Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
Haematologica ; 99(5): 858-64, 2014 May.
Article in English | MEDLINE | ID: mdl-24334295

ABSTRACT

PDK1 is a master kinase that activates at least six protein kinase groups including AKT, PKC and S6K and is a potential target in the treatment of a range of malignancies. Here we show overexpression of PDK1 in over 40% of myelomonocytic acute leukemia patients. Overexpression of PDK1 occurred uniformly throughout the leukemic population, including putative leukemia-initiating cells. Clinical outcome analysis revealed PDK1 overexpression was associated with poorer treatment outcome. Primary acute myeloid leukemia blasts over-expressing PDK1 showed improved in vitro survival and ectopic expression of PDK1 promoted the survival of myeloid cell lines. Analysis of PDK1 target kinases revealed that PDK1 overexpression was most closely associated with increased phosphorylation of PKC isoenzymes and inhibition of PKC strongly inhibited the survival advantage of PDK1 over-expressing cells. Membrane localization studies implicated PKCα as a major target for PDK1 in this disease. PDK1 over-expressing blasts showed differential sensitivity to PDK1 inhibition (in the low micromolar range) suggesting oncogene addiction, whilst normal bone marrow progenitors were refractory to PDK1 inhibition at effective inhibitor concentrations. PDK1 inhibition also targeted subpopulations of leukemic blasts with a putative leukemia-initiating cell phenotype. Together these data show that overexpression of PDK1 is common in acute myelomonocytic leukemia and is associated with poorer treatment outcome, probably arising from the cytoprotective function of PDK1. We also show that therapeutic targeting of PDK1 has the potential to be both an effective and selective treatment for these patients, and is also compatible with current treatment regimes.


Subject(s)
Gene Expression , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Protein Kinase C/metabolism , Protein Serine-Threonine Kinases/genetics , Cell Survival/genetics , Cells, Cultured , Enzyme Activation , Gene Expression Regulation, Leukemic/drug effects , Humans , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/mortality , Neoplasm Staging , Patient Outcome Assessment , Phosphorylation , Prognosis , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyruvate Dehydrogenase Acetyl-Transferring Kinase
2.
Blood ; 122(19): 3322-30, 2013 Nov 07.
Article in English | MEDLINE | ID: mdl-24089327

ABSTRACT

Excessive production of reactive oxygen species (ROS) is frequently observed in cancer and is known to strongly influence hematopoietic cell function. Here we report that extracellular ROS production is strongly elevated (mean >10-fold) in >60% of acute myeloid leukemia (AML) patients and that this increase is attributable to constitutive activation of nicotinamide adenine dinucleotide phosphate oxidases (NOX). In contrast, overproduction of mitochondrial ROS was rarely observed. Elevated ROS was found to be associated with lowered glutathione levels and depletion of antioxidant defense proteins. We also show for the first time that the levels of ROS generated were able to strongly promote the proliferation of AML cell lines, primary AML blasts, and, to a lesser extent, normal CD34(+) cells, and that the response to ROS is limited by the activation of the oxidative stress pathway mediated though p38(MAPK). Consistent with this, we observed that p38(MAPK) responses were attenuated in patients expressing high levels of ROS. These data show that overproduction of NOX-derived ROS can promote the proliferation of AML blasts and that they also develop mechanisms to suppress the stress signaling that would normally limit this response. Together these adaptations would be predicted to confer a competitive advantage to the leukemic clone.


Subject(s)
Leukemia, Myeloid, Acute/metabolism , Leukocytes, Mononuclear/metabolism , NADPH Oxidases/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , p38 Mitogen-Activated Protein Kinases/metabolism , Antigens, CD34/genetics , Antigens, CD34/metabolism , Apoptosis , Case-Control Studies , Cell Proliferation , Gene Expression Regulation, Leukemic , Glutathione/metabolism , Humans , Hydrogen Peroxide/metabolism , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Leukocytes, Mononuclear/pathology , NADPH Oxidases/genetics , Oxidative Stress , Primary Cell Culture , p38 Mitogen-Activated Protein Kinases/genetics
3.
Blood ; 115(6): 1238-46, 2010 Feb 11.
Article in English | MEDLINE | ID: mdl-20007804

ABSTRACT

Excessive production of reactive oxygen species (ROS) is a feature of human malignancy and is often triggered by activation of oncogenes such as activated Ras. ROS act as second messengers and can influence a variety of cellular process including growth factor responses and cell survival. We have examined the contribution of ROS production to the effects of N-Ras(G12D) and H-Ras(G12V) on normal human CD34(+) progenitor cells. Activated Ras strongly up-regulated the production of both superoxide and hydrogen peroxide through the stimulation of NADPH oxidase (NOX) activity, without affecting the expression of endogenous antioxidants or the production of mitochondrially derived ROS. Activated Ras also promoted both the survival and the growth factor-independent proliferation of CD34(+) cells. Using oxidase inhibitors and antioxidants, we found that excessive ROS production by these cells did not contribute to their enhanced survival; rather, ROS promoted their growth factor-independent proliferation. Although Ras-induced ROS production specifically activated the p38(MAPK) oxidative stress response, this failed to induce expression of the cell-cycle inhibitor, p16(INK4A); instead, ROS promoted the expression of D cyclins. These data are the first to show that excessive ROS production in the context of oncogene activation can promote proliferative responses in normal human hematopoietic progenitor cells.


Subject(s)
Antigens, CD34/metabolism , Cell Proliferation , Genes, ras/physiology , Hematopoietic Stem Cells/cytology , Intercellular Signaling Peptides and Proteins/metabolism , Reactive Oxygen Species/metabolism , Animals , Antioxidants/metabolism , Blotting, Western , Cells, Cultured , Electron Spin Resonance Spectroscopy , Flow Cytometry , Hematopoietic Stem Cells/metabolism , Humans , Hydrogen Peroxide/metabolism , Mice , Mitochondria/metabolism , NADPH Oxidases/metabolism , Oxidative Stress , Signal Transduction , Superoxides/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
4.
Blood ; 109(10): 4461-9, 2007 May 15.
Article in English | MEDLINE | ID: mdl-17255356

ABSTRACT

Although hyperactivation of Ras is a common feature of myeloid malignancy, its role in subverting hematopoiesis is unclear. We have examined the influence of Ras on normal human uncommitted myeloid subsets and show that expression of this oncogene strongly favors monocyte lineage selection in bipotential granulocyte/macrophage progenitors while inhibiting colony formation in other uncommitted subsets. Ras also promoted monocytic differentiation but not the proliferation of these cells. The mechanism through which Ras drives monocyte lineage selection was dependent on PKC activity and Ras was found to promote the expression, membrane translocation, and phosphorylation of conventional and novel PKC isoforms. We further show that Ras promoted the expression of the AGC kinase master regulator, PDK1, which maintains the stability and activity of PKC isoforms. Consistent with this, overexpression of PDK1 itself promoted monocyte colony formation and translocation of PKC. Overexpression of PDK1 was found to be a common feature of acute myeloid leukemia (45% of patients) and was closely associated with hyperphosphorylation of PKC. These data demonstrate that Ras is able to promote monocyte lineage selection via PKC and show for the first time the involvement of the kinase master regulator, PDK1, in both lineage specification and in human leukemia.


Subject(s)
Cell Differentiation/genetics , Cell Lineage/physiology , Monocytes/cytology , Protein Kinase C/physiology , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins p21(ras)/physiology , 3-Phosphoinositide-Dependent Protein Kinases , Acute Disease , Cells, Cultured , Fetal Blood/cytology , Gene Expression Regulation, Leukemic , Hematopoietic Stem Cells/cytology , Humans , Infant, Newborn , Isoenzymes/metabolism , Leukemia, Myeloid/genetics , Protein Kinase C/genetics , Protein Kinase C/metabolism , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins p21(ras)/genetics
5.
Mol Cell Biol ; 26(10): 3966-75, 2006 May.
Article in English | MEDLINE | ID: mdl-16648489

ABSTRACT

Hyperactivation of Ras is one of the most common abnormalities in acute myeloid leukemia. In experimental models, Ras inhibits myeloid differentiation, which is characteristic of leukemia; however, the mechanism through which it disrupts hematopoiesis is poorly understood. In multipotent FDCP-mix cells, Ras inhibits terminal neutrophil differentiation, thereby indefinitely extending their proliferative potential. Ras also strongly promotes the sensitivity of these cells to granulocyte-macrophage colony-stimulating factor (GM-CSF). Using this model, we have dissected the signaling elements downstream of Ras to determine their relative contribution to the dysregulation of hematopoiesis. Cells expressing Ras mutants selectively activating Raf (Ras*T35S) or phosphatidylinositol 3-kinase (Ras*Y40C) did not significantly affect differentiation or proliferative capacity, whereas Ras*E37G (which selectively activates RalGEFs) perpetuated proliferation and blocked neutrophil development in a manner similar to that of Ras. Correspondingly, expression of constitutively active versions of these effectors confirmed the overriding importance of Ral guanine nucleotide exchange factors. Cells expressing Ras demonstrated hyperactivation of Ral, which itself was able to exactly mimic the phenotype of Ras, including hypersensitivity to GM-CSF. Conversely, dominant negative Ral promoted spontaneous neutrophil development. Ral, in turn, appears to influence differentiation through multiple effectors. These data show, for the first time, the importance of Ral in regulating differentiation and self-renewal in hematopoietic cells.


Subject(s)
Cell Differentiation , Genes, ras , Leukemia, Myeloid/physiopathology , ral Guanine Nucleotide Exchange Factor/physiology , ras Proteins/physiology , Animals , Blotting, Western , Cell Culture Techniques , Cell Line, Tumor , Cell Proliferation , Clone Cells , Dose-Response Relationship, Drug , Enzyme Activation , Gene Expression Regulation, Leukemic , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Leukemia, Myeloid/genetics , Mice , Mutation , Neutrophils/cytology , Neutrophils/metabolism , Retroviridae/genetics , ral Guanine Nucleotide Exchange Factor/genetics
6.
Biotechnol Prog ; 21(3): 953-8, 2005.
Article in English | MEDLINE | ID: mdl-15932279

ABSTRACT

Though both low-speed centrifugation and the use of fibronectin (Retronectin) fragments increase gene transduction efficiency, they still do not overcome the adverse effects of the presence of virus-containing medium (VCM). In this study, we improved transduction efficiency of primitive human hematopoietic cells by optimizing the conditions for preadsorbing culture dishes with retrovirus using a centrifugation protocol allowing subsequent infection to be carried out in the absence of VCM. We also demonstrate that preadsorbing tissue culture plates with retrovirus is dependent on the volume of VCM used for preadsorption and the length of centrifugation and the type of plasticware used but not on the temperature of centrifugation (4-33 degrees C). Direct exposure of CD34+ target cells to VCM depletes the primitive CD34+CD38neg subpopulation by more than 30%, whereas the optimized VCM-free infection protocol targets this population with equivalent efficiency but had no detrimental effects on CD34+CD38neg frequency. In summary, we demonstrate a high-frequency transduction protocol which preserves the therapeutically relevant primitive subpopulation of human hematopoietic cells.


Subject(s)
Cell Culture Techniques/methods , Hematopoietic Stem Cells/physiology , Hematopoietic Stem Cells/virology , Recombinant Proteins/biosynthesis , Retroviridae/genetics , Transduction, Genetic/methods , Cells, Cultured , Centrifugation/methods , Humans
7.
Blood ; 101(2): 624-32, 2003 Jan 15.
Article in English | MEDLINE | ID: mdl-12393523

ABSTRACT

The t(8;21) translocation, which encodes the AML1-ETO fusion protein (now known as RUNX1-CBF2T1), is one of the most frequent translocations in acute myeloid leukemia, although its role in leukemogenesis is unclear. Here, we report that exogenous expression of AML1-ETO in human CD34(+) cells severely disrupts normal erythropoiesis, resulting in virtual abrogation of erythroid colony formation. In contrast, in bulk liquid culture of purified erythroid cells, we found that while AML1-ETO initially inhibited proliferation during early (erythropoietin [EPO]-independent) erythropoiesis, growth inhibition gave way to a sustained EPO-independent expansion of early erythroid cells that continued for more than 60 days, whereas control cultures became growth arrested after 10 to 13 days (at the EPO-dependent stage of development). Phenotypic analysis showed that although these cells were CD13(-) and CD34(-), unlike control cultures, these cells failed to up-regulate CD36 or to down-regulate CD33, suggesting that expression of AML1-ETO suppressed the differentiation of these cells and allowed extensive self-renewal to occur. In the early stages of this expansion, addition of EPO was able to promote both phenotypic (CD36(+), CD33(-), glycophorin A(+)) and morphologic differentiation of these cells, almost as effectively as in control cultures. However, with extended culture, cells expressing AML1-ETO became refractory to addition of this cytokine, suggesting that a block in differentiation had been established. These data demonstrate the capacity of AML1-ETO to promote the self-renewal of human hematopoietic cells and therefore support a causal role for t(8;21) translocations in leukemogenesis.


Subject(s)
Erythroid Precursor Cells/drug effects , Oncogene Proteins, Fusion/pharmacology , Transcription Factors/pharmacology , Antigens, CD34 , Cell Culture Techniques , Cell Cycle/drug effects , Cell Differentiation/drug effects , Cell Division/drug effects , Core Binding Factor Alpha 2 Subunit , Erythroblasts/cytology , Erythroblasts/drug effects , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/metabolism , Erythropoiesis/drug effects , Fetal Blood , Humans , Leukemia/etiology , Oncogene Proteins, Fusion/genetics , RUNX1 Translocation Partner 1 Protein , Transcription Factors/genetics , Transduction, Genetic
8.
Blood ; 100(12): 4185-92, 2002 Dec 01.
Article in English | MEDLINE | ID: mdl-12393454

ABSTRACT

RAS mutations are one of the most frequent molecular abnormalities associated with myeloid leukemia and preleukemia, yet there is a poor understanding of how they contribute to the pathogenesis of these conditions. Here, we describe the consequences of ectopic mutant N-Ras (N-Ras*) expression on normal human erythropoiesis. We show that during early (erythropoietin [EPO]-independent) erythropoiesis, N-Ras* promoted the amplification of a phenotypically primitive but functionally defective subpopulation of CD34(+) erythroblasts. N-Ras* also up-regulated the expression of megakaryocyte antigens on human erythroblasts. Although early erythroblasts expressing N-Ras* were able to respond to erythropoietin and generate mature progeny, this occurred with greatly reduced efficiency, probably explaining the poor colony growth characteristics of these cells. We further report that this oncogene promoted the expression and activation of protein kinase C (PKC) and that the effects of N-Ras* on erythropoiesis could be abrogated or attenuated by inhibition of PKC. Similarly, the effects of this oncogene could be partially mimicked by treatment with PKC agonist. Together, these data suggest that expression of N-Ras* is able to subvert the normal developmental cues that regulate erythropoiesis by activating PKC. This gives rise to phenotypic and functional abnormalities commonly observed in preleukemia, suggesting a direct link between RAS mutations and the pathogenesis of preleukemia.


Subject(s)
Erythropoiesis/drug effects , Mutation , Protein Kinase C/physiology , ras Proteins/pharmacology , Antigens, CD/drug effects , Antigens, CD/metabolism , Antigens, CD34/drug effects , Cell Division/drug effects , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/drug effects , Erythroid Precursor Cells/immunology , Fetal Blood , Humans , Immunophenotyping , Platelet Membrane Glycoprotein IIb/drug effects , Preleukemia/etiology , Protein Kinase C/antagonists & inhibitors , Transduction, Genetic , ras Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...