Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
2.
Nat Chem Biol ; 9(12): 840-848, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24161946

RESUMO

Efforts to develop more effective therapies for acute leukemia may benefit from high-throughput screening systems that reflect the complex physiology of the disease, including leukemia stem cells (LSCs) and supportive interactions with the bone marrow microenvironment. The therapeutic targeting of LSCs is challenging because LSCs are highly similar to normal hematopoietic stem and progenitor cells (HSPCs) and are protected by stromal cells in vivo. We screened 14,718 compounds in a leukemia-stroma co-culture system for inhibition of cobblestone formation, a cellular behavior associated with stem-cell function. Among those compounds that inhibited malignant cells but spared HSPCs was the cholesterol-lowering drug lovastatin. Lovastatin showed anti-LSC activity in vitro and in an in vivo bone marrow transplantation model. Mechanistic studies demonstrated that the effect was on target, via inhibition of HMG-CoA reductase. These results illustrate the power of merging physiologically relevant models with high-throughput screening.


Assuntos
Antineoplásicos/farmacologia , Ensaios de Seleção de Medicamentos Antitumorais/métodos , Leucemia , Células-Tronco Neoplásicas/efeitos dos fármacos , Linhagem Celular Tumoral , Células-Tronco Hematopoéticas , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Lovastatina/farmacologia , Células-Tronco Neoplásicas/citologia , Células-Tronco Neoplásicas/fisiologia
3.
Blood ; 119(22): 5211-4, 2012 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-22510873

RESUMO

Oncogenic activating mutations in NOTCH1 occur in more than 50% of T-cell acute lymphoblastic leukemias (T-ALLs). In the present study, we describe a novel mechanism of NOTCH1 activation in T-ALL in which a deletion removing the 5' portion of NOTCH1 abolishes the negative regulatory control of the extracellular domain and leads to constitutively active NOTCH1 signaling. Polypeptides translated from truncated transcripts encoded by the NOTCH1 deletion allele retain the transmembrane domain of the receptor and are constitutively cleaved by the γ-secretase complex, resulting in high levels of NOTCH1 signaling that can be effectively blocked by γ-secretase inhibitors. Our results expand the spectrum of oncogenic lesions activating NOTCH1 signaling in human T-ALL.


Assuntos
Alelos , Proteínas de Neoplasias/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Receptor Notch1/genética , Transdução de Sinais/genética , Adulto , Feminino , Humanos , Masculino , Proteínas de Neoplasias/metabolismo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Estrutura Terciária de Proteína , Receptor Notch1/metabolismo , Deleção de Sequência
4.
Curr Opin Hematol ; 20(4): 369-73, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23695450

RESUMO

PURPOSE OF REVIEW: Early T-cell precursor (ETP) leukaemias have been recently recognized as a form of T-cell acute lymphoblastic leukaemia (T-ALL) with a poor prognosis. The purpose of this review is to outline the most recent advances in the biology, genetics and prognostic significance of this aggressive disease. RECENT FINDINGS: Detailed immunophenotypic analyses have defined ETP T-ALLs as a distinct group of T-ALL with a poor prognosis. Transcriptionally, ETP T-ALLs and early immature T-ALLs, a broader group of tumours characterized by very early arrest in T-cell differentiation, are most related to haematopoietic stem cells and myeloid progenitors. Consistently, these leukaemias show lower frequencies of prototypical T-ALL lesions such as CDKN2A/B deletions and activating mutations in NOTCH1 and show a higher prevalence of mutations typically associated with the pathogenesis of acute myeloid leukaemias (AMLs). SUMMARY: ETP and early immature T-ALLs are characterized by a very early differentiation arrest and show unique genetic and transcriptional features that overlap both with T-ALL and with AML. Given the unique biology and poor prognosis associated with the ETP T-ALL group, there is an urgent need of new tailored therapeutic strategies for the treatment of this disease.


Assuntos
Leucemia-Linfoma Linfoblástico de Células T Precursoras , Biomarcadores Tumorais/metabolismo , Perfilação da Expressão Gênica , Humanos , Imunofenotipagem , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/imunologia , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Prognóstico , Linfócitos T/imunologia , Linfócitos T/metabolismo , Transcrição Gênica
5.
Blood Adv ; 6(9): 2835-2846, 2022 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-35081257

RESUMO

Bleeding in patients with acute myeloid leukemia (AML) receiving intensive induction chemotherapy is multifactorial and contributes to early death. We sought to define the incidence and risk factors of grade 4 bleeding to support strategies for risk mitigation. Bleeding events were retrospectively assessed between day-14 and day +60 of induction treatment according to the World Health Organization (WHO) bleeding assessment scale, which includes grade 4 bleeding as fatal, life-threatening, retinal with visual impairment, or involving the central nervous system. Predictors were considered pretreatment or prior to grade 4 bleeding. Using multivariable competing-risk regression analysis with grade 4 bleeding as the primary outcome, we identified risk factors in the development cohort (n = 341), which were tested in an independent cohort (n = 143). Grade 4 bleeding occurred in 5.9% and 9.8% of patients in the development and validation cohort, respectively. Risk factors that were independently associated with grade 4 bleeding included baseline platelet count ≤40 × 109/L compared with >40 × 109/L, and baseline international normalized ratio of prothrombin time (PT-INR) >1.5 or 1.3 > 1.5 compared with ≤1.3. These variables were allocated points, which allowed for stratification of patients with low- and high-risk for grade 4 bleeding. Cumulative incidence of grade 4 bleeding at day+60 was significantly higher among patients with high- vs low-risk (development: 31 ± 7% vs 2 ± 1%; P < .001; validation: 25 ± 9% vs 7 ± 2%; P = .008). In both cohorts, high bleeding risk was associated with disseminated intravascular coagulation (DIC) and proliferative disease. We developed and validated a simple risk model for grade 4 bleeding, which enables the development of rational risk mitigation strategies to improve early mortality of intensive induction treatment.


Assuntos
Coagulação Intravascular Disseminada , Leucemia Mieloide Aguda , Hemorragia/epidemiologia , Hemorragia/etiologia , Humanos , Quimioterapia de Indução/efeitos adversos , Leucemia Mieloide Aguda/complicações , Leucemia Mieloide Aguda/tratamento farmacológico , Estudos Retrospectivos
6.
Blood Adv ; 6(6): 1671-1683, 2022 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-35073571

RESUMO

Chronic lymphocytic leukemia (CLL), the most common leukemia worldwide, is associated with increased COVID-19 mortality. Previous studies suggest only a portion of vaccinated CLL patients develop severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike antibodies. Whether the elicited antibodies are functional and/or accompanied by functional T-cell responses is unknown. This prospective cohort study included patients with CLL who received SARS-CoV-2 and PCV13 vaccines (not concurrently). The primary cohort included adults with CLL off therapy. Coprimary outcomes were serologic response to SARS-CoV-2 (receptor binding domain [RBD] immunoassay) and PCV13 vaccines (23-serotype IgG assay). Characterization of SARS-CoV-2 antibodies and their functional activity and assessment of functional T-cell responses was performed. Sixty percent (18/30) of patients demonstrated serologic responses to SARS-CoV-2 vaccination, appearing more frequent among treatment-naïve patients (72%). Among treatment-naïve patients, an absolute lymphocyte count ≤24 000/µL was associated with serologic response (94% vs 14%; P < .001). On interferon-γ release assays, 80% (16/20) of patients had functional spike-specific T-cell responses, including 78% (7/9) with a negative RBD immunoassay, a group enriched for prior B-cell-depleting therapies. A bead-based multiplex immunoassay identified antibodies against wild-type and variant SARS-CoV-2 (α, ß, γ, and δ) in all tested patients and confirmed Fc-receptor binding and effector functions of these antibodies. Of 11 patients with negative RBD immunoassay after vaccination, 6 (55%) responded to an additional mRNA-based vaccine dose. The PCV13 serologic response rate was 29% (8/28). Our data demonstrate that SARS-CoV-2 vaccination induces functional T-cell and antibody responses in patients with CLL and provides the framework for investigating the molecular mechanisms and clinical benefit of these responses. This trial was registered at www.clinicaltrials.gov as #NCT05007860.


Assuntos
COVID-19 , Leucemia Linfocítica Crônica de Células B , Adulto , Anticorpos Neutralizantes , COVID-19/prevenção & controle , Vacinas contra COVID-19 , Humanos , Imunogenicidade da Vacina , Leucemia Linfocítica Crônica de Células B/terapia , Estudos Prospectivos , SARS-CoV-2
7.
JCI Insight ; 6(3)2021 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-33351783

RESUMO

The cohesin complex plays an essential role in chromosome maintenance and transcriptional regulation. Recurrent somatic mutations in the cohesin complex are frequent genetic drivers in cancer, including myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML). Here, using genetic dependency screens of stromal antigen 2-mutant (STAG2-mutant) AML, we identified DNA damage repair and replication as genetic dependencies in cohesin-mutant cells. We demonstrated increased levels of DNA damage and sensitivity of cohesin-mutant cells to poly(ADP-ribose) polymerase (PARP) inhibition. We developed a mouse model of MDS in which Stag2 mutations arose as clonal secondary lesions in the background of clonal hematopoiesis driven by tet methylcytosine dioxygenase 2 (Tet2) mutations and demonstrated selective depletion of cohesin-mutant cells with PARP inhibition in vivo. Finally, we demonstrated a shift from STAG2- to STAG1-containing cohesin complexes in cohesin-mutant cells, which was associated with longer DNA loop extrusion, more intermixing of chromatin compartments, and increased interaction with PARP and replication protein A complex. Our findings inform the biology and therapeutic opportunities for cohesin-mutant malignancies.


Assuntos
Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/metabolismo , Reparo do DNA/genética , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Mutação , Síndromes Mielodisplásicas/genética , Síndromes Mielodisplásicas/metabolismo , Animais , Linhagem Celular Tumoral , Cromatina/genética , Cromatina/metabolismo , Dano ao DNA , Modelos Animais de Doenças , Feminino , Humanos , Células K562 , Leucemia Mieloide Aguda/tratamento farmacológico , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos Mutantes , Camundongos SCID , Camundongos Transgênicos , Síndromes Mielodisplásicas/tratamento farmacológico , Proteínas Nucleares/genética , Ftalazinas/farmacologia , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Células U937 , Ensaios Antitumorais Modelo de Xenoenxerto , Coesinas
8.
Blood ; 112(8): 3373-82, 2008 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-18663146

RESUMO

To study the impact of oncogenic K-Ras on T-cell leukemia/lymphoma development and progression, we made use of a conditional K-Ras(G12D) murine knockin model, in which oncogenic K-Ras is expressed from its endogenous promoter. Transplantation of whole bone marrow cells that express oncogenic K-Ras into wild-type recipient mice resulted in a highly penetrant, aggressive T-cell leukemia/lymphoma. The lymphoblasts were composed of a CD4/CD8 double-positive population that aberrantly expressed CD44. Thymi of primary donor mice showed reduced cellularity, and immunophenotypic analysis demonstrated a block in differentiation at the double-negative 1 stage. With progression of disease, approximately 50% of mice acquired Notch1 mutations within the PEST domain. Of note, primary lymphoblasts were hypersensitive to gamma-secretase inhibitor treatment, which is known to impair Notch signaling. This inhibition was Notch-specific as assessed by down-regulation of Notch1 target genes and intracellular cleaved Notch. We also observed that the oncogenic K-Ras-induced T-cell disease was responsive to rapamycin and inhibitors of the RAS/MAPK pathway. These data indicate that patients with T-cell leukemia with K-Ras mutations may benefit from therapies that target the NOTCH pathway alone or in combination with inhibition of the PI3K/AKT/MTOR and RAS/MAPK pathways.


Assuntos
Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Regulação da Expressão Gênica , Genes ras , Leucemia de Células T/genética , Linfoma de Células T/genética , Receptor Notch1/genética , Proteínas ras/fisiologia , Animais , Células da Medula Óssea/citologia , Diferenciação Celular , Humanos , Receptores de Hialuronatos/biossíntese , Camundongos , Camundongos Transgênicos , Mutação
9.
Mol Cell Biol ; 26(24): 9232-43, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17030602

RESUMO

Following organ injury, morphogenic epithelial responses can vary depending on local cell density. In the present study, the role of cell confluence in determining the responsiveness of renal epithelial cells to the dedifferentiating morphogenic signals of hepatocyte growth factor (HGF) was examined. Increasing confluence resulted in a greater tendency of cells to organize into epithelial tubes and a significant decrease in migratory responsiveness to HGF. Analysis of downstream signaling revealed that the HGF receptor c-Met was equally activated in confluent and nonconfluent cells following HGF stimulation but that phosphoinositide 3-kinase-dependent activation of Akt and Rac were selectively diminished in confluent cells. In nonconfluent cells treated with HGF, the high level of Akt activation resulted in inhibitory phosphorylation of glycogen synthase kinase 3beta (GSK-3beta) and increased beta-catenin nuclear signaling. In contrast, confluent cells, in which HGF-stimulated Akt activation was diminished, displayed less inhibitory phosphorylation of GSK-3beta and less nuclear signaling by beta-catenin. Overexpression of beta-catenin (SA), which cannot be phosphorylated by GSK-3beta and targeted for ubiquitination, significantly increased migration in fully confluent cells. Thus, cells maintained at high confluence selectively downregulate signaling events such as Rac activation and beta-catenin-dependent transcription that would otherwise promote cell dedifferentiation and migration.


Assuntos
Diferenciação Celular/fisiologia , Movimento Celular/fisiologia , Células Epiteliais/citologia , Fator de Crescimento de Hepatócito/fisiologia , beta Catenina/fisiologia , Animais , Contagem de Células , Técnicas de Cultura de Células , Proliferação de Células , Células Cultivadas , Regulação para Baixo/fisiologia , Fator de Crescimento de Hepatócito/antagonistas & inibidores , Túbulos Renais Coletores/citologia , Túbulos Renais Coletores/metabolismo , Camundongos , Transdução de Sinais/fisiologia , beta Catenina/antagonistas & inibidores
10.
Nat Med ; 21(10): 1182-9, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26390244

RESUMO

Activating mutations in NOTCH1 are common in T cell acute lymphoblastic leukemia (T-ALL). Here we identify glutaminolysis as a critical pathway for leukemia cell growth downstream of NOTCH1 and a key determinant of the response to anti-NOTCH1 therapies in vivo. Mechanistically, inhibition of NOTCH1 signaling in T-ALL induces a metabolic shutdown, with prominent inhibition of glutaminolysis and triggers autophagy as a salvage pathway supporting leukemia cell metabolism. Consequently, inhibition of glutaminolysis and inhibition of autophagy strongly and synergistically enhance the antileukemic effects of anti-NOTCH1 therapy in mice harboring T-ALL. Moreover, we demonstrate that Pten loss upregulates glycolysis and consequently rescues leukemic cell metabolism, thereby abrogating the antileukemic effects of NOTCH1 inhibition. Overall, these results identify glutaminolysis as a major node in cancer metabolism controlled by NOTCH1 and as therapeutic target for the treatment of T-ALL.


Assuntos
Antineoplásicos/farmacologia , Resistencia a Medicamentos Antineoplásicos , Leucemia-Linfoma Linfoblástico de Células T Precursoras/tratamento farmacológico , Receptor Notch1/antagonistas & inibidores , Animais , Glutamina/metabolismo , Camundongos , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo
11.
Nat Genet ; 46(2): 166-70, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24413734

RESUMO

Peripheral T cell lymphomas (PTCLs) are a heterogeneous and poorly understood group of non-Hodgkin lymphomas. Here we combined whole-exome sequencing of 12 tumor-normal DNA pairs, RNA sequencing analysis and targeted deep sequencing to identify new genetic alterations in PTCL transformation. These analyses identified highly recurrent epigenetic factor mutations in TET2, DNMT3A and IDH2 as well as a new highly prevalent RHOA mutation encoding a p.Gly17Val alteration present in 22 of 35 (67%) angioimmunoblastic T cell lymphoma (AITL) samples and in 8 of 44 (18%) PTCL, not otherwise specified (PTCL-NOS) samples. Mechanistically, the RHOA Gly17Val protein interferes with RHOA signaling in biochemical and cellular assays, an effect potentially mediated by the sequestration of activated guanine-exchange factor (GEF) proteins. In addition, we describe new and recurrent, albeit less frequent, genetic defects including mutations in FYN, ATM, B2M and CD58 implicating SRC signaling, impaired DNA damage response and escape from immune surveillance mechanisms in the pathogenesis of PTCL.


Assuntos
Epigênese Genética/genética , Linfoma de Células T Periférico/genética , Proteínas Proto-Oncogênicas c-fyn/genética , Proteína rhoA de Ligação ao GTP/genética , Proteínas Mutadas de Ataxia Telangiectasia/genética , Sequência de Bases , Antígenos CD58/genética , Biologia Computacional , DNA (Citosina-5-)-Metiltransferases/genética , DNA Metiltransferase 3A , Proteínas de Ligação a DNA/genética , Dioxigenases , Escherichia coli , Exoma/genética , Imunofluorescência , Células HEK293 , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Isocitrato Desidrogenase/genética , Dados de Sequência Molecular , Mutação de Sentido Incorreto/genética , Proteínas Proto-Oncogênicas/genética , Análise de Sequência de RNA
12.
J Exp Med ; 208(13): 2571-9, 2011 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-22162831

RESUMO

Early immature T cell acute lymphoblastic leukemias (T-ALLs) account for ~5-10% of pediatric T-ALLs and are associated with poor prognosis. However, the genetic defects that drive the biology of these tumors remain largely unknown. In this study, analysis of microarray gene expression signatures in adult T-ALL demonstrated a high prevalence of early immature leukemias and revealed a close relationship between these tumors and myeloid leukemias. Many adult immature T-ALLs harbored mutations in myeloid-specific oncogenes and tumor suppressors including IDH1, IDH2, DNMT3A, FLT3, and NRAS. Moreover, we identified ETV6 mutations as a novel genetic lesion uniquely present in immature adult T-ALL. Our results demonstrate that early immature adult T-ALL represents a heterogeneous category of leukemias characterized by the presence of overlapping myeloid and T-ALL characteristics, and highlight the potential role of ETV6 mutations in these tumors.


Assuntos
Mutação , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Proteínas Proto-Oncogênicas c-ets/genética , Proteínas Proto-Oncogênicas c-ets/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Adulto , Linhagem Celular Tumoral , Feminino , Perfilação da Expressão Gênica , Regulação Leucêmica da Expressão Gênica/genética , Genes Supressores de Tumor , Humanos , Lactente , Masculino , Análise de Sequência com Séries de Oligonucleotídeos , Variante 6 da Proteína do Fator de Translocação ETS
13.
Cancer Cell ; 17(6): 584-96, 2010 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-20541703

RESUMO

We report a Jak2V617F knockin mouse myeloproliferative neoplasm (MPN) model resembling human polycythemia vera (PV). The MPN is serially transplantable and we demonstrate that the hematopoietic stem cell (HSC) compartment has the unique capacity for disease initiation but does not have a significant selective competitive advantage over wild-type HSCs. In contrast, myeloid progenitor populations are expanded and skewed toward the erythroid lineage, but cannot transplant the disease. Treatment with a JAK2 kinase inhibitor ameliorated the MPN phenotype, but did not eliminate the disease-initiating population. These findings provide insights into the consequences of JAK2 activation on HSC differentiation and function and have the potential to inform therapeutic approaches to JAK2V617F-positive MPN.


Assuntos
Substituição de Aminoácidos , Células-Tronco Hematopoéticas/patologia , Janus Quinase 2/genética , Janus Quinase 2/metabolismo , Transtornos Mieloproliferativos/genética , Transtornos Mieloproliferativos/patologia , Animais , Antígenos CD/metabolismo , Medula Óssea/patologia , Células da Medula Óssea/efeitos dos fármacos , Transplante de Medula Óssea , Contagem de Células , Diferenciação Celular/genética , Modelos Animais de Doenças , Células Precursoras Eritroides/metabolismo , Células Precursoras Eritroides/patologia , Eritropoetina/farmacologia , Expressão Gênica/genética , Perfilação da Expressão Gênica , Hematócrito , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/metabolismo , Heterozigoto , Humanos , Janus Quinase 2/antagonistas & inibidores , Células Progenitoras de Megacariócitos/metabolismo , Células Progenitoras de Megacariócitos/patologia , Células Progenitoras de Megacariócitos e Eritrócitos/efeitos dos fármacos , Células Progenitoras de Megacariócitos e Eritrócitos/metabolismo , Células Progenitoras de Megacariócitos e Eritrócitos/patologia , Células Progenitoras de Megacariócitos e Eritrócitos/transplante , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células Progenitoras Mieloides/metabolismo , Células Progenitoras Mieloides/patologia , Transtornos Mieloproliferativos/tratamento farmacológico , Policitemia Vera/genética , Policitemia Vera/patologia , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Pirrolidinas/farmacologia , Pirrolidinas/uso terapêutico , Baço/efeitos dos fármacos , Baço/patologia , Sulfonamidas/farmacologia , Sulfonamidas/uso terapêutico , Análise de Sobrevida
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA