Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 9 de 9
1.
Hum Mutat ; 43(3): 316-327, 2022 03.
Article En | MEDLINE | ID: mdl-34882875

Hereditary papillary renal cell carcinoma (HPRC) is a rare inherited renal cancer syndrome characterized by bilateral and multifocal papillary type 1 renal tumors (PRCC1). Activating germline pathogenic variants of the MET gene were identified in HPRC families. We reviewed the medical and molecular records of a large French series of 158 patients screened for MET oncogenic variants. MET pathogenic and likely pathogenic variants rate was 12.4% with 40.6% among patients with familial PRCC1 and 5% among patients with sporadic PRCC1. The phenotype in cases with MET pathogenic and likely pathogenic variants was characteristic: PRCC1 tumors were mainly bilateral (84.3%) and multifocal (87.5%). Histologically, six out of seven patients with MET pathogenic variant harbored biphasic squamoid alveolar PRCC. Genetic screening identified one novel pathogenic variant MET c.3389T>C, p.(Leu1130Ser) and three novel likely pathogenic variants: MET c.3257A>T, p.(His1086Leu); MET c.3305T>C, p.(Ile1102Thr) and MET c.3373T>G, p.(Cys1125Gly). Functional assay confirmed their oncogenic effect as they induced an abnormal focus formation. The genotype-phenotype correlation between MET pathogenic variants and PRCC1 presentation should encourage to widen the screening, especially toward nonfamilial PRCC1. This precise phenotype also constitutes a strong argument for the classification of novel missense variants within the tyrosine kinase domain when functional assays are not accessible.


Carcinoma, Renal Cell , Kidney Neoplasms , Neoplastic Syndromes, Hereditary , Proto-Oncogene Proteins c-met , Carcinoma, Renal Cell/genetics , Female , Germ Cells/metabolism , Humans , Kidney Neoplasms/genetics , Male , Phenotype , Proto-Oncogene Proteins c-met/genetics
2.
Blood ; 132(5): 469-483, 2018 08 02.
Article En | MEDLINE | ID: mdl-29891534

Chuvash polycythemia is an autosomal recessive form of erythrocytosis associated with a homozygous p.Arg200Trp mutation in the von Hippel-Lindau (VHL) gene. Since this discovery, additional VHL mutations have been identified in patients with congenital erythrocytosis, in a homozygous or compound-heterozygous state. VHL is a major tumor suppressor gene, mutations in which were first described in patients presenting with VHL disease, which is characterized by the development of highly vascularized tumors. Here, we identify a new VHL cryptic exon (termed E1') deep in intron 1 that is naturally expressed in many tissues. More importantly, we identify mutations in E1' in 7 families with erythrocytosis (1 homozygous case and 6 compound-heterozygous cases with a mutation in E1' in addition to a mutation in VHL coding sequences) and in 1 large family with typical VHL disease but without any alteration in the other VHL exons. In this study, we show that the mutations induced a dysregulation of VHL splicing with excessive retention of E1' and were associated with a downregulation of VHL protein expression. In addition, we demonstrate a pathogenic role for synonymous mutations in VHL exon 2 that altered splicing through E2-skipping in 5 families with erythrocytosis or VHL disease. In all the studied cases, the mutations differentially affected splicing, correlating with phenotype severity. This study demonstrates that cryptic exon retention and exon skipping are new VHL alterations and reveals a novel complex splicing regulation of the VHL gene. These findings open new avenues for diagnosis and research regarding the VHL-related hypoxia-signaling pathway.


Exons , Genetic Predisposition to Disease , Mutation , Polycythemia/genetics , RNA Splicing , Von Hippel-Lindau Tumor Suppressor Protein/genetics , von Hippel-Lindau Disease/genetics , Adolescent , Adult , Child , Female , Heterozygote , Humans , Male , Middle Aged , Pedigree , Polycythemia/classification , Polycythemia/pathology , Young Adult , von Hippel-Lindau Disease/pathology
3.
Mod Pathol ; 31(6): 974-983, 2018 06.
Article En | MEDLINE | ID: mdl-29410489

Hereditary leiomyomatosis and renal cell carcinoma syndrome is characterized by an increased risk of agressive renal cell carcinoma, often of type 2 papillary histology, and is caused by FH germline mutations. A prominent eosinophilic macronucleolus with a perinucleolar clear halo is distinctive of hereditary leiomyomatosis and renal cell carcinoma syndrome-associated renal cell carcinoma according to the 2012 ISUP and 2016 WHO kidney tumor classification. From an immunohistochemistry perspective, tumors are often FH-negative and S-(2-succino)-cysteine (2SC) positive. We performed a pathology review of 24 renal tumors in 23 FH mutation carriers, and compared them to 12 type 2 papillary renal cell carcinomas from FH wild-type patients. Prominent eosinophilic nucleoli with perinucleolar halos were present in almost all FH-deficient renal cell carcinomas (23/24). Unexpectedly, they were also present in 58% of type 2 papillary renal cell carcinomas from wild-type patients. Renal cell carcinoma in mutation carriers displayed a complex architecture with multiple patterns, typically papillary, tubulopapillary, and tubulocystic, but also sarcomatoid and rhabdoid. Such pattern diversity was not seen in non-carriers. FH/2SC immunohistochemistry was informative as all hereditary leiomyomatosis and renal cell carcinoma-associated renal cell carcinomas were either FH- or 2SC+. For FH and 2SC immunohistochemistries taken separately, sensitivity of negative anti-FH immunohistochemistry was 87.5% and specificity was 100%. For positive anti-2SC immunohistochemistry, sensitivity, and specificity were 91.7% and 91.7%, respectively. All FH wild-type renal cell carcinoma were FH-positive, and all but one were 2SC-negative. In conclusion, multiplicity of architectural patterns, rhabdoid/sarcomatoid components and combined FH/2SC staining, but not prominent eosinophilic nucleoli with perinucleolar halos, differentiate hereditary leiomyomatosis and renal cell carcinoma-associated renal cell carcinoma from type 2 papillary renal cell carcinoma with efficient FH gene. Our findings are crucial in identifying who should be referred to Cancer Genetics clinics for genetic counseling and testing.


Carcinoma, Renal Cell/diagnosis , Fumarate Hydratase/metabolism , Kidney Neoplasms/diagnosis , Leiomyomatosis/diagnosis , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Diagnosis, Differential , Fumarate Hydratase/genetics , Genetic Predisposition to Disease , Germ-Line Mutation , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Leiomyomatosis/genetics , Leiomyomatosis/metabolism , Leiomyomatosis/pathology
4.
Oncotarget ; 7(51): 85306-85317, 2016 Dec 20.
Article En | MEDLINE | ID: mdl-27852035

Von Hippel-Lindau (VHL) disease is an autosomal dominant hereditary cancer syndrome that predisposes affected individuals to the development of multiple benign and malignant tumors. One of the main manifestations of VHL is renal cell carcinoma (RCC). RCC is increasingly being treated with targeted therapies, which offer an alternative treatment option for patients with VHL disease. This study investigated the effectiveness of sunitinib in VHL patients with advanced tumors or tumors unsuitable for surgery.This multicenter, phase II, open-label study from the PREDIR VHL network, treated patients with genetically-confirmed advanced VHL disease with oral sunitinib (50 mg/day for 28 days then a 2-week rest period) until progression. Lesions were performed using magnetic resonance imaging (MRI) and computed tomographic (CT) scan. The primary endpoint was objective response rate; secondary endpoints included tolerability and overall survival.All five patients showed stable disease as best response at 6 months. Two patients showed impressive transitory clinical improvement during early cycles. No patient died during sunitinib treatment. Reasons for discontinuing sunitinib therapy were disease progression (n=1), unacceptable toxicity (n=3) and lack of clinical improvement after 7 cycles (10.5 months) with unacceptable toxicity (n=1).In conclusion, sunitinib was of limited benefit in patients with advanced VHL disease, but had better efficacy against metastatic RCC than other VHL-related lesions. Treatment-related toxicity is an important limiting factor in this frail patient population. New agents with different mechanisms of action are required to treat this disease.


Antineoplastic Agents/therapeutic use , Carcinoma, Renal Cell/drug therapy , Indoles/therapeutic use , Kidney Neoplasms/drug therapy , Neoplasms/drug therapy , Pyrroles/therapeutic use , von Hippel-Lindau Disease/drug therapy , Aged , Aged, 80 and over , Carcinoma, Renal Cell/etiology , Carcinoma, Renal Cell/mortality , Female , France , Humans , Kidney Neoplasms/etiology , Kidney Neoplasms/mortality , Male , Middle Aged , Neoplasm Metastasis , Neoplasm Staging , Neoplasms/etiology , Neoplasms/mortality , Prospective Studies , Sunitinib , Survival Analysis , Treatment Outcome , von Hippel-Lindau Disease/complications , von Hippel-Lindau Disease/mortality
5.
J Med Genet ; 52(6): 426-30, 2015 Jun.
Article En | MEDLINE | ID: mdl-25911086

BACKGROUND: Many cases of familial renal cell carcinoma (RCC) remain unexplained by mutations in the known predisposing genes or shared environmental factors. There are therefore additional, still unidentified genes involved in familial RCC. PBRM1 is a tumour suppressor gene and somatic mutations are found in 30-45% of sporadic clear cell (cc) RCC. METHODS: We selected 35 unrelated patients with unexplained personal history of ccRCC and at least one affected first-degree relative, and sequenced the PBRM1 gene. RESULTS: A germline frameshift mutation (c.3998_4005del [p.Asp1333Glyfs]) was found in one patient. The patient's mother, his sister and one niece also had ccRCC. The mutation co-segregated with the disease as the three affected relatives were carriers, while an unaffected sister was not, according with autosomal-dominant transmission. Somatic studies supported these findings, as we observed both loss of heterozygosity for the mutation and loss of protein expression in renal tumours. CONCLUSIONS: We show for the first time that an inherited mutation in PBRM1 predisposes to RCC. International studies are necessary to estimate the contribution of PBRM1 to RCC susceptibility, estimate penetrance and then integrate the gene into routine clinical practice.


Carcinoma, Renal Cell/genetics , Genetic Predisposition to Disease , Germ-Line Mutation , Kidney Neoplasms/genetics , Nuclear Proteins/genetics , Transcription Factors/genetics , Carcinoma, Renal Cell/diagnosis , DNA Mutational Analysis , DNA-Binding Proteins , Exons , Female , Heterozygote , Humans , Immunohistochemistry , Kidney Neoplasms/diagnosis , Male , Nuclear Proteins/metabolism , Pedigree , Transcription Factors/metabolism
6.
Orphanet J Rare Dis ; 9: 163, 2014 Oct 29.
Article En | MEDLINE | ID: mdl-25519458

BACKGROUND: The Birt-Hogg-Dubé syndrome is a rare cancer susceptibility syndrome characterised by renal tumours, lung cysts and pneumothoraces, and fibrofolliculomas. It is caused by dominantly inherited mutations in FLCN. Our objective was to report renal tumour characteristics in a large series of patients with the Birt-Hogg-Dubé syndrome. METHODS: We studied French Birt-Hogg-Dubé patients with a history of renal tumour. RESULTS: We included 33 patients with 21 distinct germline FLCN mutations. Median age at diagnosis of first renal tumour was 46, and age varied from 20 to 83. Twenty cases had one renal tumour, the remainder had two or more tumours. Most cases (23/33, 70%) had oncocytoma or renal cell carcinoma of the chromophobe or hybrid chromophobe-oncocytoma type, three had clear cell carcinoma (9%), and the other seven had carcinoma of papillary, undifferentiated or undetermined histology. Four cases had metastatic disease, although none died of it. CONCLUSIONS: Age at renal tumour diagnosis was highly variable, highlighting the need for regular surveillance from young adulthood to old age. Most cases had tumour types classically associated with Birt-Hogg-Dubé, i.e. oncocytoma or renal cell carcinoma of the chromophobe or hybrid type. Nevertheless, 9% had clear cell renal cell carcinoma. Geneticists, urologists and oncologists should therefore be alert to the possibility of Birt-Hogg-Dubé in patients with renal cell carcinoma of clear cell histology, especially if there are associated manifestations. Finally, the behaviour of metastatic carcinoma seemed more indolent than in sporadic renal cancers.


Adenoma, Oxyphilic/diagnosis , Birt-Hogg-Dube Syndrome/diagnosis , Carcinoma, Renal Cell/diagnosis , Kidney Neoplasms/diagnosis , Adenoma, Oxyphilic/epidemiology , Adenoma, Oxyphilic/genetics , Adult , Aged , Aged, 80 and over , Birt-Hogg-Dube Syndrome/epidemiology , Birt-Hogg-Dube Syndrome/genetics , Carcinoma, Renal Cell/epidemiology , Carcinoma, Renal Cell/genetics , Female , Humans , Kidney Neoplasms/epidemiology , Kidney Neoplasms/genetics , Male , Middle Aged , Retrospective Studies , Young Adult
7.
J Urol ; 185(6): 2056-60, 2011 Jun.
Article En | MEDLINE | ID: mdl-21496837

PURPOSE: Patients with von Hippel-Lindau disease frequently have early, multiple and recurrent renal cell carcinoma. Renal cell carcinoma treatment, which must prevent metastatic disease and spare nephrons, has changed in the last 2 decades. We evaluated renal cell carcinoma treatments in the long term in a large series of patients with von Hippel-Lindau disease. MATERIALS AND METHODS: We retrospectively evaluated the use and results of surgery and radio frequency ablation in patients with von Hippel-Lindau followed at our institution between 1988 and 2009. Renal anatomical survival was analyzed according to 3 periods, including 1) 1988 to 1994--the learning phase of nephron sparing surgery, 2) 1995 to 2003--routine nephron sparing surgery and 3) 2004 to 2009--the emergence of radio frequency ablation. RESULTS: A first renal cell carcinoma was treated at a mean age of 38 years (range 15 to 67) in 113 patients with von Hippel-Lindau disease. During a median followup of 7.2 years 251 therapeutic procedures were performed in a total of 176 kidneys. We observed a shift of first line renal cell carcinoma treatment with time, that is nephrectomy in 52% of cases in period 1, tumorectomy in 75% in period 2 and radio frequency ablation in 43% in period 3. The shift paralleled improved renal survival. While nephron sparing surgery was primarily done for lesions greater than 30 mm, radio frequency ablation was used to treat less numerous and smaller ipsilateral lesions but they required more frequent intervention. Radio frequency ablation became the most widely used second or third line procedure and allowed renal salvage in 8 patients. CONCLUSIONS: Nephron sparing surgery and more recently radio frequency ablation enable earlier treatment of smaller tumors and are associated with a significant improved renal prognosis in patients with von Hippel-Lindau disease.


Carcinoma, Renal Cell/surgery , Catheter Ablation , Kidney Neoplasms/surgery , Nephrectomy/methods , von Hippel-Lindau Disease/surgery , Adolescent , Adult , Aged , Carcinoma, Renal Cell/etiology , Female , Humans , Kidney Neoplasms/etiology , Male , Middle Aged , Nephrons , Retrospective Studies , Young Adult , von Hippel-Lindau Disease/complications
8.
J Med Genet ; 48(4): 226-34, 2011 Apr.
Article En | MEDLINE | ID: mdl-21398687

BACKGROUND: Hereditary leiomyomatosis and renal cell cancer (HLRCC) is an autosomal dominant disorder predisposing humans to cutaneous and uterine leiomyomas; in 20% of affected families, type 2 papillary renal cell cancers (PRCCII) also occur with aggressive course and poor prognosis. HLRCC results from heterozygous germline mutations in the tumour suppressor fumarate hydratase (FH) gene. METHODS: As part of the French National Cancer Institute (INCa) 'Inherited predispositions to kidney cancer' network, sequence analysis and a functional study of FH were preformed in 56 families with clinically proven or suspected HLRCC and in 23 patients with isolated PRCCII (5 familial and 18 sporadic). RESULTS: The study identified 32 different germline FH mutations (15 missense, 6 frameshifts, 4 nonsense, 1 deletion/insertion, 5 splice site, and 1 complete deletion) in 40/56 (71.4%) families with proven or suspected HLRCC and in 4/23 (17.4%) probands with PRCCII alone, including 2 sporadic cases. 21 of these were novel and all were demonstrated as deleterious by significant reduction of FH enzymatic activity. In addition, 5 asymptomatic parents in 3 families were confirmed as carrying disease-causing mutations. CONCLUSIONS: This study identified and characterised 21 novel FH mutations and demonstrated that PRCCII can be the only one manifestation of HLRCC. Due to the incomplete penetrance of HLRCC, the authors propose to extend the FH mutation analysis to every patient with PRCCII occurring before 40 years of age or when renal tumour harbours characteristic histologic features, in order to discover previously ignored HLRCC affected families.


Carcinoma, Renal Cell/genetics , Fumarate Hydratase/genetics , Kidney Neoplasms/genetics , Mutation , Adult , Aged , Cell Line, Tumor , Codon, Nonsense , Female , Frameshift Mutation , Gene Deletion , Gene Rearrangement , Genotype , Germ-Line Mutation , Humans , INDEL Mutation , Leiomyomatosis/congenital , Leiomyomatosis/genetics , Male , Middle Aged , Mutation, Missense , Neoplastic Syndromes, Hereditary , Pedigree , Skin Neoplasms , Uterine Neoplasms
9.
Mol Cell Biol ; 24(5): 1870-83, 2004 Mar.
Article En | MEDLINE | ID: mdl-14966269

Analysis of cis-regulatory elements is central to understanding the genomic program for development. The scl/tal-1 transcription factor is essential for lineage commitment to blood cell formation and previous studies identified an scl enhancer (the +18/19 element) which was sufficient to target the vast majority of hematopoietic stem cells, together with hematopoietic progenitors and endothelium. Moreover, expression of scl under control of the +18/19 enhancer rescued blood progenitor formation in scl(-/-) embryos. However, here we demonstrate by using a knockout approach that, within the endogenous scl locus, the +18/19 enhancer is not necessary for the initiation of scl transcription or for the formation of hematopoietic cells. These results led to the identification of a bifunctional 5' enhancer (-3.8 element), which targets expression to hematopoietic progenitors and endothelium, contains conserved critical Ets sites, and is bound by Ets family transcription factors, including Fli-1 and Elf-1. These data demonstrate that two geographically distinct but functionally related enhancers regulate scl transcription in hematopoietic progenitors and endothelial cells and suggest that enhancers with dual hematopoietic-endothelial activity may represent a general strategy for regulating blood and endothelial development.


DNA-Binding Proteins/metabolism , Enhancer Elements, Genetic , Hematopoiesis/physiology , Hematopoietic Stem Cells/physiology , Proto-Oncogene Proteins/metabolism , Transcription Factors/metabolism , Animals , Base Sequence , Basic Helix-Loop-Helix Transcription Factors , Cell Lineage , DNA-Binding Proteins/genetics , Embryo, Mammalian/anatomy & histology , Embryo, Mammalian/physiology , Gene Expression Regulation, Developmental , Genes, Reporter , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Nuclear Proteins , Proto-Oncogene Protein c-fli-1 , Proto-Oncogene Proteins/genetics , Sequence Alignment , T-Cell Acute Lymphocytic Leukemia Protein 1 , Trans-Activators/metabolism , Transcription Factors/genetics , Transcription, Genetic
...