Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where N. Lannoy is active.

Publication


Featured researches published by N. Lannoy.


Human Genetics | 1997

Novel germline mutations in the APC gene and their phenotypic spectrum in familial adenomatous polyposis kindreds.

Corinne Walon; Alex Kartheuser; Geneviève Michils; M Smaers; N. Lannoy; Patrick Ngounou; Guy Mertens; Christine Verellen-Dumoulin

Abstract Among 23 germline mutations identified in the APC screening of 45 familial adenomatous polyposis (FAP) patients, we have found 10 different novel frameshift mutations in 11 apparently unrelated patients. In two cases, an additional missense mutation was detected. One previously described as a causative germline mutation (S2621C), associated with a 1-bp insertion (4684insA) on the opposite allele, did not segregate with the FAP phenotype in the family and was therefore considered as being non-pathogenic. The other (Z1625H) was located 2 codons before a 1-bp deletion (4897delC). Both mutations were transmitted together from an FAP father to his affected son. The FAP phenotype of these 10 novel truncating mutations was clinically documented within their kindreds. Important variability was observed in the phenotype. Interestingly, we noted that a mutation (487insT) localized at the boundary of the 5’ attenuated APC phenotype region in two unrelated families resulted in classical polyposis. A clear-cut genotype-phenotype correlation could be drawn in only two instances. In one family, a 4684insA mutation led to a mild polyposis associated with early inherited osteomas and, in the family bearing the double mutation (Z1625H+4897delC), the phenotype was obviously a 3′ attenuated type. Our data illustrate the wide genetic and phenotypic heterogeneity of this condition between and within the families, making the establishment of correlations complex and any prediction in this disease difficult, although targeting the mutation site may be helpful in some specific cases.


Haemophilia | 2009

Identification of de novo deletion in the factor VIII gene by MLPA technique in two girls with isolated factor VIII deficiency

N. Lannoy; I. Abinet; Karin Dahan; Cédric Hermans

Summary.  Blood coagulation evaluation performed preoperatively in two unrelated girls with isolated prolongation of the activated partial thromboplastin time (APTT) and no family history of bleeding disorder revealed a mild factor VIII deficiency. Quantitative and qualitative defect of von Willebrand factor was not present. Genetic analysis of the F8 gene identified no mutations. In contrast, quantitative gene screening using multiplex ligation dependent probe amplification (MLPA) revealed a large heterozygous deletion of the F8 gene in both patients consistent with a carrier status of sporadic severe haemophilia A. This report illustrates that MLPA technique represents an efficient method to screen for large F8 gene deletions in sporadic undiagnosed carriers of haemophilia A.


European Journal of Human Genetics | 2013

Intron 22 homologous regions are implicated in exons 1–22 duplications of the F8 gene

N. Lannoy; Bernard Grisart; Stéphane Eeckhoudt; Christine Verellen-Dumoulin; Catherine Lambert; Miikka Vikkula; Cédric Hermans

The intron 22 inversion found in up to 50% of severe hemophilia A patients results from a recombination between three intron 22 homologous copies (int22h). This study evaluated the implication of these copies in the formation of extended duplications comprising exons 1–22 of the factor 8 (F8) gene and their association with hemophilia and mental retardation. Two hemophilic patients with moderate and severe phenotypes and a third nonhemophilic patient with developmental delay were studied. All exhibited a duplication of F8 gene exons 1–22 identified by multiplex ligation-dependent probe amplification along with abnormal patterns on Southern blotting and unexpected long-range PCR amplification. Breakpoint analysis using array comparative genomic hybridization was performed to delimit the extent of these rearrangements. These duplications were bounded on one side by the F8 intragenic int22h-1 repeat and on the other side by extragenic int22h-2 or int22h-3 copies. However, the simultaneous identification of a second duplication containing F8 gene exons 2–14 for the moderate patient and the classical intron 22 inversion for the severe patient are considered in this study as the genetic causal defects of hemophilia. This study shows that the well-known int22h copies are involved in extended duplications comprising F8 gene exons 1–22. These specific duplications are probably not responsible for hemophilia and intellectual disability, but should be carefully considered in genetic counseling, while continuing to investigate the causal mutation of hemophilia.


Haemophilia | 2010

The 'royal disease'- haemophilia A or B? A haematological mystery is finally solved

N. Lannoy; Cédric Hermans

Summary.  ‘History can change blood. And blood can change the course of history’. Haemophilia is an illustration of this, as this congenital hereditary coagulation disorder, passed through the majority of royal European families at the beginning of the 20th century by Queen Victoria of England and Empress of the Indies, had indisputable political consequences, which led to one of the most defining moments of contemporary history: the Bolshevik Revolution. Today, none of Queen Victoria’s living descendents carry haemophilia. Because of this, the characterization of haemophilia (deficit of either factor VIII or XI) and the identification of the causal mutation are rendered impossible. In 1991, a tomb containing the remains of Czar Nicolas II’s entire family was discovered. A second tomb was discovered in 2007, allowing Russian and American scientists to fill in this gap in medical history. Following a scientific approach combining current genetic experimentation tools and the development of biological information technology, researchers were able to identify each body, allowing them to obtain precious genetic material from the young Czar Alexis, who was stricken by the disease, which revealed a causal substitution in the splice acceptor site of exon 4 in the F9 gene. This mutation that is responsible for haemophilia B had traumatized European royal families throughout the 20th century!


Haemophilia | 2012

Computational and molecular approaches for predicting unreported causal missense mutations in Belgian patients with haemophilia A.

N. Lannoy; I. Abinet; A Bosmans; Catherine Lambert; Christiane Vermylen; Cédric Hermans

Summary.  Haemophilia A (HA) is caused by widespread mutations in the factor VIII gene. The high spontaneous mutation rate of this gene means that roughly 40% of HA mutations are private. This study aimed to describe the approaches used to confirm private disease‐causing mutations in a cohort of Belgian HA patients. We studied 148 unrelated HA families for the presence of intron 22 and intron 1 inversion by Southern blotting and polymerase chain reaction (PCR). Multiplex ligation‐dependent probe amplification (MLPA) assay was used to detect large genomic rearrangements. Detection of point mutations was performed by DNA sequencing. Predicting the causal impact of new non‐synonymous changes was studied by two general strategies: (i) molecular approaches such as family cosegregation, evaluation of the implicated codon based on phylogenic separated species and absence of the mutation in the general Belgian population, and (ii) bioinformatics approaches to analyse the potential functional consequences of missense mutations. Among the 148 HA patients, in addition to common intron 22 and intron 1 inversions as well as large deletions or duplications, 67 different point mutations were identified, of which 42 had been reported in the HAMSTeRS database, and 25 were novel including 10 null variants for which RNA analyses confirmed the causal effect of mutations located in a splice site consensus and 15 missense mutations whose causality was demonstrated by molecular approaches and bioinformatics. This article reports several strategies to evaluate the deleterious consequences of unreported F8 substitutions in a large cohort of HA patients.


Hormone and Metabolic Research | 2012

Prevalence and spectrum of SDHx mutations in pheochromocytoma and paraganglioma in patients from Belgium: an update

Alexandre Persu; N. Lannoy; Dominique Maiter; Antonella Mendola; Pauline Montigny; Philippe Oriot; W Vinck; Pierre Garin; Marc Hamoir; Miikka Vikkula

Since the early 2000s, the prevalence and spectrum of mutations in genes encoding subunits of succinate dehydrogenase (SDHx) were reported in large cohorts of patients with pheochromocytoma (PC) and paraganglioma (PGL) from most Western countries. Unfortunately, in Belgium, no equivalent work was performed thus far. Therefore, the aim of the work was to look for mutations in SDHx genes and genotype-phenotype correlations in patients with PC and/or PGL from Belgium. Screening of the coding parts of SDHx genes and deletion search were performed in all patients with PC and/or PGL referred to the -Cliniques Universitaires Saint-Luc from 05/2003 to 05/2011. Genetic screening was performed in 59 unrelated head and neck (hn)PGLs (8 fami-lial) and 53 PCs (7 extra-adrenal; 3 metastatic). In hnPGLs, 10 different SDHD mutations (3 substitutions, 5 deletions, 2 splice site mutations) were detected in 16 patients, including 7 familial cases and 9 apparently sporadic cases. In the same subset, we found 8 different SDHB mutations (5 substitutions, 1 splice site mutation, 1 deletion, 1 duplication) in 10 patients with sporadic hnPGL without evidence of malignancy. No SDHx mutation was detected in patients harboring PCs and no SDHC mutation whatsoever. In conclusion, in our multicentric database of PC-PGLs from Belgium, (i) the prevalence of SDHx mutations was high in hnPGLs (44% in the whole subset, 37% of apparently sporadic cases); (ii) in sporadic cases, the prevalence of SDHB mutations was high (20%), similar to that of SDHD (18%); and (iii) no SDHx mutation was found in a subset of mostly adrenal, benign PCs.


Thrombosis Research | 2016

Five int22h homologous copies at the Xq28 locus identified in intron22 inversion type 3 of the Factor VIII gene

N. Lannoy; Marie Ravoet; Bernard Grisart; Mathilde Fretigny; Miikka Vikkula; Cédric Hermans

Five int22h homologous copies at the Xq28 locus identified in intron22 inversion type 3 of the Factor VIII gene.


Thrombosis Research | 2015

Overrepresentation of missense mutations in mild hemophilia A patients from Belgium: founder effect or independent occurrence?

N. Lannoy; Catherine Lambert; Miikka Vikkula; Cédric Hermans

Roughly 40% of observed mutations responsible for hemophilia A (HA) are novel and present in either a single family or a limited number of unrelated families. During routine diagnostic analysis of 73 unrelated Belgian patients with mild HA, 4 out of 43 different mutations (p.Ser2030Asn, p.Arg2178Cys, p.Arg2178His, and p.Pro2311His) were detected in more than one family, representing 35% of total identified mutations. To discriminate between an independent recurrence or a founder effect, an analysis of intra- and -extragenic single nucleotide polymorphisms (SNPs) and short tandem repeats (STRs) flanking the F8 gene was conducted. SNP haplotype and microsatellite analysis revealed strong evidence that p.Ser2030Asn and p.Pro2311His mutations were probably associated with a founder effect. The two other mutations localized in an F8 cytosine-phosphate-guanine (CpG) site likely resulted from recurrent de novo events. This study suggests that missense mutations producing C-to-T or G-to-A substitutions in CpG dinucleotide can occur de novo with more repetition than other causal substitutions that do not affect the CpG site. Analysis of F8 database implied that CpG sites throughout the F8 gene are not all mutated with the same frequency. Causes are still unknown and remain to be identified.


Haemophilia | 2015

Tandem inversion duplication within F8 Intron 1 associated with mild haemophilia A.

N. Lannoy; Claude Bandelier; Bernard Grisart; Michel Reginster; Elisabeth Ronge-Collard; Miikka Vikkula; Cédric Hermans

In approximately 90% of mild haemophilia A (HA) patients, a missense mutation can be identified using complete gene sequencing. In this study, multiplex ligation‐dependent probe amplification analysis was performed as a second step in 10 French‐speaking Belgian with mild HA presenting no detectable causal mutation by complete sequencing of the factor VIII (FVIII) (F8) genes 26 exons and its 1.2 kb of contiguous promoter sequence. This gene dosage technique enabled the detection of exon 1 duplications of F8 in three apparently unrelated subjects. Using array‐comparative genomic hybridization, breakpoint analysis delimited the duplication extent to 210 kb in the F8 intron 1 and VBP1 gene intragenic position. We postulated that the rearrangement responsible for this duplication, never before reported, could be attributed to a symmetrical tandem inversion duplication, resulting in a large 233 kb rearrangement of F8 intron 1. This rearranged intron should lead to the production of a small number of normal mRNA transcripts in relation to the mild HA phenotype. Our analysis of the entire F8 mRNA from index case 1, particularly the segment containing exons 1–9, revealed normal amplification and sequencing. Reduced plasma FVIII antigen levels caused by cross‐reacting material is associated with a quantitative deficiency of plasma FVIII. Male patients were unresponsive to desmopressin (1‐deamino‐8‐D‐arginine vasopressin). All patients displayed identical F8 haplotypes, despite not being related, which suggests a possible founder effect caused by a 210 kb duplication involving F8 exon 1.


Journal of The American Society of Nephrology | 2000

CF Gene and Cystic Fibrosis Transmembrane Conductance Regulator Expression in Autosomal Dominant Polycystic Kidney Disease

Alexandre Persu; Olivier Devuyst; N. Lannoy; Roland Materne; Godela Brosnahan; Patricia A. Gabow; Yves Pirson; Christine Verellen-Dumoulin

Collaboration


Dive into the N. Lannoy's collaboration.

Top Co-Authors

Avatar

Cédric Hermans

Catholic University of Leuven

View shared research outputs
Top Co-Authors

Avatar

Catherine Lambert

Cliniques Universitaires Saint-Luc

View shared research outputs
Top Co-Authors

Avatar

Miikka Vikkula

Université catholique de Louvain

View shared research outputs
Top Co-Authors

Avatar

Christine Verellen-Dumoulin

Université catholique de Louvain

View shared research outputs
Top Co-Authors

Avatar

Alex Kartheuser

Cliniques Universitaires Saint-Luc

View shared research outputs
Top Co-Authors

Avatar

Alexandre Persu

Université catholique de Louvain

View shared research outputs
Top Co-Authors

Avatar

Christine Dumoulin

Cliniques Universitaires Saint-Luc

View shared research outputs
Top Co-Authors

Avatar

Corinne Walon

Université catholique de Louvain

View shared research outputs
Top Co-Authors

Avatar

I. Abinet

Cliniques Universitaires Saint-Luc

View shared research outputs
Top Co-Authors

Avatar

M Smaers

Université catholique de Louvain

View shared research outputs
Researchain Logo
Decentralizing Knowledge