Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Nat Genet ; 24(3): 266-70, 2000 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10700180

RESUMO

Haematopoietic development is regulated by nuclear protein complexes that coordinate lineage-specific patterns of gene expression. Targeted mutagenesis in embryonic stem cells and mice has revealed roles for the X-linked gene Gata1 in erythrocyte and megakaryocyte differentiation. GATA-1 is the founding member of a family of DNA-binding proteins that recognize the motif WGATAR through a conserved multifunctional domain consisting of two C4-type zinc fingers. Here we describe a family with X-linked dyserythropoietic anaemia and thrombocytopenia due to a substitution of methionine for valine at amino acid 205 of GATA-1. This highly conserved valine is necessary for interaction of the amino-terminal zinc finger of GATA-1 with its essential cofactor, FOG-1 (for friend of GATA-1; refs 9-12). We show that the V205M mutation abrogates the interaction between Gata-1 and Fog-1, inhibiting the ability of Gata-1 to rescue erythroid differentiation in an erythroid cell line deficient for Gata-1 (G1E). Our findings underscore the importance of FOG-1:Gata-1 associations in both megakaryocyte and erythroid development, and suggest that other X-linked anaemias or thrombocytopenias may be caused by defects in GATA1.


Assuntos
Anemia Diseritropoética Congênita/genética , Criptorquidismo/genética , Proteínas de Ligação a DNA/genética , Mutação Puntual , Trombocitopenia/genética , Fatores de Transcrição/genética , Cromossomo X/genética , Adulto , Sequência de Aminoácidos , Substituição de Aminoácidos , Criança , Sequência Consenso , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/fisiologia , Fatores de Ligação de DNA Eritroide Específicos , Feminino , Fator de Transcrição GATA1 , Hematopoese/genética , Humanos , Recém-Nascido , Masculino , Modelos Moleculares , Dados de Sequência Molecular , Linhagem , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/genética , Trombocitopenia/congênito , Fatores de Transcrição/química , Fatores de Transcrição/deficiência , Fatores de Transcrição/fisiologia , Dedos de Zinco/genética
2.
bioRxiv ; 2023 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-38106192

RESUMO

Chromothripsis, the process of catastrophic shattering and haphazard repair of chromosomes, is a common event in cancer. Whether chromothripsis might constitute an actionable molecular event amenable to therapeutic targeting remains an open question. We describe recurrent chromothripsis of chromosome 21 in a subset of patients in blast phase of a myeloproliferative neoplasm (BP-MPN), which alongside other structural variants leads to amplification of a region of chromosome 21 in ∼25% of patients ('chr21amp'). We report that chr21amp BP-MPN has a particularly aggressive and treatment-resistant phenotype. The chr21amp event is highly clonal and present throughout the hematopoietic hierarchy. DYRK1A , a serine threonine kinase and transcription factor, is the only gene in the 2.7Mb minimally amplified region which showed both increased expression and chromatin accessibility compared to non-chr21amp BP-MPN controls. We demonstrate that DYRK1A is a central node at the nexus of multiple cellular functions critical for BP-MPN development, including DNA repair, STAT signalling and BCL2 overexpression. DYRK1A is essential for BP-MPN cell proliferation in vitro and in vivo , and DYRK1A inhibition synergises with BCL2 targeting to induce BP-MPN cell apoptosis. Collectively, these findings define the chr21amp event as a prognostic biomarker in BP-MPN and link chromothripsis to a druggable target.

3.
Science ; 265(5180): 1866-9, 1994 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-8091213

RESUMO

Individual small nuclear ribonucleoproteins (snRNPs) U1, U2, and U4/U6 were removed from nuclear extracts of HeLa cells by antisense affinity depletion. Addition of a highly purified preparation of SR proteins fully restored splicing activity in reactions depleted of U1 snRNP but did not reconstitute splicing in reactions depleted of the other snRNPs. Affinity selection experiments revealed that spliceosomes lacking U1 snRNA formed in the U1 snRNP-depleted reactions reconstituted with SR proteins. Thus, high concentrations of SR proteins facilitate the assembly of precursor messenger RNA (pre-mRNA) into a spliceosome in the absence of interactions with U1 snRNP.


Assuntos
Precursores de RNA/genética , Splicing de RNA , Proteínas de Ligação a RNA/metabolismo , Ribonucleoproteína Nuclear Pequena U1/metabolismo , Sequência de Bases , Globinas/genética , Dados de Sequência Molecular , RNA Nuclear Pequeno/genética , RNA Nuclear Pequeno/metabolismo , Ribonucleoproteína Nuclear Pequena U2/metabolismo , Spliceossomos/metabolismo
4.
Oncogene ; 26(47): 6803-15, 2007 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-17934487

RESUMO

Red blood cells and megakaryocytes arise from a common precursor, the megakaryocyte-erythroid progenitor and share many regulators including the transcription factors GATA-1 and GFI-1B and signaling molecules such as JAK2 and STAT5. These lineages also share the distinction of being associated with rare, but aggressive malignancies that have very poor prognoses. In this review, we will briefly summarize features of normal development of red blood cells and megakaryocytes and also highlight events that lead to their leukemic transformation. It is clear that much more work needs to be done to improve our understanding of the unique biology of these leukemias and to pave the way for novel targeted therapeutics.


Assuntos
Transformação Celular Neoplásica/patologia , Células Eritroides/citologia , Células Eritroides/patologia , Megacariócitos/citologia , Megacariócitos/patologia , Animais , Transformação Celular Neoplásica/genética , Humanos , Leucemia/genética , Leucemia/patologia
5.
Leukemia ; 20(7): 1261-9, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16673016

RESUMO

Chromosome condensation is essential for proper segregation of duplicated sister chromatids in mitosis. Mammalian erythroid maturation is also associated with gradual nuclear condensation. However, few proteins that are directly involved in chromosome condensation during erythropoiesis have been identified. In this report, we show that MTB (more than blood), which was initially isolated in a yeast two-hybrid screen for proteins that interact with the basic helix-loop-helix (bHLH) protein stem cell leukemia (SCL), and later identified as the murine homolog of the condensin II subunit CAP-G2, participates in erythroid cell development. MTB interacts with SCL and another hematopoietic bHLH protein, E12, and is recruited to the nucleus by SCL and E12. In addition, MTB can repress SCL/E12-mediated transcriptional activation. Consistent with the model that MTB may function together with SCL/E12 heterodimer during erythroid cell development, MTB is highly expressed in the erythroid lineage and is upregulated upon erythroid differentiation. Moreover, overexpression of MTB promotes the terminal differentiation of the murine erythroleukemia erythroid cell line. Together, these findings demonstrate that the condensin II subunit MTB/mCAP-G2 plays a novel function during erythropoiesis and suggest that key hematopoietic transcription factors such as SCL and E12 may regulate the terminal differentiation of hematopoietic cells through the interaction with condensin complexes.


Assuntos
Células Eritroides/citologia , Hematopoese/fisiologia , Leucemia Eritroblástica Aguda/fisiopatologia , Proteínas/genética , Transcrição Gênica/fisiologia , Adenosina Trifosfatases/genética , Sequência de Aminoácidos , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Células COS , Proteínas de Ciclo Celular , Diferenciação Celular/fisiologia , Linhagem da Célula/fisiologia , Núcleo Celular/metabolismo , Chlorocebus aethiops , Proteínas de Ligação a DNA/genética , Leucemia/metabolismo , Leucemia/fisiopatologia , Leucemia Eritroblástica Aguda/metabolismo , Camundongos , Dados de Sequência Molecular , Complexos Multiproteicos/genética , Proteínas/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-kit/genética , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Homologia de Sequência de Aminoácidos , Proteína 1 de Leucemia Linfocítica Aguda de Células T , Fatores de Transcrição TCF/metabolismo , Proteína 1 Semelhante ao Fator 7 de Transcrição , Técnicas do Sistema de Duplo-Híbrido
6.
Leukemia ; 31(11): 2458-2467, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28270692

RESUMO

Imetelstat (GRN163L) is a specific telomerase inhibitor that has demonstrated clinical activity in patients with myeloproliferative neoplasms (MPN) and in patients with solid tumors. The antitumor effects were associated with the development of thrombocytopenia, one of the common side effects observed in patients treated with imetelstat. The events underlying these adverse effects are not apparent. In this report, we investigated the potential mechanisms that account for imetelstat's beneficial effects in MPN patients and the manner by which imetelstat treatment leads to a reduction in platelet numbers. Using a well-established system of ex vivo megakaryopoiesis, we demonstrated that imetelestat treatment affects normal megakaryocyte (MK) development by exclusively delaying maturation of MK precursor cells. By contrast, additional stages along MPN MK development were affected by imetelstat resulting in reduced numbers of assayable colony-forming unit MK and impaired MK maturation. In addition, treatment with imetelstat inhibited the secretion of fibrogenic growth factors by malignant but not by normal MK. Our results indicate that the delay observed in normal MK maturation may account for imetelstat-induced thrombocytopenia, while the more global effects of imetelstat on several stages along the hierarchy of MPN megakaryopoiesis may be responsible for the favorable clinical outcomes reported in MPN patients.


Assuntos
Inibidores Enzimáticos/farmacologia , Indóis/farmacologia , Megacariócitos/efeitos dos fármacos , Niacinamida/análogos & derivados , Telomerase/antagonistas & inibidores , Humanos , Megacariócitos/citologia , Niacinamida/farmacologia , Oligonucleotídeos , Poliploidia
7.
Blood Cancer J ; 7(6): e572, 2017 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-28622305

RESUMO

Myelofibrosis (MF) is characterized by hyperactivation of thrombopoietin (TPO) signaling, which induces a RPS14 deficiency that de-regulates GATA1 in megakaryocytes by hampering its mRNA translation. As mice carrying the hypomorphic Gata1low mutation, which reduces the levels of Gata1 mRNA in megakaryocytes, develop MF, we investigated whether the TPO axis is hyperactive in this model. Gata1low mice contained two times more Tpo mRNA in liver and TPO in plasma than wild-type littermates. Furthermore, Gata1low LSKs expressed levels of Mpl mRNA (five times greater than normal) and protein (two times lower than normal) similar to those expressed by LSKs from TPO-treated wild-type mice. Gata1low marrow and spleen contained more JAK2/STAT5 than wild-type tissues, an indication that these organs were reach of TPO-responsive cells. Moreover, treatment of Gata1low mice with the JAK inhibitor ruxolitinib reduced their splenomegaly. Also in Gata1low mice activation of the TPO/MPL axis was associated with a RSP14 deficiency and a discordant microarray ribosome signature (reduced RPS24, RPS26 and SBDS expression). Finally, electron microscopy revealed that Gata1low megakaryocytes contained poorly developed endoplasmic reticulum with rare polysomes. In summary, Gata1low mice are a bona fide model of MF, which recapitulates the hyperactivation of the TPO/MPL/JAK2 axis observed in megakaryocytes from myelofibrotic patients.


Assuntos
Fator de Transcrição GATA1/metabolismo , Mielofibrose Primária/genética , Proteínas Ribossômicas/genética , Trombopoetina/metabolismo , Animais , Modelos Animais de Doenças , Feminino , Fator de Transcrição GATA1/genética , Humanos , Masculino , Camundongos , Mielofibrose Primária/patologia
8.
Leukemia ; 31(3): 712-719, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27554164

RESUMO

Acute myelogenous leukemia (AML) is a high-risk hematopoietic malignancy caused by a variety of mutations, including genes encoding the cohesin complex. Recent studies have demonstrated that reduction in cohesin complex levels leads to enhanced self-renewal in hematopoietic stem and progenitors (HSPCs). We sought to delineate the molecular mechanisms by which cohesin mutations promote enhanced HSPC self-renewal as this represents a critical initial step during leukemic transformation. We verified that RNAi against the cohesin subunit Rad21 causes enhanced self-renewal of HSPCs in vitro through derepression of polycomb repressive complex 2 (PRC2) target genes, including Hoxa7 and Hoxa9. Importantly, knockdown of either Hoxa7 or Hoxa9 suppressed self-renewal, implying that both are critical downstream effectors of reduced cohesin levels. We further demonstrate that the cohesin and PRC2 complexes interact and are bound in close proximity to Hoxa7 and Hoxa9. Rad21 depletion resulted in decreased levels of H3K27me3 at the Hoxa7 and Hoxa9 promoters, consistent with Rad21 being critical to proper gene silencing by recruiting the PRC2 complex. Our data demonstrates that the cohesin complex regulates PRC2 targeting to silence Hoxa7 and Hoxa9 and negatively regulate self-renewal. Our studies identify a novel epigenetic mechanism underlying leukemogenesis in AML patients with cohesin mutations.


Assuntos
Autorrenovação Celular/genética , Repressão Epigenética , Regulação da Expressão Gênica , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Neoplasias/genética , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Aneuploidia , Animais , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proliferação de Células , Proteínas Cromossômicas não Histona/química , Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/metabolismo , Análise por Conglomerados , Proteínas de Ligação a DNA , Deleção de Genes , Perfilação da Expressão Gênica , Histonas/metabolismo , Proteínas de Homeodomínio/metabolismo , Camundongos , Modelos Biológicos , Família Multigênica , Complexos Multiproteicos/metabolismo , Células Mieloides/citologia , Células Mieloides/metabolismo , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas/genética , Coesinas
9.
Leukemia ; 30(9): 1816-23, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27285583

RESUMO

Children with Down syndrome (DS) are at a 20-fold increased risk for acute lymphoblastic leukemia (DS-ALL). Although the etiology of this higher risk of developing leukemia remains largely unclear, the recent identification of CRLF2 (cytokine receptor like factor 2) and JAK2 mutations and study of the effect of trisomy of Hmgn1 and Dyrk1a (dual-specificity tyrosine phosphorylation-regulated kinase 1A) on B-cell development have shed significant new light on the disease process. Here we focus on the clinical features, biology and genetics of ALL in children with DS. We review the unique characteristics of DS-ALL on both the clinical and molecular levels and discuss the differences in treatments and outcomes in ALL in children with DS compared with those without DS. The identification of new biological insights is expected to pave the way for novel targeted therapies.


Assuntos
Síndrome de Down/complicações , Leucemia-Linfoma Linfoblástico de Células Precursoras/patologia , Linfócitos B , Criança , Humanos , Terapia de Alvo Molecular/métodos , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/etiologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Resultado do Tratamento
10.
Blood Cancer J ; 6(9): e478, 2016 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-27689744

RESUMO

FAXDC2 (fatty acid hydroxylase domain containing 2) is a member of the fatty acid hydroxylase superfamily. Given the important role of fatty acids in megakaryocytes, we have studied the role of this gene in the development of this lineage. Here we show that the expression of FAXDC2 is constantly elevated during megakaryocyte maturation. In contrast, FAXDC2 is significantly downregulated in acute myeloid leukemia and acute megakaryoblastic leukemia. Moreover, FAXDC2 overexpression promotes the differentiation of megakaryocytic cell lines and primary cells, whereas its knockdown disrupts their maturation. Mechanism study shows that FAXDC2 overexpression enhances extracellular signal-regulated kinase (ERK) signaling and increases RUNX1 (Runt-related transcription factor 1) expression. FAXDC2 also restores megakaryocytic differentiation in cells exposed to an ERK inhibitor or those expressing a dominant negative form of RUNX1. Finally, FAXDC2 overexpression leads to an increase in sphingolipid GM3 synthase, suggesting a potential role of FAXDC2 in lipid metabolism that increases ERK signaling and facilitates megakaryocyte differentiation. Together, these results show that FAXDC2 plays a novel role in development of megakaryocytes and its dysregulation may contribute to abnormal hematopoietic cell development in leukemia.

11.
Oncogene ; 34(5): 537-45, 2015 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-24632603

RESUMO

The Aurora kinases, which include Aurora A (AURKA), Aurora B (AURKB) and Aurora C (AURKC), are serine/threonine kinases required for the control of mitosis (AURKA and AURKB) and meiosis (AURKC). Since their discovery nearly 20 years ago, Aurora kinases have been studied extensively in cell and cancer biology. Several early studies found that Aurora kinases are amplified and overexpressed at the transcript and protein level in various malignancies, including several types of leukemia. These discoveries and others provided a rationale for the development of small-molecule inhibitors of Aurora kinases as leukemia therapies. The first generation of Aurora kinase inhibitors did not fare well in clinical trials, owing to poor efficacy and high toxicity. However, the creation of second-generation, highly selective Aurora kinase inhibitors has increased the enthusiasm for targeting these proteins in leukemia. This review will describe the functions of each Aurora kinase, summarize their involvement in leukemia and discuss inhibitor development and efficacy in leukemia clinical trials.


Assuntos
Aurora Quinase A/genética , Aurora Quinase B/genética , Aurora Quinase C/genética , Leucemia/genética , Aurora Quinase A/antagonistas & inibidores , Aurora Quinase B/antagonistas & inibidores , Aurora Quinase C/antagonistas & inibidores , Ciclo Celular/genética , Ensaios Clínicos como Assunto , Humanos , Leucemia/tratamento farmacológico , Leucemia/patologia , Meiose/genética , Mitose/genética , Bibliotecas de Moléculas Pequenas/uso terapêutico
12.
Cell Death Dis ; 5: e988, 2014 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-24407241

RESUMO

GATA1 mutations are tightly associated with transient myeloproliferative disorder (TMD) and acute megakaryoblstic leukemia (AMKL) in children with Down syndrome. Numerous genes are altered in GATA-1-deficient megakaryocytes, which may contribute to the hyperproliferation and abnormal terminal differentiation of these malignant cells. In this study, we demonstrate that Pstpip2 is a GATA-1-repressed gene that controls megakaryopoiesis. Ectopic expression of PSTPIP2 impaired megakaryocytic differentiation as evidenced by a decrease of CD41 expression and reduced DNA content in K562 cells. PSTPIP2 overexpression also caused enhanced activation of Src family kinases and subsequently reduced ERK phosphorylation. Consistently, PSTPIP2 knockdown showed the opposite effect on differentiation and signaling. Moreover, the W232A mutant of PSTPIP2, defective in its interaction with PEST family phosphatases that recruit c-Src terminal kinase (CSK) to suppress Src family kinases, failed to inhibit differentiation and lost its ability to enhance Src family kinases or reduce ERK phosphorylation. In fact, the W232A mutant of PSTPIP2 promoted megakaryocyte differentiation. These observations suggest that PSTPIP2 recruiting PEST phosphatases somehow blocked CSK activity and led to enhanced activation of Src family kinases and reduced ERK phosphorylation, which ultimately repressed megakaryocyte differentiation. Supporting this idea, PSTPIP2 interacted with LYN and the expression of a dominant negative LYN (LYN DN) overwhelmed the inhibitory effect of PSTPIP2 on differentiation and ERK signaling. In addition, a constitutively active LYN (LYN CA) normalized the enhanced megakaryocyte differentiation and repressed ERK signaling in PSTPIP2 knockdown cells. Finally, we found that PSTPIP2 repressed ERK signaling, differentiation, and proliferation and verified that PSTPIP2 upregulation repressed megakaryocyte development in primary mouse bone marrow cells. Our study thus reveals a novel mechanism by which dysregulation of PSTPIP2 due to GATA-1 deficiency may contribute to abnormal megakaryocyte proliferation and differentiation in pathogenesis of related diseases.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Diferenciação Celular , Proteínas do Citoesqueleto/genética , Regulação para Baixo , Fator de Transcrição GATA1/deficiência , Fator de Transcrição GATA1/metabolismo , Megacariócitos/citologia , Quinases da Família src/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Células Cultivadas , Proteínas do Citoesqueleto/metabolismo , Fator de Transcrição GATA1/genética , Regulação da Expressão Gênica , Humanos , Células K562 , Megacariócitos/enzimologia , Megacariócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação , Transdução de Sinais , Quinases da Família src/genética
13.
Leukemia ; 27(6): 1339-47, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23380710

RESUMO

The requirement that leukemic Gata1 mutations be present in cells harboring trisomy 21 led to the discovery that overexpression of ERG drives aberrant megakaryopoiesis. Given that constitutive PI3K/AKT signaling is a frequent component of hematologic malignancies and the relationship between AKT and Notch in this lineage, we studied the crosstalk between AKT signaling and ERG in megakaryopoiesis. We discovered that constitutive AKT signaling is associated with a dramatic increase in apoptosis of WT megakaryocytes (MKs), but that overexpression of ERG blocks AKT-induced death. We further found that Gata1 mutations protect MKs from activated AKT-induced apoptosis. As a consequence, however, the enhanced signaling inhibits differentiation of Gata1 mutant, but not WT, MKs. Gata1 mutant cells that overexpress ERG with hyperactive AKT are characterized by diminished FOXO1/3a expression and an increased dependency on the c-Jun pathway similar to that seen in acute megakaryoblastic leukemia (AMKL) cell lines, acute myeloid leukemia (AML) with knockdown of FOXO3a, or AML with expression of myristoylated Akt. Additionally, we found that the AKT allosteric inhibitor MK2206 caused reduced cell viability and proliferation of AMKL cell lines. The contribution of aberrant AKT signaling during the ontogeny of Down syndrome-transient myeloproliferative disorder/AMKL indicates that AKT is a therapeutic target in this form of AML.


Assuntos
Fator de Transcrição GATA1/metabolismo , Hematopoese , Leucemia Megacarioblástica Aguda/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transativadores/metabolismo , Linhagem Celular Tumoral , Fator de Transcrição GATA1/genética , Humanos , Leucemia Megacarioblástica Aguda/genética , Mutação , Fosforilação , Ligação Proteica , Regulador Transcricional ERG
14.
Leukemia ; 27(9): 1882-90, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23748344

RESUMO

The majority of patients with BCR-ABL1-negative myeloproliferative neoplasms (MPN) harbor mutations in JAK2 or MPL, which lead to constitutive activation of the JAK/STAT, PI3K and ERK signaling pathways. JAK inhibitors by themselves are inadequate in producing selective clonal suppression in MPN and are associated with hematopoietic toxicities. MK-2206 is a potent allosteric AKT inhibitor that was well tolerated, including no evidence of myelosuppression, in a phase I study of solid tumors. Herein, we show that inhibition of PI3K/AKT signaling by MK-2206 affected the growth of both JAK2V617F- or MPLW515L-expressing cells via reduced phosphorylation of AKT and inhibition of its downstream signaling molecules. Moreover, we demonstrate that MK-2206 synergizes with ruxolitinib in suppressing the growth of JAK2V617F-mutant SET2 cells. Importantly, MK-2206 suppressed colony formation from hematopoietic progenitor cells in patients with primary myelofibrosis and alleviated hepatosplenomegaly and reduced megakaryocyte burden in the bone marrows, livers and spleens of mice with MPLW515L-induced MPN. Together, these findings establish AKT as a rational therapeutic target in the MPNs.


Assuntos
Transtornos Mieloproliferativos/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Animais , Apoptose/efeitos dos fármacos , Medula Óssea/metabolismo , Medula Óssea/patologia , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Feminino , Proteínas de Fusão bcr-abl/deficiência , Compostos Heterocíclicos com 3 Anéis/farmacologia , Compostos Heterocíclicos com 3 Anéis/uso terapêutico , Humanos , Janus Quinase 2/antagonistas & inibidores , Janus Quinase 2/genética , Fígado/metabolismo , Fígado/patologia , Megacariócitos/efeitos dos fármacos , Megacariócitos/metabolismo , Camundongos , Mutação , Transtornos Mieloproliferativos/tratamento farmacológico , Transtornos Mieloproliferativos/genética , Transtornos Mieloproliferativos/patologia , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/uso terapêutico , Transdução de Sinais/efeitos dos fármacos , Baço/efeitos dos fármacos , Baço/metabolismo
15.
Blood Cancer J ; 1(11): e40, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22829077

RESUMO

Polycythemia vera (PV), essential thrombocythemia and primary myelofibrosis, are myeloproliferative neoplasms (MPNs) with distinct clinical features and are associated with the JAK2V617F mutation. To identify genomic anomalies involved in the pathogenesis of these disorders, we profiled 87 MPN patients using Affymetrix 250K single-nucleotide polymorphism (SNP) arrays. Aberrations affecting chr9 were the most frequently observed and included 9pLOH (n=16), trisomy 9 (n=6) and amplifications of 9p13.3-23.3 (n=1), 9q33.1-34.13 (n=1) and 9q34.13 (n=6). Patients with trisomy 9 were associated with elevated JAK2V617F mutant allele burden, suggesting that gain of chr9 represents an alternative mechanism for increasing JAK2V617F dosage. Gene expression profiling of patients with and without chr9 abnormalities (+9, 9pLOH), identified genes potentially involved in disease pathogenesis including JAK2, STAT5B and MAPK14. We also observed recurrent gains of 1p36.31-36.33 (n=6), 17q21.2-q21.31 (n=5) and 17q25.1-25.3 (n=5) and deletions affecting 18p11.31-11.32 (n=8). Combined SNP and gene expression analysis identified aberrations affecting components of a non-canonical PRC2 complex (EZH1, SUZ12 and JARID2) and genes comprising a 'HSC signature' (MLLT3, SMARCA2 and PBX1). We show that NFIB, which is amplified in 7/87 MPN patients and upregulated in PV CD34+ cells, protects cells from apoptosis induced by cytokine withdrawal.

16.
Blood Cancer J ; 1: e7, 2011 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-23471017

RESUMO

Immediately following the 2010 annual American Society of Hematology (ASH) meeting, the 5th International Post-ASH Symposium on Chronic Myelogenous Leukemia and BCR-ABL1-Negative Myeloproliferative Neoplasms (MPNs) took place on 7-8 December 2010 in Orlando, Florida, USA. During this meeting, the most recent advances in laboratory research and clinical practice, including those that were presented at the 2010 ASH meeting, were discussed among recognized authorities in the field. The current paper summarizes the proceedings of this meeting in BCR-ABL1-negative MPN. We provide a detailed overview of new mutations with putative epigenetic effects (TET oncogene family member 2 (TET2), additional sex comb-like 1 (ASXL1), isocitrate dehydrogenase (IDH) and enhancer of zeste homolog 2 (EZH2)) and an update on treatment with Janus kinase (JAK) inhibitors, pomalidomide, everolimus, interferon-α, midostaurin and cladribine. In addition, the new 'Dynamic International Prognostic Scoring System (DIPSS)-plus' prognostic model for primary myelofibrosis (PMF) and the clinical relevance of distinguishing essential thrombocythemia from prefibrotic PMF are discussed.

17.
Leukemia ; 24(11): 1920-6, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20882051

RESUMO

Survivin is an inhibitor of apoptosis protein family member that has an essential role in cellular proliferation as a component of the chromosome passenger complex. Survivin is highly expressed in embryos and in proliferating adult tissues, but it is not expressed in most differentiated cells. During tumorigenesis, however, survivin expression is dramatically upregulated. Although many studies have shown that survivin is required for cancer cells, the extent to which survivin contributes to the initiation of tumors is unknown. Here we show that transgenic mice that overexpress survivin in hematopoietic cells are at an increased risk of hematologic tumors. In examining how survivin might contribute to tumorigenesis, we observed that hematopoietic cells engineered to overexpress survivin are less susceptible to apoptosis. We conclude that survivin may promote tumorigenesis by imparting a survival advantage to cells that acquire additional genetic lesions.


Assuntos
Neoplasias Hematológicas/genética , Proteínas Associadas aos Microtúbulos/genética , Regulação para Cima , Animais , Apoptose/genética , Diferenciação Celular , Sobrevivência Celular , Citometria de Fluxo , Fator de Transcrição GATA1/genética , Regulação Neoplásica da Expressão Gênica , Predisposição Genética para Doença , Neoplasias Hematológicas/patologia , Células-Tronco Hematopoéticas/fisiologia , Humanos , Proteínas Inibidoras de Apoptose , Camundongos , Camundongos Endogâmicos BALB C , Proteínas de Neoplasias/genética , Regiões Promotoras Genéticas , Baço/imunologia , Survivina , Linfócitos T/imunologia
18.
Genes Dev ; 9(18): 2314-23, 1995 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-7557384

RESUMO

The full set of consensus sequences at the 5' splice site is recognized during splicing of pre-mRNA in extracts depleted of U1 snRNP. High concentrations of HeLa SR proteins or purified SC35 alone promote the splicing of specific RNA substrates, bypassing the requirement for U1 snRNP in formation of the U2 snRNP-pre-mRNA complex. Under these conditions, mutations in the substrate that increase the sequence complementarity between U6 snRNA and the 5' splice site region can facilitate splicing. This provides additional strong evidence that U1 snRNP is not essential for splicing. Thus, the consensus sequence at the 5' splice site is probably recognized twice during splicing of most introns; however, some pre-mRNAs could potentially be processed in the absence of interactions with U1 snRNP in regions of the nucleus containing high concentrations of SR protein.


Assuntos
Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Precursores de RNA/metabolismo , Splicing de RNA , RNA Mensageiro/biossíntese , RNA Nuclear Pequeno/metabolismo , Ribonucleoproteínas , Sequência de Bases , Sistema Livre de Células , Células HeLa , Humanos , Cinética , Modelos Genéticos , Dados de Sequência Molecular , Proteínas de Ligação a RNA , Ribonucleoproteína Nuclear Pequena U1/deficiência , Fatores de Processamento de Serina-Arginina
19.
Proc Natl Acad Sci U S A ; 94(1): 133-6, 1997 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-8990173

RESUMO

Assembly of the mammalian spliceosome is known to proceed in an ordered fashion through several discrete complexes, but the mechanism of this assembly process may not be universal. In an early step, pre-mRNAs are committed to the splicing pathway through association with U1 small nuclear ribonucleoprotein (snRNP) and non-snRNP splicing factors, including U2AF and members of the SR protein family. As a means of studying the steps of spliceosome assembly, we have prepared HeLa nuclear extracts specifically depleted of the splicing factor U2AF. Surprisingly, the SR protein SC35 can functionally substitute for U2AF65 in the reconstitution of pre-mRNA splicing in U2AF-depleted extracts. This reconstitution is substrate-specific and is reminiscent of the SC35-mediated reconstitution of splicing in extracts depleted of U1 snRNP. However, SC35 reconstitution of splicing in U2AF-depleted extracts is dependent on the presence of functional U1 snRNP. These observations suggest that there are at least three distinguishable mechanisms for the binding of U2 snRNP to the pre-mRNA, including U2AF-dependent and -independent pathways.


Assuntos
Núcleo Celular/metabolismo , Proteínas Nucleares/metabolismo , Splicing de RNA , Proteínas de Ligação a RNA/metabolismo , Ribonucleoproteínas/metabolismo , Spliceossomos/metabolismo , Células HeLa , Humanos , Modelos Genéticos , Precursores de RNA/metabolismo , RNA Mensageiro/metabolismo , Fatores de Processamento de Serina-Arginina , Fator de Processamento U2AF
20.
Mol Cell ; 3(2): 219-28, 1999 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10078204

RESUMO

GATA-1 and FOG (Friend of GATA-1) are each essential for erythroid and megakaryocyte development. FOG, a zinc finger protein, interacts with the amino (N) finger of GATA-1 and cooperates with GATA-1 to promote differentiation. To determine whether this interaction is critical for GATA-1 action, we selected GATA-1 mutants in yeast that fail to interact with FOG but retain normal DNA binding, as well a compensatory FOG mutant that restores interaction. These novel GATA-1 mutants do not promote erythroid differentiation of GATA-1- erythroid cells. Differentiation is rescued by the second-site FOG mutant. Thus, interaction of FOG with GATA-1 is essential for the function of GATA-1 in erythroid differentiation. These findings provide a paradigm for dissecting protein-protein associations involved in mammalian development.


Assuntos
Proteínas de Transporte/metabolismo , Proteínas de Ligação a DNA/metabolismo , Eritropoese/fisiologia , Proteínas Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Sequência de Aminoácidos , Animais , Proteínas de Transporte/genética , Diferenciação Celular , DNA/metabolismo , Proteínas de Ligação a DNA/genética , Células Precursoras Eritroides/metabolismo , Fatores de Ligação de DNA Eritroide Específicos , Fator de Transcrição GATA1 , Regulação da Expressão Gênica , Teste de Complementação Genética , Substâncias Macromoleculares , Camundongos , Dados de Sequência Molecular , Proteínas Nucleares/genética , Proteínas Recombinantes de Fusão/metabolismo , Saccharomyces cerevisiae/genética , Relação Estrutura-Atividade , Fatores de Transcrição/genética , Transfecção , Dedos de Zinco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA