Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Genes Chromosomes Cancer ; 60(8): 586-590, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33896072

RESUMO

Atypical teratoid/rhabdoid tumor (AT/RT) is a malignant brain tumor predominantly occurring in infants. Biallelic SMARCB1 mutations causing loss of nuclear SMARCB1/INI1 protein expression represent the characteristic genetic lesion. Pathogenic SMARCB1 mutations comprise single nucleotide variants, small insertions/deletions, large deletions, which may be also present in the germline (rhabdoid tumor predisposition syndrome 1), as well as somatic copy-number neutral loss of heterozygosity (LOH). In some SMARCB1-deficient AT/RT underlying biallelic mutations cannot be identified. Here we report the case of a 24-months-old girl diagnosed with a large brain tumor. The malignant rhabdoid tumor showed loss of nuclear SMARCB1/INI1 protein expression and the diagnosis of AT/RT was confirmed by DNA methylation profiling. While FISH, MLPA, Sanger sequencing and DNA methylation data-based imbalance analysis did not disclose alterations affecting SMARCB1, OncoScan array analysis revealed a 28.29 Mb sized region of copy-number neutral LOH on chromosome 22q involving the SMARCB1 locus. Targeted next-generation sequencing did also not detect a single nucleotide variant but instead revealed insertion of an AluY element into exon 2 of SMARCB1. Specific PCR-based Sanger sequencing verified the Alu insertion (SMARCB1 c.199_200 Alu ins) resulting in a frame-shift truncation not present in the patient's germline. In conclusion, transposable element insertion represents a hitherto not widely recognized mechanism of SMARCB1 disruption in AT/RT, which might not be detected by several widely applied conventional diagnostics assays. This finding has particular clinical implications, if rhabdoid predisposition syndrome 1 is suspected, but germline SMARCB1 alterations cannot be identified.


Assuntos
Neoplasias Encefálicas/genética , Tumor Rabdoide/genética , Proteína SMARCB1/genética , Teratoma/genética , Neoplasias Encefálicas/patologia , Elementos de DNA Transponíveis , Feminino , Humanos , Lactente , Mutagênese Insercional , Tumor Rabdoide/patologia , Teratoma/patologia
2.
Acta Neuropathol ; 142(2): 361-374, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34003336

RESUMO

Loss of nuclear SMARCB1 (INI1/hSNF5/BAF47) protein expression due to biallelic mutations of the SMARCB1 tumor suppressor gene is a hallmark of atypical teratoid/rhabdoid tumors (ATRT), but the presence of cytoplasmic SMARCB1 protein in these tumors has not yet been described. In a series of 102 primary ATRT, distinct cytoplasmic SMARCB1 staining on immunohistochemistry was encountered in 19 cases (19%) and was highly over-represented in cases showing pathogenic sequence variants leading to truncation or mutation of the C-terminal part of SMARCB1 (15/19 vs. 4/83; Chi-square: 56.04, p = 1.0E-10) and, related to this, in tumors of the molecular subgroup ATRT-TYR (16/36 vs. 3/66; Chi-square: 24.47, p = 7.6E-7). Previous reports have indicated that while SMARCB1 lacks a bona fide nuclear localization signal, it harbors a masked nuclear export signal (NES) and that truncation of the C-terminal region results in unmasking of this NES leading to cytoplasmic localization. To determine if cytoplasmic localization found in ATRT is due to unmasking of NES, we generated GFP fusions of one of the SMARCB1 truncating mutations (p.Q318X) found in the tumors along with a p.L266A mutation, which was shown to disrupt the interaction of SMARCB1-NES with exportin-1. We found that while the GFP-SMARCB1(Q318X) mutant localized to the cytoplasm, the double mutant GFP-SMARCB1(Q318X;L266A) localized to the nucleus, confirming NES requirement for cytoplasmic localization. Furthermore, cytoplasmic SMARCB1(Q318X) was unable to cause senescence as determined by morphological observations and by senescence-associated ß-galactosidase assay, while nuclear SMARCB1(Q318X;L266A) mutant regained this function. Selinexor, a selective exportin-1 inhibitor, was effective in inhibiting the nuclear export of SMARCB1(Q318X) and caused rapid cell death in rhabdoid tumor cells. In conclusion, inhibition of nuclear export restores nuclear localization and residual tumor suppressor function of truncated SMARCB1. Therapies aimed at preventing nuclear export of mutant SMARCB1 protein may represent a promising targeted therapy in ATRT harboring truncating C-terminal SMARCB1 mutations.


Assuntos
Transporte Ativo do Núcleo Celular/fisiologia , Neoplasia Residual/genética , Tumor Rabdoide/metabolismo , Proteína SMARCB1/metabolismo , Neoplasias do Sistema Nervoso Central/genética , Neoplasias do Sistema Nervoso Central/metabolismo , Pré-Escolar , Feminino , Genes Supressores de Tumor/fisiologia , Humanos , Lactente , Masculino , Mutação/genética , Neoplasia Residual/metabolismo , Neoplasias Neuroepiteliomatosas/genética , Neoplasias Neuroepiteliomatosas/metabolismo , Tumor Rabdoide/genética , Proteína SMARCB1/genética , Teratoma/genética
3.
Acta Neuropathol ; 139(2): 277-286, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31732806

RESUMO

Atypical teratoid/rhabdoid tumor (ATRT) is a highly malignant brain tumor predominantly occurring in infants. Mutations of the SMARCB1 gene are the characteristic genetic lesion. SMARCB1-mutant tumors in adolescents and adults are rare and may show uncommon histopathological and clinical features. Here we report seven SMARCB1-deficient intracranial tumors sharing distinct clinical, histopathological and molecular features. Median age of the four females and three males was 40 years (range 15-61 years). All tumors were located in the pineal region. Histopathologically, these tumors displayed spindled and epithelioid cells embedded in a desmoplastic stroma alternating with a variable extent of a loose myxoid matrix. All cases showed loss of nuclear SMARCB1/INI1 protein expression, expression of EMA and CD34 was frequent and the Ki67/MIB1 proliferation index was low in the majority of cases (median 3%). Three cases displayed heterozygous SMARCB1 deletions and two cases a homozygous SMARCB1 deletion. On sequencing, one tumor showed a 2 bp deletion in exon 4 (c.369_370del) and one a short duplication in exon 3 (c.237_276dup) both resulting in frameshift mutations. Most DNA methylation profiles were not classifiable using the Heidelberg Brain Tumor Classifier (version v11b4). By unsupervised t-SNE analysis and hierarchical clustering analysis, however, all tumors grouped closely together and showed similarities with ATRT-MYC. After a median observation period of 48 months, three patients were alive with stable disease, whereas one patient experienced tumor progression and three patients had succumbed to disease. In conclusion, our series represents an entity with distinct clinical, histopathological and molecular features showing epigenetic similarities with ATRT-MYC. We propose the designation desmoplastic myxoid tumor (DMT), SMARCB1-mutant, for these tumors.


Assuntos
Neoplasias Encefálicas/genética , Mutação/genética , Glândula Pineal , Tumor Rabdoide/genética , Tumor Rabdoide/patologia , Proteína SMARCB1/genética , Adolescente , Adulto , Fatores Etários , Neoplasias Encefálicas/mortalidade , Neoplasias Encefálicas/patologia , Estudos de Coortes , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Tumor Rabdoide/mortalidade , Taxa de Sobrevida , Adulto Jovem
4.
Am J Pathol ; 186(12): 3285-3296, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27765635

RESUMO

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.


Assuntos
Neoplasias de Bainha Neural/genética , Neurilemoma/genética , Neurofibroma/genética , Neurofibromatoses/genética , Neurofibromatose 1/genética , Neoplasias Cutâneas/genética , alfa Catenina/genética , Adolescente , Adulto , Idoso , Cromossomos Humanos Par 22/genética , Hibridização Genômica Comparativa , Transição Epitelial-Mesenquimal , Feminino , Humanos , Hibridização in Situ Fluorescente , Masculino , Pessoa de Meia-Idade , Monossomia , Neoplasias de Bainha Neural/patologia , Neurilemoma/patologia , Neurofibroma/patologia , Neurofibromatoses/patologia , Neurofibromatose 1/patologia , Células de Schwann/metabolismo , Células de Schwann/patologia , Neoplasias Cutâneas/patologia , Proteínas Supressoras de Tumor/genética , Adulto Jovem
5.
Genes Chromosomes Cancer ; 55(12): 925-931, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27356182

RESUMO

Small cell undifferentiated (SCUD) hepatoblastoma is a rare variant of hepatoblastoma with poor outcome and loss of INI1 expression, sharing this with malignant rhabdoid tumors (MRT). We studied all tumors from the files of the Kiel Pediatric Tumor Registry (KTR) with the initial diagnosis of SCUD and MRT. After re-review, we performed immunistochemistry, fluorescence in situ hybridization, and multiplex ligation dependent probe amplification for loss of expression and deletion of INI1/SMARCB1 in 23 tumors. Morphologically, 12 of the tumors had a small cell morphology, 9 showed the typical picture of MRT, and 2 were composed of both small cells and rhabdoid cells. All but 1 of the 23 tumors showed loss of INI1 protein expression by immunohistochemistry. Nineteen of the INI1 negative tumors were analyzed by FISH technique and all showed a deletion of the INI1/SMARCB1 gene (17 homozygous deletions, 2 heterozygous deletions). We investigated 14 of these cases by multiplex ligation dependent probe amplification and verified the deletions in all cases. In conclusion, we postulate that SCUD hepatoblastoma is not a hepatoblastoma but represents a malignant rhabdoid tumor of the liver. © 2016 Wiley Periodicals, Inc.


Assuntos
Biomarcadores Tumorais/genética , Deleção de Genes , Hepatoblastoma/genética , Neoplasias Hepáticas/genética , Neoplasias Pulmonares/genética , Tumor Rabdoide/genética , Proteína SMARCB1/genética , Carcinoma de Pequenas Células do Pulmão/genética , Adolescente , Diferenciação Celular , Pré-Escolar , Feminino , Seguimentos , Hepatoblastoma/metabolismo , Hepatoblastoma/patologia , Humanos , Imuno-Histoquímica , Hibridização in Situ Fluorescente , Lactente , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Masculino , Estadiamento de Neoplasias , Tumor Rabdoide/metabolismo , Tumor Rabdoide/patologia , Carcinoma de Pequenas Células do Pulmão/metabolismo , Carcinoma de Pequenas Células do Pulmão/patologia
6.
Z Geburtshilfe Neonatol ; 221(1): 39-42, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27383044

RESUMO

We report on 3 male neonates with hereditary ADAMTS13 deficiency (Upshaw Schulman syndrome, USS), the inherited form of thrombotic thrombocytopenic purpura (TTP). 2 presented shortly after birth with thrombocytopenia followed by microangiopathic Coombs-negative haemolytic anaemia. Both initially received antibiotic treatment for suspected infection-associated thrombocytopenia. In one patient's brother, the first bout of incipient TTP did not occur before 6 months of age, despite the same genetic defect. ADAMTS13 activity was<5%, compound heterozygous mutations were found in all patients. USS constitutes a differential diagnosis to thrombocytopenia caused by disseminated intravascular coagulation in neonatal septicaemia. Administration of fresh frozen plasma usually resolves acute bouts of the disease. In some cases of thrombocytopenia of unknown origin in infancy, the resolution of signs and symptoms after infusion of plasma may point towards the diagnosis.


Assuntos
Coagulação Intravascular Disseminada/complicações , Coagulação Intravascular Disseminada/diagnóstico , Sepse Neonatal/complicações , Sepse Neonatal/diagnóstico , Púrpura Trombocitopênica Trombótica/diagnóstico , Antibacterianos/administração & dosagem , Transfusão de Componentes Sanguíneos/métodos , Terapia Combinada/métodos , Diagnóstico Diferencial , Coagulação Intravascular Disseminada/terapia , Humanos , Recém-Nascido , Doenças do Recém-Nascido/diagnóstico , Doenças do Recém-Nascido/terapia , Masculino , Sepse Neonatal/terapia , Plasma , Púrpura Trombocitopênica Trombótica/terapia
7.
Pediatr Blood Cancer ; 63(1): 168-70, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26275168

RESUMO

Sickle cell disease is among hereditary diseases with evidence that early diagnoses and treatment improves the clinical outcome. So far sickle cell disease has not been included in the German newborn screening program despite immigration from countries with populations at risk. To determine the birth prevalence we tested 17,018 newborns. High pressure liquid chromatography and subsequent molecular-genetic testing were used for the detection and confirmation of hemoglobin variants. The frequency of sickle cell disease-consistent genotypes was one in 2,385 newborns. Duffy-blood group typing showed evidence that affected children were likely of Sub-Saharan ancestry. An inclusion of sickle cell disease into the German newborn screening seems reasonable.


Assuntos
Anemia Falciforme/epidemiologia , Anemia Falciforme/prevenção & controle , Triagem Neonatal , Genótipo , Alemanha/epidemiologia , Humanos , Recém-Nascido , Triagem Neonatal/métodos , Prevalência
8.
Pediatr Blood Cancer ; 63(8): 1451-3, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27092963

RESUMO

We report the successful use of multiplex ligation-dependent probe amplification (MLPA) to detect heterozygous loss of SMARCB1/INI1/SNF5 in the germ line of an infant with a huge posterior fossa tumor. MLPA and Sanger sequencing of the SMARCB1 gene in the germ line may be useful for the initial diagnosis in a defined subgroup of infants with rhabdoid tumors, in which biopsies cannot be performed.


Assuntos
Neoplasias do Sistema Nervoso Central/diagnóstico , Neoplasias do Sistema Nervoso Central/genética , Predisposição Genética para Doença , Tumor Rabdoide/diagnóstico , Tumor Rabdoide/genética , Proteína SMARCB1/genética , Neoplasias do Sistema Nervoso Central/mortalidade , Deleção de Genes , Humanos , Lactente , Reação em Cadeia da Polimerase Multiplex , Tumor Rabdoide/mortalidade
10.
Childs Nerv Syst ; 32(6): 1157-61, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26747622

RESUMO

INTRODUCTION: Atypical teratoid/rhabdoid tumor (AT/RT) is an aggressive embryonal tumor of the central nervous system with a generally dismal prognosis, especially in patients younger than 12 months. DISCUSSION: We here describe the unusual case of an infant with AT/RT with long-term survival despite low-cumulative dose chemotherapy after subtotal resection. Due to a poor neurological situation and an unfavorable oncological prognosis, therapy was halted after two partial surgical resections and four of the nine chemotherapy courses recommended by the European Rhabdoid Registry, without the patient receiving either radiotherapy or high-dose chemotherapy. The patient is alive without evidence of disease 52 months after diagnosis. CONCLUSION: This case report highlights that independent prognostic factors are urgently needed for optimizing treatment stratification and preventing overtreatment.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/cirurgia , Procedimentos Neurocirúrgicos/métodos , Tumor Rabdoide , Terapia Combinada , Feminino , Humanos , Lactente , Imageamento por Ressonância Magnética , Tumor Rabdoide/tratamento farmacológico , Tumor Rabdoide/mortalidade , Tumor Rabdoide/cirurgia
11.
Pediatr Blood Cancer ; 62(5): 897-900, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25663425
13.
Genes Chromosomes Cancer ; 52(2): 185-90, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23074045

RESUMO

Atypical teratoid/rhabdoid tumor (AT/RT) is a rare malignant pediatric brain tumor characterized by genetic alterations affecting the SMARCB1 (hSNF5/INI1) locus in chromosome band 22q11.2. To identify potential additional genetic alterations, high-resolution genome-wide analysis was performed using a molecular inversion probe single-nucleotide polymorphism (MIP SNP) assay (Affymetrix OncoScan formalin-fixed paraffin-embedded express) on DNA isolated from 18 formalin-fixed paraffin-embedded archival samples. Alterations affecting the SMARCB1 locus could be demonstrated by MIP SNP in 15 out of 16 evaluable cases (94%). These comprised five tumors with homozygous deletions, six tumors with heterozygous deletions, and four tumors with copy number neutral loss of heterozygosity (LOH) involving chromosome band 22q11.2. Remarkably, MIB SNP analysis did not yield any further recurrent chromosomal gains, losses, or copy neutral LOH. On MIP SNP screening for somatic mutations, the presence of a SMARCB1 mutation (c.472C>T p.R158X) was confirmed, but no recurrent mutations of other cancer relevant genes could be identified. Results of fluorescence in situ hybridization, multiplex ligation-dependent probe amplification, and SMARCB1 sequencing were highly congruent with that of the MIP SNP assay. In conclusion, these data further suggest the absence of recurrent genomic alterations other than SMARCB1 in AT/RT.


Assuntos
Proteínas Cromossômicas não Histona/genética , Proteínas de Ligação a DNA/genética , Mutação , Tumor Rabdoide/genética , Fatores de Transcrição/genética , Pré-Escolar , Aberrações Cromossômicas , Cromossomos Humanos Par 22/genética , Variações do Número de Cópias de DNA , Análise Mutacional de DNA , Feminino , Deleção de Genes , Estudo de Associação Genômica Ampla , Genótipo , Humanos , Hibridização in Situ Fluorescente , Lactente , Recém-Nascido , Perda de Heterozigosidade , Masculino , Reação em Cadeia da Polimerase Multiplex , Polimorfismo de Nucleotídeo Único , Proteína SMARCB1
14.
Am J Hum Genet ; 86(2): 279-84, 2010 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-20137775

RESUMO

Rhabdoid tumors of early infancy are highly aggressive with consequent poor prognosis. Most cases show inactivation of the SMARCB1 (also known as INI1 and hSNF5) tumor suppressor, a core member of the ATP-dependent SWI/SNF chromatin-remodeling complex. Familial cases, described as rhabdoid tumor predisposition syndrome (RTPS), have been linked to heterozygous SMARCB1 germline mutations. We identified inactivation of another member of the SWI/SNF chromatin-remodeling complex, its ATPase subunit SMARCA4 (also known as BRG1), due to a SMARCA4/BRG1 germline mutation and loss of heterozygosity by uniparental disomy in the tumor cells of two sisters with rhabdoid tumors lacking SMARCB1 mutations. SMARCA4 is thus a second member of the SWI/SNF complex involved in cancer predisposition. Its general involvement in other tumor entities remains to be established.


Assuntos
Códon sem Sentido/genética , DNA Helicases/genética , Inativação Gênica , Predisposição Genética para Doença , Mutação em Linhagem Germinativa/genética , Proteínas Nucleares/genética , Tumor Rabdoide/genética , Fatores de Transcrição/genética , Sequência de Bases , DNA Helicases/química , Análise Mutacional de DNA , Evolução Fatal , Feminino , Humanos , Imuno-Histoquímica , Lactente , Imageamento por Ressonância Magnética , Masculino , Dados de Sequência Molecular , Proteínas Nucleares/química , Linhagem , Tumor Rabdoide/patologia , Síndrome , Fatores de Transcrição/química
15.
Ann Hematol ; 92(3): 387-94, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23180434

RESUMO

Mutations in transmembrane protease, serine 6 (TMPRSS6) cause iron refractory iron deficiency anemia (IRIDA). Parenteral iron administration may slightly improve hemoglobin level but is troublesome for patients. Optimal treatment has yet to be determined. We identified five patients from four independent families displaying the IRIDA picture with truncating biallelic mutations in TMPRSS6, one of which is novel. Liver iron determined by superconducting quantum interference device biosusceptometry ranged from 390 to 720 µg Fe/g wet weight (normal range 100-500; n = 3). Intestinal iron absorption (12 and 32 %, normal range 10-50; n = 2) and 59Fe erythrocyte incorporation after ingestion of 59Fe (57 and 38 %, normal range 70-90; n = 2) were inadequately low for iron-deficient anemic individuals. Baseline serum erythropoietin was elevated or borderline high in four patients. Administration of recombinant human erythropoietin (rhEPO) at up to 273 and 188 U/kg body weight/week alone did not improve anemia or result in a decrease of urinary hepcidin in two individuals. In conclusion, the ability of exogenous rhEPO to increase hemoglobin level appears to be impaired in IRIDA.


Assuntos
Anemia Ferropriva/tratamento farmacológico , Anemia Ferropriva/genética , Eritropoetina/administração & dosagem , Fenótipo , Adolescente , Anemia Ferropriva/sangue , Criança , Eritropoetina/sangue , Feminino , Humanos , Masculino , Proteínas de Membrana/genética , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/sangue , Serina Endopeptidases/genética , Adulto Jovem
16.
Blood ; 115(23): 4894-901, 2010 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-20351307

RESUMO

Among the different phenotypes of von Willebrand disease (VWD) type 2A, we identified a particular subgroup with a high frequency of 29%, characterized by a relative decrease of large von Willebrand factor (VWF) multimers and decreased A Disintegrin And Metalloproteinase with ThromboSpondin type 1 motifs, member 13 (ADAMTS13)-mediated proteolysis previously described in a single family as VWD type IIE (VWD2A/IIE). Phenotype and genotype of 57 patients from 38 unrelated families displaying a particular multimer pattern resembling the original VWD2A/IIE were studied. Pathogenicity of candidate mutations was confirmed by expression studies and phenotypic characterization of recombinant mutants. Specific mutations were identified in all patients. Twenty-two different mutations, most of them affecting cysteine residues, 17 of them being novel, are clustering mainly in the VWF D3 domain and correlate with the VWD2A/IIE phenotype. An intracellular retention of most mutants and/or a defect of multimerization seem to be the main pathogenic molecular mechanisms. ADAMTS13 proteolysis of mutant VWF was not different from wild-type VWF in a static assay, suggesting that reduced in vivo proteolysis is not an intrinsic property of mutant VWF. Our study identified a distinct VWD subtype with a common molecular background which contributes significantly to the heterogeneous spectrum of VWD.


Assuntos
Mutação de Sentido Incorreto , Multimerização Proteica/genética , Doença de von Willebrand Tipo 2/genética , Doença de von Willebrand Tipo 2/metabolismo , Fator de von Willebrand/genética , Fator de von Willebrand/metabolismo , Proteínas ADAM/genética , Proteínas ADAM/metabolismo , Proteína ADAMTS13 , Motivos de Aminoácidos , Estudos de Casos e Controles , Desintegrinas/genética , Desintegrinas/metabolismo , Família , Feminino , Expressão Gênica , Genótipo , Humanos , Masculino , Fenótipo , Estrutura Terciária de Proteína , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
17.
Acta Neuropathol ; 132(1): 149-51, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27067307
18.
Genes Chromosomes Cancer ; 49(2): 176-81, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19902524

RESUMO

The SMARCB1 gene status in 50 patients with atypical teratoid rhabdoid tumor and/or malignant rhabdoid tumor recruited to a German registry was prospectively analyzed with FISH and PCR. Altogether we found 40 SMARCB1 mutations in 28 patients. Two patients were positive for SMARCB1 staining at immunochemistry. Germline mutations were identified in 10 of 41 patients with CNS disease, including three large heterozygous deletions, six truncating mutations and one donor splice site mutation. No missense mutation was identified. Analysis of first degree relatives did not detect any carriers. Mutations were distributed over the SMARCB1-gene without particular clustering. No germline mutation was found in nine patients without CNS disease. Patients with germline mutation had a lower median age at diagnosis in comparison to those without detectable germline mutation (5.5 vs. 13 months, P = 0.001), a higher rate of primary multicentric CNS disease (5/10 vs. 5/36) and synchronous or metachronous mixed CNS and extracranial disease (4/10 vs. 1/36). Two year overall survival was 0% in patients with germline mutation and 48% in those without detectable germline mutation (P < 0.001). Patients with germline mutation of SMARCB1 manifest at an early age and have a very high risk for progression which has to be considered with respect to the outcome of further treatment studies.


Assuntos
Proteínas Cromossômicas não Histona/genética , Proteínas de Ligação a DNA/genética , Mutação , Tumor Rabdoide/genética , Fatores de Transcrição/genética , Éxons/genética , Família , Feminino , Mutação em Linhagem Germinativa , Humanos , Imuno-Histoquímica , Hibridização in Situ Fluorescente , Leucócitos/patologia , Masculino , Reação em Cadeia da Polimerase , Tumor Rabdoide/classificação , Proteína SMARCB1 , Deleção de Sequência
19.
Clin Appl Thromb Hemost ; 27: 10760296211021171, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34184557

RESUMO

Left atrial (LA) thrombus formation is the presumed origin of thromboembolic complications in patients with atrial fibrillation (AF). Beyond clinical risk factors, the factors causing formation of LA thrombi are not well known. In this case-control study, we analyzed clinical characteristics and genetic thrombophilia markers (factor V Leiden (FVL), prothrombin G20210A (FIIV), Tyr2561 variant of von Willebrand factor (VWF-V)) in 42 patients with AF and LA thrombus (LAT) and in 68 control patients with AF without LAT (CTR). Patients with LAT had more clinical conditions predisposing to stroke (mean CHA2DS2-VASc-score 3.4 ± 1.5 vs. 1.9 ± 1.4; P < 0.001), a higher LA volume (96 ± 32 vs. 76 ± 21 ml, P = 0.002) and lower LA appendage emptying velocity (0.21 ± 0.11vs. 0.43 ± 0.19 m/s, P < 0.001). Prevalence of FVL, FIIV and VWF-V mutations was not different, but in the subgroup of patients <65 years (y) there was a tendency for a higher incidence of VWF-V with a prevalence of 27% (LAT <65 y) vs. 7% (CTR <65 y, P = 0.066). These findings warrant further investigation of the VWF-V as a risk factor for LA thrombogenesis in younger patients.


Assuntos
Fibrilação Atrial/complicações , Ecocardiografia Transesofagiana/métodos , Trombose/genética , Idoso , Estudos de Casos e Controles , Humanos , Medição de Risco
20.
Am J Surg Pathol ; 45(9): 1228-1234, 2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-33739782

RESUMO

Atypical teratoid/rhabdoid tumor (AT/RT) is a highly malignant central nervous system tumor predominantly occurring in infants that may also arise in older children and adults. Rare secondary AT/RT developing from other tumors such as pleomorphic xanthoastrocytoma (PXA) are on record, but AT/RT presenting with molecular features of PXA have not been described. Here, we report 3 malignant central nervous system tumors in children (10, 13, and 18 y old). All tumors were located in the temporal lobe. In 2 cases, there was no history of a low-grade precursor lesion; in 1 case anaplastic PXA had been diagnosed 3 months earlier. Histopathologically, all tumors were composed of RT cells and showed frank signs of malignancy as well as loss of nuclear SMARCB1/INI1 protein expression. Two cases displayed homozygous deletions of the SMARCB1 region while the third case showed an exon 7 mutation (c.849_850delGT; p.Met283Ilefs*77). Of note, DNA methylation profiles did not group with AT/RT or other tumor entities using the Heidelberg Brain Tumor Classifier (version v11b4). By unsupervised t-distributed stochastic neighbor embedding analysis and hierarchical clustering analysis, however, all tumors clearly grouped with PXA. Genome-wide copy number analysis revealed homozygous CDNK2A/B deletions and gains of whole chromosome 7. BRAF V600E mutations could be demonstrated in all cases. In conclusion, the possibility of AT/RT with molecular features of PXA needs to be taken into account and warrants molecular characterization of AT/RT especially in older children. Since treatments targeting mutated BRAF are available, identification of such cases may also have therapeutic consequences.


Assuntos
Astrocitoma/genética , Neoplasias Encefálicas/genética , Tumor Rabdoide/genética , Teratoma/genética , Adolescente , Criança , Feminino , Humanos , Mutação , Proteínas Proto-Oncogênicas B-raf/genética , Proteína SMARCB1/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA