1
|
Avagliano L, Castiglioni S, Lettieri A, Parodi C, Di Fede E, Taci E, Grazioli P, Colombo EA, Gervasini C, Massa V. Intrauterine growth in chromatinopathies: A long road for better understanding and for improving clinical management. Birth Defects Res 2024; 116:e2383. [PMID: 38984779 DOI: 10.1002/bdr2.2383] [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: 02/23/2024] [Revised: 06/12/2024] [Accepted: 06/25/2024] [Indexed: 07/11/2024]
Abstract
BACKGROUND Chromatinopathies are a heterogeneous group of genetic disorders caused by pathogenic variants in genes coding for chromatin state balance proteins. Remarkably, many of these syndromes present unbalanced postnatal growth, both under- and over-, although little has been described in the literature. Fetal growth measurements are common practice in pregnancy management and values within normal ranges indicate proper intrauterine growth progression; on the contrary, abnormalities in intrauterine fetal growth open the discussion of possible pathogenesis affecting growth even in the postnatal period. METHODS Among the numerous chromatinopathies, we have selected six of the most documented in the literature offering evidence about two fetal overgrowth (Sotos and Weaver syndrome) and four fetal undergrowth syndromes (Bohring Opitz, Cornelia de Lange, Floating-Harbor, and Meier Gorlin syndrome), describing their molecular characteristics, maternal biochemical results and early pregnancy findings, prenatal ultrasound findings, and postnatal characteristics. RESULTS/CONCLUSION To date, the scarce data in the literature on prenatal findings are few and inconclusive, even though these parameters may contribute to a more rapid and accurate diagnosis, calling for a better and more detailed description of pregnancy findings.
Collapse
Affiliation(s)
| | - Silvia Castiglioni
- Department of Health Sciences, Università Degli Studi di Milano, Milan, Italy
| | - Antonella Lettieri
- Department of Health Sciences, Università Degli Studi di Milano, Milan, Italy
| | - Chiara Parodi
- Department of Health Sciences, Università Degli Studi di Milano, Milan, Italy
| | - Elisabetta Di Fede
- Department of Health Sciences, Università Degli Studi di Milano, Milan, Italy
- Aldo Ravelli Center for Neurotechnology and Experimental Brain Therapeutics, Università Degli Studi di Milano, Milan, Italy
| | - Esi Taci
- Department of Health Sciences, Università Degli Studi di Milano, Milan, Italy
- Aldo Ravelli Center for Neurotechnology and Experimental Brain Therapeutics, Università Degli Studi di Milano, Milan, Italy
| | - Paolo Grazioli
- Department of Health Sciences, Università Degli Studi di Milano, Milan, Italy
| | - Elisa Adele Colombo
- Department of Health Sciences, Università Degli Studi di Milano, Milan, Italy
| | - Cristina Gervasini
- Department of Health Sciences, Università Degli Studi di Milano, Milan, Italy
- Aldo Ravelli Center for Neurotechnology and Experimental Brain Therapeutics, Università Degli Studi di Milano, Milan, Italy
| | - Valentina Massa
- Department of Health Sciences, Università Degli Studi di Milano, Milan, Italy
- Aldo Ravelli Center for Neurotechnology and Experimental Brain Therapeutics, Università Degli Studi di Milano, Milan, Italy
| |
Collapse
|
2
|
Dobrzynski W, Stawinska-Dudek J, Moryto N, Lipka D, Mikulewicz M. Floating-Harbor Syndrome: A Systematic Literature Review and Case Report. J Clin Med 2024; 13:3435. [PMID: 38929963 PMCID: PMC11204875 DOI: 10.3390/jcm13123435] [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: 04/25/2024] [Revised: 06/08/2024] [Accepted: 06/09/2024] [Indexed: 06/28/2024] Open
Abstract
Floating-Harbor syndrome (FHS) is an extremely rare genetic disorder connected with a distinctive facial appearance, various skeletal malformations, delayed bone age, and expressive language delays. It is caused by heterozygous mutations in the Snf2-related CREBBP activator protein (SRCAP) gene. The aim of this paper is to describe the case of a 14-year-old male with FHS, referring to a review of the literature, and to collect all reported symptoms. In addition, the orthodontic treatment of the patient is described. For this, the electronic databases PubMed and Scopus were searched using the keyword "Floating-Harbor syndrome". Similar to previous cases in the literature, the patient presented with short stature; a triangular face with a large bulbous nose; deep-set eyes and narrow eyelid gaps; a wide mouth with a thin vermilion border of the upper lip; and dorsally rotated, small ears. They also presented some less-described symptoms, such as macrodontia and micrognathia. Moreover, mild mental retardation, microcephaly, and delayed psychomotor development were found. On the basis of an extraoral, intraoral examination, X-rays, and CBCT, he was diagnosed with overbite, canine class I and angle class III, on both sides. To the best of our knowledge, orthodontic treatment of this disease has not been assessed in detail so far, so this is the first case.
Collapse
Affiliation(s)
- Wojciech Dobrzynski
- Department of Dentofacial Orthopaedics and Orthodontics, Division of Facial Abnormalities, Medical University of Wroclaw, 50-425 Wroclaw, Poland;
| | - Julia Stawinska-Dudek
- Student Scientific Group, Department of Dentofacial Orthopaedics and Orthodontics, Division of Facial Abnormalities, Medical University of Wroclaw, 50-425 Wroclaw, Poland; (J.S.-D.); (N.M.); (D.L.)
| | - Natalia Moryto
- Student Scientific Group, Department of Dentofacial Orthopaedics and Orthodontics, Division of Facial Abnormalities, Medical University of Wroclaw, 50-425 Wroclaw, Poland; (J.S.-D.); (N.M.); (D.L.)
| | - Dominika Lipka
- Student Scientific Group, Department of Dentofacial Orthopaedics and Orthodontics, Division of Facial Abnormalities, Medical University of Wroclaw, 50-425 Wroclaw, Poland; (J.S.-D.); (N.M.); (D.L.)
| | - Marcin Mikulewicz
- Department of Dentofacial Orthopaedics and Orthodontics, Division of Facial Abnormalities, Medical University of Wroclaw, 50-425 Wroclaw, Poland;
| |
Collapse
|
3
|
Morlino S, Vaccaro L, Leone MP, Nardella G, Bisceglia L, Ortore RP, Verzicco G, Cassano L, Castori M, Cacchiarelli D, Micale L. Combined exome and whole transcriptome sequencing identifies a de novo intronic SRCAP variant causing DEHMBA syndrome with severe sleep disorder. J Hum Genet 2024; 69:287-290. [PMID: 38448605 DOI: 10.1038/s10038-024-01240-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 02/09/2024] [Accepted: 02/27/2024] [Indexed: 03/08/2024]
Abstract
Rare heterozygous variants in exons 33-34 of the SRCAP gene are associated with Floating-Harbor syndrome and have a dominant-negative mechanism of action. At variance, heterozygous null alleles falling in other parts of the same gene cause developmental delay, hypotonia, musculoskeletal defects, and behavioral abnormalities (DEHMBA) syndrome. We report an 18-year-old man with DEHMBA syndrome and obstructive sleep apnea, who underwent exome sequencing (ES) and whole transcriptome sequencing (WTS) on peripheral blood. Trio analysis prioritized the de novo heterozygous c.5658+5 G > A variant. WTS promptly demostrated four different abnormal transcripts affecting >40% of the reads, three of which leading to a frameshift. This study demonstrated the efficacy of a combined ES-WTS approach in solving undiagnosed cases. We also speculated that sleep respiratory disorder may be an underdiagnosed complication of DEHMBA syndrome.
Collapse
Affiliation(s)
- Silvia Morlino
- Division of Medical Genetics, Fondazione IRCCS-Casa Sollievo della Sofferenza, Viale Cappuccini snc, 71013, San Giovanni Rotondo, Foggia, Italy
| | - Lorenzo Vaccaro
- Armenise/Harvard Laboratory of Integrative Genomics, Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
- Department of Translational Medicine, University of Naples "Federico II", Naples, Italy
| | - Maria Pia Leone
- Division of Medical Genetics, Fondazione IRCCS-Casa Sollievo della Sofferenza, Viale Cappuccini snc, 71013, San Giovanni Rotondo, Foggia, Italy
| | - Grazia Nardella
- Division of Medical Genetics, Fondazione IRCCS-Casa Sollievo della Sofferenza, Viale Cappuccini snc, 71013, San Giovanni Rotondo, Foggia, Italy
| | - Luigi Bisceglia
- Division of Medical Genetics, Fondazione IRCCS-Casa Sollievo della Sofferenza, Viale Cappuccini snc, 71013, San Giovanni Rotondo, Foggia, Italy
| | - Rocco Pio Ortore
- Division of Maxillofacial Surgery and Otolaryngology, Fondazione IRCCS-Casa Sollievo della Sofferenza, San Giovanni Rotondo, 71013, Foggia, Italy
| | - Giannandrea Verzicco
- Division of Maxillofacial Surgery and Otolaryngology, Fondazione IRCCS-Casa Sollievo della Sofferenza, San Giovanni Rotondo, 71013, Foggia, Italy
| | - Lazzaro Cassano
- Division of Maxillofacial Surgery and Otolaryngology, Fondazione IRCCS-Casa Sollievo della Sofferenza, San Giovanni Rotondo, 71013, Foggia, Italy
| | - Marco Castori
- Division of Medical Genetics, Fondazione IRCCS-Casa Sollievo della Sofferenza, Viale Cappuccini snc, 71013, San Giovanni Rotondo, Foggia, Italy.
| | - Davide Cacchiarelli
- Armenise/Harvard Laboratory of Integrative Genomics, Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
- Department of Translational Medicine, University of Naples "Federico II", Naples, Italy
- Genomics and Experimental Medicine Program, Scuola Superiore Meridionale, Naples, Italy
| | - Lucia Micale
- Division of Medical Genetics, Fondazione IRCCS-Casa Sollievo della Sofferenza, Viale Cappuccini snc, 71013, San Giovanni Rotondo, Foggia, Italy
| |
Collapse
|
4
|
White-Brown A, Choufani S, Weksberg R, Dyment D. Missense variant in SRCAP with distinct DNA methylation signature associated with non-FLHS SRCAP-related neurodevelopmental disorder. Am J Med Genet A 2023; 191:2640-2646. [PMID: 37340855 DOI: 10.1002/ajmg.a.63329] [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: 02/17/2023] [Revised: 05/18/2023] [Accepted: 05/26/2023] [Indexed: 06/22/2023]
Abstract
Floating-Harbor syndrome (FLHS) is a neurodevelopmental disorder (NDD) caused by truncating variants in exons 33 and 34 of the SNF2-related CREBBP activator protein gene (SRCAP). Truncating variants proximal to this location in SRCAP result in a non-FLHS SRCAP-associated NDD; an overlapping but distinct NDD characterized by developmental delay with or without intellectual disability (ID), hypotonia, normal stature, and behavioral and psychiatric issues. Here, we report a young woman who initially presented in childhood with significant delays in speech and mild ID. In young adulthood, she developed schizophrenia. On physical examination, she had facial features suggestive of 22q11 deletion syndrome. After non-diagnostic chromosomal microarray and trio exome sequencing (ES), a re-analysis of trio ES data identified a de novo missense variant in SRCAP that was proximal to the FLHS critical region. Subsequent DNA methylation studies showed the unique methylation signature associated with pathogenic sequence variants in non-FLHS SRCAP-related NDD. This clinical report describes an individual with non-FLHS SRCAP-related NDD caused by an SRCAP missense variant, and it also demonstrates the clinical utility of ES re-analysis and DNA methylation analysis for undiagnosed patients, in particular, those with variants of uncertain significance.
Collapse
Affiliation(s)
- Alexandre White-Brown
- Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Sanaa Choufani
- Genetics and Genome Biology Program, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Rosanna Weksberg
- Genetics and Genome Biology Program, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
- Division of Clinical and Metabolic Genetics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - David Dyment
- Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, Ontario, Canada
- Department of Genetics, Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada
| |
Collapse
|
5
|
St John M, Tripathi T, Morgan AT, Amor DJ. To speak may draw on epigenetic writing and reading: Unravelling the complexity of speech and language outcomes across chromatin-related neurodevelopmental disorders. Neurosci Biobehav Rev 2023; 152:105293. [PMID: 37353048 DOI: 10.1016/j.neubiorev.2023.105293] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/11/2023] [Accepted: 06/20/2023] [Indexed: 06/25/2023]
Abstract
Speech and language development are complex neurodevelopmental processes that are incompletely understood, yet current evidence suggests that speech and language disorders are prominent in those with disorders of chromatin regulation. This review aimed to unravel what is known about speech and language outcomes for individuals with chromatin-related neurodevelopmental disorders. A systematic literature search following PRISMA guidelines was conducted on 70 chromatin genes, to identify reports of speech/language outcomes across studies, including clinical reports, formal subjective measures, and standardised/objective measures. 3932 studies were identified and screened and 112 were systematically reviewed. Communication impairment was core across chromatin disorders, and specifically, chromatin writers and readers appear to play an important role in motor speech development. Identification of these relationships is important because chromatin disorders show promise as therapeutic targets due to the capacity for epigenetic modification. Further research is required using standardised and formal assessments to understand the nuanced speech/language profiles associated with variants in each gene, and the influence of chromatin dysregulation on the neurobiology of speech and language development.
Collapse
Affiliation(s)
- Miya St John
- Speech and Language, Murdoch Children's Research Institute, Parkville, VIC, Australia; Department of Audiology and Speech Pathology, University of Melbourne, VIC, Australia.
| | - Tanya Tripathi
- Neurodisability and Rehabilitation, Murdoch Children's Research Institute, Parkville, VIC, Australia.
| | - Angela T Morgan
- Speech and Language, Murdoch Children's Research Institute, Parkville, VIC, Australia; Department of Audiology and Speech Pathology, University of Melbourne, VIC, Australia; Speech Genomics Clinic, Royal Children's Hospital, Parkville, VIC, Australia.
| | - David J Amor
- Neurodisability and Rehabilitation, Murdoch Children's Research Institute, Parkville, VIC, Australia; Speech Genomics Clinic, Royal Children's Hospital, Parkville, VIC, Australia; Department of Paediatrics, University of Melbourne, VIC, Australia.
| |
Collapse
|
6
|
Toni L, Plachy L, Dusatkova P, Amaratunga SA, Elblova L, Sumnik Z, Kolouskova S, Snajderova M, Obermannova B, Pruhova S, Lebl J. The Genetic Landscape of Children Born Small for Gestational Age with Persistent Short Stature. Horm Res Paediatr 2023; 97:40-52. [PMID: 37019085 DOI: 10.1159/000530521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 03/24/2023] [Indexed: 04/07/2023] Open
Abstract
INTRODUCTION Among children born small for gestational age, 10-15% fail to catch up and remain short (SGA-SS). The underlying mechanisms are mostly unknown. We aimed to decipher genetic aetiologies of SGA-SS within a large single-centre cohort. METHODS Out of 820 patients treated with growth hormone (GH), 256 were classified as SGA-SS (birth length and/or birth weight <-2 SD for gestational age and life-minimum height <-2.5 SD). Those with the DNA triplet available (child and both parents) were included in the study (176/256). Targeted testing (karyotype/FISH/MLPA/specific Sanger sequencing) was performed if a specific genetic disorder was clinically suggestive. All remaining patients underwent MS-MLPA to identify Silver-Russell syndrome, and those with unknown genetic aetiology were subsequently examined using whole-exome sequencing or targeted panel of 398 growth-related genes. Genetic variants were classified using ACMG guidelines. RESULTS The genetic aetiology was elucidated in 74/176 (42%) children. Of these, 12/74 (16%) had pathogenic or likely pathogenic (P/LP) gene variants affecting pituitary development (LHX4, OTX2, PROKR2, PTCH1, POU1F1), the GH-IGF-1 or IGF-2 axis (GHSR, IGFALS, IGF1R, STAT3, HMGA2), 2/74 (3%) the thyroid axis (TRHR, THRA), 17/74 (23%) the cartilaginous matrix (ACAN, various collagens, FLNB, MATN3), and 7/74 (9%) the paracrine chondrocyte regulation (FGFR3, FGFR2, NPR2). In 12/74 (16%), we revealed P/LP affecting fundamental intracellular/intranuclear processes (CDC42, KMT2D, LMNA, NSD1, PTPN11, SRCAP, SON, SOS1, SOX9, TLK2). SHOX deficiency was found in 7/74 (9%), Silver-Russell syndrome in 12/74 (16%) (11p15, UPD7), and miscellaneous chromosomal aberrations in 5/74 (7%) children. CONCLUSIONS The high diagnostic yield sheds a new light on the genetic landscape of SGA-SS, with a central role for the growth plate with substantial contributions from the GH-IGF-1 and thyroid axes and intracellular regulation and signalling.
Collapse
Affiliation(s)
- Ledjona Toni
- Department of Paediatrics, 2nd Faculty of Medicine, Charles University in Prague and Motol University Hospital, Prague, Czechia
| | - Lukas Plachy
- Department of Paediatrics, 2nd Faculty of Medicine, Charles University in Prague and Motol University Hospital, Prague, Czechia
| | - Petra Dusatkova
- Department of Paediatrics, 2nd Faculty of Medicine, Charles University in Prague and Motol University Hospital, Prague, Czechia
| | - Shenali Anne Amaratunga
- Department of Paediatrics, 2nd Faculty of Medicine, Charles University in Prague and Motol University Hospital, Prague, Czechia
| | - Lenka Elblova
- Department of Paediatrics, 2nd Faculty of Medicine, Charles University in Prague and Motol University Hospital, Prague, Czechia
| | - Zdenek Sumnik
- Department of Paediatrics, 2nd Faculty of Medicine, Charles University in Prague and Motol University Hospital, Prague, Czechia
| | - Stanislava Kolouskova
- Department of Paediatrics, 2nd Faculty of Medicine, Charles University in Prague and Motol University Hospital, Prague, Czechia
| | - Marta Snajderova
- Department of Paediatrics, 2nd Faculty of Medicine, Charles University in Prague and Motol University Hospital, Prague, Czechia
| | - Barbora Obermannova
- Department of Paediatrics, 2nd Faculty of Medicine, Charles University in Prague and Motol University Hospital, Prague, Czechia
| | - Stepanka Pruhova
- Department of Paediatrics, 2nd Faculty of Medicine, Charles University in Prague and Motol University Hospital, Prague, Czechia
| | - Jan Lebl
- Department of Paediatrics, 2nd Faculty of Medicine, Charles University in Prague and Motol University Hospital, Prague, Czechia
| |
Collapse
|
7
|
Chohra I, Chung K, Giri S, Malgrange B. ATP-Dependent Chromatin Remodellers in Inner Ear Development. Cells 2023; 12:cells12040532. [PMID: 36831199 PMCID: PMC9954591 DOI: 10.3390/cells12040532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 01/30/2023] [Accepted: 02/01/2023] [Indexed: 02/11/2023] Open
Abstract
During transcription, DNA replication and repair, chromatin structure is constantly modified to reveal specific genetic regions and allow access to DNA-interacting enzymes. ATP-dependent chromatin remodelling complexes use the energy of ATP hydrolysis to modify chromatin architecture by repositioning and rearranging nucleosomes. These complexes are defined by a conserved SNF2-like, catalytic ATPase subunit and are divided into four families: CHD, SWI/SNF, ISWI and INO80. ATP-dependent chromatin remodellers are crucial in regulating development and stem cell biology in numerous organs, including the inner ear. In addition, mutations in genes coding for proteins that are part of chromatin remodellers have been implicated in numerous cases of neurosensory deafness. In this review, we describe the composition, structure and functional activity of these complexes and discuss how they contribute to hearing and neurosensory deafness.
Collapse
|
8
|
Fu MP, Merrill SM, Sharma M, Gibson WT, Turvey SE, Kobor MS. Rare diseases of epigenetic origin: Challenges and opportunities. Front Genet 2023; 14:1113086. [PMID: 36814905 PMCID: PMC9939656 DOI: 10.3389/fgene.2023.1113086] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 01/24/2023] [Indexed: 02/09/2023] Open
Abstract
Rare diseases (RDs), more than 80% of which have a genetic origin, collectively affect approximately 350 million people worldwide. Progress in next-generation sequencing technology has both greatly accelerated the pace of discovery of novel RDs and provided more accurate means for their diagnosis. RDs that are driven by altered epigenetic regulation with an underlying genetic basis are referred to as rare diseases of epigenetic origin (RDEOs). These diseases pose unique challenges in research, as they often show complex genetic and clinical heterogeneity arising from unknown gene-disease mechanisms. Furthermore, multiple other factors, including cell type and developmental time point, can confound attempts to deconvolute the pathophysiology of these disorders. These challenges are further exacerbated by factors that contribute to epigenetic variability and the difficulty of collecting sufficient participant numbers in human studies. However, new molecular and bioinformatics techniques will provide insight into how these disorders manifest over time. This review highlights recent studies addressing these challenges with innovative solutions. Further research will elucidate the mechanisms of action underlying unique RDEOs and facilitate the discovery of treatments and diagnostic biomarkers for screening, thereby improving health trajectories and clinical outcomes of affected patients.
Collapse
Affiliation(s)
- Maggie P. Fu
- Department of Medical Genetics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada,Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, BC, Canada,BC Children’s Hospital Research Institute, Vancouver, BC, Canada
| | - Sarah M. Merrill
- Department of Medical Genetics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada,Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, BC, Canada,BC Children’s Hospital Research Institute, Vancouver, BC, Canada
| | - Mehul Sharma
- BC Children’s Hospital Research Institute, Vancouver, BC, Canada,Department of Pediatrics, Faculty of Medicine, BC Children’s Hospital, University of British Columbia, Vancouver, BC, Canada
| | - William T. Gibson
- Department of Medical Genetics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada,BC Children’s Hospital Research Institute, Vancouver, BC, Canada
| | - Stuart E. Turvey
- BC Children’s Hospital Research Institute, Vancouver, BC, Canada,Department of Pediatrics, Faculty of Medicine, BC Children’s Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Michael S. Kobor
- Department of Medical Genetics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada,Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, BC, Canada,BC Children’s Hospital Research Institute, Vancouver, BC, Canada,*Correspondence: Michael S. Kobor,
| |
Collapse
|
9
|
Hokken-Koelega ACS, van der Steen M, Boguszewski MCS, Cianfarani S, Dahlgren J, Horikawa R, Mericq V, Rapaport R, Alherbish A, Braslavsky D, Charmandari E, Chernausek SD, Cutfield WS, Dauber A, Deeb A, Goedegebuure WJ, Hofman PL, Isganatis E, Jorge AA, Kanaka-Gantenbein C, Kashimada K, Khadilkar V, Luo XP, Mathai S, Nakano Y, Yau M. International Consensus Guideline on Small for Gestational Age (SGA): Etiology and Management from Infancy to Early Adulthood. Endocr Rev 2023; 44:539-565. [PMID: 36635911 PMCID: PMC10166266 DOI: 10.1210/endrev/bnad002] [Citation(s) in RCA: 41] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 10/31/2022] [Accepted: 01/10/2023] [Indexed: 01/14/2023]
Abstract
This International Consensus Guideline was developed by experts in the field of SGA of 10 pediatric endocrine societies worldwide. A consensus meeting was held and 1300 articles formed the basis for discussions. All experts voted about the strengths of the recommendations. The guideline gives new and clinically relevant insights into the etiology of short stature after SGA birth, including novel knowledge about (epi)genetic causes. Besides, it presents long-term consequences of SGA birth and new treatment options, including treatment with gonadotropin-releasing hormone agonist (GnRHa) in addition to growth hormone (GH) treatment, and the metabolic and cardiovascular health of young adults born SGA after cessation of childhood-GH-treatment in comparison with appropriate control groups. To diagnose SGA, accurate anthropometry and use of national growth charts are recommended. Follow-up in early life is warranted and neurodevelopment evaluation in those at risk. Excessive postnatal weight gain should be avoided, as this is associated with an unfavorable cardio-metabolic health profile in adulthood. Children born SGA with persistent short stature < -2.5 SDS at age 2 years or < -2 SDS at age of 3-4 years, should be referred for diagnostic work-up. In case of dysmorphic features, major malformations, microcephaly, developmental delay, intellectual disability and/or signs of skeletal dysplasia, genetic testing should be considered. Treatment with 0.033-0.067 mg GH/kg/day is recommended in case of persistent short stature at age of 3-4 years. Adding GnRHa treatment could be considered when short adult height is expected at pubertal onset. All young adults born SGA require counseling to adopt a healthy lifestyle.
Collapse
Affiliation(s)
- Anita C S Hokken-Koelega
- Department of Pediatrics, subdivision of Endocrinology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Manouk van der Steen
- Department of Pediatrics, subdivision of Endocrinology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | | | - Stefano Cianfarani
- Department of Systems Medicine, University of Rome 'Tor Vergata', Children's Hospital, Rome, Italy.,Diabetology and Growth Disorders Unit, IRCCS "Bambino Gesù" Children's Hospital, Rome, Italy.,Department of Women's and Children's Health, Karolinska Institute, Stockholm, Sweden
| | - Jovanna Dahlgren
- Department of Pediatrics, the Sahlgrenska Academy, the University of Gothenburg and Queen Silvia Children's Hospital, Gothenburg, Sweden
| | - Reiko Horikawa
- Division of Endocrinology and Metabolism, National Center for Child Health and Development, Tokyo, Japan
| | - Veronica Mericq
- Institute of Maternal and Child Research, faculty of Medicine, University of Chile
| | - Robert Rapaport
- Icahn School of Medicine, Division of Pediatric Endocrinology, Mount Sinai Kravis Children's Hospital, New York, NY, USA
| | | | - Debora Braslavsky
- Centro de Investigaciones Endocrinológicas "Dr. Cesar Bergadá" (CEDIE), División de Endocrinología, Hospital de Niños Dr. Ricardo Gutiérrez, Buenos Aires, Argentina
| | - Evangelia Charmandari
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, 'Aghia Sophia' Children's Hospital, 11527, Athens, Greece.,Division of Endocrinology and Metabolism, Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
| | - Steven D Chernausek
- Department of Pediatrics, Section of Diabetes and Endocrinology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Wayne S Cutfield
- Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Andrew Dauber
- Division of Endocrinology, Children's National Hospital, Washington, DC 20012, USA
| | - Asma Deeb
- Paediatric Endocrine Division, Sheikh Shakhbout Medical City and College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Wesley J Goedegebuure
- Department of Pediatrics, subdivision of Endocrinology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Paul L Hofman
- Liggins Institute, University of Auckland, Auckland, New Zealand
| | | | - Alexander A Jorge
- Unidade de Endocrinologia Genética (LIM25) do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, Brazil
| | - Christina Kanaka-Gantenbein
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, 'Aghia Sophia' Children's Hospital, 11527, Athens, Greece
| | - Kenichi Kashimada
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | | | - Xiao-Ping Luo
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sarah Mathai
- Department of Pediatrics, Christian Medical College, Vellore, India
| | - Yuya Nakano
- Department of Pediatrics, Showa University School of Medicine, Tokyo, Japan
| | - Mabel Yau
- Icahn School of Medicine, Division of Pediatric Endocrinology, Mount Sinai Kravis Children's Hospital, New York, NY, USA
| |
Collapse
|
10
|
Boycott KM, Hartley T, Kernohan KD, Dyment DA, Howley H, Innes AM, Bernier FP, Brudno M. Care4Rare Canada: Outcomes from a decade of network science for rare disease gene discovery. Am J Hum Genet 2022; 109:1947-1959. [PMID: 36332610 PMCID: PMC9674964 DOI: 10.1016/j.ajhg.2022.10.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 10/04/2022] [Indexed: 11/06/2022] Open
Abstract
The past decade has witnessed a rapid evolution in rare disease (RD) research, fueled by the availability of genome-wide (exome and genome) sequencing. In 2011, as this transformative technology was introduced to the research community, the Care4Rare Canada Consortium was launched: initially as FORGE, followed by Care4Rare, and Care4Rare SOLVE. Over what amounted to three eras of diagnosis and discovery, the Care4Rare Consortium used exome sequencing and, more recently, genome and other 'omic technologies to identify the molecular cause of unsolved RDs. We achieved a diagnostic yield of 34% (623/1,806 of participating families), including the discovery of deleterious variants in 121 genes not previously associated with disease, and we continue to study candidate variants in novel genes for 145 families. The Consortium has made significant contributions to RD research, including development of platforms for data collection and sharing and instigating a Canadian network to catalyze functional characterization research of novel genes. The Consortium was instrumental to implementing genome-wide sequencing as a publicly funded test for RD diagnosis in Canada. Despite the successes of the past decade, the challenge of solving all RDs remains enormous, and the work is far from over. We must leverage clinical and 'omic data for secondary use, develop tools and policies to support safe data sharing, continue to explore the utility of new and emerging technologies, and optimize research protocols to delineate complex disease mechanisms. Successful approaches in each of these realms is required to offer diagnostic clarity to all families with RDs.
Collapse
Affiliation(s)
- Kym M. Boycott
- Children’s Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, ON K1H 8L1, Canada,Corresponding author
| | - Taila Hartley
- Children’s Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, ON K1H 8L1, Canada
| | - Kristin D. Kernohan
- Children’s Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, ON K1H 8L1, Canada
| | - David A. Dyment
- Children’s Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, ON K1H 8L1, Canada
| | - Heather Howley
- Children’s Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, ON K1H 8L1, Canada
| | - A. Micheil Innes
- Department of Medical Genetics and Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Francois P. Bernier
- Department of Medical Genetics and Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Michael Brudno
- Department of Computer Science, University of Toronto, Toronto, ON M5S 2E4, Canada
| | | |
Collapse
|
11
|
Zhao B, Madden JA, Lin J, Berry GT, Wojcik MH, Zhao X, Brand H, Talkowski M, Lee EA, Agrawal PB. A neurodevelopmental disorder caused by a novel de novo SVA insertion in exon 13 of the SRCAP gene. Eur J Hum Genet 2022; 30:1083-1087. [PMID: 35768521 PMCID: PMC9437004 DOI: 10.1038/s41431-022-01137-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 04/24/2022] [Accepted: 06/14/2022] [Indexed: 12/24/2022] Open
Abstract
Pathogenic variants in the SRCAP (SNF2-related CREBBP activator protein) gene, which encodes a chromatin-remodeling ATPase, cause neurodevelopmental disorders including Floating Harbor syndrome (FLHS). Here, we report the discovery of a de novo transposon insertion in SRCAP exon 13 from trio genome sequencing in a 28-year-old female with failure to thrive, developmental delay, mood disorder and seizure disorder. The insertion was a full-length (~2.8 kb), antisense-oriented SVA insertion relative to the SRCAP transcript, bearing a 5' transduction and hallmarks of target-primed reverse transcription. The 20-bp 5' transduction allowed us to trace the source SVA element to an intron of a long non-coding RNA on chromosome 12, which is highly expressed in testis. RNA sequencing and qRT-PCR confirmed significant depletion of SRCAP expression and low-level exon skipping in the proband. This case highlights a novel disease-causing structural variant and the importance of transposon analysis in a clinical diagnostic setting.
Collapse
Affiliation(s)
- Boxun Zhao
- Division of Genetics and Genomics, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
- Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, MA, USA
- The Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Jill A Madden
- Division of Genetics and Genomics, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
- Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, MA, USA
| | - Jasmine Lin
- Division of Genetics and Genomics, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
- Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, MA, USA
- Division of Newborn Medicine, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Gerard T Berry
- Division of Genetics and Genomics, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
- Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, MA, USA
| | - Monica H Wojcik
- Division of Genetics and Genomics, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
- Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, MA, USA
- The Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Division of Newborn Medicine, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Xuefang Zhao
- Program in Medical and Population Genetics and Stanley Center for Psychiatric Research, The Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - Harrison Brand
- Program in Medical and Population Genetics and Stanley Center for Psychiatric Research, The Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - Michael Talkowski
- Program in Medical and Population Genetics and Stanley Center for Psychiatric Research, The Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - Eunjung Alice Lee
- Division of Genetics and Genomics, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA.
- Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, MA, USA.
- The Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| | - Pankaj B Agrawal
- Division of Genetics and Genomics, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA.
- Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, MA, USA.
- The Broad Institute of Harvard and MIT, Cambridge, MA, USA.
- Division of Newborn Medicine, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
12
|
Allen DZ, Roy S. Otologic manifestations of Floating Harbor Syndrome (FHS): A case series and a review of the literature. OTOLARYNGOLOGY CASE REPORTS 2022. [DOI: 10.1016/j.xocr.2022.100406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
|
13
|
Turkunova ME, Barbitoff YA, Serebryakova EA, Polev DE, Berseneva OS, Bashnina EB, Baranov VS, Glotov OS, Glotov AS. Molecular Genetics and Pathogenesis of the Floating Harbor Syndrome: Case Report of Long-Term Growth Hormone Treatment and a Literature Review. Front Genet 2022; 13:846101. [PMID: 35664296 PMCID: PMC9157637 DOI: 10.3389/fgene.2022.846101] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 04/11/2022] [Indexed: 11/16/2022] Open
Abstract
Introduction: Floating Harbor syndrome (FHS) is an extremely rare disorder, with slightly more than a hundred cases reported worldwide. FHS is caused by heterozygous mutations in the SRCAP gene; however, little is known about the pathogenesis of FHS or the effectiveness of its treatment. Methods: Whole-exome sequencing (WES) was performed for the definitive molecular diagnosis of the disease. Identified variants were validated using Sanger sequencing. In addition, systematic literature and public data on genetic variation in SRCAP and the effects of growth hormone (GH) treatment was conducted. Results: We herein report the first case of FHS in the Russian Federation. The male proband presented with most of the typical phenotypic features of FHS, including short stature, skeletal and facial features, delayed growth and bone age, high pitched voice, and intellectual impairment. The proband also had partial growth hormone deficiency. We report the history of treatment of the proband with GH, which resulted in modest improvement in growth prior to puberty. WES revealed a pathogenic c.7466C>G (p.Ser2489*) mutation in the last exon of the FHS-linked SRCAP gene. A systematic literature review and analysis of available genetic variation datasets highlighted an unusual distribution of pathogenic variants in SRCAP and confirmed the lack of pathogenicity for variants outside of exons 33 and 34. Finally, we suggested a new model of FHS pathogenesis which provides possible basis for the dominant negative nature of FHS-causing mutations and explains limited effects of GH treatment in FHS. Conclusion: Our findings expand the number of reported FHS cases and provide new insights into disease genetics and the efficiency of GH therapy for FHS patients.
Collapse
Affiliation(s)
- Mariia E. Turkunova
- Federal State Budget Institution of Higher Education “North-Western State Medical University Named After I.I Mechnikov” Under the Ministry of Public Health of the Russian Federation, Saint-Petersburg, Russia
| | - Yury A. Barbitoff
- Department of Genomic Medicine, D.O.Ott Research Institute of Obstetrics, Gynaecology and Reproductology, St. Petersburg, Russia
- Bioinformatics Institute, St. Petersburg, Russia
- Department of Genetics and Biotechnology, St. Petersburg State University, St. Petersburg, Russia
| | - Elena A. Serebryakova
- Department of Genomic Medicine, D.O.Ott Research Institute of Obstetrics, Gynaecology and Reproductology, St. Petersburg, Russia
- City Center for Medical Genetics, St. Petersburg, Russia
| | - Dmitrii E. Polev
- Department of Genomic Medicine, D.O.Ott Research Institute of Obstetrics, Gynaecology and Reproductology, St. Petersburg, Russia
| | - Olga S. Berseneva
- Federal State Budget Institution of Higher Education “North-Western State Medical University Named After I.I Mechnikov” Under the Ministry of Public Health of the Russian Federation, Saint-Petersburg, Russia
| | - Elena B. Bashnina
- Federal State Budget Institution of Higher Education “North-Western State Medical University Named After I.I Mechnikov” Under the Ministry of Public Health of the Russian Federation, Saint-Petersburg, Russia
| | - Vladislav S. Baranov
- Department of Genomic Medicine, D.O.Ott Research Institute of Obstetrics, Gynaecology and Reproductology, St. Petersburg, Russia
| | - Oleg S. Glotov
- Department of Genomic Medicine, D.O.Ott Research Institute of Obstetrics, Gynaecology and Reproductology, St. Petersburg, Russia
- Children’s Scientific and Clinical Center for Infectious Diseases of the Federal Medical and Biological Agency, St. Petersburg, Russia
| | - Andrey S. Glotov
- Department of Genomic Medicine, D.O.Ott Research Institute of Obstetrics, Gynaecology and Reproductology, St. Petersburg, Russia
- Laboratory of Biobanking and Genomic Medicine of Institute of Translation Biomedicine, St. Petersburg State University, Saint-Petersburg, Russia
- *Correspondence: Andrey S. Glotov,
| |
Collapse
|
14
|
Messina G, Prozzillo Y, Delle Monache F, Santopietro MV, Atterrato MT, Dimitri P. The ATPase SRCAP is associated with the mitotic apparatus, uncovering novel molecular aspects of Floating-Harbor syndrome. BMC Biol 2021; 19:184. [PMID: 34474679 PMCID: PMC8414691 DOI: 10.1186/s12915-021-01109-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 07/22/2021] [Indexed: 11/10/2022] Open
Abstract
Background A variety of human genetic diseases is known to be caused by mutations in genes encoding chromatin factors and epigenetic regulators, such as DNA or histone modifying enzymes and members of ATP-dependent chromatin remodeling complexes. Floating-Harbor syndrome is a rare genetic disease affecting human development caused by dominant truncating mutations in the SRCAP gene, which encodes the ATPase SRCAP, the core catalytic subunit of the homonymous chromatin-remodeling complex. The main function of the SRCAP complex is to promote the exchange of histone H2A with the H2A.Z variant. According to the canonical role played by the SRCAP protein in epigenetic regulation, the Floating-Harbor syndrome is thought to be a consequence of chromatin perturbations. However, additional potential physiological functions of SRCAP have not been sufficiently explored. Results We combined cell biology, reverse genetics, and biochemical approaches to study the subcellular localization of the SRCAP protein and assess its involvement in cell cycle progression in HeLa cells. Surprisingly, we found that SRCAP associates with components of the mitotic apparatus (centrosomes, spindle, midbody), interacts with a plethora of cytokinesis regulators, and positively regulates their recruitment to the midbody. Remarkably, SRCAP depletion perturbs both mitosis and cytokinesis. Similarly, DOM-A, the functional SRCAP orthologue in Drosophila melanogaster, is found at centrosomes and the midbody in Drosophila cells, and its depletion similarly affects both mitosis and cytokinesis. Conclusions Our findings provide first evidence suggesting that SRCAP plays previously undetected and evolutionarily conserved roles in cell division, independent of its functions in chromatin regulation. SRCAP may participate in two different steps of cell division: by ensuring proper chromosome segregation during mitosis and midbody function during cytokinesis. Moreover, our findings emphasize a surprising scenario whereby alterations in cell division produced by SRCAP mutations may contribute to the onset of Floating-Harbor syndrome. Supplementary Information The online version contains supplementary material available at 10.1186/s12915-021-01109-x.
Collapse
Affiliation(s)
- Giovanni Messina
- Dipartimento di Biologia e Biotecnologie "Charles Darwin" Sapienza Università di Roma, Via dei Sardi, 70, Roma, Italy. .,Istituto Pasteur Italia Fondazione Cenci-Bolognetti, Viale Regina Elena, 291, 00161, Roma, Italy.
| | - Yuri Prozzillo
- Dipartimento di Biologia e Biotecnologie "Charles Darwin" Sapienza Università di Roma, Via dei Sardi, 70, Roma, Italy
| | - Francesca Delle Monache
- Dipartimento di Biologia e Biotecnologie "Charles Darwin" Sapienza Università di Roma, Via dei Sardi, 70, Roma, Italy
| | - Maria Virginia Santopietro
- Dipartimento di Biologia e Biotecnologie "Charles Darwin" Sapienza Università di Roma, Via dei Sardi, 70, Roma, Italy
| | - Maria Teresa Atterrato
- Dipartimento di Biologia e Biotecnologie "Charles Darwin" Sapienza Università di Roma, Via dei Sardi, 70, Roma, Italy
| | - Patrizio Dimitri
- Dipartimento di Biologia e Biotecnologie "Charles Darwin" Sapienza Università di Roma, Via dei Sardi, 70, Roma, Italy.
| |
Collapse
|
15
|
Nogueira E, Garma C, Lobo C, Del Olmo B, Arroyo JM, Gómez I. Severe developmental expressive language disorder due to a frameshift mutation in exon 18 of SRCAP gene, far away from the mutational hotspot in exons 33 and 34 associated to the Floating-Harbor syndrome. Neurol Sci 2021; 42:4349-4352. [PMID: 34213696 DOI: 10.1007/s10072-021-05423-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 06/17/2021] [Indexed: 01/13/2023]
Affiliation(s)
- Enrique Nogueira
- Eurofins-Megalab Molecular Diagnostics Laboratory, Madrid, Spain. .,Genetics Service of the Hospital La Zarzuela, Madrid, Spain. .,Genetics Service of the Hospital San Rafael, Madrid, Spain.
| | - Carmen Garma
- Eurofins-Megalab Molecular Diagnostics Laboratory, Madrid, Spain.,Genetics Service of the Hospital La Zarzuela, Madrid, Spain.,Genetics Service of the Hospital San Rafael, Madrid, Spain
| | - Concepción Lobo
- Eurofins-Megalab Molecular Diagnostics Laboratory, Madrid, Spain
| | - Beatriz Del Olmo
- Eurofins-Megalab Molecular Diagnostics Laboratory, Madrid, Spain
| | | | - Iván Gómez
- Eurofins-Megalab Molecular Diagnostics Laboratory, Madrid, Spain.,Genetics Service of the Hospital La Zarzuela, Madrid, Spain
| |
Collapse
|
16
|
Proteomic analysis of bone marrow-derived mesenchymal stem cell extracellular vesicles from healthy donors: implications for proliferation, angiogenesis, Wnt signaling, and the basement membrane. Stem Cell Res Ther 2021; 12:328. [PMID: 34090527 PMCID: PMC8180068 DOI: 10.1186/s13287-021-02405-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 05/19/2021] [Indexed: 02/07/2023] Open
Abstract
Background Bone marrow-derived mesenchymal stem cells (BM-MSCs) have shown therapeutic potential in various in vitro and in vivo studies in cutaneous wound healing. Furthermore, there are ubiquitous studies highlighting the pro-regenerative effects of BM-MSC extracellular vesicles (BM-MSC EVs). The similarities and differences in BM-MSC EV cargo among potential healthy donors are not well understood. Variation in EV protein cargo is important to understand, as it may be useful in identifying potential therapeutic applications in clinical trials. We hypothesized that the donors would share both important similarities and differences in cargo relating to cell proliferation, angiogenesis, Wnt signaling, and basement membrane formation—processes shown to be critical for effective cutaneous wound healing. Methods We harvested BM-MSC EVs from four healthy human donors who underwent strict screening for whole bone marrow donation and further Good Manufacturing Practices-grade cell culture expansion for candidate usage in clinical trials. BM-MSC EV protein cargo was determined via mass spectrometry and Proteome Discoverer software. Corresponding proteomic networks were analyzed via the UniProt Consortium and STRING consortium databases. Results More than 3000 proteins were identified in each of the donors, sharing > 600 proteins among all donors. Despite inter-donor variation in protein identities, there were striking similarities in numbers of proteins per biological functional category. In terms of biologic function, the proteins were most associated with transport of ions and proteins, transcription, and the cell cycle, relating to cell proliferation. The donors shared essential cargo relating to angiogenesis, Wnt signaling, and basement membrane formation—essential processes in modulating cutaneous wound repair. Conclusions Healthy donors of BM-MSC EVs contain important similarities and differences among protein cargo that may play important roles in their pro-regenerative functions. Further studies are needed to correlate proteomic signatures to functional outcomes in cutaneous repair.
Collapse
|
17
|
Sub-genic intolerance, ClinVar, and the epilepsies: A whole-exome sequencing study of 29,165 individuals. Am J Hum Genet 2021; 108:965-982. [PMID: 33932343 DOI: 10.1016/j.ajhg.2021.04.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 04/08/2021] [Indexed: 12/23/2022] Open
Abstract
Both mild and severe epilepsies are influenced by variants in the same genes, yet an explanation for the resulting phenotypic variation is unknown. As part of the ongoing Epi25 Collaboration, we performed a whole-exome sequencing analysis of 13,487 epilepsy-affected individuals and 15,678 control individuals. While prior Epi25 studies focused on gene-based collapsing analyses, we asked how the pattern of variation within genes differs by epilepsy type. Specifically, we compared the genetic architectures of severe developmental and epileptic encephalopathies (DEEs) and two generally less severe epilepsies, genetic generalized epilepsy and non-acquired focal epilepsy (NAFE). Our gene-based rare variant collapsing analysis used geographic ancestry-based clustering that included broader ancestries than previously possible and revealed novel associations. Using the missense intolerance ratio (MTR), we found that variants in DEE-affected individuals are in significantly more intolerant genic sub-regions than those in NAFE-affected individuals. Only previously reported pathogenic variants absent in available genomic datasets showed a significant burden in epilepsy-affected individuals compared with control individuals, and the ultra-rare pathogenic variants associated with DEE were located in more intolerant genic sub-regions than variants associated with non-DEE epilepsies. MTR filtering improved the yield of ultra-rare pathogenic variants in affected individuals compared with control individuals. Finally, analysis of variants in genes without a disease association revealed a significant burden of loss-of-function variants in the genes most intolerant to such variation, indicating additional epilepsy-risk genes yet to be discovered. Taken together, our study suggests that genic and sub-genic intolerance are critical characteristics for interpreting the effects of variation in genes that influence epilepsy.
Collapse
|
18
|
Truncating SRCAP variants outside the Floating-Harbor syndrome locus cause a distinct neurodevelopmental disorder with a specific DNA methylation signature. Am J Hum Genet 2021; 108:1053-1068. [PMID: 33909990 PMCID: PMC8206150 DOI: 10.1016/j.ajhg.2021.04.008] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 03/31/2021] [Indexed: 02/08/2023] Open
Abstract
Truncating variants in exons 33 and 34 of the SNF2-related CREBBP activator protein (SRCAP) gene cause the neurodevelopmental disorder (NDD) Floating-Harbor syndrome (FLHS), characterized by short stature, speech delay, and facial dysmorphism. Here, we present a cohort of 33 individuals with clinical features distinct from FLHS and truncating (mostly de novo) SRCAP variants either proximal (n = 28) or distal (n = 5) to the FLHS locus. Detailed clinical characterization of the proximal SRCAP individuals identified shared characteristics: developmental delay with or without intellectual disability, behavioral and psychiatric problems, non-specific facial features, musculoskeletal issues, and hypotonia. Because FLHS is known to be associated with a unique set of DNA methylation (DNAm) changes in blood, a DNAm signature, we investigated whether there was a distinct signature associated with our affected individuals. A machine-learning model, based on the FLHS DNAm signature, negatively classified all our tested subjects. Comparing proximal variants with typically developing controls, we identified a DNAm signature distinct from the FLHS signature. Based on the DNAm and clinical data, we refer to the condition as “non-FLHS SRCAP-related NDD.” All five distal variants classified negatively using the FLHS DNAm model while two classified positively using the proximal model. This suggests divergent pathogenicity of these variants, though clinically the distal group presented with NDD, similar to the proximal SRCAP group. In summary, for SRCAP, there is a clear relationship between variant location, DNAm profile, and clinical phenotype. These results highlight the power of combined epigenetic, molecular, and clinical studies to identify and characterize genotype-epigenotype-phenotype correlations.
Collapse
|
19
|
Dilshat R, Vu HN, Steingrímsson E. Epigenetic regulation during melanocyte development and homeostasis. Exp Dermatol 2021; 30:1033-1050. [PMID: 34003523 DOI: 10.1111/exd.14391] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 04/09/2021] [Accepted: 05/09/2021] [Indexed: 12/26/2022]
Abstract
Melanocytes originate in the neural crest as precursor cells which then migrate and proliferate to reach their destination where they differentiate into pigment-producing cells. Melanocytes not only determine the colour of hair, skin and eyes but also protect against the harmful effects of UV irradiation. The establishment of the melanocyte lineage is regulated by a defined set of transcription factors and signalling pathways that direct the specific gene expression programmes underpinning melanoblast specification, survival, migration, proliferation and differentiation. In addition, epigenetic modifiers and replacement histones play key roles in regulating gene expression and its timing during the different steps of this process. Here, we discuss the evidence for the role of epigenetic regulators in melanocyte development and function and how they interact with transcription factors and signalling pathways to establish and maintain this important cell lineage.
Collapse
Affiliation(s)
- Ramile Dilshat
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, BioMedical Center, University of Iceland, Reykjavik, Iceland
| | - Hong Nhung Vu
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, BioMedical Center, University of Iceland, Reykjavik, Iceland
| | - Eiríkur Steingrímsson
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, BioMedical Center, University of Iceland, Reykjavik, Iceland
| |
Collapse
|
20
|
Agrawal S, Gupta S, Neupane S, Das D. Floating harbour syndrome with medial entropion: a rare association and brief review. BMJ Case Rep 2021; 14:14/5/e242125. [PMID: 33962930 PMCID: PMC8108651 DOI: 10.1136/bcr-2021-242125] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Affiliation(s)
- Sahil Agrawal
- Ophthalmology, Dr. Rajendra Prasad Centre for Ophthalmic Sciences, AIIMS, Delhi, New Delhi, India
| | - Saloni Gupta
- Ophthalmology, Northern Railway Central Hospital New Delhi, New Delhi, Delhi, India
| | - Swechya Neupane
- Ophthalmology, Dr. Rajendra Prasad Centre for Ophthalmic Sciences, AIIMS, Delhi, New Delhi, India
| | - Deepsekhar Das
- Ophthalmology, Dr. Rajendra Prasad Centre for Ophthalmic Sciences, AIIMS, Delhi, New Delhi, India
| |
Collapse
|
21
|
Hou C, Xie L, Qiu Q, Lin H, Liu W, Sun X, Zhang Y, Xu M, Li Y, Xiao T. Generation of an induced pluripotent stem cell line from a Chinese Han infant with floating-harbor syndrome accompanied with dilated cardiomyopathy. Stem Cell Res 2021; 51:102182. [PMID: 33517121 DOI: 10.1016/j.scr.2021.102182] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 12/22/2020] [Accepted: 01/12/2021] [Indexed: 11/18/2022] Open
Abstract
Floating-harbor syndrome, are mainly caused by heterozygous truncating mutations in SRCAP. To our best knowledge, the mutation (c.452_453del) located in the fifth exon of SRCAP, has not been reported yet. Herein, an induced pluripotent stem cell (iPSC) line was generated from the peripheral blood mononuclear cells of an infant with floating-harbor syndrome accompanied with dilated cardiomyopathy through Sendaivirus-mediated reprogramming. These iPSCs have excellent cellular features, including stable amplification, pluripotent markers expression, and spontaneous differentiation into three germ layers, and a normal karyotype. These iPSCs provide a suitable cell model to study the mechanism of Floating-harbor syndrome.
Collapse
Affiliation(s)
- Cuilan Hou
- Department of Cardiology, Shanghai Children's Hospital, Shanghai Jiaotong University, Shanghai 200062, China; NHC Key Laboratory of Medical Embryogenesis and Developmental Molecular Biology, Shanghai Key Laboratory of Embryo and Reproduction Engineering, Shanghai 200062, China.
| | - Lijian Xie
- Department of Cardiology, Shanghai Children's Hospital, Shanghai Jiaotong University, Shanghai 200062, China
| | - Qingzhu Qiu
- Department of Cardiology, Shanghai Children's Hospital, Shanghai Jiaotong University, Shanghai 200062, China
| | - Hao Lin
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200062, China
| | - Wei Liu
- Department of Cardiology, Shanghai Children's Hospital, Shanghai Jiaotong University, Shanghai 200062, China
| | - Xiaomin Sun
- Department of Cardiology, Shanghai Children's Hospital, Shanghai Jiaotong University, Shanghai 200062, China
| | - Yongwei Zhang
- Department of Cardiology, Shanghai Children's Hospital, Shanghai Jiaotong University, Shanghai 200062, China
| | - Meng Xu
- Department of Cardiology, Shanghai Children's Hospital, Shanghai Jiaotong University, Shanghai 200062, China
| | - Yun Li
- Department of Cardiology, Shanghai Children's Hospital, Shanghai Jiaotong University, Shanghai 200062, China; NHC Key Laboratory of Medical Embryogenesis and Developmental Molecular Biology, Shanghai Key Laboratory of Embryo and Reproduction Engineering, Shanghai 200062, China.
| | - Tingting Xiao
- Department of Cardiology, Shanghai Children's Hospital, Shanghai Jiaotong University, Shanghai 200062, China; NHC Key Laboratory of Medical Embryogenesis and Developmental Molecular Biology, Shanghai Key Laboratory of Embryo and Reproduction Engineering, Shanghai 200062, China.
| |
Collapse
|
22
|
Bo H, Jiang L, Zheng J, Sun J. Floating-Harbor Syndrome Treated With Recombinant Human Growth Hormone: A Case Report and Literature Review. Front Pediatr 2021; 9:747353. [PMID: 34805044 PMCID: PMC8602871 DOI: 10.3389/fped.2021.747353] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 10/13/2021] [Indexed: 11/29/2022] Open
Abstract
Introduction: We aimed to summarize the clinical characteristics of Floating-Harbor syndrome (FHS) and the effect of recombinant human growth hormone (rhGH) to increase height. Methods: The clinical manifestations, gene sequencing results, treatment, and regression of one child with FHS were reported at the Department of Pediatrics, General Hospital of Tianjin Medical University, in July 2020. PubMed was searched using the keyword "Floating-Harbor Syndrome" up to March 2021 to obtain clinical information on children with FHS for review. Results: The child, who was a male aged 6 years and 9 months, presented to the clinic with main complaints of delayed language development since childhood and a short stature for 4 years. The child's short stature, peculiar facial features, delayed language development, and delayed bone development were considered alongside genetic testing and Sanger sequencing to verify the results. A heterozygous mutation (c.7401delC; p.Ile2468Phefs*7) was identified in exon 34 of the SRCAP gene, which was a frameshift mutation, and Sanger verification showed that neither parent had this mutation. The child was administered subcutaneous injection of rhGH (0.13 U/kg/day) and was followed up regularly. At the time of writing, the child had been treated for 6 months and was 7 years and 3 months old with a height of 106.3 cm (-3.69 SDS), which was a height increase of 6.3 cm. The patient did not complain of discomfort during treatment and presented normal laboratory tests results. Twenty-two children with FHS treated with rhGH were included in the literature review, and most of these patients demonstrated an increase in height SDS without adverse effects. Conclusion: Short stature, delayed skeletal maturation, impaired language expression, intellectual deficits, and peculiar facial features are the main clinical features of FHS. rhGH can be used as a treatment to increase height in patients with FHS, but its effectiveness and safety still need to be monitored in larger sample sizes over longer periods of time.
Collapse
Affiliation(s)
- Hui Bo
- Jinghai Clinical College of Tianjin Medical University, Tianjin, China.,Department of Pediatrics, Jinghai District Hospital, Tianjin, China
| | - Lihong Jiang
- Department of Pediatrics, General Hospital of Tianjin Medical University, Tianjin, China
| | - Jiaqi Zheng
- Department of Pediatrics, General Hospital of Tianjin Medical University, Tianjin, China
| | - Jie Sun
- Department of Pediatrics, General Hospital of Tianjin Medical University, Tianjin, China
| |
Collapse
|
23
|
Ercoskun P, Yuce-Kahraman C. Novel Findings in Floating-Harbor Syndrome and a Mini-Review of the Literature. Mol Syndromol 2020; 12:52-56. [PMID: 33776628 DOI: 10.1159/000512050] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 10/03/2020] [Indexed: 11/19/2022] Open
Abstract
Floating-Harbor syndrome (FHS) is a rare autosomal dominant genetic disorder characterized by proportionate short stature with delayed bone maturation, lack of expressive language, and distinctive facial features including a large nose, long eyelashes, deeply set eyes, and triangular face. Mutations in the SRCAP gene cause truncated SNF2-related CREBBP activator protein (SRCAP) and lead to FHS. SRCAP is one of several proteins that act as coactivator for the CREB-binding protein which is associated with Rubinstein-Taybi syndrome (RSTS). This condition likely explains the phenotypic overlap between FHS and RSTS. Herein, we report on a patient with FHS who also had dystrophic toenails, preauricular skin tag, and nasolacrimal duct obstruction which is also defined in patients with RSTS. In summary, the fact that especially nasolacrimal duct obstruction has also been observed in RSTS reinforces the idea that this finding is one of the features of FHS. Assessment of the lacrimal system and examination of skin and nails should be suggested in patients with FHS.
Collapse
Affiliation(s)
- Pelin Ercoskun
- Department of Medical Genetics, Faculty of Medicine, Ataturk University, Erzurum, Turkey
| | - Cigdem Yuce-Kahraman
- Department of Medical Genetics, Faculty of Medicine, Ataturk University, Erzurum, Turkey
| |
Collapse
|
24
|
Inoue T, Nakamura A, Iwahashi-Odano M, Tanase-Nakao K, Matsubara K, Nishioka J, Maruo Y, Hasegawa Y, Suzumura H, Sato S, Kobayashi Y, Murakami N, Nakabayashi K, Yamazawa K, Fuke T, Narumi S, Oka A, Ogata T, Fukami M, Kagami M. Contribution of gene mutations to Silver-Russell syndrome phenotype: multigene sequencing analysis in 92 etiology-unknown patients. Clin Epigenetics 2020; 12:86. [PMID: 32546215 PMCID: PMC7298762 DOI: 10.1186/s13148-020-00865-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Accepted: 05/14/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Silver-Russell syndrome (SRS) is characterized by growth failure and dysmorphic features. Major (epi)genetic causes of SRS are loss of methylation on chromosome 11p15 (11p15 LOM) and maternal uniparental disomy of chromosome 7 (upd(7)mat). However, IGF2, CDKN1C, HMGA2, and PLAG1 mutations infrequently cause SRS. In addition, other imprinting disturbances, pathogenic copy number variations (PCNVs), and monogenic disorders sometimes lead to SRS phenotype. This study aimed to clarify the frequency and clinical features of the patients with gene mutations among etiology-unknown patients with SRS phenotype. RESULTS Multigene sequencing was performed in 92 out of 336 patients referred to us for genetic testing for SRS. The clinical features of the patients were evaluated based on the Netchine-Harbison clinical scoring system. None of the patients showed 11p15 LOM, upd(7)mat, abnormal methylation levels for six differentially methylated regions (DMRs), namely, PLAGL1:alt-TSS-DMR on chromosome 6, KCNQ1OT1:TSS-DMR on chromosome 11, MEG3/DLK1:IG-DMR on chromosome 14, MEG3:TSS-DMR on chromosome 14, SNURF:TSS-DMR on chromosome 15, and GNAS A/B:TSS-DMR on chromosome 20, PCNVs, or maternal uniparental disomy of chromosome 16. Using next-generation sequencing and Sanger sequencing, we screened four SRS-causative genes and 406 genes related to growth failure and/or skeletal dysplasia. We identified four pathogenic or likely pathogenic variants in responsible genes for SRS (4.3%: IGF2 in two patients, CDKN1C, and PLAG1), and five pathogenic variants in causative genes for known genetic syndromes presenting with growth failure (5.4%: IGF1R abnormality (IGF1R), SHORT syndrome (PIK3R1), Floating-Harbor syndrome (SRCAP), Pitt-Hopkins syndrome (TCF4), and Noonan syndrome (PTPN11)). Functional analysis indicated the pathogenicity of the CDKN1C variant. The variants we detected in CDKN1C and PLAG1 were the second and third variants leading to SRS, respectively. Our patients with CDKN1C and PLAG1 variants showed similar phenotypes to previously reported patients. Furthermore, our data confirmed IGF1R abnormality, SHORT syndrome, and Floating-Harbor syndrome are differential diagnoses of SRS because of the shared phenotypes among these syndromes and SRS. On the other hand, the patients with pathogenic variants in causative genes for Pitt-Hopkins syndrome and Noonan syndrome were atypical of these syndromes and showed partial clinical features of SRS. CONCLUSIONS We identified nine patients (9.8%) with pathogenic or likely pathogenic variants out of 92 etiology-unknown patients with SRS phenotype. This study expands the molecular spectrum of SRS phenotype.
Collapse
Affiliation(s)
- Takanobu Inoue
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo, 157-8535 Japan
- Department of Pediatrics, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655 Japan
| | - Akie Nakamura
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo, 157-8535 Japan
- Department of Pediatrics, Hokkaido University Graduate School of Medicine, Kita15, Nishi7, Kita-Ku, Sapporo, 060-8648 Japan
| | - Megumi Iwahashi-Odano
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo, 157-8535 Japan
| | - Kanako Tanase-Nakao
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo, 157-8535 Japan
| | - Keiko Matsubara
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo, 157-8535 Japan
| | - Junko Nishioka
- Department of Pediatrics and Child Health, Kurume University School of Medicine, 67 Asahi-Machi, Kurume, 830-0011 Japan
| | - Yoshihiro Maruo
- Department of Pediatrics, Shiga University of Medical Science, Seta Tsukinowa-cho, Otsu, 520-2192 Japan
| | - Yukihiro Hasegawa
- Division of Endocrinology and Metabolism, Tokyo Metropolitan Children’s Medical Center, 2-8-29 Musashidai, Fuchu, Tokyo, 183-8561 Japan
| | - Hiroshi Suzumura
- Department of Pediatrics, Dokkyo Medical University, 880 Kitakobayashi, Mibu, 321-0293 Japan
| | - Seiji Sato
- Department of Pediatrics, Saitama City Hospital, 2460, Mimuro, Midori-ku, Saitama, 336-8522 Japan
| | - Yoshiyuki Kobayashi
- Department of Pediatrics, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553 Japan
| | - Nobuyuki Murakami
- Department of Pediatrics, Dokkyo Medical University Saitama Medical Center, 2-1-50, Minamikoshigaya, Koshigaya, 343-8555 Japan
| | - Kazuhiko Nakabayashi
- Department of Maternal-Fetal Biology, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo, 157-8535 Japan
| | - Kazuki Yamazawa
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo, 157-8535 Japan
- Medical Genetics Center, National Hospital Organization Tokyo Medical Center, 2-5-1 Higashigaoka, Meguro-ku, Tokyo, 152-8902 Japan
| | - Tomoko Fuke
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo, 157-8535 Japan
| | - Satoshi Narumi
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo, 157-8535 Japan
| | - Akira Oka
- Department of Pediatrics, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655 Japan
| | - Tsutomu Ogata
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo, 157-8535 Japan
- Department of Pediatrics, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, 431-3192 Japan
| | - Maki Fukami
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo, 157-8535 Japan
| | - Masayo Kagami
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo, 157-8535 Japan
| |
Collapse
|
25
|
Greenberg RS, Long HK, Swigut T, Wysocka J. Single Amino Acid Change Underlies Distinct Roles of H2A.Z Subtypes in Human Syndrome. Cell 2020; 178:1421-1436.e24. [PMID: 31491386 DOI: 10.1016/j.cell.2019.08.002] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 03/27/2019] [Accepted: 08/01/2019] [Indexed: 12/31/2022]
Abstract
The developmental disorder Floating-Harbor syndrome (FHS) is caused by heterozygous truncating mutations in SRCAP, a gene encoding a chromatin remodeler mediating incorporation of histone variant H2A.Z. Here, we demonstrate that FHS-associated mutations result in loss of SRCAP nuclear localization, alter neural crest gene programs in human in vitro models and Xenopus embryos, and cause craniofacial defects. These defects are mediated by one of two H2A.Z subtypes, H2A.Z.2, whose knockdown mimics and whose overexpression rescues the FHS phenotype. Selective rescue by H2A.Z.2 is conferred by one of the three amino acid differences between the H2A.Z subtypes, S38/T38. We further show that H2A.Z.1 and H2A.Z.2 genomic occupancy patterns are qualitatively similar, but quantitatively distinct, and H2A.Z.2 incorporation at AT-rich enhancers and expression of their associated genes are both sensitized to SRCAP truncations. Altogether, our results illuminate the mechanism underlying a human syndrome and uncover selective functions of H2A.Z subtypes during development.
Collapse
Affiliation(s)
- Rachel S Greenberg
- Department of Chemical and Systems Biology and Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Hannah K Long
- Department of Chemical and Systems Biology and Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Tomek Swigut
- Department of Chemical and Systems Biology and Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Joanna Wysocka
- Department of Chemical and Systems Biology and Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
26
|
Son HW, Lee JE, Oh SH, Keum C, Chung WY. Effects of long-term growth hormone therapy in a girl with Floating-Harbor syndrome. Ann Pediatr Endocrinol Metab 2020; 25:126-131. [PMID: 32615693 PMCID: PMC7336260 DOI: 10.6065/apem.1938144.072] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 05/31/2019] [Indexed: 12/12/2022] Open
Abstract
Floating-Harbor syndrome is a rare autosomal dominant disorder that presents with short stature, facial dysmorphism, significantly delayed bone age, skeletal abnormalities, speech and language problems, and intellectual disabilities. Although short stature is one of the main clinical manifestations, use of growth hormone therapy in Floating-Harbor syndrome patients has been limited. Only a few reports have investigated the response to growth hormone therapy with regard to final adult height. We report the case of a 7-year-old girl with FloatingHarbor syndrome and a heterozygous mutation, c.7330C > T (p.Arg2444*), in the SRCAP gene. The patient exhibited dysmorphic facial features, severe intellectual disabilities, obsessive-compulsive and aggressive behaviors, and short stature without growth hormone deficiency. Her height standard deviation score improved after 55 months of growth hormone therapy.
Collapse
Affiliation(s)
- Hyun Woo Son
- Department of Pediatrics, Inje University Busan Paik Hospital, Busan, Korea
| | - Jeong Eun Lee
- Department of Pediatrics, Inje University Busan Paik Hospital, Busan, Korea
| | - Seung Hwan Oh
- Department of Diagnostic Laboratory Medicine, Inje University Busan Paik Hospital, Busan, Korea
| | - Changwon Keum
- Rare Genetic Disease Research Center, 3 Billion, Seoul, Korea
| | - Woo Yeong Chung
- Department of Pediatrics, Inje University Busan Paik Hospital, Busan, Korea,Address for correspondence: Woo Yeong Chung, MD, PhD Department of Pediatrics, Inje University Busan Paik Hospital, Inje University College of Medicine, 75 BokJi-ro, Busanjin-gu, Busan 47392, Korea Tel: +82-51-890-6280 Fax: +82-51-895-7785 E-mail:
| |
Collapse
|
27
|
Homma TK, Freire BL, Honjo R, Dauber A, Funari MFA, Lerario AM, Albuquerque EVA, Vasques GA, Bertola DR, Kim CA, Malaquias AC, Jorge AAL. Growth and Clinical Characteristics of Children with Floating-Harbor Syndrome: Analysis of Current Original Data and a Review of the Literature. Horm Res Paediatr 2020; 92:115-123. [PMID: 31715605 DOI: 10.1159/000503782] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 09/28/2019] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Floating-Harbor syndrome (FHS) is a rare condition characterized by dysmorphic facial features, short stature, and expressive language delay. OBJECTIVE The aim of this study was to describe a cohort of patients with FHS and review the literature about the response to recombinant human growth hormone (rhGH) therapy. METHODS Anthropometric and laboratory data from 7 patients with FHS were described. The molecular diagnosis was established by multigene analysis. Moreover, we reviewed the literature concerning patients with FHS treated with rhGH. RESULTS All 7 patients were born small for gestational age. At first evaluation, 6 patients had a height standard deviation score (SDS) ≤-2 and 1 had short stature in relation to their target height. Bone age was usually delayed, which rapidly advanced during puberty. Nonspecific skeletal abnormalities were frequently noticed, and normal to elevated plasma IGF-I levels were observed in all except 1 patient with growth hormone deficiency. Information about 20 patients with FHS treated with rhGH was analyzed (4 from our cohort and 16 from the literature). The median height changes during the treatment period (approx. 2.9 years) were 1.1 SDS (range from -0.4 to 3.1). Nontreated patients had an adult height SDS of -4.1 ± 1.2 (n = 10) versus -2.6 ± 0.8 SDS (n = 7, p 0.012) for treated patients. CONCLUSION We observed a laboratory profile compatible with IGF-1 insensitivity in some patients with FHS. Nevertheless, our study suggests that children with FHS may be considered as candidates for rhGH therapy. Further studies are necessary to establish the real benefit and safety of rhGH therapy in these patients.
Collapse
Affiliation(s)
- Thais K Homma
- Unidade de Endocrinologia Genetica, Laboratorio de Endocrinologia Celular e Molecular LIM25, Disciplina de Endocrinologia da Faculdade de Medicina da Universidade de Sao Paulo (FMUSP), Sao Paulo, Brazil.,Unidade de Endocrinologia do Desenvolvimento, Laboratorio de Hormonios e Genetica Molecular LIM42, Hospital das Clínicas da Faculdade de Medicina da Universidade de Sao Paulo (FMUSP), Sao Paulo, Brazil
| | - Bruna L Freire
- Unidade de Endocrinologia Genetica, Laboratorio de Endocrinologia Celular e Molecular LIM25, Disciplina de Endocrinologia da Faculdade de Medicina da Universidade de Sao Paulo (FMUSP), Sao Paulo, Brazil.,Unidade de Endocrinologia do Desenvolvimento, Laboratorio de Hormonios e Genetica Molecular LIM42, Hospital das Clínicas da Faculdade de Medicina da Universidade de Sao Paulo (FMUSP), Sao Paulo, Brazil
| | - Rachel Honjo
- Unidade de Genetica do Instituto da Criança, Hospital das Clínicas da Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Andrew Dauber
- Division of Endocrinology, Children's National Health System, Washington, District of Columbia, USA
| | - Mariana F A Funari
- Unidade de Endocrinologia do Desenvolvimento, Laboratorio de Hormonios e Genetica Molecular LIM42, Hospital das Clínicas da Faculdade de Medicina da Universidade de Sao Paulo (FMUSP), Sao Paulo, Brazil
| | - Antonio M Lerario
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan, Ann Arbor, Michigan, USA
| | - Edoarda V A Albuquerque
- Unidade de Endocrinologia Genetica, Laboratorio de Endocrinologia Celular e Molecular LIM25, Disciplina de Endocrinologia da Faculdade de Medicina da Universidade de Sao Paulo (FMUSP), Sao Paulo, Brazil
| | - Gabriela A Vasques
- Unidade de Endocrinologia Genetica, Laboratorio de Endocrinologia Celular e Molecular LIM25, Disciplina de Endocrinologia da Faculdade de Medicina da Universidade de Sao Paulo (FMUSP), Sao Paulo, Brazil
| | - Debora R Bertola
- Unidade de Genetica do Instituto da Criança, Hospital das Clínicas da Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Chong A Kim
- Unidade de Genetica do Instituto da Criança, Hospital das Clínicas da Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Alexsandra C Malaquias
- Unidade de Endocrinologia Genetica, Laboratorio de Endocrinologia Celular e Molecular LIM25, Disciplina de Endocrinologia da Faculdade de Medicina da Universidade de Sao Paulo (FMUSP), Sao Paulo, Brazil.,Unidade de Endocrinologia Pediatrica, Departamento de Pediatria, Irmandade da Santa Casa de Misericórdia de São Paulo, Faculdade de Ciências Médicas da Santa Casa de São Paulo, Sao Paulo, Brazil
| | - Alexander A L Jorge
- Unidade de Endocrinologia Genetica, Laboratorio de Endocrinologia Celular e Molecular LIM25, Disciplina de Endocrinologia da Faculdade de Medicina da Universidade de Sao Paulo (FMUSP), Sao Paulo, Brazil, .,Unidade de Endocrinologia do Desenvolvimento, Laboratorio de Hormonios e Genetica Molecular LIM42, Hospital das Clínicas da Faculdade de Medicina da Universidade de Sao Paulo (FMUSP), Sao Paulo, Brazil,
| |
Collapse
|
28
|
Squeo GM, Augello B, Massa V, Milani D, Colombo EA, Mazza T, Castellana S, Piccione M, Maitz S, Petracca A, Prontera P, Accadia M, Della Monica M, Di Giacomo MC, Melis D, Selicorni A, Giglio S, Fischetto R, Di Fede E, Malerba N, Russo M, Castori M, Gervasini C, Merla G. Customised next-generation sequencing multigene panel to screen a large cohort of individuals with chromatin-related disorder. J Med Genet 2020; 57:760-768. [PMID: 32170002 DOI: 10.1136/jmedgenet-2019-106724] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 02/11/2020] [Accepted: 02/19/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND The regulation of the chromatin state by epigenetic mechanisms plays a central role in gene expression, cell function, and maintenance of cell identity. Hereditary disorders of chromatin regulation are a group of conditions caused by abnormalities of the various components of the epigenetic machinery, namely writers, erasers, readers, and chromatin remodelers. Although neurological dysfunction is almost ubiquitous in these disorders, the constellation of additional features characterizing many of these genes and the emerging clinical overlap among them indicate the existence of a community of syndromes. The introduction of high-throughput next generation sequencing (NGS) methods for testing multiple genes simultaneously is a logical step for the implementation of diagnostics of these disorders. METHODS We screened a heterogeneous cohort of 263 index patients by an NGS-targeted panel, containing 68 genes associated with more than 40 OMIM entries affecting chromatin function. RESULTS This strategy allowed us to identify clinically relevant variants in 87 patients (32%), including 30 for which an alternative clinical diagnosis was proposed after sequencing analysis and clinical re-evaluation. CONCLUSION Our findings indicate that this approach is effective not only in disorders with locus heterogeneity, but also in order to anticipate unexpected misdiagnoses due to clinical overlap among cognate disorders. Finally, this work highlights the utility of a prompt diagnosis in such a clinically and genetically heterogeneous group of disorders that we propose to group under the umbrella term of chromatinopathies.
Collapse
Affiliation(s)
- Gabriella Maria Squeo
- Division of Medical Genetics, IRCCS Ospedale Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Bartolomeo Augello
- Division of Medical Genetics, IRCCS Ospedale Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Valentina Massa
- Dipartimento di Scienze della Salute, Universita degli Studi di Milano Dipartimento di Scienze della Salute, Milano, Italy
| | - Donatella Milani
- UOSD Pediatria ad alta intensità di cura, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Elisa Adele Colombo
- Dipartimento di Scienze della Salute, Universita degli Studi di Milano Dipartimento di Scienze della Salute, Milano, Italy
| | - Tommaso Mazza
- Bioinformatics Unit, IRCCS Ospedale Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Stefano Castellana
- Bioinformatics Unit, IRCCS Ospedale Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Maria Piccione
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Silvia Maitz
- Clinical Pediatric Genetics Unit, Pediatrics Clinics, MBBM Foundation, Hospital San Gerardo, Monza, Italy
| | - Antonio Petracca
- Division of Medical Genetics, IRCCS Ospedale Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Paolo Prontera
- Medical Genetics Unit, University of Perugia Hospital SM della Misericordia, Perugia, Italy
| | - Maria Accadia
- Medical Genetics Service, Hospital "Cardinale G. Panico", Tricase, Italy
| | - Matteo Della Monica
- Medical Genetics Unit, Cardarelli Hospital, Largo A Cardarelli, Napoli, Italy
| | | | - Daniela Melis
- Department of Translational Medical Science, Section of Pediatrics, University of Naples Federico II, Napoli, Italy
| | - Angelo Selicorni
- Pediatric Department, ASST Lariana, Sant'Anna General Hospital, Como, Italy
| | - Sabrina Giglio
- Department of Biomedical, Experimental and Clinical Sciences 'Mario Serio', Medical Genetics Unit, University Hospital Meyer, Firenze, Italy
| | - Rita Fischetto
- Metabolic Diseases, Clinical Genetics and Diabetology Unit, Paediatric Hospital Giovanni XXIII, Bari, Italy
| | - Elisabetta Di Fede
- Dipartimento di Scienze della Salute, Universita degli Studi di Milano Dipartimento di Scienze della Salute, Milano, Italy
| | - Natascia Malerba
- Division of Medical Genetics, IRCCS Ospedale Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Matteo Russo
- Division of Medical Genetics, IRCCS Ospedale Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Marco Castori
- Division of Medical Genetics, IRCCS Ospedale Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Cristina Gervasini
- Dipartimento di Scienze della Salute, Universita degli Studi di Milano Dipartimento di Scienze della Salute, Milano, Italy
| | - Giuseppe Merla
- Division of Medical Genetics, IRCCS Ospedale Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| |
Collapse
|
29
|
Li RM, Lu YC, Li Z, Wang JY, Chang J, Lei SQ, Zeng Q, Sang YM. [Floating-Harbor syndrome: a case report and literature review]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2019; 21:1208-1211. [PMID: 31874661 PMCID: PMC7389003 DOI: 10.7499/j.issn.1008-8830.2019.12.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 09/06/2019] [Indexed: 06/10/2023]
Abstract
Floating-Harbor syndrome (FHS) is an autosomal dominant genetic disease caused by SRCAP mutation. This article reports the clinical features of a boy with FHS. The boy, aged 11 years and 7 months, attended the hospital due to short stature for more than 8 years and had the clinical manifestations of unusual facial features (triangularly shaped face, thin lips and long eyelashes), skeletal dysplasia (curvature finger), expressive language disorder, and retardation of bone age. Genetic detection revealed a novel heterozygous mutation, c.7330 C>T(p.R2444X), in the SRCAP gene. The boy was diagnosed with FHS based on these clinical manifestations and gene detection results. FHS is rare in clinical practice, which may lead to missed diagnosis and misdiagnosis, and gene detection may help with the clinical diagnosis of FHS in children.
Collapse
Affiliation(s)
- Rong-Min Li
- Baoding Key Laboratory of Clinical Research on Children's Respiratory and Digestive Diseases/Baoding Children's Hospital, Baoding, Hebei 071000.
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Intracranial vascular pathology in two further patients with Floating-Harbor syndrome: Proposals for cerebrovascular disease risk management. Eur J Med Genet 2019; 63:103785. [PMID: 31605816 DOI: 10.1016/j.ejmg.2019.103785] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 09/15/2019] [Accepted: 10/06/2019] [Indexed: 01/22/2023]
Abstract
Floating-Harbor syndrome (FHS) is a rare, heritable disorder caused by variants in the SRCAP gene. Most individuals with FHS have characteristic facial features, short stature, and speech and language impairment. Although FHS has been likely under-diagnosed due to a combination of lack of recognition of the clinical phenotype and limited access to genomic testing, it is a rare condition with around 100 individuals reported in the medical literature. Case series have been biased towards younger individuals (vast majority <20 years of age) meaning that it has been challenging to provide accurate medical advice for affected individuals in adulthood. We report two young adults with FHS who presented with intracranial haemorrhage likely secondary to cerebrovascular aneurysms, with devastating consequences, making a total of four FHS patients reported with significant cerebrovascular abnormalities. Three of four patients had hypertension, at least one in conjunction with normal renal structure. We consider possible relationships between hypertension, renal pathology and aneurysms in the context of FHS, and consider mechanisms through which disruption of the SRCAP protein may lead to vascular pathology. We recommend that clinicians should have a low threshold to investigate symptoms suggestive of cerebrovascular disease in FHS. We advise that patients with FHS should have annual blood pressure monitoring from adolescence, renal ultrasound at diagnosis repeated in adulthood, and timely investigation of any neurological symptoms. For patients with FHS, particularly with hypertension, we advise that clinicians should consider at least one MRA (Magnetic Resonance Imaging with Angiography) to check for cerebral aneurysms.
Collapse
|
31
|
Ko J, Pomerantz JH, Perry H, Shieh JT, Slavotinek AM, Oberoi S, Klein OD. Case Report of Floating-Harbor Syndrome With Bilateral Cleft Lip. Cleft Palate Craniofac J 2019; 57:132-136. [PMID: 31248274 DOI: 10.1177/1055665619858257] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Floating-Harbor syndrome (FHS) is a rare genetic disorder caused by heterozygous mutations in the Snf2-related CREBBP activator protein (SRCAP) gene. The syndrome is characterized by proportional short stature, delayed bone maturation, delayed speech development, and facial dysmorphism. Submucous cleft palate and cleft lip have been reported in FHS, but to our knowledge orofacial clefting in this condition has not been assessed in detail. Here, we report on a case of bilateral cleft lip in a patient with FHS confirmed by exome sequencing.
Collapse
Affiliation(s)
- Jaemin Ko
- Program in Craniofacial Biology and Division of Craniofacial Anomalies, Department of Orofacial Sciences, University of California, San Francisco, CA, USA
| | - Jason H Pomerantz
- Program in Craniofacial Biology and Division of Craniofacial Anomalies, Department of Orofacial Sciences, University of California, San Francisco, CA, USA.,Division of Plastic and Reconstructive Surgery, Department of Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Hazel Perry
- Program in Craniofacial Biology and Division of Craniofacial Anomalies, Department of Orofacial Sciences, University of California, San Francisco, CA, USA
| | - Joseph T Shieh
- Division of Medical Genetics, Department of Pediatrics, and Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA
| | - Anne M Slavotinek
- Division of Medical Genetics, Department of Pediatrics, and Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA
| | - Snehlata Oberoi
- Program in Craniofacial Biology and Division of Craniofacial Anomalies, Department of Orofacial Sciences, University of California, San Francisco, CA, USA
| | - Ophir D Klein
- Program in Craniofacial Biology and Division of Craniofacial Anomalies, Department of Orofacial Sciences, University of California, San Francisco, CA, USA.,Division of Medical Genetics, Department of Pediatrics, and Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
32
|
Zhang S, Chen S, Qin H, Yuan H, Pi Y, Yang Y, Huang H, Li G, Sun Y, Wang Z, Ma H, Fu X, Zhou T, Wang J, Zhang H, Shen Y. Novel genotypes and phenotypes among Chinese patients with Floating-Harbor syndrome. Orphanet J Rare Dis 2019; 14:144. [PMID: 31200758 PMCID: PMC6570847 DOI: 10.1186/s13023-019-1111-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 06/04/2019] [Indexed: 12/11/2022] Open
Abstract
Background Floating-Harbor syndrome (FHS) is a rare syndromic short stature disorder caused by truncating variants in SRCAP. Few Chinese FHS patients had been reported so far and limited knowledge regarding the benefit of growth hormone treatment existed. Methods We ascertained 12 short stature patients with molecularly confirmed diagnosis of FHS by whole exome sequencing. We performed a comprehensive clinical evaluation for all patients and assessed the responsiveness of growth hormone treatment in a subset of the patients. Results Five distinct pathogenic/likely pathogenic variants were identified in 12 independent FHS patients including two previously reported variants (c.7303C > T/p.Arg2435Ter and c.7330C > T/p.Arg2444Ter) and three novel variants (c.7189G > T/p.Glu2397Ter, c.7245_7246delAT/p.Ser2416ArgfsTer26 and c.7466C > G/p.Ser2489Ter). The c.7303C > T/p.Arg2435Ter mutation appears more common in Chinese FHS patients. The clinical presentations of Chinese FHS patients are very similar to those of previously reported patients of different ethnicities. Yet we noticed micropenis and ear abnormalities in multiple patients, suggesting that these may be novel phenotypes of Floating-Harbor syndrome. Eight patients (one with GH deficiency, one with undetermined GH level, six without GH deficiency) underwent growth hormone treatment, 3 patients had good responses, one with modest and two with poor responses. Conclusion We described novel genotypes and phenotypes in a Chinese FHS patient cohort. We showed that about half of FHS patients exhibited modest to good response to GH treatment regardless of their respective GH deficiency status. We didn’t find any correlation between different mutations and response to GH treatment. Electronic supplementary material The online version of this article (10.1186/s13023-019-1111-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Shujie Zhang
- Department of Medical Genetics and Molecular Diagnostic Laboratory, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200000, People's Republic of China.,Department of Genetics and Metabolism, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530003, People's Republic of China
| | - Shaoke Chen
- Department of Genetics and Metabolism, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530003, People's Republic of China
| | - Haisong Qin
- Department of Genetics and Metabolism, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530003, People's Republic of China
| | - Haiming Yuan
- Dongguan Maternal and Child Health Care Hospital, Dongguan, 523120, People's Republic of China
| | - Yalei Pi
- Department of pediatrics, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, People's Republic of China
| | - Yu Yang
- Department of Endocrinology, Metabolism, and Genetics, Jiangxi Provincial Children's Hospital, Nanchang, 330006, People's Republic of China
| | - Hui Huang
- Department of Endocrinology, Metabolism, and Genetics, Jiangxi Provincial Children's Hospital, Nanchang, 330006, People's Republic of China
| | - Guimei Li
- Department of Pediatrics Endocrinology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021, People's Republic of China
| | - Yan Sun
- Department of Pediatrics Endocrinology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021, People's Republic of China
| | - Zhihua Wang
- Department of Endocrinology, Genetics and Metabolism, Xi'an Children's Hospital Affiliated with the School of Medicine, Xi'an Jiaotong University, Xi'an, 710000, People's Republic of China
| | - Huamei Ma
- Department of Pediatrics, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, People's Republic of China
| | - Xiaoling Fu
- Department of Pediatrics, The Peoples Hospital of The Guizhou Province, Guiyang, 550002, People's Republic of China
| | - Ting Zhou
- Department of Endocrinology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, People's Republic of China
| | - Jian Wang
- Department of Medical Genetics and Molecular Diagnostic Laboratory, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200000, People's Republic of China
| | - Huifeng Zhang
- Department of pediatrics, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, People's Republic of China.
| | - Yiping Shen
- Department of Medical Genetics and Molecular Diagnostic Laboratory, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200000, People's Republic of China. .,Department of Genetics and Metabolism, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530003, People's Republic of China. .,Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
33
|
Shields LBE, Peppas DS, Rosenberg E. Renal Calculus in Floating-Harbor Syndrome: A Case Report. J Pediatr Health Care 2019; 33:97-101. [PMID: 30205917 DOI: 10.1016/j.pedhc.2018.07.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 07/12/2018] [Accepted: 07/18/2018] [Indexed: 11/24/2022]
Abstract
Floating-Harbor syndrome is a rare condition marked by short stature and delayed bone age, characteristic facial features, and speech impairment. Floating-Harbor syndrome commonly results from a sporadic genetic mutation. Renal abnormalities have rarely been encountered. We report the first patient with Floating-Harbor syndrome who spontaneously passed a renal calculus consisting of calcium oxalate monohydrate and calcium oxalate dihydrate. A renal ultrasound showed echotexture within the renal pyramids, hydronephrosis, and a cyst. Pediatric nurse practitioners should be alert to the unique features associated with Floating-Harbor syndrome and be prepared to monitor and treat the renal abnormalities that may accompany this uncommon condition.
Collapse
|
34
|
Finken MJJ, van der Steen M, Smeets CCJ, Walenkamp MJE, de Bruin C, Hokken-Koelega ACS, Wit JM. Children Born Small for Gestational Age: Differential Diagnosis, Molecular Genetic Evaluation, and Implications. Endocr Rev 2018; 39:851-894. [PMID: 29982551 DOI: 10.1210/er.2018-00083] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Accepted: 06/21/2018] [Indexed: 12/25/2022]
Abstract
Children born small for gestational age (SGA), defined as a birth weight and/or length below -2 SD score (SDS), comprise a heterogeneous group. The causes of SGA are multifactorial and include maternal lifestyle and obstetric factors, placental dysfunction, and numerous fetal (epi)genetic abnormalities. Short-term consequences of SGA include increased risks of hypothermia, polycythemia, and hypoglycemia. Although most SGA infants show catch-up growth by 2 years of age, ∼10% remain short. Short children born SGA are amenable to GH treatment, which increases their adult height by on average 1.25 SD. Add-on treatment with a gonadotropin-releasing hormone agonist may be considered in early pubertal children with an expected adult height below -2.5 SDS. A small birth size increases the risk of later neurodevelopmental problems and cardiometabolic diseases. GH treatment does not pose an additional risk.
Collapse
Affiliation(s)
- Martijn J J Finken
- Department of Pediatrics, VU University Medical Center, MB Amsterdam, Netherlands
| | - Manouk van der Steen
- Department of Pediatrics, Erasmus University Medical Center/Sophia Children's Hospital, CN Rotterdam, Netherlands
| | - Carolina C J Smeets
- Department of Pediatrics, Erasmus University Medical Center/Sophia Children's Hospital, CN Rotterdam, Netherlands
| | - Marie J E Walenkamp
- Department of Pediatrics, VU University Medical Center, MB Amsterdam, Netherlands
| | - Christiaan de Bruin
- Department of Pediatrics, Leiden University Medical Center, RC Leiden, Netherlands
| | - Anita C S Hokken-Koelega
- Department of Pediatrics, Erasmus University Medical Center/Sophia Children's Hospital, CN Rotterdam, Netherlands
| | - Jan M Wit
- Department of Pediatrics, Leiden University Medical Center, RC Leiden, Netherlands
| |
Collapse
|
35
|
Floating-Harbor Syndrome: Presentation of the First Romanian Patient with a SRCAP Mutation and Review of the Literature. Balkan J Med Genet 2018; 21:83-86. [PMID: 30425916 PMCID: PMC6231312 DOI: 10.2478/bjmg-2018-0005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Floating-Harbor syndrome (FHS) is a rare autosomal dominant syndrome characterized by short stature with delayed bone age, retarded speech development, intellectual disability and dysmorphic facial features. Recently, dominant mutations almost exclusively clustered in the final exon of the Snf2-related CREBBP activator protein (SRCAP) gene were identified to cause FHS. Here, we report a boy with short stature, speech delay, mild intellectual disability, dysmorphic features, and with genetically confirmed FHS. To the best of our knowledge, this is the first molecularly confirmed case with this syndrome reported in Romania. An intensive program of cognitive and speech stimulation, as well as yearly neurological, psychological, ophthalmological, otorhinolaryngological, pediatric and endocrinological monitoring for our patient were designed. We propose a checklist of clinical features suggestive of FHS, based on the main clinical features, in order to facilitate the diagnosis and clinical management of this rare condition.
Collapse
|
36
|
Choi EM, Lee DH, Kang SJ, Shim YJ, Kim HS, Kim JS, Jeong JI, Ha JS, Jang JH. The first Korean case with Floating-Harbor syndrome with a novel SRCAP mutation diagnosed by targeted exome sequencing. KOREAN JOURNAL OF PEDIATRICS 2018; 61:403-406. [PMID: 30304910 PMCID: PMC6313083 DOI: 10.3345/kjp.2018.06289] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Accepted: 07/31/2018] [Indexed: 12/02/2022]
Abstract
Floating-Harbor syndrome is a rare autosomal dominant genetic disorder associated with SRCAP mutation. To date, approximately 50 cases of Floating-Harbor syndrome have been reported, but none have been reported in Korea yet. Floating-Harbor syndrome is characterized by delayed bony maturation, unique facial features, and language impairment. Here, we present a 6-year-old boy with a triangular face, deep-set protruding eyes, low-set ears, wide nose with narrow nasal bridge, short philtrum, long thin lips, clinodactyly, and developmental delay that was transferred to our pediatric clinic for genetic evaluation. He showed progressive delay in the area of language and cognition-adaption as he grew. He had previously undergone chromosomal analysis at another hospital due to his language delay, but his karyotype was normal. We performed targeted exome sequencing, considering several syndromes with similar phenotypes. Library preparation was performed with the TruSight One sequencing panel, which enriches the sample for about 4,800 genes of clinical relevance. Massively parallel sequencing was conducted with NextSeq. An identified variant was confirmed by Sanger sequencing of the patient and his parents. Finally, the patient was confirmed as the first Korean case of Floating-Harbor syndrome with a novel SRCAP (Snf2 related CREBBP activator protein) mutation (c.7732dupT, p.Ser2578Phefs*6), resulting in early termination of the protein; it was not found in either of his healthy parents or a control population. To our knowledge, this is the first study to describe a boy with Floating-Harbor syndrome with a novel SRCAP mutation diagnosed by targeted exome sequencing in Korea.
Collapse
Affiliation(s)
- Eun Mi Choi
- Department of Pediatrics, Keimyung University Dongsan Medical Center, Keimyung University School of Medicine, Daegu, Korea
| | - Dong Hyun Lee
- Department of Pediatrics, Keimyung University Dongsan Medical Center, Keimyung University School of Medicine, Daegu, Korea
| | - Seok Jin Kang
- Department of Pediatrics, Keimyung University Dongsan Medical Center, Keimyung University School of Medicine, Daegu, Korea
| | - Ye Jee Shim
- Department of Pediatrics, Keimyung University Dongsan Medical Center, Keimyung University School of Medicine, Daegu, Korea
| | - Heung Sik Kim
- Department of Pediatrics, Keimyung University Dongsan Medical Center, Keimyung University School of Medicine, Daegu, Korea
| | - Jun Sik Kim
- Department of Pediatrics, Keimyung University Dongsan Medical Center, Keimyung University School of Medicine, Daegu, Korea
| | - Jong In Jeong
- Department of Otorhinolaryngology, Keimyung University Dongsan Medical Center, Keimyung University School of Medicine, Daegu, Korea
| | - Jung-Sook Ha
- Department of Laboratory Medicine, Keimyung University Dongsan Medical Center, Keimyung University School of Medicine, Daegu, Korea
| | | |
Collapse
|
37
|
Moccia A, Martin DM. Nervous system development and disease: A focus on trithorax related proteins and chromatin remodelers. Mol Cell Neurosci 2018; 87:46-54. [PMID: 29196188 PMCID: PMC5828982 DOI: 10.1016/j.mcn.2017.11.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 11/08/2017] [Accepted: 11/27/2017] [Indexed: 01/12/2023] Open
Abstract
The nervous system comprises many different cell types including neurons, glia, macrophages, and immune cells, each of which is defined by specific patterns of gene expression, morphology, function, and anatomical location. Establishment of these complex and highly regulated cell fates requires spatial and temporal coordination of gene transcription. Open chromatin (euchromatin) allows transcription factors to interact with gene promoters and activate lineage specific genes, whereas closed chromatin (heterochromatin) remains inaccessible to transcriptional activation. Changes in the genome-wide distribution of euchromatin accompany transcriptional plasticity that allows the diversity of mature cell fates to be generated during development. In the past 20years, many new genes and gene families have been identified to participate in regulation of chromatin accessibility. These genes include chromatin remodelers that interact with Trithorax group (TrxG) and Polycomb group (PcG) proteins to activate or repress transcription, respectively. Here we review the role of TrxG proteins in neurodevelopment and disease.
Collapse
Affiliation(s)
- Amanda Moccia
- Department of Human Genetics, The University of Michigan Medical School, Ann Arbor, MI 48109, United States
| | - Donna M Martin
- Department of Human Genetics, The University of Michigan Medical School, Ann Arbor, MI 48109, United States; Department of Pediatrics and Communicable Diseases, The University of Michigan Medical School, Ann Arbor, MI 48109, United States.
| |
Collapse
|
38
|
Milani D, Scuvera G, Gatti M, Tolva G, Bonarrigo F, Esposito S, Gervasini C. Perthes disease: A new finding in Floating-Harbor syndrome. Am J Med Genet A 2018; 176:703-706. [PMID: 29383823 DOI: 10.1002/ajmg.a.38605] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 10/26/2017] [Accepted: 12/16/2017] [Indexed: 11/11/2022]
Abstract
Floating-Harbor Syndrome (FHS; OMIM #136140) is an ultra-rare autosomal dominant genetic condition characterized by expressive language delay, short stature with delayed bone mineralization, a triangular face with a prominent nose, and deep-set eyes, and hand anomalies. First reported in 1973, FHS is associated with mutations in the SRCAP gene, which encodes SNF2-related CREBBP activator protein. Mutations in the CREBBP gene cause Rubinstein-Taybi Syndrome (RSTS; OMIM #180849, #613684), another rare disease characterized by broad thumbs and halluces, facial dysmorphisms, short stature, and intellectual disability, which has a phenotypic overlap with FHS. We describe a case of FHS associated with a novel SRCAP mutation and characterized by Perthes disease, a skeletal anomaly described in approximately 3% of patients with RSTS. Thus Perthes disease can be added to the list of clinical features that overlap between FHS and RSTS.
Collapse
Affiliation(s)
- Donatella Milani
- Pediatric Highly Intensive Care Unit, Department of Pathophysiology and Transplantation University of Milan, Fondazione IRCSS Ca' Granda Ospedale Maggiorte Policlinico, Milan, Italy
| | - Giulietta Scuvera
- Pediatric Highly Intensive Care Unit, Department of Pathophysiology and Transplantation University of Milan, Fondazione IRCSS Ca' Granda Ospedale Maggiorte Policlinico, Milan, Italy
| | - Marta Gatti
- Pediatric Highly Intensive Care Unit, Department of Pathophysiology and Transplantation University of Milan, Fondazione IRCSS Ca' Granda Ospedale Maggiorte Policlinico, Milan, Italy
| | - Gianluca Tolva
- Pediatric Highly Intensive Care Unit, Department of Pathophysiology and Transplantation University of Milan, Fondazione IRCSS Ca' Granda Ospedale Maggiorte Policlinico, Milan, Italy
| | - Francesca Bonarrigo
- Pediatric Highly Intensive Care Unit, Department of Pathophysiology and Transplantation University of Milan, Fondazione IRCSS Ca' Granda Ospedale Maggiorte Policlinico, Milan, Italy
| | - Susanna Esposito
- Pediatric Clinic, Università degli Studi di Perugia, Perugia, Italy
| | - Cristina Gervasini
- Genetica Medica, Dip. Scienze della Salute, Università degli Studi di Milano, Milano, Italy
| |
Collapse
|
39
|
Vardarajan BN, Tosto G, Lefort R, Yu L, Bennett DA, De Jager PL, Barral S, Reyes-Dumeyer D, Nagy PL, Lee JH, Cheng R, Medrano M, Lantigua R, Rogaeva E, St George-Hyslop P, Mayeux R. Ultra-rare mutations in SRCAP segregate in Caribbean Hispanic families with Alzheimer disease. Neurol Genet 2017; 3:e178. [PMID: 28852706 PMCID: PMC5570674 DOI: 10.1212/nxg.0000000000000178] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 06/29/2017] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To identify rare coding variants segregating with late-onset Alzheimer disease (LOAD) in Caribbean Hispanic families. METHODS Whole-exome sequencing (WES) was completed in 110 individuals from 31 Caribbean Hispanic families without APOE ε4 homozygous carriers. Rare coding mutations segregating in families were subsequently genotyped in additional families and in an independent cohort of Caribbean Hispanic patients and controls. SRCAP messenger RNA (mRNA) expression was assessed in whole blood from mutation carriers with LOAD, noncarriers with LOAD, and healthy elderly controls, and also from autopsied brains in 2 clinical neuropathologic cohort studies of aging and dementia. RESULTS Ten ultra-rare missense mutations in the Snf2-related CREBBP, activator protein (SRCAP), were found in 12 unrelated families. Compared with the frequency in Caribbean Hispanic controls and the Latino population in the Exome Aggregation Consortium, the frequency of SRCAP mutations among Caribbean Hispanic patients with LOAD was significantly enriched (p = 1.19e-16). mRNA expression of SRCAP in whole blood was significantly lower in mutation carriers with LOAD, while the expression in whole blood and in the brain was significantly higher in nonmutation carriers with LOAD. Brain expression also correlated with clinical and neuropathologic endophenotypes. CONCLUSIONS WES in Caribbean Hispanic families with LOAD revealed ultra-rare missense mutations in SRCAP, a gene expressed in the brain and mutated in Floating-Harbor syndrome. SRCAP is a potent coactivator of the CREB-binding protein and a regulator of DNA damage response involving ATP-dependent chromatin remodeling. We hypothesize that increased expression in LOAD suggests a compensatory mechanism altered in mutation carriers.
Collapse
Affiliation(s)
- Badri N Vardarajan
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain (B.N.V., G.T., R. Lefort, P.L.D.J., S.B., D.R.-D., J.H.L., R.C., R. Lantigua, R.M.); Gertrude H. Sergievsky Center (B.N.V., G.T., S.B., D.R.-D., J.H.L., R.C., R.M.); Department of Neurology (P.L.D.J., S.B., R.M.), Department of Psychiatry (R.M.), Department of Systems Biology (B.N.V.), Department of Medicine (R. Lantigua), and Department of Pathology and Cell Biology (R. Lefort, P.L.N.), College of Physicians and Surgeons, Columbia University, New York Presbyterian Hospital; Department of Epidemiology (J.H.L., R.M.), School of Public Health, Columbia University, New York; Tanz Centre for Research in Neurodegenerative Diseases (E.R., P.S.G.-H.) and Department of Medicine (E.R., P.S.G.-H.), University of Toronto, Krembil Discovery Tower, ON, Canada; Department of Clinical Neurosciences (P.S.G.-H.), Cambridge Institute for Medical Research, University of Cambridge, UK; Rush Alzheimer's Disease Center (L.Y., D.A.B.), Rush University Medical Center, Chicago, IL; Program in Medical and Population Genetics (P.L.D.J.), Broad Institute, Cambridge, MA; and School of Medicine (M.M.), Mother and Teacher Pontifical Catholic University, Santiago, Dominican Republic
| | - Giuseppe Tosto
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain (B.N.V., G.T., R. Lefort, P.L.D.J., S.B., D.R.-D., J.H.L., R.C., R. Lantigua, R.M.); Gertrude H. Sergievsky Center (B.N.V., G.T., S.B., D.R.-D., J.H.L., R.C., R.M.); Department of Neurology (P.L.D.J., S.B., R.M.), Department of Psychiatry (R.M.), Department of Systems Biology (B.N.V.), Department of Medicine (R. Lantigua), and Department of Pathology and Cell Biology (R. Lefort, P.L.N.), College of Physicians and Surgeons, Columbia University, New York Presbyterian Hospital; Department of Epidemiology (J.H.L., R.M.), School of Public Health, Columbia University, New York; Tanz Centre for Research in Neurodegenerative Diseases (E.R., P.S.G.-H.) and Department of Medicine (E.R., P.S.G.-H.), University of Toronto, Krembil Discovery Tower, ON, Canada; Department of Clinical Neurosciences (P.S.G.-H.), Cambridge Institute for Medical Research, University of Cambridge, UK; Rush Alzheimer's Disease Center (L.Y., D.A.B.), Rush University Medical Center, Chicago, IL; Program in Medical and Population Genetics (P.L.D.J.), Broad Institute, Cambridge, MA; and School of Medicine (M.M.), Mother and Teacher Pontifical Catholic University, Santiago, Dominican Republic
| | - Roger Lefort
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain (B.N.V., G.T., R. Lefort, P.L.D.J., S.B., D.R.-D., J.H.L., R.C., R. Lantigua, R.M.); Gertrude H. Sergievsky Center (B.N.V., G.T., S.B., D.R.-D., J.H.L., R.C., R.M.); Department of Neurology (P.L.D.J., S.B., R.M.), Department of Psychiatry (R.M.), Department of Systems Biology (B.N.V.), Department of Medicine (R. Lantigua), and Department of Pathology and Cell Biology (R. Lefort, P.L.N.), College of Physicians and Surgeons, Columbia University, New York Presbyterian Hospital; Department of Epidemiology (J.H.L., R.M.), School of Public Health, Columbia University, New York; Tanz Centre for Research in Neurodegenerative Diseases (E.R., P.S.G.-H.) and Department of Medicine (E.R., P.S.G.-H.), University of Toronto, Krembil Discovery Tower, ON, Canada; Department of Clinical Neurosciences (P.S.G.-H.), Cambridge Institute for Medical Research, University of Cambridge, UK; Rush Alzheimer's Disease Center (L.Y., D.A.B.), Rush University Medical Center, Chicago, IL; Program in Medical and Population Genetics (P.L.D.J.), Broad Institute, Cambridge, MA; and School of Medicine (M.M.), Mother and Teacher Pontifical Catholic University, Santiago, Dominican Republic
| | - Lei Yu
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain (B.N.V., G.T., R. Lefort, P.L.D.J., S.B., D.R.-D., J.H.L., R.C., R. Lantigua, R.M.); Gertrude H. Sergievsky Center (B.N.V., G.T., S.B., D.R.-D., J.H.L., R.C., R.M.); Department of Neurology (P.L.D.J., S.B., R.M.), Department of Psychiatry (R.M.), Department of Systems Biology (B.N.V.), Department of Medicine (R. Lantigua), and Department of Pathology and Cell Biology (R. Lefort, P.L.N.), College of Physicians and Surgeons, Columbia University, New York Presbyterian Hospital; Department of Epidemiology (J.H.L., R.M.), School of Public Health, Columbia University, New York; Tanz Centre for Research in Neurodegenerative Diseases (E.R., P.S.G.-H.) and Department of Medicine (E.R., P.S.G.-H.), University of Toronto, Krembil Discovery Tower, ON, Canada; Department of Clinical Neurosciences (P.S.G.-H.), Cambridge Institute for Medical Research, University of Cambridge, UK; Rush Alzheimer's Disease Center (L.Y., D.A.B.), Rush University Medical Center, Chicago, IL; Program in Medical and Population Genetics (P.L.D.J.), Broad Institute, Cambridge, MA; and School of Medicine (M.M.), Mother and Teacher Pontifical Catholic University, Santiago, Dominican Republic
| | - David A Bennett
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain (B.N.V., G.T., R. Lefort, P.L.D.J., S.B., D.R.-D., J.H.L., R.C., R. Lantigua, R.M.); Gertrude H. Sergievsky Center (B.N.V., G.T., S.B., D.R.-D., J.H.L., R.C., R.M.); Department of Neurology (P.L.D.J., S.B., R.M.), Department of Psychiatry (R.M.), Department of Systems Biology (B.N.V.), Department of Medicine (R. Lantigua), and Department of Pathology and Cell Biology (R. Lefort, P.L.N.), College of Physicians and Surgeons, Columbia University, New York Presbyterian Hospital; Department of Epidemiology (J.H.L., R.M.), School of Public Health, Columbia University, New York; Tanz Centre for Research in Neurodegenerative Diseases (E.R., P.S.G.-H.) and Department of Medicine (E.R., P.S.G.-H.), University of Toronto, Krembil Discovery Tower, ON, Canada; Department of Clinical Neurosciences (P.S.G.-H.), Cambridge Institute for Medical Research, University of Cambridge, UK; Rush Alzheimer's Disease Center (L.Y., D.A.B.), Rush University Medical Center, Chicago, IL; Program in Medical and Population Genetics (P.L.D.J.), Broad Institute, Cambridge, MA; and School of Medicine (M.M.), Mother and Teacher Pontifical Catholic University, Santiago, Dominican Republic
| | - Philip L De Jager
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain (B.N.V., G.T., R. Lefort, P.L.D.J., S.B., D.R.-D., J.H.L., R.C., R. Lantigua, R.M.); Gertrude H. Sergievsky Center (B.N.V., G.T., S.B., D.R.-D., J.H.L., R.C., R.M.); Department of Neurology (P.L.D.J., S.B., R.M.), Department of Psychiatry (R.M.), Department of Systems Biology (B.N.V.), Department of Medicine (R. Lantigua), and Department of Pathology and Cell Biology (R. Lefort, P.L.N.), College of Physicians and Surgeons, Columbia University, New York Presbyterian Hospital; Department of Epidemiology (J.H.L., R.M.), School of Public Health, Columbia University, New York; Tanz Centre for Research in Neurodegenerative Diseases (E.R., P.S.G.-H.) and Department of Medicine (E.R., P.S.G.-H.), University of Toronto, Krembil Discovery Tower, ON, Canada; Department of Clinical Neurosciences (P.S.G.-H.), Cambridge Institute for Medical Research, University of Cambridge, UK; Rush Alzheimer's Disease Center (L.Y., D.A.B.), Rush University Medical Center, Chicago, IL; Program in Medical and Population Genetics (P.L.D.J.), Broad Institute, Cambridge, MA; and School of Medicine (M.M.), Mother and Teacher Pontifical Catholic University, Santiago, Dominican Republic
| | - Sandra Barral
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain (B.N.V., G.T., R. Lefort, P.L.D.J., S.B., D.R.-D., J.H.L., R.C., R. Lantigua, R.M.); Gertrude H. Sergievsky Center (B.N.V., G.T., S.B., D.R.-D., J.H.L., R.C., R.M.); Department of Neurology (P.L.D.J., S.B., R.M.), Department of Psychiatry (R.M.), Department of Systems Biology (B.N.V.), Department of Medicine (R. Lantigua), and Department of Pathology and Cell Biology (R. Lefort, P.L.N.), College of Physicians and Surgeons, Columbia University, New York Presbyterian Hospital; Department of Epidemiology (J.H.L., R.M.), School of Public Health, Columbia University, New York; Tanz Centre for Research in Neurodegenerative Diseases (E.R., P.S.G.-H.) and Department of Medicine (E.R., P.S.G.-H.), University of Toronto, Krembil Discovery Tower, ON, Canada; Department of Clinical Neurosciences (P.S.G.-H.), Cambridge Institute for Medical Research, University of Cambridge, UK; Rush Alzheimer's Disease Center (L.Y., D.A.B.), Rush University Medical Center, Chicago, IL; Program in Medical and Population Genetics (P.L.D.J.), Broad Institute, Cambridge, MA; and School of Medicine (M.M.), Mother and Teacher Pontifical Catholic University, Santiago, Dominican Republic
| | - Dolly Reyes-Dumeyer
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain (B.N.V., G.T., R. Lefort, P.L.D.J., S.B., D.R.-D., J.H.L., R.C., R. Lantigua, R.M.); Gertrude H. Sergievsky Center (B.N.V., G.T., S.B., D.R.-D., J.H.L., R.C., R.M.); Department of Neurology (P.L.D.J., S.B., R.M.), Department of Psychiatry (R.M.), Department of Systems Biology (B.N.V.), Department of Medicine (R. Lantigua), and Department of Pathology and Cell Biology (R. Lefort, P.L.N.), College of Physicians and Surgeons, Columbia University, New York Presbyterian Hospital; Department of Epidemiology (J.H.L., R.M.), School of Public Health, Columbia University, New York; Tanz Centre for Research in Neurodegenerative Diseases (E.R., P.S.G.-H.) and Department of Medicine (E.R., P.S.G.-H.), University of Toronto, Krembil Discovery Tower, ON, Canada; Department of Clinical Neurosciences (P.S.G.-H.), Cambridge Institute for Medical Research, University of Cambridge, UK; Rush Alzheimer's Disease Center (L.Y., D.A.B.), Rush University Medical Center, Chicago, IL; Program in Medical and Population Genetics (P.L.D.J.), Broad Institute, Cambridge, MA; and School of Medicine (M.M.), Mother and Teacher Pontifical Catholic University, Santiago, Dominican Republic
| | - Peter L Nagy
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain (B.N.V., G.T., R. Lefort, P.L.D.J., S.B., D.R.-D., J.H.L., R.C., R. Lantigua, R.M.); Gertrude H. Sergievsky Center (B.N.V., G.T., S.B., D.R.-D., J.H.L., R.C., R.M.); Department of Neurology (P.L.D.J., S.B., R.M.), Department of Psychiatry (R.M.), Department of Systems Biology (B.N.V.), Department of Medicine (R. Lantigua), and Department of Pathology and Cell Biology (R. Lefort, P.L.N.), College of Physicians and Surgeons, Columbia University, New York Presbyterian Hospital; Department of Epidemiology (J.H.L., R.M.), School of Public Health, Columbia University, New York; Tanz Centre for Research in Neurodegenerative Diseases (E.R., P.S.G.-H.) and Department of Medicine (E.R., P.S.G.-H.), University of Toronto, Krembil Discovery Tower, ON, Canada; Department of Clinical Neurosciences (P.S.G.-H.), Cambridge Institute for Medical Research, University of Cambridge, UK; Rush Alzheimer's Disease Center (L.Y., D.A.B.), Rush University Medical Center, Chicago, IL; Program in Medical and Population Genetics (P.L.D.J.), Broad Institute, Cambridge, MA; and School of Medicine (M.M.), Mother and Teacher Pontifical Catholic University, Santiago, Dominican Republic
| | - Joseph H Lee
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain (B.N.V., G.T., R. Lefort, P.L.D.J., S.B., D.R.-D., J.H.L., R.C., R. Lantigua, R.M.); Gertrude H. Sergievsky Center (B.N.V., G.T., S.B., D.R.-D., J.H.L., R.C., R.M.); Department of Neurology (P.L.D.J., S.B., R.M.), Department of Psychiatry (R.M.), Department of Systems Biology (B.N.V.), Department of Medicine (R. Lantigua), and Department of Pathology and Cell Biology (R. Lefort, P.L.N.), College of Physicians and Surgeons, Columbia University, New York Presbyterian Hospital; Department of Epidemiology (J.H.L., R.M.), School of Public Health, Columbia University, New York; Tanz Centre for Research in Neurodegenerative Diseases (E.R., P.S.G.-H.) and Department of Medicine (E.R., P.S.G.-H.), University of Toronto, Krembil Discovery Tower, ON, Canada; Department of Clinical Neurosciences (P.S.G.-H.), Cambridge Institute for Medical Research, University of Cambridge, UK; Rush Alzheimer's Disease Center (L.Y., D.A.B.), Rush University Medical Center, Chicago, IL; Program in Medical and Population Genetics (P.L.D.J.), Broad Institute, Cambridge, MA; and School of Medicine (M.M.), Mother and Teacher Pontifical Catholic University, Santiago, Dominican Republic
| | - Rong Cheng
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain (B.N.V., G.T., R. Lefort, P.L.D.J., S.B., D.R.-D., J.H.L., R.C., R. Lantigua, R.M.); Gertrude H. Sergievsky Center (B.N.V., G.T., S.B., D.R.-D., J.H.L., R.C., R.M.); Department of Neurology (P.L.D.J., S.B., R.M.), Department of Psychiatry (R.M.), Department of Systems Biology (B.N.V.), Department of Medicine (R. Lantigua), and Department of Pathology and Cell Biology (R. Lefort, P.L.N.), College of Physicians and Surgeons, Columbia University, New York Presbyterian Hospital; Department of Epidemiology (J.H.L., R.M.), School of Public Health, Columbia University, New York; Tanz Centre for Research in Neurodegenerative Diseases (E.R., P.S.G.-H.) and Department of Medicine (E.R., P.S.G.-H.), University of Toronto, Krembil Discovery Tower, ON, Canada; Department of Clinical Neurosciences (P.S.G.-H.), Cambridge Institute for Medical Research, University of Cambridge, UK; Rush Alzheimer's Disease Center (L.Y., D.A.B.), Rush University Medical Center, Chicago, IL; Program in Medical and Population Genetics (P.L.D.J.), Broad Institute, Cambridge, MA; and School of Medicine (M.M.), Mother and Teacher Pontifical Catholic University, Santiago, Dominican Republic
| | - Martin Medrano
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain (B.N.V., G.T., R. Lefort, P.L.D.J., S.B., D.R.-D., J.H.L., R.C., R. Lantigua, R.M.); Gertrude H. Sergievsky Center (B.N.V., G.T., S.B., D.R.-D., J.H.L., R.C., R.M.); Department of Neurology (P.L.D.J., S.B., R.M.), Department of Psychiatry (R.M.), Department of Systems Biology (B.N.V.), Department of Medicine (R. Lantigua), and Department of Pathology and Cell Biology (R. Lefort, P.L.N.), College of Physicians and Surgeons, Columbia University, New York Presbyterian Hospital; Department of Epidemiology (J.H.L., R.M.), School of Public Health, Columbia University, New York; Tanz Centre for Research in Neurodegenerative Diseases (E.R., P.S.G.-H.) and Department of Medicine (E.R., P.S.G.-H.), University of Toronto, Krembil Discovery Tower, ON, Canada; Department of Clinical Neurosciences (P.S.G.-H.), Cambridge Institute for Medical Research, University of Cambridge, UK; Rush Alzheimer's Disease Center (L.Y., D.A.B.), Rush University Medical Center, Chicago, IL; Program in Medical and Population Genetics (P.L.D.J.), Broad Institute, Cambridge, MA; and School of Medicine (M.M.), Mother and Teacher Pontifical Catholic University, Santiago, Dominican Republic
| | - Rafael Lantigua
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain (B.N.V., G.T., R. Lefort, P.L.D.J., S.B., D.R.-D., J.H.L., R.C., R. Lantigua, R.M.); Gertrude H. Sergievsky Center (B.N.V., G.T., S.B., D.R.-D., J.H.L., R.C., R.M.); Department of Neurology (P.L.D.J., S.B., R.M.), Department of Psychiatry (R.M.), Department of Systems Biology (B.N.V.), Department of Medicine (R. Lantigua), and Department of Pathology and Cell Biology (R. Lefort, P.L.N.), College of Physicians and Surgeons, Columbia University, New York Presbyterian Hospital; Department of Epidemiology (J.H.L., R.M.), School of Public Health, Columbia University, New York; Tanz Centre for Research in Neurodegenerative Diseases (E.R., P.S.G.-H.) and Department of Medicine (E.R., P.S.G.-H.), University of Toronto, Krembil Discovery Tower, ON, Canada; Department of Clinical Neurosciences (P.S.G.-H.), Cambridge Institute for Medical Research, University of Cambridge, UK; Rush Alzheimer's Disease Center (L.Y., D.A.B.), Rush University Medical Center, Chicago, IL; Program in Medical and Population Genetics (P.L.D.J.), Broad Institute, Cambridge, MA; and School of Medicine (M.M.), Mother and Teacher Pontifical Catholic University, Santiago, Dominican Republic
| | - Ekaterina Rogaeva
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain (B.N.V., G.T., R. Lefort, P.L.D.J., S.B., D.R.-D., J.H.L., R.C., R. Lantigua, R.M.); Gertrude H. Sergievsky Center (B.N.V., G.T., S.B., D.R.-D., J.H.L., R.C., R.M.); Department of Neurology (P.L.D.J., S.B., R.M.), Department of Psychiatry (R.M.), Department of Systems Biology (B.N.V.), Department of Medicine (R. Lantigua), and Department of Pathology and Cell Biology (R. Lefort, P.L.N.), College of Physicians and Surgeons, Columbia University, New York Presbyterian Hospital; Department of Epidemiology (J.H.L., R.M.), School of Public Health, Columbia University, New York; Tanz Centre for Research in Neurodegenerative Diseases (E.R., P.S.G.-H.) and Department of Medicine (E.R., P.S.G.-H.), University of Toronto, Krembil Discovery Tower, ON, Canada; Department of Clinical Neurosciences (P.S.G.-H.), Cambridge Institute for Medical Research, University of Cambridge, UK; Rush Alzheimer's Disease Center (L.Y., D.A.B.), Rush University Medical Center, Chicago, IL; Program in Medical and Population Genetics (P.L.D.J.), Broad Institute, Cambridge, MA; and School of Medicine (M.M.), Mother and Teacher Pontifical Catholic University, Santiago, Dominican Republic
| | - Peter St George-Hyslop
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain (B.N.V., G.T., R. Lefort, P.L.D.J., S.B., D.R.-D., J.H.L., R.C., R. Lantigua, R.M.); Gertrude H. Sergievsky Center (B.N.V., G.T., S.B., D.R.-D., J.H.L., R.C., R.M.); Department of Neurology (P.L.D.J., S.B., R.M.), Department of Psychiatry (R.M.), Department of Systems Biology (B.N.V.), Department of Medicine (R. Lantigua), and Department of Pathology and Cell Biology (R. Lefort, P.L.N.), College of Physicians and Surgeons, Columbia University, New York Presbyterian Hospital; Department of Epidemiology (J.H.L., R.M.), School of Public Health, Columbia University, New York; Tanz Centre for Research in Neurodegenerative Diseases (E.R., P.S.G.-H.) and Department of Medicine (E.R., P.S.G.-H.), University of Toronto, Krembil Discovery Tower, ON, Canada; Department of Clinical Neurosciences (P.S.G.-H.), Cambridge Institute for Medical Research, University of Cambridge, UK; Rush Alzheimer's Disease Center (L.Y., D.A.B.), Rush University Medical Center, Chicago, IL; Program in Medical and Population Genetics (P.L.D.J.), Broad Institute, Cambridge, MA; and School of Medicine (M.M.), Mother and Teacher Pontifical Catholic University, Santiago, Dominican Republic
| | - Richard Mayeux
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain (B.N.V., G.T., R. Lefort, P.L.D.J., S.B., D.R.-D., J.H.L., R.C., R. Lantigua, R.M.); Gertrude H. Sergievsky Center (B.N.V., G.T., S.B., D.R.-D., J.H.L., R.C., R.M.); Department of Neurology (P.L.D.J., S.B., R.M.), Department of Psychiatry (R.M.), Department of Systems Biology (B.N.V.), Department of Medicine (R. Lantigua), and Department of Pathology and Cell Biology (R. Lefort, P.L.N.), College of Physicians and Surgeons, Columbia University, New York Presbyterian Hospital; Department of Epidemiology (J.H.L., R.M.), School of Public Health, Columbia University, New York; Tanz Centre for Research in Neurodegenerative Diseases (E.R., P.S.G.-H.) and Department of Medicine (E.R., P.S.G.-H.), University of Toronto, Krembil Discovery Tower, ON, Canada; Department of Clinical Neurosciences (P.S.G.-H.), Cambridge Institute for Medical Research, University of Cambridge, UK; Rush Alzheimer's Disease Center (L.Y., D.A.B.), Rush University Medical Center, Chicago, IL; Program in Medical and Population Genetics (P.L.D.J.), Broad Institute, Cambridge, MA; and School of Medicine (M.M.), Mother and Teacher Pontifical Catholic University, Santiago, Dominican Republic
| |
Collapse
|
40
|
WDR26 Haploinsufficiency Causes a Recognizable Syndrome of Intellectual Disability, Seizures, Abnormal Gait, and Distinctive Facial Features. Am J Hum Genet 2017; 101:139-148. [PMID: 28686853 DOI: 10.1016/j.ajhg.2017.06.002] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 05/30/2017] [Indexed: 11/21/2022] Open
Abstract
We report 15 individuals with de novo pathogenic variants in WDR26. Eleven of the individuals carry loss-of-function mutations, and four harbor missense substitutions. These 15 individuals comprise ten females and five males, and all have intellectual disability with delayed speech, a history of febrile and/or non-febrile seizures, and a wide-based, spastic, and/or stiff-legged gait. These subjects share a set of common facial features that include a prominent maxilla and upper lip that readily reveal the upper gingiva, widely spaced teeth, and a broad nasal tip. Together, these features comprise a recognizable facial phenotype. We compared these features with those of chromosome 1q41q42 microdeletion syndrome, which typically contains WDR26, and noted that clinical features are consistent between the two subsets, suggesting that haploinsufficiency of WDR26 contributes to the pathology of 1q41q42 microdeletion syndrome. Consistent with this, WDR26 loss-of-function single-nucleotide mutations identified in these subjects lead to nonsense-mediated decay with subsequent reduction of RNA expression and protein levels. We derived a structural model of WDR26 and note that missense variants identified in these individuals localize to highly conserved residues of this WD-40-repeat-containing protein. Given that WDR26 mutations have been identified in ∼1 in 2,000 of subjects in our clinical cohorts and that WDR26 might be poorly annotated in exome variant-interpretation pipelines, we would anticipate that this disorder could be more common than currently appreciated.
Collapse
|
41
|
Abstract
Short stature is a common and heterogeneous condition that is often genetic in etiology. For most children with genetic short stature, the specific molecular causes remain unknown; but with advances in exome/genome sequencing and bioinformatics approaches, new genetic causes of growth disorders have been identified, contributing to the understanding of the underlying molecular mechanisms of longitudinal bone growth and growth failure. Identifying new genetic causes of growth disorders has the potential to improve diagnosis, prognostic accuracy, and individualized management, and help avoid unnecessary testing for endocrine and other disorders.
Collapse
Affiliation(s)
- Youn Hee Jee
- Program in Developmental Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, CRC, Room 1-3330, 10 Center Drive MSC 1103, Bethesda, MD 20892-1103, USA.
| | - Anenisia C Andrade
- Division of Pediatric Endocrinology, Department of Women's and Children's Health, Karolinska Institutet, Karolinska University Hospital, Solnavägen 1, Solna 171 77, Sweden
| | - Jeffrey Baron
- Program in Developmental Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, CRC, Room 1-3330, 10 Center Drive MSC 1103, Bethesda, MD 20892-1103, USA
| | - Ola Nilsson
- Division of Pediatric Endocrinology, Department of Women's and Children's Health, Karolinska Institutet, Karolinska University Hospital, Solnavägen 1, Solna 171 77, Sweden; University Hospital, Örebro University, Södra Grev Rosengatan, Örebro 701 85, Sweden
| |
Collapse
|
42
|
Treatment of Moyamoya Disease and Unruptured Intracranial Aneurysm in Floating-Harbor Syndrome. World Neurosurg 2017; 104:1049.e1-1049.e6. [PMID: 28549641 DOI: 10.1016/j.wneu.2017.05.083] [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: 03/23/2017] [Revised: 05/12/2017] [Accepted: 05/13/2017] [Indexed: 11/23/2022]
Abstract
BACKGROUND This is the first report of the successful treatment of moyamoya disease and an unruptured intracranial aneurysm in a patient with Floating-Harbor syndrome. CASE DESCRIPTION A 35-year-old, phenotypically syndromic woman presented with signs and symptoms consistent with ischemic stroke. Magnetic resonance imaging and catheter angiography confirmed diagnosis of moyamoya and a 6-mm basilar apex artery aneurysm (BAA). She underwent right-sided craniotomy for direct and indirect revascularization by means of superficial temporal artery-middle cerebral artery bypass and encephalo-duro-arterio-synangiosis. Three months later, she underwent stent-assisted coiling of the BAA. At 9 months, the patient remains without her preoperative neurological deficits. Interval catheter angiography confirms revascularization of her right hemisphere and obliteration of her BAA. CONCLUSIONS We present the first case of diagnosis and treatment of moyamoya disease and BAA in a patient with Floating-Harbor syndrome. Due to a paucity of literature on this rare disorder, there has not been an associated link between Floating-Harbor syndrome and cerebrovascular disease. Our report and literature review suggest that these patients may be prone to cerebrovascular disorders and should be followed closely with neurovascular imaging.
Collapse
|
43
|
The defining DNA methylation signature of Floating-Harbor Syndrome. Sci Rep 2016; 6:38803. [PMID: 27934915 PMCID: PMC5146968 DOI: 10.1038/srep38803] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 11/15/2016] [Indexed: 11/08/2022] Open
Abstract
Floating-Harbor syndrome (FHS) is an autosomal dominant genetic condition characterized by short stature, delayed osseous maturation, expressive language impairment, and unique facial dysmorphology. We previously identified mutations in the chromatin remodeling protein SRCAP (SNF2-related CBP Activator Protein) as the cause of FHS. SRCAP has multiple roles in chromatin and transcriptional regulation; however, specific epigenetic consequences of SRCAP mutations remain to be described. Using high resolution genome-wide DNA methylation analysis, we identified a unique and highly specific DNA methylation "epi-signature" in the peripheral blood of individuals with FHS. Both hyper and hypomethylated loci are distributed across the genome, preferentially occurring in CpG islands. Clonal bisulfite sequencing of two hypermethylated (FIGN and STPG2) and two hypomethylated (MYO1F and RASIP1) genes confirmed these findings. The identification of a unique methylation signature in FHS provides further insight into the biological function of SRCAP and provides a unique biomarker for this disorder.
Collapse
|
44
|
Ganetzky RD, Bedoukian E, Deardorff MA, Ficicioglu C. Argininosuccinic Acid Lyase Deficiency Missed by Newborn Screen. JIMD Rep 2016; 34:43-47. [PMID: 27515243 DOI: 10.1007/8904_2016_2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Revised: 06/10/2016] [Accepted: 06/14/2016] [Indexed: 12/30/2022] Open
Abstract
Argininosuccinic acid lyase (ASL) deficiency, caused by mutations in the ASL gene (OMIM: 608310) is a urea cycle disorder that has pleiotropic presentations. On the mild end, ASL deficiency can manifest as nonspecific neurocognitive abnormalities without readily identifiable signs to differentiate it from other causes of intellectual disability or learning disabilities. Dietary management and arginine supplementation, if initiated early, may ameliorate symptoms.Because of the nonspecific nature of the symptoms and the possibility for therapeutic management, ASL deficiency is part of the recommended uniform screening panel for newborn screening in the USA. We report here a case of ASL deficiency that was missed on newborn screening in the USA.The case reported here has two known pathogenic mutations - one with no residual activity and one with reported 10% residual activity. Review of this newborn screening results showed subtle elevation of citrulline, overlapping the normal range. These findings suggest that newborn screening may be missing other patients with ASL deficiency with at least one hypomorphic allele. This case was diagnosed incidentally, but in retrospect had symptoms best attributed in full or in part to his ASA deficiency, including protein aversion, developmental delay, and seizures. This case highlights the importance of considering ASL deficiency in patients with nonspecific abnormal neurocognitive signs, such as epilepsy and developmental delay, even when newborn screening was normal.
Collapse
Affiliation(s)
- Rebecca D Ganetzky
- Department of Pediatrics, Division of Human Genetics, The Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, 3501 Civic Center Blvd, Philadelphia, PA, 19104, USA
| | - Emma Bedoukian
- Department of Pediatrics, Individualized Medical Genetics Center, The Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, 3501 Civic Center Blvd, Philadelphia, PA, 19104, USA
| | - Matthew A Deardorff
- Department of Pediatrics, Division of Human Genetics, The Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, 3501 Civic Center Blvd, Philadelphia, PA, 19104, USA
- Department of Pediatrics, Individualized Medical Genetics Center, The Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, 3501 Civic Center Blvd, Philadelphia, PA, 19104, USA
| | - Can Ficicioglu
- Department of Pediatrics, Division of Human Genetics, The Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, 3501 Civic Center Blvd, Philadelphia, PA, 19104, USA.
| |
Collapse
|
45
|
Amita M, Srivastava P, Agarwal D, Phadke SR. Floating Harbor Syndrome. Indian J Pediatr 2016; 83:896-7. [PMID: 27206688 DOI: 10.1007/s12098-016-2153-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 05/09/2016] [Indexed: 11/26/2022]
Affiliation(s)
- Moirangthem Amita
- Department of Medical Genetics, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, 226014, India
| | - Priyanka Srivastava
- Department of Medical Genetics, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, 226014, India
| | - Divya Agarwal
- Department of Medical Genetics, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, 226014, India
| | - Shubha R Phadke
- Department of Medical Genetics, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, 226014, India.
| |
Collapse
|
46
|
Messina G, Atterrato MT, Dimitri P. When chromatin organisation floats astray: theSrcapgene and Floating–Harbor syndrome. J Med Genet 2016; 53:793-797. [DOI: 10.1136/jmedgenet-2016-103842] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2016] [Revised: 02/29/2016] [Accepted: 03/29/2016] [Indexed: 01/19/2023]
|
47
|
Wit JM, Oostdijk W, Losekoot M, van Duyvenvoorde HA, Ruivenkamp CAL, Kant SG. MECHANISMS IN ENDOCRINOLOGY: Novel genetic causes of short stature. Eur J Endocrinol 2016; 174:R145-73. [PMID: 26578640 DOI: 10.1530/eje-15-0937] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 11/16/2015] [Indexed: 12/17/2022]
Abstract
The fast technological development, particularly single nucleotide polymorphism array, array-comparative genomic hybridization, and whole exome sequencing, has led to the discovery of many novel genetic causes of growth failure. In this review we discuss a selection of these, according to a diagnostic classification centred on the epiphyseal growth plate. We successively discuss disorders in hormone signalling, paracrine factors, matrix molecules, intracellular pathways, and fundamental cellular processes, followed by chromosomal aberrations including copy number variants (CNVs) and imprinting disorders associated with short stature. Many novel causes of GH deficiency (GHD) as part of combined pituitary hormone deficiency have been uncovered. The most frequent genetic causes of isolated GHD are GH1 and GHRHR defects, but several novel causes have recently been found, such as GHSR, RNPC3, and IFT172 mutations. Besides well-defined causes of GH insensitivity (GHR, STAT5B, IGFALS, IGF1 defects), disorders of NFκB signalling, STAT3 and IGF2 have recently been discovered. Heterozygous IGF1R defects are a relatively frequent cause of prenatal and postnatal growth retardation. TRHA mutations cause a syndromic form of short stature with elevated T3/T4 ratio. Disorders of signalling of various paracrine factors (FGFs, BMPs, WNTs, PTHrP/IHH, and CNP/NPR2) or genetic defects affecting cartilage extracellular matrix usually cause disproportionate short stature. Heterozygous NPR2 or SHOX defects may be found in ∼3% of short children, and also rasopathies (e.g., Noonan syndrome) can be found in children without clear syndromic appearance. Numerous other syndromes associated with short stature are caused by genetic defects in fundamental cellular processes, chromosomal abnormalities, CNVs, and imprinting disorders.
Collapse
Affiliation(s)
- Jan M Wit
- Departments of PaediatricsClinical GeneticsLeiden University Medical Center, PO Box 9600, 2300 RC Leiden, The Netherlands
| | - Wilma Oostdijk
- Departments of PaediatricsClinical GeneticsLeiden University Medical Center, PO Box 9600, 2300 RC Leiden, The Netherlands
| | - Monique Losekoot
- Departments of PaediatricsClinical GeneticsLeiden University Medical Center, PO Box 9600, 2300 RC Leiden, The Netherlands
| | - Hermine A van Duyvenvoorde
- Departments of PaediatricsClinical GeneticsLeiden University Medical Center, PO Box 9600, 2300 RC Leiden, The Netherlands
| | - Claudia A L Ruivenkamp
- Departments of PaediatricsClinical GeneticsLeiden University Medical Center, PO Box 9600, 2300 RC Leiden, The Netherlands
| | - Sarina G Kant
- Departments of PaediatricsClinical GeneticsLeiden University Medical Center, PO Box 9600, 2300 RC Leiden, The Netherlands
| |
Collapse
|
48
|
Abstract
The next-generation sequencing revolution has substantially increased our understanding of the mutated genes that underlie complex neurodevelopmental disease. Exome sequencing has enabled us to estimate the number of genes involved in the etiology of neurodevelopmental disease, whereas targeted sequencing approaches have provided the means for quick and cost-effective sequencing of thousands of patient samples to assess the significance of individual genes. By leveraging such technologies and clinical exome sequencing, a genotype-first approach has emerged in which patients with a common genotype are first identified and then clinically reassessed as a group. This approach has proven a powerful methodology for refining disease subtypes. We propose that the molecular characterization of these genetic subtypes has important implications for diagnostics and also for future drug development. Classifying patients into subgroups with a common genetic etiology and applying treatments tailored to the specific molecular defect they carry is likely to improve management of neurodevelopmental disease in the future.
Collapse
|
49
|
Basel-Vanagaite L, Wolf L, Orin M, Larizza L, Gervasini C, Krantz I, Deardoff M. Recognition of the Cornelia de Lange syndrome phenotype with facial dysmorphology novel analysis. Clin Genet 2016; 89:557-63. [DOI: 10.1111/cge.12716] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 12/07/2015] [Accepted: 12/09/2015] [Indexed: 11/29/2022]
Affiliation(s)
- L. Basel-Vanagaite
- Medical Genetics Department; Schneider Children's Medical Center of Israel, Rabin Medical Center; Petah Tikva Israel
- Felsenstein Medical Research Center; Petah Tikva Israel
- Tel Aviv University; Tel Aviv Israel
- FDNA Inc.; Boston, MA USA
| | - L. Wolf
- Tel Aviv University; Tel Aviv Israel
- FDNA Inc.; Boston, MA USA
| | | | - L. Larizza
- Laboratory of Medical Cytogenetics and Molecular Genetics; Istituto Auxologico Italiano; Milan Italy
- Department of Health Sciences, Medical Genetics; University of Milano; Milan Italy
| | - C. Gervasini
- Laboratory of Medical Cytogenetics and Molecular Genetics; Istituto Auxologico Italiano; Milan Italy
- Department of Health Sciences, Medical Genetics; University of Milano; Milan Italy
| | - I.D. Krantz
- Division of Human Molecular Genetics; The Children's Hospital of Philadelphia; Philadelphia PA USA
- The Perelman School of Medicine; University of Pennsylvania; Philadelphia PA USA
| | - M.A. Deardoff
- Division of Human Molecular Genetics; The Children's Hospital of Philadelphia; Philadelphia PA USA
- The Perelman School of Medicine; University of Pennsylvania; Philadelphia PA USA
| |
Collapse
|
50
|
Yagi H, Takagi M, Narumi S, Hasegawa T, Nishimura G, Hasegawa Y. Stippled calcification in an infant with a recurrent SRCAP gene mutation. Am J Med Genet A 2016; 170A:1088-91. [PMID: 26788936 DOI: 10.1002/ajmg.a.37516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 12/04/2015] [Indexed: 11/10/2022]
Affiliation(s)
- Hiroko Yagi
- Division of Genetic Research, Tokyo Metropolitan Children's Medical Center, Tokyo, Japan.,Division of Endocrinology and Metabolism, Tokyo Metropolitan Children's Medical Center, Tokyo, Japan
| | - Masaki Takagi
- Division of Genetic Research, Tokyo Metropolitan Children's Medical Center, Tokyo, Japan.,Division of Endocrinology and Metabolism, Tokyo Metropolitan Children's Medical Center, Tokyo, Japan
| | - Satoshi Narumi
- Department of Pediatrics, Keio University School of Medicine, Tokyo, Japan
| | - Tomonobu Hasegawa
- Department of Pediatrics, Keio University School of Medicine, Tokyo, Japan
| | - Gen Nishimura
- Division of Radiology, Tokyo Metropolitan Children's Medical Center, Tokyo, Japan
| | - Yukihiro Hasegawa
- Division of Genetic Research, Tokyo Metropolitan Children's Medical Center, Tokyo, Japan.,Division of Endocrinology and Metabolism, Tokyo Metropolitan Children's Medical Center, Tokyo, Japan
| |
Collapse
|