/22/2023]
|
18
|
Guzeva VI, Okhrim IV, Guzeva OV, Guzeva VV. Phenotypic and neuroimaging differentiation of polymicrogiry in children. Zh Nevrol Psikhiatr Im S S Korsakova 2019; 119:14-20. [DOI: 10.17116/jnevro201911904114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
19
|
Harris S, Gilmore K, Hardisty E, Lyerly AD, Vora NL. Ethical and counseling challenges in prenatal exome sequencing. Prenat Diagn 2018; 38:897-903. [PMID: 30171820 DOI: 10.1002/pd.5353] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 08/18/2018] [Accepted: 08/25/2018] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Ethical and counseling challenges are expected with the introduction of prenatal whole exome sequencing. In this study, we describe specific challenges identified through the UNC-Chapel Hill Prenatal Exome Sequencing Study. METHODS Participants were a subset of women participating in the fetal exome study, which has enrolled 73 mother-father-fetus trios in pregnancies diagnosed with structural anomalies and normal standard genetic testing results. In this descriptive study, cases were reviewed by members of the research team, including a bioethicist, to identify counseling challenges. Illustrative cases were chosen by group consensus. RESULTS Four illustrative cases were identified for further analysis. Challenges included need for adequate counseling and informed consent, challenges in prenatal variant interpretation, performing prenatal diagnosis in subsequent pregnancies, inability to identify a genetic etiology, and identifying parental secondary findings. CONCLUSION Our study illustrates several challenges identified in an ongoing prenatal exome study. While genomic medicine is a powerful tool for prenatal diagnosis, it is important that clinicians understand the ethical implications and parental perceptions of this testing modality.
Collapse
Affiliation(s)
- Sarah Harris
- University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Kelly Gilmore
- Department of Obstetrics and Gynecology, Division of Maternal Fetal Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Emily Hardisty
- Department of Obstetrics and Gynecology, Division of Maternal Fetal Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Anne Drapkin Lyerly
- Department of Social Medicine and Center for Bioethics, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Neeta L Vora
- Department of Obstetrics and Gynecology, Division of Maternal Fetal Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| |
Collapse
|
20
|
Functional characterization of biallelic RTTN variants identified in an infant with microcephaly, simplified gyral pattern, pontocerebellar hypoplasia, and seizures. Pediatr Res 2018; 84:435-441. [PMID: 29967526 PMCID: PMC6258334 DOI: 10.1038/s41390-018-0083-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 05/15/2018] [Accepted: 05/25/2018] [Indexed: 01/06/2023]
Abstract
BACKGROUND Biallelic deleterious variants in RTTN, which encodes rotatin, are associated with primary microcephaly, polymicrogyria, seizures, intellectual disability, and primordial dwarfism in human infants. METHODS AND RESULTS We performed exome sequencing of an infant with primary microcephaly, pontocerebellar hypoplasia, and intractable seizures and his healthy, unrelated parents. We cultured the infant's fibroblasts to determine primary ciliary phenotype. RESULTS We identified biallelic variants in RTTN in the affected infant: a novel missense variant and a rare, intronic variant that results in aberrant transcript splicing. Cultured fibroblasts from the infant demonstrated reduced length and number of primary cilia. CONCLUSION Biallelic variants in RTTN cause primary microcephaly in infants. Functional characterization of primary cilia length and number can be used to determine pathogenicity of RTTN variants.
Collapse
|
21
|
Sydor AM, Coyaud E, Rovelli C, Laurent E, Liu H, Raught B, Mennella V. PPP1R35 is a novel centrosomal protein that regulates centriole length in concert with the microcephaly protein RTTN. eLife 2018; 7:37846. [PMID: 30168418 PMCID: PMC6141234 DOI: 10.7554/elife.37846] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 08/21/2018] [Indexed: 01/02/2023] Open
Abstract
Centrosome structure, function, and number are finely regulated at the cellular level to ensure normal mammalian development. Here, we characterize PPP1R35 as a novel bona fide centrosomal protein and demonstrate that it is critical for centriole elongation. Using quantitative super-resolution microscopy mapping and live-cell imaging we show that PPP1R35 is a resident centrosomal protein located in the proximal lumen above the cartwheel, a region of the centriole that has eluded detailed characterization. Loss of PPP1R35 function results in decreased centrosome number and shortened centrioles that lack centriolar distal and microtubule wall associated proteins required for centriole elongation. We further demonstrate that PPP1R35 acts downstream of, and forms a complex with, RTTN, a microcephaly protein required for distal centriole elongation. Altogether, our study identifies a novel step in the centriole elongation pathway centered on PPP1R35 and elucidates downstream partners of the microcephaly protein RTTN.
Collapse
Affiliation(s)
| | - Etienne Coyaud
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Cristina Rovelli
- Cell Biology Program, The Hospital for Sick Children, Toronto, Canada
| | - Estelle Laurent
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Helen Liu
- Cell Biology Program, The Hospital for Sick Children, Toronto, Canada
| | - Brian Raught
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada.,Department of Medical Biophysics, University of Toronto, Ontario, Canada
| | - Vito Mennella
- Cell Biology Program, The Hospital for Sick Children, Toronto, Canada.,Department of Biochemistry, University of Toronto, Ontario, Canada
| |
Collapse
|
22
|
Cavallin M, Bery A, Maillard C, Salomon LJ, Bole C, Reilly ML, Nitschké P, Boddaert N, Bahi-Buisson N. Recurrent RTTN mutation leading to severe microcephaly, polymicrogyria and growth restriction. Eur J Med Genet 2018; 61:755-758. [PMID: 30121372 DOI: 10.1016/j.ejmg.2018.08.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 08/03/2018] [Accepted: 08/09/2018] [Indexed: 10/28/2022]
Abstract
Autosomal recessive missense Rotatin (RTTN) mutations are responsible for syndromic forms of malformation of cortical development, ranging from isolated polymicrogyria to microcephaly associated with primordial dwarfism and other major malformations. We identified, by trio based whole exome sequencing, a homozygous missense mutation in the RTTN gene (c.2953A > G; p.(Arg985Gly)) in one Moroccan patient from a consanguineous family. The patient showed early onset primary microcephaly, detected in the fetal period, postnatal growth restriction, encephalopathy with hyperkinetic movement disorders and self-injurious behavior with sleep disturbance. Brain MRI showed an extensive dysgyria associated with nodular heterotopia, large interhemispheric arachnoid cyst and corpus callosum hypoplasia.
Collapse
Affiliation(s)
- Mara Cavallin
- Laboratory of Embryology and Genetics of Congenital Malformations, INSERM UMR1163, Imagine Institute, Paris, France; Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, Paris, France; Pediatric Neurology APHP- Necker Enfants Malades University Hospital, Paris, France
| | - Amandine Bery
- Laboratory of Embryology and Genetics of Congenital Malformations, INSERM UMR1163, Imagine Institute, Paris, France
| | - Camille Maillard
- Laboratory of Embryology and Genetics of Congenital Malformations, INSERM UMR1163, Imagine Institute, Paris, France; Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, Paris, France
| | - Laurent J Salomon
- Department of Obstetrics and Fetal Medicine APHP- Necker Enfants Malades University Hospital, Paris, France
| | - Christine Bole
- Genomic Core Facility, INSERM UMR1163, Imagine Institute, Paris, France
| | - Madeline Louise Reilly
- Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, Paris, France; Laboratory of Inherited Kidney Diseases, INSERM UMR1163, Imagine Institute, Paris, France; Paris Diderot University, 75013, Paris, France
| | - Patrick Nitschké
- Bioinformatic Core Facility, INSERM UMR1163, Imagine Institute, Paris, France
| | - Nathalie Boddaert
- Pediatric Radiology, APHP-Necker Enfants Malades University Hospital, Paris, France; Image- Institut Imagine, INSERM UMR1163, Université Paris Descartes, Hôpital Necker Enfants Malades, Paris, France
| | - Nadia Bahi-Buisson
- Laboratory of Embryology and Genetics of Congenital Malformations, INSERM UMR1163, Imagine Institute, Paris, France; Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, Paris, France; Pediatric Neurology APHP- Necker Enfants Malades University Hospital, Paris, France; Centre de Référence, Déficiences Intellectuelles de Causes Rares, APHP- Necker Enfants Malades University Hospital, Paris, France.
| |
Collapse
|
23
|
Biallelic mutations in RTTN are associated with microcephaly, short stature and a wide range of brain malformations. Eur J Med Genet 2018; 61:733-737. [PMID: 29883675 DOI: 10.1016/j.ejmg.2018.06.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 05/07/2018] [Accepted: 06/02/2018] [Indexed: 12/12/2022]
Abstract
Biallelic mutations in the RTTN gene have been reported in association with microcephaly, short stature, developmental delay and malformations of cortical development. RTTN mutations have previously shown to link aberrant ciliary function with abnormal development and organization of the human cerebral cortex. We here report three individuals from two unrelated families with novel mutations in the RTTN gene. The phenotype consisted of microcephaly, short stature, pachygyria or polymicrogyria, colpocephaly, hypoplasia of the corpus callosum and superior vermis. These findings provide further confirmation of the phenotype related to pathogenic variants in RTTN.
Collapse
|
24
|
Genetics and mechanisms leading to human cortical malformations. Semin Cell Dev Biol 2018; 76:33-75. [DOI: 10.1016/j.semcdb.2017.09.031] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 09/21/2017] [Accepted: 09/21/2017] [Indexed: 02/06/2023]
|
25
|
Chartier S, Alby C, Boutaud L, Thomas S, Elkhartoufi N, Martinovic J, Kaplan J, Benachi A, Lacombe D, Sonigo P, Drunat S, Vekemans M, Agenor J, Encha Razavi F, Attie-Bitach T. A neuropathological study of novel RTTN gene mutations causing a familial microcephaly with simplified gyral pattern. Birth Defects Res 2018; 110:598-602. [PMID: 29356416 DOI: 10.1002/bdr2.1204] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2017] [Revised: 01/04/2018] [Accepted: 01/04/2018] [Indexed: 12/31/2022]
Abstract
BACKGROUND The RTTN gene encodes Rotatin, a large centrosomal protein involved in ciliary functions. RTTN mutations have been reported in seven families and are associated with two phenotypes: polymicrogyria associated with seizures and primary microcephaly associated with primordial dwarfism. CASE A targeted exome sequencing of morbid genes causing cerebral malformations identified novel RTTN compound heterozygous mutations in a family where three pregnancies were terminated because a severe fetal microcephaly was diagnosed. An autopsy performed on the second sib showed moderate growth restriction and a microcephaly with simplified gyral pattern. The histopathological study discovered a malformed cortical plate. CONCLUSIONS The present study confirms the involvement of RTTN gene mutations in microcephaly with simplified gyral pattern and describes the observed abnormal neuropathological findings.
Collapse
Affiliation(s)
- Suzanne Chartier
- Service d'Histologie-Embryologie-Cytogénétique, Hôpital Necker-Enfants Malades, APHP, Paris, France
| | - Caroline Alby
- Paris Sorbonne Cité, Université Paris Descartes, Paris, France.,INSERM U1163, Hôpital Necker-Enfants Malades, Institut Imagine, Paris, France.,Service de Génétique Médicale, Hôpital Necker-Enfants Malades, APHP, Paris, France
| | - Lucile Boutaud
- Service d'Histologie-Embryologie-Cytogénétique, Hôpital Necker-Enfants Malades, APHP, Paris, France.,Paris Sorbonne Cité, Université Paris Descartes, Paris, France.,INSERM U1163, Hôpital Necker-Enfants Malades, Institut Imagine, Paris, France
| | - Sophie Thomas
- Paris Sorbonne Cité, Université Paris Descartes, Paris, France.,INSERM U1163, Hôpital Necker-Enfants Malades, Institut Imagine, Paris, France
| | - Nadia Elkhartoufi
- Service d'Histologie-Embryologie-Cytogénétique, Hôpital Necker-Enfants Malades, APHP, Paris, France
| | - Jelena Martinovic
- Unité de fœtopathologie, Hôpital Antoine-Béclère, APHP, Clamart, France
| | - Josseline Kaplan
- Laboratoire de Génétique Moléculaire, Hôpital Necker-Enfants Malades, APHP, Paris, France
| | - Alexandra Benachi
- Service de Gynécologie-Obstétrique, Hôpital Antoine-Béclère, APHP, Université Paris Sud, Clamart, France
| | - Didier Lacombe
- Service de Génétique Médicale, CHU de Bordeaux, Bordeaux, France.,INSERM U1211, Université de Bordeaux, Bordeaux, France
| | - Pascale Sonigo
- Service de Radiologie Pédiatrique, Hôpital Necker-Enfants Malades, APHP, Paris, France
| | - Séverine Drunat
- Service de Génétique Moléculaire, Hôpital Robert-Debré, APHP, Paris, France.,INSERM U1141, Hôpital Robert Debré, Paris, France
| | - Michel Vekemans
- Service d'Histologie-Embryologie-Cytogénétique, Hôpital Necker-Enfants Malades, APHP, Paris, France.,Paris Sorbonne Cité, Université Paris Descartes, Paris, France.,INSERM U1163, Hôpital Necker-Enfants Malades, Institut Imagine, Paris, France
| | - Joël Agenor
- Service Pluridisciplinaire de Diagnostic Prénatal, Nouméa, France
| | - Férechté Encha Razavi
- Service d'Histologie-Embryologie-Cytogénétique, Hôpital Necker-Enfants Malades, APHP, Paris, France.,Paris Sorbonne Cité, Université Paris Descartes, Paris, France.,INSERM U1163, Hôpital Necker-Enfants Malades, Institut Imagine, Paris, France
| | - Tania Attie-Bitach
- Service d'Histologie-Embryologie-Cytogénétique, Hôpital Necker-Enfants Malades, APHP, Paris, France.,Paris Sorbonne Cité, Université Paris Descartes, Paris, France.,INSERM U1163, Hôpital Necker-Enfants Malades, Institut Imagine, Paris, France
| |
Collapse
|
26
|
Weisz Hubshman M, Broekman S, van Wijk E, Cremers F, Abu-Diab A, Khateb S, Tzur S, Lagovsky I, Smirin-Yosef P, Sharon D, Haer-Wigman L, Banin E, Basel-Vanagaite L, de Vrieze E. Whole-exome sequencing reveals POC5 as a novel gene associated with autosomal recessive retinitis pigmentosa. Hum Mol Genet 2017; 27:614-624. [DOI: 10.1093/hmg/ddx428] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 12/08/2017] [Indexed: 01/01/2023] Open
|
27
|
Goffinet AM. The evolution of cortical development: the synapsid-diapsid divergence. Development 2017; 144:4061-4077. [PMID: 29138289 DOI: 10.1242/dev.153908] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The cerebral cortex covers the rostral part of the brain and, in higher mammals and particularly humans, plays a key role in cognition and consciousness. It is populated with neuronal cell bodies distributed in radially organized layers. Understanding the common and lineage-specific molecular mechanisms that orchestrate cortical development and evolution are key issues in neurobiology. During evolution, the cortex appeared in stem amniotes and evolved divergently in two main branches of the phylogenetic tree: the synapsids (which led to present day mammals) and the diapsids (reptiles and birds). Comparative studies in organisms that belong to those two branches have identified some common principles of cortical development and organization that are possibly inherited from stem amniotes and regulated by similar molecular mechanisms. These comparisons have also highlighted certain essential features of mammalian cortices that are absent or different in diapsids and that probably evolved after the synapsid-diapsid divergence. Chief among these is the size and multi-laminar organization of the mammalian cortex, and the propensity to increase its area by folding. Here, I review recent data on cortical neurogenesis, neuronal migration and cortical layer formation and folding in this evolutionary perspective, and highlight important unanswered questions for future investigation.
Collapse
Affiliation(s)
- Andre M Goffinet
- University of Louvain, Avenue Mounier, 73 Box B1.73.16, B1200 Brussels, Belgium
| |
Collapse
|
28
|
Chen HY, Wu CT, Tang CJC, Lin YN, Wang WJ, Tang TK. Human microcephaly protein RTTN interacts with STIL and is required to build full-length centrioles. Nat Commun 2017; 8:247. [PMID: 28811500 PMCID: PMC5558016 DOI: 10.1038/s41467-017-00305-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 06/21/2017] [Indexed: 11/09/2022] Open
Abstract
Mutations in many centriolar protein-encoding genes cause primary microcephaly. Using super-resolution and electron microscopy, we find that the human microcephaly protein, RTTN, is recruited to the proximal end of the procentriole at early S phase, and is located at the inner luminal walls of centrioles. Further studies demonstrate that RTTN directly interacts with STIL and acts downstream of STIL-mediated centriole assembly. CRISPR/Cas9-mediated RTTN gene knockout in p53-deficient cells induce amplification of primitive procentriole bodies that lack the distal-half centriolar proteins, POC5 and POC1B. Additional analyses show that RTTN serves as an upstream effector of CEP295, which mediates the loading of POC1B and POC5 to the distal-half centrioles. Interestingly, the naturally occurring microcephaly-associated mutant, RTTN (A578P), shows a low affinity for STIL binding and blocks centriole assembly. These findings reveal that RTTN contributes to building full-length centrioles and illuminate the molecular mechanism through which the RTTN (A578P) mutation causes primary microcephaly. Mutations in many centriolar protein-encoding genes cause primary microcephaly. Here the authors show that human microcephaly protein RTTN directly interacts with STIL and acts downstream of STIL-mediated centriole assembly, contributing to building full-length centrioles
Collapse
Affiliation(s)
- Hsin-Yi Chen
- Graduate Institution of Life Sciences, National Defense Medical Center, Taipei, Taiwan.,Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Chien-Ting Wu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.,Taiwan International Graduate Program in Interdisciplinary Neuroscience, National Yang-Ming University and Academia Sinica, Taipei, Taiwan
| | - Chieh-Ju C Tang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yi-Nan Lin
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Won-Jing Wang
- Institute of Biochemistry and Molecular Biology, College of Life Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Tang K Tang
- Graduate Institution of Life Sciences, National Defense Medical Center, Taipei, Taiwan. .,Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan. .,Taiwan International Graduate Program in Interdisciplinary Neuroscience, National Yang-Ming University and Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
29
|
Naseer MI, Rasool M, Sogaty S, Chaudhary RA, Mansour HM, Chaudhary AG, Abuzenadah AM, Al-Qahtani MH. A novel WDR62 mutation causes primary microcephaly in a large consanguineous Saudi family. Ann Saudi Med 2017; 37:148-153. [PMID: 28377545 PMCID: PMC6150548 DOI: 10.5144/0256-4947.2017.148] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Primary microcephaly (MCPH) is a rare developmental defect characterized by impaired cognitive functions, retarded neurodevelopment and reduced brain size. It is genetically heterogeneous and more than 17 genes so far have been identified that are associated with this disease. OBJECTIVE To study the genetic defect in a consanguineous Saudi family with primary microcephaly. DESIGN Cross-sectional clinical genetics study of a Saudi family. SETTING Medical genomics research center. PATIENTS AND METHODS Blood samples collected from six members of a family of healthy consanguineous parents were analyzed by whole exome sequencing to identify the underlying pathogenic mutations in two members of the family (23-year-old female and 7-year-old male) who presented with primary microcephaly, intellectual disability, delayed psychomotor development and walking difficulty, speech impedi-ments and seizures. MAIN OUTCOME MEASURE(S) Detection of mutation in the WD repeat domain 62 (WDR62) gene in a family segregating autosomal recessive primary microcephaly. RESULTS The exome variant analysis identified a novel missense mutation (c.3878C > A) in WDR62 gene in exon 30 resulting in amino acid change from alanine to aspartate (p.Ala1293Asp). Further validation in the affected patients and healthy members of family and 100 unrelated healthy persons as controls confirmed it to be pathogenic. CONCLUSIONS Functional impairment of the WDR62 gene can lead to severe neurodevelopmental de-fects, brain malformations and reduced head size. A missense mutation of exon 30 changed alanine to aspartate in the WDR62 protein leading to the typical MCPH phenotype. LIMITATIONS Mutation was identified in a single family.
Collapse
Affiliation(s)
- Muhammad Imran Naseer
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mahmood Rasool
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Sameera Sogaty
- Department of Medical Genetics, King Fahad General Hospital, Jeddah, Saudi Arabia
| | - Rukhaa Adeel Chaudhary
- Faculty of Applied Medical Sciences, Medical Laboratory Technology, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Haifa Mansour Mansour
- Faculty of Applied Medical Sciences, Medical Laboratory Technology, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Adeel G. Chaudhary
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Adel M. Abuzenadah
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohammad H. Al-Qahtani
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
30
|
Grandone A, Torella A, Santoro C, Giugliano T, Del Vecchio Blanco F, Mutarelli M, Cirillo M, Cirillo G, Piluso G, Capristo C, Festa A, Marzuillo P, Miraglia Del Giudice E, Perrone L, Nigro V. Expanding the phenotype of RTTN variations: a new family with primary microcephaly, severe growth failure, brain malformations and dermatitis. Clin Genet 2016; 90:445-450. [PMID: 26940245 DOI: 10.1111/cge.12771] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Revised: 02/26/2016] [Accepted: 03/01/2016] [Indexed: 01/03/2023]
Abstract
Primary autosomal recessive microcephaly (MCPH) is a developmental disorder characterized by prenatal onset of abnormal brain growth. MCPH occurs both alone and as part of a broad range of neurodevelopmental syndromes with or without cortical malformations and growth retardation. Here we report a consanguineous Moroccan family with two siblings affected by severe primary microcephaly, failure to thrive, congenital dermatitis and severe developmental delay. Brain magnetic resonance imaging showed lissencephaly of frontal lobes and periventricular heterotopia of the gray matter. We performed both Comparative Genomic Hybridization array and whole exome sequencing (WES) analyses of the kindred. No quantitative defects were detected. However, WES identified a new homozygous missense variation in the penultimate nucleotide of exon 23 of RTTN gene (c.2953A>G;pArg985Gly). cDNA sequencing revealed two abnormal spliced products, one lacking only exon 23 and the other lacking exons 22 and 23 (out-of-frame). RTTN is a protein involved in cilia structure and function. Homozygous mutations in RTTN gene have been described in bilateral diffuse isolated polymicrogyria and, more recently, in microcephalic primordial dwarfism (PD). We found a novel homozygous mutation in RTTN associated with microcephalic PD as well as complex brain malformations and congenital dermatitis, thus expanding the phenotypic spectrum of both RTTN-associated diseases and ciliary dysfunction.
Collapse
Affiliation(s)
- A Grandone
- Dipartimento della Donna, del Bambino, di Chirurgia Generale e Specialistica, Seconda Università degli Studi di Napoli, Naples, Italy
| | - A Torella
- Dipartimento di Biochimica, Biofisica e Patologia Generale, Seconda Università degli Studi di Napoli, Naples, Italy
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli (NA), Italy
| | - C Santoro
- Dipartimento della Donna, del Bambino, di Chirurgia Generale e Specialistica, Seconda Università degli Studi di Napoli, Naples, Italy
| | - T Giugliano
- Dipartimento di Biochimica, Biofisica e Patologia Generale, Seconda Università degli Studi di Napoli, Naples, Italy
| | - F Del Vecchio Blanco
- Dipartimento di Biochimica, Biofisica e Patologia Generale, Seconda Università degli Studi di Napoli, Naples, Italy
| | - M Mutarelli
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli (NA), Italy
| | - M Cirillo
- Dipartimento di Scienze Mediche, Chirurgiche, Neurologiche, Metaboliche e dell'Invecchiamento, Seconda Università degli Studi di Napoli, Naples, Italy
| | - G Cirillo
- Dipartimento della Donna, del Bambino, di Chirurgia Generale e Specialistica, Seconda Università degli Studi di Napoli, Naples, Italy
| | - G Piluso
- Dipartimento di Biochimica, Biofisica e Patologia Generale, Seconda Università degli Studi di Napoli, Naples, Italy
| | - C Capristo
- Dipartimento della Donna, del Bambino, di Chirurgia Generale e Specialistica, Seconda Università degli Studi di Napoli, Naples, Italy
| | - A Festa
- Dipartimento della Donna, del Bambino, di Chirurgia Generale e Specialistica, Seconda Università degli Studi di Napoli, Naples, Italy
| | - P Marzuillo
- Dipartimento della Donna, del Bambino, di Chirurgia Generale e Specialistica, Seconda Università degli Studi di Napoli, Naples, Italy
| | - E Miraglia Del Giudice
- Dipartimento della Donna, del Bambino, di Chirurgia Generale e Specialistica, Seconda Università degli Studi di Napoli, Naples, Italy
| | - L Perrone
- Dipartimento della Donna, del Bambino, di Chirurgia Generale e Specialistica, Seconda Università degli Studi di Napoli, Naples, Italy
| | - V Nigro
- Dipartimento di Biochimica, Biofisica e Patologia Generale, Seconda Università degli Studi di Napoli, Naples, Italy. ,
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli (NA), Italy. ,
| |
Collapse
|
31
|
Shamseldin HE, Maddirevula S, Faqeih E, Ibrahim N, Hashem M, Shaheen R, Alkuraya FS. Increasing the sensitivity of clinical exome sequencing through improved filtration strategy. Genet Med 2016; 19:593-598. [PMID: 27711071 DOI: 10.1038/gim.2016.155] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 08/24/2016] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Clinical exome sequencing (CES) has greatly improved the diagnostic process for individuals with suspected genetic disorders. However, the majority remains undiagnosed after CES. Although understanding potential reasons for this limited sensitivity is critical for improving the delivery of clinical genomics, research in this area has been limited. MATERIALS AND METHODS We first calculated the theoretical maximum sensitivity of CES by analyzing >100 families in whom a Mendelian phenotype is mapped to a single locus. We then tested the hypothesis that positional mapping can limit the search space and thereby facilitate variant interpretation by reanalyzing 33 families with "negative" CES and applying positional mapping. RESULTS We found that >95% of families who map to a single locus harbored genic (as opposed to intergenic) variants that are potentially identifiable by CES. Our reanalysis of "negative" CES revealed likely causal variants in the majority (88%). Several of these solved cases have undergone negative whole-genome sequencing. CONCLUSION The discrepancy between the theoretical maximum and the actual clinical sensitivity of CES is primarily in the variant filtration rather than the variant capture and sequencing phase. The solution to negative CES is not necessarily in expanding the coverage but rather in devising approaches that improve variant filtration. We suggest that positional mapping is one such approach.Genet Med advance online publication 06 October 2016.
Collapse
Affiliation(s)
- Hanan E Shamseldin
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Sateesh Maddirevula
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Eissa Faqeih
- Department of Pediatric Subspecialties, Children's Hospital, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Niema Ibrahim
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Mais Hashem
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Ranad Shaheen
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Fowzan S Alkuraya
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia.,Department of Anatomy and Cell Biology, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| |
Collapse
|
32
|
Mutations in CIT, encoding citron rho-interacting serine/threonine kinase, cause severe primary microcephaly in humans. Hum Genet 2016; 135:1191-7. [PMID: 27503289 DOI: 10.1007/s00439-016-1722-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2016] [Accepted: 08/02/2016] [Indexed: 01/08/2023]
Abstract
Primary microcephaly is a clinical phenotype in which the head circumference is significantly reduced at birth due to abnormal brain development, primarily at the cortical level. Despite the marked genetic heterogeneity, most primary microcephaly-linked genes converge on mitosis regulation. Two consanguineous families segregating the phenotype of severe primary microcephaly, spasticity and failure to thrive had overlapping autozygomes in which exome sequencing identified homozygous splicing variants in CIT that segregate with the phenotype within each family. CIT encodes citron, an effector of the Rho signaling that is required for cytokinesis specifically in proliferating neuroprogenitors, as well as for postnatal brain development. In agreement with the critical role assigned to the kinase domain in effecting these biological roles, we show that both splicing variants predict variable disruption of this domain. The striking phenotypic overlap between CIT-mutated individuals and the knockout mice and rats that are specifically deficient in the kinase domain supports the proposed causal link between CIT mutation and primary microcephaly in humans.
Collapse
|
33
|
Alkuraya FS. Discovery of mutations for Mendelian disorders. Hum Genet 2016; 135:615-23. [DOI: 10.1007/s00439-016-1664-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 03/28/2016] [Indexed: 12/11/2022]
|
34
|
Shamseldin HE, Bennett AH, Alfadhel M, Gupta V, Alkuraya FS. GOLGA2, encoding a master regulator of golgi apparatus, is mutated in a patient with a neuromuscular disorder. Hum Genet 2016; 135:245-251. [PMID: 26742501 DOI: 10.1007/s00439-015-1632-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 12/25/2015] [Indexed: 01/21/2023]
Abstract
Golgi apparatus (GA) is a membrane-bound organelle that serves a multitude of critical cellular functions including protein secretion and sorting, and cellular polarity. Many Mendelian diseases are caused by mutations in genes encoding various components of GA. GOLGA2 encodes GM130, a necessary component for the assembly of GA as a single complex, and its deficiency has been found to result in severe cellular phenotypes. We describe the first human patient with a homozygous apparently loss of function mutation in GOLGA2. The phenotype is a neuromuscular disorder characterized by developmental delay, seizures, progressive microcephaly, and muscular dystrophy. Knockdown of golga2 in zebrafish resulted in severe skeletal muscle disorganization and microcephaly recapitulating loss of function human phenotype. Our data suggest an important developmental role of GM130 in humans and zebrafish.
Collapse
Affiliation(s)
- Hanan E Shamseldin
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Alexis H Bennett
- Division of Genetics, Brigham and Women's Hospital and Department of Genetics, Children's Hospital Boston, Harvard Medical School, Boston, MA, USA
| | - Majid Alfadhel
- Genetics Division, Department of Pediatrics, King Saud bin Abdulaziz University for Health Science, King Abdulaziz Medical City, Riyadh, Saudi Arabia
| | - Vandana Gupta
- Division of Genetics, Brigham and Women's Hospital and Department of Genetics, Children's Hospital Boston, Harvard Medical School, Boston, MA, USA
| | - Fowzan S Alkuraya
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
- Department of Anatomy and Cell Biology, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| |
Collapse
|