Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
2.
Free Radic Res ; 47(1): 20-34, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23009217

ABSTRACT

The concept of reactive oxygen species (ROS) being produced via the activation of specific oncogenes provides a basis for generating genomic instability and pro-survival signalling in tumour cells. The purpose of this study was to identify downstream targets of NADPH oxidase (Nox)-derived ROS signalling in acute myeloid leukaemia cells, by performing a proteomic analysis utilizing two-dimensional phosphotyrosine immunoblotting. The majority of the targets identified were cytoskeletal-associated proteins including Ezrin, a known regulator of the cytoskeleton, which was examined further. The study demonstrated that inhibition of Nox enzymes, using diphenyleneiodonium chloride in the acute myeloid leukaemia cell line MOLM-13, resulted in a decrease in Ezrin tyrosine phosphorylation and also triggered a shift in Ezrin sub-cellular localization as detected by immunofluorescence. The change in Ezrin localization coincided with altered cell morphology, observed using scanning electron microscopy and a decreased ability to migrate through a polycarbonate transwell membrane. Similar effects were observed upon inhibition of the oncogenic receptor tyrosine kinase FLT3 using the staurosporine derivate PKC412, implicating a role for FLT3 as an upstream regulator of Ezrin. Our results indicate that FLT3 drives production of ROS by Nox, which stimulates changes in Ezrin tyrosine phosphorylation and localization via redox regulation of Src. Furthermore, inhibition of FLT3 signalling leads to alterations in MOLM-13 cell morphology and has a significant influence on cell motility.


Subject(s)
Cell Movement/physiology , Cytoskeletal Proteins/metabolism , Leukemia, Myelomonocytic, Acute/enzymology , Leukemia, Myelomonocytic, Acute/pathology , fms-Like Tyrosine Kinase 3/metabolism , Cell Line , Cell Movement/drug effects , Cytoskeletal Proteins/genetics , HL-60 Cells , Humans , Leukemia, Myelomonocytic, Acute/genetics , Leukemia, Myelomonocytic, Acute/metabolism , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/metabolism , Onium Compounds/pharmacology , Oxidation-Reduction , Phosphorylation , Reactive Oxygen Species/metabolism , Signal Transduction , Staurosporine/analogs & derivatives , Staurosporine/pharmacology , Transfection , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , fms-Like Tyrosine Kinase 3/genetics
3.
Biochem Pharmacol ; 80(10): 1507-16, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20705060

ABSTRACT

Aberrant signal transduction by mutant or overexpressed protein kinases has emerged as a promising target for treatment of acute myeloid leukemia (AML). We here present a novel low molecular weight kinase inhibitor, AKN-032, targeting the FMS-like tyrosine kinase 3 (FLT3) and discovered in a new type of screening funnel combining the target therapy approach with sequential cellular screens. AKN-032 was identified among 150 selected hits from three different high throughput kinase screens. Further characterization showed inhibitory activity on FLT3 enzyme with an IC(50) of 70 nM. Western blot analysis revealed reduced autophosphorylation of the FLT3-receptor in AML cell line MV4-11 cells after exposure to AKN-032. Flow cytometry disclosed cytotoxic activity against MV4-11, but not against non-malignant 3T3-L1 fibroblast cells. Using a fluorometric microculture cytotoxicity assay, AKN-032 was tested against 15 cell lines and displayed a potent cytotoxic activity in AML cell lines MV4-11 (IC(50)=0.4 µM) and Kasumi-1 (IC(50)=2.3 µM). AKN-032 was also highly cytotoxic in tumor cells from AML patients in vitro. Furthermore, AKN-032 demonstrated significant antileukemic effect in a relatively resistant in vivo hollow fiber mouse model. No major toxicity was observed in the animals. In conclusion, AKN-032 is a promising new kinase inhibitor with significant in vivo and in vitro activity in AML. Results from the hollow fiber mouse assay suggest a favorable toxicity profile. Future studies will focus on pharmacokinetic properties, toxicity as well as further clarifying the mechanisms of action of AKN-032 in AML.


Subject(s)
Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Leukemia, Myelomonocytic, Acute/drug therapy , Pyrazines/chemistry , Pyrazines/therapeutic use , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , Adult , Aged , Animals , Antineoplastic Agents/adverse effects , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Epithelial Cells/drug effects , Epithelial Cells/enzymology , Female , Flow Cytometry , Humans , Leukemia, Myelomonocytic, Acute/enzymology , Leukemia, Myelomonocytic, Acute/pathology , Male , Mice , Middle Aged , Molecular Structure , Pyrazines/adverse effects , Pyrazines/pharmacology , Xenograft Model Antitumor Assays , Young Adult
4.
Ann Hematol ; 88(11): 1089-97, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19296110

ABSTRACT

Impact of FLT3 receptor tyrosine kinase activation via internal tandem duplication (ITD) of the juxtamembrane region on outcome of acute myeloid leukemia (AML) is still controversial. Recent researches reveal a role of FLT3 in monocyte differentiation in hematopoiesis. We analyzed the clinical impact of FLT3 alterations in adult AML patients excluding acute promyelocytic leukemia (APL) who received induction chemotherapy according to morphologic classification. Retrospective review of medical records from three centers in Korea between 1997 and 2007 was performed. Polymerase chain reaction was performed on genomic DNA derived from blood samples of patients before induction chemotherapy for FLT3-ITD detection. We assessed overall survival (OS), first disease-free survival (1-DFS), and response to induction chemotherapy. One hundred eighty-four patients (median age 49.1 years, range 16.0-76.5) with AML excluding APL received induction chemotherapy from three centers. FLT3-ITD was detected in 22 patients. One hundred forty-one patients were below age 60. One hundred seventy-nine patients received induction chemotherapy with cytarabine and idarubicin (AId) regimen. One hundred nineteen patients achieved complete remission (CR) after first induction chemotherapy. FLT3-ITD was not related to achievement of CR. 1-DFS was longer in patients without FLT3-ITD (median 1-DFS 16.5 vs. 8.5 months, p = 0.025). 1-DFS was not different according to FLT3-ITD status in nonmonocyte lineage leukemia (p = 0.355), while 1-DFS was shorter in monocyte lineage leukemia for FLT3-ITD positive patients (20.9 vs. 2.4 months, p < 0.001). FLT3-ITD had no impact on OS except for monocyte lineage, where OS was significantly shorter in FLT3-ITD positive group (39.4 vs. 6.0 months, p = 0.026). Moreover FLT3-ITD was stronger prognostic factors in monocyte lineage AML than risk stratification based on cytogenetics. Status of FLT3-ITD should be analyzed differently in AML patients according to morphologic profile. FLT3-ITD is a predictive and prognostic marker only in monocyte lineage patients. This result suggests an existence of distinct subset of monocyte lineage AML with leukemogenesis involving FLT3 activating pathway.


Subject(s)
Leukemia, Monocytic, Acute/genetics , Leukemia, Myeloid/classification , Leukemia, Myelomonocytic, Acute/genetics , Monocytes/pathology , Myelopoiesis/genetics , Tandem Repeat Sequences , fms-Like Tyrosine Kinase 3/genetics , Adolescent , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Lineage , Cell Transformation, Neoplastic/genetics , Disease-Free Survival , Exons/genetics , Female , Humans , Introns/genetics , Kaplan-Meier Estimate , Korea/epidemiology , Leukemia, Monocytic, Acute/drug therapy , Leukemia, Monocytic, Acute/enzymology , Leukemia, Monocytic, Acute/mortality , Leukemia, Myelomonocytic, Acute/drug therapy , Leukemia, Myelomonocytic, Acute/enzymology , Leukemia, Myelomonocytic, Acute/mortality , Male , Middle Aged , Prognosis , Protein Structure, Tertiary , Young Adult , fms-Like Tyrosine Kinase 3/analysis , fms-Like Tyrosine Kinase 3/chemistry
5.
Stem Cells ; 24(5): 1174-84, 2006 May.
Article in English | MEDLINE | ID: mdl-16410383

ABSTRACT

Ligand-mediated activation of the FMS-like tyrosine kinase 3 (FLT3) receptor is important for normal proliferation of primitive hematopoietic cells. However, activating mutations in FLT3 induce ligand-independent downstream signaling that promotes oncogenesis through pathways involved in proliferation, differentiation, and survival. FLT3 mutations are identified as the most frequent genetic abnormality in acute myeloid leukemia and are also observed in other leukemias. Multiple small-molecule inhibitors are under development to target aberrant FLT3 activity that confers a poor prognosis in patients.


Subject(s)
Hematopoiesis , Leukemia, Myelomonocytic, Acute/enzymology , Leukemia, Myelomonocytic, Acute/genetics , fms-Like Tyrosine Kinase 3/genetics , Clinical Trials as Topic , Humans , Mutation , Protein Kinase Inhibitors/pharmacology , Signal Transduction , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , fms-Like Tyrosine Kinase 3/metabolism
6.
Histopathology ; 48(3): 247-57, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16430471

ABSTRACT

AIMS: To analyse and characterize the ultrastructural morphology of normal tissue mast cells (MC) and neoplastic bone marrow MC. METHODS: We have examined the ultrastructure and cytomorphological features of MC derived from cord blood cells, neoplastic bone marrow MC in patients with systemic mastocytosis (SM, n = 4), myelomastocytic leukaemia (MML, n = 2), mast cell leukaemia (MCL, n = 2) and tryptase-positive acute myeloid leukaemia (AML, n = 4). RESULTS: Based on their ultrastructure and morphology, four distinct cell types could be delineated: (i) mature well-granulated tissue MC exhibiting a round central nucleus; (ii) atypical MC type I with oval nuclei, hypogranulated cytoplasm, and prominent surface projections; (iii) immature atypical MC with bi- or polylobed nuclei (atypical MC type II = promastocytes); and (iv) metachromatic blasts. Type I atypical MC were detected in a patient with indolent SM, whereas type II MC and metachromatic blasts were primarily found in MML, MCL and tryptase-positive AML. In all samples examined, the identity of MC could be reconfirmed by immunoelectron microscopy, irrespective of the stage of cell maturation or the disease variant, all types of MC contained tryptase in their cytoplasmic granules. CONCLUSION: Immunoelectron microscopy may be a helpful approach in confirming the identity of neoplastic MC in myeloid neoplasms.


Subject(s)
Leukemia, Myeloid/pathology , Leukemia, Myelomonocytic, Acute/pathology , Mast Cells/enzymology , Mast Cells/ultrastructure , Mastocytosis, Systemic/pathology , Serine Endopeptidases/analysis , Adult , Aged , Cell Nucleus/ultrastructure , Cytoplasm/enzymology , Cytoplasm/ultrastructure , Female , Fetal Blood/cytology , Humans , Leukemia, Mast-Cell/enzymology , Leukemia, Mast-Cell/pathology , Leukemia, Myeloid/enzymology , Leukemia, Myelomonocytic, Acute/enzymology , Male , Mast Cells/pathology , Mastocytosis, Systemic/enzymology , Microscopy, Electron , Microscopy, Immunoelectron , Middle Aged , Tryptases
7.
Cancer Res ; 65(7): 2890-8, 2005 Apr 01.
Article in English | MEDLINE | ID: mdl-15805291

ABSTRACT

We investigated the antileukemic activity and molecular mechanisms of action of a newly synthesized ring-substituted diindolylmethane derivative, 1,1-bis[3'-(5-methoxyindolyl)]-1-(p-t-butylphenyl) methane (DIM #34), in acute myelogenous leukemia (AML) cells. DIM #34 inhibited AML cell growth via the induction of apoptosis and abrogated clonogenic growth of primary AML samples. Exposure to DIM #34 induced loss of mitochondrial inner transmembrane potential, release of cytochrome c into the cytosol, and caspase activation. Bcl-2-overexpressing, Bax knockout, and caspase-9-deficient cells were partially resistant to cell death, suggesting the involvement of the intrinsic apoptotic pathway. Furthermore, DIM #34 transiently inhibited the phosphorylation and activity of the extracellular signal-regulated kinase and abrogated Bcl-2 phosphorylation. Because other methylene-substituted diindolylmethane analogues have been shown to transactivate the nuclear receptor peroxisome proliferator-activated receptor gamma (PPARgamma), we studied the role of PPARgamma in apoptosis induction. Cotreatment of cells with a selective PPARgamma antagonist or with retinoid X receptor and retinoic acid receptor ligands partially modulated apoptosis when combined with DIM #34, suggesting PPARgamma receptor-dependent and receptor-independent cell death. Together, these findings suggest that diindolylmethanes are a new class of compounds that selectively induce apoptosis in AML cells through the modulation of the extracellular signal-regulated kinase and PPARgamma signaling pathways.


Subject(s)
Apoptosis/drug effects , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Indoles/pharmacology , Leukemia, Myeloid/drug therapy , Protein Kinase Inhibitors/pharmacology , Acute Disease , Apoptosis/physiology , Caspases/metabolism , Enzyme Activation/drug effects , HL-60 Cells , Humans , Jurkat Cells , Leukemia, Monocytic, Acute/drug therapy , Leukemia, Monocytic, Acute/enzymology , Leukemia, Monocytic, Acute/pathology , Leukemia, Myeloid/enzymology , Leukemia, Myeloid/pathology , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/enzymology , Leukemia, Myeloid, Acute/pathology , Leukemia, Myelomonocytic, Acute/drug therapy , Leukemia, Myelomonocytic, Acute/enzymology , Leukemia, Myelomonocytic, Acute/pathology , MAP Kinase Signaling System/drug effects , PPAR gamma/metabolism , Proto-Oncogene Proteins c-bcl-2/physiology , U937 Cells
9.
Malays J Pathol ; 26(2): 111-6, 2004 Dec.
Article in English | MEDLINE | ID: mdl-16329563

ABSTRACT

INTRODUCTION: Myeloperoxidase (MPO) is present in azurophilic granules which appear in the promyelocyte stage of differentiation and is expressed in granulomonocytic cells. MPO is usually detected by cytochemistry. The demonstration of peroxidase in at least 3% of bone marrow blasts defines an acute leukaemia as acute myeloblastic leukaemia (AML). MPO is important in distinguishing acute myeloblastic leukaemia (AML) from acute lymphoblastic leukaemia (ALL). It is difficult to diagnose AML with minimal evidence of myeloid differentiation (AML- M0) by conventional light microscopy. However, these AML-M0 blasts can be detected by monoclonal antibodies. Anti-MPO recognizes the enzymatically inactive precursor forms of MPO. There are a few commercially available monoclonal antibodies against MPO. In this study, we evaluated two monoclonal antibodies against MPO from different commercial sources. METHODS: Anti-MPO were purchased from Dako (Denmark) and Becton Dickinson, BD (California, USA). MPO detection was done using the permeabilisation-staining technique, followed by analysis with flow cytometer (FASCalibur, California, USA). RESULTS: 63 cases of acute leukaemias (38 ALL and 25 AML) were studied. Anti-MPO by Dako showed that 12/38 (31.6%) of ALL cases were positive, but all these cases were clear-cut negative for anti-MPO from BD. 24/38 (63.2%) of these ALL cases were associated with aberrant expression of myeloid antigens. However, only 8/24 (33.3%) cases with aberrant myeloid antigen expression showed positive reaction to anti-MPO (Dako). 23/25 (92%) of AML showed concordance results for both anti-MPO by Dako and BD. CONCLUSION: Anti-MPO is a useful and reliable marker for the diagnosis of AML. However, this study had demonstrated that results vary with the monoclonal antibody used in ALL cases. Anti-MPO (Dako) had shown false positive result in 31.6% of ALL cases whereas anti-MPO (BD) had shown consistent negative result in ALL cases.


Subject(s)
Antibodies, Monoclonal , Flow Cytometry/methods , Leukemia, Myelomonocytic, Acute/diagnosis , Peroxidase/analysis , Precursor Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Cloning, Molecular , Cross-Sectional Studies , Humans , Leukemia, Myelomonocytic, Acute/enzymology , Peroxidase/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/enzymology
10.
Hematol J ; 4(1): 26-30, 2003.
Article in English | MEDLINE | ID: mdl-12692517

ABSTRACT

INTRODUCTION: Classically, the monocytic component of acute myelomonocytic (FAB-M4) and acute monocytic/monoblastic (FAB-M5) leukemias is demonstrated by nonspecific esterase positivity in cytochemical stainings. We have previously demonstrated that non-specific esterases from normal monocytes can be determined by a chemiluminescent method. In the present study, we investigated whether this assay can also determine the monocytic component of FAB-M4 and FAB-M5 and distinguish these acute myeloid leukemia (AML) categories. MATERIALS AND METHODS: Bone marrow samples were obtained from 66 patients with AML (M0, two cases; M1, 12 cases; M2, 13 cases; M3, 10 cases; M4, 11 cases; M5, 12 cases; M6, two cases; M7, four cases). Cells were incubated with a standard reaction mixture and chemiluminescence was measured for 10 min. Two parameters were assessed, the peak (PLE) and the integrated light emission (ILE). RESULTS: Both PLE and ILE were higher in FAB-M4 and FAB-M5 subtypes compared to other AML subtypes (P<0.001). In addition, the classification of AML cases into FAB-M4, FAB-M5 and nonmonocytic subtypes based on ILE analysis was concordant with alpha-naphthyl acetate esterase (ANAE) in 97% of cases (kappa coefficient 0.94, P<0.001). CONCLUSIONS: These findings indicate that this chemiluminescent assay was able to determine the monocytic component of FAB-M4 and FAB-M5 cells, and the classification of AML subtypes based on chemiluminescent analysis strongly agreed with the cytochemical ANAE-staining. In conclusion, this chemiluminescent assay is a simple, fast and objective method, which may be useful as an alternative tool in the differential diagnosis of AML subtypes.


Subject(s)
Bone Marrow/pathology , Leukemia, Monocytic, Acute/pathology , Leukemia, Myelomonocytic, Acute/pathology , Luminescent Measurements , Monocytes/pathology , Neoplastic Stem Cells/pathology , Acute Disease , Adolescent , Adult , Aged , Aged, 80 and over , Benzoates/metabolism , Carboxylesterase , Carboxylic Ester Hydrolases/analysis , Child , Child, Preschool , Cryopreservation , Diagnosis, Differential , Female , Horseradish Peroxidase/metabolism , Humans , Leukemia, Monocytic, Acute/diagnosis , Leukemia, Monocytic, Acute/enzymology , Leukemia, Myeloid/pathology , Leukemia, Myelomonocytic, Acute/diagnosis , Leukemia, Myelomonocytic, Acute/enzymology , Male , Middle Aged , Monocytes/enzymology , Neoplasm Proteins/analysis , Neoplastic Stem Cells/enzymology , Sodium Fluoride/pharmacology
11.
Nat Genet ; 34(2): 148-50, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12717436

ABSTRACT

We report here that individuals with Noonan syndrome and juvenile myelomonocytic leukemia (JMML) have germline mutations in PTPN11 and that somatic mutations in PTPN11 account for 34% of non-syndromic JMML. Furthermore, we found mutations in PTPN11 in a small percentage of individuals with myelodysplastic syndrome (MDS) and de novo acute myeloid leukemia (AML). Functional analyses documented that the two most common mutations in PTPN11 associated with JMML caused a gain of function.


Subject(s)
Leukemia, Myeloid, Acute/enzymology , Leukemia, Myeloid, Acute/genetics , Leukemia, Myelomonocytic, Acute/enzymology , Leukemia, Myelomonocytic, Acute/genetics , Mutation , Myelodysplastic Syndromes/enzymology , Myelodysplastic Syndromes/genetics , Protein Tyrosine Phosphatases/genetics , Animals , COS Cells , Child , Humans , Intracellular Signaling Peptides and Proteins , Leukemia, Myelomonocytic, Acute/complications , Noonan Syndrome/complications , Noonan Syndrome/enzymology , Noonan Syndrome/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Protein Tyrosine Phosphatases/metabolism , Transfection
12.
Clin Cancer Res ; 8(7): 2240-5, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12114426

ABSTRACT

PURPOSE: Carbonic anhydrases (CAs) are key enzymes that regulate acid-base homeostasis in both normal and pathological conditions. Recent studies have shown that they are functionally involved in the growth and invasion of cancer cells. However, there are only a few publications on CAs in hematological malignancies. EXPERIMENTAL DESIGN: Here we investigated the presence of CA isozymes in six malignant hematopoietic cell lines and malignant blast cells of bone marrow samples collected from patients with acute myeloid leukemia, acute lymphoblastic leukemia, or chronic myelomonocytic leukemia. RESULTS: Because three of the malignant hematopoietic cell lines expressed CA II, we also set out to examine its expression in a series of bone marrow samples. Positive reactions were found in 16 of 26 cases (62%) of acute myeloid leukemia, 11 of 15 cases (73%) of acute lymphoblastic leukemia, and 1 of 2 cases (50%) of chronic myelomonocytic leukemia. CONCLUSIONS: The results indicate that CA II expression is not restricted to only one cell lineage but may result from a genetic aberration that occurs in both myeloid and lymphatic lineages or in their progenitor cell. Because CA II is expressed in most patients with leukemic blast cells, CA inhibitors may prove to be of value as an adjunct to chemotherapy for such cancers.


Subject(s)
Carbonic Anhydrase II/metabolism , Leukemia, Myeloid, Acute/enzymology , Leukemia, Myelomonocytic, Acute/enzymology , Leukemia, Myelomonocytic, Chronic/enzymology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/enzymology , Acute Disease , Anemia, Refractory, with Excess of Blasts/metabolism , Antigens, CD34/metabolism , Bone Marrow/metabolism , Follow-Up Studies , Humans , Immunoenzyme Techniques , Tumor Cells, Cultured
13.
Anticancer Drugs ; 12(5): 441-51, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11395572

ABSTRACT

In order to investigate the mechanisms of drug resistance arising in tumor cells, we investigated the capacity of fluoroquinolones to inhibit the in vitro growth of WEHI-3B monomyelocytic leukemia cells and then we established a variant of this line (currently maintained in the absence of drug). The line, named WEHI-3B/CPX, expresses a specific resistance to ciprofloxacin (CPX; resistance index=17.3+/-2.2), and does not show cross-resistance with other fluoroquinolones, camptothecin and topoisomerase II inhibitors such as doxorubicin, etoposide and teniposide. Although a little decrease in intracellular accumulation of CPX is observed in WEHI-3B/CPX cells, these cells do not express MDR or LRP markers, and the resistance is not circumvented by verapamil. Purified nuclear extracts from WEHI-3B and WEHI-3B/CPX cells were tested for topoisomerase I catalytic activity and checking in vitro topoisomerase I sensitivity to CPX and camptothecin inhibition, but no difference was observed. As the treatment with CPX showed that the resistant cell line suffers a significantly lower number of breaks in the DNA molecule we also addressed our investigations to the topoisomerase II-dependent DNA cleavage that, in the resistant clone, was found dramatically less susceptible to be enhanced by CPX both in pre-strand and post-strand DNA passage conditions. WEHI-3B/CPX cells do not express any character of multidrug resistance and represent a rare case of specific drug resistance to CPX. The specific resistance to CPX observed in these cells is related to a functional decrease of topoisomerase II cleavage activity. It could be consequent to a decreased binding affinity of CPX for the topoisomerase II--DNA complex or to a decreased affinity or specificity of topoisomerase II for its DNA cleavage sites.


Subject(s)
Anti-Infective Agents/pharmacology , Ciprofloxacin/pharmacology , DNA Topoisomerases, Type II/metabolism , DNA, Neoplasm/metabolism , Leukemia, Myelomonocytic, Acute/enzymology , Tumor Cells, Cultured/drug effects , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Camptothecin/pharmacology , Cell Division/drug effects , Drug Resistance, Neoplasm , Tumor Cells, Cultured/enzymology , Tumor Cells, Cultured/pathology
14.
Blood ; 97(9): 2830-8, 2001 May 01.
Article in English | MEDLINE | ID: mdl-11313278

ABSTRACT

Treatment of different human leukemia cell variants with the anthracycline adriamycin was associated with a rapid activation of the proteasome. Thus, proliferating U937, TUR, and retrodifferentiated U937 cells exhibited a 4.3-fold, 5.8-fold, and 4.3-fold proteasome activation within 15 minutes after adriamycin treatment, respectively. In contrast, little if any proteasome activation was detectable in a growth-arrested differentiated U937 population following adriamycin treatment. Further analysis of this mechanism revealed a significant reduction of adriamycin-induced proteasome activity after inhibition of poly(ADP-ribose) polymerase (PARP) by 3-aminobenzamide (3-ABA) in the proliferating leukemic cell types. These findings suggested that PARP is involved in the regulation of drug-induced proteasome activation. Indeed, anti-PARP immunoprecipitation experiments of adriamycin-treated cells revealed increasing levels of coprecipitated, enzymatically active proteasome particularly in the proliferating cell variants in contrast to the differentiated U937 cells, with a maximum after 15 minutes, and sensitivity to PARP inhibition by 3-ABA. The specific role of the PARP was investigated in U937 and TUR cell clones stably transfected with a constitutively active antisense PARP (asPARP) vector. Thus, asPARP-TUR cells developed a 25-fold increased sensitivity to adriamycin treatment. Furthermore, we investigated leukemic blasts isolated from acute myelogenous leukemia patients and obtained a similarly enhanced proteasome activity after adriamycin treatment, which was dependent on the PARP and thus could be coprecipitated with anti-PARP antibodies. Transient transfection of leukemic blasts with the asPARP vector significantly reduced the adriamycin-induced proteasome activation. These data suggest that the PARP-associated nuclear proteasome activation represents a potential target within chemotherapeutic defense mechanisms developed by leukemia cells.


Subject(s)
Antineoplastic Agents/pharmacology , Cysteine Endopeptidases/metabolism , Doxorubicin/pharmacology , Leukemia, Myelomonocytic, Acute/drug therapy , Leukemia, Myelomonocytic, Acute/enzymology , Multienzyme Complexes/metabolism , Antineoplastic Agents/therapeutic use , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Doxorubicin/therapeutic use , Enzyme Activation/drug effects , Humans , Oligonucleotides, Antisense , Proteasome Endopeptidase Complex , Transfection , U937 Cells
15.
Cancer Res ; 61(1): 2-7, 2001 Jan 01.
Article in English | MEDLINE | ID: mdl-11196162

ABSTRACT

Histone deacetylase (HDAC)-dependent transcriptional repression of the retinoic acid (RA)-signaling pathway underlies the differentiation block of acute promyelocytic leukemia. RA treatment relieves transcriptional repression and triggers differentiation of acute promyelocytic leukemia blasts, leading to disease remission. We report that transcriptional repression of RA signaling is a common mechanism in acute myeloid leukemias (AMLs). HDAC inhibitors restored RA-dependent transcriptional activation and triggered terminal differentiation of primary blasts from 23 AML patients. Accordingly, we show that AML1/ETO, the commonest AML-associated fusion protein, is an HDAC-dependent repressor of RA signaling. These findings relate alteration of the RA pathway to myeloid leukemogenesis and underscore the potential of transcriptional/differentiation therapy in AML.


Subject(s)
Antineoplastic Agents/pharmacology , Histone Deacetylase Inhibitors , Leukemia, Myeloid, Acute/enzymology , Leukemia, Myeloid, Acute/pathology , Leukemia, Myelomonocytic, Acute/enzymology , Leukemia, Myelomonocytic, Acute/pathology , Signal Transduction/drug effects , Tretinoin/pharmacology , Acetylation , Cell Differentiation/drug effects , Cell Differentiation/physiology , Core Binding Factor Alpha 2 Subunit , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Leukemic/drug effects , Histone Deacetylases/physiology , Histones/metabolism , Humans , Hydroxamic Acids/pharmacology , Leukemia, Myeloid, Acute/genetics , Leukemia, Myelomonocytic, Acute/genetics , Oncogene Proteins, Fusion/biosynthesis , Oncogene Proteins, Fusion/genetics , RUNX1 Translocation Partner 1 Protein , Signal Transduction/physiology , Transcription Factors/biosynthesis , Transcription Factors/genetics , Transcriptional Activation/drug effects , Transcriptional Activation/physiology , Tretinoin/physiology , Tumor Cells, Cultured
16.
Blood ; 90(7): 2730-7, 1997 Oct 01.
Article in English | MEDLINE | ID: mdl-9326240

ABSTRACT

Myeloperoxidase (MPO) catalyzes a reaction between chloride and hydrogen peroxide to generate hypochlorous acid and other reactive compounds that have been linked to DNA damage. The MPO gene is expressed at high levels in normal myeloid precursors and in acute myeloid leukemias (AMLs) which are clonal derivatives of myeloid precursors that have lost the ability to differentiate into mature blood cells. Two MPO alleles differ at -463 G/A within a cluster of nuclear receptor binding sites in an Alu element. The -463 G creates a stronger SP1 binding site and retinoic acid (RA) response element (RARE) in the allele termed Sp. In this study, we investigate potential links between MPO genotype, MPO expression level, and myeloid leukemia. The SpSp MPO genotype is shown to correlate with increased MPO mRNA levels in primary myeloid leukemia cells. This higher-expressing SpSp genotype is further shown to be overrepresented in acute promyelocytic leukemia-M3 (APL-M3) and AML-M4, suggesting that higher levels of MPO are associated with an increased risk for this subset of leukemias.


Subject(s)
Leukemia, Myelomonocytic, Acute/etiology , Leukemia, Promyelocytic, Acute/etiology , Peroxidase/physiology , Alleles , Binding Sites , Cell Transformation, Neoplastic/genetics , DNA Damage , Disease Susceptibility , Female , Gene Frequency , Genotype , Humans , Leukemia, Myelomonocytic, Acute/enzymology , Leukemia, Myelomonocytic, Acute/genetics , Leukemia, Promyelocytic, Acute/enzymology , Leukemia, Promyelocytic, Acute/genetics , Male , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Peroxidase/biosynthesis , Peroxidase/genetics , Polymerase Chain Reaction , RNA, Messenger/biosynthesis , RNA, Neoplasm/biosynthesis , Risk , Tretinoin/pharmacology
17.
Leukemia ; 11(4): 479-84, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9096686

ABSTRACT

Mitogen-activated protein (MAP) kinase appears to be one of the key regulators of cell proliferation and differentiation. Very little, however, has been revealed as to how MAP kinase is involved in leukemogenesis. We have studied the activation of the MAP kinase pathway in 100 human primary leukemia cells including 73 acute myelogenous leukemias (AMLs). Forty acute leukemia samples (40% of the total), including 37 AML samples (51% of AML), showed activation of MAP kinase as revealed by the mobility shift of the phosphorylated form of the protein and by in vitro kinase assay. This activation was correlated with MAP kinase kinase activity in these cells. In contrast, none of 14 chronic myelogenous leukemia samples showed the activation of MAP kinase. These results suggest that the MAP kinase pathway is constitutively activated in a subset of primary acute leukemias, and thus indicate the possible role of the constitutively activated MAP kinase in leukemogenesis.


Subject(s)
Genes, ras/physiology , Leukemia/enzymology , Mitogen-Activated Protein Kinases , Protein Kinases/physiology , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Enzyme Activation , Humans , Leukemia, Myelomonocytic, Acute/enzymology , Mitogen-Activated Protein Kinase 1 , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinase Kinases , Phosphorylation , Protein Kinases/genetics , Tumor Cells, Cultured
18.
J Chemother ; 7(3): 224-9, 1995 Jun.
Article in English | MEDLINE | ID: mdl-7562019

ABSTRACT

High levels of O6-alkylguanine-DNA-alkyltransferase (OGAT) can, at least in part, account for tumor cell resistance to O6-alkylguanine alkylating agents, including triazene compounds. A pilot clinical study indicates that dacarbazine can induce a marked decrease of leukemic blasts in patients affected by acute myelogenous leukemia (AML) with low pretreatment levels of OGAT activity. In this study we show a synergistic antitumor effect between cisplatin (CDDP) and temozolomide (an in vitro active analog of dacarbazine), following combined in vitro treatment of leukemic blasts. Synergistic effect appears to be CDDP-dose dependent. In vivo treatment of leukemic patients with CDDP was followed by a reduction of OGAT activity in 2 out 3 cases. These data point out that CDDP could be a good candidate for depleting OGAT protein of leukemic cells, thus reversing tumor cell resistance to dacarbazine.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cisplatin/therapeutic use , Leukemia/drug therapy , Triazenes/therapeutic use , Adult , Aged , Blast Crisis/drug therapy , Blast Crisis/enzymology , Cisplatin/administration & dosage , Dacarbazine/administration & dosage , Dacarbazine/analogs & derivatives , Dacarbazine/therapeutic use , Dose-Response Relationship, Drug , Drug Synergism , Female , Humans , In Vitro Techniques , Leukemia/enzymology , Leukemia, Megakaryoblastic, Acute/drug therapy , Leukemia, Megakaryoblastic, Acute/enzymology , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/enzymology , Leukemia, Myelomonocytic, Acute/drug therapy , Leukemia, Myelomonocytic, Acute/enzymology , Male , Methyltransferases/metabolism , Middle Aged , O(6)-Methylguanine-DNA Methyltransferase , Pilot Projects , Remission Induction , Temozolomide , Treatment Outcome , Triazenes/administration & dosage , Triazenes/pharmacology
19.
Br J Haematol ; 88(2): 357-63, 1994 Oct.
Article in English | MEDLINE | ID: mdl-7803282

ABSTRACT

Using an ultrastructural immunogold method, we performed a quantitative study on cellular lysozyme (LZ) content in young normal bone marrow cells and in 14 cases of acute myeloid leukaemia (AML) of the M2, M3, M4 and M5 types. In five cases of M2 we found significantly lower LZ content than in normal promyelocytes and than in nine cases of M3, M4 and M5. In M3, M4 and M5 cells a very high LZ content was observed whereas the serum LZ activity was high in M4 and M5 and normal in M3. The intragranular LZ content was especially high in M5 and in most granules of M4 cells. The immunogold reaction (IGR) for LZ was also performed in cells previously reacted for myeloperoxidase (MPO). In M2 the granules showed definite positive MPO reactivity and low LZ density (granulocytic pattern), whereas in M5 we found high granular LZ content and weak or almost negative MPO activity (monocytic pattern). In M4 we found 'granulocytic' and 'monocytic' type of granules in the same cell. The IGR for LZ performed in post-embedded M5 cells which were previously subjected to phagocytosis of latex particles, showed granules that had moved toward the phagosome, releasing LZ without degranulation. The above findings and those showing normal serum LZ in M3 despite their high cellular LZ content, definitely indicate that only leukaemic M4 and M5 cells secrete LZ into their environment, explaining the high serum LZ observed in those leukaemias.


Subject(s)
Bone Marrow/enzymology , Leukemia, Myeloid/enzymology , Muramidase/analysis , Acute Disease , Bone Marrow/ultrastructure , Gold Colloid , Humans , Leukemia, Monocytic, Acute/enzymology , Leukemia, Myeloid/pathology , Leukemia, Myeloid, Acute/enzymology , Leukemia, Myelomonocytic, Acute/enzymology , Leukemia, Promyelocytic, Acute/enzymology , Microscopy, Electron , Peroxidase/analysis , Phagocytosis
SELECTION OF CITATIONS
SEARCH DETAIL
...