Network


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

Hotspot


Dive into the research topics where Fabien Nevo is active.

Publication


Featured researches published by Fabien Nevo.


The New England Journal of Medicine | 2011

INF2 Mutations in Charcot–Marie–Tooth Disease with Glomerulopathy

Olivia Boyer; Fabien Nevo; Emmanuelle Plaisier; Benoît Funalot; Olivier Gribouval; Geneviève Benoit; Evelyne Huynh Cong; Christelle Arrondel; Marie-Josèphe Tête; Rodrick Montjean; Laurence Richard; Alexandre Karras; Claire Pouteil-Noble; Leila Balafrej; Alain Bonnardeaux; Guillaume Canaud; Christophe Charasse; Jacques Dantal; Georges Deschênes; Patrice Deteix; Odile Dubourg; Philippe Petiot; Dominique Pouthier; Eric LeGuern; Anne Guiochon-Mantel; Isabelle Broutin; Marie-Claire Gubler; Sophie Saunier; Pierre Ronco; Jean-Michel Vallat

BACKGROUNDnCharcot-Marie-Tooth neuropathy has been reported to be associated with renal diseases, mostly focal segmental glomerulosclerosis (FSGS). However, the common mechanisms underlying the neuropathy and FSGS remain unknown. Mutations in INF2 were recently identified in patients with autosomal dominant FSGS. INF2 encodes a formin protein that interacts with the Rho-GTPase CDC42 and myelin and lymphocyte protein (MAL) that are implicated in essential steps of myelination and myelin maintenance. We therefore hypothesized that INF2 may be responsible for cases of Charcot-Marie-Tooth neuropathy associated with FSGS.nnnMETHODSnWe performed direct genotyping of INF2 in 16 index patients with Charcot-Marie-Tooth neuropathy and FSGS who did not have a mutation in PMP22 or MPZ, encoding peripheral myelin protein 22 and myelin protein zero, respectively. Histologic and functional studies were also conducted.nnnRESULTSnWe identified nine new heterozygous mutations in 12 of the 16 index patients (75%), all located in exons 2 and 3, encoding the diaphanous-inhibitory domain of INF2. Patients presented with an intermediate form of Charcot-Marie-Tooth neuropathy as well as a glomerulopathy with FSGS on kidney biopsy. Immunohistochemical analysis revealed strong INF2 expression in Schwann-cell cytoplasm and podocytes. Moreover, we demonstrated that INF2 colocalizes and interacts with MAL in Schwann cells. The INF2 mutants perturbed the INF2-MAL-CDC42 pathway, resulting in cytoskeleton disorganization, enhanced INF2 binding to CDC42 and mislocalization of INF2, MAL, and CDC42.nnnCONCLUSIONSnINF2 mutations appear to cause many cases of FSGS-associated Charcot-Marie-Tooth neuropathy, showing that INF2 is involved in a disease affecting both the kidney glomerulus and the peripheral nervous system. These findings provide new insights into the pathophysiological mechanisms linking formin proteins to podocyte and Schwann-cell function. (Funded by the Agence Nationale de la Recherche and others.).


Journal of The American Society of Nephrology | 2011

Mutations in INF2 Are a Major Cause of Autosomal Dominant Focal Segmental Glomerulosclerosis

Olivia Boyer; Geneviève Benoit; Olivier Gribouval; Fabien Nevo; Marie-Josèphe Tête; Jacques Dantal; Brigitte Gilbert-Dussardier; Guy Touchard; Alexandre Karras; Claire Presne; Jean-Pierre Grünfeld; Christophe Legendre; Dominique Joly; Philippe Rieu; Nabil Mohsin; Thierry Hannedouche; Valérie Moal; Marie-Claire Gubler; Isabelle Broutin; Géraldine Mollet; Corinne Antignac

The recent identification of mutations in the INF2 gene, which encodes a member of the formin family of actin-regulating proteins, in cases of familial FSGS supports the importance of an intact actin cytoskeleton in podocyte function. To determine better the prevalence of INF2 mutations in autosomal dominant FSGS, we screened 54 families (78 patients) and detected mutations in 17% of them. All mutations were missense variants localized to the N-terminal diaphanous inhibitory domain of the protein, a region that interacts with the C-terminal diaphanous autoregulatory domain, thereby competing for actin monomer binding and inhibiting depolymerization. Six of the seven distinct altered residues localized to an INF2 region that corresponded to a subdomain of the mDia1 diaphanous inhibitory domain reported to co-immunoprecipitate with IQ motif-containing GTPase-activating protein 1 (IQGAP1). In addition, we evaluated 84 sporadic cases but detected a mutation in only one patient. In conclusion, mutations in INF2 are a major cause of autosomal dominant FSGS. Because IQGAP1 interacts with crucial podocyte proteins such as nephrin and PLCε1, the identification of mutations that may alter the putative INF2-IQGAP1 interaction provides additional insight into the pathophysiologic mechanisms linking formin proteins to podocyte dysfunction and FSGS.


Kidney International | 2009

Clinical and epidemiological assessment of steroid-resistant nephrotic syndrome associated with the NPHS2 R229Q variant

Eduardo Machuca; Aurélie Hummel; Fabien Nevo; Jacques Dantal; Frank Martinez; Essam Al-Sabban; Véronique Baudouin; Laurent Abel; Jean-Pierre Grünfeld; Corinne Antignac

Mutations of NPHS2, encoding podocin, are the main cause of autosomal recessive steroid-resistant nephrotic syndrome (NS) presenting in childhood. Adult-onset steroid-resistant NS has been described in patients heterozygous for a pathogenic NPHS2 mutation together with the p.R229Q variant. To determine the frequency and the phenotype of patients carrying the p.R229Q variant, we sequenced the complete coding region of NPHS2 in 455 families (546 patients) non-responsive to immunosuppressive therapy or without relapse after transplantation. Among affected Europeans, the p.R229Q allele was significantly more frequent compared to control individuals. Thirty-six patients from 27 families (11 families from Europe and 14 from South America) were compound heterozygotes for the p.R229Q variant and one pathogenic mutation. These patients had significantly later onset of NS and end stage renal disease than patients with two pathogenic mutations. Among 119 patients diagnosed with NS presenting after 18 years of age, 18 patients were found to have one pathogenic mutation and p.R229Q, but none had two pathogenic mutations. Our study shows that compound heterozygosity for p.R229Q is associated with adult-onset steroid-resistant NS, mostly among patients of European and South American origin. Screening for the p.R229Q variant is recommended in these patients along with further NPHS2 mutation analysis in those carrying the variant.


Journal of The American Society of Nephrology | 2008

Nephrin Mutations Can Cause Childhood-Onset Steroid-Resistant Nephrotic Syndrome

Aurélie Philippe; Fabien Nevo; Ernie L. Esquivel; Dalia Reklaityte; Olivier Gribouval; Marie-Josèphe Tête; Chantal Loirat; Jacques Dantal; Michel Fischbach; Claire Pouteil-Noble; Stéphane Decramer; Martin Hoehne; Thomas Benzing; Marina Charbit; Patrick Niaudet; Corinne Antignac

Classically, infants with mutations in NPHS1, which encodes nephrin, present with nephrotic syndrome within the first 3 mo of life (congenital nephrotic syndrome of the Finnish-type), and children with mutations in NPHS2, which encodes podocin, present later with steroid-resistant nephrotic syndrome. Recently, however, NPHS2 mutations have been identified in children with congenital nephrotic syndrome. Whether NPHS1 mutations similarly account for some cases of childhood steroid-resistant nephrotic syndrome is unknown. In this study, 160 patients who belonged to 142 unrelated families and presented with nephrotic syndrome at least 3 mo after birth were screened for NPHS1 variants once mutations in NPHS2 had been excluded. Compound heterozygous NPHS1 mutations were identified in one familial case and nine sporadic cases. Mutations included protein-truncating nonsense and frameshift mutations, as well as splice-site and missense variants. Mutations were classified as severe or mild using prediction algorithms and functional assays. Most missense variants trafficked normally to the plasma membrane and maintained the ability to form nephrin homodimers and to heterodimerize with NEPH1, suggesting retained function. The presence of at least one mild mutation in these patients likely explains the later onset and milder course of disease. These results broaden the spectrum of renal disease related to nephrin mutations.


Journal of The American Society of Nephrology | 2010

Genotype-phenotype correlations in non-Finnish congenital nephrotic syndrome.

Eduardo Machuca; Geneviève Benoit; Fabien Nevo; Marie-Josèphe Tête; Olivier Gribouval; Audrey Pawtowski; Per Brandström; Chantal Loirat; Patrick Niaudet; Marie-Claire Gubler; Corinne Antignac

Mutations in NPHS1, which encodes nephrin, are the main causes of congenital nephrotic syndrome (CNS) in Finnish patients, whereas mutations in NPHS2, which encodes podocin, are typically responsible for childhood-onset steroid-resistant nephrotic syndrome in European populations. Genotype-phenotype correlations are not well understood in non-Finnish patients. We evaluated the clinical presentation, kidney histology, and disease progression in non-Finnish CNS cases by mutational screening in 107 families (117 cases) by sequencing the entire coding regions of NPHS1, NPHS2, PLCE1, WT1, LAMB2, PDSS2, COQ2, and NEPH1. We found that CNS describes a heterogeneous group of disorders in non-Finnish populations. We identified nephrin and podocin mutations in most families and only rarely found mutations in genes implicated in other hereditary forms of NS. In approximately 20% of cases, we could not identify the underlying genetic cause. Consistent with the major role of nephrin at the slit diaphragm, NPHS1 mutations associated with an earlier onset of disease and worse renal outcomes than NPHS2 mutations. Milder cases resulting from mutant NPHS1 had either two mutations in the cytoplasmic tail or two missense mutations in the extracellular domain, including at least one that preserved structure and function. In addition, we extend the spectrum of known NPHS1 mutations by describing long NPHS1 deletions. In summary, these data demonstrate that CNS is not a distinct clinical entity in non-Finnish populations but rather a clinically and genetically heterogeneous group of disorders.


Nature Genetics | 2014

Mutation-dependent recessive inheritance of NPHS2-associated steroid-resistant nephrotic syndrome

Kalman Tory; Dóra K. Menyhárd; Stéphanie Woerner; Fabien Nevo; Olivier Gribouval; Andrea Kerti; Pál Stráner; Christelle Arrondel; Evelyne Huynh Cong; Tivadar Tulassay; Géraldine Mollet; András Perczel; Corinne Antignac

Monogenic disorders result from defects in a single gene. According to Mendels laws, these disorders are inherited in either a recessive or dominant fashion. Autosomal-recessive disorders require a disease-causing variant on both alleles, and according to our current understanding, their pathogenicities are not influenced by each other. Here we present an autosomal-recessive disorder, nephrotic syndrome type 2 (MIM 600995), in which the pathogenicity of an NPHS2 allele encoding p.Arg229Gln depends on the trans-associated mutation. We show that, contrary to expectations, this allele leads to a disease phenotype only when it is associated specifically with certain 3′ NPHS2 mutations because of an altered heterodimerization and mislocalization of the encoded p.Arg229Gln podocin. The disease-associated 3′ mutations exert a dominant-negative effect on p.Arg229Gln podocin but behave as recessive alleles when associated with wild-type podocin. Therefore, the transmission rates for couples carrying the disease-associated mutations and p.Arg229Gln may be substantially different from those expected in autosomal-recessive disorders.


Journal of Medical Genetics | 2010

Mutational analysis of the PLCE1 gene in steroid resistant nephrotic syndrome

Olivia Boyer; Geneviève Benoit; Olivier Gribouval; Fabien Nevo; Audrey Pawtowski; Ilmay Bilge; Zelal Bircan; Georges Deschênes; Lisa M. Guay-Woodford; Michelle Hall; Marie-Alice Macher; Kenza Soulami; Constantinos J. Stefanidis; Robert Weiss; Chantal Loirat; Marie-Claire Gubler; Corinne Antignac

Background Mutations in the PLCE1 gene encoding phospholipase C epsilon 1 (PLCɛ1) have been recently described in patients with early onset nephrotic syndrome (NS) and diffuse mesangial sclerosis (DMS). In addition, two cases of PLCE1 mutations associated with focal segmental glomerulosclerosis (FSGS) and later NS onset have been reported. Method In order to better assess the spectrum of phenotypes associated with PLCE1 mutations, mutational analysis was performed in a worldwide cohort of 139 patients (95 familial cases belonging to 68 families and 44 sporadic cases) with steroid resistant NS presenting at a median age of 23.0u2005months (range 0–373). Results Homozygous or compound heterozygous mutations were identified in 33% (8/24) of DMS cases. PLCE1 mutations were found in 8% (6/78) of FSGS cases without NPHS2 mutations. Nine were novel mutations. No clear genotype–phenotype correlation was observed, with either truncating or missense mutations detected in both DMS and FSGS, and leading to a similar renal evolution. Surprisingly, three unaffected and unrelated individuals were also found to carry the homozygous mutations identified in their respective families. Conclusion PLCE1 is a major gene of DMS and is mutated in a non-negligible proportion of FSGS cases without NPHS2 mutations. Although additional variants in 19 candidate genes (16 other PLC genes, BRAF,IQGAP1 and NPHS1) were not identified, it is speculated that other modifier genes or environmental factors may play a role in the renal phenotype variability observed in individuals bearing PLCE1 mutations. This observation needs to be considered in the genetic counselling offered to patients.


Human Mutation | 2014

NPHS2 mutations in steroid-resistant nephrotic syndrome: A mutation update and the associated phenotypic spectrum

Karim Bouchireb; Olivia Boyer; Olivier Gribouval; Fabien Nevo; Evelyne Huynh-Cong; Vincent Morinière; Raphaëlle Campait; Elisabet Ars; Damien Brackman; Jacques Dantal; Philippe Eckart; Maddalena Gigante; Beata S. Lipska; Aurélia Liutkus; André Mégarbané; Nabil Mohsin; Fatih Ozaltin; Moin A. Saleem; Franz Schaefer; Kenza Soulami; Roser Torra; Nicolas Garcelon; Géraldine Mollet; Karin Dahan; Corinne Antignac

Mutations in the NPHS2 gene encoding podocin are implicated in an autosomal‐recessive form of nonsyndromic steroid‐resistant nephrotic syndrome in both pediatric and adult patients. Patients with homozygous or compound heterozygous mutations commonly present with steroid‐resistant nephrotic syndrome before the age of 6 years and rapidly progress to end‐stage kidney disease with a very low prevalence of recurrence after renal transplantation. Here, we reviewed all the NPHS2 mutations published between October 1999 and September 2013, and also all novel mutations identified in our personal cohort and in international genetic laboratories. We identified 25 novel pathogenic mutations in addition to the 101 already described. The mutations are distributed along the entire coding region and lead to all kinds of alterations including 53 missense, 17 nonsense, 11 small insertions, 26 small deletions, 16 splicing, two indel mutations, and one mutation in the stop codon. In addition, 43 variants were classified as variants of unknown significance, as these missense changes were exclusively described in the heterozygous state and/or considered benign by prediction software. Genotype–phenotype analyses established correlations between specific variants and age at onset, ethnicity, or clinical evolution. We created a Web database using the Leiden Open Variation Database (www.lovd.nl/NPHS2) software that will allow the inclusion of future reports.


Pediatric Nephrology | 2010

Analysis of recessive CD2AP and ACTN4 mutations in steroid-resistant nephrotic syndrome

Geneviève Benoit; Eduardo Machuca; Fabien Nevo; Olivier Gribouval; David Lepage; Corinne Antignac

Mutations in podocyte genes have been identified in patients with steroid-resistant nephrotic syndrome (SRNS). Point mutations in the ACTN4 gene cause an autosomal dominant form of human focal segmental glomerular sclerosis (FSGS); however, reports of CD2AP mutations remain scarce. Based on the phenotype of Actn4 and Cd2ap null mice, we aimed to define the role of recessive CD2AP and ACTN4 mutations in a cohort of children with SRNS for which NPHS1, NPHS2, and PLCE1 mutations had been previously excluded. CD2AP and ACTN4 mutational analysis was performed in 42 children from 35 unrelated families. The median age of disease onset was 20xa0(range 0–102) months. Sixteen patients reached end-stage kidney disease at a median age of 84xa0(range 4–161) months. Renal histology showed FSGS lesions and minimal glomerular changes in 49% and 20% of patients, respectively. Microsatellite marker analysis excluded linkage to the CD2AP locus in 26 families and to the ACTN4 locus in 31 families. No disease-causing mutations were identified in the remaining families. Recessive CD2AP and ACTN4 mutations are rare in children with SRNS. The absence of mutations in this study suggests that there are other genetic causes of SRNS that still need to be identified.


Pediatric Nephrology | 2008

A novel Wilms’ tumor 1 gene mutation in a child with severe renal dysfunction and persistent renal blastema

Nicole Wagner; Kay-Dietrich Wagner; Mickael Afanetti; Fabien Nevo; Corinne Antignac; Jean-François Michiels; Andreas Schedl; Etienne Bérard

The Wilms’ tumor suppressor gene WT1 is an important regulator of development. Mutations in this gene have been associated with Wilms’ tumor, Frasier syndrome, and Denys–Drash syndrome, as well as isolated glomerular disease. Here we report the case of a 4-month-old girl, who presented with end-stage renal disease, thrombopenia, anemia, and cardiac hypertrophy accompanied by severe hypertension. Histological analysis of kidney biopsies revealed a massive and diffuse nephroblastomatosis with a dramatic reduction in the number of glomeruli. Although no normal cortical nephrons could be detected, medullary organization was nearly normal. Sequence analysis demonstrated a heterozygous nonsense mutation in exonxa09 of WT1, which leads to a truncation of the WT1 protein at the beginning of zinc fingerxa03. Given the requirement of WT1 for normal development of the kidney and heart, these data raise the hypothesis that the mutation identified was responsible for the severe phenotype observed in our patient.

Collaboration


Dive into the Fabien Nevo's collaboration.

Top Co-Authors

Avatar

Olivier Gribouval

Paris Descartes University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Marie-Josèphe Tête

Necker-Enfants Malades Hospital

View shared research outputs
Top Co-Authors

Avatar

Olivia Boyer

Necker-Enfants Malades Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Eduardo Machuca

Pontifical Catholic University of Chile

View shared research outputs
Top Co-Authors

Avatar

Alexandre Karras

Necker-Enfants Malades Hospital

View shared research outputs
Top Co-Authors

Avatar

Audrey Pawtowski

Necker-Enfants Malades Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge