1
|
Currao P, Balzarini M, Pruna D, Marica M, Soddu C, Marras M, Pavanello M, Satta S, Savasta S. Vascular Abnormalities and Neurofibromatosis Type 1: A Paediatric Case Series. J Child Neurol 2025; 40:49-60. [PMID: 39380389 DOI: 10.1177/08830738241284081] [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] [Indexed: 10/10/2024]
Abstract
Neurofibromatosis type 1 (NF1) is a multisystemic neurocutaneous disease caused by a heterozygous mutation of the NF1 gene that encodes neurofibromin. Complications include vascular and neurologic abnormalities such as moyamoya syndrome, a cerebrovascular disorder with progressive occlusion of the large intracranial arteries, leading to ischemic events and the formation of abnormal vascular networks. Stenosis of the renal artery is another frequent complication of neurofibromatosis type 1, and it represents the most common cause of secondary hypertension in these patients. The purpose of the article is to describe the clinical manifestations of neurofibromatosis type 1 vasculopathy in 4 patients presenting with a wide range of neurologic and reno-vascular manifestations, as well as to examine current diagnostic management and follow-up, current therapeutic options, and to discuss further perspectives in terms of screening, diagnosis, and treatment.
Collapse
Affiliation(s)
- Paolo Currao
- Pediatric Clinic, "Microcitemico - A. Cao" Pediatric Hospital, University of Cagliari, Cagliari, Italy
| | - Marta Balzarini
- Pediatric Service, San Michele Hospital, ARNAS Brotzu Cagliari, Cagliari, Italy
| | - Dario Pruna
- Department of Pediatric Neurology, "Microcitemico - A. Cao" Pediatric Hospital, ASL Cagliari, Cagliari, Italy
| | - Monica Marica
- Pediatric Clinic, "Microcitemico - A. Cao" Pediatric Hospital, University of Cagliari, Cagliari, Italy
| | - Consolata Soddu
- Pediatric Clinic, "Microcitemico - A. Cao" Pediatric Hospital, University of Cagliari, Cagliari, Italy
| | - Mariangela Marras
- Department of Pediatric Radiology, "Microcitemico - A. Cao" Hospital, ASL Cagliari, Cagliari, Italy
| | - Marco Pavanello
- Department of Neurosurgery, IRCCS Istituto Giannina Gaslini, Via Gerolamo Gaslini, Genoa, Italy
| | - Stefania Satta
- Department of Medical Science and Public Health, University of Cagliari, Laboratory of Genetics and Genomics, "Microcitemico - A. Cao" Pediatric Hospital, ASL Cagliari, Cagliari, Italy
| | - Salvatore Savasta
- Pediatric Clinic, "Microcitemico - A. Cao" Pediatric Hospital, University of Cagliari, Cagliari, Italy
| |
Collapse
|
2
|
Boulter JH, Szuflita NS, Keating RF, Magge SN. Outcomes of surgical revascularization for pediatric moyamoya disease and syndrome. Childs Nerv Syst 2024; 40:2449-2456. [PMID: 38753003 PMCID: PMC11269374 DOI: 10.1007/s00381-024-06393-w] [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: 10/19/2023] [Accepted: 04/02/2024] [Indexed: 07/25/2024]
Abstract
PURPOSE Moyamoya disease and syndrome represent rare entities characterized by progressive stenosis and/or occlusion of the intracranial blood vessels. We present our series of patients with moyamoya disease and syndrome stratified by underlying disease and analyze differences in presentation and outcome following surgical revascularization. METHODS This was an Institutional Review Board (IRB) approved, retrospective review of all patients surgically revascularized by the senior author (SNM) while at Children's National Hospital in Washington, DC. Demographic data, presenting symptoms and severity, surgical details, and functional and radiographic outcomes were obtained and analyzed for differences among the underlying cohorts of moyamoya disease and syndrome as well as by unilateral or bilateral disease and index or non-index surgeries. RESULTS Twenty-two patients were identified with the following underlying diseases: six with idiopathic moyamoya disease, six with sickle cell anemia, five with trisomy 21, and five with neurofibromatosis type 1. Thirty hemispheres were revascularized with a significantly reduced rate of stroke from 3.19 strokes/year (SD = 3.10) to 0.13 strokes/year (SD = 0.25), p = 0.03. When analyzed by underlying cause of moyamoya syndrome, patients with neurofibromatosis type 1 were found to be significantly less likely than the other subtypes of moyamoya syndrome to have had either a clinical stroke (0.0% neurofibromatosis type 1 vs. 100.0% sickle cell, 60.0% trisomy 21, or 83.3% moyamoya disease, p < 0.01) or radiographic stroke (0.0% neurofibromatosis type 1 vs. 100.0% sickle cell, 60.0% trisomy 21, or 83.3% moyamoya disease, p < 0.01) at time of presentation. Patients with moyamoya syndrome associated with sickle cell disease were more likely to present with clinical and radiographic strokes. Additionally, patients with bilateral disease demonstrated no difference in final functional outcome compared to patients with unilateral disease (mRS 0.73 (SD = 1.33) vs. 1.29 (SD = 1.60), p = 0.63). CONCLUSION Indirect surgical revascularization decreases stroke risk for pediatric patients with different forms of moyamoya disease and moyamoya syndrome. Additionally, these data suggest that sickle cell anemia-associated moyamoya syndrome may represent a more aggressive variant, while neurofibromatosis type 1 may represent a more benign variant.
Collapse
Affiliation(s)
- Jason H Boulter
- Division of Neurosurgery, Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - Nicholas S Szuflita
- Division of Neurosurgery, Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - Robert F Keating
- Division of Neurosurgery, Children's National Hospital, Washington, DC, USA
| | - Suresh N Magge
- Division of Neurosurgery, Children's National Hospital, Washington, DC, USA.
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
3
|
Savad S, Modarressi MH, Younesi S, Seifi-Alan M, Samadaian N, Masoomy M, Dianatpour M, Norouzi S, Amidi S, Boroumand A, Ashrafi MR, Ronagh A, Eslami M, Hashemnejad M, Nourian S, Mohammadi S, Taheri Amin MM, Heidari M, Seifi-Alan M, Shojaaldini Ardakani H, Aghamahdi F, Khalilian S, Ghafouri-Fard S. A Comprehensive Overview of NF1 Mutations in Iranian Patients. Neuromolecular Med 2024; 26:28. [PMID: 38954284 DOI: 10.1007/s12017-024-08790-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 04/30/2024] [Indexed: 07/04/2024]
Abstract
Neurofibromatosis type 1 (NF1) is a genetic disorder caused by mutations in the NF1 gene. This disorder shows nearly complete penetrance and high phenotypic variability. We used the whole-exome sequencing technique to identify mutations in 32 NF1 cases from 22 Iranian families. A total of 31 variants, including 30 point mutations and one large deletion, were detected. In eight cases, variants were inherited, while they were sporadic in the remaining. Seven novel variants, including c.5576 T > G, c.6658_6659insC, c.2322dupT, c.92_93insAA, c.4360C > T, c.3814C > T, and c.4565_4566delinsC, were identified. The current study is the largest in terms of the sample size of Iranian NF1 cases with identified mutations. The results can broaden the spectrum of NF1 mutations and facilitate the process of genetic counseling in the affected families.
Collapse
Affiliation(s)
| | | | - Sarang Younesi
- Prenatal Screening Department, Nilou Laboratory, Tehran, Iran
| | - Mahnaz Seifi-Alan
- Cardiovascular Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | | | - Mona Masoomy
- Applied Biotechnology Research Center, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mehdi Dianatpour
- Stem Cell and Transgenic Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Human Genetic, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | | | | | - Mahmoud Reza Ashrafi
- Pediatric Neurology Division, Children's Medical Center, Pediatrics Center of Excellence, Ataxia Clinic, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Ronagh
- Department of Pediatrics Neurologists, Shahid Bahonar Hospital, Alborz University of Medical Sciences, Karaj, Iran
| | - Maryam Eslami
- Applied Biotechnology Research Center, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Department of Genetics, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Maryam Hashemnejad
- Department of Obstetrics and Gynecology, School of Medicine, Kamali Hospital, Alborz University of Medical Sciences, Karaj, Iran
| | - Shahab Nourian
- Department of Pediatrics Endocrinology and Metabolisms, Emam Ali Hospital, Alborz University of Medical Sciences and Health Services, Karaj, Iran
| | - Sanaz Mohammadi
- Comprehensive Medical Genetics Center, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Morteza Heidari
- Pediatric Neurology Division, Children's Medical Center, Pediatrics Center of Excellence, Ataxia Clinic, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahin Seifi-Alan
- Cardiovascular Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | | | - Fatemeh Aghamahdi
- Department of Pediatrics, Alborz University of Medical Sciences, Karaj, Iran
| | - Sheyda Khalilian
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
4
|
Lehman LL, Ullrich NJ. Cerebral Vasculopathy in Children with Neurofibromatosis Type 1. Cancers (Basel) 2023; 15:5111. [PMID: 37894478 PMCID: PMC10605225 DOI: 10.3390/cancers15205111] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 10/09/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
Cerebrovascular abnormalities are a severe and often underrecognized complication of childhood neurofibromatosis type 1 (NF1). There are no prospective studies of cerebral vasculopathy in NF1; thus, the estimated frequency of vasculopathy varies between studies. The data is difficult to interpret due to the retrospective data collection and variability in whether imaging is done based on screening/surveillance or due to acute neurologic symptoms. The prevalent NF1-associated cerebral vasculopathy is moyamoya syndrome (MMS). Vascular changes can present without symptoms or with acute TIA or stroke-like symptoms or a range of progressive neurologic deficits. Advanced imaging may enhance sensitivity of neuroimaging in children. Medical and/or surgical interventions may prevent short- and long-term complications. Challenges for establishment of a screening protocol for cerebral vasculopathy in children with NF1 include the relatively large number of patients with NF1, the potential need for sedation to achieve quality imaging and the broad age range at time of detection for cerebral vascular changes. The goal of this review is to present the epidemiology, clinical presentation, imaging features and medical/surgical management of cerebral arteriopathies in children with NF1.
Collapse
Affiliation(s)
- Laura L Lehman
- Department of Neurology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Nicole J Ullrich
- Department of Neurology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
5
|
Carotenuto M, Messina G, Esposito M, Santoro C, Iacono D, Spruyt K. Polysomnographic study in pediatric neurofibromatosis type 1. Front Neurol 2023; 14:1213430. [PMID: 37538252 PMCID: PMC10394094 DOI: 10.3389/fneur.2023.1213430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 06/29/2023] [Indexed: 08/05/2023] Open
Abstract
Background Neurofibromatosis type 1 (NF1) is a genetic disease that alters neurodevelopment. We aimed to analyze the sleep macrostructure of a sample of children affected by NF1 without neurocognitive co-morbidities and MRI reports of unidentified bright objects (UBOs). Methods A 100 pre-pubertal children participated in the cross-sectional study: 50 subjects were children diagnosed with NF1 and 50 subjects were typically developing healthy children (TDC). All participants underwent polysomnographic evaluation through which conventional sleep parameters were collected: Total sleep time (TST), Sleep latency (SOL), first REM latency (FRL), number of stage shifts/h (SS/h), number of awakenings/h (AWN/h), wake after sleep onset (WASO%), sleep efficiency percentage (SE%), percentage of sleep time spent in sleep stages 1 (N1%) and 2 (N2%), slow-wave sleep (N3%), and REM sleep (REM%). Additionally, nocturnal respiratory events such as apnea/hypopnea index (AHI), oxygen desaturation index (ODI), and periodic limb movement index (PLMI) were recorded. Results Neurofibromatosis type 1 children showed a reduction in sleep duration parameters (TST; p < 0.001), sleep efficiency (SE%; p < 0.001), and stage N2% (p < 0.001). Moreover, the number of awakenings per hour (AWN/h), wake after sleep onset (WASO%), and respiratory events such as AHI, ODI, and PLMI resulted higher in NF1 vs. TDC children. Conclusion The data showed that the sleep macrostructure differs between NF1 and TDC children. These findings suggest that the evaluation of sleep may provide useful support in corroborating the diagnosis and offers additional therapeutic management perspectives in NF1 and genetic neurodevelopmental disorders in general.
Collapse
Affiliation(s)
- Marco Carotenuto
- Sleep Lab for Developmental Age, Clinic of Child and Adolescent Neuropsychiatry, Department of Mental and Physical Health and Preventive Medicine, Child and Adolescent Neuropsychiatry Clinic, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Giovanni Messina
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Maria Esposito
- Sleep Lab for Developmental Age, Clinic of Child and Adolescent Neuropsychiatry, Department of Mental and Physical Health and Preventive Medicine, Child and Adolescent Neuropsychiatry Clinic, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Claudia Santoro
- Sleep Lab for Developmental Age, Clinic of Child and Adolescent Neuropsychiatry, Department of Mental and Physical Health and Preventive Medicine, Child and Adolescent Neuropsychiatry Clinic, University of Campania "Luigi Vanvitelli", Naples, Italy
- Department of Women's and Children's Health, and General and Specialized Surgery, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Diego Iacono
- Neuropathology Research, Biomedical Research Institute of New Jersey, BRInj, Cedar Knolls, NJ, United States
- Department of Pediatrics, Neuropathology Research, Mid-Atlantic Neonatology Associates (MANA), Atlantic Health System (AHS), Morristown, NJ, United States
| | - Karen Spruyt
- NeuroDiderot INSERM, Université de Paris, Paris, France
| |
Collapse
|
6
|
Ognibene M, Scala M, Iacomino M, Schiavetti I, Madia F, Traverso M, Guerrisi S, Di Duca M, Caroli F, Baldassari S, Tappino B, Romano F, Uva P, Vozzi D, Chelleri C, Piatelli G, Diana MC, Zara F, Capra V, Pavanello M, De Marco P. Moyamoya Vasculopathy in Neurofibromatosis Type 1 Pediatric Patients: The Role of Rare Variants of RNF213. Cancers (Basel) 2023; 15:cancers15061916. [PMID: 36980803 PMCID: PMC10047491 DOI: 10.3390/cancers15061916] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/15/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023] Open
Abstract
Neurofibromatosis type 1 (NF1) is a neurocutaneous disorder caused by mutations in NF1 gene, coding for neurofibromin 1. NF1 can be associated with Moyamoya disease (MMD), and this association, typical of paediatric patients, is referred to as Moyamoya syndrome (MMS). MMD is a cerebral arteriopathy characterized by the occlusion of intracranial arteries and collateral vessel formation, which increase the risk of ischemic and hemorrhagic events. RNF213 gene mutations have been associated with MMD, so we investigated whether rare variants of RNF213 could act as genetic modifiers of MMS phenotype in a pediatric cohort of 20 MMS children, 25 children affected by isolated MMD and 47 affected only by isolated NF1. By next-generation re-sequencing (NGS) of patients' DNA and gene burden tests, we found that RNF213 seems to play a role only for MMD occurrence, while it does not appear to be involved in the increased risk of Moyamoya for MMS patients. We postulated that the loss of neurofibromin 1 can be enough for the excessive proliferation of vascular smooth muscle cells, causing Moyamoya arteriopathy associated with NF1. Further studies will be crucial to support these findings and to elucidate the possible role of other genes, enhancing our knowledge about pathogenesis and treatment of MMS.
Collapse
Affiliation(s)
- Marzia Ognibene
- U.O.C. Genetica Medica, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy
| | - Marcello Scala
- U.O.C. Genetica Medica, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, Università Degli Studi di Genova, 16145 Genova, Italy
| | - Michele Iacomino
- U.O.C. Genetica Medica, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy
| | - Irene Schiavetti
- Dipartimento di Scienze della Salute, Università di Genova, 16132 Genova, Italy
| | - Francesca Madia
- U.O.C. Genetica Medica, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy
| | - Monica Traverso
- U.O.C. Neurologia Pediatrica e Malattie Muscolari, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy
| | - Sara Guerrisi
- U.O.C. Genetica Medica, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy
| | - Marco Di Duca
- U.O.C. Genetica Medica, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy
| | - Francesco Caroli
- U.O.C. Genetica Medica, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy
| | - Simona Baldassari
- U.O.C. Genetica Medica, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy
| | - Barbara Tappino
- LABSIEM (Laboratory for the Study of Inborn Errors of Metabolism), IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy
| | - Ferruccio Romano
- U.O.C. Genomica e Genetica Clinica, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy
| | - Paolo Uva
- Unità di Bioinformatica Clinica, Direzione Scientifica, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy
| | - Diego Vozzi
- Genomic Facility, Istituto Italiano di Tecnologia, 16163 Genova, Italy
| | - Cristina Chelleri
- U.O.C. Neurologia Pediatrica e Malattie Muscolari, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy
| | - Gianluca Piatelli
- U.O.C. Neurochirurgia, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy
| | - Maria Cristina Diana
- U.O.C. Neurologia Pediatrica e Malattie Muscolari, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy
| | - Federico Zara
- U.O.C. Genetica Medica, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy
| | - Valeria Capra
- U.O.C. Genomica e Genetica Clinica, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy
| | - Marco Pavanello
- U.O.C. Neurochirurgia, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy
| | - Patrizia De Marco
- U.O.C. Genetica Medica, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy
| |
Collapse
|
7
|
Peduto C, Zanobio M, Nigro V, Perrotta S, Piluso G, Santoro C. Neurofibromatosis Type 1: Pediatric Aspects and Review of Genotype-Phenotype Correlations. Cancers (Basel) 2023; 15:1217. [PMID: 36831560 PMCID: PMC9954221 DOI: 10.3390/cancers15041217] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/12/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023] Open
Abstract
Neurofibromatosis type 1 (NF1) is an autosomal dominant condition, with a birth incidence of approximately 1:2000-3000, caused by germline pathogenic variants in NF1, a tumor suppressor gene encoding neurofibromin, a negative regulator of the RAS/MAPK pathway. This explains why NF1 is included in the group of RASopathies and shares several clinical features with Noonan syndrome. Here, we describe the main clinical characteristics and complications associated with NF1, particularly those occurring in pediatric age. NF1 has complete penetrance and shows wide inter- and intrafamilial phenotypic variability and age-dependent appearance of manifestations. Clinical presentation and history of NF1 are multisystemic and highly unpredictable, especially in the first years of life when penetrance is still incomplete. In this scenario of extreme phenotypic variability, some genotype-phenotype associations need to be taken into consideration, as they strongly impact on genetic counseling and prognostication of the disease. We provide a synthetic review, based on the most recent literature data, of all known genotype-phenotype correlations from a genetic and clinical perspective. Molecular diagnosis is fundamental for the confirmation of doubtful clinical diagnoses, especially in the light of recently revised diagnostic criteria, and for the early identification of genotypes, albeit few, that correlate with specific phenotypes.
Collapse
Affiliation(s)
- Cristina Peduto
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via Luigi de Crecchio 7, 80138 Naples, Italy
| | - Mariateresa Zanobio
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via Luigi de Crecchio 7, 80138 Naples, Italy
| | - Vincenzo Nigro
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via Luigi de Crecchio 7, 80138 Naples, Italy
- Telethon Institute of Genetics and Medicine (TIGEM), Via Campi Flegrei 34, 80078 Pozzuoli, Italy
| | - Silverio Perrotta
- Department of Women’s and Children’s Health and General and Specialized Surgery, University of Campania “Luigi Vanvitelli”, Via Luigi de Crecchio 2, 80138 Naples, Italy
| | - Giulio Piluso
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via Luigi de Crecchio 7, 80138 Naples, Italy
| | - Claudia Santoro
- Department of Women’s and Children’s Health and General and Specialized Surgery, University of Campania “Luigi Vanvitelli”, Via Luigi de Crecchio 2, 80138 Naples, Italy
- Clinic of Child and Adolescent Neuropsychiatry, Department of Physical and Mental Health, and Preventive Medicine, University of Campania “Luigi Vanvitelli”, Largo Madonna delle Grazie 1, 80138 Naples, Italy
| |
Collapse
|
8
|
Chaves Rabelo N, Gomes ME, de Oliveira Moraes I, Cantagalli Pfisterer J, Loss de Morais G, Antunes D, Caffarena ER, Llerena Jr J, Gonzalez S. RASopathy Cohort of Patients Enrolled in a Brazilian Reference Center for Rare Diseases: A Novel Familial LZTR1 Variant and Recurrent Mutations. Appl Clin Genet 2022; 15:153-170. [PMID: 36304179 PMCID: PMC9595068 DOI: 10.2147/tacg.s372761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 09/03/2022] [Indexed: 11/05/2022] Open
Abstract
Purpose Noonan syndrome and related disorders are genetic conditions affecting 1:1000-2000 individuals. Variants causing hyperactivation of the RAS/MAPK pathway lead to phenotypic overlap between syndromes, in addition to an increased risk of pediatric tumors. DNA sequencing methods have been optimized to provide a molecular diagnosis for clinical and genetic heterogeneity conditions. This work aimed to investigate the genetic basis in RASopathy patients through Next Generation Sequencing in a Reference Center for Rare Diseases (IFF/Fiocruz) and implement the precision medicine at a public health institute in Brazil. Patients and Methods This study comprises 26 cases with clinical suspicion of RASopathies. Sanger sequencing was used to screen variants in exons usually affected in the PTPN11 and HRAS genes for cases with clinical features of Noonan and Costello syndrome, respectively. Posteriorly, negative and new cases with clinical suspicion of RASopathy were analyzed by clinical or whole-exome sequencing. Results Molecular analysis revealed recurrent variants and a novel LZTR1 missense variant: 24 unrelated individuals with pathogenic variants [PTPN11(11), NF1(2), SOS1(2), SHOC2(2), HRAS(1), BRAF(1), LZTR (1), RAF1(1), KRAS(1), RIT1(1), a patient with co-occurrence of PTPN11 and NF1 mutations (1)]; familial cases carrying a known pathogenic variant in PTPN11 (mother-two children), and a previously undescribed paternally inherited variant in LZTR1. The comparative modeling analysis of the novel LZTR1 variant p.Pro225Leu showed local and global changes in the secondary and tertiary structures, showing a decrease of about 1% in the β-sheet content. Furthermore, evolutionary conservation indicated that Pro225 is in a highly conserved region, as observed for known dominant pathogenic variants in this protein. Conclusion Bringing precision medicine through NGS towards congenital syndromes promotes a better understanding of complex clinical and/or undiagnosed cases. The National Policy for Rare Diseases in Brazil emphasizes the importance of incorporating and optimizing diagnostic methodologies in the Unified Brazilian Health System (SUS). Therefore, this work is an important step for the NGS inclusion in diagnostic genetic routine in the public health system.
Collapse
Affiliation(s)
- Natana Chaves Rabelo
- Centro de Genética Médica IFF/Fiocruz, Rio de Janeiro, RJ, Brazil,Centro de Referência para Doenças Raras IFF/Fiocruz, Rio de Janeiro, RJ, Brazil,Laboratório de Medicina Genômica IFF/Fiocruz, Rio de Janeiro, RJ, Brazil
| | - Maria Eduarda Gomes
- Centro de Genética Médica IFF/Fiocruz, Rio de Janeiro, RJ, Brazil,Centro de Referência para Doenças Raras IFF/Fiocruz, Rio de Janeiro, RJ, Brazil,Laboratório de Medicina Genômica IFF/Fiocruz, Rio de Janeiro, RJ, Brazil
| | - Isabelle de Oliveira Moraes
- Centro de Genética Médica IFF/Fiocruz, Rio de Janeiro, RJ, Brazil,Centro de Referência para Doenças Raras IFF/Fiocruz, Rio de Janeiro, RJ, Brazil,Laboratório de Medicina Genômica IFF/Fiocruz, Rio de Janeiro, RJ, Brazil
| | - Juliana Cantagalli Pfisterer
- Centro de Genética Médica IFF/Fiocruz, Rio de Janeiro, RJ, Brazil,Centro de Referência para Doenças Raras IFF/Fiocruz, Rio de Janeiro, RJ, Brazil,Laboratório de Medicina Genômica IFF/Fiocruz, Rio de Janeiro, RJ, Brazil
| | | | - Deborah Antunes
- Laboratório de Genômica Funcional e Bioinformática, Instituto Oswaldo Cruz/Fiocruz, Rio de Janeiro, RJ, Brazil
| | - Ernesto Raúl Caffarena
- Grupo de Biofísica Computacional e Modelagem Molecular, Programa de Computação Científica, Fundação Oswaldo Cruz/Fiocruz, Rio de Janeiro, RJ, Brazil
| | - Juan Llerena Jr
- Centro de Genética Médica IFF/Fiocruz, Rio de Janeiro, RJ, Brazil,Centro de Referência para Doenças Raras IFF/Fiocruz, Rio de Janeiro, RJ, Brazil,Faculdade de Medicina de Petrópolis, FASE, Petrópolis, RJ, Brazil,INAGEMP, Rio de Janeiro, RJ, Brazil,Correspondence: Juan Llerena Jr, Email
| | - Sayonara Gonzalez
- Centro de Genética Médica IFF/Fiocruz, Rio de Janeiro, RJ, Brazil,Centro de Referência para Doenças Raras IFF/Fiocruz, Rio de Janeiro, RJ, Brazil,Laboratório de Medicina Genômica IFF/Fiocruz, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
9
|
Moyamoya disease emerging as an immune-related angiopathy. Trends Mol Med 2022; 28:939-950. [DOI: 10.1016/j.molmed.2022.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 08/17/2022] [Accepted: 08/18/2022] [Indexed: 11/22/2022]
|
10
|
Brosius SN, Vossough A, Fisher MJ, Lang SS, Beslow LA, George BJ, Ichord R. Characteristics of Moyamoya Syndrome in Pediatric Patients With Neurofibromatosis Type 1. Pediatr Neurol 2022; 134:85-92. [PMID: 35849956 DOI: 10.1016/j.pediatrneurol.2022.05.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 05/09/2022] [Accepted: 05/30/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND Moyamoya syndrome (MMS) is a progressive cerebral arteriopathy with increased incidence in children with neurofibromatosis type 1 (NF1). Despite the potential for significant neurological morbidity including stroke, little is known about the natural history, and no guidelines exist for screening and management of NF1-associated MMS. METHODS We identified 152 literature cases of children aged ≤18 years with NF1-associated MMS. A meta-analysis was performed evaluating clinical and neuroimaging findings and patient outcomes. Data from 19 patients with NF1-associated MMS from our center treated from January 1995 to July 2020 were abstracted via chart review and similarly analyzed for clinical and neuroimaging features. RESULTS Meta-analysis of literature cases showed a median age of MMS diagnosis of 6 years (interquartile range 3 to 10.8 years). Optic pathway gliomas were more common in patients with MMS (42%) compared with historical prevalence. Stroke or transient ischemic attack (TIA) was present at diagnosis in 46%. TIA and stroke were more common in patients with bilateral versus unilateral MMS (62% vs 34%, P = 0.001) and in children aged <4 years versus those aged ≥4 years (61% vs 40%, P = 0.02). Compared with the literature cases, our cohort was more frequently asymptomatic (42% vs 25%) and less likely to present with TIA or stroke (32% vs 46%) at diagnosis. CONCLUSIONS These data suggest there is an aggressive form of MMS in children with NF1 <4 years of age. Therefore, early screening should be considered to facilitate early detection and treatment of cerebral arteriopathy.
Collapse
Affiliation(s)
- Stephanie N Brosius
- Department of Pediatrics, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania; Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania.
| | - Arastoo Vossough
- Department of Radiology, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania; Division of Neuroradiology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Michael J Fisher
- Department of Pediatrics, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania; Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Shih-Shan Lang
- Department of Neurosurgery, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania; Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Lauren A Beslow
- Department of Pediatrics, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania; Department of Neurology, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania; Division of Child Neurology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Brandon J George
- Jefferson College of Population Health, Thomas Jefferson University, Philadelphia, Pennsylvania; Division of Biostatistics, Department of Pharmacology and Experimental Therapeutics, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Rebecca Ichord
- Department of Pediatrics, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania; Department of Neurology, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania; Division of Child Neurology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| |
Collapse
|
11
|
Correspondence. Retina 2022; 42:e35-e36. [PMID: 35877969 DOI: 10.1097/iae.0000000000003532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
12
|
Reply. Retina 2022; 42:e36-e38. [PMID: 35877970 DOI: 10.1097/iae.0000000000003533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
13
|
Genotype-Phenotype Correlations in Neurofibromatosis Type 1: Identification of Novel and Recurrent NF1 Gene Variants and Correlations with Neurocognitive Phenotype. Genes (Basel) 2022; 13:genes13071130. [PMID: 35885913 PMCID: PMC9316015 DOI: 10.3390/genes13071130] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 06/14/2022] [Accepted: 06/17/2022] [Indexed: 02/01/2023] Open
Abstract
Neurofibromatosis type 1 (NF1) is one of the most common genetic tumor predisposition syndrome, caused by mutations in the NF1. To date, few genotype-phenotype correlations have been discerned in NF1, due to a highly variable clinical presentation. We aimed to study the molecular spectrum of NF1 and genotype-phenotype correlations in a monocentric study cohort of 85 NF1 patients (20 relatives, 65 sporadic cases). Clinical data were collected at the time of the mutation analysis and reviewed for accuracy in this investigation. An internal phenotypic categorization was applied. The 94% of the patients enrolled showed a severe phenotype with at least one systemic complication and a wide range of associated malignancies. Spine deformities were the most common complications in this cohort. We also reported 66 different NF1 mutations, of which 7 are novel mutations. Correlation analysis identified a slight significant inverse correlation between age at diagnosis and delayed acquisition of psychomotor skills with residual multi-domain cognitive impairment. Odds ratio with 95% confidence interval showed a higher prevalence of learning disabilities in patients carrying frameshift mutations. Overall, our results aim to offer an interesting contribution to studies on the genotype–phenotype of NF1 and in genetic management and counselling.
Collapse
|
14
|
Venkataramany AS, Schieffer KM, Lee K, Cottrell CE, Wang PY, Mardis ER, Cripe TP, Chandler DS. Alternative RNA Splicing Defects in Pediatric Cancers: New Insights in Tumorigenesis and Potential Therapeutic Vulnerabilities. Ann Oncol 2022; 33:578-592. [PMID: 35339647 DOI: 10.1016/j.annonc.2022.03.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 03/15/2022] [Accepted: 03/16/2022] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Compared to adult cancers, pediatric cancers are uniquely characterized by a genomically stable landscape and lower tumor mutational burden. However, alternative splicing, a global cellular process that produces different mRNA/protein isoforms from a single mRNA transcript, has been increasingly implicated in the development of pediatric cancers. DESIGN We review the current literature on the role of alternative splicing in adult cancer, cancer predisposition syndromes, and pediatric cancers. We also describe multiple splice variants identified in adult cancers and confirmed through comprehensive genomic profiling in our institutional cohort of rare, refractory and relapsed pediatric and adolescent young adult cancer patients. Finally, we summarize the contributions of alternative splicing events to neoantigens and chemoresistance and prospects for splicing-based therapies. RESULTS Published dysregulated splicing events can be categorized as exon inclusion, exon exclusion, splicing factor upregulation, or splice site alterations. We observe these phenomena in cancer predisposition syndromes (Lynch syndrome, Li-Fraumeni syndrome, CHEK2) and pediatric leukemia (B-ALL), sarcomas (Ewing sarcoma, rhabdomyosarcoma, osteosarcoma), retinoblastoma, Wilms tumor, and neuroblastoma. Within our institutional cohort, we demonstrate splice variants in key regulatory genes (CHEK2, TP53, PIK3R1, MDM2, KDM6A, NF1) that resulted in exon exclusion or splice site alterations, which were predicted to impact functional protein expression and promote tumorigenesis. Differentially spliced isoforms and splicing proteins also impact neoantigen creation and treatment resistance, such as imatinib or glucocorticoid regimens. Additionally, splice-altering strategies with the potential to change the therapeutic landscape of pediatric cancers include antisense oligonucleotides, adeno-associated virus gene transfers, and small molecule inhibitors. CONCLUSIONS Alternative splicing plays a critical role in the formation and growth of pediatric cancers, and our institutional cohort confirms and highlights the broad spectrum of affected genes in a variety of cancers. Further studies that elucidate the mechanisms of disease-inducing splicing events will contribute toward the development of novel therapeutics.
Collapse
Affiliation(s)
- A S Venkataramany
- Biomedical Sciences Graduate Program, The Ohio State University, Columbus, Ohio, United States; Medical Scientist Training Program, The Ohio State University, Columbus, Ohio, United States
| | - K M Schieffer
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, Ohio, United States
| | - K Lee
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, Ohio, United States; Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio, United States; Department of Pathology, The Ohio State University College of Medicine, Columbus, Ohio, United States
| | - C E Cottrell
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, Ohio, United States; Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio, United States; Department of Pathology, The Ohio State University College of Medicine, Columbus, Ohio, United States
| | - P Y Wang
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio, United States; Center for Childhood Cancer and Blood Diseases, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio, United States
| | - E R Mardis
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, Ohio, United States; Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio, United States
| | - T P Cripe
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio, United States; Center for Childhood Cancer and Blood Diseases, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio, United States; Division of Hematology, Oncology and Blood and Marrow Transplant, Department of Pediatrics, The Ohio State University, Columbus, Ohio, United States
| | - D S Chandler
- Center for Childhood Cancer and Blood Diseases, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio, United States; Molecular, Cellular and Developmental Biology Graduate Program and The Center for RNA Biology, The Ohio State University, Columbus, Ohio, United States.
| |
Collapse
|
15
|
Po' C, Nosadini M, Zedde M, Pascarella R, Mirone G, Cicala D, Rosati A, Cosi A, Toldo I, Colombatti R, Martelli P, Iodice A, Accorsi P, Giordano L, Savasta S, Foiadelli T, Sanfilippo G, Lafe E, Thyrion FZ, Polonara G, Campa S, Raviglione F, Scelsa B, Bova SM, Greco F, Cordelli DM, Cirillo L, Toni F, Baro V, Causin F, Frigo AC, Suppiej A, Sainati L, Azzolina D, Agostini M, Cesaroni E, De Carlo L, Di Rosa G, Esposito G, Grazian L, Morini G, Nicita F, Operto FF, Pruna D, Ragazzi P, Rollo M, Spalice A, Striano P, Skabar A, Lanterna LA, Carai A, Marras CE, Manara R, Sartori S. Pediatric Moyamoya Disease and Syndrome in Italy: A Multicenter Cohort. Front Pediatr 2022; 10:892445. [PMID: 35601411 PMCID: PMC9120837 DOI: 10.3389/fped.2022.892445] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 04/05/2022] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND Moyamoya is a rare progressive cerebral arteriopathy, occurring as an isolated phenomenon (moyamoya disease, MMD) or associated with other conditions (moyamoya syndrome, MMS), responsible for 6-10% of all childhood strokes and transient ischemic attacks (TIAs). METHODS We conducted a retrospective multicenter study on pediatric-onset MMD/MMS in Italy in order to characterize disease presentation, course, management, neuroradiology, and outcome in a European country. RESULTS A total of 65 patients (34/65 women) with MMD (27/65) or MMS (38/65) were included. About 18% (12/65) of patients were asymptomatic and diagnosed incidentally during investigations performed for an underlying condition (incMMS), whereas 82% (53/65) of patients with MMD or MMS were diagnosed due to the presence of neurological symptoms (symptMMD/MMS). Of these latter, before diagnosis, 66% (43/65) of patients suffered from cerebrovascular events with or without other manifestations (ischemic stroke 42%, 27/65; TIA 32%, 21/65; and no hemorrhagic strokes), 18% (12/65) of them reported headache (in 4/12 headache was not associated with any other manifestation), and 26% (17/65) of them experienced multiple phenotypes (≥2 among: stroke/TIA/seizures/headache/others). Neuroradiology disclosed ≥1 ischemic lesion in 67% (39/58) of patients and posterior circulation involvement in 51% (30/58) of them. About 73% (47/64) of patients underwent surgery, and 69% (45/65) of them received aspirin, but after diagnosis, further stroke events occurred in 20% (12/61) of them, including operated patients (11%, 5/47). Between symptom onset and last follow-up, the overall patient/year incidence of stroke was 10.26% (IC 95% 7.58-13.88%). At last follow-up (median 4 years after diagnosis, range 0.5-15), 43% (26/61) of patients had motor deficits, 31% (19/61) of them had intellectual disability, 13% (8/61) of them had epilepsy, 11% (7/61) of them had behavioral problems, and 25% (13/52) of them had mRS > 2. The proportion of final mRS > 2 was significantly higher in patients with symptMMD/MMS than in patients with incMMS (p = 0.021). Onset age <4 years and stroke before diagnosis were significantly associated with increased risk of intellectual disability (p = 0.0010 and p = 0.0071, respectively) and mRS > 2 at follow-up (p = 0.0106 and p = 0.0009, respectively). CONCLUSIONS Moyamoya is a severe condition that may affect young children and frequently cause cerebrovascular events throughout the disease course, but may also manifest with multiple and non-cerebrovascular clinical phenotypes including headache (isolated or associated with other manifestations), seizures, and movement disorder. Younger onset age and stroke before diagnosis may associate with increased risk of worse outcome (final mRS > 2).
Collapse
Affiliation(s)
- Chiara Po'
- Paediatric Neurology and Neurophysiology Unit, Department of Women's and Children's Health, University Hospital of Padova, Padova, Italy.,Department of Women's and Children's Health, University of Padova, Padova, Italy.,Unit of Pediatrics, AULSS 2 Marca Trevigiana, Ca' Foncello Hospital, Treviso, Italy
| | - Margherita Nosadini
- Paediatric Neurology and Neurophysiology Unit, Department of Women's and Children's Health, University Hospital of Padova, Padova, Italy.,Neuroimmunology Group, Paediatric Research Institute "Città della Speranza", Padova, Italy
| | - Marialuisa Zedde
- Neurology Unit, Stroke Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Rosario Pascarella
- Neuroradiology Unit, Arcispedale S. Maria Nuova AUSL Reggio Emilia - IRCCS, Reggio Emilia, Italy
| | - Giuseppe Mirone
- Pediatric Neurosurgery Unit, Department of Neuroscience, Santobono-Pausilipon Children's Hospital, Naples, Italy
| | - Domenico Cicala
- Pediatric Neuroradiology, Department of Neuroscience, Santobono-Pausilipon Children's Hospital, Naples, Italy
| | - Anna Rosati
- Department of Neuroscience, Children's Hospital Anna Meyer, University of Firenze, Firenze, Italy
| | - Alessandra Cosi
- Department of Neuroscience, Children's Hospital Anna Meyer, University of Firenze, Firenze, Italy
| | - Irene Toldo
- Paediatric Neurology and Neurophysiology Unit, Department of Women's and Children's Health, University Hospital of Padova, Padova, Italy
| | - Raffaella Colombatti
- Clinic of Pediatric Hematology Oncology, Department of Women's and Children's Health, University of Padova, Padova, Italy
| | - Paola Martelli
- Child Neurology and Psychiatry Unit, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Alessandro Iodice
- Child Neurology and Psychiatry Unit, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Patrizia Accorsi
- Child Neurology and Psychiatry Unit, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Lucio Giordano
- Child Neurology and Psychiatry Unit, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Salvatore Savasta
- Pediatric Clinic, IRCCS Policlinico San Matteo Foundation, University of Pavia, Pavia, Italy
| | - Thomas Foiadelli
- Pediatric Clinic, IRCCS Policlinico San Matteo Foundation, University of Pavia, Pavia, Italy
| | - Giuseppina Sanfilippo
- Department of Diagnostic and Interventional Radiology and Neuroradiology, IRCCS Policlinico San Matteo, Pavia, Italy
| | - Elvis Lafe
- Department of Diagnostic and Interventional Radiology and Neuroradiology, IRCSS Policlinico San Matteo, Pavia, Italy
| | - Federico Zappoli Thyrion
- Department of Diagnostic and Interventional Radiology and Neuroradiology, IRCSS Policlinico San Matteo, Pavia, Italy
| | - Gabriele Polonara
- Neuroradiology - Department of Odontostomatologic and Specialized Clinical Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Serena Campa
- Neuroradiology Unit, University Hospital "Ospedali Riuniti di Ancona, " Università Politecnica delle Marche, Ancona, Italy
| | | | - Barbara Scelsa
- Department of Pediatric Neurology, Vittore Buzzi Children's Hospital, Milan, Italy
| | - Stefania Maria Bova
- Department of Pediatric Neurology, Vittore Buzzi Children's Hospital, Milan, Italy
| | - Filippo Greco
- Pediatric Clinic, Department of Clinical and Experimental Medicine, University Hospital A.U.O. "Policlinico-San Marco" of Catania, Catania, Italy
| | - Duccio Maria Cordelli
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Neuropsichiatria dell'età Pediatrica, Bologna, Italy.,Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Luigi Cirillo
- Neuroradiology Unit, IRCSS "Istituto delle Scienze Neurologiche di Bologna, " Ospedale Bellaria, Bologna, Italy
| | - Francesco Toni
- Neuroradiology Unit, IRCSS "Istituto delle Scienze Neurologiche di Bologna, " Ospedale Bellaria, Bologna, Italy
| | - Valentina Baro
- Academic Neurosurgery, Department of Neurosciences, University of Padova, Padova, Italy
| | - Francesco Causin
- Neuroradiology, Department of Neurological Sciences, University of Padova, Padova, Italy
| | - Anna Chiara Frigo
- Unit of Biostatistics, Epidemiology and Public Health, Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, Padova, Italy
| | - Agnese Suppiej
- Paediatric Neurology and Neurophysiology Unit, Department of Women's and Children's Health, University Hospital of Padova, Padova, Italy.,Pediatric Section, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Laura Sainati
- Clinic of Pediatric Hematology Oncology, Department of Women's and Children's Health, University of Padova, Padova, Italy
| | - Danila Azzolina
- Unit of Biostatistics, Epidemiology and Public Health, Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, Padova, Italy
| | - Manuela Agostini
- Department of Pediatrics, Regina Margherita Children's Hospital, Torino, Italy
| | - Elisabetta Cesaroni
- Department of Child Neuropsychiatry, University Hospital Ospedali Riuniti, Ancona, Italy
| | - Luigi De Carlo
- Pediatric Section, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Gabriella Di Rosa
- Unit of Child Neurology and Psychiatry, Department of Human Pathology of the Adult and Developmental Age, Messina, Italy
| | - Giacomo Esposito
- Pediatric Neurosurgery Unit, Department of Neuroscience and Neurorehabilitation, Bambino Gesù Children's Hospital, Roma, Italy
| | - Luisa Grazian
- Unit of Pediatrics, AULSS 2 Marca Trevigiana, Ca' Foncello Hospital, Treviso, Italy
| | - Giovanna Morini
- Department of Pediatrics, Institute for Maternal and Child Health - IRCCS "Burlo Garofolo", Trieste, Italy
| | - Francesco Nicita
- Unit of Neuromuscular and Neurodegenerative Disorders, Department of Neurosciences, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.,Child Neurology Division, Department of Pediatrics, Sapienza University, Rome, Italy
| | - Francesca Felicia Operto
- Child and Adolescent Unit, Department of Medicine, Surgery and Dentistry, University of Salerno, Salerno, Italy
| | - Dario Pruna
- Neurology and Epileptology Unit, Department of Pediatric, ARNAS Brotzu, Cagliari, Italy
| | - Paola Ragazzi
- Department of Neurosurgery, "Regina Margherita" Children's Hospital, Torino, Italy
| | - Massimo Rollo
- Interventional Radiology Unit, Department of Imaging, Bambino Gesù Children's Hospital (IRCCS), Rome, Italy
| | - Alberto Spalice
- Department of Maternal Sciences, Sapienza University, Rome, Italy
| | - Pasquale Striano
- Pediatric Neurology and Muscular Diseases Unit, IRCCS "Istituto Giannina Gaslini", Genova, Italy.,Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genova, Genova, Italy
| | - Aldo Skabar
- Department of Pediatrics, Institute for Maternal and Child Health - IRCCS "Burlo Garofolo", Trieste, Italy
| | | | - Andrea Carai
- Pediatric Neurosurgery Unit, Department of Neuroscience and Neurorehabilitation, Bambino Gesù Children's Hospital, Roma, Italy
| | - Carlo Efisio Marras
- Pediatric Neurosurgery Unit, Department of Neuroscience and Neurorehabilitation, Bambino Gesù Children's Hospital, Roma, Italy
| | - Renzo Manara
- Neuroradiology Unit, Department of Neurological Sciences, University of Padova, Padova, Italy
| | - Stefano Sartori
- Paediatric Neurology and Neurophysiology Unit, Department of Women's and Children's Health, University Hospital of Padova, Padova, Italy.,Department of Women's and Children's Health, University of Padova, Padova, Italy.,Department of Neuroscience, University of Padova, Padova, Italy.,Neuroimmunology Group, Paediatric Research Institute "Città della Speranza", Padova, Italy
| |
Collapse
|
16
|
Touzé R, Abitbol MM, Bremond-Gignac D, Robert MP. RETINAL VASCULAR ABNORMALITIES IN CHILDREN WITH NEUROFIBROMATOSIS TYPE 1. Retina 2021; 41:2589-2595. [PMID: 34111884 DOI: 10.1097/iae.0000000000003234] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
PURPOSE Retinal vascular abnormalities (RVAs) have been recently described in patients with neurofibromatosis Type 1 (NF1) as vascular tortuosity, best visible on infrared imaging. This study assessed clinical RVA's characteristics in a large series of children with NF1. METHODS This retrospective observational study was conducted in children (0-18 years) with an NF1 diagnosis. Using near-infrared imaging, RVAs were classified according to the nature of vessels involvement and their degree of tortuosity. RESULTS Retinal imaging from 140 children, with a median age of 8.8 years (1.5-18), was included; 52 patients (37.1%) (81 eyes) exhibited RVAs. These RVAs comprised 96% (50/52) of simple vascular tortuosity and 17% (9/52) of a corkscrew pattern. A corkscrew pattern involved only small veins, whereas simple vascular tortuosity could affect both arteries and veins. No statistically significant age correlation was observed, but evolution of RVAs from simple vascular tortuosity to corkscrew pattern was observed in 5 cases. CONCLUSION Retinal vascular abnormalities occurred in 37.1% of children with NF1. These abnormalities may result from NF1 promoting localized tortuosity in both small arteries and veins, whereas only small second-order or tertiary-order venules evolve to a highly tortuous pattern.
Collapse
Affiliation(s)
- Romain Touzé
- Ophthalmology Department, Hôpital Universitaire Necker-Enfants Malades, Paris, France
| | - Marc M Abitbol
- Ophthalmology Department, Hôpital Universitaire Necker-Enfants Malades, Paris, France
- INSERM, UMRS 1138, Team 17, from Physiopathology of Ocular Diseases to Clinical Development, Paris University, Paris, France; and
| | - Dominique Bremond-Gignac
- Ophthalmology Department, Hôpital Universitaire Necker-Enfants Malades, Paris, France
- INSERM, UMRS 1138, Team 17, from Physiopathology of Ocular Diseases to Clinical Development, Paris University, Paris, France; and
| | - Matthieu P Robert
- Ophthalmology Department, Hôpital Universitaire Necker-Enfants Malades, Paris, France
- Borelli Centre, UMR 9010, CNRS-SSA-ENS Paris Saclay-Paris University, Paris, France
| |
Collapse
|
17
|
D'Amico A, Rosano C, Pannone L, Pinna V, Assunto A, Motta M, Ugga L, Daniele P, Mandile R, Mariniello L, Siano MA, Santoro C, Piluso G, Martinelli S, Strisciuglio P, De Luca A, Tartaglia M, Melis D. Clinical variability of neurofibromatosis 1: A modifying role of cooccurring PTPN11 variants and atypical brain MRI findings. Clin Genet 2021; 100:563-572. [PMID: 34346503 DOI: 10.1111/cge.14040] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 07/28/2021] [Accepted: 07/30/2021] [Indexed: 01/04/2023]
Abstract
Neurofibromatosis 1 (NF1) is a disorder characterized by variable expressivity caused by loss-of-function variants in NF1, encoding neurofibromin, a protein negatively controlling RAS signaling. We evaluated whether concurrent variation in proteins functionally linked to neurofibromin contribute to the variable expressivity of NF1. Parallel sequencing of a RASopathy gene panel in 138 individuals with molecularly confirmed clinical diagnosis of NF1 identified missense variants in PTPN11, encoding SHP2, a positive regulator of RAS signaling, in four subjects from three unrelated families. Three subjects were heterozygous for a gain-of-function variant and showed a severe expression of NF1 (developmental delay, multiple cerebral neoplasms and peculiar cortical MRI findings), and features resembling Noonan syndrome (a RASopathy caused by activating variants in PTPN11). Conversely, the fourth subject, who showed an attenuated presentation, carried a previously unreported PTPN11 variant that had a hypomorphic behavior in vitro. Our findings document that functionally relevant PTPN11 variants occur in a small but significant proportion of subjects with NF1 modulating disease presentation, suggesting a model in which the clinical expression of pathogenic NF1 variants is modified by concomitant dysregulation of protein(s) functionally linked to neurofibromin. We also suggest targeting of SHP2 function as an approach to treat evolutive complications of NF1.
Collapse
Affiliation(s)
- Alessandra D'Amico
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Naples, Italy.,Tortorella Private Hospital, Salerno, Italy
| | - Carmen Rosano
- Translational Medical Sciences Department, University of Naples "Federico II", Naples, Italy
| | - Luca Pannone
- Genetics and Rare Diseases Research Division, Pediatric Hospital Bambino Gesù, IRCCS, Rome, Italy
| | - Valentina Pinna
- Medical Genetics Division, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Foggia, Italy
| | - Antonia Assunto
- Translational Medical Sciences Department, University of Naples "Federico II", Naples, Italy
| | - Marialetizia Motta
- Genetics and Rare Diseases Research Division, Pediatric Hospital Bambino Gesù, IRCCS, Rome, Italy
| | - Lorenzo Ugga
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Naples, Italy.,Tortorella Private Hospital, Salerno, Italy
| | - Paola Daniele
- Medical Genetics Division, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Foggia, Italy
| | - Roberta Mandile
- Translational Medical Sciences Department, University of Naples "Federico II", Naples, Italy
| | - Lucio Mariniello
- Translational Medical Sciences Department, University of Naples "Federico II", Naples, Italy
| | - Maria Anna Siano
- Department of Medicine, Surgery and Dentistry, "Scuola Medica Salernitana", Salerno, Italy
| | - Claudia Santoro
- Referral Centre of Neurofibromatosis, Department of Woman and Child, Specialistic and General Surgery, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Giulio Piluso
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Simone Martinelli
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Pietro Strisciuglio
- Translational Medical Sciences Department, University of Naples "Federico II", Naples, Italy
| | - Alessandro De Luca
- Medical Genetics Division, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Foggia, Italy
| | - Marco Tartaglia
- Genetics and Rare Diseases Research Division, Pediatric Hospital Bambino Gesù, IRCCS, Rome, Italy
| | - Daniela Melis
- Translational Medical Sciences Department, University of Naples "Federico II", Naples, Italy.,Department of Medicine, Surgery and Dentistry, "Scuola Medica Salernitana", Salerno, Italy
| |
Collapse
|
18
|
Barreto-Duarte B, Andrade-Gomes FH, Arriaga MB, Araújo-Pereira M, Cubillos-Angulo JM, Andrade BB. Association between neurofibromatosis type 1 and cerebrovascular diseases in children: A systematic review. PLoS One 2021; 16:e0241096. [PMID: 33395412 PMCID: PMC7781472 DOI: 10.1371/journal.pone.0241096] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 12/19/2020] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Neurofibromatosis type 1 (NF-1) is an autosomal dominant disease that affects one in every 3000 individuals. This disease can present a wide range of clinical manifestations, ranging from skin abnormalities to severe vascular damage. Although not commonly recognized in the context of NF-1, cerebrovascular disease (CVD), can be often present since childhood and diagnosed just later in life. When present, NF-1-associated CVD clinical manifestations may include headache, cognitive deficits and ultimately aneurysm rupture, causing death. Thus, CVD plays an important role in the clinical manifestations, disease severity and prognosis of patients with NF-1. This systematic review aims to summarize the body of evidence linking NF-1 and CVD in children. METHODS Two independent investigators performed a systematic review on the PubMed and EMBASE search platforms, using the following key terms: "neurofibromatosis type 1", "Von Recklinghausen's disease", "children", "adolescents", "stroke", "Moyamoya disease", "vascular diseases", "cerebrovascular disorders", "aneurysm" and "congenital abnormalities". Studies focused on assessing the development of CVD in children with NF-1 were included. RESULTS Seven studies met the inclusion criteria. Twelve different clinical manifestations have been associated with cerebrovascular changes in children with NF-1; 44,5% of diagnosed patients were asymptomatic. CONCLUSION The available evidence suggests that CVDs are related with the progression of NF-1, even in the absence of a clear clinical manifestation. In addition, improved prognosis was observed when imaging tests were performed to screen for cerebrovascular alterations early during the clinical investigation. Early diagnosis of CVD in NF-1 patients foster implementation of timely interventions, directly impacting clinical outcomes.
Collapse
Affiliation(s)
- Beatriz Barreto-Duarte
- Laboratório de Inflamação e Biomarcadores, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Bahia, Brazil
- Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador, Bahia, Brazil
- Curso de Medicina, Universidade Salvador (UNIFACS), Salvador, Bahia, Brazil
- Programa de Pós-Graduação em Clínica Médica, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- * E-mail: (BBA); (BBD)
| | | | - María B. Arriaga
- Laboratório de Inflamação e Biomarcadores, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Bahia, Brazil
- Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador, Bahia, Brazil
- Curso de Medicina, Centro Universitário Faculdade de Tecnologia e Ciências (UniFTC), Salvador, Bahia, Brazil
| | - Mariana Araújo-Pereira
- Laboratório de Inflamação e Biomarcadores, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Bahia, Brazil
- Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador, Bahia, Brazil
- Faculdade de Medicina, Universidade Federal da Bahia, Salvador, Bahia, Brazil
| | - Juan Manuel Cubillos-Angulo
- Laboratório de Inflamação e Biomarcadores, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Bahia, Brazil
- Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador, Bahia, Brazil
- Curso de Medicina, Centro Universitário Faculdade de Tecnologia e Ciências (UniFTC), Salvador, Bahia, Brazil
| | - Bruno B. Andrade
- Laboratório de Inflamação e Biomarcadores, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Bahia, Brazil
- Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador, Bahia, Brazil
- Curso de Medicina, Universidade Salvador (UNIFACS), Salvador, Bahia, Brazil
- Programa de Pós-Graduação em Clínica Médica, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Curso de Medicina, Centro Universitário Faculdade de Tecnologia e Ciências (UniFTC), Salvador, Bahia, Brazil
- Faculdade de Medicina, Universidade Federal da Bahia, Salvador, Bahia, Brazil
- Curso de Medicina, Escola Bahiana de Medicina e Saúde Pública (EBMSP), Salvador, Bahia, Brazil
- * E-mail: (BBA); (BBD)
| |
Collapse
|
19
|
Feghali J, Xu R, Yang W, Liew JA, Blakeley J, Ahn ES, Tamargo RJ, Huang J. Moyamoya disease versus moyamoya syndrome: comparison of presentation and outcome in 338 hemispheres. J Neurosurg 2020; 133:1441-1449. [PMID: 31585423 DOI: 10.3171/2019.6.jns191099] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 06/28/2019] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Phenotypic differences between moyamoya disease (MMD) and moyamoya syndrome (MMS) remain unclear. The purpose of this study was to evaluate whether such differences exist when presentation, procedure-related, and outcome variables are compared quantitatively. METHODS The study cohort included 185 patients with moyamoya presenting to the Johns Hopkins Medical Institutions between 1994 and 2015. Baseline demographic, angiographic, and clinical characteristics were compared between patients with MMS and MMD, in addition to procedure-related complications and length of stay (LOS) after surgery. Stroke-free survival was compared between both disease variants after diagnosis. Kaplan-Meier analysis and Cox proportional hazards regression were used to compare stroke-free survival between surgically treated and conservatively managed hemispheres in both types of disease, while evaluating interaction between disease variant and management. RESULTS The cohort consisted of 137 patients with MMD (74%) with a bimodal age distribution and 48 patients with MMS (26%) who were mostly under 18 years of age (75%). Underlying diseases included sickle cell disease (48%), trisomy 21 (12%), neurofibromatosis (23%), and other disorders (17%). Patients with MMS were younger (p < 0.001) and less likely to be female (p = 0.034). Otherwise, baseline characteristics were statistically comparable. The rate of surgical complications was 33% in patients with MMD and 16% in patients with MMS (p = 0.097). Both groups of patients had a similar LOS after surgery (p = 0.823). Survival analysis (n = 330 hemispheres) showed similar stroke-free survival after diagnosis (p = 0.856) and lower stroke hazard in surgically managed patients in both MMD (hazard ratio [HR] 0.29, p = 0.028) and MMS (HR 0.62, p = 0.586). The disease variant (MMD vs MMS) did not affect the relationship between management approach (surgery vs conservative) and stroke hazard (p = 0.787). CONCLUSIONS MMD and MMS have largely comparable clinical and angiographic phenotypes with analogously favorable responses to surgical revascularization.
Collapse
Affiliation(s)
| | | | | | | | - Jaishri Blakeley
- 2Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland; and
| | - Edward S Ahn
- 3Department of Neurosurgery, Mayo Clinic, Rochester, Minnesota
| | | | | |
Collapse
|
20
|
Pabst L, Carroll J, Lo W, Truxal KV. Moyamoya syndrome in a child with Legius syndrome: Introducing a cerebral vasculopathy to the SPRED1 phenotype? Am J Med Genet A 2020; 185:223-227. [PMID: 33078527 DOI: 10.1002/ajmg.a.61921] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 09/22/2020] [Accepted: 09/27/2020] [Indexed: 11/11/2022]
Abstract
Legius syndrome is a disorder of the RAS and mitogen-activated protein kinase (MAPK) pathway first described in 2007 by Eric Legius, et al., that has been considered a milder phenotype than reported in the RASopathy neurofibromatosis type 1 (NF1). However, with approximately 200 cases reported in the literature, the Legius syndrome phenotype remains to be fully characterized. We report a child who presented with moyamoya syndrome and who has Legius syndrome due to a pathogenic variant in SPRED1. Vascular complications such as moyamoya syndrome have been reported in NF1. However, this association has not been reported in Legius syndrome. This child's case may represent an expansion of the clinical phenotype of Legius syndrome, and further study is needed. We emphasize the importance of obtaining neuroimaging studies in patients with Legius syndrome who present with new neurologic deficits.
Collapse
Affiliation(s)
- Lisa Pabst
- Division of Neurology, Department of Pediatrics, Nationwide Children's Hospital, Columbus, Ohio, US
| | - Jennifer Carroll
- Division of Genetic and Genomic Medicine, Department of Internal Medicine, Nationwide Children's Hospital and The Ohio State University, Columbus, Ohio, US
| | - Warren Lo
- Division of Neurology, Depts. Of Pediatrics and Neurology, The Ohio State University and Nationwide Children's Hospital, Columbus, Ohio, US
| | - Kristen V Truxal
- Division of Genetic and Genomic Medicine, Department of Pediatrics, Nationwide Children's Hospital and The Ohio State University, Columbus, Ohio, US
| |
Collapse
|
21
|
Bernardo P, Cinalli G, Santoro C. Epilepsy in NF1: a systematic review of the literature. Childs Nerv Syst 2020; 36:2333-2350. [PMID: 32613422 DOI: 10.1007/s00381-020-04710-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 05/27/2020] [Indexed: 12/11/2022]
Abstract
Epilepsy is one of the possible neurological manifestations of the neurofibromatosis type 1 (NF1) that represents the most common neurocutaneous disorder. We performed a systematic review of the literature on epilepsy associated with NF1 since 1995 in order to better define prevalence and describe type and causes of seizures. Data on type, nature of studies, number of patients, gender, and inheritance of NF1 were recorded as well as data on causes, type, EEGs, brain imaging, intellectual disability (ID), surgical treatment, and outcome of epilepsy. We identified a total of 141 references through the literature search of Pubmed and Embase. After screening, 42 records were identified, including 11617 individuals with NF1 (53% of males). Overall prevalence was estimated at 5.4% lifelong with values that seemed to be slightly lower in children, 3.7% (p 0.0016). Neither gender differences nor correlation with NF1 inheritance was found. Focal with or without bilateral tonic-clonic seizures were the most common seizure type encountered (60.9%). Structural causes were identified in half of cases (114/226). Low-grade gliomas were the most frequent associated lesions followed by mesial temporal sclerosis, malformation of cortical development, dysembryoplastic neuroepithelial tumor, and cerebrovascular lesions. In these cases, the surgical approach improved the epileptic outcome. Prevalence of epilepsy is higher in subjects with NF1 respect of the general population, with values apparently significantly lower in pediatric age. Brain tumors and cytoarchitectural abnormalities are the most frequent causes of epilepsy in this population, although many other brain complications should be taken in account.
Collapse
Affiliation(s)
- Pia Bernardo
- Pediatric Psychiatry and Neurology, Department of Neuroscience, Santobono-Pausilipon Children's Hospital, Naples, Italy
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Giuseppe Cinalli
- Pediatric Neurosurgery, Department of Neuroscience, Santobono-Pausilipon Children's Hospital, Naples, Italy
| | - Claudia Santoro
- Pediatric Psychiatry and Neurology, Department of Neuroscience, Santobono-Pausilipon Children's Hospital, Naples, Italy.
- Pediatric Neurosurgery, Department of Neuroscience, Santobono-Pausilipon Children's Hospital, Naples, Italy.
- Referral Centre of Neurofibromatosis, Department of Woman and Child, Specialistic and General Surgery, "Luigi Vanvitelli" University of Campania, Via Luigi de Crecchio, 2, 80138, Naples, Italy.
| |
Collapse
|
22
|
D'Amico A, Ugga L, Cocozza S, Giorgio SMDA, Cicala D, Santoro C, Melis D, Cinalli G, Brunetti A, Pappatà S. Multimodal evaluation of the cerebrovascular reserve in Neurofibromatosis type 1 patients with Moyamoya syndrome. Neurol Sci 2020; 42:655-663. [PMID: 32651859 PMCID: PMC7843564 DOI: 10.1007/s10072-020-04574-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 07/02/2020] [Indexed: 11/25/2022]
Abstract
Purpose Moyamoya syndrome (MMS) is a rare intracranial arterial vasculopathy which can occur in neurofibromatosis type 1 (NF1) disease, representing a cause of cerebrovascular reserve (CVR) impairment, possibly leading to ischemic stroke. Here, we evaluated noninvasive imaging techniques used to assess CVR in MMS patients, describing clinical and imaging findings in patients affected by MMS-NF1. Methods Following strict inclusion and exclusion criteria, in this retrospective observational study, we evaluated imaging data of nine consecutive MMS-NF1 patients (M/F = 5/4, mean age: 12.6 ± 4.0). Subjects underwent a multimodal evaluation of cerebral vascular status, including intracranial arterial MR Angiography (MRA), MRI perfusion with dynamic susceptibility contrast (DSC) technique, and 99mTc-hexamethylpropyleneamine oxime (HMPAO) SPECT. Results In 8 out 9 patients (88.8%, 6/8 symptomatic), time-to-peak maps were correlated with the involved cerebral hemisphere, while in 6 out 9 patients (66.6%, 5/6 symptomatic), mean transit time (MTT) maps showed correspondence with the affected cerebrovascular territories. Cerebral blood flow (CBF) calculated using DSC perfusion failed to detect the hypoperfused regions instead identified by SPECT-CBF in all patients, while MTT maps overlapped with SPECT-CBF data in all cases and time-to-peak maps in 60.0%. Conclusions Although SPECT imaging still represents the gold standard for CBF assessment, our results suggest that data obtained using DSC perfusion technique, and in particular MTT maps, might be a very useful and noninvasive tool for evaluating hemodynamic status in MMS-NF1 patients. Electronic supplementary material The online version of this article (10.1007/s10072-020-04574-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Alessandra D'Amico
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Via Pansini, 5, 80131, Naples, Italy
| | - Lorenzo Ugga
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Via Pansini, 5, 80131, Naples, Italy
| | - Sirio Cocozza
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Via Pansini, 5, 80131, Naples, Italy.
| | | | - Domenico Cicala
- Department of Pediatric Neurosurgery, Santobono-Pausilipon Children's Hospital, Naples, Italy
| | - Claudia Santoro
- Referral Centre of Neurofibromatosis, Department of Woman and Child, Specialistic and General Surgery, University "Luigi Vanvitelli", Naples, Italy
| | - Daniela Melis
- Department of Translational Medical Sciences, Section of Pediatrics, University of Naples "Federico II", Naples, Italy
| | - Giuseppe Cinalli
- Department of Pediatric Neurosurgery, Santobono-Pausilipon Children's Hospital, Naples, Italy
| | - Arturo Brunetti
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Via Pansini, 5, 80131, Naples, Italy
| | - Sabina Pappatà
- Institute of Biostructure and Bioimaging, National Research Council, Naples, Italy
| |
Collapse
|
23
|
Santoro C, Picariello S, Palladino F, Spennato P, Melis D, Roth J, Cirillo M, Quaglietta L, D’Amico A, Gaudino G, Meucci MC, Ferrara U, Constantini S, Perrotta S, Cinalli G. Retrospective Multicentric Study on Non-Optic CNS Tumors in Children and Adolescents with Neurofibromatosis Type 1. Cancers (Basel) 2020; 12:E1426. [PMID: 32486389 PMCID: PMC7353051 DOI: 10.3390/cancers12061426] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 05/22/2020] [Accepted: 05/28/2020] [Indexed: 02/01/2023] Open
Abstract
s: The natural history of non-optic central nervous system (CNS) tumors in neurofibromatosis type 1 (NF1) is largely unknown. Here, we describe prevalence, clinical presentation, treatment, and outcome of 49 non-optic CNS tumors observed in 35 pediatric patients (0-18 years). Patient- and tumor-related data were recorded. Overall survival (OS) and progression-free survival (PFS) were evaluated. Eighteen patients (51%) harbored an optic pathway glioma (OPG) and eight (23%) had multiple non-optic CNS lesions. The majority of lesions (37/49) were managed with a wait-and-see strategy, with one regression and five reductions observed. Twenty-one lesions (42.9%) required surgical treatment. Five-year OS was 85.3%. Twenty-four patients progressed with a 5-year PFS of 41.4%. Patients with multiple low-grade gliomas progressed earlier and had a lower 5-year PFS than those with one lesion only (14.3% vs. 57.9%), irrespective of OPG co-presence. Non-optic CNS tumors are common in young patients with NF1. Neither age and symptoms at diagnosis nor tumor location influenced time to progression in our series. Patients with multiple lesions tended to have a lower age at onset and to progress earlier, but with a good OS.
Collapse
Affiliation(s)
- Claudia Santoro
- Neurofibromatosis Referral Center, Department of Women’s and Children’s Health, and General and Specialized Surgery, “Luigi Vanvitelli” University of Campania, Via Luigi de Crecchio 2, 80138 Naples, Italy; (S.P.); (F.P.); (G.G.); (S.P.)
- Clinic of Child and Adolescent Neuropsychiatry, Department of Mental and Physical Health, and Preventive Medicine, “Luigi Vanvitelli” University of Campania, Largo Madonna delle Grazie 1, 80138 Naples, Italy
| | - Stefania Picariello
- Neurofibromatosis Referral Center, Department of Women’s and Children’s Health, and General and Specialized Surgery, “Luigi Vanvitelli” University of Campania, Via Luigi de Crecchio 2, 80138 Naples, Italy; (S.P.); (F.P.); (G.G.); (S.P.)
- Department of Advanced Medical and Surgical Sciences, “Luigi Vanvitelli” University of Campania, P.zza L. Miraglia 2, 80138 Naples, Italy
| | - Federica Palladino
- Neurofibromatosis Referral Center, Department of Women’s and Children’s Health, and General and Specialized Surgery, “Luigi Vanvitelli” University of Campania, Via Luigi de Crecchio 2, 80138 Naples, Italy; (S.P.); (F.P.); (G.G.); (S.P.)
| | - Pietro Spennato
- Department of Pediatric Neurosurgery, Santobono-Pausilipon Children’s Hospital, Via Mario Fiore 6, 80129 Naples, Italy; (P.S.); (M.C.M.); (G.C.)
| | - Daniela Melis
- Department of Medicine, Surgery and Dentistry, “Scuola Medica Salernitana”, Via Salvador Allende, Baronissi, 84081 Salerno, Italy;
| | - Jonathan Roth
- Department of Pediatric Neurosurgery, Dana Children’s Hospital, Tel Aviv Sourasky Medical Center, 6 Weizmann St., Tel Aviv 6423906, Israel; (J.R.); (S.C.)
| | - Mario Cirillo
- Department of Medicine, Surgery, Neurology, Metabolism and Geriatrics, “Luigi Vanvitelli” University of Campania, Piazza Luigi Miraglia 2, 80138 Naples, Italy;
| | - Lucia Quaglietta
- Department of Pediatric Oncology, Santobono-Pausilipon Children’s Hospital, Via Mario Fiore 6, 80129 Naples, Italy;
| | - Alessandra D’Amico
- Department of Advanced Biomedical Sciences, “Federico II” University of Naples, Via Sergio Pansini 5, 80100 Naples, Italy;
| | - Giuseppina Gaudino
- Neurofibromatosis Referral Center, Department of Women’s and Children’s Health, and General and Specialized Surgery, “Luigi Vanvitelli” University of Campania, Via Luigi de Crecchio 2, 80138 Naples, Italy; (S.P.); (F.P.); (G.G.); (S.P.)
| | - Maria Chiara Meucci
- Department of Pediatric Neurosurgery, Santobono-Pausilipon Children’s Hospital, Via Mario Fiore 6, 80129 Naples, Italy; (P.S.); (M.C.M.); (G.C.)
| | - Ursula Ferrara
- Section of Pediatrics, Department of Translational Medical Science, “Federico II” University of Naples, Via Sergio Pansini 5, 80100 Naples, Italy;
| | - Shlomi Constantini
- Department of Pediatric Neurosurgery, Dana Children’s Hospital, Tel Aviv Sourasky Medical Center, 6 Weizmann St., Tel Aviv 6423906, Israel; (J.R.); (S.C.)
| | - Silverio Perrotta
- Neurofibromatosis Referral Center, Department of Women’s and Children’s Health, and General and Specialized Surgery, “Luigi Vanvitelli” University of Campania, Via Luigi de Crecchio 2, 80138 Naples, Italy; (S.P.); (F.P.); (G.G.); (S.P.)
| | - Giuseppe Cinalli
- Department of Pediatric Neurosurgery, Santobono-Pausilipon Children’s Hospital, Via Mario Fiore 6, 80129 Naples, Italy; (P.S.); (M.C.M.); (G.C.)
| |
Collapse
|
24
|
Giugliano T, Santoro C, Torella A, Del Vecchio Blanco F, Grandone A, Onore ME, Melone MAB, Straccia G, Melis D, Piccolo V, Limongelli G, Buono S, Perrotta S, Nigro V, Piluso G. Clinical and Genetic Findings in Children with Neurofibromatosis Type 1, Legius Syndrome, and Other Related Neurocutaneous Disorders. Genes (Basel) 2019; 10:genes10080580. [PMID: 31370276 PMCID: PMC6722641 DOI: 10.3390/genes10080580] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 07/27/2019] [Accepted: 07/30/2019] [Indexed: 02/07/2023] Open
Abstract
Pigmentary manifestations can represent an early clinical sign in children affected by Neurofibromatosis type 1 (NF1), Legius syndrome, and other neurocutaneous disorders. The differential molecular diagnosis of these pathologies is a challenge that can now be met by combining next generation sequencing of target genes with concurrent second-level tests, such as multiplex ligation-dependent probe amplification and RNA analysis. We clinically and genetically investigated 281 patients, almost all pediatric cases, presenting with either NF1 (n = 150), only pigmentary features (café au lait macules with or without freckling; (n = 95), or clinical suspicion of other RASopathies or neurocutaneous disorders (n = 36). The causative variant was identified in 239 out of the 281 patients analyzed (85.1%), while 42 patients remained undiagnosed (14.9%). The NF1 and SPRED1 genes were mutated in 73.3% and 2.8% of cases, respectively. The remaining 8.9% carried mutations in different genes associated with other disorders. We achieved a molecular diagnosis in 69.5% of cases with only pigmentary manifestations, allowing a more appropriate clinical management of these patients. Our findings, together with the increasing availability and sharing of clinical and genetic data, will help to identify further novel genotype–phenotype associations that may have a positive impact on patient follow-up.
Collapse
Affiliation(s)
- Teresa Giugliano
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Via L. De Crecchio 7, 80138 Napoli, Italy
| | - Claudia Santoro
- Departement of Women's and Children's Health and General and Specialized Surgery, University of Campania "Luigi Vanvitelli", Via Luigi De Crecchio 2, 80138 Napoli, Italy
| | - Annalaura Torella
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Via L. De Crecchio 7, 80138 Napoli, Italy
| | - Francesca Del Vecchio Blanco
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Via L. De Crecchio 7, 80138 Napoli, Italy
| | - Anna Grandone
- Departement of Women's and Children's Health and General and Specialized Surgery, University of Campania "Luigi Vanvitelli", Via Luigi De Crecchio 2, 80138 Napoli, Italy
| | - Maria Elena Onore
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Via L. De Crecchio 7, 80138 Napoli, Italy
| | - Mariarosa Anna Beatrice Melone
- Department of Medical Sciences and Advanced Surgery, University of Campania "Luigi Vanvitelli", Piazza L. Miraglia 2, 80138 Napoli, Italy
| | - Giulia Straccia
- Department of Medical Sciences and Advanced Surgery, University of Campania "Luigi Vanvitelli", Piazza L. Miraglia 2, 80138 Napoli, Italy
| | - Daniela Melis
- Department of Pediatrics, University of Naples "Federico II", Via Pansini 5, 80131 Napoli, Italy
| | - Vincenzo Piccolo
- Dermatology Unit, University of Campania "Luigi Vanvitelli", Via Pansini 5, 80131 Napoli, Italy
| | - Giuseppe Limongelli
- Department of Translational Medicine, University of Campania "Luigi Vanvitelli", Via L. Bianchi c/o Ospedale Monaldi, 80131 Napoli, Italy
| | - Salvatore Buono
- Department of Neurosciences, "Santobono-Pausilipon" Pediatric Hospital, Via Fiore 6, 80129 Napoli, Italy
| | - Silverio Perrotta
- Departement of Women's and Children's Health and General and Specialized Surgery, University of Campania "Luigi Vanvitelli", Via Luigi De Crecchio 2, 80138 Napoli, Italy
| | - Vincenzo Nigro
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Via L. De Crecchio 7, 80138 Napoli, Italy
- Telethon Institute of Genetics and Medicine (TIGEM), Via Campi Flegrei 34, 80078 Pozzuoli, Italy
| | - Giulio Piluso
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Via L. De Crecchio 7, 80138 Napoli, Italy.
| |
Collapse
|
25
|
Sampson J, Thompson HL, Parilo DMW. Caring for children with neurofibromatosis type 1. Nursing 2019; 49:30-36. [PMID: 30839383 DOI: 10.1097/01.nurse.0000554214.17051.d9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
This article discusses the variable physical manifestations of neurofibromatosis type 1 among children in terms of presentation, disease severity, and prognosis, and addresses appropriate nursing interventions and patient teaching.
Collapse
Affiliation(s)
- Janice Sampson
- Janice Sampson is an associate professor in the School of Nursing, California State University in Sacramento, Calif. Also at California State University in Sacramento, Heather Thompson is an assistant professor in the Department of Communication Sciences and Disorders, and Denise Wall Parilo is a professor in the School of Nursing
| | | | | |
Collapse
|
26
|
Luisa SF, Rizzo A, Bedini G, Capone F, Di Lazzaro V, Nava S, Acerbi F, Rossi DS, Binelli S, Faragò G, Gioppo A, Grisoli M, Bruzzone MG, Ferroli P, Pantaleoni C, Caputi L, Gomez JV, Parati EA, Bersano A. Microduplication of 15q13.3 and Microdeletion of 18q21.32 in a Patient with Moyamoya Syndrome. Int J Mol Sci 2018; 19:ijms19113675. [PMID: 30463371 PMCID: PMC6274901 DOI: 10.3390/ijms19113675] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 11/13/2018] [Accepted: 11/16/2018] [Indexed: 01/28/2023] Open
Abstract
Moyamoya angiopathy (MA) is a cerebrovascular disease determining a progressive stenosis of the terminal part of the internal carotid arteries (ICAs) and their proximal branches and the compensatory development of abnormal “moyamoya” vessels. MA occurs as an isolated cerebral angiopathy (so-called moyamoya disease) or in association with various conditions (moyamoya syndromes) including several heritable conditions such as Down syndrome, neurofibromatosis type 1 and other genomic defects. Although the mechanism that links MA to these genetic syndromes is still unclear, it is believed that the involved genes may contribute to the disease susceptibility. Herein, we describe the case of a 43 years old woman with bilateral MA and peculiar facial characteristics, having a 484-kb microduplication of the chromosomal region 15q13.3 and a previously unreported 786 kb microdeletion in 18q21.32. This patient may have a newly-recognized genetic syndrome associated with MA. Although the relationship between these genetic variants and MA is unclear, our report would contribute to widening the genetic scenario of MA, in which not only genic mutation, but also genome unbalances are possible candidate susceptibility factors.
Collapse
Affiliation(s)
- Sciacca Francesca Luisa
- Dipartimento di Diagnostica e Tecnologia Applicata, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy.
| | - Ambra Rizzo
- Dipartimento di Diagnostica e Tecnologia Applicata, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy.
| | - Gloria Bedini
- Laboratory of Cellular Neurobiology, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy.
| | - Fioravante Capone
- Unit of Neurology, Neurophysiology, Neurobiology, Department of Medicine, Università Campus Bio-Medico di Roma, 00128 Rome, Italy.
| | - Vincenzo Di Lazzaro
- Unit of Neurology, Neurophysiology, Neurobiology, Department of Medicine, Università Campus Bio-Medico di Roma, 00128 Rome, Italy.
| | - Sara Nava
- Laboratory of Cellular Neurobiology, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy.
| | - Francesco Acerbi
- Neurosurgical Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy.
| | - Davide Sebastiano Rossi
- Neurophysiopathology Department and Epilepsy Centre, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy.
| | - Simona Binelli
- Neurophysiopathology Department and Epilepsy Centre, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy.
| | - Giuseppe Faragò
- Neuroradiological Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy.
| | - Andrea Gioppo
- Neuroradiological Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy.
| | - Marina Grisoli
- Neuroradiological Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy.
| | - Maria Grazia Bruzzone
- Neuroradiological Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy.
| | - Paolo Ferroli
- Neurosurgical Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy.
| | - Chiara Pantaleoni
- Developmental Neurology Division, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy.
| | - Luigi Caputi
- Cerebrovascular Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy.
| | - Jesus Vela Gomez
- Cerebrovascular Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy.
| | - Eugenio Agostino Parati
- Cerebrovascular Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy.
| | - Anna Bersano
- Cerebrovascular Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy.
| |
Collapse
|
27
|
Santoro C, Giugliano T, Kraemer M, Torella A, Schwitalla JC, Cirillo M, Melis D, Berlit P, Nigro V, Perrotta S, Piluso G. Whole exome sequencing identifies MRVI1 as a susceptibility gene for moyamoya syndrome in neurofibromatosis type 1. PLoS One 2018; 13:e0200446. [PMID: 30001348 PMCID: PMC6042724 DOI: 10.1371/journal.pone.0200446] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 06/26/2018] [Indexed: 12/30/2022] Open
Abstract
Background and purpose Moyamoya angiopathy is a progressive cerebral vasculopathy. The p.R4810K substitution in RNF213 has previously been linked to moyamoya disease in Asian populations. When associated with other medical conditions, such as neurofibromatosis type 1, this vasculopathy is frequently reported as moyamoya syndrome. Intriguingly, most cases of moyamoya-complicated neurofibromatosis type 1 have been described in Caucasians, inverting the population ratio observed in Asians, although prevalence of neurofibromatosis type 1 is constant worldwide. Our aim was to investigate whether, among Caucasians, additive genetic factors may contribute to the occurrence of moyamoya in neurofibromatosis type 1. Methods Whole exome sequencing was carried out on an Italian family with moyamoya-complicated neurofibromatosis type 1 to identify putative genetic modifiers independent of the NF1 locus and potentially involved in moyamoya pathogenesis. Results were validated in an unrelated family of German ancestry. Results We identified the p.P186S substitution (rs35857561) in MRVI1 that segregated with moyamoya syndrome in both the Italian and German family. Conclusions The rs35857561 polymorphism in MRVI1 may be a genetic susceptibility factor for moyamoya in European patients with neurofibromatosis type 1. MRVI1 is a functional partner of ITPR1, PRKG1 and GUCY1A3, which are involved in response to nitric oxide. Mutations in GUCY1A3 have been recently linked to a recessive syndromic form of moyamoya with esophageal achalasia.
Collapse
Affiliation(s)
- Claudia Santoro
- Dipartimento della Donna, del Bambino e di Chirurgia Generale e Specialistica, Università degli Studi della Campania “Luigi Vanvitelli”, Naples, Italy
| | - Teresa Giugliano
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania “Luigi Vanvitelli”, Naples, Italy
| | - Markus Kraemer
- Department of Neurology, Alfried Krupp Hospital, Essen, Germany
- Department of Neurology, Heinrich-Heine-University, Medical Faculty, Düsseldorf, Germany
| | - Annalaura Torella
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania “Luigi Vanvitelli”, Naples, Italy
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
| | | | - Mario Cirillo
- Dipartimento di Scienze Mediche, Chirurgiche, Neurologiche, Metaboliche e dell’Invecchiamento, Università degli Studi della Campania “Luigi Vanvitelli”, Naples, Italy
| | - Daniela Melis
- Dipartimento di Pediatria, Università degli Studi di Napoli "Federico II", Naples, Italy
| | - Peter Berlit
- Department of Neurology, Alfried Krupp Hospital, Essen, Germany
| | - Vincenzo Nigro
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania “Luigi Vanvitelli”, Naples, Italy
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
| | - Silverio Perrotta
- Dipartimento della Donna, del Bambino e di Chirurgia Generale e Specialistica, Università degli Studi della Campania “Luigi Vanvitelli”, Naples, Italy
| | - Giulio Piluso
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania “Luigi Vanvitelli”, Naples, Italy
- * E-mail:
| |
Collapse
|
28
|
Warejko JK, Schueler M, Vivante A, Tan W, Daga A, Lawson JA, Braun DA, Shril S, Amann K, Somers MJG, Rodig NM, Baum MA, Daouk G, Traum AZ, Kim HB, Vakili K, Porras D, Lock J, Rivkin MJ, Chaudry G, Smoot LB, Singh MN, Smith ER, Mane SM, Lifton RP, Stein DR, Ferguson MA, Hildebrandt F. Whole Exome Sequencing Reveals a Monogenic Cause of Disease in ≈43% of 35 Families With Midaortic Syndrome. Hypertension 2018; 71:691-699. [PMID: 29483232 PMCID: PMC5843550 DOI: 10.1161/hypertensionaha.117.10296] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 10/06/2017] [Accepted: 01/18/2018] [Indexed: 11/16/2022]
Abstract
Midaortic syndrome (MAS) is a rare cause of severe childhood hypertension characterized by narrowing of the abdominal aorta in children and is associated with extensive vascular disease. It may occur as part of a genetic syndrome, such as neurofibromatosis, or as consequence of a pathological inflammatory disease. However, most cases are considered idiopathic. We hypothesized that in a high percentage of these patients, a monogenic cause of disease may be detected by evaluating whole exome sequencing data for mutations in 1 of 38 candidate genes previously described to cause vasculopathy. We studied a cohort of 36 individuals from 35 different families with MAS by exome sequencing. In 15 of 35 families (42.9%), we detected likely causal dominant mutations. In 15 of 35 (42.9%) families with MAS, whole exome sequencing revealed a mutation in one of the genes previously associated with vascular disease (NF1, JAG1, ELN, GATA6, and RNF213). Ten of the 15 mutations have not previously been reported. This is the first report of ELN, RNF213, or GATA6 mutations in individuals with MAS. Mutations were detected in NF1 (6/15 families), JAG1 (4/15 families), ELN (3/15 families), and one family each for GATA6 and RNF213 Eight individuals had syndromic disease and 7 individuals had isolated MAS. Whole exome sequencing can provide conclusive molecular genetic diagnosis in a high fraction of individuals with syndromic or isolated MAS. Establishing an etiologic diagnosis may reveal genotype/phenotype correlations for MAS in the future and should, therefore, be performed routinely in MAS.
Collapse
Affiliation(s)
- Jillian K Warejko
- From the Department of Medicine (J.K.W., M.S., A.V., W.T., A.D., J.A.L., D.A.B., S.S., K.A., M.J.G.S., N.M.R., M.A.B., G.D., A.Z.T., D.R.S., M.A.F., F.H.), Department of Surgery (H.B.K., K.V.), Department of Cardiology (D.P., J.L., L.B.S., M.N.S.), Department of Neurology (M.J.R.), Department of Radiology (G.C.), and Department of Neurosurgery (E.R.S.), Boston Children's Hospital, Harvard Medical School, MA; Department of Pediatrics, Yale-New Haven Children's Hospital (J.K.W.) and Department of Genetics (S.M.M., R.P.L.), Yale School of Medicine, CT; Talpiot Medical Leadership Program, Sheba Medical Center, Tel-Hashomer, Israel (A.V.); and Laboratory of Human Genetics and Genomics, The Rockefeller University, New York (R.P.L.)
| | - Markus Schueler
- From the Department of Medicine (J.K.W., M.S., A.V., W.T., A.D., J.A.L., D.A.B., S.S., K.A., M.J.G.S., N.M.R., M.A.B., G.D., A.Z.T., D.R.S., M.A.F., F.H.), Department of Surgery (H.B.K., K.V.), Department of Cardiology (D.P., J.L., L.B.S., M.N.S.), Department of Neurology (M.J.R.), Department of Radiology (G.C.), and Department of Neurosurgery (E.R.S.), Boston Children's Hospital, Harvard Medical School, MA; Department of Pediatrics, Yale-New Haven Children's Hospital (J.K.W.) and Department of Genetics (S.M.M., R.P.L.), Yale School of Medicine, CT; Talpiot Medical Leadership Program, Sheba Medical Center, Tel-Hashomer, Israel (A.V.); and Laboratory of Human Genetics and Genomics, The Rockefeller University, New York (R.P.L.)
| | - Asaf Vivante
- From the Department of Medicine (J.K.W., M.S., A.V., W.T., A.D., J.A.L., D.A.B., S.S., K.A., M.J.G.S., N.M.R., M.A.B., G.D., A.Z.T., D.R.S., M.A.F., F.H.), Department of Surgery (H.B.K., K.V.), Department of Cardiology (D.P., J.L., L.B.S., M.N.S.), Department of Neurology (M.J.R.), Department of Radiology (G.C.), and Department of Neurosurgery (E.R.S.), Boston Children's Hospital, Harvard Medical School, MA; Department of Pediatrics, Yale-New Haven Children's Hospital (J.K.W.) and Department of Genetics (S.M.M., R.P.L.), Yale School of Medicine, CT; Talpiot Medical Leadership Program, Sheba Medical Center, Tel-Hashomer, Israel (A.V.); and Laboratory of Human Genetics and Genomics, The Rockefeller University, New York (R.P.L.)
| | - Weizhen Tan
- From the Department of Medicine (J.K.W., M.S., A.V., W.T., A.D., J.A.L., D.A.B., S.S., K.A., M.J.G.S., N.M.R., M.A.B., G.D., A.Z.T., D.R.S., M.A.F., F.H.), Department of Surgery (H.B.K., K.V.), Department of Cardiology (D.P., J.L., L.B.S., M.N.S.), Department of Neurology (M.J.R.), Department of Radiology (G.C.), and Department of Neurosurgery (E.R.S.), Boston Children's Hospital, Harvard Medical School, MA; Department of Pediatrics, Yale-New Haven Children's Hospital (J.K.W.) and Department of Genetics (S.M.M., R.P.L.), Yale School of Medicine, CT; Talpiot Medical Leadership Program, Sheba Medical Center, Tel-Hashomer, Israel (A.V.); and Laboratory of Human Genetics and Genomics, The Rockefeller University, New York (R.P.L.)
| | - Ankana Daga
- From the Department of Medicine (J.K.W., M.S., A.V., W.T., A.D., J.A.L., D.A.B., S.S., K.A., M.J.G.S., N.M.R., M.A.B., G.D., A.Z.T., D.R.S., M.A.F., F.H.), Department of Surgery (H.B.K., K.V.), Department of Cardiology (D.P., J.L., L.B.S., M.N.S.), Department of Neurology (M.J.R.), Department of Radiology (G.C.), and Department of Neurosurgery (E.R.S.), Boston Children's Hospital, Harvard Medical School, MA; Department of Pediatrics, Yale-New Haven Children's Hospital (J.K.W.) and Department of Genetics (S.M.M., R.P.L.), Yale School of Medicine, CT; Talpiot Medical Leadership Program, Sheba Medical Center, Tel-Hashomer, Israel (A.V.); and Laboratory of Human Genetics and Genomics, The Rockefeller University, New York (R.P.L.)
| | - Jennifer A Lawson
- From the Department of Medicine (J.K.W., M.S., A.V., W.T., A.D., J.A.L., D.A.B., S.S., K.A., M.J.G.S., N.M.R., M.A.B., G.D., A.Z.T., D.R.S., M.A.F., F.H.), Department of Surgery (H.B.K., K.V.), Department of Cardiology (D.P., J.L., L.B.S., M.N.S.), Department of Neurology (M.J.R.), Department of Radiology (G.C.), and Department of Neurosurgery (E.R.S.), Boston Children's Hospital, Harvard Medical School, MA; Department of Pediatrics, Yale-New Haven Children's Hospital (J.K.W.) and Department of Genetics (S.M.M., R.P.L.), Yale School of Medicine, CT; Talpiot Medical Leadership Program, Sheba Medical Center, Tel-Hashomer, Israel (A.V.); and Laboratory of Human Genetics and Genomics, The Rockefeller University, New York (R.P.L.)
| | - Daniela A Braun
- From the Department of Medicine (J.K.W., M.S., A.V., W.T., A.D., J.A.L., D.A.B., S.S., K.A., M.J.G.S., N.M.R., M.A.B., G.D., A.Z.T., D.R.S., M.A.F., F.H.), Department of Surgery (H.B.K., K.V.), Department of Cardiology (D.P., J.L., L.B.S., M.N.S.), Department of Neurology (M.J.R.), Department of Radiology (G.C.), and Department of Neurosurgery (E.R.S.), Boston Children's Hospital, Harvard Medical School, MA; Department of Pediatrics, Yale-New Haven Children's Hospital (J.K.W.) and Department of Genetics (S.M.M., R.P.L.), Yale School of Medicine, CT; Talpiot Medical Leadership Program, Sheba Medical Center, Tel-Hashomer, Israel (A.V.); and Laboratory of Human Genetics and Genomics, The Rockefeller University, New York (R.P.L.)
| | - Shirlee Shril
- From the Department of Medicine (J.K.W., M.S., A.V., W.T., A.D., J.A.L., D.A.B., S.S., K.A., M.J.G.S., N.M.R., M.A.B., G.D., A.Z.T., D.R.S., M.A.F., F.H.), Department of Surgery (H.B.K., K.V.), Department of Cardiology (D.P., J.L., L.B.S., M.N.S.), Department of Neurology (M.J.R.), Department of Radiology (G.C.), and Department of Neurosurgery (E.R.S.), Boston Children's Hospital, Harvard Medical School, MA; Department of Pediatrics, Yale-New Haven Children's Hospital (J.K.W.) and Department of Genetics (S.M.M., R.P.L.), Yale School of Medicine, CT; Talpiot Medical Leadership Program, Sheba Medical Center, Tel-Hashomer, Israel (A.V.); and Laboratory of Human Genetics and Genomics, The Rockefeller University, New York (R.P.L.)
| | - Kassaundra Amann
- From the Department of Medicine (J.K.W., M.S., A.V., W.T., A.D., J.A.L., D.A.B., S.S., K.A., M.J.G.S., N.M.R., M.A.B., G.D., A.Z.T., D.R.S., M.A.F., F.H.), Department of Surgery (H.B.K., K.V.), Department of Cardiology (D.P., J.L., L.B.S., M.N.S.), Department of Neurology (M.J.R.), Department of Radiology (G.C.), and Department of Neurosurgery (E.R.S.), Boston Children's Hospital, Harvard Medical School, MA; Department of Pediatrics, Yale-New Haven Children's Hospital (J.K.W.) and Department of Genetics (S.M.M., R.P.L.), Yale School of Medicine, CT; Talpiot Medical Leadership Program, Sheba Medical Center, Tel-Hashomer, Israel (A.V.); and Laboratory of Human Genetics and Genomics, The Rockefeller University, New York (R.P.L.)
| | - Michael J G Somers
- From the Department of Medicine (J.K.W., M.S., A.V., W.T., A.D., J.A.L., D.A.B., S.S., K.A., M.J.G.S., N.M.R., M.A.B., G.D., A.Z.T., D.R.S., M.A.F., F.H.), Department of Surgery (H.B.K., K.V.), Department of Cardiology (D.P., J.L., L.B.S., M.N.S.), Department of Neurology (M.J.R.), Department of Radiology (G.C.), and Department of Neurosurgery (E.R.S.), Boston Children's Hospital, Harvard Medical School, MA; Department of Pediatrics, Yale-New Haven Children's Hospital (J.K.W.) and Department of Genetics (S.M.M., R.P.L.), Yale School of Medicine, CT; Talpiot Medical Leadership Program, Sheba Medical Center, Tel-Hashomer, Israel (A.V.); and Laboratory of Human Genetics and Genomics, The Rockefeller University, New York (R.P.L.)
| | - Nancy M Rodig
- From the Department of Medicine (J.K.W., M.S., A.V., W.T., A.D., J.A.L., D.A.B., S.S., K.A., M.J.G.S., N.M.R., M.A.B., G.D., A.Z.T., D.R.S., M.A.F., F.H.), Department of Surgery (H.B.K., K.V.), Department of Cardiology (D.P., J.L., L.B.S., M.N.S.), Department of Neurology (M.J.R.), Department of Radiology (G.C.), and Department of Neurosurgery (E.R.S.), Boston Children's Hospital, Harvard Medical School, MA; Department of Pediatrics, Yale-New Haven Children's Hospital (J.K.W.) and Department of Genetics (S.M.M., R.P.L.), Yale School of Medicine, CT; Talpiot Medical Leadership Program, Sheba Medical Center, Tel-Hashomer, Israel (A.V.); and Laboratory of Human Genetics and Genomics, The Rockefeller University, New York (R.P.L.)
| | - Michelle A Baum
- From the Department of Medicine (J.K.W., M.S., A.V., W.T., A.D., J.A.L., D.A.B., S.S., K.A., M.J.G.S., N.M.R., M.A.B., G.D., A.Z.T., D.R.S., M.A.F., F.H.), Department of Surgery (H.B.K., K.V.), Department of Cardiology (D.P., J.L., L.B.S., M.N.S.), Department of Neurology (M.J.R.), Department of Radiology (G.C.), and Department of Neurosurgery (E.R.S.), Boston Children's Hospital, Harvard Medical School, MA; Department of Pediatrics, Yale-New Haven Children's Hospital (J.K.W.) and Department of Genetics (S.M.M., R.P.L.), Yale School of Medicine, CT; Talpiot Medical Leadership Program, Sheba Medical Center, Tel-Hashomer, Israel (A.V.); and Laboratory of Human Genetics and Genomics, The Rockefeller University, New York (R.P.L.)
| | - Ghaleb Daouk
- From the Department of Medicine (J.K.W., M.S., A.V., W.T., A.D., J.A.L., D.A.B., S.S., K.A., M.J.G.S., N.M.R., M.A.B., G.D., A.Z.T., D.R.S., M.A.F., F.H.), Department of Surgery (H.B.K., K.V.), Department of Cardiology (D.P., J.L., L.B.S., M.N.S.), Department of Neurology (M.J.R.), Department of Radiology (G.C.), and Department of Neurosurgery (E.R.S.), Boston Children's Hospital, Harvard Medical School, MA; Department of Pediatrics, Yale-New Haven Children's Hospital (J.K.W.) and Department of Genetics (S.M.M., R.P.L.), Yale School of Medicine, CT; Talpiot Medical Leadership Program, Sheba Medical Center, Tel-Hashomer, Israel (A.V.); and Laboratory of Human Genetics and Genomics, The Rockefeller University, New York (R.P.L.)
| | - Avram Z Traum
- From the Department of Medicine (J.K.W., M.S., A.V., W.T., A.D., J.A.L., D.A.B., S.S., K.A., M.J.G.S., N.M.R., M.A.B., G.D., A.Z.T., D.R.S., M.A.F., F.H.), Department of Surgery (H.B.K., K.V.), Department of Cardiology (D.P., J.L., L.B.S., M.N.S.), Department of Neurology (M.J.R.), Department of Radiology (G.C.), and Department of Neurosurgery (E.R.S.), Boston Children's Hospital, Harvard Medical School, MA; Department of Pediatrics, Yale-New Haven Children's Hospital (J.K.W.) and Department of Genetics (S.M.M., R.P.L.), Yale School of Medicine, CT; Talpiot Medical Leadership Program, Sheba Medical Center, Tel-Hashomer, Israel (A.V.); and Laboratory of Human Genetics and Genomics, The Rockefeller University, New York (R.P.L.)
| | - Heung Bae Kim
- From the Department of Medicine (J.K.W., M.S., A.V., W.T., A.D., J.A.L., D.A.B., S.S., K.A., M.J.G.S., N.M.R., M.A.B., G.D., A.Z.T., D.R.S., M.A.F., F.H.), Department of Surgery (H.B.K., K.V.), Department of Cardiology (D.P., J.L., L.B.S., M.N.S.), Department of Neurology (M.J.R.), Department of Radiology (G.C.), and Department of Neurosurgery (E.R.S.), Boston Children's Hospital, Harvard Medical School, MA; Department of Pediatrics, Yale-New Haven Children's Hospital (J.K.W.) and Department of Genetics (S.M.M., R.P.L.), Yale School of Medicine, CT; Talpiot Medical Leadership Program, Sheba Medical Center, Tel-Hashomer, Israel (A.V.); and Laboratory of Human Genetics and Genomics, The Rockefeller University, New York (R.P.L.)
| | - Khashayar Vakili
- From the Department of Medicine (J.K.W., M.S., A.V., W.T., A.D., J.A.L., D.A.B., S.S., K.A., M.J.G.S., N.M.R., M.A.B., G.D., A.Z.T., D.R.S., M.A.F., F.H.), Department of Surgery (H.B.K., K.V.), Department of Cardiology (D.P., J.L., L.B.S., M.N.S.), Department of Neurology (M.J.R.), Department of Radiology (G.C.), and Department of Neurosurgery (E.R.S.), Boston Children's Hospital, Harvard Medical School, MA; Department of Pediatrics, Yale-New Haven Children's Hospital (J.K.W.) and Department of Genetics (S.M.M., R.P.L.), Yale School of Medicine, CT; Talpiot Medical Leadership Program, Sheba Medical Center, Tel-Hashomer, Israel (A.V.); and Laboratory of Human Genetics and Genomics, The Rockefeller University, New York (R.P.L.)
| | - Diego Porras
- From the Department of Medicine (J.K.W., M.S., A.V., W.T., A.D., J.A.L., D.A.B., S.S., K.A., M.J.G.S., N.M.R., M.A.B., G.D., A.Z.T., D.R.S., M.A.F., F.H.), Department of Surgery (H.B.K., K.V.), Department of Cardiology (D.P., J.L., L.B.S., M.N.S.), Department of Neurology (M.J.R.), Department of Radiology (G.C.), and Department of Neurosurgery (E.R.S.), Boston Children's Hospital, Harvard Medical School, MA; Department of Pediatrics, Yale-New Haven Children's Hospital (J.K.W.) and Department of Genetics (S.M.M., R.P.L.), Yale School of Medicine, CT; Talpiot Medical Leadership Program, Sheba Medical Center, Tel-Hashomer, Israel (A.V.); and Laboratory of Human Genetics and Genomics, The Rockefeller University, New York (R.P.L.)
| | - James Lock
- From the Department of Medicine (J.K.W., M.S., A.V., W.T., A.D., J.A.L., D.A.B., S.S., K.A., M.J.G.S., N.M.R., M.A.B., G.D., A.Z.T., D.R.S., M.A.F., F.H.), Department of Surgery (H.B.K., K.V.), Department of Cardiology (D.P., J.L., L.B.S., M.N.S.), Department of Neurology (M.J.R.), Department of Radiology (G.C.), and Department of Neurosurgery (E.R.S.), Boston Children's Hospital, Harvard Medical School, MA; Department of Pediatrics, Yale-New Haven Children's Hospital (J.K.W.) and Department of Genetics (S.M.M., R.P.L.), Yale School of Medicine, CT; Talpiot Medical Leadership Program, Sheba Medical Center, Tel-Hashomer, Israel (A.V.); and Laboratory of Human Genetics and Genomics, The Rockefeller University, New York (R.P.L.)
| | - Michael J Rivkin
- From the Department of Medicine (J.K.W., M.S., A.V., W.T., A.D., J.A.L., D.A.B., S.S., K.A., M.J.G.S., N.M.R., M.A.B., G.D., A.Z.T., D.R.S., M.A.F., F.H.), Department of Surgery (H.B.K., K.V.), Department of Cardiology (D.P., J.L., L.B.S., M.N.S.), Department of Neurology (M.J.R.), Department of Radiology (G.C.), and Department of Neurosurgery (E.R.S.), Boston Children's Hospital, Harvard Medical School, MA; Department of Pediatrics, Yale-New Haven Children's Hospital (J.K.W.) and Department of Genetics (S.M.M., R.P.L.), Yale School of Medicine, CT; Talpiot Medical Leadership Program, Sheba Medical Center, Tel-Hashomer, Israel (A.V.); and Laboratory of Human Genetics and Genomics, The Rockefeller University, New York (R.P.L.)
| | - Gulraiz Chaudry
- From the Department of Medicine (J.K.W., M.S., A.V., W.T., A.D., J.A.L., D.A.B., S.S., K.A., M.J.G.S., N.M.R., M.A.B., G.D., A.Z.T., D.R.S., M.A.F., F.H.), Department of Surgery (H.B.K., K.V.), Department of Cardiology (D.P., J.L., L.B.S., M.N.S.), Department of Neurology (M.J.R.), Department of Radiology (G.C.), and Department of Neurosurgery (E.R.S.), Boston Children's Hospital, Harvard Medical School, MA; Department of Pediatrics, Yale-New Haven Children's Hospital (J.K.W.) and Department of Genetics (S.M.M., R.P.L.), Yale School of Medicine, CT; Talpiot Medical Leadership Program, Sheba Medical Center, Tel-Hashomer, Israel (A.V.); and Laboratory of Human Genetics and Genomics, The Rockefeller University, New York (R.P.L.)
| | - Leslie B Smoot
- From the Department of Medicine (J.K.W., M.S., A.V., W.T., A.D., J.A.L., D.A.B., S.S., K.A., M.J.G.S., N.M.R., M.A.B., G.D., A.Z.T., D.R.S., M.A.F., F.H.), Department of Surgery (H.B.K., K.V.), Department of Cardiology (D.P., J.L., L.B.S., M.N.S.), Department of Neurology (M.J.R.), Department of Radiology (G.C.), and Department of Neurosurgery (E.R.S.), Boston Children's Hospital, Harvard Medical School, MA; Department of Pediatrics, Yale-New Haven Children's Hospital (J.K.W.) and Department of Genetics (S.M.M., R.P.L.), Yale School of Medicine, CT; Talpiot Medical Leadership Program, Sheba Medical Center, Tel-Hashomer, Israel (A.V.); and Laboratory of Human Genetics and Genomics, The Rockefeller University, New York (R.P.L.)
| | - Michael N Singh
- From the Department of Medicine (J.K.W., M.S., A.V., W.T., A.D., J.A.L., D.A.B., S.S., K.A., M.J.G.S., N.M.R., M.A.B., G.D., A.Z.T., D.R.S., M.A.F., F.H.), Department of Surgery (H.B.K., K.V.), Department of Cardiology (D.P., J.L., L.B.S., M.N.S.), Department of Neurology (M.J.R.), Department of Radiology (G.C.), and Department of Neurosurgery (E.R.S.), Boston Children's Hospital, Harvard Medical School, MA; Department of Pediatrics, Yale-New Haven Children's Hospital (J.K.W.) and Department of Genetics (S.M.M., R.P.L.), Yale School of Medicine, CT; Talpiot Medical Leadership Program, Sheba Medical Center, Tel-Hashomer, Israel (A.V.); and Laboratory of Human Genetics and Genomics, The Rockefeller University, New York (R.P.L.)
| | - Edward R Smith
- From the Department of Medicine (J.K.W., M.S., A.V., W.T., A.D., J.A.L., D.A.B., S.S., K.A., M.J.G.S., N.M.R., M.A.B., G.D., A.Z.T., D.R.S., M.A.F., F.H.), Department of Surgery (H.B.K., K.V.), Department of Cardiology (D.P., J.L., L.B.S., M.N.S.), Department of Neurology (M.J.R.), Department of Radiology (G.C.), and Department of Neurosurgery (E.R.S.), Boston Children's Hospital, Harvard Medical School, MA; Department of Pediatrics, Yale-New Haven Children's Hospital (J.K.W.) and Department of Genetics (S.M.M., R.P.L.), Yale School of Medicine, CT; Talpiot Medical Leadership Program, Sheba Medical Center, Tel-Hashomer, Israel (A.V.); and Laboratory of Human Genetics and Genomics, The Rockefeller University, New York (R.P.L.)
| | - Shrikant M Mane
- From the Department of Medicine (J.K.W., M.S., A.V., W.T., A.D., J.A.L., D.A.B., S.S., K.A., M.J.G.S., N.M.R., M.A.B., G.D., A.Z.T., D.R.S., M.A.F., F.H.), Department of Surgery (H.B.K., K.V.), Department of Cardiology (D.P., J.L., L.B.S., M.N.S.), Department of Neurology (M.J.R.), Department of Radiology (G.C.), and Department of Neurosurgery (E.R.S.), Boston Children's Hospital, Harvard Medical School, MA; Department of Pediatrics, Yale-New Haven Children's Hospital (J.K.W.) and Department of Genetics (S.M.M., R.P.L.), Yale School of Medicine, CT; Talpiot Medical Leadership Program, Sheba Medical Center, Tel-Hashomer, Israel (A.V.); and Laboratory of Human Genetics and Genomics, The Rockefeller University, New York (R.P.L.)
| | - Richard P Lifton
- From the Department of Medicine (J.K.W., M.S., A.V., W.T., A.D., J.A.L., D.A.B., S.S., K.A., M.J.G.S., N.M.R., M.A.B., G.D., A.Z.T., D.R.S., M.A.F., F.H.), Department of Surgery (H.B.K., K.V.), Department of Cardiology (D.P., J.L., L.B.S., M.N.S.), Department of Neurology (M.J.R.), Department of Radiology (G.C.), and Department of Neurosurgery (E.R.S.), Boston Children's Hospital, Harvard Medical School, MA; Department of Pediatrics, Yale-New Haven Children's Hospital (J.K.W.) and Department of Genetics (S.M.M., R.P.L.), Yale School of Medicine, CT; Talpiot Medical Leadership Program, Sheba Medical Center, Tel-Hashomer, Israel (A.V.); and Laboratory of Human Genetics and Genomics, The Rockefeller University, New York (R.P.L.)
| | - Deborah R Stein
- From the Department of Medicine (J.K.W., M.S., A.V., W.T., A.D., J.A.L., D.A.B., S.S., K.A., M.J.G.S., N.M.R., M.A.B., G.D., A.Z.T., D.R.S., M.A.F., F.H.), Department of Surgery (H.B.K., K.V.), Department of Cardiology (D.P., J.L., L.B.S., M.N.S.), Department of Neurology (M.J.R.), Department of Radiology (G.C.), and Department of Neurosurgery (E.R.S.), Boston Children's Hospital, Harvard Medical School, MA; Department of Pediatrics, Yale-New Haven Children's Hospital (J.K.W.) and Department of Genetics (S.M.M., R.P.L.), Yale School of Medicine, CT; Talpiot Medical Leadership Program, Sheba Medical Center, Tel-Hashomer, Israel (A.V.); and Laboratory of Human Genetics and Genomics, The Rockefeller University, New York (R.P.L.)
| | - Michael A Ferguson
- From the Department of Medicine (J.K.W., M.S., A.V., W.T., A.D., J.A.L., D.A.B., S.S., K.A., M.J.G.S., N.M.R., M.A.B., G.D., A.Z.T., D.R.S., M.A.F., F.H.), Department of Surgery (H.B.K., K.V.), Department of Cardiology (D.P., J.L., L.B.S., M.N.S.), Department of Neurology (M.J.R.), Department of Radiology (G.C.), and Department of Neurosurgery (E.R.S.), Boston Children's Hospital, Harvard Medical School, MA; Department of Pediatrics, Yale-New Haven Children's Hospital (J.K.W.) and Department of Genetics (S.M.M., R.P.L.), Yale School of Medicine, CT; Talpiot Medical Leadership Program, Sheba Medical Center, Tel-Hashomer, Israel (A.V.); and Laboratory of Human Genetics and Genomics, The Rockefeller University, New York (R.P.L.)
| | - Friedhelm Hildebrandt
- From the Department of Medicine (J.K.W., M.S., A.V., W.T., A.D., J.A.L., D.A.B., S.S., K.A., M.J.G.S., N.M.R., M.A.B., G.D., A.Z.T., D.R.S., M.A.F., F.H.), Department of Surgery (H.B.K., K.V.), Department of Cardiology (D.P., J.L., L.B.S., M.N.S.), Department of Neurology (M.J.R.), Department of Radiology (G.C.), and Department of Neurosurgery (E.R.S.), Boston Children's Hospital, Harvard Medical School, MA; Department of Pediatrics, Yale-New Haven Children's Hospital (J.K.W.) and Department of Genetics (S.M.M., R.P.L.), Yale School of Medicine, CT; Talpiot Medical Leadership Program, Sheba Medical Center, Tel-Hashomer, Israel (A.V.); and Laboratory of Human Genetics and Genomics, The Rockefeller University, New York (R.P.L.).
| |
Collapse
|
29
|
Santoro C, Bernardo P, Coppola A, Pugliese U, Cirillo M, Giugliano T, Piluso G, Cinalli G, Striano S, Bravaccio C, Perrotta S. Seizures in children with neurofibromatosis type 1: is neurofibromatosis type 1 enough? Ital J Pediatr 2018; 44:41. [PMID: 29566708 PMCID: PMC5863905 DOI: 10.1186/s13052-018-0477-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 03/09/2018] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Neurofibromatosis type 1 (NF1) is related to a generally increased prevalence of seizures. The mechanism underlying the increased predisposition to seizures has not been fully elucidated. The aim of the study was to evaluate the role of NF1 in seizures pathogenesis in a cohort of children with NF1 and seizures. METHODS The medical records of 437 children (0-18 years old) with NF1 were reviewed. All children with at least one afebrile seizure were included. Demographic, clinical, neurological, NF1 mutation status, and EEG data were collected along with brain magnetic resonance imaging. Depending on etiology, structural seizures have been identified and were further classified as NF1 related or not. RESULTS Nineteen patients (4.3%; 13 males) were included. NF1 was inherited in 7 (37.5%), with 3 maternal forms. Ten children with structural seizures were identified. Seven forms were identified someway related to NF1, two of which were associated to 17q11.2 microdeletion and hypoxic-ischemic encephalopathy. Any brain lesion that could explain seizures was found in nine patients, two third of these patients had a familiar history of epilepsy. CONCLUSIONS Our results suggest seizures are more frequent in NF1 children (4.3%) than in general pediatric population (0.3-0.5%) and that are someway related to NF1 in half of patients. Facing seizures in NF1, the clinician should first exclude brain tumors but also other, and rarer NF1-related scenarios, such as hydrocephalous and vasculopathies. Children with non-structural seizures frequently had a family history of epilepsy, raising questions about the pathogenic role of NF1. They should be approached as for the general population.
Collapse
Affiliation(s)
- Claudia Santoro
- Centro di Riferimento Pediatrico delle Neurofibromatosi, Dipartimento della Donna, del Bambino e di Chirurgia Generale e Specialistica, Università degli Studi della Campania "L. Vanvitelli", Caserta, Italy.
| | - Pia Bernardo
- Dipartimento di Scienze Mediche Traslazionali, Università "Federico II", Napoli, Italy.,Centro Epilessia. Dipartimento di Neuroscienze, Scienze Riproduttive ed Odontostomatologiche, Università Federico II, Naples, Italy
| | - Antonietta Coppola
- Centro Epilessia. Dipartimento di Neuroscienze, Scienze Riproduttive ed Odontostomatologiche, Università Federico II, Naples, Italy
| | - Umberto Pugliese
- Centro di Riferimento Pediatrico delle Neurofibromatosi, Dipartimento della Donna, del Bambino e di Chirurgia Generale e Specialistica, Università degli Studi della Campania "L. Vanvitelli", Caserta, Italy
| | - Mario Cirillo
- Dipartimento di Scienze Mediche, Chirurgiche, Neurologiche, Metaboliche e dell'invecchiamento, Università degli Studi della Campania "Luigi Vanvitelli", Caserta, Italy
| | - Teresa Giugliano
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania "Luigi Vanvitelli", Caserta, Italy
| | - Giulio Piluso
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania "Luigi Vanvitelli", Caserta, Italy
| | - Giuseppe Cinalli
- Dipartimento di Neurochirurgia, Santobono-Pausilipon Children's Hospital, Naples, Italy
| | - Salvatore Striano
- Centro Epilessia. Dipartimento di Neuroscienze, Scienze Riproduttive ed Odontostomatologiche, Università Federico II, Naples, Italy
| | - Carmela Bravaccio
- Dipartimento di Scienze Mediche Traslazionali, Università "Federico II", Napoli, Italy
| | - Silverio Perrotta
- Centro di Riferimento Pediatrico delle Neurofibromatosi, Dipartimento della Donna, del Bambino e di Chirurgia Generale e Specialistica, Università degli Studi della Campania "L. Vanvitelli", Caserta, Italy
| |
Collapse
|
30
|
Hori YS, Ebisudani Y, Aoi M, Fukuhara T. Adult-Onset Hemorrhagic Quasi-Moyamoya Disease with Unilateral Steno-occlusive Lesion in a Patient with Neurofibromatosis Type 1. J Stroke Cerebrovasc Dis 2018; 27:1423-1424. [PMID: 29305273 DOI: 10.1016/j.jstrokecerebrovasdis.2017.11.025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 10/26/2017] [Accepted: 11/19/2017] [Indexed: 10/18/2022] Open
Abstract
BACKGROUND Quasi-moyamoya disease is a condition that occurs in association with a specific underlying condition or disease such as atherosclerotic disease or neurofibromatosis type 1 (NF1). Pediatric cases are frequently reported, and an ischemic and bilateral presentation is more common than a hemorrhagic and unilateral presentation. CLINICAL PRESENTATION A 39-year-old woman previously diagnosed with NF1 presented to our department with nausea and left hemiparesis. She was diagnosed with right temporal intracerebral hemorrhage by initial computed tomography. Subsequent angiography showed an occlusion of the terminal portion of the right internal carotid artery, and magnetic resonance imaging showed multiple flow voids in the right basal ganglia, suggesting quasi-moyamoya disease. The hematoma was surgically removed, and her neurological condition improved after the operation. CONCLUSIONS This is the first reported case of quasi-moyamoya disease with a rare combination of characteristics, including an adult-onset, hemorrhagic presentation and a unilateral lesion in a patient previously diagnosed with NF1.
Collapse
Affiliation(s)
- Yusuke S Hori
- Department of Neurological Surgery, National Hospital Organization Okayama Medical Center, Okayama, Japan.
| | - Yuki Ebisudani
- Department of Neurological Surgery, National Hospital Organization Okayama Medical Center, Okayama, Japan
| | - Mizuho Aoi
- Department of Neurological Surgery, National Hospital Organization Okayama Medical Center, Okayama, Japan
| | - Toru Fukuhara
- Department of Neurological Surgery, National Hospital Organization Okayama Medical Center, Okayama, Japan
| |
Collapse
|
31
|
Juvenile Moyamoya and Craniosynostosis in a Child with Deletion 1p32p31: Expanding the Clinical Spectrum of 1p32p31 Deletion Syndrome and a Review of the Literature. Int J Mol Sci 2017; 18:ijms18091998. [PMID: 28926972 PMCID: PMC5618647 DOI: 10.3390/ijms18091998] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 09/11/2017] [Accepted: 09/13/2017] [Indexed: 11/29/2022] Open
Abstract
Moyamoya angiopathy (MA) is a rare cerebrovascular disorder characterised by the progressive occlusion of the internal carotid artery. Its aetiology is uncertain, but a genetic background seems likely, given the high MA familial rate. To investigate the aetiology of craniosynostosis and juvenile moyamoya in a 14-year-old male patient, we performed an array-comparative genomic hybridisation revealing a de novo interstitial deletion of 8.5 Mb in chromosome region 1p32p31. The deletion involved 34 protein coding genes, including NF1A, whose haploinsufficiency is indicated as being mainly responsible for the 1p32-p31 chromosome deletion syndrome phenotype (OMIM 613735). Our patient also has a deleted FOXD3 of the FOX gene family of transcription factors, which plays an important role in neural crest cell growth and differentiation. As the murine FOXD3−/− model shows craniofacial anomalies and abnormal common carotid artery morphology, it can be hypothesised that FOXD3 is involved in the pathogenesis of the craniofacial and vascular defects observed in our patient. In support of our assumption, we found in the literature another patient with a syndromic form of MA who had a deletion involving another FOX gene (FOXC1). In addition to describing the clinical history of our patient, we have reviewed all of the available literature concerning other patients with a 1p32p31 deletion, including cases from the Decipher database, and we have also reviewed the genetic disorders associated with MA, which is a useful guide for the diagnosis of syndromic form of MA.
Collapse
|