Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Gynecol Oncol ; 142(2): 341-8, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27017984

RESUMO

OBJECTIVE: The cancer stem cell (CSC) paradigm hypothesizes that successful clinical eradication of CSCs may lead to durable remission for patients with ovarian cancer. Despite mounting evidence in support of ovarian CSCs, their phenotype and clinical relevance remain unclear. We and others have found high aldehyde dehydrogenase 1 (ALDH(high)) expression in a variety of normal and malignant stem cells, and sought to better characterize ALDH(high) cells in ovarian cancer. METHODS: We compared ALDH(high) to ALDH(low) cells in two ovarian cancer models representing distinct subtypes: FNAR-C1 cells, derived from a spontaneous rat endometrioid carcinoma, and the human SKOV3 cell line (described as both serous and clear cell subtypes). We assessed these populations for stem cell features then analyzed expression by microarray and qPCR. RESULTS: ALDH(high) cells displayed CSC properties, including: smaller size, quiescence, regenerating the phenotypic diversity of the cell lines in vitro, lack of contact inhibition, nonadherent growth, multi-drug resistance, and in vivo tumorigenicity. Microarray and qPCR analysis of the expression of markers reported by others to enrich for ovarian CSCs revealed that ALDH(high) cells of both models showed downregulation of CD24, but inconsistent expression of CD44, KIT and CD133. However, the following druggable targets were consistently expressed in the ALDH(high) cells from both models: mTOR signaling, her-2/neu, CD47 and FGF18/FGFR3. CONCLUSIONS: Based on functional characterization, ALDH(high) ovarian cancer cells represent an ovarian CSC population. Differential gene expression identified druggable targets that have the potential for therapeutic efficacy against ovarian CSCs from multiple subtypes.


Assuntos
Aldeído Oxirredutases/biossíntese , Células-Tronco Neoplásicas/enzimologia , Células-Tronco Neoplásicas/patologia , Neoplasias Ovarianas/enzimologia , Neoplasias Ovarianas/patologia , Retinal Desidrogenase/biossíntese , Família Aldeído Desidrogenase 1 , Aldeído Oxirredutases/genética , Animais , Processos de Crescimento Celular/fisiologia , Feminino , Humanos , Terapia de Alvo Molecular , Células-Tronco Neoplásicas/efeitos dos fármacos , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/genética , Ratos , Ratos Endogâmicos Lew , Retinal Desidrogenase/genética
2.
Cell Stem Cell ; 11(3): 346-58, 2012 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-22958930

RESUMO

Internal tandem duplication (ITD) mutations within the FMS-like tyrosine kinase-3 (FLT3) render the receptor constitutively active driving proliferation and survival in leukemic blasts. Expression of FLT3-ITD from the endogenous promoter in a murine knockin model results in progenitor expansion and a myeloproliferative neoplasm. In this study, we show that this expansion begins with overproliferation within a compartment of normally quiescent long-term hematopoietic stem cells (LT-HSCs), which become rapidly depleted. This depletion is reversible upon treatment with the small molecule inhibitor Sorafenib, which also ablates the disease. Although the normal LT-HSC has been defined as FLT3(-) by flow cytometric detection, we demonstrate that FLT3 is capable of playing a role within this compartment by examining the effects of constitutively activated FLT3-ITD. This indicates an important link between stem cell quiescence/homeostasis and myeloproliferative disease while also giving novel insight into the emergence of FLT3-ITD mutations in the evolution of leukemic transformation.


Assuntos
Neoplasias da Medula Óssea/patologia , Duplicação Gênica , Técnicas de Introdução de Genes , Células-Tronco Hematopoéticas/patologia , Homeostase , Sequências de Repetição em Tandem/genética , Tirosina Quinase 3 Semelhante a fms/genética , Animais , Medula Óssea/efeitos dos fármacos , Medula Óssea/metabolismo , Medula Óssea/patologia , Neoplasias da Medula Óssea/genética , Compartimento Celular/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Feminino , Duplicação Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/metabolismo , Homeostase/efeitos dos fármacos , Homeostase/genética , Masculino , Camundongos , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Niacinamida/análogos & derivados , Niacinamida/farmacologia , Fenótipo , Compostos de Fenilureia/farmacologia , Regiões Promotoras Genéticas/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Células da Side Population/efeitos dos fármacos , Células da Side Population/metabolismo , Células da Side Population/patologia , Transdução de Sinais/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/farmacologia , Sorafenibe , Tirosina Quinase 3 Semelhante a fms/metabolismo
3.
Blood ; 119(15): 3571-7, 2012 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-22262762

RESUMO

Relapse of acute myeloid leukemia (AML) is thought to reflect the failure of current therapies to adequately target leukemia stem cells (LSCs), the rare, resistant cells presumed responsible for maintenance of the leukemia and typically enriched in the CD34(+)CD38(-) cell population. Despite the considerable research on LSCs over the past 2 decades, the clinical significance of these cells remains uncertain. However, if clinically relevant, it is expected that LSCs would be enriched in minimal residual disease and predictive of relapse. CD34(+) subpopulations from AML patients were analyzed by flow cytometry throughout treatment. Sorted cell populations were analyzed by fluorescence in situ hybridization for leukemia-specific cytogenetic abnormalities (when present) and by transplantation into immunodeficient mice to determine self-renewal capacity. Intermediate (int) levels of aldehyde dehydrogenase (ALDH) activity reliably distinguished leukemic CD34(+)CD38(-) cells capable of engrafting immunodeficient mice from residual normal hematopoietic stem cells that exhibited relatively higher ALDH activity. Minimal residual disease detected during complete remission was enriched for the CD34(+)CD38(-)ALDH(int) leukemic cells, and the presence of these cells after therapy highly correlated with subsequent clinical relapse. ALDH activity appears to distinguish normal from leukemic CD34(+)CD38(-) cells and identifies those AML cells associated with relapse.


Assuntos
Antígenos CD34/metabolismo , Leucemia Mieloide Aguda/diagnóstico , Leucemia Mieloide Aguda/patologia , Células-Tronco Neoplásicas/patologia , ADP-Ribosil Ciclase 1/metabolismo , Adulto , Idoso , Animais , Separação Celular/métodos , Células Cultivadas , Feminino , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Camundongos Transgênicos , Pessoa de Meia-Idade , Células-Tronco Neoplásicas/metabolismo , Prognóstico , Recidiva , Adulto Jovem
4.
Am J Hematol ; 86(1): 31-7, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21132730

RESUMO

Although tyrosine kinase inhibitors have redefined the care of chronic myeloid leukemia (CML), these agents have not proved curative, likely due to resistance of the leukemia stem cells (LSC). While a number of potential therapeutic targets have emerged in CML, their expression in the LSC remains largely unknown. We therefore isolated subsets of CD34(+) stem/progenitor cells from normal donors and from patients with chronic phase or blast crisis CML. These cell subsets were then characterized based on ability to engraft immunodeficient mice and expression of candidate therapeutic targets. The CD34(+)CD38(-) CML cell population with high aldehyde dehydrogenase (ALDH) activity was the most enriched for immunodeficient mouse engrafting capacity. The putative targets: PROTEINASE 3, SURVIVIN, and hTERT were expressed only at relatively low levels by the CD34(+)CD38(-)ALDH(high) CML cells, similar to the normal CD34(+)CD38(-)ALDH(high) cells and less than in the total CML CD34(+) cells. In fact, the highest expression of these antigens was in normal, unfractionated CD34(+) cells. In contrast, PRAME and WT1 were more highly expressed by all CML CD34(+) subsets than their normal counterparts. Thus, ALDH activity appears to enrich for CML stem cells, which display an expression profile that is distinct from normal stem/progenitor cells and even the CML progenitors. Indeed, expression of a putative target by the total CD34(+) population in CML does not guarantee expression by the LSC. These expression patterns suggest that PROTEINASE 3, SURVIVIN, and hTERT are not optimal therapeutic targets in CML stem cells; whereas PRAME and WT1 seem promising.


Assuntos
Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/patologia , Leucemia Mielogênica Crônica BCR-ABL Positiva/metabolismo , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , ADP-Ribosil Ciclase 1/biossíntese , Adulto , Idoso , Aldeído Desidrogenase/biossíntese , Animais , Antígenos CD34/biossíntese , Antígenos de Neoplasias/biossíntese , Antígenos de Neoplasias/genética , Feminino , Células-Tronco Hematopoéticas/imunologia , Humanos , Proteínas Inibidoras de Apoptose , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Leucemia Mielogênica Crônica BCR-ABL Positiva/imunologia , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Proteínas Associadas aos Microtúbulos/biossíntese , Proteínas Associadas aos Microtúbulos/genética , Pessoa de Meia-Idade , Mieloblastina/biossíntese , Mieloblastina/genética , Transplante de Neoplasias , RNA Mensageiro/biossíntese , Survivina , Telomerase/biossíntese , Telomerase/genética , Proteínas WT1/biossíntese , Proteínas WT1/genética
5.
PLoS One ; 5(6): e11251, 2010 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-20582166

RESUMO

BACKGROUND: Pulmonary hypertension (PH) is a disease of multiple etiologies with several common pathological features, including inflammation and pulmonary vascular remodeling. Recent evidence has suggested a potential role for the recruitment of bone marrow-derived (BMD) progenitor cells to this remodeling process. We recently demonstrated that hypoxia-induced mitogenic factor (HIMF/FIZZ1/RELM alpha) is chemotactic to murine bone marrow cells in vitro and involved in pulmonary vascular remodeling in vivo. METHODOLOGY/PRINCIPAL FINDINGS: We used a mouse bone marrow transplant model in which lethally irradiated mice were rescued with bone marrow transplanted from green fluorescent protein (GFP)(+) transgenic mice to determine the role of HIMF in recruiting BMD cells to the lung vasculature during PH development. Exposure to chronic hypoxia and pulmonary gene transfer of HIMF were used to induce PH. Both models resulted in markedly increased numbers of BMD cells in and around the pulmonary vasculature; in several neomuscularized small (approximately 20 microm) capillary-like vessels, an entirely new medial wall was made up of these cells. We found these GFP(+) BMD cells to be positive for stem cell antigen-1 and c-kit, but negative for CD31 and CD34. Several of the GFP(+) cells that localized to the pulmonary vasculature were alpha-smooth muscle actin(+) and localized to the media layer of the vessels. This finding suggests that these cells are of mesenchymal origin and differentiate toward myofibroblast and vascular smooth muscle. Structural location in the media of small vessels suggests a functional role in the lung vasculature. To examine a potential mechanism for HIMF-dependent recruitment of mesenchymal stem cells to the pulmonary vasculature, we performed a cell migration assay using cultured human mesenchymal stem cells (HMSCs). The addition of recombinant HIMF induced migration of HMSCs in a phosphoinosotide-3-kinase-dependent manner. CONCLUSIONS/SIGNIFICANCE: These results demonstrate HIMF-dependent recruitment of BMD mesenchymal-like cells to the remodeling pulmonary vasculature.


Assuntos
Vasos Sanguíneos/citologia , Células da Medula Óssea/citologia , Hipóxia/fisiopatologia , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Pulmão/irrigação sanguínea , Animais , Western Blotting , Transplante de Medula Óssea , Quimiotaxia , Dependovirus/genética , Feminino , Vetores Genéticos , Imuno-Histoquímica , Peptídeos e Proteínas de Sinalização Intercelular/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia de Fluorescência
6.
Pathology ; 39(2): 247-51, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17454756

RESUMO

AIMS: The transgenic enhanced green fluorescent protein (EGFP) expressing 'green' mouse (C57BL/6-TgN(ACTbEGFP)1Osb) is a widely used tool in stem cell research, where the ubiquitous nature of EGFP expression is critical to track the fate of single or small groups of transplanted haematopoietic stem cells (HSC). Our aim was to investigate this assumed ubiquitous expression by performing a detailed histological survey of EGFP expression in these mice. METHODS: Fluorescent microscopy of frozen tissue sections was used to perform a detailed histological survey of the pattern of EGFP expression in these mice. Flow cytometry was also used to determine the expression pattern in blood and bone marrow. RESULTS: Three patterns of EGFP expression were noted. In most tissues there was an apparently stochastic variegation of the transgene, with individual cell types demonstrating highly variable rates of EGFP expression. Certain specific cell types such as pancreatic ductal epithelium, cerebral cortical neurones and glial cells and glomerular mesangial cells consistently lacked EGFP expression, while others, including pancreatic islet cells, expressed EGFP only at extremely low levels, barely distinguishable from background. Lastly, in the colon and stomach the pattern of EGFP expression was suggestive of clonal inactivation. Only cardiac and skeletal muscle showed near ubiquitous expression. CONCLUSIONS: These findings raise questions regarding the 'ubiquitous' expression of EGFP in these transgenic mice and suggest caution in relying overly on EGFP alone as an infallible marker of donor cell origin.


Assuntos
Pesquisa Biomédica/métodos , Linhagem da Célula , Proteínas de Fluorescência Verde/biossíntese , Modelos Animais , Células-Tronco , Animais , Citometria de Fluxo , Imunofluorescência , Proteínas de Fluorescência Verde/genética , Camundongos , Camundongos Transgênicos , Microscopia de Fluorescência
7.
Exp Hematol ; 35(2): 335-41, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17258082

RESUMO

OBJECTIVE: Our laboratory has established two unique methods to isolate murine hematopoietic stem cells on the basis of functional characteristics such as the ability of stem cells to home to bone marrow and aldehyde dehydrogenase (ALDH) activity. An essential component of both protocols is the separation of whole bone marrow into small-sized cells by counter-flow elutriation. We sought to provide the scientific community with an alternate approach to acquire our stem cells by replacing elutriation with the use of density-gradient centrifugation. METHODS: The elutriated fraction 25 population was characterized based on density using a discontinuous gradient. The long-term reconstituting potential of whole bone marrow cells collected at each density interface was determined by subjecting the fractions to the two-day homing protocol, transplanting them into lethally irradiated recipient mice, and assessing peripheral blood chimerism. We also investigated the ability of high-density bone marrow cells isolated in conjunction with the ALDH protocol to repopulate the hematopoietic system of myeloablated recipients. RESULTS: Bone marrow cells collected at the high-density interface of 1.081/1.087 g/mL (fraction 3) had the capacity for homing to marrow and the ability to provide long-term hematopoietic reconstitution. Fraction three lineage-depleted ALDH-bright cells could also engraft and provide long-term hematopoiesis at limiting dilutions. CONCLUSIONS: Density-gradient centrifugation can be used in conjunction with either of our stem cell isolation protocols to obtain cells with long-term reconstitution ability. We anticipate that this strategy will encourage and enable investigators to study the biology of HSCs isolated using functional characteristics.


Assuntos
Células da Medula Óssea/citologia , Separação Celular/métodos , Células-Tronco Hematopoéticas/citologia , Aldeído Desidrogenase/imunologia , Animais , Células da Medula Óssea/enzimologia , Células da Medula Óssea/efeitos da radiação , Centrifugação com Gradiente de Concentração/métodos , Ativação Enzimática , Feminino , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/enzimologia , Células-Tronco Hematopoéticas/efeitos da radiação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Taxa de Sobrevida , Condicionamento Pré-Transplante , Transplante Homólogo , Irradiação Corporal Total
8.
J Urol ; 176(2): 813-8, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16813953

RESUMO

PURPOSE: Using various nonphysiological tissue injury/repair models numerous studies have demonstrated the capacity of bone marrow derived cells to contribute to the repopulation of epithelial tissues following damage. To investigate whether this phenomenon might also occur during periods of physiological tissue degeneration/regeneration we compared the ability of bone marrow derived cells to rejuvenate the prostate gland in mice that were castrated and then later treated with dihydrotestosterone vs mice with prostate epithelium that had been damaged by lytic virus infection. MATERIALS AND METHODS: Using allogenic bone marrow grafts from female donor transgenic mice expressing green fluorescent protein transplanted into lethally irradiated males we were able to assess the contributions of bone marrow derived cells to recovery of the prostatic epithelium in 2 distinct systems, including 1) surgical castration followed 1 week later by dihydrotestosterone replacement and 2) intraprostatic viral injection. Eight to 10-week-old male C57/Bl6 mice were distributed among bone marrow donor-->recipient/prostate injury groups, including 5 with C57/Bl6-->C57/Bl6/no injury, 3 with green fluorescent protein-->C57/Bl6/no injury, 3 with green fluorescent protein-->C57/Bl6/vehicle injection, 4 with green fluorescent protein-->C57/Bl6/virus injection and 3 each with green fluorescent protein-->C57/Bl6/castration without and with dihydrotestosterone, respectively. Prostate tissues were harvested 3 weeks after dihydrotestosterone replacement or 14 days following intraprostatic viral injection. Prostate tissue immunofluorescence was performed with antibodies against the epithelial marker cytokeratin 5/8, the hematopoietic marker CD45 and green fluorescent protein. RESULTS: Mice that sustained prostate injury from vaccinia virus infection with concomitant severe inflammation and glandular disruption showed evidence of bone marrow derived cell reconstitution of prostate epithelium, that is approximately 4% of all green fluorescent protein positive cells in the epithelial compartment 14 days after injury expressed cytokeratin 5/8, similar to the proportion of green fluorescent protein positive cells in the prostate that no longer expressed the hematopoietic marker CD45. When prostatic degeneration/regeneration was triggered by androgen deprivation and reintroduction, no green fluorescent protein positive prostate epithelial cells were detected. CONCLUSIONS: These findings are consistent with a requirement for inflammation associated architectural destruction for the bone marrow derived cell contribution to the regeneration of prostate epithelium.


Assuntos
Células da Medula Óssea/fisiologia , Próstata/patologia , Próstata/fisiologia , Regeneração , Animais , Epitélio/patologia , Epitélio/fisiologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL
9.
Radiat Res ; 162(1): 48-55, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15222777

RESUMO

It has recently been shown that specific lineage-depleted murine hematopoietic stem cells that home to the bone marrow 2 days after transplantation of ablated primary recipients are capable of long-term engraftment and repopulation of secondary recipients. We were interested in determining whether the rate at which the ablating radiation dose was delivered to the mice affected the homing of lineage-depleted stem cells to the bone marrow and/or sites of tissue damage. Fractionated, lineage-depleted donor marrow cells were isolated and labeled with the membrane dye PKH26. Recipient mice were lethally irradiated with 11 Gy ionizing radiation using varying dose rates and were immediately injected with PKH26-labeled progenitor stem cells. With the exception of the lowest dose-rate group, all irradiated mice had an approximately fivefold (P = 0.014 to 0.025) reduction in stem cell homing to the bone marrow compared to unirradiated control animals. A fivefold reduction of stem cell homing to the spleen compared to unirradiated animals was also observed, though this was not statistically significant for any dose-rate group (P = 0.072 to 0.233). This difference in homing could not be explained by increased stem cell apoptosis/necrosis or non-marrow tissue homing to the intestine, lung or liver. We show that the dose rate at which a lethal dose of total-body radiation is delivered does not augment hematopoietic progenitor stem cell homing to the bone marrow, spleen or sites of early radiation-mediated tissue damage at either 2 or 5 days postirradiation/transplantation. The observation that greater homing was seen in unirradiated control mice calls into question the concept that adequate bone marrow stem cell homing requires radiation-induced "space" to be made in the marrow, certainly for the enriched early progenitor hematopoietic stem cells used for this set of experiments. Further experiments will be needed to determine whether these homed cells are as capable of giving rise to long-term engraftment/repopulation of the marrow of secondary recipients as they are in irradiated recipients.


Assuntos
Células-Tronco Hematopoéticas/fisiologia , Animais , Movimento Celular/efeitos da radiação , Relação Dose-Resposta à Radiação , Molécula 1 de Adesão Intercelular/análise , Molécula 1 de Adesão Intercelular/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Tolerância a Radiação , Irradiação Corporal Total
10.
Nat Cell Biol ; 6(6): 532-9, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15133469

RESUMO

Both plasticity and cell fusion have been suggested to have a role in germ-layer switching. To understand the mechanisms underlying cell fate changes, we have examined a highly enriched population of hematopoietic stem cells (HSCs) in vitro or in vivo in response to injury for liver-specific phenotypic and functional changes. Here we show that HSCs become liver cells when cocultured with injured liver separated by a barrier. Chromosomal analyses and tissue-specific gene and/or protein expression show that microenvironmental cues rather than fusion are responsible for conversion in vitro. We transplanted HSCs into liver-injured mice and observed that HSCs convert into viable hepatocytes with increasing injury. Notably, liver function was restored 2-7 d after transplantation. We conclude that HSCs contribute to the regeneration of injured liver by converting into functional hepatocytes without fusion.


Assuntos
Diferenciação Celular/fisiologia , Linhagem da Célula/genética , Camadas Germinativas/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Hepatócitos/metabolismo , Regeneração Hepática/fisiologia , Animais , Biomarcadores , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Sobrevivência Celular/genética , Células Cultivadas , Técnicas de Cocultura , Sinais (Psicologia) , Meios de Cultivo Condicionados/farmacologia , Líquido Extracelular/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento/genética , Camadas Germinativas/citologia , Sobrevivência de Enxerto/genética , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Hepatócitos/citologia , Regeneração Hepática/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ploidias , Cromossomos Sexuais/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
11.
J Biol Chem ; 279(15): 15678-87, 2004 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-14747476

RESUMO

AML1/RUNX1, a member of the core binding factor (CBF) family stimulates myelopoiesis and lymphopoiesis by activating lineage-specific genes. In addition, AML1 induces S phase entry in 32Dcl3 myeloid or Ba/F3 lymphoid cells via transactivation. We now found that AML1 levels are regulated during the cell cycle. 32Dcl3 and Ba/F3 cell cycle fractions were prepared using elutriation. Western blotting and a gel shift/supershift assay demonstrated that endogenous CBF DNA binding and AML1 levels were increased 2-4-fold in S and G(2)/M phase cells compared with G(1) cells. In addition, G(1) arrest induced by mimosine reduced AML1 protein levels. In contrast, AML1 RNA did not vary during cell cycle progression relative to actin RNA. Analysis of exogenous Myc-AML1 or AML1-ER demonstrated a significant reduction in G(1) phase cells, whereas levels of exogenous DNA binding domain alone were constant, lending support to the conclusion that regulation of AML1 protein stability contributes to cell cycle variation in endogenous AML1. However, cytokine-dependent AML1 phosphorylation was independent of cell cycle phase, and an AML1 mutant lacking two ERK phosphorylation sites was still cell cycle-regulated. Inhibition of AML1 activity with the CBFbeta-SMMHC or AML1-ETO oncoproteins reduced cyclin D3 RNA expression, and AML1 bound and activated the cyclin D3 promoter. Signals stimulating G(1) to S cell cycle progression or entry into the cell cycle in immature hematopoietic cells might do so in part by inducing AML1 expression, and mutations altering pathways regulating variation in AML1 stability potentially contribute to leukemic transformation.


Assuntos
Ciclinas/biossíntese , Proteínas de Ligação a DNA/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Fatores de Transcrição/fisiologia , Actinas/metabolismo , Animais , Northern Blotting , Western Blotting , Ciclo Celular , Linhagem Celular , Transformação Celular Neoplásica , Subunidade alfa 2 de Fator de Ligação ao Core , Ciclina D3 , Ciclinas/metabolismo , DNA/química , DNA Complementar/metabolismo , Proteínas de Ligação a DNA/metabolismo , Fase G1 , Fase G2 , Regulação da Expressão Gênica , Humanos , Camundongos , Mimosina/farmacologia , Mutação , Fosforilação , Regiões Promotoras Genéticas , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas/metabolismo , RNA/química , Fase S , Fatores de Transcrição/metabolismo , Ativação Transcricional
12.
Exp Hematol ; 31(1): 56-64, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12543107

RESUMO

OBJECTIVES: The purpose of this study was to investigate cell fates and long-term repopulating potential of a primitive hematopoietic stem cell (HSC) population (i.e., FR25Lin(-) cells) in vitro. MATERIALS AND METHODS: FR25Lin(-) cells were isolated by elutriation and cell sorting and cultured with a combination of cytokines for 7 days. Utilizing the membrane dye PKH-26, cultured cells were separated into two subsets based on their proliferation rates and assayed for progenitors and HSC. RESULTS: Fresh FR25Lin(-) cells were mostly quiescent; however, some of this population entered cell cycle after cytokine exposure reaching a peak 4 to 5 days after culture. Two subsets of cultured cells were isolated: 1) cells that had divided several times (PKH(dull) cells) and 2) cells that remained undivided or divided only once or twice (PKH(bright) cells). The PKH(dull) cells accounted for 94% of total viable cells in culture after 5 days. The PKH(dull) subset contained all the multi-potential in vivo progenitors (CFU-S) and 10 times more committed progenitors (CFU-C). Quantitative analysis of HSC engraftment from the PKH(bright) subset demonstrated stem cell maintenance. For the PKH(dull) subset, on day 5, HSC numbers increased. By day 7, increased differentiation in the PKH(dull) population supports expanding differentiation divisions. CONCLUSIONS: Our primitive HSC population underwent different types of cell divisions stimulated by cytokines, resulting in subsets with different self-renewal and differentiation potentials. This in vitro/in vivo model provides a useful tool for studies of early events during HSC self-renewal and differentiation.


Assuntos
Diferenciação Celular , Divisão Celular , Células-Tronco Hematopoéticas/citologia , Animais , Ciclo Celular/efeitos dos fármacos , Células Cultivadas/citologia , Células Cultivadas/efeitos dos fármacos , Cruzamentos Genéticos , Sinergismo Farmacológico , Feminino , Sobrevivência de Enxerto , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/efeitos dos fármacos , Humanos , Interleucina-6/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Quimera por Radiação , Proteínas Recombinantes/farmacologia , Fator de Células-Tronco/farmacologia , Trombopoetina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...