Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Leuk Lymphoma ; 45(5): 997-1008, 2004 May.
Article in English | MEDLINE | ID: mdl-15291360

ABSTRACT

Bryostatin, a macrocyclic lactone and protein kinase C (PKC) modulator, has been shown to have differentiation and anti-tumor activity against several leukemia cell lines in vitro. In this study, we demonstrated Bryostatin-induced differentiation in B-cell chronic lymphocytic leukemia (B-CLL) cells, characterized by an increase in cell size and a marked up-regulation of CD11c expression. The specific inhibitors of the extracellular signal-regulated kinase (ERK) and protein kinase C pathways, PD98059 and GF 109203X respectively, each completely blocked Bryostatin-induced differentiation of B-CLL cells, suggesting that activation of the ERK pathway plays a direct role in this process in a PKC-dependent manner. Furthermore, Bryostatin reduced both spontaneous and drug-induced apoptosis with chlorambucil, fludarabine and 2-chloro-2'-deoxyadenosine (2-Cda) in B-CLL cells. This resistance was associated with an early up-regulation of the anti-apoptotic protein Mcl-1 and post-translational phosphorylation of Bcl-2 at serine 70. The anti-apoptotic effects of Bryostatin were abrogated by GF 109203X, and to a lesser extent by the phosphatidylinositol 3-kinase (PI3-kinase) inhibitor, LY294002. Interestingly, the ERK inhibitor, PD98059 inhibited Mcl-1 expression but had little effect on Bryostatin-induced survival suggesting that the ERK pathway predominantly affects differentiation. Taken together these results present an explanation for Bryostatin-induced B-CLL cell survival in which modulation of the PKC pathway couples differentiation with an increase in antiapoptotic protein expression and calls into question the rationale for its use in the treatment of B-CLL.


Subject(s)
Lactones/pharmacology , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Neoplasm Proteins/genetics , Protein Kinase C/drug effects , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Apoptosis/drug effects , Bryostatins , Cell Differentiation/drug effects , Drug Resistance, Neoplasm , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Macrolides , Myeloid Cell Leukemia Sequence 1 Protein , Phosphorylation/drug effects , Signal Transduction , Tumor Cells, Cultured , Up-Regulation/drug effects
2.
Cancer Genet Cytogenet ; 148(1): 49-54, 2004 Jan 01.
Article in English | MEDLINE | ID: mdl-14697641

ABSTRACT

We report a case of a psu dic(6;5)(p21.3;q13) in a patient with secondary myelodysplastic syndrome (sMDS) following treatment for multiple myeloma. The abnormal chromosome was isolated by flow karyotyping and initially identified by reverse chromosome painting. The findings were then confirmed by forward painting. The value of flow karyotyping as a diagnostic technique in hematologic malignancies is discussed.


Subject(s)
Chromosome Aberrations , Chromosome Painting/methods , Chromosomes, Human, Pair 5 , Chromosomes, Human, Pair 6 , Multiple Myeloma/therapy , Myelodysplastic Syndromes/genetics , Female , Humans , In Situ Hybridization, Fluorescence , Karyotyping , Middle Aged , Multiple Myeloma/complications
3.
Br J Haematol ; 123(1): 66-71, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14510944

ABSTRACT

A cohort of 121 patients with B-cell chronic lymphocytic leukaemia (B-CLL) was investigated for a single nucleotide polymorphism in the P2X7 receptor gene (1513 A-->C), and the findings were correlated with clinical prognostic markers, in vitro sensitivity to fludarabine, expression of Bcl-2 family proteins and overall survival. The frequency of the polymorphism in B-CLL samples was not significantly different from that found in normal healthy controls (P = 0.27; Fisher's exact test). Furthermore, when the B-CLL patients were analysed according to P2X7 genotype (1513 A/A versus 1513 A/C), there was no significant difference in age at diagnosis, stage at diagnosis, lymphocyte doubling time, time to first treatment, progression-free survival and overall survival, and neither was there any evidence of bias in terms of VH gene mutational status, CD38 expression, in vitro sensitivity to fludarabine or expression of Bcl-2, Bax or Mcl-1 between the two groups. These results indicate that the 1513 A-->C polymorphism of the P2X7 gene is unlikely to play a significant role in the pathogenesis or disease progression of B-CLL.


Subject(s)
Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Polymorphism, Genetic , Receptors, Purinergic P2/genetics , Vidarabine/analogs & derivatives , Aged , Antineoplastic Agents/therapeutic use , Case-Control Studies , Disease Progression , Disease-Free Survival , Drug Resistance, Neoplasm , Female , Gene Frequency , Genes, bcl-2 , Genetic Predisposition to Disease , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Male , Middle Aged , Receptors, Purinergic P2X7 , Vidarabine/therapeutic use
4.
Leuk Lymphoma ; 44(2): 337-42, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12688354

ABSTRACT

Flavopiridol, a synthetic flavone, has been previously shown to induce apoptosis in B-cell chronic lymphocytic leukaemia (B-CLL) cells in vitro. The apoptosis was associated with a concomitant activation of caspase-3 without evidence of dependence on functional p53 or Bcl-2 family modulation. In this study, we examined flavopiridol-induced apoptosis in terms of upstream caspase activity, cell cycle distribution and signal transduction, in order to elucidate the mechanism of action of this potent cytotoxic agent. Flavopiridol-induced apoptosis was significantly abrogated by the caspase-9 inhibitor Z-LEHD-FMK (p = 0.002; paired t-test) but was not altered by the caspase-8 inhibitor Z-IETD-FMK (p = 0.37; paired t-test). There was a concentration-dependent increase in a sub G0/G1 peak indicative of apoptotic cells but if these cells were excluded by gating no other cell cycle perturbations were observed suggesting that flavopiridol is capable of inducing apoptosis in cells in all phases of the cell cycle. Significantly, apoptosis was associated with activation of p38 MAP kinase and suppression of ERK activity (p = 0.0036 and p = 0.0048, respectively; paired t-test). These results show for the first time that flavopiridol modulates specific cellular signal transduction pathways in B-CLL cells thereby altering the balance between survival and cell death signals and providing a rationale for the p53-independent nature of flavopiridol-induced apoptosis. Further work is required to identify whether combinations of conventional chemotherapeutic drugs and novel agents like flavopiridol can be used to improve patient outcomes in the treatment of B-CLL.


Subject(s)
Apoptosis/drug effects , Flavonoids/pharmacology , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Mitogen-Activated Protein Kinases/metabolism , Piperidines/pharmacology , Antineoplastic Agents/pharmacology , Caspase 8 , Caspase 9 , Caspases/metabolism , Cell Cycle , Humans , Mitogen-Activated Protein Kinases/drug effects , Resting Phase, Cell Cycle , Signal Transduction , p38 Mitogen-Activated Protein Kinases
5.
Cell Cycle ; 2(1): 53-8, 2003.
Article in English | MEDLINE | ID: mdl-12695689

ABSTRACT

Li Fraumeni syndrome (LFS) is characterised by a predisposition to the early onset of certain tumors and is associated with germline mutation of the anti-oncogene p53. In this study we analysed the in vitro responses of lymphocytes from two LFS patients to chemotherapeutic drugs in terms of apoptosis induction and the expression of key intracellular proteins that regulate this process. One of the LFS patients also suffered from B-cell chronic lymphocytic leukemia (B-CLL) and hence presented with a light-chain restricted B-cell lymphocytosis while the other patient had entirely normal blood counts. The B-lymphocytes from both LFS patients showed a marked degree of resistance to chlorambucil and fludarabine when compared to age-matched controls but were remarkably sensitive to the novel flavone, flavopiridol. Loss of function of p53 was demonstrated by a failure to induce Bax and p21 protein expression. In addition, altered basal expression patterns of Bcl-2 and Bax, two key regulators of apoptosis, were found in the LFS lymphocytes when compared with controls. These results suggest that LFS lymphocytes carrying a p53 mutation show intrinsic resistance to conventional chemotherapeutic drugs and this is associated with dysregulation of Bcl-2 family proteins. Furthermore, The innate resistance profile was similar in leukemic and non-leukemic lymphocytes and was therefore independent of genetic changes acquired during malignant transformation. Novel agents that induce p53-independent cell killing may be useful not only in the treatment of LFS-associated tumors but also drug resistant tumors in general where p53 and/or Bcl-2 family dysregulation is a feature.


Subject(s)
Apoptosis/drug effects , B-Lymphocytes/metabolism , Drug Resistance, Neoplasm/genetics , Leukemia/metabolism , Li-Fraumeni Syndrome/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Tumor Suppressor Protein p53/deficiency , Adolescent , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/genetics , B-Lymphocytes/drug effects , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/metabolism , Female , Flavonoids/pharmacology , Flavonoids/therapeutic use , Humans , Leukemia/drug therapy , Leukemia/genetics , Li-Fraumeni Syndrome/drug therapy , Li-Fraumeni Syndrome/genetics , Middle Aged , Mutation/genetics , Piperidines/pharmacology , Piperidines/therapeutic use , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics , bcl-2-Associated X Protein
6.
Blood ; 101(7): 2454-60, 2003 Apr 01.
Article in English | MEDLINE | ID: mdl-12446453

ABSTRACT

EB1089, a novel vitamin D3 analog, has been shown to have cytotoxic and antiproliferative properties in a variety of malignant cells. However, its potential as a treatment for B-cell chronic lymphocytic leukemia (B-CLL) has not been evaluated. EB1089 induced apoptosis in all of the 102 B-CLL samples tested with a mean LD(50) (the concentration of EB1089 required to kill 50% of cells) value (+/- SD) of 2.1 x 10(-8) M (+/- 1.4 x 10(-8) M). Furthermore, no significant difference was found in the cytotoxicity of EB1089 in B-CLL samples from previously treated and untreated patients (P =.1637). Induction of apoptosis was associated with a reduction in Bcl-2 and Mcl-1 protein expression, but this was evident only in the apoptotic cells. In contrast, the expression of Bax, p21, and p53 was not altered in the viable or apoptotic cells from either B- or T-lymphocyte lineages. EB1089-induced apoptosis was preceded by activation of p38 mitogen-activated protein (MAP) kinase and suppression of extracellular signal-regulated kinase (ERK) activity, and this was associated with downstream activation of caspase-3. The pancaspase inhibitor (Z-VAD-FMK) and the caspase-9 inhibitor (Z-LEHD-FMK) were able to partially abrogate the apoptotic effects of EB1089 but did not affect the phosphorylation of p38 MAP kinase or the suppression of ERK. The B-CLL cells in the study were shown to highly express vitamin D receptor, but an additional receptor-independent mechanism of cell killing cannot be ruled out at this stage. These findings show that EB1089 is a potent apoptosis-inducing agent in B-CLL cells and may be useful in the treatment of B-CLL patients, particularly those with p53 mutations or drug-resistant disease.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Calcitriol/analogs & derivatives , Calcitriol/pharmacology , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Mitogen-Activated Protein Kinases/metabolism , Antineoplastic Agents/administration & dosage , Calcitriol/administration & dosage , Case-Control Studies , Cell Cycle/drug effects , Dose-Response Relationship, Drug , Enzyme Activation , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Mitogen-Activated Protein Kinases/physiology , Proto-Oncogene Proteins c-bcl-2/drug effects , Tumor Suppressor Protein p53/drug effects , p38 Mitogen-Activated Protein Kinases
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.
Leuk Lymphoma ; 43(10): 2003-9, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12481899

ABSTRACT

Our previous work demonstrated that antisense oligonucleotides complementary to Bcl-2 mRNA sequences were able to reduce Bcl-2 protein expression in B-cell chronic lymphocytic leukaemia (B-CLL) cells. Furthermore, the reduction in Bcl-2 expression led to an increase in apoptotic cell death that was associated with an increase in Bax expression. In this present study antisense oligonucleotides directed towards the Bax translation initiation sequence were employed to determine whether Bcl-2 and Bax protein expression were interdependent upon one another and whether Bcl-2 antisense-induced apoptosis was mediated through a p53-dependent cell death pathway. The antisense oligonucleotide down-regulated Bax protein expression between 23.7 and 68.8% in the 20 B-CLL samples tested and significantly inhibited apoptotic cell death when compared to controls (p = 0.0001). Furthermore, these changes were independent of Bcl-2 expression suggesting that the previously observed increase in Bax expression following exposure of B-CLL cells to Bcl-2 antisense oligonucleotides was probably caused by the induction of apoptosis rather than a rheostatic response to the reduction in Bcl-2 protein expression. This notion was substantiated by the fact that p21 expression was induced in Bcl-2 antisense-treated B-CLL cells, a finding consistent with p53 activation.


Subject(s)
Apoptosis/drug effects , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Oligonucleotides, Antisense/pharmacology , Proto-Oncogene Proteins c-bcl-2/physiology , Proto-Oncogene Proteins/genetics , Flow Cytometry , Humans , Lymphocytes/drug effects , Proto-Oncogene Proteins/physiology , RNA, Messenger/antagonists & inhibitors , Tumor Suppressor Protein p53/metabolism , bcl-2-Associated X Protein
9.
Eur J Haematol ; 69(4): 227-35, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12431242

ABSTRACT

The aim of this study was to investigate the effects of all-trans retinoic acid (ATRA) on apoptosis induction, Bcl-2 family protein expression, and differentiation in B-cell chronic lymphocytic leukaemia (B-CLL) cells. ATRA induced apoptosis in all the B-CLL samples tested, and this was accompanied by a specific reduction in Bcl-2 and Mcl-1 protein expression in the apoptotic cells. In contrast, Bax, p21, and p53 expression was not altered in either the viable or apoptotic B-CLL cells, inferring that ATRA utilises a p53-independent cell death pathway. Caspase-3 activation was shown to be a prerequisite for ATRA-induced apoptosis, which was inhibited by the pan-caspase inhibitor Z-VAD-FMK and the caspase-9 inhibitor Z-LEHD-FMK. In addition, the retinoic acid receptor (RAR) antagonist AGN194310 failed to abrogate the apoptotic effects of ATRA, indicating that RAR binding was not necessary for ATRA-induced apoptosis. Furthermore, there was no evidence of ATRA-induced differentiation of the B-CLL cells in this study either in terms of altered morphology or immunophenotype. In summary these data indicate that ATRA induces apoptosis via the intrinsic apoptotic pathway, and this is independent of RAR binding, p53 activation, and cellular differentiation in B-CLL cells.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Caspases/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Tretinoin/pharmacology , Antineoplastic Agents/therapeutic use , Caspase 3 , Cell Differentiation/drug effects , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptors, Retinoic Acid/metabolism , Signal Transduction/drug effects , Tretinoin/therapeutic use , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism
10.
Leuk Res ; 26(11): 997-1005, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12363468

ABSTRACT

We recorded elevated numbers of circulating myeloid and erythroid colony-forming cells in 15 adult patients with acute myeloid leukaemia (AML) who presented with high blood white cell counts. Since leukaemic blasts from three of these patients were Philadelphia chromosome-positive (Ph+), we were able to determine if blood progenitors from these particular patients arose from the leukaemic clone or from residual normal progenitors. Blasts and colonies were intensively investigated using a combination of cell surface marker analysis by flow cytometry, RT-PCR and interphase fluorescence in situ hybridization (FISH). FISH detected rearrangements within the major breakpoint BCR (M-BCR) region in blasts and in some myeloid and erythroid colonies from patients 1 and 2. The minor breakpoint (m-BCR) region was detected in blasts and in some myeloid and erythroid colonies from patient 3. RT-PCR detected long b2a2 BCR-ABL transcripts in blasts from patients 1 and 2, although misspliced short e1a2 transcripts were also seen in patient 1. Only e1a2 transcripts were found in blasts from patient 3. Flow sorting demonstrated the B-cell marker CD19 on blasts and on a proportion of myeloid and erythroid progenitors from patients 1 and 3. RT-PCR also detected IgH rearrangements, further evidence of B-cell differentiation, in blasts from these two patients. We conclude that both normal and clonal circulating progenitor numbers can be raised in both M-BCR and m-BCR Ph+ AML. The underlying cause, perhaps efflux from a congested marrow, may be common to AML patients with a high blood white cell count.


Subject(s)
Erythroid Precursor Cells/metabolism , Fusion Proteins, bcr-abl/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Myeloid Progenitor Cells/metabolism , Protein-Tyrosine Kinases , Proto-Oncogene Proteins , Acute Disease , Adult , Biomarkers, Tumor , Case-Control Studies , Colony-Forming Units Assay , DNA, Neoplasm/analysis , Flow Cytometry , Fusion Proteins, bcr-abl/blood , Genes, Immunoglobulin/genetics , Humans , In Situ Hybridization, Fluorescence , Karyotyping , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukocyte Count , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcr , RNA, Messenger/genetics , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Reverse Transcriptase Polymerase Chain Reaction
11.
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
...