Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Nat Commun ; 13(1): 2485, 2022 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-35585047

RESUMO

The utility of cancer whole genome and transcriptome sequencing (cWGTS) in oncology is increasingly recognized. However, implementation of cWGTS is challenged by the need to deliver results within clinically relevant timeframes, concerns about assay sensitivity, reporting and prioritization of findings. In a prospective research study we develop a workflow that reports comprehensive cWGTS results in 9 days. Comparison of cWGTS to diagnostic panel assays demonstrates the potential of cWGTS to capture all clinically reported mutations with comparable sensitivity in a single workflow. Benchmarking identifies a minimum of 80× as optimal depth for clinical WGS sequencing. Integration of germline, somatic DNA and RNA-seq data enable data-driven variant prioritization and reporting, with oncogenic findings reported in 54% more patients than standard of care. These results establish key technical considerations for the implementation of cWGTS as an integrated test in clinical oncology.


Assuntos
Perfilação da Expressão Gênica , Neoplasias , Criança , Estudos de Viabilidade , Perfilação da Expressão Gênica/métodos , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Humanos , Neoplasias/diagnóstico , Neoplasias/genética , Estudos Prospectivos , Transcriptoma/genética , Sequenciamento Completo do Genoma/métodos , Adulto Jovem
2.
Nat Med ; 6(12): 1335-40, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11100117

RESUMO

Chronic hypoxia, a hallmark of many tumors, is associated with angiogenesis and tumor progression. Strategies to treat tumors have been developed in which tumor cells are targeted with drugs or gene-therapy vectors specifically activated under hypoxic conditions. Here we report a different approach, in which the normal transcriptional response to hypoxia is selectively disrupted. Our data indicate that specific blockade of the interaction of hypoxia-inducible factor with the CH1 domain of its p300 and CREB binding protein transcriptional coactivators leads to attenuation of hypoxia-inducible gene expression and diminution of tumor growth. Thus, disrupting the normal co-activational response to hypoxia may be a new and useful therapeutic strategy.


Assuntos
Hipóxia Celular/genética , Proteínas de Ligação a DNA/metabolismo , Regulação Neoplásica da Expressão Gênica , Neoplasias Experimentais/terapia , Proteínas Nucleares/metabolismo , Transativadores/metabolismo , Fatores de Transcrição , Animais , Sítios de Ligação , Proteína de Ligação a CREB , Proteína p300 Associada a E1A , Terapia Genética/métodos , Fator 1 Induzível por Hipóxia , Subunidade alfa do Fator 1 Induzível por Hipóxia , Camundongos , Camundongos Nus , Neovascularização Patológica , Ligação Proteica , Transcrição Gênica
3.
Science ; 292(5523): 1907-10, 2001 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-11397944

RESUMO

How cytokines control differentiation of helper T (TH) cells is controversial. We show that T-bet, without apparent assistance from interleukin 12 (IL-12)/STAT4, specifies TH1 effector fate by targeting chromatin remodeling to individual interferon-gamma (IFN-gamma) alleles and by inducing IL-12 receptor beta2 expression. Subsequently, it appears that IL-12/STAT4 serves two essential functions in the development of TH1 cells: as growth signal, inducing survival and cell division; and as trans-activator, prolonging IFN-gamma synthesis through a genetic interaction with the coactivator, CREB-binding protein. These results suggest that a cytokine does not simply induce TH fate choice but instead may act as an essential secondary stimulus that mediates selective survival of a lineage.


Assuntos
Interferon gama/biossíntese , Interleucina-12/metabolismo , Células Th1/imunologia , Fatores de Transcrição/metabolismo , Alelos , Animais , Proteína de Ligação a CREB , Diferenciação Celular , Divisão Celular , Linhagem da Célula , Células Cultivadas , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica , Histonas/metabolismo , Interferon gama/genética , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Proteínas Nucleares/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Interleucina/metabolismo , Receptores de Interleucina-12 , Fator de Transcrição STAT4 , Transdução de Sinais , Proteínas com Domínio T , Células Th1/citologia , Células Th1/metabolismo , Transativadores/metabolismo , Fatores de Transcrição/genética
5.
Cancer Res ; 50(22): 7307-17, 1990 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-2121336

RESUMO

Although agents which act in a cell cycle phase specific manner are commonly used in the clinic and in basic research, it is as yet unclear why these agents are cytotoxic. In this paper, we examine the cellular events associated with the cytotoxicity of aphidicolin and vincristine in CHO strain AA8 cells. Cell killing resulting from aphidicolin treatment was found to require a period of inhibition-free growth following removal of the drug and was associated with characteristic aberrant mitotic processes. The cytotoxic effects of aphidicolin could be antagonized by the concomitant inhibition of protein synthesis with cycloheximide in the period of DNA synthesis inhibition. Cell killing resulting from treatment with vincristine was associated with the aberrant segregation of nuclear material and the formation of multiple partial nuclei. Vincristine cytotoxicity was found to be antagonized by concomitant administration of cycloheximide or cytochalasin D. These data support a hypothesis that the cytotoxic effects of cell cycle phase specific agents do not derive directly from their biochemical actions per se. We propose that cell death results from processes that are evoked by dissociation of normally integrated cell cycle events, and that dissociation involves replicative/mitotic events in the case of aphidicolin and karyokinetic/nuclear reformation events in the case of vincristine.


Assuntos
Ciclo Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Diterpenos/toxicidade , Vincristina/toxicidade , Animais , Afidicolina , Cricetinae , Cricetulus , Cicloeximida/farmacologia , Citocalasina D/farmacologia , Feminino , Técnicas In Vitro , Mitose/efeitos dos fármacos , Ovário , Fatores de Tempo
6.
Cancer Res ; 54(19): 5064-70, 1994 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-7923118

RESUMO

Chinese hamster ovary (and many rodent cell lines) transiently delay mitosis and progress into a second cell cycle without undergoing cytokinesis when treated with Colcemid, whereas HeLaS3 (and most human cell lines) arrest permanently in mitosis. We have discussed these differences and their consequences for cell survival under cell cycle-perturbing conditions within the context of mitotic checkpoint control (Schimke et al., Cold Spring Harbor Symp. Quant. Biol., 56: 417-425, 1991). Here, we report studies with mouse BALB/3T3 cell populations which, by the criterion of response to Colcemid, constitute a heterogeneous population with respect to mitotic checkpoint control. Clonal and subclonal populations retain population heterogeneity but with a bias for enrichment of cell populations that respond as do HeLaS3 cells. We have analyzed clones for their propensity for gene amplification as assessed by a stepwise increment selection protocol in methotrexate and report that there are significant differences in amplification propensities that correlate with differences in mitotic checkpoint control properties.


Assuntos
Amplificação de Genes , Mitose , Células 3T3 , Animais , Linhagem Celular Transformada , Resistência a Medicamentos , Metotrexato/farmacologia , Camundongos , Tetra-Hidrofolato Desidrogenase/genética
7.
Leukemia ; 30(1): 190-9, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26202935

RESUMO

Currently available combination chemotherapy for acute myeloid leukemia (AML) often fails to result in long-term remissions, emphasizing the need for novel therapeutic strategies. We reasoned that targeted inhibition of a prominent nuclear exporter, XPO1/CRM1, could eradicate self-renewing leukemia-initiating cells (LICs) whose survival depends on timely XPO1-mediated transport of specific protein and RNA cargoes. Using an immunosuppressed mouse model bearing primary patient-derived AML cells, we demonstrate that selinexor (KPT-330), an oral antagonist of XPO1 that is currently in clinical trials, has strong activity against primary AML cells while sparing normal stem and progenitor cells. Importantly, limiting dilution transplantation assays showed that this cytotoxic activity is not limited to the rapidly proliferating bulk population of leukemic cells but extends to the LICs, whose inherent drug resistance and unrestricted self-renewal capacity has been implicated in the difficulty of curing AML patients with conventional chemotherapy alone.


Assuntos
Hidrazinas/farmacologia , Carioferinas/antagonistas & inibidores , Leucemia Mieloide Aguda/tratamento farmacológico , Células-Tronco Neoplásicas/efeitos dos fármacos , Receptores Citoplasmáticos e Nucleares/antagonistas & inibidores , Triazóis/farmacologia , Animais , Humanos , Terapia de Imunossupressão , Leucemia Mieloide Aguda/patologia , Camundongos , Ensaios Antitumorais Modelo de Xenoenxerto , Proteína Exportina 1
8.
Leukemia ; 28(1): 155-65, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23588715

RESUMO

The key nuclear export protein CRM1/XPO1 may represent a promising novel therapeutic target in human multiple myeloma (MM). Here we showed that chromosome region maintenance 1 (CRM1) is highly expressed in patients with MM, plasma cell leukemia cells and increased in patient cells resistant to bortezomib treatment. CRM1 expression also correlates with increased lytic bone and shorter survival. Importantly, CRM1 knockdown inhibits MM cell viability. Novel, oral, irreversible selective inhibitors of nuclear export (SINEs) targeting CRM1 (KPT-185, KPT-330) induce cytotoxicity against MM cells (ED50<200 nM), alone and cocultured with bone marrow stromal cells (BMSCs) or osteoclasts (OC). SINEs trigger nuclear accumulation of multiple CRM1 cargo tumor suppressor proteins followed by growth arrest and apoptosis in MM cells. They further block c-myc, Mcl-1, and nuclear factor κB (NF-κB) activity. SINEs induce proteasome-dependent CRM1 protein degradation; concurrently, they upregulate CRM1, p53-targeted, apoptosis-related, anti-inflammatory and stress-related gene transcripts in MM cells. In SCID mice with diffuse human MM bone lesions, SINEs show strong anti-MM activity, inhibit MM-induced bone lysis and prolong survival. Moreover, SINEs directly impair osteoclastogenesis and bone resorption via blockade of RANKL-induced NF-κB and NFATc1, with minimal impact on osteoblasts and BMSCs. These results support clinical development of SINE CRM1 antagonists to improve patient outcome in MM.


Assuntos
Carioferinas/antagonistas & inibidores , Mieloma Múltiplo/terapia , Osteoclastos/patologia , Receptores Citoplasmáticos e Nucleares/antagonistas & inibidores , Humanos , Mieloma Múltiplo/patologia , Proteína Exportina 1
9.
Leukemia ; 27(1): 66-74, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22847027

RESUMO

Drugs that target the chief mediator of nuclear export, chromosome region maintenance 1 protein (CRM1) have potential as therapeutics for leukemia, but existing CRM1 inhibitors show variable potencies and a broad range of cytotoxic effects. Here, we report the structural analysis and antileukemic activity of a new generation of small-molecule inhibitors of CRM1. Designated selective inhibitors of nuclear export (SINE), these compounds were developed using molecular modeling to screen a small virtual library of compounds against the nuclear export signal (NES) groove of CRM1. The 2.2-Å crystal structure of the CRM1-Ran-RanBP1 complex bound to KPT-251, a representative molecule of this class of inhibitors, shows that the drug occupies part of the groove in CRM1 that is usually occupied by the NES, but penetrates much deeper into the groove and blocks CRM1-directed protein export. SINE inhibitors exhibit potent antileukemic activity, inducing apoptosis at nanomolar concentrations in a panel of 14 human acute myeloid leukemia (AML) cell lines representing different molecular subtypes of the disease. When administered orally to immunodeficient mice engrafted with human AML cells, KPT-251 had potent antileukemic activity with negligible toxicity to normal hematopoietic cells. Thus, KPT-SINE CRM1 antagonists represent a novel class of drugs that warrant further testing in AML patients.


Assuntos
Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Antineoplásicos/farmacologia , Carioferinas/química , Leucemia Mieloide Aguda/tratamento farmacológico , Sinais de Exportação Nuclear , Proteínas Nucleares/metabolismo , Receptores Citoplasmáticos e Nucleares/química , Proteína ran de Ligação ao GTP/metabolismo , Animais , Antineoplásicos/química , Apoptose , Western Blotting , Ciclo Celular , Núcleo Celular/metabolismo , Proliferação de Células , Células Cultivadas , Cristalização , Cristalografia por Raios X , Feminino , Células-Tronco Hematopoéticas , Humanos , Subunidade gama Comum de Receptores de Interleucina/fisiologia , Carioferinas/metabolismo , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Proteínas Nucleares/química , Ligação Proteica , Receptores Citoplasmáticos e Nucleares/metabolismo , Bibliotecas de Moléculas Pequenas , Ensaios Antitumorais Modelo de Xenoenxerto , Proteína ran de Ligação ao GTP/química , Proteína Exportina 1
10.
Leukemia ; 27(4): 852-60, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23235717

RESUMO

Mixed lineage leukemia (MLL)-fusion proteins can induce acute myeloid leukemias (AMLs) from either hematopoietic stem cells (HSCs) or granulocyte-macrophage progenitors (GMPs), but it remains unclear whether the cell of origin influences the biology of the resultant leukemia. MLL-AF9-transduced single HSCs or GMPs could be continuously replated, but HSC-derived clones were more likely than GMP-derived clones to initiate AML in mice. Leukemia stem cells derived from either HSCs or GMPs had a similar immunophenotype consistent with a maturing myeloid cell (LGMP). Gene expression analyses demonstrated that LGMP inherited gene expression programs from the cell of origin including high-level Evi-1 expression in HSC-derived LGMP. The gene expression signature of LGMP derived from HSCs was enriched in poor prognosis human MLL-rearranged AML in three independent data sets. Moreover, global 5'-mC levels were elevated in HSC-derived leukemias as compared with GMP-derived leukemias. This mirrored a difference seen in 5'-mC between MLL-rearranged human leukemias that are either EVI1 positive or EVI1 negative. Finally, HSC-derived leukemias were more resistant to chemotherapy than GMP-derived leukemias. These data demonstrate that the cell of origin influences the gene expression profile, the epigenetic state and the drug response in AML, and that these differences can account for clinical heterogeneity within a molecularly defined group of leukemias.


Assuntos
Leucemia Mieloide Aguda/genética , Proteína de Leucina Linfoide-Mieloide/genética , Adulto , Animais , Antineoplásicos/farmacologia , Citarabina/farmacologia , Perfilação da Expressão Gênica , Histona-Lisina N-Metiltransferase , Humanos , Camundongos , Camundongos Endogâmicos C57BL
11.
Leukemia ; 26(5): 985-90, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22182920

RESUMO

Drug resistance is a growing area of concern. It has been shown that a small, residual pool of leukemic CD34+ progenitor cells can survive in the marrow microenvironment of chronic myeloid leukemia (CML) patients after years of kinase inhibitor treatment. Bone marrow (BM) stroma has been implicated in the long-term survival of leukemic cells, and contributes to the expansion and proliferation of both transformed and normal hematopoietic cells. Mechanistically, we found that CML cells expressed CXCR4, and that plerixafor diminished BCR-ABL-positive cell migration and reduced adhesion of these cells to extra cellular-matrix components and to BM stromal cells in vitro. Moreover, plerixafor decreased the drug resistance of CML cells induced by co-culture with BM stromal cells in vitro. Using a functional mouse model of progressive and residual disease, we demonstrated the ability of the CXCR4 inhibitor, plerixafor, to mobilize leukemic cells in vivo, such that a plerixafor-nilotinib combination reduced the leukemia burden in mice significantly below the baseline level suppression exhibited by a moderate-to-high dose of nilotinib as single agent. These results support the idea of using CXCR4 inhibition in conjunction with targeted tyrosine kinase inhibition to override drug resistance in CML and suppress or eradicate residual disease.


Assuntos
Antineoplásicos/uso terapêutico , Células da Medula Óssea/efeitos dos fármacos , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Pirimidinas/uso terapêutico , Receptores CXCR4/antagonistas & inibidores , Células Estromais/efeitos dos fármacos , Animais , Benzilaminas , Células da Medula Óssea/metabolismo , Células da Medula Óssea/patologia , Adesão Celular , Linhagem Celular Tumoral , Proliferação de Células , Técnicas de Cocultura , Ciclamos , Resistencia a Medicamentos Antineoplásicos , Citometria de Fluxo , Compostos Heterocíclicos/farmacologia , Humanos , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , Camundongos , Células Estromais/metabolismo , Células Estromais/patologia
12.
Leukemia ; 26(3): 481-9, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21860432

RESUMO

Myeloproliferative neoplasms are characterized by overproduction of myeloid lineage cells with frequent acquisition of oncogenic JAK2V617F kinase mutations. The molecular mechanisms that regulate energy requirements in these diseases are poorly understood. Transformed cells tend to rely on fermentation instead of more efficient oxidative phosphorylation for energy production. Our data in JAK2V617F-transformed cells show that growth and metabolic activity were strictly dependent on the presence of glucose. Uptake of glucose and cell surface expression of the glucose transporter Glut1 required the oncogenic tyrosine kinase. Importantly, JAK2V617F as well as active STAT5 increased the expression of the inducible rate-limiting enzyme 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3), which controls glycolytic flux through 6-phosphofructo-1-kinase. PFKFB3 was required for JAK2V617F-dependent lactate production, oxidative metabolic activity and glucose uptake. Targeted knockdown of PFKFB3 also limited cell growth under normoxic and hypoxic conditions and blocked in vivo tumor formation in mice. Overall, these data suggest that inducible PFKFB3 is required for increased growth, metabolic activity and is regulated through active JAK2 and STAT5. Novel therapies that specifically block PFKFB3 activity or expression would, therefore, be expected to inhibit JAK2/STAT5-dependent malignancies and related cancers.


Assuntos
Frutose-Bifosfatase/genética , Frutose-Bifosfatase/metabolismo , Janus Quinase 2/genética , Janus Quinase 2/metabolismo , Fosfofrutoquinase-2/genética , Animais , Linhagem Celular Tumoral , Proliferação de Células , Transformação Celular Neoplásica/genética , Feminino , Glucose/metabolismo , Transportador de Glucose Tipo 1/genética , Transportador de Glucose Tipo 1/metabolismo , Humanos , Camundongos , Camundongos SCID , Fosfofrutoquinase-2/metabolismo , Fator de Transcrição STAT5/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Leukemia ; 26(10): 2233-44, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22469781

RESUMO

Acute myeloid leukemia (AML) progenitors are frequently characterized by activating mutations in the receptor tyrosine kinase Fms-like tyrosine kinase-3 (FLT3). Protein tyrosine kinases are integral components of signaling cascades that have a role in both FLT3-mediated transformation as well as viability pathways that are advantageous to leukemic cell survival. The bone marrow microenvironment can diminish AML sensitivity to tyrosine kinase inhibitors. We hypothesized that inhibition of protein kinases in addition to FLT3 may be effective in overriding drug resistance in AML. We used a cell-based model mimicking stromal protection as part of an unbiased high-throughput chemical screen to identify kinase inhibitors with the potential to override microenvironment-mediated drug resistance in mutant FLT3-positive AML. Several related multi-targeted kinase inhibitors, including dasatinib, with the capability of reversing microenvironment-induced resistance to FLT3 inhibition were identified and validated. We validated synergy in vitro and demonstrated effective combination potential in vivo. In particular Janus kinase inhibitors were effective in overriding stromal protection and potentiating FLT3 inhibition in primary AML and cell lines. These results hint at a novel concept of using combination therapy to override drug resistance in mutant FLT3-positive AML in the bone marrow niche and suppress or eradicate residual disease.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Janus Quinases/antagonistas & inibidores , Leucemia Mieloide Aguda/tratamento farmacológico , Mutação , Inibidores de Proteínas Quinases/administração & dosagem , Tirosina Quinase 3 Semelhante a fms/genética , Animais , Dasatinibe , Resistencia a Medicamentos Antineoplásicos , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Camundongos , Pirimidinas/administração & dosagem , Fator de Transcrição STAT5/metabolismo , Estaurosporina/administração & dosagem , Estaurosporina/análogos & derivados , Células Estromais/fisiologia , Tiazóis/administração & dosagem , Tirosina Quinase 3 Semelhante a fms/antagonistas & inibidores
14.
Leukemia ; 24(12): 2100-9, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20844561

RESUMO

Drug resistance is a growing concern with clinical use of tyrosine kinase inhibitors. Utilizing in vitro models of intrinsic drug resistance and stromal-mediated chemoresistance, as well as functional mouse models of progressive and residual disease, we attempted to develop a potential therapeutic approach designed to suppress leukemia recurrence following treatment with selective kinase inhibitors. The novel IAP inhibitor, LCL161, [corrected] was observed to potentiate the effects of tyrosine kinase inhibition against leukemic disease both in the absence and presence of a stromal-protected [corrected] environment. LCL161 enhanced the proapoptotic effects of nilotinib and PKC412, against leukemic disease in vitro and potentiated the activity of both kinase inhibitors against leukemic disease in vivo. In addition, LCL161 synergized in vivo with nilotinib to reduce leukemia burden significantly below the baseline level suppression exhibited by a moderate-to-high dose of nilotinib. Finally, LCL161 displayed antiproliferative effects against cells characterized by intrinsic resistance to tyrosine kinase inhibitors as a result of expression of point mutations in the protein targets of drug inhibition. These results support the idea of using IAP inhibitors in conjunction with targeted tyrosine kinase inhibition to override drug resistance and suppress or eradicate residual disease.


Assuntos
Proteínas Inibidoras de Apoptose/antagonistas & inibidores , Leucemia/tratamento farmacológico , Inibidores de Proteínas Quinases/uso terapêutico , Tirosina Quinase 3 Semelhante a fms/antagonistas & inibidores , Animais , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos , Sinergismo Farmacológico , Humanos , Leucemia/patologia , Camundongos , Proteínas Proto-Oncogênicas c-abl/antagonistas & inibidores , Pirimidinas/uso terapêutico , Estaurosporina/análogos & derivados , Estaurosporina/uso terapêutico
15.
Leukemia ; 22(1): 66-77, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17851551

RESUMO

Human leukemias harboring chromosomal translocations involving the mixed lineage leukemia (MLL, HRX, ALL-1) gene possess high-level expression, and frequent activating mutations of the receptor tyrosine kinase FLT3. We used a murine bone marrow transplant model to assess cooperation between MLL translocation and FLT3 activation. We demonstrate that MLL-AF9 expression induces acute myelogenous leukemia (AML) in approximately 70 days, whereas the combination of MLL-AF9 and FLT3-ITD does so in less than 30 days. Secondary transplantation of splenic cells from diseased mice established that leukemia stem cells are present at a very high frequency of approximately 1:100 in both diseases. Importantly, prospectively isolated granulocyte macrophage progenitors (GMPs) coinfected with MLL-AF9 and FLT3-ITD give rise to a similar AML, with shorter latency than from GMP transduced with MLL-AF9 alone. Cooperation between MLL-AF9 and FLT3-ITD was further verified by real-time assessment of leukemogenesis using noninvasive bioluminescence imaging. We used this model to demonstrate that MLL-AF9/FLT3-ITD-induced leukemias are sensitive to FLT3 inhibition in a 2-3 week in vivo assay. These data show that activated FLT3 cooperates with MLL-AF9 to accelerate onset of an AML from whole bone marrow as well as a committed hematopoietic progenitor, and provide a new genetically defined model system that should prove useful for rapid assessment of potential therapeutics in vivo.


Assuntos
Modelos Animais de Doenças , Leucemia Mieloide Aguda/etiologia , Proteína de Leucina Linfoide-Mieloide/genética , Proteínas de Fusão Oncogênica/genética , Tirosina Quinase 3 Semelhante a fms/genética , Animais , Southern Blotting , Western Blotting , Transplante de Medula Óssea , Proliferação de Células , Feminino , Granulócitos/citologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Imunofenotipagem , Imunoprecipitação , Leucemia Mieloide Aguda/patologia , Luciferases/metabolismo , Macrófagos/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação , Proteína de Leucina Linfoide-Mieloide/metabolismo , Proteínas de Fusão Oncogênica/metabolismo , Regiões Promotoras Genéticas , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sequências de Repetição em Tandem , Transfecção , Células Tumorais Cultivadas , Tirosina Quinase 3 Semelhante a fms/metabolismo
17.
J Biol Chem ; 268(31): 23072-80, 1993 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-8226824

RESUMO

We have shown previously that there are significant differences between mammalian cell lines in response to disruption of the assembly of the mitotic spindle apparatus (Kung, A. L., Sherwood, S. W., and Schimke, R. T. (1990) Proc. Natl. Acad. Sci. U. S. A. 87, 9553-9557). In this paper we report that there are also significant differences between mammalian cell lines in response to the inhibition of DNA synthesis. In HeLa S3 cells protein synthesis is down-regulated, and cellular growth is arrested in response to the inhibition of DNA synthesis. Upon release from inhibition and resumption of normal growth, cellular viability is maintained near untreated control levels. In contrast, Chinese hamster ovary cells continue to accumulate protein and continue to undergo cellular growth during the period of DNA synthesis inhibition. Cyclin B levels accumulate throughout the period of inhibition and rapidly exceed normal levels at mitosis. The degree of aberrant growth during the period of transient DNA synthesis inhibition is directly related to the degree of subsequent cytotoxicity. If protein accumulation and cellular growth are limited with partially inhibitory levels of cycloheximide during the period of DNA synthesis inhibition, the cytotoxic effects are abolished. These results support the concept that aberrant growth and accumulation of proteins during a transient period of DNA synthesis inhibition are primary determinants of subsequent cell killing.


Assuntos
Afidicolina/farmacologia , Células CHO/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Ciclinas/metabolismo , Replicação do DNA/efeitos dos fármacos , Células HeLa/efeitos dos fármacos , Biossíntese de Proteínas , Animais , Divisão Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Cricetinae , Cicloeximida/farmacologia , DNA/biossíntese , Humanos , Técnicas In Vitro , Especificidade da Espécie , Fatores de Tempo
18.
Proc Natl Acad Sci U S A ; 87(24): 9553-7, 1990 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-2263610

RESUMO

This paper reports that there are major differences between mammalian cell lines in the propensity to progress into subsequent cell cycles when mitosis is inhibited with agents that disrupt the assembly of the mitotic spindle apparatus (Colcemid, nocodazole, and taxol). Human HeLa S3 cells, which represent one extreme, remain arrested in mitosis, with elevated levels of cyclin B and p34cdc2 kinase activity. In Chinese hamster ovary cells, at the other extreme, the periodic rise and fall of cyclin B levels and p34cdc2 kinase activity is only transiently inhibited in the absence of mitosis. The cells progress into subsequent cell cycles, without dividing, resulting in serial doublings of cellular DNA content. In general, the propensity to progress into subsequent cell cycles in the absence of mitosis appears to be species related, such that human cell lines remain permanently blocked in a mitotic state, whereas rodent cell lines are only transiently inhibited when spindle assembly is disrupted. We interpret these results to indicate that in mammalian cell lines there exists a checkpoint which serves to couple cell cycle progression to the completion of certain karyokinetic events. Furthermore, either such a checkpoint exists in some cell lines but not in others or the stringency of the control mechanism varies among different cell lines.


Assuntos
Ciclo Celular , Animais , Proteína Quinase CDC2/metabolismo , Ciclo Celular/efeitos dos fármacos , Linhagem Celular , Cricetinae , Cricetulus , Demecolcina/farmacologia , Feminino , Células HeLa/citologia , Humanos , Cinética , Camundongos , Mitose/efeitos dos fármacos , Ovário
19.
Proc Natl Acad Sci U S A ; 90(8): 3353-7, 1993 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-8386372

RESUMO

The cytotoxic neutral cysteine protease inhibitor N-acetylleucylleucylnorleucinal (ALLN) inhibits cell-cycle progression in CHO cells, affecting the G1/S and metaphase-anaphase transition points, as well as S phase. Mitotic arrest induced by ALLN is associated with the inhibition of cyclin B degradation. At mitosis-inhibiting concentrations of ALLN, cells undergo nuclear-envelope breakdown, spindle formation, chromosome condensation, and congression to the metaphase plate. However, normal anaphase events do not occur, and cells arrest in a metaphase configuration for a prolonged period. Steady-state levels of cyclin B increase to greater than normal mitotic levels, and cyclin B is not degraded for an extended period. Histone H1 kinase activity remains elevated during mitotic arrest. Duration of mitotic arrest depends on ALLN concentration; high concentrations (> 50 micrograms/ml) produce a prolonged mitotic arrest, whereas at lower concentrations, cells are transiently delayed through mitosis (up to 4-12 hr), after which they undergo aberrant cell division resulting in randomly sized daughter cells containing variable amounts of DNA. Cyclin B degradation fails to occur, and histone H1 kinase remains activated for the duration of mitotic arrest at all ALLN concentrations.


Assuntos
Ciclo Celular/efeitos dos fármacos , Ciclinas/metabolismo , Inibidores de Cisteína Proteinase/farmacologia , Leupeptinas/farmacologia , Animais , Células CHO , Cricetinae , Citometria de Fluxo , Imunofluorescência , Cinética , Mitose/efeitos dos fármacos , Protamina Quinase/metabolismo , Fatores de Tempo
20.
Cell ; 102(6): 839-48, 2000 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-11030627

RESUMO

Complexes containing p300, but not CBP, and the nuclear proto-oncoprotein SYT were detected in confluent cultures of G1-arrested cells but not in sparse cells or during S or G2. SYT sequences constitute the N-terminal segment of a fusion oncogene product, SYT-SSX, routinely detected in synovial sarcoma, an aggressive human tumor. SYT/p300 complex formation promotes cell adhesion to a fibronectin matrix, as reflected by compromise of this process in cells expressing SYT dl mutants that retain p300 binding activity and in the primary fibroblasts of p300 but not CBP heterozygous null mice. The mechanism linking the action of SYT/p300 complexes to adhesion function is, at least in part, transcription activation-independent and results in proper activation of beta1 integrin, a major adhesion receptor.


Assuntos
Proteínas Nucleares/metabolismo , Proteínas/genética , Proteínas/metabolismo , Membrana Sinovial/citologia , Membrana Sinovial/metabolismo , Transativadores/metabolismo , Animais , Adesão Celular/fisiologia , Linhagem Celular , Núcleo Celular/metabolismo , Proteína p300 Associada a E1A , Feminino , Feto/citologia , Fibroblastos/citologia , Fibronectinas/metabolismo , Fase G1 , Dosagem de Genes , Expressão Gênica/fisiologia , Haplorrinos , Humanos , Rim/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Peso Molecular , Mutagênese/fisiologia , Proteínas/química , Proteínas Proto-Oncogênicas , Proteínas Repressoras
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA