Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 77
Filtrer
1.
Anticancer Res ; 42(3): 1455-1463, 2022 Mar.
Article de Anglais | MEDLINE | ID: mdl-35220239

RÉSUMÉ

BACKGROUND/AIM: Polymorphous adenocarcinoma (PAC) is a low-grade salivary gland malignancy in contrast to variants with papillary (PAP) or cribriform (CASG) architecture and confers the second most common malignancy of minor salivary glands. Our study aimed to identify prognostic factors and to evaluate histomorphological and molecular diagnostic criteria of PACs. PATIENTS AND METHODS: A series of 155 PACs, including 10 PAPs and 12 CASGs from the population-based Cancer Registry of North Rhine-Westphalia (LKR-NRW) and the Hamburg Salivary Gland Reference Centre (HRC) were analyzed. RESULTS: One fifth of the tumors were located in the major salivary glands and PACS/CASGS invariably lacked p40 expression. Fifty-two percent of PACs showed a PRKD1 E710D mutation. Ordinary PACs had a disease-specific 10-year survival probability of 97% compared to 90% when combining PAPs and CASGs. T-stage at diagnosis was a prognostic factor with 98% for stages T1/T2 versus 75% for T3/T4. CONCLUSION: Diagnostic algorithms for the PAC/CASG spectrum of tumors need to be improved and should include molecular markers.


Sujet(s)
Adénocarcinome papillaire , Adénocarcinome , Marqueurs biologiques tumoraux , Tumeurs des glandes salivaires , Adénocarcinome/composition chimique , Adénocarcinome/génétique , Adénocarcinome/mortalité , Adénocarcinome/anatomopathologie , Adénocarcinome papillaire/composition chimique , Adénocarcinome papillaire/génétique , Adénocarcinome papillaire/mortalité , Adénocarcinome papillaire/anatomopathologie , Adolescent , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Marqueurs biologiques tumoraux/analyse , Marqueurs biologiques tumoraux/génétique , Enfant , Femelle , Allemagne , Humains , Mâle , Adulte d'âge moyen , Mutation , Stadification tumorale , Enregistrements , Tumeurs des glandes salivaires/composition chimique , Tumeurs des glandes salivaires/génétique , Tumeurs des glandes salivaires/mortalité , Tumeurs des glandes salivaires/anatomopathologie , Facteurs sexuels , Facteurs temps , Charge tumorale , Jeune adulte
2.
Blood Cancer J ; 11(5): 102, 2021 05 26.
Article de Anglais | MEDLINE | ID: mdl-34039950

RÉSUMÉ

Epstein-Barr virus (EBV)-associated diffuse large B-cell lymphoma not otherwise specified (DLBCL NOS) constitute a distinct clinicopathological entity in the current World Health Organization (WHO) classification. However, its genomic features remain sparsely characterized. Here, we combine whole-genome sequencing (WGS), targeted amplicon sequencing (tNGS), and fluorescence in situ hybridization (FISH) from 47 EBV + DLBCL (NOS) cases to delineate the genomic landscape of this rare disease. Integrated WGS and tNGS analysis clearly distinguished this tumor type from EBV-negative DLBCL due to frequent mutations in ARID1A (45%), KMT2A/KMT2D (32/30%), ANKRD11 (32%), or NOTCH2 (32%). WGS uncovered structural aberrations including 6q deletions (5/8 patients), which were subsequently validated by FISH (14/32 cases). Expanding on previous reports, we identified recurrent alterations in CCR6 (15%), DAPK1 (15%), TNFRSF21 (13%), CCR7 (11%), and YY1 (6%). Lastly, functional annotation of the mutational landscape by sequential gene set enrichment and network propagation predicted an effect on the nuclear factor κB (NFκB) pathway (CSNK2A2, CARD10), IL6/JAK/STAT (SOCS1/3, STAT3), and WNT signaling (FRAT1, SFRP5) alongside aberrations in immunological processes, such as interferon response. This first comprehensive description of EBV + DLBCL (NOS) tumors substantiates the evidence of its pathobiological independence and helps stratify the molecular taxonomy of aggressive lymphomas in the effort for future therapeutic strategies.


Sujet(s)
Infections à virus Epstein-Barr/complications , Lymphome B diffus à grandes cellules/génétique , Lymphome B diffus à grandes cellules/virologie , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Aberrations des chromosomes , Femelle , Réseaux de régulation génique , Herpèsvirus humain de type 4/isolement et purification , Séquençage nucléotidique à haut débit , Humains , Hybridation fluorescente in situ , Mâle , Adulte d'âge moyen , Mutation , Séquençage du génome entier , Jeune adulte
3.
Am J Pathol ; 186(12): 3285-3296, 2016 12.
Article de Anglais | MEDLINE | ID: mdl-27765635

RÉSUMÉ

Neurofibromas and schwannomas are benign Schwann cell-derived peripheral nerve sheath tumors arising sporadically and within neurofibromatoses. Multiple tumors are a hallmark of neurofibromatosis type 1 (NF1) and type 2 (NF2) and schwannomatosis. Neurofibromas in NF1 and schwannomas in NF2 or schwannomatosis are defined by distinctive molecular hits. Among these, multiple hybrid neurofibromas/schwannomas may also appear, not yet being defined by a molecular background. We therefore performed molecular analysis of 22 hybrid neurofibromas/schwannomas using array comparative genomic hybridization, immunohistochemistry, quantitative RT-PCR, and functional analyses of cultured Schwann cells. Furthermore, we analyzed SMARCB1 by fluorescence in situ hybridization and multiplex ligation-dependent probe. Monosomy 22 was identified in 44% of tumors of tested patients with hybrid neurofibromas/schwannomas. In addition, in a single case, we detected focal deletion of the α-T-catenin/CTNNA3 gene (10q21.3). To further characterize this candidate, transient knockdown of α-T-catenin in Schwann cells was performed. CTNNA3 depleted cells showed cytoskeletal abnormalities and reduced E-cadherin expression, indicating epithelial-mesenchymal transition-like abnormalities. To conclude, we uncovered loss of chromosome 22 in almost half of all cases with hybrid neurofibromas/schwannomas of patients with multiple peripheral nerve sheath tumors. We tagged α-T-catenin/CTNNA3 as a novel candidate gene. Our functional investigations might indicate involvement of α-T-catenin/CTNNA3 in the biology of peripheral nerve sheath tumors.


Sujet(s)
Tumeurs des gaines nerveuses/génétique , Neurinome/génétique , Neurofibrome/génétique , Neurofibromatoses/génétique , Neurofibromatose de type 1/génétique , Tumeurs cutanées/génétique , alpha-Caténine/génétique , Adolescent , Adulte , Sujet âgé , Chromosomes humains de la paire 22/génétique , Hybridation génomique comparative , Transition épithélio-mésenchymateuse , Femelle , Humains , Hybridation fluorescente in situ , Mâle , Adulte d'âge moyen , Monosomie , Tumeurs des gaines nerveuses/anatomopathologie , Neurinome/anatomopathologie , Neurofibrome/anatomopathologie , Neurofibromatoses/anatomopathologie , Neurofibromatose de type 1/anatomopathologie , Cellules de Schwann/métabolisme , Cellules de Schwann/anatomopathologie , Tumeurs cutanées/anatomopathologie , Protéines suppresseurs de tumeurs/génétique , Jeune adulte
5.
Br J Haematol ; 157(6): 702-8, 2012 Jun.
Article de Anglais | MEDLINE | ID: mdl-22469134

RÉSUMÉ

Hodgkin and Reed/Sternberg (HRS) cells in classical Hodgkin lymphoma (cHL) show constitutive activation of nuclear factor (NF)-κB. Several genetic lesions contribute to this deregulated NF-κB activity. Here, we analysed two further NF-κB regulators for genetic lesions, the inhibitory factor TRAF3 and the key signalling component of the alternative NF-κB pathway, MAP3K14 (NIK). Single nucleotide polymorphism (SNP) array analysis of cHL cell lines revealed a uniparental disomy of the long arm of chromosome 14 associated with a biallelic deletion of TRAF3 located on this chromosome in cell line U-HO1. Cloning of the deletion breakpoint showed a 123 371 bp deletion. No inactivating mutations of TRAF3 were found in six other cHL cell lines or in microdissected HRS cells from seven cHL. However, in primary cHL samples interphase cytogenetic analyses revealed signal patterns indicating monoallelic deletion of TRAF3 in 3/20 other cases. SNP array analysis revealed a gain of copy number for MAP3K14 in three cHL cell lines. Gains of MAP3K14 were detected in 5/16 cases of primary cHL. In conclusion, in rare instances, HRS cells harbour inactivating mutations of the TRAF3 gene and recurrently show gains of MAP3K14, indicating that more components of NF-κB signalling show genetic lesions in HRS cells than previously known.


Sujet(s)
Maladie de Hodgkin/génétique , Polymorphisme de nucléotide simple , Protein-Serine-Threonine Kinases/génétique , Facteur-3 associé aux récepteurs de TNF/génétique , Adolescent , Adulte , Sujet âgé , Lignée cellulaire tumorale , Enfant , Analyse cytogénétique , Femelle , Délétion de gène , Dosage génique , Maladie de Hodgkin/métabolisme , Humains , Mâle , Adulte d'âge moyen , Protein-Serine-Threonine Kinases/métabolisme , Protéines proto-oncogènes/génétique , Protéines proto-oncogènes/métabolisme , Transduction du signal/génétique , Facteur-3 associé aux récepteurs de TNF/métabolisme , Transactivateurs/génétique , Transactivateurs/métabolisme ,
6.
Genes Chromosomes Cancer ; 51(4): 338-43, 2012 Apr.
Article de Anglais | MEDLINE | ID: mdl-22420028

RÉSUMÉ

Translocations involving immunoglobulin (IG) loci are the hallmarks of several subtypes of B-cell lymphoma. Common to these translocations is that cellular proto-oncogenes come under the influence of IG regulatory elements leading to deregulated expression. In case of a breakpoint in the IGH switch region, oncogene activation can take place on both derivative chromosomes, which means that in principle one translocation can result in concurrent activation of two genes. By fluorescence in situ hybridization (FISH), we identified a case of leukemic B-cell lymphoma in a child with an IGH break and unknown partner. Subsequent long-distance inverse PCR revealed fusion of IGH Sl in 14q32 and the 50 region of CBFA2T3 in 16q24.3, suggesting presence of the t(14;16)(q32;q24.3). Candidate oncogenes targeted through this translocation are CBFA2T3 and ACSF3, which could be activated on der(16) and der(14), respectively. FISH screening of a population-based cohort of B-cell lymphomas from a prospective trial for the treatment of lymphoma in childhood (BFM-NHL) identified additionally a follicular lymphoma Grade 3/diffuse large B-cell lymphoma with IGH-CBFA2T3/ACSF3 juxtaposition. Both lymphomas shared expression of CD10 and CD20 in the absence of TdT, suggesting a germinal center (GC) B-cell origin. Our data indicate that the CBFA2T3/ACSF3 locus is a novel recurrent oncogenic target of IGH translocations, which might contribute to the pathogenesis of pediatric GC-derived B-cell lymphoma.


Sujet(s)
Coenzyme A ligases/génétique , Gènes de chaine lourde d'immunoglobuline , Lymphome B/génétique , Protéines de répression/génétique , Translocation génétique , Protéines suppresseurs de tumeurs/génétique , Adolescent , Antigènes CD20/biosynthèse , Enfant , Chromosomes humains de la paire 14/génétique , Chromosomes humains de la paire 16/génétique , Femelle , Centre germinatif/anatomopathologie , Centre germinatif/physiologie , Humains , Hybridation fluorescente in situ , Lymphome B/anatomopathologie , Lymphome folliculaire/génétique , Mâle , Néprilysine/biosynthèse
7.
J Matern Fetal Neonatal Med ; 25(2): 200-2, 2012 Feb.
Article de Anglais | MEDLINE | ID: mdl-21476793

RÉSUMÉ

INTRODUCTION: We report on a prenatally diagnosed de novo small supernumerary marker chromosome (sSMC) derived form chromosome 18. Molecular cytogenetic studies led to information about the clinical relevance of the sSMC-induced chromosomal imbalance. As prenatal ultrasound was normal, detailed information with respect to prenatal counseling of the parents was necessary. In general, detection of an sSMC requires as much information on the exact genetic content with its possible impact on the phenotype as achievable. MATERIAL AND METHODS: Amniocentesis was performed in a 37-year-old Gravida IV Para II with a history of an induced abortion due to a prenatally diagnosed trisomy 21. Fluorescence in situ hybridization quick test gave hint on a possible mosaic trisomy 18, whereas the conventional banding cytogenetic analysis revealed an sSMC. The amount of euchromatin was estimated to be less than 5 MB. CONCLUSIONS: sSMC are rare, being present in less than 0.08% of all pregnancies. Going together with an abnormal ultrasound, counseling of the parents is relatively easy to perform. In cases of normal prenatal ultrasound, profound knowledge about the surplus genetic content is necessary for the estimation of the fetal outcome prognosis. In the present case, detailed molecular cytogenetics techniques led the parents to continue the pregnancy.


Sujet(s)
Chromosomes humains de la paire 18 , Mosaïcisme , Échographie prénatale , Adulte , Amniocentèse , Femelle , Conseil génétique , Humains , Grossesse
8.
Eur J Med Genet ; 55(1): 49-55, 2012 Jan.
Article de Anglais | MEDLINE | ID: mdl-21893220

RÉSUMÉ

A 9-year-old girl born to healthy parents showed manifestations suggestive of ataxia telangiectasia (AT), such as short stature, sudden short bouts of horizontal and rotary nystagmus, a weak and dysarthric voice, rolling gait, unstable posture, and atactic movements. She did not show several cardinal features typical of AT such as frequent, severe infections of the respiratory tract. In contrast, she showed symptoms not generally related to AT, including microcephaly, profound motor and mental retardation, small hands and feet, severely and progressively reduced muscle tone with slackly protruding abdomen and undue drooling, excess fat on her upper arms, and severe oligoarthritis. A cranial MRI showed no cerebellar hypoplasia and other abnormalities. In peripheral blood samples she carried a de novo duplication of 3.14 Mb in chromosomal region 19q12 containing six annotated genes, UQCRFS1, VSTM2B, POP4, PLEKHF1, CCNE1, and ZNF536, and a de novo mosaic inversion 14q11q32 (96% of metaphases). In a saliva-derived DNA sample only the duplication in 19q12 was detected, suggesting that the rearrangements in blood lymphocytes were acquired. These findings reinforced the suspicion that she had AT. AT was confirmed by strongly elevated serum AFP levels, cellular radiosensitivity and two inherited mutations in the ATM gene (c.510_511delGT; paternal origin and c.2922-50_2940del69; maternal origin). This case suggest that a defective ATM-dependent DNA damage response may entail additional stochastic genomic rearrangements. Screening for genomic rearrangements appears indicated in patients suspected of defective DNA damage responses.


Sujet(s)
Ataxie-télangiectasie/génétique , Duplication chromosomique , Chromosomes humains de la paire 19/génétique , Protéines régulatrices de l'apoptose/génétique , Ataxie-télangiectasie/diagnostic , Ataxie-télangiectasie/anatomopathologie , Protéines mutées dans l'ataxie-télangiectasie , Protéines du cycle cellulaire/génétique , Enfant , Inversion chromosomique , Altération de l'ADN , Protéines de liaison à l'ADN/génétique , Femelle , Humains , Lymphocytes/cytologie , Troubles mentaux/génétique , Troubles mentaux/anatomopathologie , Métaphase , Microcéphalie/génétique , Microcéphalie/anatomopathologie , Protein-Serine-Threonine Kinases/génétique , Salive/cytologie , Protéines suppresseurs de tumeurs/génétique
9.
Acta Haematol ; 127(1): 16-9, 2012.
Article de Anglais | MEDLINE | ID: mdl-21986343

RÉSUMÉ

Chromosomal abnormalities, like deletions, amplifications, inversions or translocations, are recurrent features in haematological malignancies. However, the precise molecular breakpoints are frequently not determined. Here we describe a rapid analysis of genetic imbalances combining fine tiling comparative genomic hybridization (FT-CGH) and ligation-mediated PCR (LM-PCR). We clarified an inv(14)(q11q32) in a case of T cell acute lymphoblastic leukaemia with a breakpoint in the TRA/D in 68% of cells detected by fluorescence in situ hybridization. FT-CGH showed several mono- and biallelic losses within TRA/D. LM-PCR disclosed a TRA/D rearrangement on one allele. The other allele revealed an inv(14)(q11q32), joining TRDD2 at 21,977,000 of 14q11 together with the IGH locus at 105,948,000 and 3'-sequence of TRAC at 22,092,000 joined together with IGHV4-61 at 106,166,000. This sensitive approach can unravel complex chromosomal abnormalities in patient samples with a limited amount of aberrant cells and may lead to better diagnostic and therapeutic options.


Sujet(s)
Aberrations des chromosomes , Réaction en chaîne par ligase , Leucémie-lymphome lymphoblastique à précurseurs T/génétique , Adulte , Humains , Mâle , Hybridation d'acides nucléiques , Leucémie-lymphome lymphoblastique à précurseurs T/diagnostic , Sensibilité et spécificité
10.
Br J Haematol ; 156(4): 499-507, 2012 Feb.
Article de Anglais | MEDLINE | ID: mdl-22171799

RÉSUMÉ

Approximately 70% of chronic lymphocytic leukaemia (CLL) patients present with early stage disease, therefore defining which patients will progress and require treatment is a major clinical challenge. Here, we present the largest study of prognostic markers ever carried out in Binet stage A patients (n = 1154) with a median follow-up of 8 years. We assessed the prognostic impact of lymphocyte doubling time (LDT), immunoglobulin gene (IGHV) mutation status, CD38 expression, ZAP-70 expression and fluorescence in situ hybridization (FISH) cytogenetics with regards to time to first treatment (TTFT) and overall survival (OS). Univariate analysis revealed LDT as the most prognostic parameter for TTFT, with IGHV mutation status most prognostic for OS. CD38 expression, ZAP-70 expression and FISH were also prognostic variables; combinations of these markers increased prognostic power in concordant cases. Multivariate analysis revealed that only LDT, IGHV mutation status, CD38 and age at diagnosis were independent prognostic variables for TTFT and OS. Therefore, IGHV mutation status and CD38 expression have independent prognostic value in early stage CLL and should be performed as part of the routine diagnostic workup. ZAP-70 expression and FISH were not independent prognostic markers in early stage disease and can be omitted at diagnosis but FISH analysis should be undertaken at disease progression to direct treatment strategy.


Sujet(s)
Antigènes CD38/génétique , Région variable d'immunoglobuline/génétique , Leucémie chronique lymphocytaire à cellules B/génétique , ZAP-70 Protein-tyrosine kinase/génétique , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Régulation de l'expression des gènes dans la leucémie , Humains , Estimation de Kaplan-Meier , Leucémie chronique lymphocytaire à cellules B/mortalité , Leucémie chronique lymphocytaire à cellules B/anatomopathologie , Adulte d'âge moyen , Mutation , Stadification tumorale , Pronostic , Facteurs temps , Jeune adulte
11.
Proc Natl Acad Sci U S A ; 108(30): 12461-6, 2011 Jul 26.
Article de Anglais | MEDLINE | ID: mdl-21746927

RÉSUMÉ

The chromosomal translocation t(11;14)(q13;q32) leading to cyclin-D1 overexpression plays an essential role in the development of mantle cell lymphoma (MCL), an aggressive tumor that remains incurable with current treatment strategies. Cyclin-D1 has been postulated as an effective therapeutic target, but the evaluation of this target has been hampered by our incomplete understanding of its oncogenic functions and by the lack of valid MCL murine models. To address these issues, we generated a cyclin-D1-driven mouse model in which cyclin-D1 expression can be regulated externally. These mice developed cyclin-D1-expressing lymphomas capable of recapitulating features of human MCL. We found that cyclin-D1 inactivation was not sufficient to induce lymphoma regression in vivo; however, using a combination of in vitro and in vivo assays, we identified a novel prosurvival cyclin-D1 function in MCL cells. Specifically, we found that cyclin-D1, besides increasing cell proliferation through deregulation of the cell cycle at the G(1)-S transition, sequestrates the proapoptotic protein BAX in the cytoplasm, thereby favoring BCL2's antiapoptotic function. Accordingly, cyclin-D1 inhibition sensitized the lymphoma cells to apoptosis through BAX release. Thus, genetic or pharmacologic targeting of cyclin-D1 combined with a proapoptotic BH3 mimetic synergistically killed the cyclin-D1-expressing murine lymphomas, human MCL cell lines, and primary lymphoma cells. Our study identifies a role of cyclin-D1 in deregulating apoptosis in MCL cells, and highlights the potential benefit of simultaneously targeting cyclin-D1 and survival pathways in patients with MCL. This effective combination therapy also might be exploited in other cyclin-D1-expressing tumors.


Sujet(s)
Cycline D1/métabolisme , Lymphome à cellules du manteau/métabolisme , Protéine Bax/métabolisme , Animaux , Antinéoplasiques/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Dérivés du biphényle/pharmacologie , Cycle cellulaire , Lignée cellulaire tumorale , Survie cellulaire , Cycline D1/antagonistes et inhibiteurs , Cycline D1/génétique , Modèles animaux de maladie humaine , Amplification de gène , Gènes bcl-2 , Humains , Lymphome à cellules du manteau/étiologie , Lymphome à cellules du manteau/anatomopathologie , Lymphome à cellules du manteau/thérapie , Souris , Nitrophénols/pharmacologie , Pipérazines/pharmacologie , Sulfonamides/pharmacologie , Tests d'activité antitumorale sur modèle de xénogreffe , Protéine Bax/antagonistes et inhibiteurs
12.
Eur J Med Genet ; 54(5): e505-9, 2011.
Article de Anglais | MEDLINE | ID: mdl-21736959

RÉSUMÉ

Interstitial deletions of chromosome 14 have rarely been described. We report on a boy in whom a 2 Mb deletion in 14q13 was discovered by array CGH. The deletion was a de novo event. The boy presented with asymmetrical growth retardation at birth. There was severe developmental delay with muscular hypotonia and focal epilepsy with apneic episodes progressing to serial tonic seizures. At the age of 3 3/12 years he was diagnosed with pneumonia. In the further course he developed symptoms of hemophagocytic lymphohistiocytosis. He died due to organ failure. Herein the clinical findings are compared to patients with cytogenetic visible deletions encompassing the region deleted in the proband and the possible connection with the deleted genes.


Sujet(s)
Délétion de segment de chromosome , Chromosomes humains de la paire 14/génétique , Incapacités de développement/génétique , Lymphohistiocytose hémophagocytaire/génétique , Enfant d'âge préscolaire , Hybridation génomique comparative , Issue fatale , Humains , Hybridation fluorescente in situ , Nourrisson , Nouveau-né , Caryotypage , Lymphohistiocytose hémophagocytaire/complications , Lymphohistiocytose hémophagocytaire/anatomopathologie , Lymphohistiocytose hémophagocytaire/thérapie , Mâle , Phénotype
13.
Blood ; 118(8): 2239-42, 2011 Aug 25.
Article de Anglais | MEDLINE | ID: mdl-21705501

RÉSUMÉ

Imatinib-resistant tyrosine kinase (TK) fusions involving FGFR1, JAK2, or FLT3 are rare but recurrent in patients with eosinophilia-associated neoplasms. We report here 2 male patients with ETV6-FLT3(+) myeloid/lymphoid neoplasms with eosinophilia who were treated with the multitargeted TK inhibitors sunitinib and sorafenib. Patient 1 achieved rapid complete hematologic response and complete cytogenetic response after 3 months of taking sunitinib. A secondary blast phase caused by clonal evolution was diagnosed after 6 months. He achieved a second complete hematologic response after taking sorafenib but relapsed 2 months later. An N841K point mutation within the TK domain of FLT3, previously reported in acute myeloid leukemia and potentially conferring resistance to sorafenib, was subsequently identified. Patient 2 was heavily pretreated according to the initial diagnosis of T-lymphoblastic lymphoma and died in sunitinib-induced pancytopenia. This report highlights the importance of a careful diagnostic workup for eosinophilia-associated neoplasms to evaluate the possibility of TK inhibitor therapy.


Sujet(s)
Tumeurs hématologiques/traitement médicamenteux , Tumeurs hématologiques/génétique , Protéines de fusion oncogènes/génétique , Protéines proto-oncogènes c-ets/génétique , Protéines de répression/génétique , Tyrosine kinase-3 de type fms/antagonistes et inhibiteurs , Tyrosine kinase-3 de type fms/génétique , Adulte , Séquence d'acides aminés , Antinéoplasiques/usage thérapeutique , Séquence nucléotidique , Benzènesulfonates/usage thérapeutique , ADN tumoral/génétique , Résistance aux médicaments antinéoplasiques/génétique , Éosinophilie/complications , Éosinophilie/traitement médicamenteux , Éosinophilie/génétique , Tumeurs hématologiques/complications , Humains , Indoles/usage thérapeutique , Mâle , Adulte d'âge moyen , Données de séquences moléculaires , Nicotinamide/analogues et dérivés , Fusion oncogène , Phénylurées , Mutation ponctuelle , Inhibiteurs de protéines kinases/usage thérapeutique , Pyridines/usage thérapeutique , Pyrroles/usage thérapeutique , Sorafénib , Sunitinib ,
14.
Am J Surg Pathol ; 35(6): 933-5, 2011 Jun.
Article de Anglais | MEDLINE | ID: mdl-21566516

RÉSUMÉ

Atypical teratoid/rhabdoid tumors (AT/RTs) are highly aggressive brain tumors of early childhood poorly responding to therapy. The majority of cases show inactivation of SMARCB1 (INI1, hSNF5, BAF47), a core member of the adenosine triphosphate (ATP)-dependent SWI/SNF chromatin-remodeling complex. We here report the case of a supratentorial AT/RT in a 9-month-old boy, which showed retained SMARCB1 staining on immunohistochemistry and lacked genetic alterations of SMARCB1. Instead, the tumor showed loss of protein expression of another SWI/SNF chromatin-remodeling complex member, the ATPase subunit SMARCA4 (BRG1) due to a homozygous SMARCA4 mutation [c.2032C>T (p.Q678X)]. Our findings highlight the role of SMARCA4 in the pathogenesis of SMARCB1-positive AT/RT and the usefulness of antibodies directed against SMARCA4 in this diagnostic setting.


Sujet(s)
Tumeurs du cerveau/anatomopathologie , Protéines chromosomiques nonhistones/génétique , Codon non-sens , Helicase/génétique , Protéines de liaison à l'ADN/génétique , Protéines nucléaires/génétique , Tumeur rhabdoïde/anatomopathologie , Tératome/anatomopathologie , Facteurs de transcription/génétique , Tumeurs du cerveau/génétique , Tumeurs du cerveau/thérapie , Protéines chromosomiques nonhistones/métabolisme , Association thérapeutique , Protéines de liaison à l'ADN/métabolisme , Issue fatale , Extinction de l'expression des gènes , Humains , Nourrisson , Mâle , Tumeur rhabdoïde/génétique , Tumeur rhabdoïde/thérapie , Protéine SMARCB1 , Tératome/génétique , Tératome/thérapie , Facteurs de transcription/métabolisme
15.
Blood ; 118(1): 139-47, 2011 Jul 07.
Article de Anglais | MEDLINE | ID: mdl-21487109

RÉSUMÉ

The prognosis of germinal center-derived B-cell (GCB) lymphomas, including follicular lymphoma and diffuse large-B-cell lymphoma (DLBCL), strongly depends on age. Children have a more favorable outcome than adults. It is not known whether this is because of differences in host characteristics, treatment protocols, or tumor biology, including the presence of chromosomal alterations. By screening for novel IGH translocation partners in pediatric and adult lymphomas, we identified chromosomal translocations juxtaposing the IRF4 oncogene next to one of the immunoglobulin (IG) loci as a novel recurrent aberration in mature B-cell lymphoma. FISH revealed 20 of 427 lymphomas to carry an IG/IRF4-fusion. Those were predominantly GCB-type DLBCL or follicular lymphoma grade 3, shared strong expression of IRF4/MUM1 and BCL6, and lacked PRDM1/BLIMP1 expression and t(14;18)/BCL2 breaks. BCL6 aberrations were common. The gene expression profile of IG/IRF4-positive lymphomas differed from other subtypes of DLBCL. A classifier for IG/IRF4 positivity containing 27 genes allowed accurate prediction. IG/IRF4 positivity was associated with young age and a favorable outcome. Our results suggest IRF4 translocations to be primary alterations in a molecularly defined subset of GCB-derived lymphomas. The probability for this subtype of lymphoma significantly decreases with age, suggesting that diversity in tumor biology might contribute to the age-dependent differences in prognosis of lymphoma.


Sujet(s)
Gènes de chaine lourde d'immunoglobuline/génétique , Centre germinatif/anatomopathologie , Facteurs de régulation d'interféron/génétique , Lymphome B/génétique , Lymphome B/anatomopathologie , Translocation génétique , Adolescent , Adulte , Âge de début , Sujet âgé , Séquence nucléotidique , Enfant , Enfant d'âge préscolaire , Chromosomes humains de la paire 14 , Chromosomes humains de la paire 6 , Femelle , Gènes de chaine lourde d'immunoglobuline/immunologie , Humains , Facteurs de régulation d'interféron/immunologie , Lymphome B/immunologie , Mâle , Adulte d'âge moyen , Données de séquences moléculaires , Protéines de fusion oncogènes/génétique , Pronostic , Jeune adulte
18.
Cancer Genet Cytogenet ; 203(2): 149-60, 2010 Dec.
Article de Anglais | MEDLINE | ID: mdl-21156227

RÉSUMÉ

To understand the influence of chromosomal alterations on gene expression in a genome-wide view, chromosomal imbalances detected by single nucleotide polymorphism (SNP) chips were compared with global gene expression in 16 cases of chronic lymphocytic leukemia (CLL). A strong concordance between chromosomal gain or loss and increased or reduced expression of genes in the affected regions was found, respectively. Regions of uniparental disomy (UPD) were rare and had usually no consistent influence on gene expression, but in one instance, a large UPD was associated with a downregulation of most genes in the affected chromosome. The frequently deleted miRNAs, MIRN15A and MIRN16-1, did not show a reduced expression in cases with monoallelic deletions. The BCL2 protein, considered to be downregulated by these miRNAs, was upregulated not only in CLL with biallelic deletion of MIRN15A and MIRN16-1, but also in cases with monoallelic deletion. This suggests a complex regulation of BCL2 levels in CLL cells. Taken together, in CLL, a global gene dosage effect exists for chromosomal gains and deletions and in some instances for UPDs. We did not confirm a consistent correlation between MIRN15A and MIRN16-1 expression levels and BCL2 protein levels, indicating a complex regulation of BCL2 expression.


Sujet(s)
Dosage génique , Régulation de l'expression des gènes dans la leucémie , Leucémie chronique lymphocytaire à cellules B/génétique , Polymorphisme de nucléotide simple , Protéines mutées dans l'ataxie-télangiectasie , Protéines du cycle cellulaire/génétique , Cartographie chromosomique , DEAD-box RNA helicases/génétique , Protéines de liaison à l'ADN/génétique , Régulation de l'expression des gènes , Humains , Hybridation fluorescente in situ , microARN/métabolisme , Protein-Serine-Threonine Kinases/génétique , Protéines proto-oncogènes c-bcl-2/génétique , RT-PCR , Protéines suppresseurs de tumeurs/génétique , Disomie uniparentale/génétique , Régulation positive
19.
Mol Cytogenet ; 3: 21, 2010 Nov 05.
Article de Anglais | MEDLINE | ID: mdl-21054846

RÉSUMÉ

BACKGROUND: Typical Williams-Beuren syndrome (WBS) is commonly caused by a ~1.5 Mb - ~1.8 Mb heterozygous deletion of contiguous genes at chromosome region 7q11.23. The majority of WBS cases occurs sporadically but few familial cases of autosomal dominant inheritance have been reported. Recent data demonstrated the existence of the paracentric inversion polymorphism at the WBS critical region in 7q11.23 in some of the progenitors transmitting the chromosome which shows the deletion in the affected child. In parents having a child affected by WBS the prevalence of such a structural variant has been reported to be much higher (~25- ~30%) than in the general population (~1- ~6%). However, in these previously reported studies only a limited number of randomly selected patients and non transmitting parents of WBS patients were used as controls, but without specification of any clinical data. Therefore we have undertaken a German population-based molecular cytogenetic investigation. We evaluated the incidence of the paracentric inversion polymorphism at 7q11.23 analyzing interphase nuclei of lymphocytes using a three color fluorescence in situ hybridization (FISH) probe. RESULTS: FISH analysis was carried out on couples with a child affected by WBS as compared to a population sample composed of different normal individuals: Control group I: couples with two healthy children, control group II: couples with fertility problems, planning ICSI and control group III: couples with two healthy children and one child with a chromosome aberration, not involving region 7q11.23. The three color FISH assay showed that the frequency of the paracentric inversion polymorphism at 7q11.23 in couples with a child affected by WBS was 20.8% (5 out of 24 pairs) as compared to 8.3% (2 out of 24 pairs, control group I), 25% (4 out of 16 pairs, control group II) and 9.1% (1 out of 11 pairs, control group III), respectively (total 7 out of 51 pairs, 13.8%). The frequencies differed between the groups, but this was statistically not significant (p > 0.05, Fisher's test). CONCLUSION: Our results do not support the hypothesis that the paracentric inversion polymorphism at 7q11.23 is a major predisposing factor for the WBS deletion.

SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE