Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
J Cell Mol Med ; 25(18): 9060-9065, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34402163

RESUMO

BCL2-associated athanogene-1 (BAG1) is a multi-functional protein that is found deregulated in several solid cancers and in paediatric acute myeloid leukaemia. The investigation of BAG1 isoforms expression and intracellular localization in B-cell acute lymphoblastic leukaemia (B-ALL) patient-derived specimens revealed that BAG1 levels decrease during disease remission, compared to diagnosis, but drastically increase at relapse. In particular, at diagnosis both BAG1-L and BAG1-M isoforms are mainly nuclear, while during remission the localization pattern changes, having BAG1-M almost exclusively in the cytosol indicating its potential cytoprotective role in B-ALL. In addition, knockdown of BAG1/BAG3 induces cell apoptosis and G1-phase cell cycle arrest and, more intriguingly, shapes cell response to chemotherapy. BAG1-depleted cells show an increased sensitivity to the common chemotherapeutic agents, dexamethasone or daunorubicin, and to the BCL2 inhibitor ABT-737. Moreover, the BAG1 inhibitor Thio-2 induces a cytotoxic effect on RS4;11 cells both in vitro and in a zebrafish xenograft model and strongly synergizes with pan-BCL inhibitors. Collectively, these data sustain BAG1 deregulation as a critical event in assuring survival advantage to B-ALL cells.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Reguladoras de Apoptose/metabolismo , Proteínas de Ligação a DNA/metabolismo , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Leucemia-Linfoma Linfoblástico de Células Precursoras , Fatores de Transcrição/metabolismo , Antineoplásicos , Humanos , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Cultura Primária de Células , Células Tumorais Cultivadas
2.
Oncogene ; 40(4): 746-762, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33247204

RESUMO

Leukemias are routinely sub-typed for risk/outcome prediction and therapy choice using acquired mutations and chromosomal rearrangements. Down syndrome acute lymphoblastic leukemia (DS-ALL) is characterized by high frequency of CRLF2-rearrangements, JAK2-mutations, or RAS-pathway mutations. Intriguingly, JAK2 and RAS-mutations are mutually exclusive in leukemic sub-clones, causing dichotomy in therapeutic target choices. We prove in a cell model that elevated CRLF2 in combination with constitutionally active JAK2 is sufficient to activate wtRAS. On primary clinical DS-ALL samples, we show that wtRAS-activation is an obligatory consequence of mutated/hyperphosphorylated JAK2. We further prove that CRLF2-ligand TSLP boosts the direct binding of active PTPN11 to wtRAS, providing the molecular mechanism for the wtRAS activation. Pre-inhibition of RAS or PTPN11, but not of PI3K or JAK-signaling, prevented TSLP-induced RAS-GTP boost. Cytotoxicity assays on primary clinical DS-ALL samples demonstrated that, regardless of mutation status, high-risk leukemic cells could only be killed using RAS-inhibitor or PTPN11-inhibitor, but not PI3K/JAK-inhibitors, suggesting a unified treatment target for up to 80% of DS-ALL. Importantly, protein activities-based principal-component-analysis multivariate clusters analyzed for independent outcome prediction using Cox proportional-hazards model showed that protein-activity (but not mutation-status) was independently predictive of outcome, demanding a paradigm-shift in patient-stratification strategy for precision therapy in high-risk ALL.


Assuntos
Mutação , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Proteínas ras/fisiologia , Animais , Citocinas/fisiologia , Humanos , Janus Quinase 2/genética , Janus Quinase 2/fisiologia , Camundongos , Fosfatidilinositol 3-Quinases/fisiologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Proteína Tirosina Fosfatase não Receptora Tipo 11/fisiologia , Receptores de Citocinas/genética , Transdução de Sinais/fisiologia , Serina-Treonina Quinases TOR/fisiologia , Proteínas ras/antagonistas & inibidores , Proteínas ras/genética
4.
Br J Haematol ; 180(5): 680-693, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29359790

RESUMO

Relapse remains the leading cause of treatment failure in children with acute lymphoblastic leukaemia (ALL) undergoing allogeneic haematopoietic stem cell transplantation (HSCT). We retrospectively investigated the prognostic role of minimal residual disease (MRD) before and after HSCT in 119 children transplanted in complete remission (CR). MRD was measured by polymerase chain reaction in bone marrow samples collected pre-HSCT and during the first and third trimesters after HSCT (post-HSCT1 and post-HSCT3). The overall event-free survival (EFS) was 50%. The cumulative incidence of relapse and non-relapse mortality was 41% and 9%. Any degree of detectable pre-HSCT MRD was associated with poor outcome: EFS was 39% and 18% in patients with MRD positivity <1 × 10-3 and ≥1 × 10-3 , respectively, versus 73% in MRD-negative patients (P < 0·001). This effect was maintained in different disease remissions, but low-level MRD had a very strong negative impact only in patients transplanted in second or further CR. Also, MRD after HSCT enabled patients to be stratified, with increasing MRD between post-HSCT1 and post-HSCT3 clearly defining cohorts with a different outcome. MRD is an important prognostic factor both before and after transplantation. Given that MRD persistence after HSCT is associated with dismal outcome, these patients could benefit from early discontinuation of immunosuppression, or pre-emptive immuno-therapy.


Assuntos
Transplante de Células-Tronco Hematopoéticas/métodos , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Adolescente , Criança , Pré-Escolar , Intervalo Livre de Doença , Feminino , Transplante de Células-Tronco Hematopoéticas/mortalidade , Humanos , Lactente , Masculino , Recidiva Local de Neoplasia/etiologia , Neoplasia Residual , Reação em Cadeia da Polimerase , Leucemia-Linfoma Linfoblástico de Células Precursoras/mortalidade , Análise de Sobrevida , Transplante Homólogo , Resultado do Tratamento
5.
Nat Commun ; 8(1): 593, 2017 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-28928446

RESUMO

Group 2 innate lymphoid cells (ILC2s) are involved in human diseases, such as allergy, atopic dermatitis and nasal polyposis, but their function in human cancer remains unclear. Here we show that, in acute promyelocytic leukaemia (APL), ILC2s are increased and hyper-activated through the interaction of CRTH2 and NKp30 with elevated tumour-derived PGD2 and B7H6, respectively. ILC2s, in turn, activate monocytic myeloid-derived suppressor cells (M-MDSCs) via IL-13 secretion. Upon treating APL with all-trans retinoic acid and achieving complete remission, the levels of PGD2, NKp30, ILC2s, IL-13 and M-MDSCs are restored. Similarly, disruption of this tumour immunosuppressive axis by specifically blocking PGD2, IL-13 and NKp30 partially restores ILC2 and M-MDSC levels and results in increased survival. Thus, using APL as a model, we uncover a tolerogenic pathway that may represent a relevant immunosuppressive, therapeutic targetable, mechanism operating in various human tumour types, as supported by our observations in prostate cancer.Group 2 innate lymphoid cells (ILC2s) modulate inflammatory and allergic responses, but their function in cancer immunity is still unclear. Here the authors show that, in acute promyelocytic leukaemia, tumour-activated ILC2s secrete IL-13 to induce myeloid-derived suppressor cells and support tumour growth.


Assuntos
Antígenos B7/imunologia , Linfócitos/imunologia , Células Supressoras Mieloides/imunologia , Receptor 3 Desencadeador da Citotoxicidade Natural/imunologia , Prostaglandina D2/imunologia , Células A549 , Animais , Antineoplásicos/uso terapêutico , Antígenos B7/metabolismo , Linhagem Celular Tumoral , Modelos Animais de Doenças , Células HL-60 , Células Hep G2 , Humanos , Imunidade Inata/imunologia , Interleucina-13/imunologia , Interleucina-13/metabolismo , Leucemia Promielocítica Aguda/tratamento farmacológico , Leucemia Promielocítica Aguda/imunologia , Leucemia Promielocítica Aguda/metabolismo , Linfócitos/metabolismo , Camundongos Endogâmicos C57BL , Células Supressoras Mieloides/metabolismo , Receptor 3 Desencadeador da Citotoxicidade Natural/metabolismo , Prostaglandina D2/metabolismo , Ligação Proteica , Tretinoína/uso terapêutico
6.
Oncotarget ; 8(26): 42398-42413, 2017 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-28415578

RESUMO

ERG-related leukemia is a B cell precursor acute lymphoblastic leukemia (BCP ALL) subtype characterized by aberrant expression of DUX4 and ERG transcription factors, and highly recurrent ERG intragenic deletions. ERG-related patients have remarkably favorable outcome despite a high incidence of inauspicious IKZF1 aberrations.We describe clinical and genomic features of the ERG-related cases in an unselected cohort of B-other BCP ALL pediatric patients enrolled in the AIEOP ALL 2000 therapeutic protocol. We report a small noncoding RNA signature specific of ERG-related group, with up-regulation of miR-125b-2 cluster on chromosome 21 and several snoRNAs in the Prader-Willi locus at 15q11.2, including the orphan SNORD116 cluster.


Assuntos
Expressão Gênica , MicroRNAs/genética , Família Multigênica , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/metabolismo , RNA Nucleolar Pequeno/genética , Regulador Transcricional ERG/metabolismo , Adolescente , Antineoplásicos/uso terapêutico , Biomarcadores , Criança , Pré-Escolar , Aberrações Cromossômicas , Cromossomos Humanos Par 15 , Cromossomos Humanos Par 21/genética , Estudos de Coortes , Feminino , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Lactente , Masculino , Síndrome de Prader-Willi/genética , Síndrome de Prader-Willi/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras B/tratamento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patologia , Locos de Características Quantitativas , Deleção de Sequência , Regulador Transcricional ERG/genética , Transcriptoma , Resultado do Tratamento
7.
Sci Rep ; 6: 34449, 2016 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-27698462

RESUMO

To induce and sustain the leukaemogenic process, MLL-AF4+ leukaemia seems to require very few genetic alterations in addition to the fusion gene itself. Studies of infant and paediatric patients with MLL-AF4+ B cell precursor acute lymphoblastic leukaemia (BCP-ALL) have reported mutations in KRAS and NRAS with incidences ranging from 25 to 50%. Whereas previous studies employed Sanger sequencing, here we used next generation amplicon deep sequencing for in depth evaluation of RAS mutations in 36 paediatric patients at diagnosis of MLL-AF4+ leukaemia. RAS mutations including those in small sub-clones were detected in 63.9% of patients. Furthermore, the mutational analysis of 17 paired samples at diagnosis and relapse revealed complex RAS clone dynamics and showed that the mutated clones present at relapse were almost all originated from clones that were already detectable at diagnosis and survived to the initial therapy. Finally, we showed that mutated patients were indeed characterized by a RAS related signature at both transcriptional and protein levels and that the targeting of the RAS pathway could be of beneficial for treatment of MLL-AF4+ BCP-ALL clones carrying somatic RAS mutations.


Assuntos
GTP Fosfo-Hidrolases/genética , Sequenciamento de Nucleotídeos em Larga Escala , Leucemia/genética , Proteínas de Membrana/genética , Mutação , Proteína de Leucina Linfoide-Mieloide , Proteínas de Fusão Oncogênica , Proteínas Proto-Oncogênicas p21(ras)/genética , Feminino , GTP Fosfo-Hidrolases/metabolismo , Humanos , Leucemia/metabolismo , Leucemia/patologia , Masculino , Proteínas de Membrana/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/metabolismo
8.
Nat Commun ; 5: 4654, 2014 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-25105841

RESUMO

Children with Down syndrome (DS) and acute lymphoblastic leukaemia (ALL) have poorer survival and more relapses than non-DS children with ALL, highlighting an urgent need for deeper mechanistic understanding of DS-ALL. Here, using full-exome or cancer genes-targeted sequencing of 42 ALL samples from 39 DS patients, we uncover driver mutations in RAS, (KRAS and NRAS) recurring to a similar extent (15/42) as JAK2 (12/42) mutations or P2RY8-CRLF2 fusions (14/42). RAS mutations are almost completely mutually exclusive with JAK2 mutations (P=0.016), driving a combined total of two-thirds of analysed cases. Clonal architecture analysis reveals that both RAS and JAK2 drove sub-clonal expansions primarily initiated by CRLF2 rearrangements, and/or mutations in chromatin remodellers and lymphocyte differentiation factors. Remarkably, in 2/3 relapsed cases, there is a switch from a primary JAK2- or PTPN11-mutated sub-clone to a RAS-mutated sub-clone in relapse. These results provide important new insights informing the patient stratification strategies for targeted therapeutic approaches for DS-ALL.


Assuntos
Síndrome de Down/genética , Janus Quinase 2/genética , Mutação , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Proteínas ras/metabolismo , Criança , Cromatina/química , Análise Mutacional de DNA , Síndrome de Down/metabolismo , Feminino , Deleção de Genes , Regulação Leucêmica da Expressão Gênica , Humanos , Linfócitos/citologia , Masculino , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Receptores de Citocinas/genética
9.
Blood ; 122(4): 554-61, 2013 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-23733339

RESUMO

Some neonates with Down syndrome (DS) are diagnosed with self-regressing transient myeloproliferative disorder (TMD), and 20% to 30% of those progress to acute megakaryoblastic leukemia (AMKL). We performed exome sequencing in 7 TMD/AMKL cases and copy-number analysis in these and 10 additional cases. All TMD/AMKL samples contained GATA1 mutations. No exome-sequenced TMD/AMKL sample had other recurrently mutated genes. However, 2 of 5 TMD cases, and all AMKL cases, showed mutations/deletions other than GATA1, in genes proven as transformation drivers in non-DS leukemia (EZH2, APC, FLT3, JAK1, PARK2-PACRG, EXT1, DLEC1, and SMC3). One patient at the TMD stage revealed 2 clonal expansions with different GATA1 mutations, of which 1 clone had an additional driver mutation. Interestingly, it was the other clone that gave rise to AMKL after accumulating mutations in 7 other genes. Data suggest that GATA1 mutations alone are sufficient for clonal expansions, and additional driver mutations at the TMD stage do not necessarily predict AMKL progression. Later in infancy, leukemic progression requires "third-hit driver" mutations/somatic copy-number alterations found in non-DS leukemias. Putative driver mutations affecting WNT (wingless-related integration site), JAK-STAT (Janus kinase/signal transducer and activator of transcription), or MAPK/PI3K (mitogen-activated kinase/phosphatidylinositol-3 kinase) pathways were found in all cases, aberrant activation of which converges on overexpression of MYC.


Assuntos
Transformação Celular Neoplásica/genética , Síndrome de Down/genética , Leucemia Megacarioblástica Aguda/genética , Transtornos Mieloproliferativos/genética , Progressão da Doença , Síndrome de Down/complicações , Exoma/genética , Predisposição Genética para Doença/genética , Estudo de Associação Genômica Ampla , Instabilidade Genômica/genética , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Humanos , Lactente , Recém-Nascido , Leucemia Megacarioblástica Aguda/complicações , Leucemia Megacarioblástica Aguda/patologia , Análise em Microsséries , Transtornos Mieloproliferativos/complicações , Transtornos Mieloproliferativos/patologia , Polimorfismo de Nucleotídeo Único , Transcriptoma
10.
Haematologica ; 98(4): 602-10, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23100280

RESUMO

MicroRNA-34b down-regulation in acute myeloid leukemia was previously shown to induce CREB overexpression, thereby causing leukemia proliferation in vitro and in vivo. The role of microRNA-34b and CREB in patients with myeloid malignancies has never been evaluated. We examined microRNA-34b expression and the methylation status of its promoter in cells from patients diagnosed with myeloid malignancies. We used gene expression profiling to identify signatures of myeloid transformation. We established that microRNA-34b has suppressor ability and that CREB has oncogenic potential in primary bone marrow cell cultures and in vivo. MicroRNA-34b was found to be up-regulated in pediatric patients with juvenile myelomonocytic leukemia (n=17) and myelodysplastic syndromes (n=28), but was down-regulated in acute myeloid leukemia patients at diagnosis (n=112). Our results showed that hypermethylation of the microRNA-34b promoter occurred in 66% of cases of acute myeloid leukemia explaining the low microRNA-34b levels and CREB overexpression, whereas preleukemic myelodysplastic syndromes and juvenile myelomonocytic leukemia were not associated with hypermethylation or CREB overexpression. In paired samples taken from the same patients when they had myelodysplastic syndrome and again during the subsequent acute myeloid leukemia, we confirmed microRNA-34b promoter hypermethylation at leukemia onset, with 103 CREB target genes differentially expressed between the two disease stages. This subset of CREB targets was confirmed to associate with high-risk myelodysplastic syndromes in a separate cohort of patients (n=20). Seventy-eight of these 103 CREB targets were also differentially expressed between healthy samples (n=11) and de novo acute myeloid leukemia (n=72). Further, low microRNA-34b and high CREB expression levels induced aberrant myelopoiesis through CREB-dependent pathways in vitro and in vivo. In conclusion, we suggest that microRNA-34b controls CREB expression and contributes to myeloid transformation from both healthy bone marrow and myelodysplastic syndromes. We identified a subset of CREB target genes that represents a novel transcriptional network that may control myeloid transformation.


Assuntos
Metilação de DNA , Regulação Leucêmica da Expressão Gênica , MicroRNAs/genética , Células Mieloides/metabolismo , Regiões Promotoras Genéticas/genética , Doença Aguda , Adolescente , Animais , Transformação Celular Neoplásica/genética , Células Cultivadas , Criança , Pré-Escolar , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Perfilação da Expressão Gênica , Células HL-60 , Humanos , Lactente , Recém-Nascido , Subunidade gama Comum de Receptores de Interleucina/deficiência , Subunidade gama Comum de Receptores de Interleucina/genética , Leucemia Mieloide/genética , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Síndromes Mielodisplásicas/genética , Células Mieloides/patologia
11.
Pharmacogenomics ; 13(16): 1905-16, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23215883

RESUMO

AIM: In the AIEOP-BFM 2000 trial, 15% of pediatric patients treated according to risk-adapted polychemotherapeutic regimens relapsed. The present study aimed to investigate the influence of GST-M1 and GST-T1 deletions on clinical outcome of children with acute lymphoblastic leukemia treated according to the AIEOP-BFM ALL 2000 study protocol. MATERIALS & METHODS: A novel-design, two-phase study was applied to select a subsample of 614 children to be genotyped for the deletions of GST genes. Cumulative incidence of relapse was then estimated by weighted Kaplan-Meier analysis, and the Cox model was applied to evaluate the effect of GST-M1 and GST-T1 isoenzyme deletions on relapse. RESULTS: No overall effect was found, but the GST-M1 deletion was associated with better clinical outcome within prednisone poor-responder patients (hazard ratio [HR]: 0.45; 95% CI: 0.23-0.91; p = 0.026), whereas the GST-T1 deletion was associated with worse outcome in the standard-risk group (HR: 4.62; 95% CI: 1.04-20.6; p = 0.045) and within prednisone good responders (HR: 1.62; 95% CI: 1.02-2.58; p = 0.041). CONCLUSION: Our results show that GST-M1 and GST-T1 homozygous deletions have opposite correlation with relapse, the former being protective and the latter unfavourable in specific subsets of acute lymphoblastic leukemia patients.


Assuntos
Deleção de Genes , Glutationa Transferase/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras , Resultado do Tratamento , Criança , Pré-Escolar , Feminino , Genótipo , Glutationa Transferase/metabolismo , Homozigoto , Humanos , Lactente , Itália , Estimativa de Kaplan-Meier , Masculino , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/patologia , Prognóstico , Modelos de Riscos Proporcionais , Recidiva
12.
Br J Haematol ; 157(2): 197-200, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22221250

RESUMO

Children with Down syndrome have a 20- to 50-fold increased risk of acute lymphocytic or myeloid leukaemia. Whole or partial gains of chromosome 21 have been described in multiple childhood leukaemias, and have recently been reported as a likely primary event in B-precursor-acute lymphoblastic leukaemia. It is unclear which amplified gene(s) on chromosome 21 play a key role in leukaemia progression. We describe a minimal amplified segment within the so-called 'Down syndrome critical region' shared between two cases of AML-M0; a Down syndrome, and a constitutionally normal individual. Interestingly, the amplified region does not include the oncogenes RUNX1, ETS2 and ERG.


Assuntos
Subunidade alfa 2 de Fator de Ligação ao Core/genética , Síndrome de Down/genética , Loci Gênicos , Leucemia Mieloide Aguda/genética , Proteína Proto-Oncogênica c-ets-2/genética , Transativadores/genética , Adolescente , Adulto , Feminino , Humanos , Masculino , Regulador Transcricional ERG
15.
Oncol Rep ; 18(2): 417-23, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17611665

RESUMO

Hypermethylation of CpG islands is the most well defined epigenetic change in neoplasia and plays an important role in the inactivation or silencing of cancer related genes. DLX genes (1-7), with large CpG islands in their 5' region, are implicated in a number of processes among which haematopoiesis. They are characterized by highly dynamic spatio-temporal expression and supposed to be involved in resistance to apoptosis of several tumor cell lines. In acute lymphoblastic leukemia (ALL) hypermethylation is a common phenomenon frequently associated with poor prognosis in specific genetic childhood leukemia subgroups. These data together with the presence of large CpG islands in the up-stream regions of the DLX genes make them attractive candidates for methylation regulated gene expression and leukemia related aberrancies. To validate the role of DLX genes in paediatric B-ALL cells, we studied two cell lines and two groups of patients with paediatric chromosomal rearrangements: MLL-AF4 and TEL-AML1, respectively. Analysis of methylation and gene expression patterns of DLX3 in 64 specimens of B-lineage ALL revealed that DLX3 presents aberrant methylation in paediatric B-ALL patients. In vitro experiments with 5-Aza-2'dC on leukemia cell lines, confirmed by Western blot analysis, indicated that the methylation of DLX3 CpG islands has a functional role and interferes with the DLX3 gene and DLX3 protein expression in B-ALL cells. Importantly, hypermethylation of DLX3 significantly reduces its expression in MLL-AF4 rearranged leukemias while methylation is almost absent in TEL-AML1 positive ALL specimens. These results show that differential DLX3 methylation could be a new epigenetic marker for genotypic B-cell leukemia subgroup with high-risk features.


Assuntos
Linfoma de Burkitt/patologia , Metilação de DNA , Proteínas de Homeodomínio/genética , Proteína de Leucina Linfoide-Mieloide/genética , Proteínas de Fusão Oncogênica/genética , Regiões Promotoras Genéticas/genética , Fatores de Transcrição/genética , Western Blotting , Linfoma de Burkitt/genética , Linfoma de Burkitt/metabolismo , Linhagem Celular Tumoral , Criança , Regulação para Baixo , Regulação Neoplásica da Expressão Gênica , Células HL-60 , Proteínas de Homeodomínio/metabolismo , Humanos , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Fatores de Transcrição/metabolismo , Células U937
16.
Blood ; 106(4): 1419-22, 2005 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-15870172

RESUMO

Nucleophosmin (NPM) is a nucleocytoplasmic shuttling protein involved in leukemia-associated chromosomal translocations, and it regulates the alternate reading frame (ARF)-p53 tumor-suppressor pathway. Recently, it has been demonstrated that mutations of the NPM1 gene alter the protein at its C-terminal, causing its cytoplasmic localization. Cytoplasmic NPM was detected in 35% of adult patients with primary non-French-American-British (FAB) classification M3 acute myeloid leukemia (AML), associated mainly with normal karyotype. We evaluated the prevalence of the NPM1 gene mutation in non-M3 childhood AML patients enrolled in the ongoing Associazione Italiana di Ematologia e Oncologia Pediatrica (AIEOP-AML02) protocol in Italy. NPM1 mutations were found in 7 (6.5%) of 107 successfully analyzed patients. NPM1-mutated patients carried a normal karyotype (7/26, 27.1%) and were older in age. Thus, the NPM1 mutation is a frequent abnormality in AML patients without known genetic marker; the mutation may represent a new target to monitor minimal residual disease in AML and a potential candidate for alternative and targeted treatments.


Assuntos
Leucemia Mieloide Aguda/genética , Mutação , Proteínas Nucleares/genética , Adolescente , Fatores Etários , Sequência de Bases , Criança , Pré-Escolar , Citoplasma/química , Análise Mutacional de DNA , Éxons , Feminino , Humanos , Cariotipagem , Masculino , Dados de Sequência Molecular , Nucleofosmina
17.
Br J Haematol ; 129(3): 333-9, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15842656

RESUMO

Somatic mutations in PTPN11, the gene encoding the transducer SHP-2, have emerged as a novel class of lesions that upregulate RAS signalling and contribute to leukaemogenesis. In a recent study of 69 children and adolescents with de novo acute myeloid leukaemia (AML), we documented a non-random distribution of PTPN11 mutations among French-American-British (FAB) subtypes. Lesions were restricted to FAB-M5 cases, where they were relatively common (four of 12 cases). Here, we report on the results of a molecular screening performed on 181 additional unselected patients, enrolled in participating institutions of the Associazione Italiana Ematologia Oncologia Pediatrica-AML Study Group, to provide a more accurate picture of the prevalence, spectrum and distribution of PTPN11 mutations in childhood AML and to investigate their clinical relevance. We concluded that PTPN11 defects do not represent a frequent event in this heterogeneous group of malignancies (4.4%), although they recur in a considerable percentage of patients with FAB-M5 (18%). PTPN11 lesions rarely occur in other subtypes. Within the FAB-M5 group no clear association of PTPN11 mutations with any clinical variable was evident. Nearly two third of the patients with this subtype were found to harbour an activating mutation in PTPN11, NRAS, KRAS2 or FLT3.


Assuntos
Leucemia Mieloide/genética , Mutação , Proteínas de Neoplasias/genética , Proteínas Tirosina Fosfatases/genética , Doença Aguda , Adolescente , Criança , Pré-Escolar , Feminino , Genes ras/genética , Humanos , Lactente , Peptídeos e Proteínas de Sinalização Intracelular , Leucemia Monocítica Aguda/sangue , Leucemia Monocítica Aguda/genética , Leucemia Mieloide/sangue , Contagem de Leucócitos , Masculino , Proteína Tirosina Fosfatase não Receptora Tipo 11 , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas p21(ras) , Receptores Proteína Tirosina Quinases/genética , Tirosina Quinase 3 Semelhante a fms , Proteínas ras
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...