92 results on '"D. Vidaud"'
Search Results
2. SPRED1 germline mutations caused a neurofibromatosis type 1 overlapping phenotype
- Author
-
E Pasmant, A Sabbagh, N Hanna, J Masliah-Planchon, E Jolly, P Goussard, P Ballerini, F Cartault, S Barbarot, J Landman-Parker, N Soufir, B Parfait, M Vidaud, P Wolkenstein, D Vidaud, R N F France, Genetique et Biotherapies des Maladies Degeneratives et Proliferatives du Systeme Nerveux (Inserm U745), Institut des sciences du Médicament -Toxicologie - Chimie - Environnement (IFR71), Institut National de la Santé et de la Recherche Médicale (INSERM)-Ecole Nationale Supérieure de Chimie de Paris - Chimie ParisTech-PSL (ENSCP), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Centre National de la Recherche Scientifique (CNRS)-Institut de Recherche pour le Développement (IRD)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Ecole Nationale Supérieure de Chimie de Paris - Chimie ParisTech-PSL (ENSCP), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Centre National de la Recherche Scientifique (CNRS)-Institut de Recherche pour le Développement (IRD)-Université Paris Descartes - Paris 5 (UPD5)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM), Service de Biochimie et Génétique Moléculaire, Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Université Paris Diderot - Paris 7 (UPD7)-Hôpital Beaujon [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP), Service d'hématologie-immunologie-oncologie pédiatrique [CHU Trousseau], Université Pierre et Marie Curie - Paris 6 (UPMC)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-CHU Trousseau [APHP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Sorbonne Université (SU), Service de génétique, Centre hospitalier Félix Guyon, Bellepierre, Service de dermatologie [Nantes], Université de Nantes (UN)-Centre hospitalier universitaire de Nantes (CHU Nantes), Service de biochimie hormonale, métabolique et génétique, Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-AP-HP - Hôpital Bichat - Claude Bernard [Paris], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Université Paris Diderot - Paris 7 (UPD7), Service de dermatologie [Mondor], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpital Henri Mondor-Université Paris-Est Créteil Val-de-Marne - Paris 12 (UPEC UP12), Institut de Recherche pour le Développement (IRD)-Ecole Nationale Supérieure de Chimie de Paris - Chimie ParisTech-PSL (ENSCP), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Institut de Recherche pour le Développement (IRD)-Ecole Nationale Supérieure de Chimie de Paris - Chimie ParisTech-PSL (ENSCP), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM), CHU Trousseau [APHP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU), Genetique et Biotherapies des Maladies Degeneratives et Proliferatives du Systeme Nerveux ( Inserm U745 ), Institut des sciences du Médicament -Toxicologie - Chimie - Environnement ( IFR71 ), Institut National de la Santé et de la Recherche Médicale ( INSERM ) -Ecole Nationale Supérieure de Chimie de Paris- Chimie ParisTech-PSL ( ENSCP ) -Centre National de la Recherche Scientifique ( CNRS ) -Institut de Recherche pour le Développement ( IRD ) -Université Paris Descartes - Paris 5 ( UPD5 ) -Institut National de la Santé et de la Recherche Médicale ( INSERM ) -Ecole Nationale Supérieure de Chimie de Paris- Chimie ParisTech-PSL ( ENSCP ) -Centre National de la Recherche Scientifique ( CNRS ) -Institut de Recherche pour le Développement ( IRD ) -Université Paris Descartes - Paris 5 ( UPD5 ) -Université Paris Descartes - Paris 5 ( UPD5 ) -Institut National de la Santé et de la Recherche Médicale ( INSERM ), Assistance publique - Hôpitaux de Paris (AP-HP)-Université Paris Diderot - Paris 7 ( UPD7 ) -Hôpital Beaujon, Service d'hématologie-immunologie-oncologie pédiatrique, Université Pierre et Marie Curie - Paris 6 ( UPMC ) -Assistance publique - Hôpitaux de Paris (AP-HP)-CHU Trousseau [APHP], Université de Nantes ( UN ) -Centre hospitalier universitaire de Nantes ( CHU Nantes ), Assistance publique - Hôpitaux de Paris (AP-HP)-AP-HP - Hôpital Bichat - Claude Bernard [Paris]-Université Paris Diderot - Paris 7 ( UPD7 ), Assistance publique - Hôpitaux de Paris (AP-HP)-Hôpital Henri Mondor-Université Paris-Est Créteil Val-de-Marne - Paris 12 ( UPEC UP12 ), and Peer, Hal
- Subjects
Adult ,Male ,medicine.medical_specialty ,congenital, hereditary, and neonatal diseases and abnormalities ,Neurofibromatosis 1 ,Adolescent ,DNA Mutational Analysis ,education ,Gene Dosage ,medicine.disease_cause ,Germline ,03 medical and health sciences ,0302 clinical medicine ,Germline mutation ,Molecular genetics ,Internal medicine ,Genetics ,medicine ,Humans ,Genetic Predisposition to Disease ,Neurofibromatosis ,Child ,Germ-Line Mutation ,Genetics (clinical) ,Adaptor Proteins, Signal Transducing ,Aged ,030304 developmental biology ,Aged, 80 and over ,Legius syndrome ,0303 health sciences ,Mutation ,Neurofibromin 1 ,biology ,Intracellular Signaling Peptides and Proteins ,Membrane Proteins ,Middle Aged ,medicine.disease ,Phenotype ,Pedigree ,3. Good health ,nervous system diseases ,Endocrinology ,Child, Preschool ,biology.protein ,Female ,030217 neurology & neurosurgery - Abstract
Germline loss-of-function mutations in the SPRED1 gene have recently been identified in patients fulfilling the National Institutes of Health (NIH) diagnostic criteria for neurofibromatosis type 1 (NF1) but with no NF1 (neurofibromin 1) mutation found, suggesting a neurofibromatosis type 1-like syndrome.61 index cases with NF1 clinical diagnosis but no identifiable NF1 mutation were screened for SPRED1 mutation.We describe one known SPRED1 mutation (c.190CT leading to p.Arg64Stop) and four novel mutations (c.637CT leading to p.Gln213Stop, c.2TC leading to p.Met1Thr, c.46CT leading to p.Arg16Stop, and c.1048_1060del leading to p.Gly350fs) in five French families. Their NF1-like phenotype was characterised by a high prevalence of café-au-lait spots, freckling, learning disability, and an absence of neurofibromas and Lisch nodules in agreement with the original description. However, we did not observe Noonan-like dysmorphy. It is noteworthy that one patient with the p.Arg16Stop mutation developed a monoblastic acute leukaemia.In our series, SPRED1 mutations occurred with a prevalence of 0.5% in NF1 patients and in 5% of NF1 patients displaying an NF1-like phenotype. SPRED1 mutated patients did not display any specific dermatologic features that were not present in NF1 patients, except for the absence of neurofibromas that seem to be a specific clinical feature of NF1. The exact phenotypic spectrum and the putative complications of this NF1 overlapping syndrome, in particular haematological malignancies, remain to be further characterised. NIH diagnostic criteria for NF1 must be revised in view of this newly characterised Legius syndrome in order to establish a specific genetic counselling.
- Published
- 2009
- Full Text
- View/download PDF
3. Molecular basis for antithrombin III type I deficiency: three novel mutations located in exon IV
- Author
-
D Vidaud, Joseph Emmerich, Martine Aiach, Pierre Sié, Martine Alhenc-Gelas, and ME Sirieix
- Subjects
Genetics ,Mutation ,Immunology ,Antithrombin ,Antithrombin III deficiency ,Cell Biology ,Hematology ,Biology ,medicine.disease ,medicine.disease_cause ,Biochemistry ,Molecular biology ,Stop codon ,Frameshift mutation ,Exon ,medicine ,Missense mutation ,Gene ,medicine.drug - Abstract
Antithrombin III (AT III) type I deficiencies are characterized by a 50% decrease of both immunoreactive and functional protein and carry a high risk of thrombotic complication. We have studied the molecular basis for such deficiencies by asymmetric polymerase chain reaction amplification and direct sequencing of the seven exons and of the intron-exon junction of the AT III gene. Three different mutations were observed in the exon IV: a 4-bp deletion, a 2-bp deletion, and a nucleotide insertion. Each of these mutations results in a frameshift introducing premature stop codons at positions 313, 309, and 232, respectively. These results were confirmed by dot-blot analysis with allele-specific oligonucleotide probes. Furthermore, no mutation was observed in the other six exons. The comparison of the type of mutations observed by our group in six cases of type I deficiencies and in 16 cases of type II heparin binding site variants deficiencies suggests that the former are caused by heterogeneous molecular abnormalities while the latter are caused by recurrent missense mutations.
- Published
- 1991
- Full Text
- View/download PDF
4. Important role of arginine 129 in heparin-binding site of antithrombin III. Identification of a novel mutation arginine 129 to glutamine
- Author
-
Eric Clauser, P. Molho-Sabatier, J N Fiessinger, P de Moerloose, Sophie Gandrille, Martine Aiach, D Vidaud, R Caso, and David A. Lane
- Subjects
chemistry.chemical_classification ,Arginine ,Binding protein ,Antithrombin ,Cell Biology ,Heparin ,Biology ,Biochemistry ,Molecular biology ,Amino acid ,Thrombin ,Affinity chromatography ,chemistry ,medicine ,Binding site ,Molecular Biology ,medicine.drug - Abstract
An hereditary abnormal antithrombin III (ATIII Geneva) with defective heparin cofactor activity was characterized by DNA single strand amplification and subsequent direct sequencing. ATIII Geneva was found to have a G to A transition in Exon IIIa leading to an Arg-129 to Gln mutation. This amino acid is part of the ATIII region comprising residues 114-154, which contains the highest proportion of basic residues (Arg or Lys), and is known from chemical modification studies to be involved in heparin binding. The variant protein did not bind heparin-Sepharose and was isolated from the propositus plasma by immunoaffinity chromatography. High affinity (for ATIII) heparin had only a minimal effect on thrombin and activated factor X inhibition by the purified abnormal ATIII. Taken together, these results demonstrate an important role for Arg-129 in the binding and interaction of ATIII with heparin of high affinity. We propose that a cooperation between Lys-125, Arg-129, Lys-136, and Arg-47 exposed at the surface of the inhibitor allows the binding of the essential pentasaccharide domain of heparin which is specific for the ATIII interaction.
- Published
- 1990
- Full Text
- View/download PDF
5. 126 EFFECTS OF IRON DEPLETION ON ARTICULAR BIOMARKERS AND JOINT SYMPTOMS IN PATIENTS WITH GENETIC HEMOCHROMATOSIS: A PROSPECTIVE, LONGITUDINAL STUDY
- Author
-
Yves Henrotin, D Vidaud, Thomas Bardin, C Eymard, Michelle Deberg, and P. Richette
- Subjects
medicine.medical_specialty ,Longitudinal study ,Pathology ,business.industry ,Biomedical Engineering ,Genetic hemochromatosis ,Gastroenterology ,Rheumatology ,Internal medicine ,medicine ,Orthopedics and Sports Medicine ,In patient ,business ,Iron depletion - Published
- 2009
- Full Text
- View/download PDF
6. Detection of more than 91% cystic fibrosis mutations in a sample of the population from Reunion Island and identification of two novel mutations (A309G, S1255L) and one novel polymorphism (L49L)
- Author
-
F, Cartault, J, Steffann, D, Vidaud, S, Bousquet, F, Lesure, M, Renouil, N, McDonell, J, Feingold, C, Beldjord, and T, Bienvenu
- Subjects
Alanine ,Polymorphism, Genetic ,Cystic Fibrosis ,Leucine ,Glycine ,Mutation, Missense ,Serine ,Cystic Fibrosis Transmembrane Conductance Regulator ,Humans ,France - Published
- 1998
7. Three novel mutations of antithrombin inducing high-molecular-mass compounds
- Author
-
Martine Alhenc-Gelas, Joseph Emmerich, G Chadeuf, Martine Aiach, D Vidaud, M. F. Aillaud, and M Gouault-Heilmann
- Subjects
Adult ,Male ,Adolescent ,Population ,Molecular Sequence Data ,medicine.disease_cause ,Compound heterozygosity ,Antithrombins ,Exon ,medicine ,Humans ,Nucleotide ,education ,Gene ,chemistry.chemical_classification ,Mutation ,education.field_of_study ,Molecular mass ,Base Sequence ,Antithrombin ,Thrombosis ,Exons ,Molecular biology ,Molecular Weight ,Biochemistry ,chemistry ,Female ,Cardiology and Cardiovascular Medicine ,Gene Deletion ,medicine.drug - Abstract
We have identified three novel mutations of the antithrombin (AT) gene in patients with thrombotic complications: a Cys 128 --> Tyr mutations, a G --> A mutation in the intervening sequence 4 (IVS4) 14 nucleotide 5' to exon 5, and a 9 bp deletion in the 3' end of exon 6 resulting in a short aberrant sequence after Arg 425. The latter mutation was associated with an Arg 47 --> His mutation in two compound heterozygous brothers. These three mutations led to the expression in the circulation of small amounts of inactive molecules with a high molecular mass in immunoblot analysis. In reducing conditions, these variant molecules had a normal molecular mass, which led us to postulate that these mutations prevent the formation of one intramolecular disulfide bond and allow the formation of intermolecular disulfide bonds. Plasma from a heterozygous patients bearing the Cys 128 --> Tyr mutation and from a compound heterozygote bearing the Arg 47 --> His mutation and the 9 bp deletion in exon 6 were passed through a heparin-sepharose column. In both cases a population of high-molecular-weight AT molecules with no binding affinity and no AT activity was separated from a population of normal molecules in the first patient, together with a population of molecules with a reduced binding affinity for heparin due to the substitution of Arg 47, in the compound heterozygote. The common feature of these three mutations is that they lead to partial misfolding and to the formation of intermolecular disulfide bonds with other plasma components, inducing the pleiotropic phenotypes observed.
- Published
- 1994
8. Met 358 to Arg mutation of alpha 1-antitrypsin associated with protein C deficiency in a patient with mild bleeding tendency
- Author
-
D Vidaud, J Yvart, J Emmerich, Martine Aiach, M. Alhenc-Gelas, and J N Fiessinger
- Subjects
Male ,medicine.drug_mechanism_of_action ,Factor Xa Inhibitor ,DNA Mutational Analysis ,Molecular Sequence Data ,Alpha (ethology) ,Serpin ,Biology ,Thrombin ,Protein C deficiency ,medicine ,Humans ,Transition (genetics) ,Base Sequence ,Antithrombin ,Protein C Deficiency ,General Medicine ,Blood Coagulation Disorders ,medicine.disease ,Molecular biology ,Pedigree ,Oligodeoxyribonucleotides ,alpha 1-Antitrypsin ,Mutation ,Oligonucleotide Probes ,Protein C ,medicine.drug ,Research Article ,Factor Xa Inhibitors - Abstract
The molecular defect responsible for a dramatic prolongation of all standard clotting tests discovered in a 15-yr-old boy has been identified. Initial investigations revealed the presence of an activated Factor X (Factor Xa) and thrombin inhibitor which copurified with alpha 1-antitrypsin (alpha 1-AT), thereby suggesting the occurrence of an alpha 1-AT variant similar to alpha 1-AT Pittsburgh. This was confirmed by dot-blot analysis and direct sequencing after amplification by the polymerase chain reaction. A G to T transition at nucleotide 10038 results in the substitution of Met to an Arg, converting alpha 1-AT into an Arg-Ser protease inhibitor (serpin) that inhibited thrombin and Factor Xa more effectively than antithrombin III. Surprisingly, there was no bleeding history in the proband. The common mutation Z, which may explain a reduced expression of the allele bearing the Arg 358 Met mutation, was not observed in the propositus' DNA. To exclude the presence of another mutation, the coding regions and intron/exon junctions were sequenced. No other mutation was found. Recently, the patient experienced his first hemorrhagic episode at the age of 17. The level of the abnormal inhibitor had increased twofold 2 mo before. The large decrease in protein C concentration may account for the mild bleeding tendency in this case, despite the presence of the alpha 1-AT Pittsburgh mutation. An abnormal protein C pattern was observed in patient's plasma, suggesting that the circulating deficiency might be due to a deleterious effect of the abnormal inhibitor on both intracellular processing and catabolism of protein C.
- Published
- 1992
9. Towards a transgenic mouse model of sickle cell disease: hemoglobin SAD
- Author
-
J L Guerquin-Kern, M.C. Garel, N Saadane, J Bardakdjian-Michau, A Pachnis, Paul-Henri Romeo, Y Blouquit, P Rouyer-Fessard, M Trudel, and D Vidaud
- Subjects
Genetically modified mouse ,Erythrocyte Indices ,Anemia ,Transgene ,Hemoglobin, Sickle ,Spleen ,Mice, Transgenic ,Anemia, Sickle Cell ,Biology ,behavioral disciplines and activities ,Peptide Mapping ,Polymerase Chain Reaction ,General Biochemistry, Genetics and Molecular Biology ,Mice ,mental disorders ,medicine ,Animals ,Trypsin ,Beta (finance) ,Molecular Biology ,Chromatography, High Pressure Liquid ,General Immunology and Microbiology ,General Neuroscience ,DNA ,medicine.disease ,Molecular biology ,Sickle cell anemia ,Globins ,Oxygen ,Disease Models, Animal ,medicine.anatomical_structure ,Phenotype ,Erythropoiesis ,Electrophoresis, Polyacrylamide Gel ,Hemoglobin ,Isoelectric Focusing ,Research Article - Abstract
In order to obtain a transgenic mouse model of sickle cell disease, we have synthesized a novel human beta-globin gene, beta SAD, designed to increase the polymerization of the transgenic human hemoglobin S (Hb S) in vivo. beta SAD (beta S-Antilles-D Punjab) includes the beta 6Val substitution of the beta S chain, as well as two other mutations, Antilles (beta 23Ile) and D Punjab (beta 121Gln) each of which promotes the polymerization of Hb S in human. The beta SAD gene and the human alpha 2-globin gene, each linked to the beta-globin locus control region (LCR) were co-introduced into the mouse germ line. In one of the five transgenic lines obtained, SAD-1, red blood cells contained 19% human Hb SAD (alpha 2 human 1 beta 2SAD) and mouse-human hybrids in addition to mouse hemoglobin. Adult SAD-1 transgenic mice were not anemic but had some abnormal features of erythrocytes and slightly enlarged spleens. Their erythrocytes displayed sickling upon deoxygenation in vitro. SAD-1 neonates were anemic and many did not survive. In order to generate adult mice with a more severe sickle cell syndrome, crosses between the SAD progeny and homozygous for beta-thalassemic mice were performed. Hemoglobin SAD was increased to 26% in beta-thal/SAD-1 mice which exhibited: (i) abnormal erythrocytes with regard to shape and density; (ii) an enlarged spleen and a high reticulocyte count indicating an increased erythropoiesis; (iii) mortality upon hypoxia; (iv) polymerization of hemolysate similar to that obtained in human homozygous sickle cell disease; and (v) anemia and mortality during development.
- Published
- 1991
10. Important role of arginine 129 in heparin-binding site of antithrombin III. Identification of a novel mutation arginine 129 to glutamine
- Author
-
S, Gandrille, M, Aiach, D A, Lane, D, Vidaud, P, Molho-Sabatier, R, Caso, P, de Moerloose, J N, Fiessinger, and E, Clauser
- Subjects
Models, Molecular ,Binding Sites ,Base Sequence ,Heparin ,Protein Conformation ,Glutamine ,Antithrombin III ,Molecular Sequence Data ,DNA ,Exons ,Arginine ,Polymerase Chain Reaction ,Kinetics ,Mutation ,Leukocytes ,Humans ,Amino Acid Sequence - Abstract
An hereditary abnormal antithrombin III (ATIII Geneva) with defective heparin cofactor activity was characterized by DNA single strand amplification and subsequent direct sequencing. ATIII Geneva was found to have a G to A transition in Exon IIIa leading to an Arg-129 to Gln mutation. This amino acid is part of the ATIII region comprising residues 114-154, which contains the highest proportion of basic residues (Arg or Lys), and is known from chemical modification studies to be involved in heparin binding. The variant protein did not bind heparin-Sepharose and was isolated from the propositus plasma by immunoaffinity chromatography. High affinity (for ATIII) heparin had only a minimal effect on thrombin and activated factor X inhibition by the purified abnormal ATIII. Taken together, these results demonstrate an important role for Arg-129 in the binding and interaction of ATIII with heparin of high affinity. We propose that a cooperation between Lys-125, Arg-129, Lys-136, and Arg-47 exposed at the surface of the inhibitor allows the binding of the essential pentasaccharide domain of heparin which is specific for the ATIII interaction.
- Published
- 1990
11. NF1 gene analysis focused on CpG-rich exons in a cohort of 93 patients with neurofibromatosis type 1 Communicated by: Jurgen Horst Online Citation: Human Mutation, Mutation in Brief #363 (2000) Online http://journals.wiley.com/1059-7794/pdf/mutation/363.pdf
- Author
-
E. Girodon Boulandet, J. Pantel, C. Cazeneuve, M. Van Gijn, D. Vidaud, S. Lemay, J. Martin, J. Zeller, J. Revuz, M. Goossens, S. Amselem, and P. Wolkenstein
- Subjects
Genetics ,Genetics (clinical) - Published
- 2000
- Full Text
- View/download PDF
12. Prenatal diagnosis for neurofibromatosis type 1 and the pitfalls of germline mosaics.
- Author
-
Pacot L, Vidaud D, Ye M, Chansavang A, Coustier A, Maillard T, Barbance C, Laurendeau I, Hébrard B, Lunati-Rozie A, Funalot B, Wolkenstein P, Vidaud M, Goldenberg A, Morice-Picard F, Hadjadj D, Parfait B, and Pasmant E
- Abstract
We report our 5-year experience in neurofibromatosis type 1 prenatal diagnosis (PND): 205 PNDs in 146 women (chorionic villus biopsies, 88% or amniocentesis, 12%). The NF1 variant was present in 85 (41%) and absent in 122 (59%) fetuses. Among 205 pregnancies (207 fetuses), 135 were carried to term (119 unaffected and 16 NF1 affected children), 69 pregnancy terminations (affected fetuses), 2 miscarriages, and 1 in utero death. The majority of PND requests came from parents with sporadic NF1. We describe two PNDs in women with mosaic NF1. In both families, direct PND showed the absence of the maternal NF1 variant in the fetus. However, microsatellite markers analysis showed that the risk haplotype had been transmitted. These rare cases of germline mosaicism illustrate the pitfall of indirect PND. Our study illustrates the crucial consequences of PND for medical and genetic counseling decisions. We also point to the challenges of germline mosaics., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
13. Breast cancer risk in NF1 -deleted patients.
- Author
-
Pacot L, Masliah-Planchon J, Petcu A, Terris B, Gauthier Villars M, Lespinasse J, Wolkenstein P, Vincent-Salomon A, Vidaud D, and Pasmant E
- Subjects
- Female, Humans, Genetic Predisposition to Disease, Breast Neoplasms epidemiology, Breast Neoplasms genetics
- Abstract
Competing Interests: Competing interests: None declared.
- Published
- 2024
- Full Text
- View/download PDF
14. Neurofibromatosis type 1 mosaicism in patients with constitutional mismatch repair deficiency.
- Author
-
Guerrini-Rousseau L, Pasmant E, Muleris M, Abbou S, Adam-De-Beaumais T, Brugieres L, Cabaret O, Colas C, Cotteret S, Decq P, Dufour C, Guillerm E, Rouleau E, Varlet P, Zili S, Vidaud D, and Grill J
- Subjects
- Female, Humans, Mosaicism, Retrospective Studies, Mismatch Repair Endonuclease PMS2 genetics, DNA Mismatch Repair genetics, Neurofibromatosis 1 diagnosis, Neurofibromatosis 1 genetics, Neoplastic Syndromes, Hereditary genetics, Brain Neoplasms genetics, Colorectal Neoplasms genetics
- Abstract
Differential diagnosis between constitutional mismatch repair deficiency (CMMRD) and neurofibromatosis type 1 (NF1 ) is crucial as treatment and surveillance differ. We report the case of a girl with a clinical diagnosis of sporadic NF1 who developed a glioblastoma. Immunohistochemistry for MMR proteins identified PMS2 loss in tumour and normal cells and WES showed the tumour had an ultra-hypermutated phenotype, supporting the diagnosis of CMMRD. Germline analyses identified two variants (one pathogenic variant and one classified as variant(s) of unknown significance) in the PMS2 gene and subsequent functional assays on blood lymphocytes confirmed the diagnosis of CMMRD. The large plexiform neurofibroma of the thigh and the freckling were however more compatible with NF1. Indeed, a NF1 PV (variant allele frequencies of 20%, 3% and 9% and in blood, skin and saliva samples, respectively) was identified confirming a mosaicism for NF1. Retrospective analysis of a French cohort identified NF1 mosaicism in blood DNA in 2 out of 22 patients with CMMRD, underlining the existence of early postzygotic PV of NF1 gene in patients with CMMRD whose tumours have been frequently reported to exhibit somatic NF1 mutations. It highlights the potential role of this pathway in the pathogenesis of CMMRD-associated gliomas and argues in favour of testing MEK inhibitors in this context., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2024. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2024
- Full Text
- View/download PDF
15. Prevalence of Endocrine Manifestations and GIST in 108 Systematically Screened Patients With Neurofibromatosis Type 1.
- Author
-
Dupuis H, Chevalier B, Cardot-Bauters C, Jannin A, Do Cao C, Ladsous M, Cortet C, Merlen E, Drouard M, Aubert S, Vidaud D, Espiard S, and Vantyghem MC
- Abstract
Context: In patients with neurofibromatosis type 1 (NF1), guidelines suggest screening for pheochromocytoma by metanephrine measurement and abdominal imaging, which may lead to the discovery of gastroenteropancreatic neuroendocrine tumors (GEP-NETs) and their differential diagnosis, gastrointestinal stromal tumors (GISTs). Other endocrine manifestations such as follicular thyroid carcinoma and primary hyperparathyroidism have also been reported in a few cases., Objective: This study aimed to describe prevalence and clinical presentation of these manifestations through systematic screening in a large cohort of patients., Methods: In this monocentric retrospective study, 108 patients with NF1 were included and screened for endocrine manifestations and GISTs. Clinical, laboratory, molecular profile, pathology, and morphologic (abdominal computed tomography scan and/or magnetic resonance imaging) and functional imaging were collected., Results: Twenty-four patients (22.2% of the cohort, 16 female, mean age 42.6 years) presented with pheochromocytomas that were unilateral in 65.5%, benign in 89.7%, and with a ganglioneural component in 20.7%. Three female patients (2.8% of the cohort, aged 42-63 years) presented with well-differentiated GEP-NETs, and 4 (3.7%) with GISTs. One patient had primary hyperparathyroidism, 1 patient had medullary microcarcinoma, and 16 patients had goiter, multinodular in 10 cases. There was no correlation between pheochromocytoma and other NF1 tumoral manifestations, nor correlations between pheochromocytoma and NF1 genotype, despite a familial clustering in one-third of patients., Conclusion: The pheochromocytoma prevalence in this NF1 cohort was higher (>20%) than previously described, confirming the interest of systematic screening, especially in young women. The prevalence of GEP-NETs and GISTs was about 3%, respectively. No phenotype-genotype correlation was observed., (© The Author(s) 2023. Published by Oxford University Press on behalf of the Endocrine Society.)
- Published
- 2023
- Full Text
- View/download PDF
16. Non-invasive prenatal diagnosis of single gene disorders with enhanced relative haplotype dosage analysis for diagnostic implementation.
- Author
-
Pacault M, Verebi C, Champion M, Orhant L, Perrier A, Girodon E, Leturcq F, Vidaud D, Férec C, Bienvenu T, Daveau R, and Nectoux J
- Subjects
- Pregnancy, Female, Humans, Haplotypes, Prenatal Diagnosis methods, Genotype, Noninvasive Prenatal Testing methods, Cell-Free Nucleic Acids
- Abstract
Non-invasive prenatal diagnosis of single-gene disorders (SGD-NIPD) has been widely accepted, but is mostly limited to the exclusion of either paternal or de novo mutations. Indeed, it is still difficult to infer the inheritance of the maternal allele from cell-free DNA (cfDNA) analysis. Based on the study of maternal haplotype imbalance in cfDNA, relative haplotype dosage (RHDO) was developed to address this challenge. Although RHDO has been shown to be reliable, robust control of statistical error and explicit delineation of critical parameters for assessing the quality of the analysis have not been fully addressed. We present here a universal and adaptable enhanced-RHDO (eRHDO) procedure through an automated bioinformatics pipeline with a didactic visualization of the results, aiming to be applied for any SGD-NIPD in routine care. A training cohort of 43 families carrying CFTR, NF1, DMD, or F8 mutations allowed the characterization and optimal setting of several adjustable data variables, such as minimum sequencing depth, type 1 and type 2 statistical errors, as well as the quality assessment of intermediate steps and final results by block score and concordance score. Validation was successfully performed on a test cohort of 56 pregnancies. Finally, computer simulations were used to estimate the effect of fetal-fraction, sequencing depth and number of informative SNPs on the quality of results. Our workflow proved to be robust, as we obtained conclusive and correctly inferred fetal genotypes in 94.9% of cases, with no false-negative or false-positive results. By standardizing data generation and analysis, we fully describe a turnkey protocol for laboratories wishing to offer eRHDO-based non-invasive prenatal diagnosis for single-gene disorders as an alternative to conventional prenatal diagnosis., Competing Interests: The authors have declared that no competing interests exist., (Copyright: © 2023 Pacault et al. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.)
- Published
- 2023
- Full Text
- View/download PDF
17. Comment on Intragenic inversions in NF1 gene as pathogenic mechanism in neurofibromatosis type 1.
- Author
-
Pacot L, Chansavang A, Jacques S, Laurendeau I, Hadjadj D, Ferkal S, Wolkenstein P, Vidaud D, and Pasmant E
- Subjects
- Humans, Genes, Neurofibromatosis 1, Neurofibromatosis 1 diagnosis, Neurofibromatosis 1 genetics, Neurofibromin 1 genetics
- Published
- 2023
- Full Text
- View/download PDF
18. Contribution of whole genome sequencing in the molecular diagnosis of mosaic partial deletion of the NF1 gene in neurofibromatosis type 1.
- Author
-
Pacot L, Pelletier V, Chansavang A, Briand-Suleau A, Burin des Roziers C, Coustier A, Maillard T, Vaucouleur N, Orhant L, Barbance C, Lermine A, Hamzaoui N, Hadjadj D, Laurendeau I, El Khattabi L, Nectoux J, Vidaud M, Parfait B, Dollfus H, Pasmant E, and Vidaud D
- Subjects
- Humans, Genes, Neurofibromatosis 1, Comparative Genomic Hybridization, Exons, Whole Genome Sequencing, Neurofibromatosis 1 diagnosis, Neurofibromatosis 1 genetics
- Abstract
Neurofibromatosis type 1 (NF1) is an autosomal dominant disease with complete penetrance but highly variable expressivity. In most patients, Next Generation Sequencing (NGS) technologies allow the identification of a loss-of-function pathogenic variant in the NF1 gene, a negative regulator of the RAS-MAPK pathway. We describe the 5-year diagnosis wandering of a patient with a clear NF1 clinical diagnosis, but no molecular diagnosis using standard molecular technologies. The patient presented with a typical NF1 phenotype but NF1 targeted NGS, NF1 transcript analysis, MLPA, and array comparative genomic hybridization failed to reveal a genetic aberration. After 5 years of unsuccessful investigations, trio WGS finally identified a de novo mosaic (VAF ~ 14%) 24.6 kb germline deletion encompassing the promoter and first exon of NF1. This case report illustrates the relevance of WGS to detect structural variants including copy number variants that would be missed by alternative approaches. The identification of the causal pathogenic variant allowed a tailored genetic counseling with a targeted non-invasive prenatal diagnosis by detecting the deletion in plasmatic cell-free DNA from the proband's pregnant partner. This report clearly highlights the need to make WGS a clinically accessible test, offering a tremendous opportunity to identify a molecular diagnosis for otherwise unsolved cases., (© 2022. The Author(s), under exclusive licence to Springer-Verlag GmbH Germany, part of Springer Nature.)
- Published
- 2023
- Full Text
- View/download PDF
19. Natural history of NF1 c.2970_2972del p.(Met992del): confirmation of a low risk of complications in a longitudinal study.
- Author
-
Forde C, Burkitt-Wright E, Turnpenny PD, Haan E, Ealing J, Mansour S, Holder M, Lahiri N, Dixit A, Procter A, Pacot L, Vidaud D, Capri Y, Gerard M, Dollfus H, Schaefer E, Quelin C, Sigaudy S, Busa T, Vera G, Damaj L, Messiaen L, Stevenson DA, Davies P, Palmer-Smith S, Callaway A, Wolkenstein P, Pasmant E, and Upadhyaya M
- Subjects
- Cafe-au-Lait Spots genetics, Genetic Association Studies, Humans, Longitudinal Studies, Neurofibroma genetics, Neurofibromatosis 1 diagnosis, Neurofibromatosis 1 genetics, Neurofibromatosis 1 pathology
- Abstract
Individuals with the three base pair deletion NM_000267.3(NF1):c.2970_2972del p.(Met992del) have been recognised to present with a milder neurofibromatosis type 1 (NF1) phenotype characterised by café-au-lait macules (CALs) and intertriginous freckling, as well as a lack of cutaneous, subcutaneous and plexiform neurofibromas and other NF1-associated complications. Examining large cohorts of patients over time with this specific genotype is important to confirm the presentation and associated risks of this variant across the lifespan. Forty-one individuals with the in-frame NF1 deletion p.Met992del were identified from 31 families. Clinicians completed a standardised clinical questionnaire for each patient and the resulting data were collated and compared to published cohorts. Thirteen patients have been previously reported, and updated clinical information has been obtained for these individuals. Both CALs and intertriginous freckling were present in the majority of individuals (26/41, 63%) and the only confirmed features in 11 (27%). 34/41 (83%) of the cohort met NIH diagnostic criteria. There was a notable absence of all NF1-associated tumour types (neurofibroma and glioma). Neurofibroma were observed in only one individual-a subcutaneous lesion (confirmed histologically). Nineteen individuals were described as having a learning disability (46%). This study confirms that individuals with p.Met992del display a mild tumoural phenotype compared to those with 'classical', clinically diagnosed NF1, and this appears to be the case longitudinally through time as well as at presentation. Learning difficulties, however, appear to affect a significant proportion of NF1 subjects with this phenotype. Knowledge of this genotype-phenotype association is fundamental to accurate prognostication for families and caregivers., (© 2021. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
20. Severe Phenotype in Patients with Large Deletions of NF1 .
- Author
-
Pacot L, Vidaud D, Sabbagh A, Laurendeau I, Briand-Suleau A, Coustier A, Maillard T, Barbance C, Morice-Picard F, Sigaudy S, Glazunova OO, Damaj L, Layet V, Quelin C, Gilbert-Dussardier B, Audic F, Dollfus H, Guerrot AM, Lespinasse J, Julia S, Vantyghem MC, Drouard M, Lackmy M, Leheup B, Alembik Y, Lemaire A, Nitschké P, Petit F, Dieux Coeslier A, Mutez E, Taieb A, Fradin M, Capri Y, Nasser H, Ruaud L, Dauriat B, Bourthoumieu S, Geneviève D, Audebert-Bellanger S, Nizon M, Stoeva R, Hickman G, Nicolas G, Mazereeuw-Hautier J, Jannic A, Ferkal S, Parfait B, Vidaud M, Members Of The Nf France Network, Wolkenstein P, and Pasmant E
- Abstract
Complete deletion of the NF1 gene is identified in 5-10% of patients with neurofibromatosis type 1 (NF1). Several studies have previously described particularly severe forms of the disease in NF1 patients with deletion of the NF1 locus, but comprehensive descriptions of large cohorts are still missing to fully characterize this contiguous gene syndrome. NF1 -deleted patients were enrolled and phenotypically characterized with a standardized questionnaire between 2005 and 2020 from a large French NF1 cohort. Statistical analyses for main NF1-associated symptoms were performed versus an NF1 reference population. A deletion of the NF1 gene was detected in 4% (139/3479) of molecularly confirmed NF1 index cases. The median age of the group at clinical investigations was 21 years old. A comprehensive clinical assessment showed that 93% (116/126) of NF1 -deleted patients fulfilled the NIH criteria for NF1. More than half had café-au-lait spots, skinfold freckling, Lisch nodules, neurofibromas, neurological abnormalities, and cognitive impairment or learning disabilities. Comparison with previously described "classic" NF1 cohorts showed a significantly higher proportion of symptomatic spinal neurofibromas, dysmorphism, learning disabilities, malignancies, and skeletal and cardiovascular abnormalities in the NF1 -deleted group. We described the largest NF1 -deleted cohort to date and clarified the more severe phenotype observed in these patients.
- Published
- 2021
- Full Text
- View/download PDF
21. Phase I study of vinblastine in combination with nilotinib in children, adolescents, and young adults with refractory or recurrent low-grade glioma.
- Author
-
Vairy S, Le Teuff G, Bautista F, De Carli E, Bertozzi AI, Pagnier A, Fouyssac F, Nysom K, Aerts I, Leblond P, Millot F, Berger C, Canale S, Paci A, Poinsignon V, Chevance A, Ezzalfani M, Vidaud D, Di Giannatale A, Hladun-Alvaro R, Petit FM, Vassal G, Geoerger B, Le Deley MC, and Grill J
- Abstract
Background: New rescue regimens are needed for pediatric refractory/recurrent low-grade glioma. Nilotinib is a tyrosine kinase inhibitor that has potential synergistic effects with vinblastine on angiogenesis, tumor cell growth, and immunomodulation., Methods: This phase I trial aimed to determine the recommended doses of this combination for phase II trials (RP2D) using the dual-agent Bayesian continual reassessment method. Nilotinib was given orally twice daily (BID) in combination with once-weekly vinblastine injections for a maximum of 12 cycles of 28 days (clinicaltrials.gov, NCT01884922)., Results: Thirty-five pediatric patients were enrolled across 4 dose levels. The median age was 7 years and 10 had neurofibromatosis type 1. Patients had received a median of 3 prior treatment lines and 25% had received more than 4 previous treatment lines. Dose-limiting toxicity (DLT) during cycle 1 was hematologic, dermatologic, and cardiovascular. The RP2D was identified at 3 mg/m
2 weekly for vinblastine with 230 mg/m2 BID for nilotinib (estimated probability of DLT = 18%; 95% credibility interval, 7-29%). Fifteen patients completed the 12 cycles; 2 stopped therapy prematurely due to toxicity and 18 due to disease progression. Three patients achieved a partial response leading to an objective response rate of 8.8% (95% confidence interval [CI], 1.9-23.7), and the disease control rate was 85.3% (95% CI, 68.9-95.1). The 12-month progression-free survival was 37.1% (95% CI, 23.2-53.67)., Conclusions: Vinblastine and nilotinib combination was mostly limited by myelosuppression and dermatologic toxicity. The efficacy of the combination at the RP2D is currently evaluated in a randomized phase II trial comparing this regimen to vinblastine alone., (© The Author(s) 2020. Published by Oxford University Press, the Society for Neuro-Oncology and the European Association of Neuro-Oncology.)- Published
- 2020
- Full Text
- View/download PDF
22. NF1-like optic pathway gliomas in children: clinical and molecular characterization of this specific presentation.
- Author
-
Lobón-Iglesias MJ, Laurendeau I, Guerrini-Rousseau L, Tauziède-Espariat A, Briand-Suleau A, Varlet P, Vidaud D, Vidaud M, Brugieres L, Grill J, and Pasmant E
- Abstract
Background: Pediatric neurofibromatosis type 1 (NF1)-associated optic pathway gliomas (OPGs) exhibit different clinico-radiological features, treatment, and outcome compared with sporadic OPGs. While NF1-associated OPGs are caused by complete loss-of-function of the NF1 gene, other genetic alterations of the RAS-MAPK pathway are frequently described in the sporadic cases. We identified a group of patients who presented OPGs with typical radiological features of NF1-associated OPGs but without the NF1 diagnostic criteria. We aim to investigate into the possible molecular mechanisms underlying this "NF1-like" pediatric OPGs presentation., Methods: We analyzed clinico-radiological features of 16 children with NF1-like OPGs and without NF1 diagnostic criteria. We performed targeted sequencing of the NF1 gene in constitutional samples ( n = 16). The RAS-MAPK pathway major genes were sequenced in OPG tumor samples ( n = 11); BRAF FISH and IHC analyses were also performed., Results: In one patient's blood and tumor samples, we identified a NF1 nonsense mutation (exon 50: c.7285C>T, p.Arg2429*) with ~8% and ~70% VAFs, respectively, suggesting a mosaic NF1 mutation limited to the brain (segmental NF1). This patient presented signs of neurodevelopmental disorder. We identified a somatic alteration of the RAS-MAPK pathway in eight tumors: four BRAF activating p.Val600Glu mutations, three BRAF:KIAA oncogenic fusions, and one putative gain-of-function complex KRAS indel inframe mutation., Conclusions: NF1-like OPGs can rarely be associated with mosaic NF1 that needs specific constitutional DNA analyses for diagnosis. Further studies are warranted to explore unknown predisposition condition leading to the NF1-like OPG presentation, particularly in patients with the association of a neurodevelopmental disorder., (© The Author(s) 2019. Published by Oxford University Press, the Society for Neuro-Oncology and the European Association of Neuro-Oncology.)
- Published
- 2019
- Full Text
- View/download PDF
23. One NF1 Mutation may Conceal Another.
- Author
-
Pacot L, Burin des Roziers C, Laurendeau I, Briand-Suleau A, Coustier A, Mayard T, Tlemsani C, Faivre L, Thomas Q, Rodriguez D, Blesson S, Dollfus H, Muller YG, Parfait B, Vidaud M, Gilbert-Dussardier B, Yardin C, Dauriat B, Derancourt C, Vidaud D, and Pasmant E
- Subjects
- Adolescent, Adult, Cafe-au-Lait Spots complications, Cafe-au-Lait Spots pathology, Child, Female, Humans, Male, Mutation, Neurofibromatosis 1 complications, Neurofibromatosis 1 pathology, Pedigree, Adaptor Proteins, Signal Transducing genetics, Cafe-au-Lait Spots genetics, Neurofibromatosis 1 genetics, Neurofibromin 1 genetics, Phenotype
- 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 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.
- Published
- 2019
- Full Text
- View/download PDF
24. NF1 mutations identify molecular and clinical subtypes of lung adenocarcinomas.
- Author
-
Tlemsani C, Pécuchet N, Gruber A, Laurendeau I, Danel C, Riquet M, Le Pimpec-Barthes F, Fabre E, Mansuet-Lupo A, Damotte D, Alifano M, Luscan A, Rousseau B, Vidaud D, Varin J, Parfait B, Bieche I, Leroy K, Laurent-Puig P, Terris B, Blons H, Vidaud M, and Pasmant E
- Subjects
- Adenocarcinoma of Lung genetics, Adult, Aged, Aged, 80 and over, Female, High-Throughput Nucleotide Sequencing methods, Humans, Lung Neoplasms genetics, MAP Kinase Signaling System, Male, Middle Aged, Neoplasm Staging, Prognosis, Retrospective Studies, Sequence Analysis, DNA, Sequence Deletion, Smoking adverse effects, Smoking epidemiology, Smoking genetics, Survival Analysis, Adenocarcinoma of Lung pathology, Lung Neoplasms pathology, Mutation, Neurofibromin 1 genetics
- Abstract
The tumor suppressor gene neurofibromin 1 (NF1) is a major regulator of the RAS-MAPK pathway. NF1 mutations occur in lung cancer but were not extensively explored. We hypothesized that NF1-mutated tumors could define a specific population with a distinct clinical and molecular profile. We performed NF1 sequencing using next generation sequencing (NGS) in 154 lung adenocarcinoma surgical specimens with known KRAS, EGFR, TP53, BRAF, HER2, and PIK3CA status, to evaluate the molecular and clinical specificities of NF1-mutated lung cancers. Clinical data were retrospectively collected, and their associations with molecular profiles assessed. In this series, 24 tumors were NF1 mutated (17.5%) and 11 were NF1 deleted (8%). There was no mutation hotspot. NF1 mutations were rarely associated with other RAS-MAPK pathway mutations. Most of patients with NF1 alterations were males (74.3%) and smokers (74.3%). Overall survival and disease-free survival were statistically better in patients with NF1 alterations (N = 34) than in patients with KRAS mutations (N = 30) in univariate analysis. Our results confirm that NF1 is frequently mutated and represents a distinct molecular and clinical subtype of lung adenocarcinoma., (© 2019 The Authors. Cancer Medicine published by John Wiley & Sons Ltd.)
- Published
- 2019
- Full Text
- View/download PDF
25. Clinical and Molecular Features of 5 European Multigenerational Families With Moyamoya Angiopathy.
- Author
-
Grangeon L, Guey S, Schwitalla JC, Bergametti F, Arnould M, Corpechot M, Hadjadj J, Riant F, Aloui C, Drunat S, Vidaud D, Tournier-Lasserve E, and Kraemer M
- Subjects
- Adolescent, Adult, Age of Onset, Aged, Alleles, Child, Europe, Female, Gene Frequency, Genetic Association Studies, Humans, Male, Middle Aged, Moyamoya Disease genetics, Pedigree, Phosphoprotein Phosphatases genetics, Exome Sequencing, Young Adult, Adenosine Triphosphatases genetics, Genetic Predisposition to Disease, Moyamoya Disease diagnosis, Mutation, Ubiquitin-Protein Ligases genetics
- Abstract
Background and Purpose Moyamoya angiopathy (MMA) is a rare cerebral vasculopathy outside of Asia. In Japanese patients, a vast majority of patients carry the founder p.R4810K variant in the RNF213 gene, and familial cases are around 10%. In European patients, data about familial occurrence are limited. The aim of this study was to characterize the clinical and molecular features of several European families with a parent-to-child transmission of MMA. Methods Out of 126 MMA probands referred, we identified 113 sporadic probands and 13 familial probands. Segregation analysis showed a vertical parent-to-child pattern of inheritance in the families of 5 of these probands. All 5 families were of German or Dutch ancestry. We investigated the clinical features of affected members and used whole-exome sequencing to screen RNF213 and 13 genes involved in Mendelian MMA and to identify genes recurrently mutated in these families. Results Twelve affected MMA patients were identified, including 9 females and 3 males. Age at clinical onset ranged from 11 to 65 years. In 3 of 5 families, associated livedo racemosa was found. We did not detect any deleterious variants in the 13 known MMA genes. RNF213 rare missense variants predicted to be pathogenic were detected in all affected members of 2 of these families, as well as 2 candidate variants of the PALD1 gene. Conclusions Nonsyndromic MMA was identified in 5 European families, including 2 to 3 clinically affected cases segregating with a parent-to-child pattern of inheritance in each family. Molecular screening detected rare deleterious variants within RNF213 and PALD1 in all affected members of 2 of these 5 families, as well as in some clinically unaffected members. Altogether these data raise the difficult and, to date unanswered, question of the medical indication of presymptomatic screening.
- Published
- 2019
- Full Text
- View/download PDF
26. The molecular landscape of glioma in patients with Neurofibromatosis 1.
- Author
-
D'Angelo F, Ceccarelli M, Tala, Garofano L, Zhang J, Frattini V, Caruso FP, Lewis G, Alfaro KD, Bauchet L, Berzero G, Cachia D, Cangiano M, Capelle L, de Groot J, DiMeco F, Ducray F, Farah W, Finocchiaro G, Goutagny S, Kamiya-Matsuoka C, Lavarino C, Loiseau H, Lorgis V, Marras CE, McCutcheon I, Nam DH, Ronchi S, Saletti V, Seizeur R, Slopis J, Suñol M, Vandenbos F, Varlet P, Vidaud D, Watts C, Tabar V, Reuss DE, Kim SK, Meyronet D, Mokhtari K, Salvador H, Bhat KP, Eoli M, Sanson M, Lasorella A, and Iavarone A
- Subjects
- Adolescent, Adult, Antigens, Neoplasm metabolism, Brain Neoplasms immunology, Child, Child, Preschool, Cohort Studies, DNA Methylation genetics, Female, Germ-Line Mutation genetics, Glioma immunology, Humans, Male, Middle Aged, Neurofibromin 1 genetics, Reproducibility of Results, T-Lymphocytes immunology, Transcriptome genetics, X-linked Nuclear Protein genetics, Young Adult, Brain Neoplasms complications, Brain Neoplasms genetics, Glioma complications, Glioma genetics, Neurofibromatosis 1 complications, Neurofibromatosis 1 genetics
- Abstract
Neurofibromatosis type 1 (NF1) is a common tumor predisposition syndrome in which glioma is one of the prevalent tumors. Gliomagenesis in NF1 results in a heterogeneous spectrum of low- to high-grade neoplasms occurring during the entire lifespan of patients. The pattern of genetic and epigenetic alterations of glioma that develops in NF1 patients and the similarities with sporadic glioma remain unknown. Here, we present the molecular landscape of low- and high-grade gliomas in patients affected by NF1 (NF1-glioma). We found that the predisposing germline mutation of the NF1 gene was frequently converted to homozygosity and the somatic mutational load of NF1-glioma was influenced by age and grade. High-grade tumors harbored genetic alterations of TP53 and CDKN2A, frequent mutations of ATRX associated with Alternative Lengthening of Telomere, and were enriched in genetic alterations of transcription/chromatin regulation and PI3 kinase pathways. Low-grade tumors exhibited fewer mutations that were over-represented in genes of the MAP kinase pathway. Approximately 50% of low-grade NF1-gliomas displayed an immune signature, T lymphocyte infiltrates, and increased neo-antigen load. DNA methylation assigned NF1-glioma to LGm6, a poorly defined Isocitrate Dehydrogenase 1 wild-type subgroup enriched with ATRX mutations. Thus, the profiling of NF1-glioma defined a distinct landscape that recapitulates a subset of sporadic tumors.
- Published
- 2019
- Full Text
- View/download PDF
27. Targeted next-generation sequencing for differential diagnosis of neurofibromatosis type 2, schwannomatosis, and meningiomatosis.
- Author
-
Louvrier C, Pasmant E, Briand-Suleau A, Cohen J, Nitschké P, Nectoux J, Orhant L, Zordan C, Goizet C, Goutagny S, Lallemand D, Vidaud M, Vidaud D, Kalamarides M, and Parfait B
- Subjects
- Biomarkers, Tumor, Chromosomal Proteins, Non-Histone genetics, DNA-Binding Proteins genetics, Diagnosis, Differential, Follow-Up Studies, Humans, Meningeal Neoplasms genetics, Meningioma genetics, Neurilemmoma genetics, Neurofibromatoses genetics, Neurofibromatosis 2 genetics, Neurofibromin 2 genetics, Prognosis, Prospective Studies, Repressor Proteins genetics, Retrospective Studies, SMARCB1 Protein genetics, Skin Neoplasms genetics, Transcription Factors genetics, Genes, Tumor Suppressor, High-Throughput Nucleotide Sequencing methods, Meningeal Neoplasms diagnosis, Meningioma diagnosis, Mutation, Neurilemmoma diagnosis, Neurofibromatoses diagnosis, Neurofibromatosis 2 diagnosis, Skin Neoplasms diagnosis
- Abstract
Background: Clinical overlap between neurofibromatosis type 2 (NF2), schwannomatosis, and meningiomatosis can make clinical diagnosis difficult. Hence, molecular investigation of germline and tumor tissues may improve the diagnosis., Methods: We present the targeted next-generation sequencing (NGS) of NF2, SMARCB1, LZTR1, SMARCE1, and SUFU tumor suppressor genes, using an amplicon-based approach. We analyzed blood DNA from a cohort of 196 patients, including patients with NF2 (N = 79), schwannomatosis (N = 40), meningiomatosis (N = 12), and no clearly established diagnosis (N = 65). Matched tumor DNA was analyzed when available. Forty-seven NF2-/SMARCB1-negative schwannomatosis patients and 27 NF2-negative meningiomatosis patients were also evaluated., Results: A NF2 variant was found in 41/79 (52%) NF2 patients. SMARCB1 or LZTR1 variants were identified in 5/40 (12.5%) and 13/40 (∼32%) patients in the schwannomatosis cohort. Potentially pathogenic variants were found in 12/65 (18.5%) patients with no clearly established diagnosis. A LZTR1 variant was identified in 16/47 (34%) NF2/SMARCB1-negative schwannomatosis patients. A SMARCE1 variant was found in 3/39 (∼8%) meningiomatosis patients. No SUFU variant was found in the cohort. NGS was an effective and sensitive method to detect mutant alleles in blood or tumor DNA of mosaic NF2 patients. Interestingly, we identified a 4-hit mechanism resulting in the complete NF2 loss-of-function combined with SMARCB1 and LZTR1 haploinsufficiency in two-thirds of tumors from NF2 patients., Conclusions: Simultaneous investigation of NF2, SMARCB1, LZTR1, and SMARCE1 is a key element in the differential diagnosis of NF2, schwannomatosis, and meningiomatosis. The targeted NGS strategy is suitable for the identification of NF2 mosaicism in blood and for the investigation of tumors from these patients.
- Published
- 2018
- Full Text
- View/download PDF
28. SETD2 and DNMT3A screen in the Sotos-like syndrome French cohort.
- Author
-
Tlemsani C, Luscan A, Leulliot N, Bieth E, Afenjar A, Baujat G, Doco-Fenzy M, Goldenberg A, Lacombe D, Lambert L, Odent S, Pasche J, Sigaudy S, Buffet A, Violle-Poirsier C, Briand-Suleau A, Laurendeau I, Chin M, Saugier-Veber P, Vidaud D, Cormier-Daire V, Vidaud M, Pasmant E, and Burglen L
- Abstract
Background: Heterozygous NSD1 mutations were identified in 60%-90% of patients with Sotos syndrome. Recently, mutations of the SETD2 and DNMT3A genes were identified in patients exhibiting only some Sotos syndrome features. Both NSD1 and SETD2 genes encode epigenetic 'writer' proteins that catalyse methylation of histone 3 lysine 36 (H3K36me). The DNMT3A gene encodes an epigenetic 'reader' protein of the H3K36me chromatin mark., Methods: We aimed at confirming the implication of DNMT3A and SETD2 mutations in an overgrowth phenotype, through a comprehensive targeted-next generation sequencing (NGS) screening in 210 well-phenotyped index cases with a Sotos-like phenotype and no NSD1 mutation, from a French cohort., Results: Six unreported heterozygous likely pathogenic variants in DNMT3A were identified in seven patients: two nonsense variants and four de novo missense variants. One de novo unreported heterozygous frameshift variant was identified in SETD2 in one patient. All the four DNMT3A missense variants affected DNMT3A functional domains, suggesting a potential deleterious impact. DNMT3A -mutated index cases shared similar clinical features including overgrowth phenotype characterised by postnatal tall stature (≥+2SD), macrocephaly (≥+2SD), overweight or obesity at older age, intellectual deficiency and minor facial features. The phenotype associated with SETD2 mutations remains to be described more precisely. The p.Arg882Cys missense de novo constitutional DNMT3A variant found in two patients is the most frequent DNMT3A somatic mutation in acute leukaemia., Conclusions: Our results illustrate the power of targeted NGS to identify rare disease-causing variants. These observations provided evidence for a unifying mechanism (disruption of apposition and reading of the epigenetic chromatin mark H3K36me) that causes an overgrowth syndrome phenotype. Further studies are needed in order to assess the role of SETD2 and DNMT3A in intellectual deficiency without overgrowth., Competing Interests: Competing interests: None declared., (Published by the BMJ Publishing Group Limited. For permission to use (where not already granted under a licence) please go to http://www.bmj.com/company/products-services/rights-and-licensing/.)
- Published
- 2016
- Full Text
- View/download PDF
29. Copy number variants and rasopathies: germline KRAS duplication in a patient with syndrome including pigmentation abnormalities.
- Author
-
Gilbert-Dussardier B, Briand-Suleau A, Laurendeau I, Bilan F, Cavé H, Verloes A, Vidaud M, Vidaud D, and Pasmant E
- Subjects
- Adolescent, Cafe-au-Lait Spots genetics, Child, Chromosomes, Human, Pair 12 genetics, Female, Humans, Intracellular Signaling Peptides and Proteins genetics, MAP Kinase Kinase 2 genetics, Male, Mitogen-Activated Protein Kinases genetics, Noonan Syndrome genetics, Protein Tyrosine Phosphatase, Non-Receptor Type 11 genetics, Proto-Oncogene Proteins c-raf genetics, DNA Copy Number Variations genetics, Mutation genetics, Proto-Oncogene Proteins p21(ras) genetics
- Abstract
RAS/MAPK pathway germline mutations were described in Rasopathies, a class of rare genetic syndromes combining facial abnormalities, heart defects, short stature, skin and genital abnormalities, and mental retardation. The majority of the mutations identified in the Rasopathies are point mutations which increase RAS/MAPK pathway signaling. Duplications encompassing RAS/MAPK pathway genes (PTPN11, RAF1, MEK2, or SHOC2) were more rarely described. Here we report, a syndromic familial case of a 12p duplication encompassing the dosage sensitive gene KRAS, whose phenotype overlapped with rasopathies. The patient was referred because of a history of mild learning disabilities, small size, facial dysmorphy, and pigmentation abnormalities (café-au-lait and achromic spots, and axillar lentigines). This phenotype was reminiscent of rasopathies. No mutation was identified in the most common genes associated with Noonan, cardio-facio-cutaneous, Legius, and Costello syndromes, as well as neurofibromatosis type 1. The patient constitutional DNA exhibited a ~10.5 Mb duplication at 12p, including the KRAS gene. The index case's mother carried the same chromosome abnormality and also showed development delay with short stature, and numerous café-au-lait spots. Duplication of the KRAS gene may participate in the propositus phenotype, in particular of the specific pigmentation abnormalities. Array-CGH or some other assessment of gene/exon CNVs of RAS/MAPK pathway genes should be considered in the evaluation of individuals with rasopathies.
- Published
- 2016
- Full Text
- View/download PDF
30. Neurofibromatosis Type 1 Molecular Diagnosis: The RNA Point of View.
- Author
-
Pasmant E and Vidaud D
- Subjects
- Humans, Intracellular Signaling Peptides and Proteins, Membrane Proteins genetics, Neurofibromatosis 1 genetics, RNA
- Published
- 2016
- Full Text
- View/download PDF
31. Growth Hormone Deficiency in a Child with Neurofibromatosis-Noonan Syndrome.
- Author
-
Vurallı D, Gönç N, Vidaud D, Özön A, Alikaşifoğlu A, and Kandemir N
- Subjects
- Adolescent, DNA Mutational Analysis, Dwarfism, Pituitary etiology, Female, Human Growth Hormone adverse effects, Humans, Neurofibromatoses physiopathology, Noonan Syndrome physiopathology, Phenotype, Prognosis, Dwarfism, Pituitary diagnosis, Genes, Neurofibromatosis 1, Human Growth Hormone deficiency, Mutation genetics, Neurofibromatoses complications, Noonan Syndrome complications
- Abstract
Neurofibromatosis-Noonan syndrome (NFNS) is a distinct entity which shows the features of both NF1 (neurofibromatosis 1) and Noonan syndrome (NS). While growth hormone deficiency (GHD) has been relatively frequently identified in NF1 and NS patients, there is limited experience in NFNS cases. The literature includes only one case report of a NFNS patient having GHD and that report primarily focuses on the dermatological lesions that accompany the syndrome and not on growth hormone (GH) treatment. Here, we present a 13-year-old girl who had clinical features of NFNS with a mutation in the NF1 gene. The case is the first NFNS patient reported in the literature who was diagnosed to have GHD and who received GH treatment until reaching final height. The findings in this patient show that short stature is a feature of NFNS and can be caused by GHD. Patients with NFNS who show poor growth should be evaluated for GHD.
- Published
- 2016
- Full Text
- View/download PDF
32. RAS MAPK inhibitors deregulation in leukemia.
- Author
-
Pasmant E, Vidaud D, and Ballerini P
- Published
- 2015
- Full Text
- View/download PDF
33. Diagnosis of Constitutional Mismatch Repair-Deficiency Syndrome Based on Microsatellite Instability and Lymphocyte Tolerance to Methylating Agents.
- Author
-
Bodo S, Colas C, Buhard O, Collura A, Tinat J, Lavoine N, Guilloux A, Chalastanis A, Lafitte P, Coulet F, Buisine MP, Ilencikova D, Ruiz-Ponte C, Kinzel M, Grandjouan S, Brems H, Lejeune S, Blanché H, Wang Q, Caron O, Cabaret O, Svrcek M, Vidaud D, Parfait B, Verloes A, Knappe UJ, Soubrier F, Mortemousque I, Leis A, Auclair-Perrossier J, Frébourg T, Fléjou JF, Entz-Werle N, Leclerc J, Malka D, Cohen-Haguenauer O, Goldberg Y, Gerdes AM, Fedhila F, Mathieu-Dramard M, Hamelin R, Wafaa B, Gauthier-Villars M, Bourdeaut F, Sheridan E, Vasen H, Brugières L, Wimmer K, Muleris M, and Duval A
- Subjects
- Adaptor Proteins, Signal Transducing genetics, Adenosine Triphosphatases genetics, Adult, Brain Neoplasms drug therapy, Brain Neoplasms genetics, Brain Neoplasms metabolism, Brain Neoplasms pathology, Caco-2 Cells, Case-Control Studies, Colorectal Neoplasms drug therapy, Colorectal Neoplasms genetics, Colorectal Neoplasms metabolism, Colorectal Neoplasms pathology, Colorectal Neoplasms, Hereditary Nonpolyposis drug therapy, Colorectal Neoplasms, Hereditary Nonpolyposis genetics, Colorectal Neoplasms, Hereditary Nonpolyposis metabolism, Colorectal Neoplasms, Hereditary Nonpolyposis pathology, DNA Mutational Analysis, DNA Repair Enzymes genetics, DNA-Binding Proteins genetics, Female, Genetic Predisposition to Disease, HCT116 Cells, Heredity, Humans, Lymphocytes metabolism, Male, Methylation, Mismatch Repair Endonuclease PMS2, Multiplex Polymerase Chain Reaction, MutL Protein Homolog 1, MutS Homolog 2 Protein genetics, Neoplastic Syndromes, Hereditary drug therapy, Neoplastic Syndromes, Hereditary genetics, Neoplastic Syndromes, Hereditary metabolism, Neoplastic Syndromes, Hereditary pathology, Nuclear Proteins genetics, Phenotype, Predictive Value of Tests, Reproducibility of Results, Transfection, Young Adult, Antineoplastic Agents, Alkylating therapeutic use, Biomarkers, Tumor genetics, Brain Neoplasms diagnosis, Colorectal Neoplasms diagnosis, Colorectal Neoplasms, Hereditary Nonpolyposis diagnosis, Drug Resistance, Neoplasm, Genetic Testing methods, Germ-Line Mutation, Lymphocytes drug effects, Microsatellite Instability, Neoplastic Syndromes, Hereditary diagnosis
- Abstract
Background & Aims: Patients with bi-allelic germline mutations in mismatch repair (MMR) genes (MLH1, MSH2, MSH6, or PMS2) develop a rare but severe variant of Lynch syndrome called constitutional MMR deficiency (CMMRD). This syndrome is characterized by early-onset colorectal cancers, lymphomas or leukemias, and brain tumors. There is no satisfactory method for diagnosis of CMMRD because screens for mutations in MMR genes are noninformative for 30% of patients. MMR-deficient cancer cells are resistant to genotoxic agents and have microsatellite instability (MSI), due to accumulation of errors in repetitive DNA sequences. We investigated whether these features could be used to identify patients with CMMRD., Methods: We examined MSI by PCR analysis and tolerance to methylating or thiopurine agents (functional characteristics of MMR-deficient tumor cells) in lymphoblastoid cells (LCs) from 3 patients with CMMRD and 5 individuals with MMR-proficient LCs (controls). Using these assays, we defined experimental parameters that allowed discrimination of a series of 14 patients with CMMRD from 52 controls (training set). We then used the same parameters to assess 23 patients with clinical but not genetic features of CMMRD., Results: In the training set, we identified parameters, based on MSI and LC tolerance to methylation, that detected patients with CMMRD vs controls with 100% sensitivity and 100% specificity. Among 23 patients suspected of having CMMRD, 6 had MSI and LC tolerance to methylation (CMMRD highly probable), 15 had neither MSI nor LC tolerance to methylation (unlikely to have CMMRD), and 2 were considered doubtful for CMMRD based on having only 1 of the 2 features., Conclusion: The presence of MSI and tolerance to methylation in LCs identified patients with CMMRD with 100% sensitivity and specificity. These features could be used in diagnosis of patients., (Copyright © 2015 AGA Institute. Published by Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
34. Neurofibromatosis type 1 molecular diagnosis: what can NGS do for you when you have a large gene with loss of function mutations?
- Author
-
Pasmant E, Parfait B, Luscan A, Goussard P, Briand-Suleau A, Laurendeau I, Fouveaut C, Leroy C, Montadert A, Wolkenstein P, Vidaud M, and Vidaud D
- Subjects
- Adaptor Proteins, Signal Transducing, Computational Biology methods, Exons, Gene Duplication, Genes, Neurofibromatosis 1, High-Throughput Nucleotide Sequencing, Humans, Intracellular Signaling Peptides and Proteins genetics, Membrane Proteins genetics, Mosaicism, Mutation, Reproducibility of Results, Retrospective Studies, Sequence Deletion, Neurofibromatosis 1 diagnosis, Neurofibromatosis 1 genetics
- Abstract
Molecular diagnosis of neurofibromatosis type 1 (NF1) is challenging owing to the large size of the tumour suppressor gene NF1, and the lack of mutation hotspots. A somatic alteration of the wild-type NF1 allele is observed in NF1-associated tumours. Genetic heterogeneity in NF1 was confirmed in patients with SPRED1 mutations. Here, we present a targeted next-generation sequencing (NGS) of NF1 and SPRED1 using a multiplex PCR approach (230 amplicons of ∼150 bp) on a PGM sequencer. The chip capacity allowed mixing 48 bar-coded samples in a 4-day workflow. We validated the NGS approach by retrospectively testing 30 NF1-mutated samples, and then prospectively analysed 279 patients in routine diagnosis. On average, 98.5% of all targeted bases were covered by at least 20X and 96% by at least 100X. An NF1 or SPRED1 alteration was found in 246/279 (88%) and 10/279 (4%) patients, respectively. Genotyping throughput was increased over 10 times, as compared with Sanger, with ∼90[euro ] for consumables per sample. Interestingly, our targeted NGS approach also provided quantitative information based on sequencing depth allowing identification of multiexons deletion or duplication. We then addressed the NF1 somatic mutation detection sensitivity in mosaic NF1 patients and tumours.
- Published
- 2015
- Full Text
- View/download PDF
35. NF1 single and multi-exons copy number variations in neurofibromatosis type 1.
- Author
-
Imbard A, Pasmant E, Sabbagh A, Luscan A, Soares M, Goussard P, Blanché H, Laurendeau I, Ferkal S, Vidaud M, Pinson S, Bellanne-Chantelot C, Vidaud D, Wolkenstein P, and Parfait B
- Subjects
- Adolescent, Adult, Child, Comparative Genomic Hybridization, Female, Gene Order, Humans, Male, Middle Aged, Mutation, Neurofibromatosis 1 diagnosis, Young Adult, DNA Copy Number Variations, Exons, Genes, Neurofibromatosis 1, Neurofibromatosis 1 genetics
- Abstract
Neurofibromatosis type 1 (NF1) is caused by dominant loss-of-function mutations of the tumor suppressor NF1 containing 57 constitutive coding exons. A huge number of different pathogenic NF1 alterations has been reported. The aim of the present study was to evaluate the usefulness of a multiplex ligation-dependent probe amplification (MLPA) approach in NF1 patients to detect single and multi-exon NF1 gene copy number variations. A genotype-phenotype correlation was then performed in NF1 patients carrying these types of genetic alterations. Among 565 NF1 index cases from the French NF1 cohort, single and multi-exon deletions/duplications screening identified NF1 partial deletions/duplications in 22 patients (~4%) using MLPA analysis. Eight single exon deletions, 11 multiple exons deletions, 1 complex rearrangement and 2 duplications were identified. All results were confirmed using a custom array-CGH. MLPA and custom array-CGH allowed the identification of rearrangements that were missed by cDNA/DNA sequencing or microsatellite analysis. We then performed a targeted next-generation sequencing of NF1 that allowed confirmation of all 22 rearrangements. No clear genotype-phenotype correlations were found for the most clinically significant disease features of NF1 in patients with single and multi-exons NF1 gene copy number changes.
- Published
- 2015
- Full Text
- View/download PDF
36. SPRED1, a RAS MAPK pathway inhibitor that causes Legius syndrome, is a tumour suppressor downregulated in paediatric acute myeloblastic leukaemia.
- Author
-
Pasmant E, Gilbert-Dussardier B, Petit A, de Laval B, Luscan A, Gruber A, Lapillonne H, Deswarte C, Goussard P, Laurendeau I, Uzan B, Pflumio F, Brizard F, Vabres P, Naguibvena I, Fasola S, Millot F, Porteu F, Vidaud D, Landman-Parker J, and Ballerini P
- Subjects
- Adaptor Proteins, Signal Transducing, Adolescent, Cafe-au-Lait Spots complications, Cafe-au-Lait Spots pathology, Child, Child, Preschool, Female, Gene Expression Regulation, Neoplastic, Genes, Tumor Suppressor, Humans, Infant, Infant, Newborn, Intracellular Signaling Peptides and Proteins biosynthesis, Leukemia, Myeloid, Acute complications, Leukemia, Myeloid, Acute pathology, Loss of Heterozygosity genetics, Male, Membrane Proteins biosynthesis, Mutation, Neurofibromin 1 genetics, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma complications, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma pathology, Cafe-au-Lait Spots genetics, Genes, ras genetics, Intracellular Signaling Peptides and Proteins genetics, Leukemia, Myeloid, Acute genetics, Membrane Proteins genetics, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma genetics
- Abstract
Constitutional dominant loss-of-function mutations in the SPRED1 gene cause a rare phenotype referred as neurofibromatosis type 1 (NF1)-like syndrome or Legius syndrome, consisted of multiple café-au-lait macules, axillary freckling, learning disabilities and macrocephaly. SPRED1 is a negative regulator of the RAS MAPK pathway and can interact with neurofibromin, the NF1 gene product. Individuals with NF1 have a higher risk of haematological malignancies. SPRED1 is highly expressed in haematopoietic cells and negatively regulates haematopoiesis. SPRED1 seemed to be a good candidate for leukaemia predisposition or transformation. We performed SPRED1 mutation screening and expression status in 230 paediatric lymphoblastic and acute myeloblastic leukaemias (AMLs). We found a loss-of-function frameshift SPRED1 mutation in a patient with Legius syndrome. In this patient, the leukaemia blasts karyotype showed a SPRED1 loss of heterozygosity, confirming SPRED1 as a tumour suppressor. Our observation confirmed that acute leukaemias are rare complications of the Legius syndrome. Moreover, SPRED1 was significantly decreased at RNA and protein levels in the majority of AMLs at diagnosis compared with normal or paired complete remission bone marrows. SPRED1 decreased expression correlated with genetic features of AML. Our study reveals a new mechanism which contributes to deregulate RAS MAPK pathway in the vast majority of paediatric AMLs.
- Published
- 2015
- Full Text
- View/download PDF
37. Atypical hematologic and renal manifestations in neurofibromatosis type I: coincidence or pathophysiological link?
- Author
-
Van-Gils J, Harambat J, Jubert C, Vidaud D, Llanas B, Perel Y, Lacombe D, and Goizet C
- Subjects
- Adolescent, Adult, Cafe-au-Lait Spots diagnosis, Cafe-au-Lait Spots genetics, Child, Humans, Male, Mutation, Missense, Nephrotic Syndrome genetics, Neurofibromatosis 1 genetics, Neurofibromin 1 genetics, Pedigree, Proto-Oncogene Mas, Nephrotic Syndrome diagnosis, Neurofibromatosis 1 diagnosis
- Abstract
Neurofibromatosis type 1 (NF1) is an autosomal dominant, multi-system, neurocutaneous disorder that predisposes to the development of benign and malignant tumors with a birth incidence rate of 1 in 2500-3000. 50% of cases are sporadic. The diagnosis is exclusively based on clinical assessment with clinical diagnostic criteria such as café-au-lait spots, neurofibromas, axillary or groin freckling, Lisch nodules, optic pathway glioma, bony dysplasia and first-degree relative with NF1. We report a family with NF1 in which two members presented atypical clinical features in addition to the classical diagnostic criteria. Three relatives affected by NF1, a father and two of his three sons, are described. The clinical diagnosis was originally worn in all three cases, with the association many spots café-au -lait over the entire body and some axillary freckling as well as first-degree relative. One case presented an Acute Myeloid Leukemia (AML) type 2 at 10 years of age diagnosed before the revelation of bicytopenia associated pallor and isolated asthenia. A second case presented a nephrotic syndrome at 4 years of age due to the association of hydrops with headache and asthenia. Direct sequencing of NF1 led to identify the familial mutation, a previously unreported heterozygous missense mutation c.3443C > A, p.Ala1148Glu in exon 20 which segregated with all three affected patients. The family described in this report confirms the high clinical variability of NF1, even intrafamilial, and raises the question as to whether rare features such as AML and nephrotic syndrome are associated with NF1. Some NF1 patients presenting glomerular diseases or AML have rarely been reported, but due to the small number of cases described the mechanisms underlying these associations are poorly understood. However, it seems important to be aware of the possible occurrence of nephritic syndrome and/or malignant blood diseases in NF1 patients., (Copyright © 2014 Elsevier Masson SAS. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
38. [PRC2 alterations in NF1-associated malignant peripheral nerve sheath tumors: schwann cells with no complex].
- Author
-
Luscan A, Vidaud D, Ortonne N, Wolkenstein P, Vidaud M, and Pasmant E
- Subjects
- Biomarkers, Tumor metabolism, Gene Expression Regulation, Neoplastic, Histone Acetyltransferases metabolism, Histone Methyltransferases, Histone-Lysine N-Methyltransferase metabolism, Histones metabolism, Humans, Neurilemmoma pathology, Polycomb Repressive Complex 2 metabolism, Schwann Cells pathology, Carcinogenesis genetics, Genes, Neurofibromatosis 1 physiology, Mutation, Neurilemmoma genetics, Polycomb Repressive Complex 2 genetics, Schwann Cells metabolism
- Published
- 2014
- Full Text
- View/download PDF
39. The activation of the WNT signaling pathway is a Hallmark in neurofibromatosis type 1 tumorigenesis.
- Author
-
Luscan A, Shackleford G, Masliah-Planchon J, Laurendeau I, Ortonne N, Varin J, Lallemand F, Leroy K, Dumaine V, Hivelin M, Borderie D, De Raedt T, Valeyrie-Allanore L, Larousserie F, Terris B, Lantieri L, Vidaud M, Vidaud D, Wolkenstein P, Parfait B, Bièche I, Massaad C, and Pasmant E
- Subjects
- Biomarkers metabolism, Cell Line, Tumor, Endothelial Cells metabolism, Epithelial-Mesenchymal Transition, Fibroblasts metabolism, Gene Expression, Gene Expression Profiling, Humans, Immunophenotyping, Mast Cells metabolism, Neurofibroma genetics, Neurofibroma metabolism, Neurofibroma pathology, Neurofibromatosis 1 pathology, Neurofibromin 1 genetics, Neurofibromin 1 metabolism, RNA, Messenger genetics, Reproducibility of Results, Schwann Cells metabolism, Stem Cells metabolism, Cell Transformation, Neoplastic genetics, Cell Transformation, Neoplastic metabolism, Neurofibromatosis 1 genetics, Neurofibromatosis 1 metabolism, Wnt Signaling Pathway
- Abstract
Purpose: The hallmark of neurofibromatosis type 1 (NF1) is the onset of dermal or plexiform neurofibromas, mainly composed of Schwann cells. Plexiform neurofibromas can transform into malignant peripheral nerve sheath tumors (MPNST) that are resistant to therapies., Experimental Design: The aim of this study was to identify an additional pathway in the NF1 tumorigenesis. We focused our work on Wnt signaling that is highly implicated in cancer, mainly in regulating the proliferation of cancer stem cells. We quantified mRNAs of 89 Wnt pathway genes in 57 NF1-associated tumors including dermal and plexiform neurofibromas and MPNSTs. Expression of two major stem cell marker genes and five major epithelial-mesenchymal transition marker genes was also assessed. The expression of significantly deregulated Wnt genes was then studied in normal human Schwann cells, fibroblasts, endothelial cells, and mast cells and in seven MPNST cell lines., Results: The expression of nine Wnt genes was significantly deregulated in plexiform neurofibromas in comparison with dermal neurofibromas. Twenty Wnt genes showed altered expression in MPNST biopsies and cell lines. Immunohistochemical studies confirmed the Wnt pathway activation in NF1-associated MPNSTs. We then confirmed that the knockdown of NF1 in Schwann cells but not in epithelial cells provoked the activation of Wnt pathway by functional transfection assays. Furthermore, we showed that the protein expression of active β-catenin was increased in NF1-silenced cell lines. Wnt pathway activation was strongly associated to both cancer stem cell reservoir and Schwann-mesenchymal transition., Conclusion: We highlighted the implication of Wnt pathway in NF1-associated tumorigenesis., (©2013 AACR.)
- Published
- 2014
- Full Text
- View/download PDF
40. MicroRNAome profiling in benign and malignant neurofibromatosis type 1-associated nerve sheath tumors: evidences of PTEN pathway alterations in early NF1 tumorigenesis.
- Author
-
Masliah-Planchon J, Pasmant E, Luscan A, Laurendeau I, Ortonne N, Hivelin M, Varin J, Valeyrie-Allanore L, Dumaine V, Lantieri L, Leroy K, Parfait B, Wolkenstein P, Vidaud M, Vidaud D, and Bièche I
- Subjects
- Argonaute Proteins genetics, Cell Line, Tumor, Cluster Analysis, Humans, Neurofibroma, Plexiform genetics, Neurofibroma, Plexiform pathology, Proteins genetics, RNA-Binding Proteins, Ribonuclease III genetics, Sequence Homology, Nucleic Acid, Time Factors, Gene Expression Profiling, MicroRNAs genetics, Neurofibromatosis 1 genetics, Neurofibromatosis 1 pathology, PTEN Phosphohydrolase metabolism, Signal Transduction genetics
- Abstract
Background: Neurofibromatosis type 1 (NF1) is a common dominant tumor predisposition syndrome affecting 1 in 3,500 individuals. The hallmarks of NF1 are the development of peripheral nerve sheath tumors either benign (dermal and plexiform neurofibromas) or malignant (MPNSTs)., Results: To comprehensively characterize the role of microRNAs in NF1 tumorigenesis, we analyzed 377 miRNAs expression in a large panel of dermal and plexiform neurofibromas, and MPNSTs. The most significantly upregulated miRNA in plexiform neurofibromas was miR-486-3p that targets the major tumor suppressor gene, PTEN. We confirmed PTEN downregulation at mRNA level. In plexiform neurofibromas, we also report aberrant expression of four miRNAs involved in the RAS-MAPK pathway (miR-370, miR-143, miR-181a, and miR-145). In MPNSTs, significant deregulated miRNAs were involved in PTEN repression (miR-301a, miR-19a, and miR-106b), RAS-MAPK pathway regulation (Let-7b, miR-195, and miR-10b), mesenchymal transition (miR-200c, let-7b, miR-135a, miR-135b, and miR-9), HOX genes expression (miR-210, miR-196b, miR-10a, miR-10b, and miR-9), and cell cycle progression (miR-195, let-7b, miR-20a, miR-210, miR-129-3p, miR-449a, and miR-106b)., Conclusion: We confirmed the implication of PTEN in genesis of plexiform neurofibromas and MPNSTs in NF1. Markedly deregulated miRNAs might have potential diagnostic or prognostic value and could represent novel strategies for effective pharmacological therapies of NF1 tumors.
- Published
- 2013
- Full Text
- View/download PDF
41. Neurofibromatosis type 1: from genotype to phenotype.
- Author
-
Pasmant E, Vidaud M, Vidaud D, and Wolkenstein P
- Subjects
- Alleles, Animals, Disease Models, Animal, Genes, Modifier, Genetic Heterogeneity, Humans, Mice, Mutation, Neurofibromatosis 1 pathology, Genetic Association Studies, Genotype, Neurofibromatosis 1 genetics, Phenotype
- Abstract
Although neurofibromatosis 1 (NF1) is a common Mendelian disorder with autosomal-dominant inheritance, its expression is highly variable and unpredictable. Many NF1 patients have been genotyped but few allele-phenotype correlations have been identified. NF1 genotype-phenotype correlations are difficult to identify because of the complexity of the NF1 phenotype, its strong age dependency, the relatedness of many clinical features and the huge heterogeneity of pathogenic NF1 mutations. Some NF1 patients with a given NF1 mutation may develop very severe disease while others with the same mutation have only mild symptoms. This phenotypic variability may be due to both modifier genes and environmental factors. Recent targeted strategies have identified several interesting candidate modifier genes.
- Published
- 2012
- Full Text
- View/download PDF
42. First description of ABCB4 gene deletions in familial low phospholipid-associated cholelithiasis and oral contraceptives-induced cholestasis.
- Author
-
Pasmant E, Goussard P, Baranes L, Laurendeau I, Quentin S, Ponsot P, Consigny Y, Farges O, Condat B, Vidaud D, Vidaud M, Chen JM, and Parfait B
- Subjects
- ATP Binding Cassette Transporter, Subfamily B deficiency, Adult, Base Sequence, Cholestasis, Intrahepatic chemically induced, Chromosomes, Human, Pair 7, Comparative Genomic Hybridization, Female, Humans, Male, Middle Aged, Pedigree, Syndrome, Young Adult, ATP Binding Cassette Transporter, Subfamily B genetics, Cholelithiasis genetics, Cholestasis, Intrahepatic genetics, Contraceptives, Oral adverse effects, Gallbladder Diseases genetics, Gene Deletion, Pregnancy Complications genetics
- Abstract
The wide clinical spectrum of the ABCB4 gene (ATP-binding cassette subfamily B member 4) deficiency syndromes in humans includes low phospholipid-associated cholelithiasis (LPAC), intrahepatic cholestasis of pregnancy (ICP), oral contraceptives-induced cholestasis (CIC), and progressive familial intrahepatic cholestasis type 3 (PFIC3). No ABCB4 mutations are found in a significant proportion of patients with these syndromes. In the present study, 102 unrelated adult patients with LPAC (43 patients) or CIC/ICP (59 patients) were screened for ABCB4 mutations using DNA sequencing. Heterozygous ABCB4 point or short insertion/deletion mutations were found in 37% (16/43) of the LPAC patients and in 27% (16/59) of the ICP/CIC patients. High-resolution gene dosage methodologies were used in the 70 negative patients. Here, we describe for the first time ABCB4 partial or complete heterozygous deletions in 7% (3/43) of the LPAC patients, and in 2% (1/59) of the ICP/CIC patients. Our observations urge to systematically test patients with LPAC, ICP/CIC, and also children with PFIC3 for the presence of ABCB4 deletions using molecular tools allowing detection of gross rearrangements. In clinical practice, a comprehensive ABCB4 alteration-screening algorithm will permit the use of ABCB4 genotyping to confirm the diagnosis of LPAC or ICP/CIC, and allow familial testing. An early diagnosis of these biliary diseases may be beneficial because of the preventive effect of ursodeoxycholic acid on biliary complications. Further comparative studies of patients with well-characterized genotypes (including deletions) and phenotypes will help determine whether ABCB4 mutation types influence clinical outcomes.
- Published
- 2012
- Full Text
- View/download PDF
43. Role of noncoding RNA ANRIL in genesis of plexiform neurofibromas in neurofibromatosis type 1.
- Author
-
Pasmant E, Sabbagh A, Masliah-Planchon J, Ortonne N, Laurendeau I, Melin L, Ferkal S, Hernandez L, Leroy K, Valeyrie-Allanore L, Parfait B, Vidaud D, Bièche I, Lantieri L, Wolkenstein P, and Vidaud M
- Subjects
- Adult, Aged, Chromosome Deletion, Cohort Studies, Comparative Genomic Hybridization, Female, France, Gene Expression Regulation, Neoplastic, Gene Frequency, Genetic Predisposition to Disease, Genome-Wide Association Study, Genotype, Humans, Linkage Disequilibrium, Male, Middle Aged, Phenotype, RNA, Long Noncoding, RNA, Messenger metabolism, RNA, Untranslated genetics, Reverse Transcriptase Polymerase Chain Reaction, Chromosomes, Human, Pair 9, Cyclin-Dependent Kinase Inhibitor p15 genetics, Cyclin-Dependent Kinase Inhibitor p16 genetics, Gene Deletion, Neurofibroma, Plexiform genetics, Neurofibromatosis 1 genetics, Polymorphism, Single Nucleotide, RNA, Untranslated metabolism
- Abstract
Background: Neurofibromatosis type 1 (NF1) is a tumor predisposition syndrome with a worldwide birth incidence of one in 2500. Genetic factors unrelated to the NF1 locus are thought to influence the number of plexiform neurofibromas (PNFs) in patients with NF1, but no factors have been identified to date., Methods: We used high-resolution array comparative genomic hybridization of tissue from 22 PNFs obtained from 18 NF1 patients to identify modifier genes involved in PNF development. We used a family-based association test for five previously identified cancer-susceptibility tag single-nucleotide polymorphisms (rs1063192, rs2151280, rs2218220, rs10757257, and rs7023329) located in chromosomal region 9p21.3 in 1105 subjects (740 NF1 patients and 365 non-affected relatives) from 306 families. To confirm the functional role of rs2151280, we used real-time quantitative reverse transcription-polymerase chain reaction to analyze the expression of cyclin-dependent kinase inhibitor 2A (CDKN2A), CDKN2B, alternate reading frame (ARF), and antisense noncoding RNA in the INK4 locus (ANRIL) in the peripheral blood of 124 NF1 patients. Relationships between CDKN2A, CDKN2B, ARF, and ANRIL expression and the rs2151280 genotype were tested by the Kruskal-Wallis test. All statistical tests were two-sided., Results: In NF1-associated PNFs, 9p21.3 deletions (including the CDKN2A/B-ANRIL locus) were found as the only recurrent somatic alterations. Single-nucleotide polymorphism rs2151280 (located in ANRIL) was statistically significantly associated with the number of PNFs (P < .001) in NF1 patients. In addition, allele T of rs2151280 was statistically significantly associated with reduced ANRIL transcript levels (P < .001), suggesting that modulation of ANRIL expression mediates PNF susceptibility., Conclusion: Identification of ANRIL as a modifier gene in NF1 may offer clues to the molecular pathogenesis of PNFs, particularly neurofibroma formation, and emphasizes the unanticipated role of large noncoding RNA in activation of critical regulators of tumor development.
- Published
- 2011
- Full Text
- View/download PDF
44. Identification of genes potentially involved in the increased risk of malignancy in NF1-microdeleted patients.
- Author
-
Pasmant E, Masliah-Planchon J, Lévy P, Laurendeau I, Ortonne N, Parfait B, Valeyrie-Allanore L, Leroy K, Wolkenstein P, Vidaud M, Vidaud D, and Bièche I
- Subjects
- Carrier Proteins genetics, Cell Line, Tumor, GTPase-Activating Proteins genetics, Gene Expression Profiling, Gene Expression Regulation, Neoplastic, Genetic Predisposition to Disease, Humans, Membrane Proteins genetics, MicroRNAs genetics, Open Reading Frames genetics, Risk Factors, Schwann Cells metabolism, Ubiquitin-Protein Ligases, Biomarkers, Tumor genetics, Gene Deletion, Genes, Neurofibromatosis 1, Nerve Sheath Neoplasms genetics
- Abstract
Patients with NF1 microdeletion develop more neurofibromas at a younger age, and have an increased risk of malignant peripheral nerve sheath tumors (MPNSTs). We postulated that the increased risk of malignancy could be due to inactivation, in addition to NF1, of a second tumor suppressor gene located in the typical 1.4-Mb microdeletion found in most of the microdeleted patients. We investigated the expression of NF1, the other 16 protein-coding genes and the 2 microRNAs located in the 1.4-Mb microdeletion by means of real-time quantitative reverse-transcription polymerase chain reaction (RT-PCR) in a large series of human dermal and plexiform neurofibromas and MPNSTs. Five genes were significantly upregulated: OMG and SUZ12 in plexiform neurofibromas and ATAD5, EVI2A and C17orf79 in MPNSTs. More interestingly, two genes were significantly downregulated (RNF135 and CENTA2) in tumor Schwann cells from MPNST biopsies and in MPNST cell lines. This study points to the involvement of several genes (particularly RNF135 and CENTA2) in the increased risk of malignancy observed in NF1-microdeleted patients.
- Published
- 2011
- Full Text
- View/download PDF
45. [The amazing story of ANRIL, a long non-coding RNA].
- Author
-
Pasmant E, Laurendeau I, Sabbagh A, Parfait B, Vidaud M, Vidaud D, and Bièche I
- Subjects
- Cardiovascular Diseases genetics, Genes, Tumor Suppressor, Homeostasis genetics, Humans, Neoplasms genetics, RNA, Untranslated genetics, Tumor Suppressor Protein p14ARF genetics, RNA, Untranslated physiology
- Published
- 2010
- Full Text
- View/download PDF
46. Epithelial-to-mesenchymal transition and oncogenic Ras expression in resistance to the protein kinase Cbeta inhibitor enzastaurin in colon cancer cells.
- Author
-
Serova M, Astorgues-Xerri L, Bieche I, Albert S, Vidaud M, Benhadji KA, Emami S, Vidaud D, Hammel P, Theou-Anton N, Gespach C, Faivre S, and Raymond E
- Subjects
- Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Apoptosis drug effects, Apoptosis genetics, Carcinoma drug therapy, Carcinoma genetics, Carcinoma pathology, Cell Dedifferentiation drug effects, Cell Dedifferentiation genetics, Colonic Neoplasms drug therapy, Colonic Neoplasms genetics, Drug Evaluation, Preclinical, Drug Resistance, Neoplasm drug effects, Drug Resistance, Neoplasm genetics, Epithelial Cells drug effects, Gene Expression Regulation, Neoplastic drug effects, HCT116 Cells, HT29 Cells, Humans, Indoles therapeutic use, Mesenchymal Stem Cells drug effects, Mesenchymal Stem Cells pathology, Protein Kinase C beta, Protein Kinase Inhibitors pharmacology, Protein Kinase Inhibitors therapeutic use, Tumor Cells, Cultured, Colonic Neoplasms pathology, Epithelial Cells pathology, Genes, ras genetics, Indoles pharmacology, Protein Kinase C antagonists & inhibitors
- Abstract
Identifying molecular factors of sensitivity and resistance of cancer cells to enzastaurin, a drug inhibiting protein kinase C (PKC) beta, remains a major challenge to improve its clinical development. Investigating the cellular effects of enzastaurin in a panel of 20 human cancer cell lines, we found that most cells displaying oncogenic K-Ras mutations also display resistance to enzastaurin. Wild-type (WT) K-Ras cancer cells displaying high sensitivity to enzastaurin also expressed high mRNA levels of epithelial markers, such as E-cadherin (CDH1), and low mRNA expressions of mesenchymal markers, such as vimentin, N-cadherin (CDH2), and other genes frequently expressed in mesenchymal transition such as ZEB1, TWIST, SLUG, SNAIL, and TGFbeta. WT K-Ras enzastaurin-resistant cells also expressed high levels of mesenchymal markers. Based on this observation, the effects of enzastaurin were investigated in epithelial colon COLO205-S cells that expressed WT Ras/Raf and its derived COLO205-R mesenchymal counterpart selected for resistance to most PKC modulators and displaying oncogenic K-Ras (G13D/exon 2). In COLO205-S cells, inhibition of phosphorylated PKCbeta led to the inactivation of AKT and glycogen synthase kinase 3beta and was associated with apoptosis without significant effect on cell cycle progression. In COLO205-R cells, enzastaurin induced mainly necrosis at high concentrations. In COLO205-R cells, a strong activation of extracellular signal-regulated kinase 1/2 possibly due to oncogenic K-Ras was predominantly associated with transcription of potent antiapoptotic genes, such as BCL2, GADD45B, and CDKN1A, as well as the multidrug resistance gene ABCB1. From this study, colon cancer cells undergoing apoptosis under enzastaurin exposure seem to frequently express a WT Ras and an epithelial phenotype.
- Published
- 2010
- Full Text
- View/download PDF
47. Increase in type II collagen turnover after iron depletion in patients with hereditary haemochromatosis.
- Author
-
Richette P, Eymard C, Deberg M, Vidaud D, de Kerguenec C, Valla D, Vicaut E, Bardin T, and Henrotin Y
- Subjects
- Adult, Biomarkers blood, Biomarkers metabolism, Collagen Type II blood, Collagen Type II genetics, Female, Ferritins blood, Hemochromatosis genetics, Hemochromatosis metabolism, Humans, Iron metabolism, Male, Middle Aged, Phlebotomy methods, Cartilage, Articular metabolism, Collagen Type II metabolism, Ferritins metabolism, Hemochromatosis therapy, Iron Deficiencies
- Abstract
Objective: To determine the effects of iron depletion on serum levels of joint biomarkers and on joint symptoms in patients with hereditary haemochromatosis (HH)., Methods: Levels of biomarkers were measured in 18 patients with HH at the time of diagnosis and after iron depletion. The markers were type II collagen degradation (Coll2-1) and its nitrated form (Coll2-1NO(2)), type II procollagen synthesis (CPII), MPO, COMP and HA. For each patient, demographic data were collected and the global joint pain (visual analogue scale) was assessed before and after iron depletion by phlebotomy., Results: A total of 18 patients [10 males; mean (s.d.) age 48 (11) years] were homozygous for the C282Y mutation. No patient had liver dysfunction. Ferritin level before iron removal was 627.5 (range 133-3276) microg/l, and duration of the iron depletion phase was 295 (70-670) days. Serum levels of both Coll2-1 and CPII were significantly increased from diagnosis after iron depletion: 80.1 (55.6-113.5) vs 96.0 (48.8-136.3) nM (P = 0.004) and 731.4 (374.2-1012.3) vs 812.8 (535.8-1165.6) ng/ml (P = 0.03), respectively. Levels of other biomarkers were not modified by iron depletion. Ferritin level, which at baseline was correlated with body iron store (r = 0.63; P = 0.008), was significantly correlated with HA level measured before iron depletion (r = 0.60; P = 0.01). Global joint pain was not correlated with ferritin concentration and did not significantly decrease after iron depletion: 43 (19-73) vs 36 (16-67) mm (P = 0.07)., Conclusions: In patients with HH, cartilage homoeostasis is modified by iron excess and an increase in type II collagen turnover occurs after excess iron removal.
- Published
- 2010
- Full Text
- View/download PDF
48. Detection and characterization of NF1 microdeletions by custom high resolution array CGH.
- Author
-
Pasmant E, Sabbagh A, Masliah-Planchon J, Haddad V, Hamel MJ, Laurendeau I, Soulier J, Parfait B, Wolkenstein P, Bièche I, Vidaud M, and Vidaud D
- Subjects
- Humans, Microsatellite Repeats genetics, Mutation genetics, Neurofibromatosis 1 genetics, Polymerase Chain Reaction, Comparative Genomic Hybridization methods, Neurofibromin 1 genetics
- Abstract
In 5% to 10% of cases, neurofibromatosis type 1 is caused by microdeletions scattered across the entire NF1 gene and various neighboring genes. The phenotype appears to be more severe in patients with NF1 microdeletions than in patients with NF1 single point mutations. We have developed a new method for detecting and characterizing NF1 microdeletions based on a custom high-resolution oligonucleotide array comparative genomic hybridization by using the custom 8x15K Agilent array format. The array comprised a total of 14,207 oligonucleotide probes spanning the whole of chromosome 17, including 12,314 probes spanning an approximately 8 Mb interval surrounding the NF1 locus. We validated this approach by testing NF1 microdeleted DNA samples previously characterized by means of microsatellites and real-time PCR methods. Our array comparative genomic hybridization provided enough information for subsequent long-range PCR and nucleotide sequencing of the microdeletion endpoints. Unlike previously described methods, our array comparative genomic hybridization was able to unambiguously differentiate between the three types of microdeletions (type I, type II, and atypical) and to characterize atypical microdeletions. Further comparative studies of patients with well-characterized genotypes and phenotypes and different microdeletions sizes and breakpoints will help determine whether haploinsufficiency of deleted genes and/or genes rearrangements influence clinical outcomes.
- Published
- 2009
- Full Text
- View/download PDF
49. SPRED1 disorder and predisposition to leukemia in children.
- Author
-
Pasmant E, Ballerini P, Lapillonne H, Perot C, Vidaud D, Leverger G, and Landman-Parker J
- Subjects
- Adaptor Proteins, Signal Transducing, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Appendicitis complications, Cell Transformation, Neoplastic genetics, Genetic Predisposition to Disease, Humans, Infant, Intracellular Signaling Peptides and Proteins genetics, Intracellular Signaling Peptides and Proteins physiology, Leukemia, Monocytic, Acute drug therapy, MAP Kinase Signaling System, Male, Membrane Proteins genetics, Membrane Proteins physiology, Pseudomonas Infections complications, Remission Induction, Sepsis complications, Genes, ras, Heart Defects, Congenital genetics, Intracellular Signaling Peptides and Proteins deficiency, Leukemia, Monocytic, Acute genetics, Membrane Proteins deficiency, Neurocutaneous Syndromes genetics
- Published
- 2009
- Full Text
- View/download PDF
50. SPRED1 germline mutations caused a neurofibromatosis type 1 overlapping phenotype.
- Author
-
Pasmant E, Sabbagh A, Hanna N, Masliah-Planchon J, Jolly E, Goussard P, Ballerini P, Cartault F, Barbarot S, Landman-Parker J, Soufir N, Parfait B, Vidaud M, Wolkenstein P, Vidaud D, and France RN
- Subjects
- Adaptor Proteins, Signal Transducing, Adolescent, Adult, Aged, Aged, 80 and over, Child, Child, Preschool, DNA Mutational Analysis, Female, Gene Dosage, Genetic Predisposition to Disease, Humans, Male, Middle Aged, Pedigree, Germ-Line Mutation, Intracellular Signaling Peptides and Proteins genetics, Membrane Proteins genetics, Neurofibromatosis 1 genetics, Neurofibromin 1 genetics
- Abstract
Objective: Germline loss-of-function mutations in the SPRED1 gene have recently been identified in patients fulfilling the National Institutes of Health (NIH) diagnostic criteria for neurofibromatosis type 1 (NF1) but with no NF1 (neurofibromin 1) mutation found, suggesting a neurofibromatosis type 1-like syndrome., Methods: 61 index cases with NF1 clinical diagnosis but no identifiable NF1 mutation were screened for SPRED1 mutation., Results: We describe one known SPRED1 mutation (c.190C>T leading to p.Arg64Stop) and four novel mutations (c.637C>T leading to p.Gln213Stop, c.2T>C leading to p.Met1Thr, c.46C>T leading to p.Arg16Stop, and c.1048_1060del leading to p.Gly350fs) in five French families. Their NF1-like phenotype was characterised by a high prevalence of café-au-lait spots, freckling, learning disability, and an absence of neurofibromas and Lisch nodules in agreement with the original description. However, we did not observe Noonan-like dysmorphy. It is noteworthy that one patient with the p.Arg16Stop mutation developed a monoblastic acute leukaemia., Conclusions: In our series, SPRED1 mutations occurred with a prevalence of 0.5% in NF1 patients and in 5% of NF1 patients displaying an NF1-like phenotype. SPRED1 mutated patients did not display any specific dermatologic features that were not present in NF1 patients, except for the absence of neurofibromas that seem to be a specific clinical feature of NF1. The exact phenotypic spectrum and the putative complications of this NF1 overlapping syndrome, in particular haematological malignancies, remain to be further characterised. NIH diagnostic criteria for NF1 must be revised in view of this newly characterised Legius syndrome in order to establish a specific genetic counselling.
- Published
- 2009
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.