1
|
Kilic S, Bove J, So BN, Whitman MC. Strabismus in Genetic Syndromes: A Review. Clin Exp Ophthalmol 2025; 53:302-330. [PMID: 39948700 DOI: 10.1111/ceo.14507] [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: 09/24/2024] [Revised: 01/29/2025] [Accepted: 01/29/2025] [Indexed: 04/03/2025]
Abstract
Strabismus is a feature of many genetic syndromes, with highly variable penetrance. The congenital cranial dysinnervation disorders (CCDDs) result in paralytic strabismus, with limited eye movements. CCDDs result from either deficits in differentiation of the cranial motor neuron precursors or from abnormal axon guidance of the cranial nerves. Although most individuals with comitant strabismus are otherwise healthy, strabismus is a variable feature of many genetic syndromes, most commonly those associated with intellectual disability. We review 255 genetic syndromes in which strabismus has been described and discuss the variable penetrance. The association with intellectual disability and neurological disorders underscores the likely neurological basis of strabismus, but the variable penetrance emphasises the complexity of strabismus pathophysiology. The syndromes described here mostly result from loss of function or change in function of the responsible genes; one hypothesis is that nonsyndromic strabismus may result from altered expression or regulation of the same genes.
Collapse
Affiliation(s)
- Seyda Kilic
- Department of Ophthalmology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Jillian Bove
- Department of Ophthalmology, Boston Children's Hospital, Boston, Massachusetts, USA
- Boston Orthoptic Fellowship Program, Boston, Massachusetts, USA
| | | | - Mary C Whitman
- Department of Ophthalmology, Boston Children's Hospital, Boston, Massachusetts, USA
- Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, USA
- F.M. Kirby Neuroscience Center, Boston Children's Hospital, Boston, Massachusetts, USA
| |
Collapse
|
2
|
Makker S, Gagnon BR, Trew I, Mougios V, Hanna A, Cale JM, McIntosh CS. A Patient Case of Malan Syndrome Involving 19p13.2 Deletion of NFIX with Longitudinal Follow-Up and Future Prospectives. J Clin Med 2024; 13:6575. [PMID: 39518712 PMCID: PMC11546576 DOI: 10.3390/jcm13216575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 10/17/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
Background and Objectives: Malan syndrome is a rare overgrowth syndrome resulting from NFIX haploinsufficiency due to heterozygous loss-of-function mutations or microdeletions of NFIX on chromosome 19 at p13.2. Phenotypic presentation can vary but is characterized by macrocephaly, long and slender body habitus, skeletal abnormalities, and intellectual disability. Methods: Here, we report on the presentation, management, and development of a patient with Malan syndrome, highlighting the clinical and behavioral aspects of this syndrome, therapeutic interventions employed, and the course of disease over a 15-year period. We review medical records, cytogenetic analysis and neuropsychologic testing results, as well as speech pathology, optometric, and medical reports. In addition, we discuss personalized therapeutic strategies that could potentially be exploited in the future for such overgrowth syndromes. Results: To our knowledge, this is the first longitudinal follow-up report of a case of Malan syndrome to highlight the clinical course, interventions employed, and resulting improvements in neurocognitive function over time. Conclusions: This case highlights the importance of early diagnosis, intervention, and preventative care in overgrowth syndromes, as well as the potential for therapeutic intervention in the future.
Collapse
Affiliation(s)
| | - Bernadine R. Gagnon
- Edward D. Mysak Clinic for Communication Disorders, Teachers College, Columbia University, New York, NY 10027, USA;
| | - Isabella Trew
- Centre for Molecular Medicine and Innovative Therapeutics, Health Futures Institute, Murdoch University, Murdoch, WA 6150, Australia; (I.T.); (J.M.C.)
- Perron Institute for Neurological and Translational Science, Nedlands, WA 6009, Australia
| | - Vivian Mougios
- Action Potential Institute, 145 W 96th S, New York, NY 10025, USA;
| | - Anne Hanna
- Gold Coast Optometric Vision Performance, Oyster Bay, NY 11771, USA;
| | - Jessica M. Cale
- Centre for Molecular Medicine and Innovative Therapeutics, Health Futures Institute, Murdoch University, Murdoch, WA 6150, Australia; (I.T.); (J.M.C.)
- Perron Institute for Neurological and Translational Science, Nedlands, WA 6009, Australia
| | - Craig S. McIntosh
- Centre for Molecular Medicine and Innovative Therapeutics, Health Futures Institute, Murdoch University, Murdoch, WA 6150, Australia; (I.T.); (J.M.C.)
- Perron Institute for Neurological and Translational Science, Nedlands, WA 6009, Australia
| |
Collapse
|
3
|
林 雪, 全 昱, 贺 海, 彭 镜. [ NFIX gene mutation causes Marshall-Smith syndrome in a pair of identical twins and literature review]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2024; 26:750-756. [PMID: 39014953 PMCID: PMC11562048 DOI: 10.7499/j.issn.1008-8830.2401047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 05/04/2024] [Indexed: 07/18/2024]
Abstract
This article reports on the clinical and genetic characteristics of monozygotic twins with Marshall-Smith syndrome (MRSHSS) due to a mutation in the NFIX gene, along with a review of related literature. Both patients presented with global developmental delays, a prominent forehead, shallow eye sockets, and pectus excavatum. Genetic testing revealed a heterozygous splicing site mutation c.697+1G>A in both children, with parents showing wild-type at this locus. According to the guidelines of the American College of Medical Genetics and Genomics, this mutation is considered likely pathogenic and has not been previously reported in the literature. A review of the literature identified 32 MRSHSS patients with splicing/frameshift mutations. Accelerated bone maturation and moderate to severe global developmental delay/intellectual disability are the primary clinical manifestations of patients with MRSHSS. Genetic testing results are crucial for the diagnosis of this condition.
Collapse
Affiliation(s)
| | | | | | - 镜 彭
- 湖南省儿童脑发育障碍性疾病临床医学研究中心,湖南长沙410008
| |
Collapse
|
4
|
Uzman CY, Gürsoy S, Hazan F. A rare cause of intellectual disability: Novel mutations of NFIX gene in two patients with clinical features of Marshall-Smith syndrome and Malan syndrome. Int J Dev Neurosci 2023. [PMID: 37336770 DOI: 10.1002/jdn.10280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 05/12/2023] [Accepted: 05/19/2023] [Indexed: 06/21/2023] Open
Abstract
Marshall-Smith syndrome (MSS) and Malan syndrome (MS) are both allelic disorders caused by mutations in the NFIX gene. MS is characterized by overgrowth, intellectual disability, distinctive facial features, and accelerated skeletal maturation. On the other hand, clinical features of MSS consist of advanced bone age, dysmorphic features, intellectual disability, and failure to thrive at birth. In this study, we presented the clinical and molecular findings of two different patients with MS and MSS as a rare cause of intellectual disability and reported two novel variants in the NFIX gene. NFIX gene sequencing revealed a novel heterozygous c.1287delC (p.G430Vfs*34) mutation in patient 1 whose clinical diagnosis was compatible with Marshall-Smith syndrome, and in the second patient, physical features consistent with Malan syndrome, was detected a heterozygous one nucleotide duplication, c.303dupC (pCys102LeufsTer17).
Collapse
Affiliation(s)
- Ceren Yılmaz Uzman
- Department of Pediatric Genetics, S.B.Ü. Dr. Behçet Uz Children's Education and Research Hospital, Izmir, Turkey
| | - Semra Gürsoy
- Department of Pediatric Genetics, S.B.Ü. Dr. Behçet Uz Children's Education and Research Hospital, Izmir, Turkey
| | - Filiz Hazan
- Department of Medical Genetics, S.B.Ü. Dr. Behçet Uz Children's Education and Research Hospital, Izmir, Turkey
| |
Collapse
|
5
|
Chaand M, Fiore C, Johnston B, D'Ippolito A, Moon DH, Carulli JP, Shearstone JR. Erythroid lineage chromatin accessibility maps facilitate identification and validation of NFIX as a fetal hemoglobin repressor. Commun Biol 2023; 6:640. [PMID: 37316562 DOI: 10.1038/s42003-023-05025-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 06/07/2023] [Indexed: 06/16/2023] Open
Abstract
Human genetics has validated de-repression of fetal gamma globin (HBG) in adult erythroblasts as a powerful therapeutic paradigm in diseases involving defective adult beta globin (HBB)1. To identify factors involved in the switch from HBG to HBB expression, we performed Assay for Transposase Accessible Chromatin with high-throughput sequencing (ATAC-seq)2 on sorted erythroid lineage cells derived from bone marrow (BM) or cord blood (CB), representing adult and fetal states, respectively. BM to CB cell ATAC-seq profile comparisons revealed genome-wide enrichment of NFI DNA binding motifs and increased NFIX promoter chromatin accessibility, suggesting that NFIX may repress HBG. NFIX knockdown in BM cells increased HBG mRNA and fetal hemoglobin (HbF) protein levels, coincident with increased chromatin accessibility and decreased DNA methylation at the HBG promoter. Conversely, overexpression of NFIX in CB cells reduced HbF levels. Identification and validation of NFIX as a new target for HbF activation has implications in the development of therapeutics for hemoglobinopathies.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jeffrey R Shearstone
- Syros Pharmaceuticals, Cambridge, MA, USA
- Scientific and Medical Writing Partners, Cambridge, MA, USA
| |
Collapse
|
6
|
Alfieri P, Montanaro FAM, Macchiaiolo M, Collotta M, Caciolo C, Galassi P, Panfili FM, Cortellessa F, Zollino M, Chinali M, Accadia M, Seri M, Bartuli A, Mammì C, Tartaglia M, Vicari S, Priolo M. Behavioral profiling in children and adolescents with Malan syndrome. FRONTIERS IN CHILD AND ADOLESCENT PSYCHIATRY 2023; 2:1106228. [PMID: 39816865 PMCID: PMC11732151 DOI: 10.3389/frcha.2023.1106228] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 01/24/2023] [Indexed: 01/18/2025]
Abstract
Malan syndrome (MALNS) is an ultra-rare genetic disorder caused by heterozygous chromosomal microdeletions involving the 19p13.2 region or loss-of-function variants in the NFIX gene. It is characterized by specific phenotypical features, intellectual disability (ID), and limitations in adaptive functioning and behavioral problems. In a previous work, we defined the cognitive, adaptive, linguistic and visuomotor ability profiles in a group of 15 MALNS individuals, providing quantitative data from standardized evaluations. Here, we further extend the characterization of MALNS by analyzing the behavioral and psychopathological comorbidities of the same cohort, administering standardized tests. Children were evaluated from October 2020 to January 2022. Retrospective data analysis was also performed. Assessment consisted of clinical observations, structured parent interviews, and parent-reported questionnaires. For each scale, comparisons between subtests were performed. Results of our analysis show that the most prevalent psychiatric comorbidities are represented by anxiety symptoms (including GAD, separation anxiety and specific phobias), ADHD, autistic symptoms, and social and attention problems. Of note, minimal or no signs of ASD were observed. In conclusion, our findings indicate that the psychopathological and behavioral comorbidities, together with cognitive impairment, language problems and sensory difficulties interfere with development, daily activities and social participation, therefore contributing to the severity of the disability associated with MALNS. Awareness of this profile by professionals and caregivers can promote prompt diagnosis and support cognitive and behavioral development.
Collapse
Affiliation(s)
- Paolo Alfieri
- Child and Adolescent Neuropsychiatry Unit, Department of Neuroscience, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Federica Alice Maria Montanaro
- Child and Adolescent Neuropsychiatry Unit, Department of Neuroscience, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Marina Macchiaiolo
- Rare Diseases and Medical Genetics Unit, University – Hospital Pediatric Department (DPUO), Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Martina Collotta
- Child and Adolescent Neuropsychiatry Unit, Department of Neuroscience, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Cristina Caciolo
- Child and Adolescent Neuropsychiatry Unit, Department of Neuroscience, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Paolo Galassi
- Child and Adolescent Neuropsychiatry Unit, Department of Neuroscience, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Filippo Maria Panfili
- School of Pediatrics, University Tor Vergata, Rome, Italy
- Academic Department of Pediatrics, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Fabiana Cortellessa
- Rare Diseases and Medical Genetics Unit, University – Hospital Pediatric Department (DPUO), Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Marcella Zollino
- Genetica Medica, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Italy
- Dipartimento Universitario Scienze Della Vita e Sanità Pubblica, Sezione di Medicina Genomica, Università Cattolica del Sacro Cuore Facoltà di Medicina e Chirurgia, Roma, Italy
| | - Marcello Chinali
- Department of Pediatric Cardiology, Bambino Gesù Children's Hospital, Rome, Italy
| | - Maria Accadia
- Medical Genetics Service, Hospital “Cardinale G. Panico”, Lecce, Italy
| | - Marco Seri
- Unit of Medical Genetics, IRCCS Azienda Ospedaliero Universitaria di Bologna, Bologna, Italy
| | - Andrea Bartuli
- Rare Diseases and Medical Genetics Unit, University – Hospital Pediatric Department (DPUO), Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Corrado Mammì
- Operative Unit of Medical Genetics Bianchi – Melacrino- Morelli Hospital, Reggio Calabria, Italy
| | - Marco Tartaglia
- Genetics and Rare Diseases Research Division, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Stefano Vicari
- Child and Adolescent Neuropsychiatry Unit, Department of Neuroscience, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
- Department of Life Sciences and Public Health, Università Cattolica Del Sacro Cuore, Rome, Italy
| | - Manuela Priolo
- Operative Unit of Medical Genetics Bianchi – Melacrino- Morelli Hospital, Reggio Calabria, Italy
| |
Collapse
|
7
|
Seed L, G N, Prentice S, Chandershekar P. Early diagnosis of Malan syndrome in an infant presenting with macrocephaly. BMJ Case Rep 2022; 15:e249391. [PMID: 36379624 PMCID: PMC9668004 DOI: 10.1136/bcr-2022-249391] [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] [Accepted: 10/31/2022] [Indexed: 11/16/2022] Open
Abstract
We present an infant with persistent macrocephaly and developmental delay. There is a wide range of differential diagnoses for this presentation, including many rare genetic conditions. Here, a diagnosis of Malan syndrome was made-a rare overgrowth syndrome caused by haploinsufficiency of NFIX and features affecting the neurological and musculoskeletal systems. Improvements in genomic medicine technologies and clinical services have revolutionised the way clinicians diagnose rare diseases. We highlight the importance of early genetic testing, particularly if there are red flag features such as developmental delay, and the need for a coordinated strategy to improve the management of rare diseases like Malan syndrome.
Collapse
Affiliation(s)
- Lydia Seed
- University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Natasha G
- Department of Neonatology, Lister Hospital, East and North Hertfordshire NHS Trust, Stevenage, UK
| | - Sarah Prentice
- Department of Neonatology, Lister Hospital, East and North Hertfordshire NHS Trust, Stevenage, UK
- Clinical Research Department, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Prathiba Chandershekar
- Department of Neonatology, Lister Hospital, East and North Hertfordshire NHS Trust, Stevenage, UK
| |
Collapse
|
8
|
Langley E, Farach LS, Mowrey K. Case Report: Novel pathogenic variant in NFIX in two sisters with Malan syndrome due to germline mosaicism. Front Genet 2022; 13:1044660. [DOI: 10.3389/fgene.2022.1044660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 10/31/2022] [Indexed: 11/11/2022] Open
Abstract
Malan syndrome is an autosomal dominant disorder caused by pathogenic variants in NFIX with less than 100 cases reported thus far. NFIX is important for stem cell proliferation, quiescence, and differentiation during development and its protein plays a role in replication, signal transduction, and transcription. As a result of pathogenic variants, symptoms of Malan syndrome include overgrowth, intellectual disability, speech delay, and dysmorphic features. Currently, the recurrence risk for this disorder is indicated at less than 1%, standard for de novo autosomal dominant disorders. Herein, we report an additional set of sisters with the same novel pathogenic variant in NFIX and clinical features consistent with Malan syndrome providing evidence of germline mosaicism. Considering the rarity of this condition in conjunction with three previous reports of germline mosaicism, it is worthwhile to investigate and re-evaluate the proper recurrence risk for this condition. This discovery would be paramount for family planning and genetic counseling practices in families with affected individuals.
Collapse
|
9
|
Alfieri P, Macchiaiolo M, Collotta M, Montanaro FAM, Caciolo C, Cumbo F, Galassi P, Panfili FM, Cortellessa F, Zollino M, Accadia M, Seri M, Tartaglia M, Bartuli A, Mammì C, Vicari S, Priolo M. Characterization of Cognitive, Language and Adaptive Profiles of Children and Adolescents with Malan Syndrome. J Clin Med 2022; 11:jcm11144078. [PMID: 35887841 PMCID: PMC9316998 DOI: 10.3390/jcm11144078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 06/16/2022] [Accepted: 06/16/2022] [Indexed: 11/17/2022] Open
Abstract
Malan Syndrome (MS) is an ultra-rare overgrowth genetic syndrome due to heterozygous variants or deletions in the Nuclear Factor I X (NFIX) gene. It is characterized by an unusual facial phenotype, generalized overgrowth, intellectual disability (ID) and behavioral problems. Even though limitations in cognitive and adaptive functioning have been previously described, systematic studies on MS cohorts are still lacking. Here, we aim to define the cognitive and adaptive behavior profile of MS children and adolescents, providing quantitative data from standardized evaluations. Subjects included in this study were evaluated from October 2020 to January 2022 and the study is based on a retrospective data archive: fifteen MS individuals were recruited and underwent evaluation with Wechsler Intelligence Scales, Leiter International Performance Scales and Griffith Mental Development Scales for cognitive profiles and with Vineland Adaptive Behavior Scales-II Edition (VABS-II) for adaptive functioning. Language skills and visuomotor integration abilities were assessed too. Comparisons and correlations between scales and subtests were performed. All the assessed MS individuals showed both low cognitive and adaptive functioning. One subject presented with mild ID, five had moderate ID and eight showed severe ID. One female toddler received a diagnosis of psychomotor delay. Linguistic skills were impaired in all individuals, with language comprehension relatively more preserved. Results revealed significant differences between VABS-II subdomains and a strong relationship between cognitive and adaptive functioning. All subjects exhibited mild to moderate ID and adaptive behavior lower than normal, with communication skills being the most affected. Regarding the daily living skills domain, personal and community subscale scores were dramatically lower than for the domestic subdomain, highlighting the importance of considering behavior within developmental and environmental contexts. Our cognitive and adaptive MS characterization provides a more accurate quantitative MS profiling, which is expected to help clinicians to better understand the complexity of this rare disorder.
Collapse
Affiliation(s)
- Paolo Alfieri
- Child and Adolescent Neuropsychiatry Unit, Department of Neuroscience, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (M.C.); (F.A.M.M.); (C.C.); (F.C.); (P.G.); (S.V.)
- Correspondence: ; Tel.: +39-668-594-721
| | - Marina Macchiaiolo
- Rare Diseases and Medical Genetics Unit, University-Hospital Pediatric Department (DPUO), Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (M.M.); (F.C.); (A.B.)
| | - Martina Collotta
- Child and Adolescent Neuropsychiatry Unit, Department of Neuroscience, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (M.C.); (F.A.M.M.); (C.C.); (F.C.); (P.G.); (S.V.)
| | - Federica Alice Maria Montanaro
- Child and Adolescent Neuropsychiatry Unit, Department of Neuroscience, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (M.C.); (F.A.M.M.); (C.C.); (F.C.); (P.G.); (S.V.)
| | - Cristina Caciolo
- Child and Adolescent Neuropsychiatry Unit, Department of Neuroscience, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (M.C.); (F.A.M.M.); (C.C.); (F.C.); (P.G.); (S.V.)
| | - Francesca Cumbo
- Child and Adolescent Neuropsychiatry Unit, Department of Neuroscience, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (M.C.); (F.A.M.M.); (C.C.); (F.C.); (P.G.); (S.V.)
| | - Paolo Galassi
- Child and Adolescent Neuropsychiatry Unit, Department of Neuroscience, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (M.C.); (F.A.M.M.); (C.C.); (F.C.); (P.G.); (S.V.)
| | - Filippo Maria Panfili
- Academic Department of Pediatrics, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy;
| | - Fabiana Cortellessa
- Rare Diseases and Medical Genetics Unit, University-Hospital Pediatric Department (DPUO), Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (M.M.); (F.C.); (A.B.)
| | - Marcella Zollino
- Genetica Medica, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy;
- Dipartimento Universitario Scienze della Vita e Sanità Pubblica, Sezione di Medicina Genomica, Università Cattolica del Sacro Cuore Facoltà di Medicina e Chirurgia, 00168 Roma, Italy
| | - Maria Accadia
- Medical Genetics Service, Hospital “Cardinale G. Panico”, 73039 Tricase, Italy;
| | - Marco Seri
- Unit of Medical Genetics, Azienda Ospedaliero Universitaria di Bologna, IRCCS, 40126 Bologna, Italy;
| | - Marco Tartaglia
- Genetics and Rare Diseases Research Division, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy;
| | - Andrea Bartuli
- Rare Diseases and Medical Genetics Unit, University-Hospital Pediatric Department (DPUO), Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (M.M.); (F.C.); (A.B.)
| | - Corrado Mammì
- Operative Unit of Medical Genetics Bianchi-Melacrino-Morelli Great Metropolitan Hospital, 89133 Reggio Calabria, Italy; (C.M.); (M.P.)
| | - Stefano Vicari
- Child and Adolescent Neuropsychiatry Unit, Department of Neuroscience, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (M.C.); (F.A.M.M.); (C.C.); (F.C.); (P.G.); (S.V.)
- Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Manuela Priolo
- Operative Unit of Medical Genetics Bianchi-Melacrino-Morelli Great Metropolitan Hospital, 89133 Reggio Calabria, Italy; (C.M.); (M.P.)
| |
Collapse
|
10
|
Macchiaiolo M, Panfili FM, Vecchio D, Gonfiantini MV, Cortellessa F, Caciolo C, Zollino M, Accadia M, Seri M, Chinali M, Mammì C, Tartaglia M, Bartuli A, Alfieri P, Priolo M. A deep phenotyping experience: up to date in management and diagnosis of Malan syndrome in a single center surveillance report. Orphanet J Rare Dis 2022; 17:235. [PMID: 35717370 PMCID: PMC9206304 DOI: 10.1186/s13023-022-02384-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 06/06/2022] [Indexed: 12/14/2022] Open
Abstract
Background Malan syndrome (MALNS) is a recently described ultrarare syndrome lacking guidelines for diagnosis, management and monitoring of evolutive complications. Less than 90 patients are reported in the literature and limited clinical information are available to assure a proper health surveillance.
Results A multidisciplinary team with high expertise in MALNS has been launched at the “Ospedale Pediatrico Bambino Gesù”, Rome, Italy. Sixteen Italian MALNS individuals with molecular confirmed clinical diagnosis of MALNS were enrolled in the program. For all patients, 1-year surveillance in a dedicated outpatient Clinic was attained. The expert panel group enrolled 16 patients and performed a deep phenotyping analysis directed to clinically profiling the disorder and performing critical revision of previously reported individuals. Some evolutive complications were also assessed. Previously unappreciated features (e.g., high risk of bone fractures in childhood, neurological/neurovegetative symptoms, noise sensitivity and Chiari malformation type 1) requiring active surveillance were identified. A second case of neoplasm was recorded. No major cardiovascular anomalies were noticed. An accurate clinical description of 9 new MALNS cases was provided. Conclusions Deep phenotyping has provided a more accurate characterization of the main clinical features of MALNS and allows broadening the spectrum of disease. A minimal dataset of clinical evaluations and follow-up timeline has been proposed for proper management of patients affected by this ultrarare disorder. Supplementary Information The online version contains supplementary material available at 10.1186/s13023-022-02384-9.
Collapse
Affiliation(s)
- Marina Macchiaiolo
- Rare Diseases and Medical Genetics Unit, University-Hospital Pediatric Department (DPUO), Bambino Gesù Children's Hospital, IRCSS, Piazza di Sant'Onofrio, 4, 00165, Rome, Italy.
| | - Filippo M Panfili
- University of Rome Tor Vergata, Rome, Italy.,Academic Department of Pediatrics, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Davide Vecchio
- Rare Diseases and Medical Genetics Unit, University-Hospital Pediatric Department (DPUO), Bambino Gesù Children's Hospital, IRCSS, Piazza di Sant'Onofrio, 4, 00165, Rome, Italy
| | - Michaela V Gonfiantini
- Rare Diseases and Medical Genetics Unit, University-Hospital Pediatric Department (DPUO), Bambino Gesù Children's Hospital, IRCSS, Piazza di Sant'Onofrio, 4, 00165, Rome, Italy
| | - Fabiana Cortellessa
- Rare Diseases and Medical Genetics Unit, University-Hospital Pediatric Department (DPUO), Bambino Gesù Children's Hospital, IRCSS, Piazza di Sant'Onofrio, 4, 00165, Rome, Italy
| | - Cristina Caciolo
- Child and Adolescent Psychiatric Unit, Department of Neuroscience, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Marcella Zollino
- Genetica Medica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,Dipartimento Universitario Scienze della Vita e Sanità Pubblica, Sezione di Medicina Genomica, Università Cattolica del Sacro Cuore Facoltà di Medicina e Chirurgia, Rome, Italy
| | - Maria Accadia
- Medical Genetics Service, Hospital "Cardinale G. Panico", Tricase, Lecce, Italy
| | - Marco Seri
- Unit of Medical Genetics, IRCCS Azienda Ospedaliero Universitaria di Bologna, Bologna, Italy
| | - Marcello Chinali
- Department of Pediatric Cardiology and Cardiac Surgery, Bambino Gesù Children's Hospital IRCSS, Rome, Italy
| | - Corrado Mammì
- Operative Unit of Medical Genetics, Bianchi-Melacrino-Morelli Hospital, V. Melacrino, 89100, Reggio Calabria, Italy
| | - Marco Tartaglia
- Genetics and Rare Disease Research Division, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Andrea Bartuli
- Rare Diseases and Medical Genetics Unit, University-Hospital Pediatric Department (DPUO), Bambino Gesù Children's Hospital, IRCSS, Piazza di Sant'Onofrio, 4, 00165, Rome, Italy
| | - Paolo Alfieri
- Child and Adolescent Psychiatric Unit, Department of Neuroscience, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Manuela Priolo
- Operative Unit of Medical Genetics, Bianchi-Melacrino-Morelli Hospital, V. Melacrino, 89100, Reggio Calabria, Italy.
| |
Collapse
|
11
|
Partyka LM. Anesthetic Considerations in an Infant With Malan Syndrome: A Case Report. A A Pract 2022; 16:e01600. [DOI: 10.1213/xaa.0000000000001600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
12
|
Marques P, Korbonits M. Approach to the Patient With Pseudoacromegaly. J Clin Endocrinol Metab 2022; 107:1767-1788. [PMID: 34792134 DOI: 10.1210/clinem/dgab789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Indexed: 11/19/2022]
Abstract
Pseudoacromegaly encompasses a heterogeneous group of conditions in which patients have clinical features of acromegaly or gigantism, but no excess of GH or IGF-1. Acromegaloid physical features or accelerated growth in a patient may prompt referral to endocrinologists. Because pseudoacromegaly conditions are rare and heterogeneous, often with overlapping clinical features, the underlying diagnosis may be challenging to establish. As many of these have a genetic origin, such as pachydermoperiostosis, Sotos syndrome, Weaver syndrome, or Cantú syndrome, collaboration is key with clinical geneticists in the diagnosis of these patients. Although rare, awareness of these uncommon conditions and their characteristic features will help their timely recognition.
Collapse
Affiliation(s)
- Pedro Marques
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, EC1M 6BQ London, UK
- Endocrinology Department, Hospital de Santa Maria, Centro Hospitalar Universitário de Lisboa Norte, Lisboa, Portugal
| | - Márta Korbonits
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, EC1M 6BQ London, UK
| |
Collapse
|
13
|
Xiang J, Ding Y, Yang F, Gao A, Zhang W, Tang H, Mao J, He Q, Zhang Q, Wang T. Genetic Analysis of Children With Unexplained Developmental Delay and/or Intellectual Disability by Whole-Exome Sequencing. Front Genet 2021; 12:738561. [PMID: 34858471 PMCID: PMC8631448 DOI: 10.3389/fgene.2021.738561] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 10/07/2021] [Indexed: 11/25/2022] Open
Abstract
Background: Whole-exome sequencing (WES) has been recommended as a first-tier clinical diagnostic test for individuals with neurodevelopmental disorders (NDDs). We aimed to identify the genetic causes of 17 children with developmental delay (DD) and/or intellectual disability (ID). Methods: WES and exome-based copy number variation (CNV) analysis were performed for 17 patients with unexplained DD/ID. Results: Single-nucleotide variant (SNV)/small insertion or deletion (Indel) analysis and exome-based CNV calling yielded an overall diagnostic rate of 58.8% (10/17), of which diagnostic SNVs/Indels accounted for 41.2% (7/17) and diagnostic CNVs accounted for 17.6% (3/17). Conclusion: Our findings expand the known mutation spectrum of genes related to DD/ID and indicate that exome-based CNV analysis could improve the diagnostic yield of patients with DD/ID.
Collapse
Affiliation(s)
- Jingjing Xiang
- Center for Reproduction and Genetics, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China.,Center for Reproduction and Genetics, Suzhou Municipal Hospital, Suzhou, China
| | - Yang Ding
- Center for Reproduction and Genetics, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China.,Center for Reproduction and Genetics, Suzhou Municipal Hospital, Suzhou, China
| | - Fei Yang
- Center for Reproduction and Genetics, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China.,Center for Reproduction and Genetics, Suzhou Municipal Hospital, Suzhou, China
| | - Ang Gao
- Center for Reproduction and Genetics, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China.,Center for Reproduction and Genetics, Suzhou Municipal Hospital, Suzhou, China
| | - Wei Zhang
- Center for Reproduction and Genetics, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China.,Center for Reproduction and Genetics, Suzhou Municipal Hospital, Suzhou, China
| | - Hui Tang
- Center for Reproduction and Genetics, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China.,Center for Reproduction and Genetics, Suzhou Municipal Hospital, Suzhou, China
| | - Jun Mao
- Center for Reproduction and Genetics, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China.,Center for Reproduction and Genetics, Suzhou Municipal Hospital, Suzhou, China
| | - Quanze He
- Center for Reproduction and Genetics, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China.,Center for Reproduction and Genetics, Suzhou Municipal Hospital, Suzhou, China
| | - Qin Zhang
- Center for Reproduction and Genetics, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China.,Center for Reproduction and Genetics, Suzhou Municipal Hospital, Suzhou, China
| | - Ting Wang
- Center for Reproduction and Genetics, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China.,Center for Reproduction and Genetics, Suzhou Municipal Hospital, Suzhou, China
| |
Collapse
|
14
|
Billar RJ, Manoubi W, Kant SG, Wijnen RMH, Demirdas S, Schnater JM. Association between pectus excavatum and congenital genetic disorders: A systematic review and practical guide for the treating physician. J Pediatr Surg 2021; 56:2239-2252. [PMID: 34039477 DOI: 10.1016/j.jpedsurg.2021.04.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 04/13/2021] [Accepted: 04/18/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Pectus excavatum (PE) could be part of a genetic disorder, which then has implications regarding comorbidity, the surgical correction of PE, and reproductive choices. However, referral of a patient presenting with PE for genetic analysis is often delayed because additional crucial clinical signs may be subtle or even missed in syndromic patients. We reviewed the literature to inventory known genetic disorders associated with PE and create a standardized protocol for clinical evaluation. METHODS A systematic literature search was performed in electronic databases. Genetic disorders were considered associated with PE if studies reported at least five cases with PE. Characteristics of each genetic disorder were extracted from the literature and the OMIM database in order to create a practical guide for the clinician. RESULTS After removal of duplicates from the initial search, 1632 citations remained. Eventually, we included 119 full text articles, representing 20 different genetic disorders. Relevant characteristics and important clinical signs of each genetic disorder were summarized providing a standardized protocol in the form of a scoring list. The most important clinical sign was a positive family history for PE and/or congenital heart defect. CONCLUSIONS Twenty unique genetic disorders have been found associated with PE. We have created a scoring list for the clinician that systematically evaluates crucial clinical signs, thereby facilitating decision making for referral to a clinical geneticist.
Collapse
Affiliation(s)
- Ryan J Billar
- Erasmus University Medical Center - Sophia Children's Hospital, department of Paediatric Surgery Rotterdam, Netherlands
| | - Wiem Manoubi
- Erasmus University Medical Centre, department of Neuroscience, Rotterdam, Netherlands
| | - Sarina G Kant
- Erasmus University Medical Centre, department of Clinical Genetics, Rotterdam, Netherlands
| | - René M H Wijnen
- Erasmus University Medical Center - Sophia Children's Hospital, department of Paediatric Surgery Rotterdam, Netherlands
| | - Serwet Demirdas
- Erasmus University Medical Centre, department of Clinical Genetics, Rotterdam, Netherlands
| | - Johannes M Schnater
- Erasmus University Medical Center - Sophia Children's Hospital, department of Paediatric Surgery Rotterdam, Netherlands.
| |
Collapse
|
15
|
Ocular manifestations of Beckwith-Wiedemann syndrome. J AAPOS 2021; 25:303-305.e1. [PMID: 34582951 DOI: 10.1016/j.jaapos.2021.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 04/12/2021] [Accepted: 04/25/2021] [Indexed: 11/22/2022]
Abstract
Beckwith-Wiedemann syndrome (BWS; OMIM #130650) is a pediatric overgrowth disorder with few known ocular manifestations. We retrospectively reviewed the medical records of patients with BWS evaluated at Bascom Palmer Eye Institute over a 10-year period and identified 5 patients, of whom 4 presented with ocular misalignment and 1 with eye rubbing. Three patients were noted to have strabismus, and 1 patient manifested with significant astigmatism. No patients received surgical intervention.
Collapse
|
16
|
Tønne E, Due-Tønnessen BJ, Mero IL, Wiig US, Kulseth MA, Vigeland MD, Sheng Y, von der Lippe C, Tveten K, Meling TR, Helseth E, Heimdal KR. Benefits of clinical criteria and high-throughput sequencing for diagnosing children with syndromic craniosynostosis. Eur J Hum Genet 2021; 29:920-929. [PMID: 33288889 PMCID: PMC8187391 DOI: 10.1038/s41431-020-00788-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 11/04/2020] [Accepted: 11/20/2020] [Indexed: 12/13/2022] Open
Abstract
An accurate diagnosis of syndromic craniosynostosis (CS) is important for personalized treatment, surveillance, and genetic counselling. We describe detailed clinical criteria for syndromic CS and the distribution of genetic diagnoses within the cohort. The prospective registry of the Norwegian National Unit for Craniofacial Surgery was used to retrieve individuals with syndromic CS born between 1 January 2002 and 30 June 2019. All individuals were assessed by a clinical geneticist and classified using defined clinical criteria. A stepwise approach consisting of single-gene analysis, comparative genomic hybridization (aCGH), and exome-based high-throughput sequencing, first filtering for 72 genes associated with syndromic CS, followed by an extended trio-based panel of 1570 genes were offered to all syndromic CS cases. A total of 381 individuals were registered with CS, of whom 104 (27%) were clinically classified as syndromic CS. Using the single-gene analysis, aCGH, and custom-designed panel, a genetic diagnosis was confirmed in 73% of the individuals (n = 94). The diagnostic yield increased to 84% after adding the results from the extended trio-based panel. Common causes of syndromic CS were found in 53 individuals (56%), whereas 26 (28%) had other genetic syndromes, including 17 individuals with syndromes not commonly associated with CS. Only 15 individuals (16%) had negative genetic analyses. Using the defined combination of clinical criteria, we detected among the highest numbers of syndromic CS cases reported, confirmed by a high genetic diagnostic yield of 84%. The observed genetic heterogeneity encourages a broad genetic approach in diagnosing syndromic CS.
Collapse
Affiliation(s)
- Elin Tønne
- Faculty of Medicine, University of Oslo, Oslo, Norway.
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway.
- Norwegian National Unit for Craniofacial Surgery, Oslo University Hospital, Oslo, Norway.
| | - Bernt Johan Due-Tønnessen
- Norwegian National Unit for Craniofacial Surgery, Oslo University Hospital, Oslo, Norway
- Department of Neurosurgery, Oslo University Hospital, Oslo, Norway
| | - Inger-Lise Mero
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Ulrikke Straume Wiig
- Norwegian National Unit for Craniofacial Surgery, Oslo University Hospital, Oslo, Norway
- Department of Neurosurgery, Oslo University Hospital, Oslo, Norway
| | - Mari Ann Kulseth
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Magnus Dehli Vigeland
- Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Ying Sheng
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Charlotte von der Lippe
- Centre for Rare Disorders, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Department of Medical Genetics, Telemark Hospital Trust, Skien, Norway
| | - Kristian Tveten
- Department of Medical Genetics, Telemark Hospital Trust, Skien, Norway
| | - Torstein Ragnar Meling
- Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Neurosurgery, Oslo University Hospital, Oslo, Norway
- Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Neurosurgery, Geneva University Hospitals, Geneva, Switzerland
| | - Eirik Helseth
- Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Neurosurgery, Oslo University Hospital, Oslo, Norway
| | - Ketil Riddervold Heimdal
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
- Norwegian National Unit for Craniofacial Surgery, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
17
|
Deconte D, dos Santos CBL, de Morais CO, Yonamine TM, Nogueira LT, Ferreira MAT, Franceschi VB, Longhi ALS, Villacis RAR, Rogatto SR, Ligabue-Braun R, Zen PRG, Rosa RFM, Fiegenbaum M. Unusual features in a child with Marshall-Smith syndrome due to a novel NFIX variant: Evidence for an abnormal protein function. GENE REPORTS 2021. [DOI: 10.1016/j.genrep.2020.100991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
18
|
Mulder RH, Neumann A, Cecil CAM, Walton E, Houtepen LC, Simpkin AJ, Rijlaarsdam J, Heijmans BT, Gaunt TR, Felix JF, Jaddoe VWV, Bakermans-Kranenburg MJ, Tiemeier H, Relton CL, van IJzendoorn MH, Suderman M. Epigenome-wide change and variation in DNA methylation in childhood: trajectories from birth to late adolescence. Hum Mol Genet 2021; 30:119-134. [PMID: 33450751 PMCID: PMC8033147 DOI: 10.1093/hmg/ddaa280] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 12/03/2020] [Accepted: 12/23/2020] [Indexed: 12/21/2022] Open
Abstract
DNA methylation (DNAm) is known to play a pivotal role in childhood health and development, but a comprehensive characterization of genome-wide DNAm trajectories across this age period is currently lacking. We have therefore performed a series of epigenome-wide association studies in 5019 blood samples collected at multiple time-points from birth to late adolescence from 2348 participants of two large independent cohorts. DNAm profiles of autosomal CpG sites (CpGs) were generated using the Illumina Infinium HumanMethylation450 BeadChip. Change over time was widespread, observed at over one-half (53%) of CpGs. In most cases, DNAm was decreasing (36% of CpGs). Inter-individual variation in linear trajectories was similarly widespread (27% of CpGs). Evidence for non-linear change and inter-individual variation in non-linear trajectories was somewhat less common (11 and 8% of CpGs, respectively). Very little inter-individual variation in change was explained by sex differences (0.4% of CpGs) even though sex-specific DNAm was observed at 5% of CpGs. DNAm trajectories were distributed non-randomly across the genome. For example, CpGs with decreasing DNAm were enriched in gene bodies and enhancers and were annotated to genes enriched in immune-developmental functions. In contrast, CpGs with increasing DNAm were enriched in promoter regions and annotated to genes enriched in neurodevelopmental functions. These findings depict a methylome undergoing widespread and often non-linear change throughout childhood. They support a developmental role for DNA methylation that extends beyond birth into late adolescence and has implications for understanding life-long health and disease. DNAm trajectories can be visualized at http://epidelta.mrcieu.ac.uk.
Collapse
Affiliation(s)
- Rosa H Mulder
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.,Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.,Institute of Education and Child Studies, Leiden University, Leiden, The Netherlands
| | - Alexander Neumann
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.,Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.,Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, Canada
| | - Charlotte A M Cecil
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.,Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.,Department of Psychology, Institute of Psychology, Psychiatry and Neuroscience, King's College London, London, UK
| | - Esther Walton
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK.,Department of Psychology, University of Bath, Bath, UK
| | - Lotte C Houtepen
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Andrew J Simpkin
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK.,School of Mathematics, Statistics and Applied Mathematics, National University of Ireland, Galway, Ireland
| | - Jolien Rijlaarsdam
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.,Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Bastiaan T Heijmans
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, The Netherlands
| | - Tom R Gaunt
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Janine F Felix
- Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.,Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Vincent W V Jaddoe
- Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.,Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | | | - Henning Tiemeier
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.,Department of Social and Behavioral Science, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Caroline L Relton
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Marinus H van IJzendoorn
- Department of Psychology, Education and Child Studies, Erasmus University Rotterdam, Rotterdam, The Netherlands.,School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Matthew Suderman
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| |
Collapse
|
19
|
Accogli A, Geraldo AF, Piccolo G, Riva A, Scala M, Balagura G, Salpietro V, Madia F, Maghnie M, Zara F, Striano P, Tortora D, Severino M, Capra V. Diagnostic Approach to Macrocephaly in Children. Front Pediatr 2021; 9:794069. [PMID: 35096710 PMCID: PMC8795981 DOI: 10.3389/fped.2021.794069] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 12/02/2021] [Indexed: 01/19/2023] Open
Abstract
Macrocephaly affects up to 5% of the pediatric population and is defined as an abnormally large head with an occipitofrontal circumference (OFC) >2 standard deviations (SD) above the mean for a given age and sex. Taking into account that about 2-3% of the healthy population has an OFC between 2 and 3 SD, macrocephaly is considered as "clinically relevant" when OFC is above 3 SD. This implies the urgent need for a diagnostic workflow to use in the clinical setting to dissect the several causes of increased OFC, from the benign form of familial macrocephaly and the Benign enlargement of subarachnoid spaces (BESS) to many pathological conditions, including genetic disorders. Moreover, macrocephaly should be differentiated by megalencephaly (MEG), which refers exclusively to brain overgrowth, exceeding twice the SD (3SD-"clinically relevant" megalencephaly). While macrocephaly can be isolated and benign or may be the first indication of an underlying congenital, genetic, or acquired disorder, megalencephaly is most likely due to a genetic cause. Apart from the head size evaluation, a detailed family and personal history, neuroimaging, and a careful clinical evaluation are crucial to reach the correct diagnosis. In this review, we seek to underline the clinical aspects of macrocephaly and megalencephaly, emphasizing the main differential diagnosis with a major focus on common genetic disorders. We thus provide a clinico-radiological algorithm to guide pediatricians in the assessment of children with macrocephaly.
Collapse
Affiliation(s)
- Andrea Accogli
- Division of Medical Genetics, Department of Medicine, McGill University Health Centre, Montreal, QC, Canada
| | - Ana Filipa Geraldo
- Diagnostic Neuroradiology Unit, Imaging Department, Centro Hospitalar Vila Nova de Gaia/Espinho, Vila Nova de Gaia, Portugal
| | - Gianluca Piccolo
- Pediatric Neurology and Neuromuscular Diseases Unit, IRCCS Giannina Gaslini Institute, Genoa, Italy
| | - Antonella Riva
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | - Marcello Scala
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | - Ganna Balagura
- Pediatric Neurology and Neuromuscular Diseases Unit, IRCCS Giannina Gaslini Institute, Genoa, Italy
| | - Vincenzo Salpietro
- Pediatric Neurology and Neuromuscular Diseases Unit, IRCCS Giannina Gaslini Institute, Genoa, Italy.,Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | - Francesca Madia
- Pediatric Clinic and Endocrinology, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Mohamad Maghnie
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy.,Pediatric Clinic and Endocrinology, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Federico Zara
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy.,Medical Genetics Unit, IRCCS Giannina Gaslini Institute, Genoa, Italy
| | - Pasquale Striano
- Pediatric Neurology and Neuromuscular Diseases Unit, IRCCS Giannina Gaslini Institute, Genoa, Italy.,Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | - Domenico Tortora
- Neuroradiology Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | | | - Valeria Capra
- Medical Genetics Unit, IRCCS Giannina Gaslini Institute, Genoa, Italy
| |
Collapse
|
20
|
Mulder PA, van Balkom IDC, Landlust AM, Priolo M, Menke LA, Acero IH, Alkuraya FS, Arias P, Bernardini L, Bijlsma EK, Cole T, Coubes C, Dapia I, Davies S, Di Donato N, Elcioglu NH, Fahrner JA, Foster A, González NG, Huber I, Iascone M, Kaiser AS, Kamath A, Kooblall K, Lapunzina P, Liebelt J, Lynch SA, Maas SM, Mammì C, Mathijssen IB, McKee S, Mirzaa GM, Montgomery T, Neubauer D, Neumann TE, Pintomalli L, Pisanti MA, Plomp AS, Price S, Salter C, Santos-Simarro F, Sarda P, Schanze D, Segovia M, Shaw-Smith C, Smithson S, Suri M, Tatton-Brown K, Tenorio J, Thakker RV, Valdez RM, Van Haeringen A, Van Hagen JM, Zenker M, Zollino M, Dunn WW, Piening S, Hennekam RC. Development, behaviour and sensory processing in Marshall-Smith syndrome and Malan syndrome: phenotype comparison in two related syndromes. JOURNAL OF INTELLECTUAL DISABILITY RESEARCH : JIDR 2020; 64:956-969. [PMID: 33034087 PMCID: PMC8957705 DOI: 10.1111/jir.12787] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 08/19/2020] [Accepted: 09/15/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND Ultrarare Marshall-Smith and Malan syndromes, caused by changes of the gene nuclear factor I X (NFIX), are characterised by intellectual disability (ID) and behavioural problems, although questions remain. Here, development and behaviour are studied and compared in a cross-sectional study, and results are presented with genetic findings. METHODS Behavioural phenotypes are compared of eight individuals with Marshall-Smith syndrome (three male individuals) and seven with Malan syndrome (four male individuals). Long-term follow-up assessment of cognition and adaptive behaviour was possible in three individuals with Marshall-Smith syndrome. RESULTS Marshall-Smith syndrome individuals have more severe ID, less adaptive behaviour, more impaired speech and less reciprocal interaction compared with individuals with Malan syndrome. Sensory processing difficulties occur in both syndromes. Follow-up measurement of cognition and adaptive behaviour in Marshall-Smith syndrome shows different individual learning curves over time. CONCLUSIONS Results show significant between and within syndrome variability. Different NFIX variants underlie distinct clinical phenotypes leading to separate entities. Cognitive, adaptive and sensory impairments are common in both syndromes and increase the risk of challenging behaviour. This study highlights the value of considering behaviour within developmental and environmental context. To improve quality of life, adaptations to environment and treatment are suggested to create a better person-environment fit.
Collapse
Affiliation(s)
- P A Mulder
- Autism Team Northern-Netherlands, Jonx Department of (Youth) Mental Health and Autism, Lentis Psychiatric Institute, Groningen, Netherlands
| | - I D C van Balkom
- Autism Team Northern-Netherlands, Jonx Department of (Youth) Mental Health and Autism, Lentis Psychiatric Institute, Groningen, Netherlands
- Rob Giel Research Centre, Department of Psychiatry, University Medical Center Groningen, Groningen, Netherlands
| | - A M Landlust
- Autism Team Northern-Netherlands, Jonx Department of (Youth) Mental Health and Autism, Lentis Psychiatric Institute, Groningen, Netherlands
| | - M Priolo
- Unità Operativa di Genetica Medica, Grande Ospedale Metropolitano Bianchi-Melacrino-Morelli, Reggio Calabria, Italy
| | - L A Menke
- Department of Paediatrics, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - I H Acero
- Genetics Unit, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - F S Alkuraya
- Saudi Human Genome Project, King Abdulaziz City for Science and Technology, and Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - P Arias
- Institute of Medical and Molecular Genetics (INGEMM), Hospital Universitario La Paz, IdiPAZ, Universidad Autónoma de Madrid, and CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain
| | - L Bernardini
- Cytogenetics Unit, Casa Sollievo della Sofferenza Foundation, San Giovanni Rotondo, Italy
| | - E K Bijlsma
- Department of Clinical Genetics, Leiden University Medical Centre, Leiden, Netherlands
| | - T Cole
- Department of Clinical Genetics, Birmingham Women's and Children's NHS Foundation Trust, Birmingham, UK
| | - C Coubes
- Département de Génétique Médicale, Hôpital Arnaud de Villeneuve, CHRU Montpellier, Montpellier, France
| | - I Dapia
- Institute of Medical and Molecular Genetics (INGEMM), Hospital Universitario La Paz, IdiPAZ, Universidad Autónoma de Madrid, and CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain
| | - S Davies
- Institute of Medical Genetics, University Hospital of Wales, Cardiff, UK
| | - N Di Donato
- Institute for Clinical Genetics, TU Dresden, Dresden, Germany
| | - N H Elcioglu
- Department of Pediatric Genetics, Marmara University Medical School, Istanbul and Eastern Mediterranean University, Mersin, Turkey
| | - J A Fahrner
- McKusick-Nathans Institute of Genetic Medicine, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - A Foster
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - N G González
- Unit Hospital Universitario Central de Asturias, Oviedo, Spain
| | - I Huber
- Sørland Hospital, Kristiansand, Norway
| | - M Iascone
- Medical Genetics Laboratory, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - A-S Kaiser
- Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | - A Kamath
- Institute of Medical Genetics, University Hospital of Wales, Cardiff, UK
| | - K Kooblall
- Academic Endocrine Unit, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - P Lapunzina
- Institute of Medical and Molecular Genetics (INGEMM), Hospital Universitario La Paz, IdiPAZ, Universidad Autónoma de Madrid, and CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain
| | - J Liebelt
- South Australian Clinical Genetics Services, Women's and Children's Hospital, North Adelaide, Australia
| | - S A Lynch
- UCD Academic Centre on Rare Diseases, School of Medicine and Medical Sciences, University College Dublin, and Clinical Genetics, Temple Street Children's University Hospital, Dublin, Ireland
| | - S M Maas
- Department of Clinical Genetics, Academic Medical Center, Amsterdam, Netherlands
| | - C Mammì
- Unità Operativa di Genetica Medica, Grande Ospedale Metropolitano Bianchi-Melacrino-Morelli, Reggio Calabria, Italy
| | - I B Mathijssen
- Department of Clinical Genetics, Academic Medical Center, Amsterdam, Netherlands
| | - S McKee
- Northern Ireland Regional Genetics Service, Belfast Health and Social Care Trust, Belfast, UK
| | - G M Mirzaa
- Center for Integrative Brain Research, Seattle Children's Research Institute, and Division of Genetic Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - T Montgomery
- Newcastle upon Tyne NHS Foundation Trust, Newcastle upon Tyne, UK
| | - D Neubauer
- Institute of Human Genetics, University Hospital Magdeburg, Magdeburg, Germany
| | - T E Neumann
- Mitteldeutscher Praxisverbund Humangenetik, Halle, Germany
| | - L Pintomalli
- Unità Operativa di Genetica Medica, Grande Ospedale Metropolitano Bianchi-Melacrino-Morelli, Reggio Calabria, Italy
| | - M A Pisanti
- Medical Genetic and Laboratory Unit, "Antonio Cardarelli" Hospital, Naples, Italy
| | - A S Plomp
- Department of Clinical Genetics, Academic Medical Center, Amsterdam, Netherlands
| | - S Price
- Department of Clinical Genetics, Northampton General Hospital NHS Trust, Northampton, UK
| | - C Salter
- Wessex Clinical Genetics Service, Princess Ann Hospital, Southampton, UK
| | - F Santos-Simarro
- Institute of Medical and Molecular Genetics (INGEMM), Hospital Universitario La Paz, IdiPAZ, Universidad Autónoma de Madrid, and CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain
| | - P Sarda
- Département de Génétique Médicale, Hôpital Arnaud de Villeneuve, CHRU Montpellier, Montpellier, France
| | - D Schanze
- Institute of Human Genetics, University Hospital Magdeburg, Magdeburg, Germany
| | - M Segovia
- CENAGEM, Centro Nacional de Genética, Buenos Aires, Argentina
| | - C Shaw-Smith
- Royal Devon and Exeter NHS Foundation Trust, Exeter, UK
| | - S Smithson
- University Hospitals Bristol NHS Trust, Bristol, UK
| | - M Suri
- Nottingham Clinical Genetics Service, Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - K Tatton-Brown
- Division of Genetics and Epidemiology, Institute of Cancer Research, London and South West Thames Regional Genetics Service, St. George's University Hospitals NHS Foundation Trust, London, UK
| | - J Tenorio
- Institute of Medical and Molecular Genetics (INGEMM), Hospital Universitario La Paz, IdiPAZ, Universidad Autónoma de Madrid, and CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain
| | - R V Thakker
- Academic Endocrine Unit, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - R M Valdez
- Genetics Unit, Hospital Militar Central "Cirujano Mayor Dr. Cosme Argerich", Buenos Aires, Argentina
| | - A Van Haeringen
- Department of Clinical Genetics, Leiden University Medical Centre, Leiden, Netherlands
| | - J M Van Hagen
- Department of Clinical Genetics, VU University Medical Centre, Amsterdam, Netherlands
| | - M Zenker
- Institute of Human Genetics, University Hospital Magdeburg, Magdeburg, Germany
| | - M Zollino
- Department of Laboratory Medicine, Institute of Medical Genetics, Catholic University, Rome, Italy
| | - W W Dunn
- Department of Occupational Therapy Education, School of Health Professions, University of Missouri, Columbia, MO, USA
| | - S Piening
- Autism Team Northern-Netherlands, Jonx Department of (Youth) Mental Health and Autism, Lentis Psychiatric Institute, Groningen, Netherlands
- Rob Giel Research Centre, Department of Psychiatry, University Medical Center Groningen, Groningen, Netherlands
| | - R C Hennekam
- Autism Team Northern-Netherlands, Jonx Department of (Youth) Mental Health and Autism, Lentis Psychiatric Institute, Groningen, Netherlands
- Department of Paediatrics, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
21
|
Sihombing NRB, Winarni TI, van Bokhoven H, van der Burgt I, de Leeuw N, Faradz SMH. Pathogenic variant in NFIX gene affecting three sisters due to paternal mosaicism. Am J Med Genet A 2020; 182:2731-2736. [PMID: 32945093 DOI: 10.1002/ajmg.a.61835] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 06/14/2020] [Accepted: 06/25/2020] [Indexed: 12/26/2022]
Abstract
We present a family with three girls presenting similar dysmorphic features, including overgrowth, intellectual disability, macrocephaly, prominent forehead, midface retrusion, strabismus, and scoliosis. Both parents were unaffected, suggesting the presence of an autosomal recessive syndrome. Following exome sequencing, a heterozygous nonsense variant was identified in the NFIX gene in all three siblings. The father appeared to have a low-grade (7%) mosaicism for this variant in his blood. Previously, de novo pathogenic variants in NFIX have been identified in Marshall-Smith syndrome and Malan syndrome, which share distinctive phenotypic features shared with the patients of the present family. This case emphasizes the importance of further molecular analysis especially in familial cases, to exclude the possibility of parental mosaicism.
Collapse
Affiliation(s)
- Nydia Rena Benita Sihombing
- Doctoral Program of Medical and Health Sciences, Faculty of Medicine, Diponegoro University, Semarang, Indonesia
| | - Tri Indah Winarni
- Division of Human Genetics, Center for Biomedical Research (CEBIOR), Faculty of Medicine, Diponegoro University, Semarang, Indonesia
| | - Hans van Bokhoven
- Department of Human Genetics, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Ineke van der Burgt
- Department of Human Genetics, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Nicole de Leeuw
- Department of Human Genetics, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Sultana M H Faradz
- Division of Human Genetics, Center for Biomedical Research (CEBIOR), Faculty of Medicine, Diponegoro University, Semarang, Indonesia
| |
Collapse
|
22
|
Expression of NFIA and NFIB within the murine spinal cord. Gene Expr Patterns 2020; 35:119098. [PMID: 32068188 DOI: 10.1016/j.gep.2020.119098] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 02/06/2020] [Accepted: 02/11/2020] [Indexed: 12/17/2022]
Abstract
The Nuclear factor I proteins comprise a family of transcription factors that are expressed in many developing and mature cell populations, including within the central nervous system. Within the embryonic mouse spinal cord, NFIA and NFIB are expressed by neural progenitor cells lining the central canal, where they act to promote astrocytic and oligodendrocytic lineage specification. Cells lining the mature spinal cord central canal retain characteristics of neural progenitor cells, but the expression of NFIA and NFIB within the mature spinal cord at a cell-type-specific level remains undefined. Here, we investigated where these two transcription factors are expressed within the adult mouse spinal cord. We reveal that both factors are expressed in similar cohorts of mature cells, including ependymal cells, interneurons and motor neurons. We also show robust and widespread expression of NFIA and NFIB within nestin-expressing cells following injury to the spinal cord. Collectively, these data provide a basis to further define what functional role(s) NFIA and NFIB play within the adult spinal cord.
Collapse
|
23
|
Oishi S, Zalucki O, Vega MS, Harkins D, Harvey TJ, Kasherman M, Davila RA, Hale L, White M, Piltz S, Thomas P, Burne THJ, Harris L, Piper M. Investigating cortical features of Sotos syndrome using mice heterozygous for Nsd1. GENES BRAIN AND BEHAVIOR 2020; 19:e12637. [PMID: 31909872 DOI: 10.1111/gbb.12637] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 01/03/2020] [Accepted: 01/04/2020] [Indexed: 12/18/2022]
Abstract
Sotos syndrome is a developmental disorder characterized by a suite of clinical features. In children, the three cardinal features of Sotos syndrome are a characteristic facial appearance, learning disability and overgrowth (height and/or head circumference > 2 SDs above average). These features are also evident in adults with this syndrome. Over 90% of Sotos syndrome patients are haploinsufficient for the gene encoding nuclear receptor-binding Su(var)3-9, Enhancer-of-zesteand Trithorax domain-containing protein 1 (NSD1). NSD1 is a histone methyltransferase that catalyzes the methylation of lysine residue 36 on histone H3. However, although the symptomology of Sotos syndrome is well established, many aspects of NSD1 biology remain unknown. Here, we assessed the expression of Nsd1 within the mouse brain, and showed a predominantly neuronal pattern of expression for this histone-modifying factor. We also generated a mouse strain lacking one allele of Nsd1 and analyzed morphological and behavioral characteristics in these mice, showing behavioral characteristics reminiscent of some of the deficits seen in Sotos syndrome patients.
Collapse
Affiliation(s)
- Sabrina Oishi
- The School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Oressia Zalucki
- The School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Michelle S Vega
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - Danyon Harkins
- The School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Tracey J Harvey
- The School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Maria Kasherman
- The School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Raul A Davila
- The School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Lauren Hale
- The School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Melissa White
- School of Biological Sciences and South Australia Genome Editing Facility, University of Adelaide, Adelaide, South Australia, Australia
| | - Sandra Piltz
- School of Biological Sciences and South Australia Genome Editing Facility, University of Adelaide, Adelaide, South Australia, Australia
| | - Paul Thomas
- School of Biological Sciences and South Australia Genome Editing Facility, University of Adelaide, Adelaide, South Australia, Australia
| | - Thomas H J Burne
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia.,Queensland Centre for Mental Health Research, The Park Centre for Mental Health, Wacol, Queensland, Australia
| | - Lachlan Harris
- The School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Michael Piper
- The School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia.,Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
24
|
Conditions of embryo culture from days 5 to 7 of development alter the DNA methylome of the bovine fetus at day 86 of gestation. J Assist Reprod Genet 2019; 37:417-426. [PMID: 31838628 DOI: 10.1007/s10815-019-01652-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 12/03/2019] [Indexed: 01/30/2023] Open
Abstract
PURPOSE We tested whether in vitro production (IVP) causes changes in DNA methylation in fetal liver and skeletal muscle and if exposure of cultured embryos to colony-stimulating factor 2 (CSF2) alters DNA methylation. METHODS Female fetuses were produced by artificial insemination or transfer of an IVP embryo. Embryos were treated from days 5 to 7 after fertilization with CSF2 or vehicle. DNA methylation in fetal liver and skeletal muscle was determined by post-bisulfite adaptor tagging-based sequencing. The degree of DNA methylation for CpG sites in 50-bp windows of the promoter region 500 bp upstream of the transcriptional start site was compared between treatments. RESULTS For liver, there were 12 genes (6% of those analyzed) in which DNA methylation was affected by treatment, with one 50-bp window per gene affected by treatment. For muscle, the degree of DNA methylation was affected by treatment for 32 windows (19% of the total windows analyzed) representing 28 distinct genes (23% of analyzed genes). For 19 of the 28 genes in muscle, the greatest deviation in DNA methylation was for the CSF2 group. CONCLUSION Results are consistent with alterations in the methylome being one of the mechanisms by which IVP can result in altered fetal development and postnatal function in the resultant offspring. In addition, results indicate that maternally derived cell-signaling molecules can regulate the pattern of DNA methylation.
Collapse
|
25
|
Bellucco FT, de Mello CB, Meloni VA, Melaragno MI. Malan syndrome in a patient with 19p13.2p13.12 deletion encompassing NFIX and CACNA1A genes: Case report and review of the literature. Mol Genet Genomic Med 2019; 7:e997. [PMID: 31574590 PMCID: PMC6900369 DOI: 10.1002/mgg3.997] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 09/16/2019] [Indexed: 11/16/2022] Open
Abstract
Background Malan syndrome is a recently introduced overgrowth disorder described in a limited number of individuals. Haploinsufficiency and also point mutations of NFIX gene have been proposed as its leading causative mechanism, however, due to the limited number of cases and different deletion sizes, genotype/phenotype correlations are still limited. Methods Here, we report the first Brazilian case of Malan syndrome caused by a 990 kb deletion in 19p13.2p13.12, focusing on clinical and behavioral aspects of the syndrome. Results The patient presented with macrocephaly, facial dysmorphisms, hypotonia, developmental delay, moderate thoracolumbar scoliosis, and seizures. The intellectual and behavioral assessments showed severe cognitive, language, and adaptive functions impairments. The 19p deleted region of our patient encompasses NFIX, CACNA1A, which seems to be related to a higher frequency of seizures among individuals with microdeletions in 19p13.2, and 15 other coding genes, including CC2D1A and NACC1, both known to be involved in neurobiological process and pathways. Conclusion Deletions involving NFIX gene should be considered in patients with overgrowth during childhood, macrocephaly, developmental delay, and seizures, as well as severe intellectual disability.
Collapse
Affiliation(s)
- Fernanda T Bellucco
- Department of Morphology and Genetics, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Claudia B de Mello
- Department of Psychobiology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Vera A Meloni
- Department of Morphology and Genetics, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Maria Isabel Melaragno
- Department of Morphology and Genetics, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
26
|
Brioude F, Toutain A, Giabicani E, Cottereau E, Cormier-Daire V, Netchine I. Overgrowth syndromes - clinical and molecular aspects and tumour risk. Nat Rev Endocrinol 2019; 15:299-311. [PMID: 30842651 DOI: 10.1038/s41574-019-0180-z] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Overgrowth syndromes are a heterogeneous group of rare disorders characterized by generalized or segmental excessive growth commonly associated with additional features, such as visceromegaly, macrocephaly and a large range of various symptoms. These syndromes are caused by either genetic or epigenetic anomalies affecting factors involved in cell proliferation and/or the regulation of epigenetic markers. Some of these conditions are associated with neurological anomalies, such as cognitive impairment or autism. Overgrowth syndromes are frequently associated with an increased risk of cancer (embryonic tumours during infancy or carcinomas during adulthood), but with a highly variable prevalence. Given this risk, syndrome-specific tumour screening protocols have recently been established for some of these conditions. Certain specific clinical traits make it possible to discriminate between different syndromes and orient molecular explorations to determine which molecular tests to conduct, despite the syndromes having overlapping clinical features. Recent advances in molecular techniques using next-generation sequencing approaches have increased the number of patients with an identified molecular defect (especially patients with segmental overgrowth). This Review discusses the clinical and molecular diagnosis, tumour risk and recommendations for tumour screening for the most prevalent generalized and segmental overgrowth syndromes.
Collapse
Affiliation(s)
- Frédéric Brioude
- Sorbonne Université, INSERM UMR_S938, Centre de Recherche Saint Antoine, AP-HP Hôpital Trousseau, Paris, France.
| | - Annick Toutain
- CHU de Tours, Hôpital Bretonneau, Service de Génétique, INSERM UMR1253, iBrain, Université de Tours, Faculté de Médecine, Tours, France
| | - Eloise Giabicani
- Sorbonne Université, INSERM UMR_S938, Centre de Recherche Saint Antoine, AP-HP Hôpital Trousseau, Paris, France
| | - Edouard Cottereau
- CHU de Tours, Hôpital Bretonneau, Service de Génétique, Tours, France
| | - Valérie Cormier-Daire
- Service de génétique clinique, Université Paris Descartes-Sorbonne Paris Cité, INSERM UMR1163, Institut Imagine, Hôpital Necker-Enfants Malades, Paris, France
| | - Irene Netchine
- Sorbonne Université, INSERM UMR_S938, Centre de Recherche Saint Antoine, AP-HP Hôpital Trousseau, Paris, France
| |
Collapse
|
27
|
Oishi S, Harkins D, Kurniawan ND, Kasherman M, Harris L, Zalucki O, Gronostajski RM, Burne THJ, Piper M. Heterozygosity for Nuclear Factor One X in mice models features of Malan syndrome. EBioMedicine 2019; 39:388-400. [PMID: 30503862 PMCID: PMC6354567 DOI: 10.1016/j.ebiom.2018.11.044] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 11/16/2018] [Accepted: 11/20/2018] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Nuclear Factor One X (NFIX) haploinsufficiency in humans results in Malan syndrome, a disorder characterized by overgrowth, macrocephaly and intellectual disability. Although clinical assessments have determined the underlying symptomology of Malan syndrome, the fundamental mechanisms contributing to the enlarged head circumference and intellectual disability in these patients remains undefined. METHODS Here, we used Nfix heterozygous mice as a model to investigate these aspects of Malan syndrome. Volumetric magnetic resonance imaging (MRI) was used to calculate the volumes of 20 brain sub regions. Diffusion tensor MRI was used to perform tractography-based analyses of the corpus callosum, hippocampal commissure, and anterior commissure, as well as structural connectome mapping of the whole brain. Immunohistochemistry examined the neocortical cellular populations. Two behavioral assays were performed, including the active place avoidance task to assess spatial navigation and learning and memory function, and the 3-chambered sociability task to examine social behaviour. FINDINGS Adult Nfix+/- mice exhibit significantly increased brain volume (megalencephaly) compared to wildtypes, with the cerebral cortex showing the highest increase. Moreover, all three forebrain commissures, in particular the anterior commissure, revealed significantly reduced fractional anisotropy, axial and radial diffusivity, and tract density intensity. Structural connectome analyses revealed aberrant connectivity between many crucial brain regions. Finally, Nfix+/- mice exhibit behavioral deficits that model intellectual disability. INTERPRETATION Collectively, these data provide a significant conceptual advance in our understanding of Malan syndrome by suggesting that megalencephaly underlies the enlarged head size of these patients, and that disrupted cortical connectivity may contribute to the intellectual disability these patients exhibit. FUND: Australian Research Council (ARC) Discovery Project Grants, ARC Fellowship, NYSTEM and Australian Postgraduate Fellowships.
Collapse
Affiliation(s)
- Sabrina Oishi
- The School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Danyon Harkins
- The School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Nyoman D Kurniawan
- The Centre for Advanced Imaging, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Maria Kasherman
- The School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia; Griffith Institute for Drug Discovery, Griffith University, Brisbane, QLD 4111, Australia
| | - Lachlan Harris
- The School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia; The Francis Crick Institute, 1 Midland Road, King's Cross, London, United Kingdom
| | - Oressia Zalucki
- The School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Richard M Gronostajski
- Department of Biochemistry, Program in Genetics, Genomics and Bioinformatics, Center of Excellence in Bioinformatics and Life Sciences, State University of New York at Buffalo, Buffalo, NY 14203, USA
| | - Thomas H J Burne
- The Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia; Queensland Centre for Mental Health Research, The Park Centre for Mental Health, Wacol, Brisbane, QLD 4076, Australia
| | - Michael Piper
- The School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia; The Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia.
| |
Collapse
|
28
|
Priolo M. Nuclear Factor One X Mice model for Malan syndrome: the less the better. EBioMedicine 2019; 39:15-16. [PMID: 30529069 PMCID: PMC6355653 DOI: 10.1016/j.ebiom.2018.11.065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 11/30/2018] [Indexed: 11/29/2022] Open
Affiliation(s)
- Manuela Priolo
- Operative Unite of Medical Genetics, Great Metropolitan Hospital Bianchi-Melacrino-Morelli, Reggio Calabria, Italy.
| |
Collapse
|
29
|
Marques P, Korbonits M. Pseudoacromegaly. Front Neuroendocrinol 2019; 52:113-143. [PMID: 30448536 DOI: 10.1016/j.yfrne.2018.11.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 10/30/2018] [Accepted: 11/14/2018] [Indexed: 01/19/2023]
Abstract
Individuals with acromegaloid physical appearance or tall stature may be referred to endocrinologists to exclude growth hormone (GH) excess. While some of these subjects could be healthy individuals with normal variants of growth or physical traits, others will have acromegaly or pituitary gigantism, which are, in general, straightforward diagnoses upon assessment of the GH/IGF-1 axis. However, some patients with physical features resembling acromegaly - usually affecting the face and extremities -, or gigantism - accelerated growth/tall stature - will have no abnormalities in the GH axis. This scenario is termed pseudoacromegaly, and its correct diagnosis can be challenging due to the rarity and variability of these conditions, as well as due to significant overlap in their characteristics. In this review we aim to provide a comprehensive overview of pseudoacromegaly conditions, highlighting their similarities and differences with acromegaly and pituitary gigantism, to aid physicians with the diagnosis of patients with pseudoacromegaly.
Collapse
Affiliation(s)
- Pedro Marques
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Márta Korbonits
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK.
| |
Collapse
|
30
|
Corredor B, Dattani M, Gertosio C, Bozzola M. Tall Stature: A Challenge for Clinicians. Curr Pediatr Rev 2019; 15:10-21. [PMID: 30394212 PMCID: PMC6696825 DOI: 10.2174/1573396314666181105092917] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 10/12/2018] [Accepted: 10/29/2018] [Indexed: 02/07/2023]
Abstract
Clinicians generally use the term "tall stature" to define a height more than two standard deviations above the mean for age and sex. In most cases, these subjects present with familial tall stature or a constitutional advance of growth which is diagnosed by excluding the other conditions associated with overgrowth. Nevertheless, it is necessary to be able to identify situations in which tall stature or an accelerated growth rate indicate an underlying disorder. A careful physical evaluation allows the classification of tall patients into two groups: those with a normal appearance and those with an abnormal appearance including disproportion or dysmorphism. In the first case, the growth rate has to be evaluated and, if it is normal for age and sex, the subjects may be considered as having familial tall stature or constitutional advance of growth or they may be obese, while if the growth rate is increased, pubertal status and thyroid function should be evaluated. In turn, tall subjects having an abnormal appearance can be divided into proportionate and disproportionate syndromic patients. Before initiating further investigations, the clinician needs to perform both a careful physical examination and growth evaluation. To exclude pathological conditions, the cause of tall stature needs to be considered, although most children are healthy and generally do not require treatment to inhibit growth progression. In particular cases, familial tall stature subject can be treated by inducing puberty early and leading to a complete fusion of the epiphyses, so final height is reached. This review aims to provide proposals about the management of tall children.
Collapse
Affiliation(s)
| | | | | | - Mauro Bozzola
- Address correspondence to this author at the Department of Internal Medicine and Therapeutics, Pediatrics and Adolescent Care Unit, University of Pavia, Strada Nuova 65, 27100 Pavia, Italy; Tel: +39 339 5469483;
E-mail:
| |
Collapse
|
31
|
Rai A, Narayanan DL, Phadke SR. Malan syndrome: Extension of genotype and phenotype spectrum. Am J Med Genet A 2018; 176:2896-2900. [PMID: 30548146 DOI: 10.1002/ajmg.a.40663] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 07/09/2018] [Accepted: 09/18/2018] [Indexed: 01/02/2023]
Abstract
Malan syndrome and Marshall-Smith syndrome (MSS) are allelic disorders caused by mutation in NFIX gene. We report a 3-year- 6 months- old female with clinical features suggestive of Malan syndrome with mutation in exon 2 of NFIX gene. NFIX gene, where most of the mutations in Malan syndrome are located. She did not have advanced bone age. The radiographs of long bones showed metaphyseal changes which were not reported previously. This study reports the first mutation proven case from India and highlights the overlap between MSS and Malan syndrome.
Collapse
Affiliation(s)
- Archana Rai
- Department of Medical Genetics, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Dhanya Lakshmi Narayanan
- Department of Medical Genetics, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Shubha R Phadke
- Department of Medical Genetics, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| |
Collapse
|
32
|
Fraser J, Essebier A, Brown AS, Davila RA, Sengar AS, Tu Y, Ensbey KS, Day BW, Scott MP, Gronostajski RM, Wainwright BJ, Boden M, Harvey TJ, Piper M. Granule neuron precursor cell proliferation is regulated by NFIX and intersectin 1 during postnatal cerebellar development. Brain Struct Funct 2018; 224:811-827. [PMID: 30511336 DOI: 10.1007/s00429-018-1801-3] [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: 03/08/2018] [Accepted: 11/24/2018] [Indexed: 01/06/2023]
Abstract
Cerebellar granule neurons are the most numerous neuronal subtype in the central nervous system. Within the developing cerebellum, these neurons are derived from a population of progenitor cells found within the external granule layer of the cerebellar anlage, namely the cerebellar granule neuron precursors (GNPs). The timely proliferation and differentiation of these precursor cells, which, in rodents occurs predominantly in the postnatal period, is tightly controlled to ensure the normal morphogenesis of the cerebellum. Despite this, our understanding of the factors mediating how GNP differentiation is controlled remains limited. Here, we reveal that the transcription factor nuclear factor I X (NFIX) plays an important role in this process. Mice lacking Nfix exhibit reduced numbers of GNPs during early postnatal development, but elevated numbers of these cells at postnatal day 15. Moreover, Nfix-/- GNPs exhibit increased proliferation when cultured in vitro, suggestive of a role for NFIX in promoting GNP differentiation. At a mechanistic level, profiling analyses using both ChIP-seq and RNA-seq identified the actin-associated factor intersectin 1 as a downstream target of NFIX during cerebellar development. In support of this, mice lacking intersectin 1 also displayed delayed GNP differentiation. Collectively, these findings highlight a key role for NFIX and intersectin 1 in the regulation of cerebellar development.
Collapse
Affiliation(s)
- James Fraser
- The School of Biomedical Sciences, The University of Queensland, Brisbane, 4072, Australia
| | - Alexandra Essebier
- The School of Chemistry and Molecular Bioscience, The University of Queensland, Brisbane, 4072, Australia
| | - Alexander S Brown
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Raul Ayala Davila
- The School of Biomedical Sciences, The University of Queensland, Brisbane, 4072, Australia
| | - Ameet S Sengar
- Program in Neurosciences & Mental Health, The Hospital for Sick Children, Toronto, M5G 0A8, Canada
| | - YuShan Tu
- Program in Neurosciences & Mental Health, The Hospital for Sick Children, Toronto, M5G 0A8, Canada
| | - Kathleen S Ensbey
- Cell and Molecular Biology Department, Translational Brain Cancer Research Laboratory, QIMR Berghofer MRI, Brisbane, QLD, 4006, Australia
| | - Bryan W Day
- Cell and Molecular Biology Department, Translational Brain Cancer Research Laboratory, QIMR Berghofer MRI, Brisbane, QLD, 4006, Australia
| | - Matthew P Scott
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Richard M Gronostajski
- Department of Biochemistry, Program in Genetics, Genomics and Bioinformatics, Center of Excellence in Bioinformatics and Life Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Brandon J Wainwright
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, 4072, Australia
| | - Mikael Boden
- The School of Chemistry and Molecular Bioscience, The University of Queensland, Brisbane, 4072, Australia
| | - Tracey J Harvey
- The School of Biomedical Sciences, The University of Queensland, Brisbane, 4072, Australia.
| | - Michael Piper
- The School of Biomedical Sciences, The University of Queensland, Brisbane, 4072, Australia. .,Queensland Brain Institute, The University of Queensland, Brisbane, 4072, Australia.
| |
Collapse
|
33
|
Schanze I, Bunt J, Lim JWC, Schanze D, Dean RJ, Alders M, Blanchet P, Attié-Bitach T, Berland S, Boogert S, Boppudi S, Bridges CJ, Cho MT, Dobyns WB, Donnai D, Douglas J, Earl DL, Edwards TJ, Faivre L, Fregeau B, Genevieve D, Gérard M, Gatinois V, Holder-Espinasse M, Huth SF, Izumi K, Kerr B, Lacaze E, Lakeman P, Mahida S, Mirzaa GM, Morgan SM, Nowak C, Peeters H, Petit F, Pilz DT, Puechberty J, Reinstein E, Rivière JB, Santani AB, Schneider A, Sherr EH, Smith-Hicks C, Wieland I, Zackai E, Zhao X, Gronostajski RM, Zenker M, Richards LJ. NFIB Haploinsufficiency Is Associated with Intellectual Disability and Macrocephaly. Am J Hum Genet 2018; 103:752-768. [PMID: 30388402 PMCID: PMC6218805 DOI: 10.1016/j.ajhg.2018.10.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 10/03/2018] [Indexed: 12/19/2022] Open
Abstract
The nuclear factor I (NFI) family of transcription factors play an important role in normal development of multiple organs. Three NFI family members are highly expressed in the brain, and deletions or sequence variants in two of these, NFIA and NFIX, have been associated with intellectual disability (ID) and brain malformations. NFIB, however, has not previously been implicated in human disease. Here, we present a cohort of 18 individuals with mild ID and behavioral issues who are haploinsufficient for NFIB. Ten individuals harbored overlapping microdeletions of the chromosomal 9p23-p22.2 region, ranging in size from 225 kb to 4.3 Mb. Five additional subjects had point sequence variations creating a premature termination codon, and three subjects harbored single-nucleotide variations resulting in an inactive protein as determined using an in vitro reporter assay. All individuals presented with additional variable neurodevelopmental phenotypes, including muscular hypotonia, motor and speech delay, attention deficit disorder, autism spectrum disorder, and behavioral abnormalities. While structural brain anomalies, including dysgenesis of corpus callosum, were variable, individuals most frequently presented with macrocephaly. To determine whether macrocephaly could be a functional consequence of NFIB disruption, we analyzed a cortex-specific Nfib conditional knockout mouse model, which is postnatally viable. Utilizing magnetic resonance imaging and histology, we demonstrate that Nfib conditional knockout mice have enlargement of the cerebral cortex but preservation of overall brain structure and interhemispheric connectivity. Based on our findings, we propose that haploinsufficiency of NFIB causes ID with macrocephaly.
Collapse
Affiliation(s)
- Ina Schanze
- Institute of Human Genetics, University Hospital Magdeburg, Otto-von-Guericke University, Magdeburg 39120, Germany
| | - Jens Bunt
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia.
| | - Jonathan W C Lim
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Denny Schanze
- Institute of Human Genetics, University Hospital Magdeburg, Otto-von-Guericke University, Magdeburg 39120, Germany
| | - Ryan J Dean
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Marielle Alders
- Department of Clinical Genetics, Academic Medical Center, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands
| | - Patricia Blanchet
- INSERM U1183, Département de Génétique Médicale, Maladies Rares et Médecine Personnalisée, Génétique clinique, CHU Montpellier, Université Montpellier, Centre de référence anomalies du développement SORO, Montpellier 34295, France
| | - Tania Attié-Bitach
- INSERM U1163, Laboratory of Embryology and Genetics of Congenital Malformations, Paris Descartes University, Sorbonne Paris Cité and Imagine Institute, Paris 75015, France
| | - Siren Berland
- Department of Medical Genetics, Haukeland University Hospital, Bergen 5021, Norway
| | - Steven Boogert
- Institute of Human Genetics, University Hospital Magdeburg, Otto-von-Guericke University, Magdeburg 39120, Germany
| | - Sangamitra Boppudi
- Institute of Human Genetics, University Hospital Magdeburg, Otto-von-Guericke University, Magdeburg 39120, Germany
| | - Caitlin J Bridges
- Institute of Human Genetics, University Hospital Magdeburg, Otto-von-Guericke University, Magdeburg 39120, Germany
| | | | - William B Dobyns
- Department of Pediatrics (Genetics), University of Washington and Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Dian Donnai
- Manchester Centre for Genomic Medicine, Manchester Academic Health Science Centre, Central Manchester University Hospitals NHS Foundation Trust; Division of Evolution and Genomic Sciences School of Biological Sciences, and University of Manchester, Manchester M13 9WL, UK
| | - Jessica Douglas
- Boston Children's Hospital - The Feingold Center, Waltham, MA 02115, USA
| | - Dawn L Earl
- Division of Genetic Medicine, Seattle Children's Hospital, Seattle, WA 98105, USA
| | - Timothy J Edwards
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia; The Faculty of Medicine Brisbane, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Laurence Faivre
- UMR1231, Génétique des Anomalies du Développement, Université de Bourgogne, Dijon 21079, France; Centre de Génétique et Centre de Référence Anomalies du Développement et Syndromes Malformatifs de l'Interrégion Est et FHU TRANSLAD, Centre Hospitalier Universitaire Dijon, Dijon 21079, France
| | - Brieana Fregeau
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - David Genevieve
- INSERM U1183, Département de Génétique Médicale, Maladies Rares et Médecine Personnalisée, Génétique clinique, CHU Montpellier, Université Montpellier, Centre de référence anomalies du développement SORO, Montpellier 34295, France
| | - Marion Gérard
- Service de Génétique, CHU de Caen - Hôpital Clémenceau, Caen Cedex 14000, France
| | - Vincent Gatinois
- INSERM U1183, Département de Génétique Médicale, Maladies Rares et Médecine Personnalisée, Génétique clinique, CHU Montpellier, Université Montpellier, Centre de référence anomalies du développement SORO, Montpellier 34295, France
| | - Muriel Holder-Espinasse
- Service de Génétique Clinique, Hôpital Jeanne de Flandre, CHU Lille, Lille 59000, France; Department of Clinical Genetics, Guy's Hospital, London SE1 9RT, UK
| | - Samuel F Huth
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Kosuke Izumi
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Division of Genetics, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Bronwyn Kerr
- Manchester Centre for Genomic Medicine, Manchester Academic Health Science Centre, Central Manchester University Hospitals NHS Foundation Trust; Division of Evolution and Genomic Sciences School of Biological Sciences, and University of Manchester, Manchester M13 9WL, UK
| | - Elodie Lacaze
- Department of genetics, Le Havre Hospital, 76600 Le Havre, France
| | - Phillis Lakeman
- Department of Clinical Genetics, Academic Medical Center, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands
| | - Sonal Mahida
- Department of Neurogenetics, Kennedy Krieger Institute, Baltimore, MD 21205, USA
| | - Ghayda M Mirzaa
- Department of Pediatrics (Genetics), University of Washington and Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Sian M Morgan
- All Wales Genetics Laboratory, Institute of Medical Genetics, University Hospital of Wales, Cardiff CF14 4XW, UK
| | - Catherine Nowak
- Boston Children's Hospital - The Feingold Center, Waltham, MA 02115, USA
| | - Hilde Peeters
- Center for Human Genetics, University Hospital Leuven, KU Leuven, Leuven 3000, Belgium
| | - Florence Petit
- Service de Génétique Clinique, Hôpital Jeanne de Flandre, CHU Lille, Lille 59000, France
| | - Daniela T Pilz
- West of Scotland Genetics Service, Queen Elizabeth University Hospital, Glasgow G51 4TF, UK
| | - Jacques Puechberty
- INSERM U1183, Département de Génétique Médicale, Maladies Rares et Médecine Personnalisée, Génétique clinique, CHU Montpellier, Université Montpellier, Centre de référence anomalies du développement SORO, Montpellier 34295, France
| | - Eyal Reinstein
- Medical Genetics Institute, Meir Medical Center, Kfar-Saba 4428164, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Jean-Baptiste Rivière
- UMR1231, Génétique des Anomalies du Développement, Université de Bourgogne, Dijon 21079, France; Centre de Génétique et Centre de Référence Anomalies du Développement et Syndromes Malformatifs de l'Interrégion Est et FHU TRANSLAD, Centre Hospitalier Universitaire Dijon, Dijon 21079, France; Child Health and Human Development Program, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Avni B Santani
- Division of Genomic Diagnostics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Anouck Schneider
- INSERM U1183, Département de Génétique Médicale, Maladies Rares et Médecine Personnalisée, Génétique clinique, CHU Montpellier, Université Montpellier, Centre de référence anomalies du développement SORO, Montpellier 34295, France
| | - Elliott H Sherr
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94158, USA
| | | | - Ilse Wieland
- Institute of Human Genetics, University Hospital Magdeburg, Otto-von-Guericke University, Magdeburg 39120, Germany
| | - Elaine Zackai
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Xiaonan Zhao
- Division of Genomic Diagnostics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Richard M Gronostajski
- Department of Biochemistry, Program in Genetics, Genomics and Bioinformatics, Center of Excellence in Bioinformatics and Life Sciences, State University of New York at Buffalo, Buffalo, NY 14203, USA
| | - Martin Zenker
- Institute of Human Genetics, University Hospital Magdeburg, Otto-von-Guericke University, Magdeburg 39120, Germany.
| | - Linda J Richards
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia; School of Biomedical Sciences, The Faculty of Medicine Brisbane, The University of Queensland, Brisbane, QLD 4072, Australia
| |
Collapse
|
34
|
Piper M, Gronostajski R, Messina G. Nuclear Factor One X in Development and Disease. Trends Cell Biol 2018; 29:20-30. [PMID: 30287093 DOI: 10.1016/j.tcb.2018.09.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 09/03/2018] [Accepted: 09/06/2018] [Indexed: 02/06/2023]
Abstract
The past decade has seen incredible advances in the field of stem cell biology that have greatly improved our understanding of development and provided important insights into pathological processes. Transcription factors (TFs) play a central role in mediating stem cell proliferation, quiescence, and differentiation. One TF that contributes to these processes is Nuclear Factor One X (NFIX). Recently, NFIX activity has been shown to be essential in multiple organ systems and to have important translational impacts for human health. Here, we describe recent studies showing the contribution of NFIX to muscle development and muscular dystrophies, hematopoiesis, cancer, and neural stem cell biology, highlighting the importance of this knowledge in the development of therapeutic targets.
Collapse
Affiliation(s)
- Michael Piper
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland 4072, Australia.
| | - Richard Gronostajski
- Department of Biochemistry, Genetics, Genomics & Bioinformatics Graduate Program, New York State Center of Excellence in Bioinformatics and Life Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Graziella Messina
- Department of Biosciences, University of Milan, via Celoria 26, 20133, Milan, Italy.
| |
Collapse
|
35
|
Two patients with 19p13.2 deletion (Malan syndrome) involving NFIX and CACNA1A with overgrowth, developmental delay, and epilepsy. Clin Dysmorphol 2018; 26:224-227. [PMID: 28557812 DOI: 10.1097/mcd.0000000000000185] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
36
|
Priolo M, Schanze D, Tatton-Brown K, Mulder PA, Tenorio J, Kooblall K, Acero IH, Alkuraya FS, Arias P, Bernardini L, Bijlsma EK, Cole T, Coubes C, Dapia I, Davies S, Di Donato N, Elcioglu NH, Fahrner JA, Foster A, González NG, Huber I, Iascone M, Kaiser AS, Kamath A, Liebelt J, Lynch SA, Maas SM, Mammì C, Mathijssen IB, McKee S, Menke LA, Mirzaa GM, Montgomery T, Neubauer D, Neumann TE, Pintomalli L, Pisanti MA, Plomp AS, Price S, Salter C, Santos-Simarro F, Sarda P, Segovia M, Shaw-Smith C, Smithson S, Suri M, Valdez RM, Van Haeringen A, Van Hagen JM, Zollino M, Lapunzina P, Thakker RV, Zenker M, Hennekam RC. Further delineation of Malan syndrome. Hum Mutat 2018; 39:1226-1237. [PMID: 29897170 PMCID: PMC6175110 DOI: 10.1002/humu.23563] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 06/05/2018] [Accepted: 06/07/2018] [Indexed: 01/02/2023]
Abstract
Malan syndrome is an overgrowth disorder described in a limited number of individuals. We aim to delineate the entity by studying a large group of affected individuals. We gathered data on 45 affected individuals with a molecularly confirmed diagnosis through an international collaboration and compared data to the 35 previously reported individuals. Results indicate that height is > 2 SDS in infancy and childhood but in only half of affected adults. Cardinal facial characteristics include long, triangular face, macrocephaly, prominent forehead, everted lower lip, and prominent chin. Intellectual disability is universally present, behaviorally anxiety is characteristic. Malan syndrome is caused by deletions or point mutations of NFIX clustered mostly in exon 2. There is no genotype‐phenotype correlation except for an increased risk for epilepsy with 19p13.2 microdeletions. Variants arose de novo, except in one family in which mother was mosaic. Variants causing Malan and Marshall‐Smith syndrome can be discerned by differences in the site of stop codon formation. We conclude that Malan syndrome has a well recognizable phenotype that usually can be discerned easily from Marshall–Smith syndrome but rarely there is some overlap. Differentiation from Sotos and Weaver syndrome can be made by clinical evaluation only.
Collapse
Affiliation(s)
- Manuela Priolo
- Unità Operativa di Genetica Medica, Grande Ospedale Metropolitano Bianchi-Melacrino-Morelli, Reggio Calabria, Italy
| | - Denny Schanze
- Institute of Human Genetics, University Hospital Magdeburg, Magdeburg, Germany
| | - Katrin Tatton-Brown
- Division of Genetics and Epidemiology, Institute of Cancer Research, London and South West Thames Regional Genetics Service, St. George's University Hospitals NHS Foundation Trust, London, UK
| | - Paul A Mulder
- Autism Team Northern-Netherlands, Jonx Department of Youth Mental Health, Lentis Psychiatric Institute, Groningen, The Netherlands
| | - Jair Tenorio
- Institute of Medical and Molecular Genetics (INGEMM), Hospital Universitario La Paz, IdiPAZ, Universidad Autónoma de Madrid, and CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain
| | - Kreepa Kooblall
- Academic Endocrine Unit, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | | | - Fowzan S Alkuraya
- Saudi Human Genome Project, King Abdulaziz City for Science and Technology, and Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Pedro Arias
- Institute of Medical and Molecular Genetics (INGEMM), Hospital Universitario La Paz, IdiPAZ, Universidad Autónoma de Madrid, and CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain
| | - Laura Bernardini
- Cytogenetics Unit, Casa Sollievo della Sofferenza Foundation, San Giovanni Rotondo, Italy
| | - Emilia K Bijlsma
- Department of Clinical Genetics, Leiden University Medical Centre, Leiden, The Netherlands
| | - Trevor Cole
- Department of Clinical Genetics, Birmingham Women's and Children's NHS Foundation Trust, Birmingham, UK
| | - Christine Coubes
- Département de Génétique Médicale, Hôpital Arnaud de Villeneuve, CHRU Montpellier, Montpellier, France
| | - Irene Dapia
- Institute of Medical and Molecular Genetics (INGEMM), Hospital Universitario La Paz, IdiPAZ, Universidad Autónoma de Madrid, and CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain
| | - Sally Davies
- Institute of Medical Genetics, University Hospital of Wales, Cardiff, UK
| | | | - Nursel H Elcioglu
- Department of Pediatric Genetics, Marmara University Medical School, Istanbul, and Eastern Mediterranean University, Mersin, Turkey
| | - Jill A Fahrner
- McKusick-Nathans Institute of Genetic Medicine, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Alison Foster
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | | | | | - Maria Iascone
- Laboratorio di Genetica Medica, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Ann-Sophie Kaiser
- Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | - Arveen Kamath
- Institute of Medical Genetics, University Hospital of Wales, Cardiff, UK
| | - Jan Liebelt
- South Australian Clinical Genetics Services, SA Pathology, North Adelaide, Australia
| | - Sally Ann Lynch
- UCD Academic Centre on Rare Diseases, School of Medicine and Medical Sciences, University College Dublin, and Clinical Genetics, Temple Street Children's University Hospital, Dublin, Ireland
| | - Saskia M Maas
- Department of Clinical Genetics, Academic Medical Center, Amsterdam, The Netherlands
| | - Corrado Mammì
- Unità Operativa di Genetica Medica, Grande Ospedale Metropolitano Bianchi-Melacrino-Morelli, Reggio Calabria, Italy
| | - Inge B Mathijssen
- Department of Clinical Genetics, Academic Medical Center, Amsterdam, The Netherlands
| | - Shane McKee
- Belfast HSC Trust, Northern Ireland Regional Genetics Service, Belfast, Northern Ireland
| | - Leonie A Menke
- Department of Pediatrics, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Ghayda M Mirzaa
- Center for Integrative Brain Research, Seattle Children's Research Institute, and Department of Human Genetics, University of Washington, Seattle, Washington
| | - Tara Montgomery
- Newcastle upon Tyne NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Dorothee Neubauer
- Institute of Human Genetics, University Hospital Magdeburg, Magdeburg, Germany
| | | | - Letizia Pintomalli
- Unità Operativa di Genetica Medica, Grande Ospedale Metropolitano Bianchi-Melacrino-Morelli, Reggio Calabria, Italy
| | | | - Astrid S Plomp
- Department of Clinical Genetics, Academic Medical Center, Amsterdam, The Netherlands
| | - Sue Price
- Department of Clinical Genetics, Northampton General Hospital NHS Trust, Northampton, UK
| | - Claire Salter
- Wessex Clinical Genetics Service, Princess Ann Hospital, Southampton, UK
| | - Fernando Santos-Simarro
- Institute of Medical and Molecular Genetics (INGEMM), Hospital Universitario La Paz, IdiPAZ, Universidad Autónoma de Madrid, and CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain
| | - Pierre Sarda
- Département de Génétique Médicale, Hôpital Arnaud de Villeneuve, CHRU Montpellier, Montpellier, France
| | - Mabel Segovia
- CENAGEM, Centro Nacional de Genética, Buenos Aires, Argentina
| | | | | | - Mohnish Suri
- Nottingham Clinical Genetics Service, Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Rita Maria Valdez
- Genetics Unit, Hospital Militar Central "Cirujano Mayor Dr. Cosme Argerich,", Buenos Aires, Argentina
| | - Arie Van Haeringen
- Department of Clinical Genetics, Leiden University Medical Centre, Leiden, The Netherlands
| | - Johanna M Van Hagen
- Department of Clinical Genetics, VU University Medical Centre, Amsterdam, The Netherlands
| | - Marcela Zollino
- Department of Laboratory Medicine, Institute of Medical Genetics, Catholic University, Rome, Italy
| | - Pablo Lapunzina
- Institute of Medical and Molecular Genetics (INGEMM), Hospital Universitario La Paz, IdiPAZ, Universidad Autónoma de Madrid, and CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain
| | - Rajesh V Thakker
- Academic Endocrine Unit, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Martin Zenker
- Institute of Human Genetics, University Hospital Magdeburg, Magdeburg, Germany
| | - Raoul C Hennekam
- Department of Pediatrics, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
37
|
Kamien B, Ronan A, Poke G, Sinnerbrink I, Baynam G, Ward M, Gibson WT, Dudding-Byth T, Scott RJ. A Clinical Review of Generalized Overgrowth Syndromes in the Era of Massively Parallel Sequencing. Mol Syndromol 2018; 9:70-82. [PMID: 29593474 DOI: 10.1159/000484532] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/23/2017] [Indexed: 12/22/2022] Open
Abstract
The overgrowth syndromes are important to diagnose, not just for accurate genetic counseling, but also for knowledge surrounding cancer surveillance and prognosis. There has been a recent expansion in the number of genes associated with a mendelian overgrowth phenotype, so this review updates previous classifications of overgrowth syndromes. We also describe a clinical and molecular approach to the investigation of individuals presenting with overgrowth. This review aims to assist the clinical diagnosis of generalized overgrowth syndromes by outlining the salient features of well-known overgrowth syndromes alongside the many syndromes that have been discovered and classified more recently. We provide key clinical "handles" to aid clinical diagnosis and a list of genes to aid with panel design when using next generation sequencing, which we believe is frequently needed due to the overlapping phenotypic features seen between overgrowth syndromes.
Collapse
Affiliation(s)
- Benjamin Kamien
- Hunter Genetics, Perth, WA, Australia.,School of Medicine and Public Health, The University of Newcastle, Perth, WA, Australia.,School of Biomedical Sciences and Pharmacy, The University of Newcastle, Newcastle, NSW, Australia
| | - Anne Ronan
- Hunter Genetics, Perth, WA, Australia.,School of Medicine and Public Health, The University of Newcastle, Perth, WA, Australia
| | - Gemma Poke
- Department of Clinical Genetics, Capital & Coast District Health Board, Wellington, New Zealand
| | - Ingrid Sinnerbrink
- Department of Clinical Genetics, Nepean Hospital, Perth, WA, Australia.,Nepean Clinical School, University of Sydney, Penrith, NSW, Australia
| | - Gareth Baynam
- Genetic Services of Western Australia, Newcastle, NSW, Australia.,Western Australian Register of Developmental Anomalies, Perth, WA, Australia.,Office of Population Health Genomics, Public Health Division, Department of Health, Government of Western Australia, Perth, WA, Australia.,School of Paediatrics and Child Health, University of Western Australia, Perth, WA, Australia.,Institute for Immunology and Infectious Diseases, Murdoch University, Perth, WA, Australia.,Telethon Kids Institute, University of Western Australia, Perth, WA, Australia.,Spatial Sciences, Department of Science and Engineering, Curtin University, Perth, WA, Australia
| | - Michelle Ward
- Genetic Services of Western Australia, Newcastle, NSW, Australia
| | - William T Gibson
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada.,BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Tracy Dudding-Byth
- Hunter Genetics, Perth, WA, Australia.,GrowUpWell Priority Research Center, Perth, WA, Australia.,School of Medicine and Public Health, The University of Newcastle, Perth, WA, Australia.,Hunter Medical Research Institute, Perth, WA, Australia
| | - Rodney J Scott
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Newcastle, NSW, Australia.,Molecular Pathology, Hunter Area Pathology Service, Perth, WA, Australia
| |
Collapse
|
38
|
19p13 microduplications encompassing NFIX are responsible for intellectual disability, short stature and small head circumference. Eur J Hum Genet 2017; 26:85-93. [PMID: 29184170 DOI: 10.1038/s41431-017-0037-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Revised: 10/11/2017] [Accepted: 10/17/2017] [Indexed: 01/17/2023] Open
Abstract
Syndromes caused by copy number variations are described as reciprocal when they result from deletions or duplications of the same chromosomal region. When comparing the phenotypes of these syndromes, various clinical features could be described as reversed, probably due to the opposite effect of these imbalances on the expression of genes located at this locus. The NFIX gene codes for a transcription factor implicated in neurogenesis and chondrocyte differentiation. Microdeletions and loss of function variants of NFIX are responsible for Sotos syndrome-2 (also described as Malan syndrome), a syndromic form of intellectual disability associated with overgrowth and macrocephaly. Here, we report a cohort of nine patients harboring microduplications encompassing NFIX. These patients exhibit variable intellectual disability, short stature and small head circumference, which can be described as a reversed Sotos syndrome-2 phenotype. Strikingly, such a reversed phenotype has already been described in patients harboring microduplications encompassing NSD1, the gene whose deletions and loss-of-function variants are responsible for classical Sotos syndrome. Even though the type/contre-type concept has been criticized, this model seems to give a plausible explanation for the pathogenicity of 19p13 microduplications, and the common phenotype observed in our cohort.
Collapse
|
39
|
Romanovskaya EV, Vikhnina MV, Grishina TV, Ivanov MP, Leonova LE, Tsvetkova EV. Transcription factors of the NF1 family: Possible mechanisms of inducible gene expression in the evolutionary lineage of multicellular animals. J EVOL BIOCHEM PHYS+ 2017. [DOI: 10.1134/s123456781702001x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
40
|
A novel mutation of NFIX causes Sotos-like syndrome (Malan syndrome) complicated with thoracic aortic aneurysm and dissection. Hum Genome Var 2017; 4:17022. [PMID: 28584646 PMCID: PMC5451486 DOI: 10.1038/hgv.2017.22] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Revised: 03/07/2017] [Accepted: 04/18/2017] [Indexed: 01/03/2023] Open
Abstract
Malan syndrome has recently been characterized to present Sotos-like phenotypes, such as intellectual disability and macrocephaly, with mutations in the NFIX gene. Herein, we report a 38-year-old patient with a novel single adenine insertion mutation in exon 2 of the NFIX gene (c.290_291insA). He developed early-onset thoracic aortic aneurysm and dissection, which was a rare complication but deserves particular attention in relatively long-lived patients with Sotos-like phenotypes.
Collapse
|
41
|
Lu Y, Chong PF, Kira R, Seto T, Ondo Y, Shimojima K, Yamamoto T. Mutations in NSD1 and NFIX in Three Patients with Clinical Features of Sotos Syndrome and Malan Syndrome. J Pediatr Genet 2017; 6:234-237. [PMID: 29142766 DOI: 10.1055/s-0037-1603194] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 04/12/2017] [Indexed: 12/19/2022]
Abstract
Mutations in nuclear receptor SET domain-containing protein 1 gene ( NSD1 ) are related to Sotos syndrome, which is characterized by overgrowth, macrocephaly, distinctive features, and neurodevelopmental disabilities. On the other hand, mutations in the nuclear factor I/X gene ( NFIX ) can lead to Malan syndrome, also known as Sotos-like syndrome, or to the Marshall-Smith syndrome. In this study, using next generation sequencing (NGS), we identified de novo mutations in NSD1 and NFIX in three patients with developmental disabilities associated with overgrowth or macrocephaly. Overall, we confirmed that clinical entities of congenital malformation syndromes can be expanded by molecular diagnoses via NGS.
Collapse
Affiliation(s)
- Yongping Lu
- Institute of Medical Genetics, Tokyo Women's Medical University, Tokyo, Japan
| | - Pin Fee Chong
- Department of Pediatric Neurology, Fukuoka Children's Hospital, Fukuoka, Japan
| | - Ryutaro Kira
- Department of Pediatric Neurology, Fukuoka Children's Hospital, Fukuoka, Japan
| | - Toshiyuki Seto
- Department of Pediatrics, Osaka City University, Osaka, Japan
| | - Yumiko Ondo
- Institute of Medical Genetics, Tokyo Women's Medical University, Tokyo, Japan
| | - Keiko Shimojima
- Institute of Medical Genetics, Tokyo Women's Medical University, Tokyo, Japan
| | - Toshiyuki Yamamoto
- Institute of Medical Genetics, Tokyo Women's Medical University, Tokyo, Japan
| |
Collapse
|
42
|
Marshall-Smith syndrome: Novel pathogenic variant and previously unreported associations with precocious puberty and aortic root dilatation. Eur J Med Genet 2017; 60:391-394. [PMID: 28442439 DOI: 10.1016/j.ejmg.2017.04.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 02/23/2017] [Accepted: 04/12/2017] [Indexed: 12/28/2022]
Abstract
Marshall-Smith Syndrome (MRSHSS) is a very rare genetic disorder characterized by failure to thrive and characteristic dysmorphic features associated with accelerated osseous maturation. We present a nine-year-old girl who was diagnosed with MRSHSS based on characteristic clinical features supported by the identification of a novel de novo pathogenic variant in the NFIX gene. The patient also presented with precocious puberty diagnosed at five years of age and had an abnormal GnRH stimulation test indicative of central precocious puberty. Central precocious puberty has not been described in association with MRSHSS previously in the medical literature and broadens our knowledge of the natural history of MRSHSS. The causes of advanced bone age in this syndrome are also reviewed. Additionally, the patient showed progressive dilatation of the aortic root. Although connective tissue abnormalities have been described in association with MRSHSS, aortic root dilatation has not. Understanding the mechanism of comorbidities such as advanced bone age and aortic root dilatation in MRSHSS patients enables future development of anticipatory guidance, preventative care measures, and treatment guidelines.
Collapse
|
43
|
Son AI, Fu X, Suto F, Liu JS, Hashimoto-Torii K, Torii M. Proteome dynamics during postnatal mouse corpus callosum development. Sci Rep 2017; 7:45359. [PMID: 28349996 PMCID: PMC5368975 DOI: 10.1038/srep45359] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 02/27/2017] [Indexed: 02/08/2023] Open
Abstract
Formation of cortical connections requires the precise coordination of numerous discrete phases. This is particularly significant with regard to the corpus callosum, whose development undergoes several dynamic stages including the crossing of axon projections, elimination of exuberant projections, and myelination of established tracts. To comprehensively characterize the molecular events in this dynamic process, we set to determine the distinct temporal expression of proteins regulating the formation of the corpus callosum and their respective developmental functions. Mass spectrometry-based proteomic profiling was performed on early postnatal mouse corpus callosi, for which limited evidence has been obtained previously, using stable isotope of labeled amino acids in mammals (SILAM). The analyzed corpus callosi had distinct proteomic profiles depending on age, indicating rapid progression of specific molecular events during this period. The proteomic profiles were then segregated into five separate clusters, each with distinct trajectories relevant to their intended developmental functions. Our analysis both confirms many previously-identified proteins in aspects of corpus callosum development, and identifies new candidates in understudied areas of development including callosal axon refinement. We present a valuable resource for identifying new proteins integral to corpus callosum development that will provide new insights into the development and diseases afflicting this structure.
Collapse
Affiliation(s)
- Alexander I Son
- Center for Neuroscience Research, Children's Research Institute, Children's National Medical Center, Washington, DC 20010, USA
| | - Xiaoqin Fu
- Center for Neuroscience Research, Children's Research Institute, Children's National Medical Center, Washington, DC 20010, USA
| | - Fumikazu Suto
- Department of Ultrastructural Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo 187-8502, Japan
| | - Judy S Liu
- Center for Neuroscience Research, Children's Research Institute, Children's National Medical Center, Washington, DC 20010, USA.,Department of Pediatrics, Pharmacology and Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA
| | - Kazue Hashimoto-Torii
- Center for Neuroscience Research, Children's Research Institute, Children's National Medical Center, Washington, DC 20010, USA.,Department of Pediatrics, Pharmacology and Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA.,Department of Neurobiology and Kavli Institute for Neuroscience, School of Medicine, Yale University, New Haven, CT 06510, USA
| | - Masaaki Torii
- Center for Neuroscience Research, Children's Research Institute, Children's National Medical Center, Washington, DC 20010, USA.,Department of Pediatrics, Pharmacology and Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA.,Department of Neurobiology and Kavli Institute for Neuroscience, School of Medicine, Yale University, New Haven, CT 06510, USA
| |
Collapse
|
44
|
Laccetta G, Moscuzza F, Michelucci A, Guzzetta A, Lunardi S, Lorenzoni F, Ghirri P. A Novel Missense Mutation of the NSD1 Gene Associated with Overgrowth in Three Generations of an Italian Family: Case Report, Differential Diagnosis, and Review of Mutations of NSD1 Gene in Familial Sotos Syndrome. Front Pediatr 2017; 5:236. [PMID: 29164086 PMCID: PMC5681921 DOI: 10.3389/fped.2017.00236] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 10/19/2017] [Indexed: 12/31/2022] Open
Abstract
Sotos syndrome (SoS) is characterized by overgrowth of prenatal onset, learning disability, and characteristic facial appearance; it is usually due to haploinsufficiency of NSD1 gene at chromosome 5q35. An Italian child was born at 37 weeks of gestation (weight 2,910 g, 25th-50th centiles; length 50 cm, 75th centile; head circumference 36 cm, 97th centile) showing cryptorchidism on the right side, hypertelorism, dolichocephaly, broad and prominent forehead, and narrow jaw; the pregnancy was worsened by maternal preeclampsia and gestational diabetes, and his mother had a previous history of four early miscarriages. The patient showed neonatal jaundice, hypotonia, feeding difficulties, frequent vomiting, and gastroesophageal reflux. After the age of 6 months, his weight, length, and head circumference were above the 97th centile; psychomotor development was delayed. At the age of 9 years, the patient showed also joint laxity and scoliosis. DNA sequence analysis of NSD1 gene detected a novel heterozygous mutation (c.521T>A, p.Val174Asp) in exon 2. The same mutant allele was also found in the mother and in the maternal grandfather of the proband; both the mother and the maternal grandfather of the proband showed isolated overgrowth with height above the 97th centile in absence of other features of SoS. At present 23 familial cases of SoS have been described (two cases with mutation in exon 2 of NSD1 gene); no familial cases of SoS with mutation of NSD1 gene and isolated overgrowth have been reported. Probably, point mutations of NSD1 gene, and particularly mutations between exon 20 and exon 23, are not likely to affect reproductive fitness. Epigenetic mechanisms and intrauterine environment may influence phenotypes, therefore genetic tests are not useful to predict the phenotype but they are indispensable for the diagnosis of SoS. This is the first Italian familial case of SoS with genetic confirmation and the third report in which a missense mutation of NSD1 gene is found in three generations of the same family.
Collapse
Affiliation(s)
- Gianluigi Laccetta
- Department of Maternal and Child Health, Division of Neonatology and Neonatal Intensive Care Unit, Santa Chiara Hospital, University of Pisa, Pisa, Italy
| | - Francesca Moscuzza
- Department of Maternal and Child Health, Division of Neonatology and Neonatal Intensive Care Unit, Santa Chiara Hospital, University of Pisa, Pisa, Italy
| | - Angela Michelucci
- Division of Cytogenetic and Molecular Biology, Azienda Ospedaliera Universitaria Pisana (AOUP), Pisa, Italy
| | - Andrea Guzzetta
- Department of Developmental Neuroscience, IRCCS Stella Maris, University of Pisa, Pisa, Italy
| | - Sara Lunardi
- Department of Maternal and Child Health, Division of Neonatology and Neonatal Intensive Care Unit, Santa Chiara Hospital, University of Pisa, Pisa, Italy
| | - Francesca Lorenzoni
- Department of Maternal and Child Health, Division of Neonatology and Neonatal Intensive Care Unit, Santa Chiara Hospital, University of Pisa, Pisa, Italy
| | - Paolo Ghirri
- Department of Maternal and Child Health, Division of Neonatology and Neonatal Intensive Care Unit, Santa Chiara Hospital, University of Pisa, Pisa, Italy
| |
Collapse
|
45
|
Abstract
Overgrowth syndromes comprise a group of disorders associated with excessive growth and other features such as facial dysmorphism, developmental delay or intellectual disability, congenital anomalies, neurological problems and an increased risk of neoplasia. Recent advances in understanding the genetic basis of overgrowth syndromes has resulted in a move away from clinical classification to molecular classification of overgrowth syndromes. This review provides a structured clinical approach to patients with this group of disorders and includes most of the currently known overgrowth syndromes.
Collapse
|
46
|
Dong HY, Zeng H, Hu YQ, Xie L, Wang J, Wang XY, Yang YF, Tan ZP. 19p13.2 Microdeletion including NFIX associated with overgrowth and intellectual disability suggestive of Malan syndrome. Mol Cytogenet 2016; 9:71. [PMID: 27688808 PMCID: PMC5034553 DOI: 10.1186/s13039-016-0282-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 09/13/2016] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND Overgrowth syndromes represent clinically and genetically heterogeneous conditions characterized by a wide spectrum of malformations, tall stature, intellectual disability and/or macrocephaly. RESULTS In a cohort of four clinically characterized patients with overgrowth syndrome without known causative gene mutation, we performed an Illumina SNP-array analysis to identify the pathogenic copy number variations. We identified two rare copy number variations harboring overgrowth syndrome related genes. Patient 1 was Malan syndrome with a 1.4 Mb 19p13.2-13.13 microdeletion including NFIX, and Patient 2 was identified as Sotos syndrome with a 1.6 Mb 5q35.2 microdeletion encompassing NSD1. CONCLUSIONS We identified two patients associated with Manlan syndrome and Sotos syndrome respectively. We also discuss the use of the microarrays-based candidate gene strategy in Mendelian disease-gene identification.
Collapse
Affiliation(s)
- Hai-Yun Dong
- Intensive Care Unit, Central South University, Changsha, Hunan Province 410011 China
| | - Hui Zeng
- Clinical Center for Gene Diagnosis and Therapy of State Key Laboratory of Medical Genetics, The Second Xiangya Hospital of Central South University, 139# Renmin Road, Changsha, Hunan 410011 China ; Department of Cardiothoracic Surgery, Central South University, Changsha, Hunan Province 410011 China
| | - Yi-Qiao Hu
- The State Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan Province 410011 China
| | - Li Xie
- Clinical Center for Gene Diagnosis and Therapy of State Key Laboratory of Medical Genetics, The Second Xiangya Hospital of Central South University, 139# Renmin Road, Changsha, Hunan 410011 China ; Department of Cardiothoracic Surgery, Central South University, Changsha, Hunan Province 410011 China
| | - Jian Wang
- Clinical Center for Gene Diagnosis and Therapy of State Key Laboratory of Medical Genetics, The Second Xiangya Hospital of Central South University, 139# Renmin Road, Changsha, Hunan 410011 China ; Department of Cardiothoracic Surgery, Central South University, Changsha, Hunan Province 410011 China
| | - Xiu-Ying Wang
- Department of Pediatrics, the Second Xiangya Hospital, Central South University, Changsha, Hunan Province 410011 China
| | - Yi-Feng Yang
- Clinical Center for Gene Diagnosis and Therapy of State Key Laboratory of Medical Genetics, The Second Xiangya Hospital of Central South University, 139# Renmin Road, Changsha, Hunan 410011 China ; Department of Cardiothoracic Surgery, Central South University, Changsha, Hunan Province 410011 China
| | - Zhi-Ping Tan
- Clinical Center for Gene Diagnosis and Therapy of State Key Laboratory of Medical Genetics, The Second Xiangya Hospital of Central South University, 139# Renmin Road, Changsha, Hunan 410011 China ; Department of Cardiothoracic Surgery, Central South University, Changsha, Hunan Province 410011 China
| |
Collapse
|
47
|
Jezela-Stanek A, Kucharczyk M, Falana K, Jurkiewicz D, Mlynek M, Wicher D, Rydzanicz M, Kugaudo M, Cieslikowska A, Ciara E, Ploski R, Krajewska-Walasek M. Malan syndrome (Sotos syndrome 2) in two patients with 19p13.2 deletion encompassing NFIX gene and novel NFIX sequence variant. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2016; 160:161-7. [DOI: 10.5507/bp.2016.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 02/03/2016] [Indexed: 01/14/2023] Open
|
48
|
Begum F, Ruczinski I, Li S, Silverman EK, Cho MH, Lynch DA, Curran-Everett D, Crapo J, Scharpf RB, Parker MM, Hetmanski JB, Beaty TH. Identifying a Deletion Affecting Total Lung Capacity Among Subjects in the COPDGene Study Cohort. Genet Epidemiol 2015; 40:81-8. [PMID: 26643968 DOI: 10.1002/gepi.21943] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2015] [Revised: 09/21/2015] [Accepted: 10/19/2015] [Indexed: 01/17/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is a progressive disease with both environmental and genetic risk factors. Genome-wide association studies (GWAS) have identified multiple genomic regions influencing risk of COPD. To thoroughly investigate the genetic etiology of COPD, however, it is also important to explore the role of copy number variants (CNVs) because the presence of structural variants can alter gene expression and can be causal for some diseases. Here, we investigated effects of polymorphic CNVs on quantitative measures of pulmonary function and chest computed tomography (CT) phenotypes among subjects enrolled in COPDGene, a multisite study. COPDGene subjects consist of roughly one-third African American (AA) and two-thirds non-Hispanic white adult smokers (with or without COPD). We estimated CNVs using PennCNV on 9,076 COPDGene subjects using Illumina's Omni-Express genome-wide marker array. We tested for association between polymorphic CNV components (defined as disjoint intervals of copy number regions) for several quantitative phenotypes associated with COPD within each racial group. Among the AAs, we identified a polymorphic CNV on chromosome 5q35.2 located between two genes (FAM153B and SIMK1, but also harboring several pseudo-genes) giving genome-wide significance in tests of association with total lung capacity (TLCCT ) as measured by chest CT scans. This is the first study of genome-wide association tests of polymorphic CNVs and TLCCT . Although the ARIC cohort did not have the phenotype of TLCCT , we found similar counts of CNV deletions and amplifications among AA and European subjects in this second cohort.
Collapse
Affiliation(s)
- Ferdouse Begum
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Ingo Ruczinski
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Shengchao Li
- Cancer Genomics Research Laboratory (CGR), Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, Maryland, United States of America
| | - Edwin K Silverman
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Michael H Cho
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - David A Lynch
- Department of Medicine, National Jewish Health, Denver, Colorado, United States of America
| | - Douglas Curran-Everett
- Division of Biostatistics and Bioinformatics, National Jewish Health, Denver, Colorado, United States of America
| | - James Crapo
- Department of Medicine, National Jewish Health, Denver, Colorado, United States of America
| | - Robert B Scharpf
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Margaret M Parker
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Jacqueline B Hetmanski
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Terri H Beaty
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| |
Collapse
|
49
|
Novel mutations of NFIX gene causing Marshall-Smith syndrome or Sotos-like syndrome: one gene, two phenotypes. Pediatr Res 2015. [PMID: 26200704 DOI: 10.1038/pr.2015.135] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND Only 15 point mutations in NFIX gene have been reported so far, nine of them cause the Marshall-Smith syndrome (MSS) and the remaining mutations lead to an overgrowth disorder with a less severe phenotype, defined as Sotos-like. METHODS The clinical findings in three patients with MSS and two patients with a Sotos-like phenotype are presented. Analysis of the NFIX gene was performed both by conventional or next-generation sequencing. RESULTS Five de novo mutations in NFIX gene were identified, four of them not previously reported. Two frameshift mutations and a donor-splice one caused MSS, while two missense mutations in the DNA binding/dimerisation domain entailed an overgrowth syndrome with some clinical features resembling Sotos syndrome, accompanied by a marfanoid habitus, very low BMI, long narrow face, or arachnodactyly. CONCLUSION Marshall-Smith mutations are scattered through exons 6-10 of NFIX gene, while most point mutations causing an overgrowth syndrome are clustered in exon 2. Clinical features of this overgrowth syndrome may well be considered an intermediate phenotype between Sotos and Marfan syndromes.
Collapse
|
50
|
Edmondson AC, Kalish JM. Overgrowth Syndromes. J Pediatr Genet 2015; 4:136-43. [PMID: 27617124 PMCID: PMC4918719 DOI: 10.1055/s-0035-1564440] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 05/20/2015] [Indexed: 01/19/2023]
Abstract
Numerous multiple malformation syndromes associated with pathologic overgrowth have been described and, for many, their molecular bases elucidated. This review describes the characteristic features of these overgrowth syndromes, as well as the current understanding of their molecular bases, intellectual outcomes, and cancer predispositions. We review syndromes such as Sotos, Malan, Marshall-Smith, Weaver, Simpson-Golabi-Behmel, Perlman, Bannayan-Riley-Ruvalcaba, PI3K-related, Proteus, Beckwith-Wiedemann, fibrous dysplasia, Klippel-Trenaunay-Weber, and Maffucci.
Collapse
Affiliation(s)
- Andrew C. Edmondson
- Division of Human Genetics, Children's Hospital of Philadelphia and Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Jennifer M. Kalish
- Division of Human Genetics, Children's Hospital of Philadelphia and Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States
| |
Collapse
|