Neurofibromatosis 1 Genetics

A special issue of Genes (ISSN 2073-4425). This special issue belongs to the section "Human Genomics and Genetic Diseases".

Deadline for manuscript submissions: closed (28 June 2019) | Viewed by 26546

Special Issue Editor


E-Mail Website
Guest Editor
1. INSERM U1016, Cochin Institute, CARPEM, Paris Descartes University, Sorbonne Paris Cité, 75006 Paris, France
2. Department of Molecular Genetics, Cochin Hospital, AP-HP, 75006 Paris, France
Interests: neurofibromatosis; cancer; molecular genetics; NGS; RAS-MAPK pathway
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues,

Neurofibromatosis type 1 (NF1) is one of the most common autosomal dominant tumor predisposition syndromes. Clinical diagnosis of this condition relies on specific diagnosis criteria outlined in the National Institutes of Health consensus development conference in 1987. The broad phenotypic spectrum and age-dependent symptoms make clinical diagnosis challenging, particularly in young individuals. NF1 is caused by heterozygous loss-of-function mutations of the tumor suppressor NF1 (Neurofibromin 1). The mutation rate at the NF1 locus is one of the highest reported in any human disorder. This observation is reflected in the finding that almost 50% of all NF1 patients exhibit a de novo NF1 mutation. A huge number of different pathogenic NF1 mutations have been reported. The absence of genotype–phenotype correlation demonstrates the minor role of the NF1 gene in the great variability of NF1 clinical expression, with the rare exceptions of (i) missense mutations in codons 844-848 and Arg1809, one short in-frame deletion (p.Met992del), and (ii) the recurrent large deletions of the NF1 locus. Evidence of modifier genes was provided by animal models and intrafamilial phenotypic correlations. Genetic heterogeneity in NF1 has been evidenced by the identification of pathogenic mutations in the SPRED1 gene causing an NF1 phenocopy, the so-called Legius syndrome. NF1 mutation detection and interpretation are challenging owing to the large size of the gene, the existence of multiple highly identical pseudogenes, the lack of mutational hotspots, and the complex mutational spectrum. Targeted next-generation sequencing (NGS) can be applied to the accurate identification of NF1 and SPRED1 variants and copy number alterations (CNAs), and to establish the unambiguous diagnosis of NF1 or Legius syndrome, particularly in mosaicism presentations of low-frequency mutations, and in uncertain clinical presentations.

This Special Issue of Genes dedicated to neurofibromatosis type 1 genetics welcomes reviews and original papers covering recent genetic research on NF1 molecular diagnosis and genetic counselling, and on the effect of mutations and modifier genes on NF1-related phenotypes; case reports highlighting testing approaches that can be utilized in several clinical scenarios may also be considered

Dr. Eric Pasmant
Guest Editor

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Genes is an international peer-reviewed open access monthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2600 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • NF1
  • SPRED1
  • neurofibromatosis type 1
  • Legius syndrome
  • mosaicism
  • genotype-phenotype correlation
  • molecular diagnosis

Published Papers (7 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

16 pages, 1766 KiB  
Article
Non-Coding RNA and Tumor Development in Neurofibromatosis Type 1: ANRIL Rs2151280 Is Associated with Optic Glioma Development and a Mild Phenotype in Neurofibromatosis Type 1 Patients
by Viviana Tritto, Luca Ferrari, Silvia Esposito, Paola Zuccotti, Donatella Bianchessi, Federica Natacci, Veronica Saletti, Marica Eoli and Paola Riva
Genes 2019, 10(11), 892; https://0-doi-org.brum.beds.ac.uk/10.3390/genes10110892 - 05 Nov 2019
Cited by 14 | Viewed by 3795
Abstract
Non-coding RNAs (ncRNAs) are known to regulate gene expression at the transcriptional and post-transcriptional levels, chromatin remodeling, and signal transduction. The identification of different species of ncRNAs, microRNAs (miRNAs), circular RNAs (circRNAs), and long ncRNAs (lncRNAs)—and in some cases, their combined regulatory function [...] Read more.
Non-coding RNAs (ncRNAs) are known to regulate gene expression at the transcriptional and post-transcriptional levels, chromatin remodeling, and signal transduction. The identification of different species of ncRNAs, microRNAs (miRNAs), circular RNAs (circRNAs), and long ncRNAs (lncRNAs)—and in some cases, their combined regulatory function on specific target genes—may help to elucidate their role in biological processes. NcRNAs’ deregulation has an impact on the impairment of physiological programs, driving cells in cancer development. We here carried out a review of literature concerning the implication of ncRNAs on tumor development in neurofibromatosis type 1 (NF1), an inherited tumor predisposition syndrome. A number of miRNAs and a lncRNA has been implicated in NF1-associated tumors, such as malignant peripheral nerve sheath tumors (MPNSTs) and astrocytoma, as well as in the pathognomonic neurofibromas. Some authors reported that the lncRNA ANRIL was deregulated in the blood of NF1 patients with plexiform neurofibromas (PNFs), even if its role should be further elucidated. We here provided original data concerning the association of a specific genotype about ANRIL rs2151280 with the presence of optic gliomas and a mild expression of the NF1 phenotype. We also detected the LOH of ANRIL in different tumors from NF1 patients, supporting the involvement of ANRIL in some NF1-associated tumors. Our results suggest that ANRIL rs2151280 may be a potential diagnostic and prognostic marker, addressing early diagnosis of optic glioma and predicting the phenotype severity in NF1 patients. Full article
(This article belongs to the Special Issue Neurofibromatosis 1 Genetics)
Show Figures

Figure 1

8 pages, 694 KiB  
Article
Clinical Presentation and Novel Pathogenic Variants among 68 Chinese Neurofibromatosis 1 Children
by Ruen Yao, Tingting Yu, Yufei Xu, Li Yu, Jiwen Wang, Xiumin Wang, Jian Wang and Yiping Shen
Genes 2019, 10(11), 847; https://0-doi-org.brum.beds.ac.uk/10.3390/genes10110847 - 26 Oct 2019
Cited by 11 | Viewed by 2289
Abstract
Background: Neurofibromatosis 1 (NF1) is one of the most common dominantly inherited genetic disorders worldwide, with an age-dependent phenotypic expression. Exploring the mutational spectrum and clinical presentation of NF1 patients at different ages from a diverse population will aid the understanding of genotype–phenotype [...] Read more.
Background: Neurofibromatosis 1 (NF1) is one of the most common dominantly inherited genetic disorders worldwide, with an age-dependent phenotypic expression. Exploring the mutational spectrum and clinical presentation of NF1 patients at different ages from a diverse population will aid the understanding of genotype–phenotype correlations. Methods: In this study, 95 Chinese children with clinical suspicion of NF1 mainly due to the presence of multiple café-au-lait macules (CALMs) were subjected to medical exome-sequencing analysis and Sanger confirmation of pathogenic variants. Clinical presentations were evaluated regarding dermatological, ocular, neurological, and behavioral features. Results: Pathogenic or likely pathogenic NF1 variants were detected in 71.6% (68/95) of patients; 20 pathogenic variants were not previously reported, indicating that Chinese NF1 patients are still understudied. Parental Sanger sequencing confirmation revealed 77.9% of de novo variants, a percentage that was much higher than expected. The presence of a higher number of NF1-related features at young ages was correlated with positive diagnostic findings. In addition to CALMs, neurological and behavioral features had a high expression among Chinese NF1 children. We attempted to correlate short stature with the locations of the pathogenic variants across the NF1 gene. It is interesting to notice that variants detected in the C-terminal region of the NF1 gene were less likely to be associated with short stature among the NF1 patients, whereas variants at the N-terminal were highly penetrant for the short stature phenotype. Conclusion: Novel NF1 pathogenic variants are yet to be uncovered in under-studied NF1 patient populations; their identification will help to reveal novel genotype–phenotype correlations. Full article
(This article belongs to the Special Issue Neurofibromatosis 1 Genetics)
Show Figures

Figure 1

13 pages, 769 KiB  
Article
The Role of Co-Deleted Genes in Neurofibromatosis Type 1 Microdeletions: An Evolutive Approach
by Larissa Brussa Reis, Andreia Carina Turchetto-Zolet, Maievi Fonini, Patricia Ashton-Prolla and Clévia Rosset
Genes 2019, 10(11), 839; https://0-doi-org.brum.beds.ac.uk/10.3390/genes10110839 - 24 Oct 2019
Cited by 3 | Viewed by 2286
Abstract
Neurofibromatosis type 1 (NF1) is a cancer predisposition syndrome that results from dominant loss-of-function mutations mainly in the NF1 gene. Large rearrangements are present in 5–10% of affected patients, generally encompass NF1 neighboring genes, and are correlated with a more severe NF1 phenotype. [...] Read more.
Neurofibromatosis type 1 (NF1) is a cancer predisposition syndrome that results from dominant loss-of-function mutations mainly in the NF1 gene. Large rearrangements are present in 5–10% of affected patients, generally encompass NF1 neighboring genes, and are correlated with a more severe NF1 phenotype. Evident genotype–phenotype correlations and the importance of the co-deleted genes are difficult to establish. In our study we employed an evolutionary approach to provide further insights into the understanding of the fundamental function of genes that are co-deleted in subjects with NF1 microdeletions. Our goal was to access the ortholog and paralog relationship of these genes in primates and verify if purifying or positive selection are acting on these genes. Fourteen genes were analyzed in twelve mammalian species. Of these, four and ten genes showed positive selection and purifying selection, respectively. The protein, RNF135, showed three sites under positive selection at the RING finger domain, which may have been selected to increase efficiency in ubiquitination routes in primates. The phylogenetic analysis suggests distinct evolutionary constraint between the analyzed genes. With these analyses, we hope to help clarify the correlation of the co-deletion of these genes and the more severe phenotype of NF1. Full article
(This article belongs to the Special Issue Neurofibromatosis 1 Genetics)
Show Figures

Figure 1

14 pages, 640 KiB  
Article
Prevalence, Type, and Molecular Spectrum of NF1 Mutations in Patients with Neurofibromatosis Type 1 and Congenital Heart Disease
by Valentina Pinna, Paola Daniele, Giulio Calcagni, Lucio Mariniello, Roberta Criscione, Chiara Giardina, Francesca Romana Lepri, Hossein Hozhabri, Angela Alberico, Stefania Cavone, Annunziata Tina Morella, Roberta Mandile, Francesca Annunziata, Niccolò Di Giosaffatte, Maria Cecilia D’Asdia, Paolo Versacci, Rossella Capolino, Pietro Strisciuglio, Sandra Giustini, Daniela Melis, Maria Cristina Digilio, Marco Tartaglia, Bruno Marino and Alessandro De Lucaadd Show full author list remove Hide full author list
Genes 2019, 10(9), 675; https://0-doi-org.brum.beds.ac.uk/10.3390/genes10090675 - 04 Sep 2019
Cited by 19 | Viewed by 4024
Abstract
The aim of this study was to assess the prevalence and type of congenital heart disease (CHD) and the associated mutation spectrum in a large series of patients with neurofibromatosis type 1 (NF1), and correlate the mutation type with the presence and subgroups [...] Read more.
The aim of this study was to assess the prevalence and type of congenital heart disease (CHD) and the associated mutation spectrum in a large series of patients with neurofibromatosis type 1 (NF1), and correlate the mutation type with the presence and subgroups of cardiac defects. The study cohort included 493 individuals with molecularly confirmed diagnosis of NF1 for whom cardiac evaluation data were available. CHD was reported in 62/493 (12.6%) patients. Among these patients, 23/62 (37.1%) had pulmonary valve stenosis/dysplasia, 20/62 (32.3%) had mitral valve anomalies, and 10/62 (16.1%) had septal defects. Other defects occurred as rare events. In this NF1 subcohort, three subjects carried a whole-gene deletion, while 59 were heterozygous for an intragenic mutation. A significantly increased prevalence of non-truncating intragenic mutations was either observed in individuals with CHD (22/59, 37.3%) or with pulmonary valve stenosis (13/20, 65.0%), when compared to individuals without CHD (89/420, 21.2%) (p = 0.038) or pulmonary valve stenosis (98/459, 21.4%) (p = 0.002). Similarly, patients with non-truncating NF1 mutations displayed two- and six-fold higher risk of developing CHD (odds ratio = 1.9713, 95% confidence interval (CI): 1.1162–3.4814, p = 0.0193) and pulmonary valve stenosis (odds ratio = 6.8411, 95% CI: 2.6574–17.6114, p = 0.0001), respectively. Noteworthy, all but one patient (19/20, 95.0%) with pulmonary valve stenosis, and 18/35 (51.4%) patients with other CHDs displayed Noonan syndrome (NS)-like features. Present data confirm the significant frequency of CHD in patients with NF1, and provide further evidence for a higher than expected prevalence of NF1 in-frame variants and NS-like characteristics in NF1 patients with CHD, particularly with pulmonary valve stenosis. Full article
(This article belongs to the Special Issue Neurofibromatosis 1 Genetics)
Show Figures

Figure 1

16 pages, 1204 KiB  
Article
Affinity Purification of NF1 Protein–Protein Interactors Identifies Keratins and Neurofibromin Itself as Binding Partners
by Rachel M. Carnes, Robert A. Kesterson, Bruce R. Korf, James A. Mobley and Deeann Wallis
Genes 2019, 10(9), 650; https://0-doi-org.brum.beds.ac.uk/10.3390/genes10090650 - 28 Aug 2019
Cited by 10 | Viewed by 4295
Abstract
Neurofibromatosis Type 1 (NF1) is caused by pathogenic variants in the NF1 gene encoding neurofibromin. Definition of NF1 protein–protein interactions (PPIs) has been difficult and lacks replication, making it challenging to define binding partners that modulate its function. We created a novel tandem [...] Read more.
Neurofibromatosis Type 1 (NF1) is caused by pathogenic variants in the NF1 gene encoding neurofibromin. Definition of NF1 protein–protein interactions (PPIs) has been difficult and lacks replication, making it challenging to define binding partners that modulate its function. We created a novel tandem affinity purification (TAP) tag cloned in frame to the 3’ end of the full-length murine Nf1 cDNA (mNf1). We show that this cDNA is functional and expresses neurofibromin, His-Tag, and can correct p-ERK/ERK ratios in NF1 null HEK293 cells. We used this affinity tag to purify binding partners with Strep-Tactin®XT beads and subsequently, identified them via mass spectrometry (MS). We found the tagged mNf1 can affinity purify human neurofibromin and vice versa, indicating that neurofibromin oligomerizes. We identify 21 additional proteins with high confidence of interaction with neurofibromin. After Metacore network analysis of these 21 proteins, eight appear within the same network, primarily keratins regulated by estrogen receptors. Previously, we have shown that neurofibromin levels negatively regulate keratin expression. Here, we show through pharmacological inhibition that this is independent of Ras signaling, as the inhibitors, selumetinib and rapamycin, do not alter keratin expression. Further characterization of neurofibromin oligomerization and binding partners could aid in discovering new neurofibromin functions outside of Ras regulation, leading to novel drug targets. Full article
(This article belongs to the Special Issue Neurofibromatosis 1 Genetics)
Show Figures

Graphical abstract

10 pages, 635 KiB  
Article
One NF1 Mutation may Conceal Another
by Laurence Pacot, Cyril Burin des Roziers, Ingrid Laurendeau, Audrey Briand-Suleau, Audrey Coustier, Théodora Mayard, Camille Tlemsani, Laurence Faivre, Quentin Thomas, Diana Rodriguez, Sophie Blesson, Hélène Dollfus, Yvon-Gauthier Muller, Béatrice Parfait, Michel Vidaud, Brigitte Gilbert-Dussardier, Catherine Yardin, Benjamin Dauriat, Christian Derancourt, Dominique Vidaud and Eric Pasmantadd Show full author list remove Hide full author list
Genes 2019, 10(9), 633; https://0-doi-org.brum.beds.ac.uk/10.3390/genes10090633 - 22 Aug 2019
Cited by 7 | Viewed by 3885
Abstract
Neurofibromatosis type 1 (NF1) is an autosomal dominant disease with complete penetrance but high variable expressivity. NF1 is caused by loss-of-function mutations in the NF1 gene, a negative regulator of the RAS-MAPK pathway. The NF1 gene has one of the highest mutation rates [...] Read more.
Neurofibromatosis type 1 (NF1) is an autosomal dominant disease with complete penetrance but high variable expressivity. NF1 is caused by loss-of-function mutations in the NF1 gene, a negative regulator of the RAS-MAPK pathway. The NF1 gene has one of the highest mutation rates in human disorders, which may explain the outbreak of independent de novo variants in the same family. Here, we report the co-occurrence of pathogenic variants in the NF1 and SPRED1 genes in six families with NF1 and Legius syndrome, using next-generation sequencing. In five of these families, we observed the co-occurrence of two independent NF1 variants. All NF1 variants were classified as pathogenic, according to the American College of Medical Genetics and Genomics and the Association for Molecular Pathology (ACMG-AMP) guidelines. In the sixth family, one sibling inherited a complete deletion of the NF1 gene from her mother and carried a variant of unknown significance in the SPRED1 gene. This variant was also present in her brother, who was diagnosed with Legius syndrome, a differential diagnosis of NF1. This work illustrates the complexity of molecular diagnosis in a not-so-rare genetic disease. Full article
(This article belongs to the Special Issue Neurofibromatosis 1 Genetics)
Show Figures

Figure 1

29 pages, 302 KiB  
Article
Clinical and Genetic Findings in Children with Neurofibromatosis Type 1, Legius Syndrome, and Other Related Neurocutaneous Disorders
by Teresa Giugliano, Claudia Santoro, Annalaura Torella, Francesca Del Vecchio Blanco, Anna Grandone, Maria Elena Onore, Mariarosa Anna Beatrice Melone, Giulia Straccia, Daniela Melis, Vincenzo Piccolo, Giuseppe Limongelli, Salvatore Buono, Silverio Perrotta, Vincenzo Nigro and Giulio Piluso
Genes 2019, 10(8), 580; https://0-doi-org.brum.beds.ac.uk/10.3390/genes10080580 - 31 Jul 2019
Cited by 28 | Viewed by 5433
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 [...] Read more.
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. Full article
(This article belongs to the Special Issue Neurofibromatosis 1 Genetics)
Show Figures

Graphical abstract

Back to TopTop