Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 145
Filter
1.
Cytopathology ; 27(6): 472-478, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27109167

ABSTRACT

OBJECTIVE: The purpose of the present study was to evaluate the reproducibility of the cytological diagnosis of endometrial lesions by the Osaki Study Group (OSG) method of new cytological diagnostic criteria using BD SurePath™ (SP)-liquid-based cytology (LBC). METHODS: This cytological classification using the OSG method consists of six categories: (i) normal endometrium (NE), (ii) endometrial glandular and stromal breakdown (EGBD), (iii) atypical endometrial cells, cannot exclude atypical endometrial hyperplasia or more (ATEC-A), (iv) adenocarcinoma including atypical endometrial hyperplasia or malignant tumour (Malignancy), (v) endometrial hyperplasia without atypia (EH) and (vi) atypical endometrial cells of undetermined significance (ATEC-US). For this study, a total 244 endometrial samplings were classified by two academic cytopathologists as follows: 147 NE cases , 36 EGBD cases , 47 Malignant cases, eight ATEC-A cases, two EH cases and four ATEC-US cases. To confirm the reproducibility of the diagnosis and to study the inter- and intra-observer agreement further, a second review round followed at 3-month intervals, which included three additional cytopathologists. RESULTS: The inter-observer agreement of NE classes improved progressively from 'good to fair' to 'excellent', with values increasing from 0.70 to 0.81. Both EGBD and Malignancy classes improved progressively from 'good to fair' to 'excellent', with values increasing from 0.62-0.63 to 0.84-0.95, respectively. The overall intra-observer agreement between the first and the second rounds was 'good to fair' to 'excellent', with values changing from 0.79 to 0.85. All kappa improvements were significant (P < 0.0001). CONCLUSION: In this study, it seemed that the use of the OSG method as the new diagnostic criteria for SP-LBC preparation, may be a valid method to improve the precision (reproducibility) of endometrial cytology.


Subject(s)
Cytodiagnosis , Endometrial Hyperplasia/diagnosis , Endometrial Neoplasms/diagnosis , Endometrium/pathology , Adult , Endometrial Hyperplasia/pathology , Endometrial Neoplasms/pathology , Female , Humans , Middle Aged , Observer Variation
2.
Cytopathology ; 27(1): 43-9, 2016 Feb.
Article in English | MEDLINE | ID: mdl-25683486

ABSTRACT

OBJECTIVES: Damage and detachment of podocytes and loss into the urine have been implicated in the progression of kidney diseases. The purpose of this study was to investigate the potential role of urine cytology based on SurePath(™) combined with immunoenzyme staining using Wilms' tumour 1 (WT1) antibody as a podocyte marker in the discrimination of normality and non-renal urinary tract disease from kidney disease. METHODS: Sixty-six patients with kidney disease, 45 patients with lower urinary tract disease and 30 healthy volunteers were examined. Urine cytology slides were prepared using the SurePath method and immunoenzyme stained with WT1 antibody, and the number of WT1-positive cells was counted. RESULTS: In kidney disease, WT1-positive cells were found in 33 (50%) of 66 samples. No WT1-positive cells were found in 45 patients with lower urinary tract disease or in 30 healthy volunteers. The positive rates for WT1 varied with disease type, but not significantly: immunoglobulin A (IgA) nephropathy, (14/23); membranous glomerulonephritis, (4/10); Henoch-Schönlein purpura nephritis, (3/5); diabetic glomerulopathy, (5/5); minor glomerular abnormality/minimal change nephrotic syndrome (0/4). CONCLUSIONS: The results suggest that WT1 immunoenzyme staining of urine cytology can be used to detect some types of kidney disease.


Subject(s)
Immunoenzyme Techniques , Kidney Diseases/diagnosis , Podocytes/chemistry , Staining and Labeling/methods , WT1 Proteins/analysis , Adult , Aged , Aged, 80 and over , Antibodies/immunology , Biomarkers/analysis , Disease Progression , Female , Humans , Kidney Diseases/urine , Male , Middle Aged , Prostatic Hyperplasia/diagnosis , Prostatic Hyperplasia/urine , Urinary Calculi/diagnosis , Urinary Calculi/urine , Urinary Tract Infections/diagnosis , Urinary Tract Infections/urine , Urine/cytology , WT1 Proteins/immunology
4.
Blood ; 91(12): 4543-53, 1998 Jun 15.
Article in English | MEDLINE | ID: mdl-9616150

ABSTRACT

Monocytes/macrophages exert a series of important functions in vivo. To facilitate detailed investigation of their functional capacity and the mechanism leading to their differentiation, several cell lines have been established from primary material. We present here a new human monoblastic cell line, designated UG3. UG3 cells are characterized by the following features. (1) UG3 cells harbor the t(9;11)(p22;q23) translocation that results in fusion of the MLL and the AF9 genes and produce the corresponding AF9-MLL and MLL-AF9 fusion transcripts. (2) UG3 cells rely on the presence of exogenous growth factors for viability and proliferation, such as interleukin-3 (IL-3), granulocyte-macrophage colony-stimulating factor (GM-CSF), granulocyte colony-stimulating factor (G-CSF), or macrophage colony-stimulating factor (M-CSF). (3) When cultured in the presence of G-CSF, UG3 cells differentiate along the granulocytic lineage, as evidenced by segmentation of nuclei and positive staining for neutrophilic alkaline phosphatase and peroxidase. (4) When cultured in the presence of GM-CSF or M-CSF, UG3 cells differentiate into mature macrophages while preserving surface expression of CD14 and CD68 and also start to release cytokines into cell-culture supernatants. Under these culture conditions, UG3 cells also take up acetylated LDL. (5) When cultured in the presence of M-CSF and IL-4, UG3 cells differentiate into osteoclast-like multinucleated giant cells capable of bone resorption and display tartrate-resistant acid phosphatase (TRAP) activity. UG3 cells thus provide features to qualify them as a useful model to further investigate the mechanism underlying these processes and also to further elucidate the functional role of mature monocytes/macrophages or osteoclasts.


Subject(s)
Cell Line , Chromosomes, Human, Pair 11 , Chromosomes, Human, Pair 9 , Interleukin-4/pharmacology , Macrophage Colony-Stimulating Factor/pharmacology , Macrophages/pathology , Monocytes/pathology , Osteoclasts/pathology , Translocation, Genetic , Adult , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Lineage , Female , Humans , Karyotyping , Leukemia, Myeloid/genetics , Leukemia, Myeloid/pathology , Macrophages/physiology , Monocytes/physiology , Osteoclasts/physiology
5.
Br J Haematol ; 98(2): 392-8, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9266938

ABSTRACT

A novel GM-CSF-dependent myeloid cell line, OHN-GM, was established from a patient who developed acute myelogenous leukaemia (AML) as a consequence of myelodysplastic syndrome (MDS). As the patient had previously received cytotoxic chemotherapy for Hodgkin's disease, the MDS and AML were probably related to such therapy. Sequential karyotypic analysis established a del(5q) as the initial cytogenetic abnormality. Additional alterations, including t(10;13)(q24;q14), had developed subsequently during disease progression. Southern blot analysis of OHN-GM cells suggested deletion of one allele of the IRF-1 gene, although no aberrant transcripts were detected. Fluorescence in situ hybridization analysis revealed the deletion of the Rb gene due to the t(10;13)(q24;q14) translocation, and Western blot analysis demonstrated the absence of Rb protein in OHN-GM cells. Finally, the OHN-GM cells exhibited two missense point mutations in highly conserved regions of the p53 gene. These observations suggest that a multistep process, involving alterations of Rb and p53 genes, may have contributed to the patient's disease development and progression. To our knowledge, OHN-GM is the first cell line derived from a therapy-related AML. These cells may aid the investigation of leukaemogenesis as well as the biology of secondary leukaemia.


Subject(s)
Genes, Retinoblastoma/genetics , Genes, p53/genetics , Leukemia, Myeloid, Acute/genetics , Antigens, Surface/analysis , Blotting, Southern , Blotting, Western , Chromosomes, Human, Pair 10 , Chromosomes, Human, Pair 13 , Humans , In Situ Hybridization, Fluorescence , Karyotyping , Leukemia, Myeloid, Acute/chemically induced , Male , Middle Aged , Myelodysplastic Syndromes/drug therapy , Point Mutation , Sequence Analysis , Translocation, Genetic , Tumor Cells, Cultured
6.
Int J Hematol ; 66(1): 35-45, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9220659

ABSTRACT

A novel factor-dependent human myeloid leukemia cell line (SAS-1) was established from a 69-year-old Japanese male suffering from CD7 and CD34 expressing acute myeloblastic leukemia (AML M2 in FAB classification). Morphological and cytochemical staining showed that SAS-1 cells were round with basophilic cytoplasm which is positive for peroxidase. Analysis of surface markers revealed that SAS-1 cells were myeloblasts derived from an immature progenitor origin, which express CD34. The consensus karyotype of the cell line was 41 XY 5q-, -7, 11p-, 12p+, -13, -14, -16, -17, -19, -22, with two markers. The proliferation of SAS-1 cells was dependent on the presence of either granulocyte-macrophage colony-stimulating factor (GM-CSF) or interleukin-3 (IL-3), and GM-CSF- and IL-3-induced proliferation was dose dependent. Neither, stem cell factor (SCF) nor granulocyte colony-stimulating factor (G-CSF) alone supported the growth of SAS-1 cells, but supported their viability for more than 4 days and arrested them in the G0/G1 phase. SCF also enhanced GM-CSF- or IL-3-induced growth, but other cytokines did not have this synergistic effect. Clonogenic assays revealed that SAS-1 cells formed 36.0 +/- 5.7 or 41.5 +/- 0.7 colonies/1000 cells in the presence of GM-CSF or IL-3, respectively. SCF also increased the number of colonies formed by GM-CSF or IL-3 treatment, while SAS-1 cells did not form colonies in the presence of SCF alone. SAS-1 cells may prove to be a useful tool for studying the regulation of the cell cycle, myeloid proliferation, and differentiation.


Subject(s)
Antigens, CD34/immunology , Antigens, CD7/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Interleukin-3/pharmacology , Leukemia, Myeloid, Acute/pathology , Tumor Cells, Cultured , Aged , Cell Division/drug effects , Genetic Markers , Humans , Karyotyping , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/immunology , Male
7.
Leukemia ; 11(4): 497-503, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9096689

ABSTRACT

We constructed a recombinant plasmid which expresses antisense src RNA after dexamethasone (Dexa) treatment, and transfected it into U937 human monoblastic leukemia cells (U937-ASRC). Induction of antisense src RNA expression diminished the amounts of c-Src and its protein tyrosine kinase (PTK) activity in U937-ASRC cells. The declines in c-Src and its PTK activity subsequently reduced the proliferation of U937-ASRC cells. To elucidate the growth signal transduction pathway downstream of c-Src, tyrosine phosphorylation of Shc was examined in U937-ASRC cells treated with Dexa. The decline in c-Src by induction of antisense src RNA expression decreased the level of tyrosine phosphorylation of Shc. Immunoprecipitated c-Src directly phosphorylated immunoprecipitated Shc on tyrosine residues in vitro. The amounts of Grb2 and Sos co-immunoprecipitated with Shc were decreased after Dexa treatment. However, the amount of Sos co-immunoprecipitated with Grb2 was apparently not affected by Dexa treatment. These results indicate that Grb2 and Sos constitutively associate with each other in U937 cells. Furthermore, the level of phosphorylation on tyrosine (204) essential for MAP kinase activation was decreased after Dexa treatment. Taken together with all these findings, it is suggested that c-Src directly phosphorylates Shc on tyrosine residues, which in turn binds to Grb2 constitutively associated with Sos to form a Shc-Grb2-Sos complex, and that the complex formation is coupled with MAP kinase activation mediated by Ras activation in U937 cells.


Subject(s)
Adaptor Proteins, Signal Transducing , Leukemia/genetics , Oligopeptides/metabolism , Peptide Fragments/metabolism , Protein Kinases/metabolism , Proteins/metabolism , RNA, Antisense/metabolism , Tyrosine/metabolism , src-Family Kinases/metabolism , Cell Line , Cell Membrane/metabolism , Dexamethasone , GRB2 Adaptor Protein , Humans , Mitogen-Activated Protein Kinase Kinases , Phosphorylation , Plasmids , Signal Transduction , Transfection , src-Family Kinases/genetics
8.
Leuk Res ; 21(2): 139-45, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9112431

ABSTRACT

Heat shock protein 70 (HSP70) protects cells from various injurious stimuli and is important in cell growth and differentiation. However, its expression in leukemia cells has not been analyzed systematically. We, therefore, investigated the expression of HSP70 in various types of leukemia cells and hematopoietic cell lines. Immunoblot analysis revealed that HSP72/73 were expressed at low levels in the acute lymphocytic leukemia cells and lymphoid cell lines examined. However, heat (43 C for 2 h) preferentially augmented HSP72/73 expression in lymphoid cells. This induction was partially blocked by the treatment with quercetin (10 microM for 24 h). Finally, heat shock following quercetin treatment synergistically induced apoptosis in a lymphoid cell line (OZ). These observations suggest the possibility of selective purging of lymphocytic leukemia cells by combination with quercetin and heat.


Subject(s)
HSP70 Heat-Shock Proteins/biosynthesis , Hyperthermia, Induced , Leukemia/metabolism , Leukemia/therapy , Quercetin/pharmacology , Apoptosis/drug effects , Apoptosis/physiology , Combined Modality Therapy , Drug Synergism , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/therapy , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/therapy , Multiple Myeloma/therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Tumor Cells, Cultured
9.
Blood ; 88(3): 955-61, 1996 Aug 01.
Article in English | MEDLINE | ID: mdl-8704254

ABSTRACT

Expression of various cytokines by cytokine gene-transduced tumor cells has been shown to increase antitumor immunity of tumor-bearing hosts. In the present study, macrophage-colony stimulating factor (M-CSF) cDNA was retrovirally transfected into Lewis lung carcinoma cells (3LL) of C57BL/6 mouse origin, and the effects of M-CSF expression were studied by inoculating syngeneic C57BL/6 mice with M-CSF-expressing 3LL cells. The mice inoculated with the lowest M-CSF-producing 3LL clone showed significant prolongation of the survival compared with wild-type 3LL-Inoculated mice, and 70% or more of the mice inoculated with 3LL clones with higher M-CSF production rejected inoculation. Mice injected with radiation-inactivated M-CSF-expressing 3LL cells before or after Inoculation of wild-type 3LL cells showed prolonged survival compared with mice injected with radiated control 3LL cells before or after transplantation of wild-type cells. In vivo depletion of effector subpopulations by injection of antibodies against CD4+ T cells, CD8+ T cells, or natural killer (NK) cells suggested involvement of NK cells and CD4+ T cells in M-CSF-mediated antitumor cytotoxicity in M-CSF-producing 3LL cells-inoculated mice. Severe combined immunodeficiency (SCID) mice with defective T- and B-cell function showed prolonged survival duration after inoculation with M-CSF-expressing 3LL cells compared with those transplanted with control 3LL cells, and this effect of M-CSF expression by 3LL-cells in SCID mice was also abolished by in vivo depletion of NK cells by antibody injection. These findings together with the previous reports that M-CSF augments antibody-dependent and-independent antitumor cytotoxicity suggest that M-CSF induces tumor immunity in this cytokine-expressing tumor-transplantation model.


Subject(s)
Carcinoma, Lewis Lung/pathology , Carcinoma/pathology , Graft Rejection , Macrophage Colony-Stimulating Factor/genetics , Macrophage Colony-Stimulating Factor/therapeutic use , T-Lymphocyte Subsets/immunology , Vaccination , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cytotoxicity, Immunologic , DNA, Complementary/genetics , Genetic Therapy , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Killer Cells, Natural/immunology , Macrophage Colony-Stimulating Factor/biosynthesis , Mice , Mice, Inbred C57BL , Mice, SCID , Neoplasm Proteins/metabolism , Neoplasm Transplantation , Recombinant Proteins/biosynthesis , Recombinant Proteins/therapeutic use , Transfection , Tumor Cells, Cultured/metabolism , Tumor Cells, Cultured/transplantation
10.
Stem Cells ; 14(4): 439-44, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8843545

ABSTRACT

To determine the roles of thrombopoietin (TPO) in platelet function in vitro, we examined the effects of TPO on platelet aggregation. Although several proteins in platelets were tyrosine-phosphorylated by TPO treatment, TPO alone was unable to induce platelet aggregation. However, the secondary wave of platelet aggregation induced by adenosine diphosphate (ADP) was enhanced by TPO in a dose-dependent manner. TPO in conjunction with ADP augmented tyrosine phosphorylation of platelet proteins, including tyrosine-phosphorylated proteins induced by TPO alone. Genistein inhibited protein-tyrosine phosphorylation in platelets induced by TPO with ADP and suppressed TPO-enhanced platelet aggregation. Moreover, tyrosine phosphorylation of MAP-kinases induced by TPO alone and TPO with ADP was consistent with TPO-enhanced platelet aggregation. These findings in the present study suggest that signal transduction involved in TPO-enhanced platelet aggregation is mediated in part by tyrosine-phosphorylated proteins, including MAP-kinases, in platelets through TPO-stimulated c-Mpl, TPO receptor.


Subject(s)
Adenosine Diphosphate/pharmacology , Blood Platelets/physiology , Platelet Aggregation/drug effects , Thrombopoietin/pharmacology , Tyrosine/metabolism , Blood Platelets/metabolism , Cells, Cultured , Humans , Phosphorylation , Proteins/metabolism
11.
Leukemia ; 10(4): 720-6, 1996 Apr.
Article in English | MEDLINE | ID: mdl-8618453

ABSTRACT

To elucidate the role of phosphatidylinositol-3-kinase (Pl3K) in erythropoietin receptor (EPOR)-mediated signaling, we examined the effects of wortmannin (WT), a specific inhibitor of Pl3K on the proliferation of erythropoietin (EPO)-induced erythroid differentiation in K562 human erythroleukemia cells. Percentage of benzidine-positive cells synthesizing hemoglobin and level of glycophorin A expression in the cells were increased after EPO treatment. EPO-enhanced Pl3K activity was suppressed by WT treatment in a dose-dependent manner and constant inhibition of Pl3K by WT interfered with both hemoglobin synthesis and glycophorin A expression promoted by EPO. Wortmannin, however, did not inhibit hemin-induced erythroid differentiation. These findings in the present study suggest that Pl3K plays a crucial role in the transducing the erythroid differentiation signal through EPOR activated by EPO-binding ib K562 cells and that the signaling pathways involved in EPO-induced erythroid differentiation differ from those involved in hemin-induced differentiation.


Subject(s)
Androstadienes/pharmacology , Enzyme Inhibitors/pharmacology , Erythropoietin/pharmacology , Hemoglobins/biosynthesis , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Cell Differentiation/drug effects , Cell Line , Gene Expression , Glycophorins/biosynthesis , Heme/pharmacology , Humans , Kinetics , Leukemia, Erythroblastic, Acute , Phosphatidylinositol 3-Kinases , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Recombinant Proteins/pharmacology , Time Factors , Tumor Cells, Cultured , Wortmannin
12.
Blood ; 85(5): 1220-8, 1995 Mar 01.
Article in English | MEDLINE | ID: mdl-7532035

ABSTRACT

Stem cell factor (SCF) is a cytokine for hematopoietic progenitor cells and plays an important role in megakaryocyte proliferation. The UT-7 cell line was established from a patient with megakaryoblastic leukemia, and its growth and survival are strictly dependent on interleukin-3 (IL-3), granulocyte-macrophage colony-stimulating factor (GM-CSF), erythropoietin (Epo), or IL-6. In this study, we showed that SCF also supported the growth of UT-7 in the absence of other cytokines and downregulated the cell surface c-kit receptors. Constitutive expression of SCF by introducing SCF expression vector made UT-7 grow factor-independently in liquid medium, but not in semisolid medium. This SCF-expressing factor-independent UT-7 (UT-7scf9) expressed the membrane bound form of SCF on their surface, but did not secrete detectable amounts of soluble SCF. UT-7scf9 formed aggregates as they grew in the absence of cytokines, and this aggregation was inhibited by adding soluble SCF into the medium. UT-7 cultured with SCF and UT-7scf9 cultured without cytokines expressed GM-CSF, and anti-GM-CSF neutralizing antibody partially inhibited their growth. These results suggest that SCF stimulated UT-7 proliferation partially through the autocrine-loop of GM-CSF, and UT-7scf9 expressed SCF mostly as a membrane-bound form, which transduces its growth signal through c-kit receptor as they aggregate by cell-to-cell interaction.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Hematopoietic Cell Growth Factors/metabolism , Hematopoietic Cell Growth Factors/pharmacology , Leukemia, Megakaryoblastic, Acute/pathology , Membrane Proteins/metabolism , Base Sequence , Cell Aggregation , Cell Communication , Cell Division/drug effects , Culture Media/pharmacology , DNA, Complementary/genetics , Down-Regulation , Gene Expression Regulation, Leukemic , Humans , Interleukin-3/biosynthesis , Molecular Sequence Data , Neoplasm Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-kit , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Colony-Stimulating Factor/metabolism , Recombinant Fusion Proteins/metabolism , Stem Cell Factor , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism
13.
Leukemia ; 8(12): 2188-93, 1994 Dec.
Article in English | MEDLINE | ID: mdl-7528860

ABSTRACT

We investigated the effect of ubenimex on the growth and differentiation of U937 cells, a histiocytic lymphoma cell line. Ubenimex is a dipeptide ((2S,3R)-3-amino-2-hydroxy-4-phenylbutyryl-L-leucine) and an inhibitor of aminopeptidase B produced by Streptomyces olivoreticuli. Ubenimex inhibited the proliferation of U937 cells in a dose-dependent manner. Ubenimex-treated U937 cells showed condensation of nuclear chromatin, increase of cytoplasmic vacuoles and more intense nonspecific esterase staining compared with untreated U937 cells. Expression of CD13 and CD68 detected by monoclonal antibodies My7 and EBM11, respectively, was enhanced by ubenimex, but the expression of CD4 detected by MT310 was significantly decreased. The effects of ubenimex on U937 cell growth inhibition and enhancement of monocytic cell surface marker expression on U937 cells were reversible when cultivated without ubenimex for more than 6 days. In addition, the bactericidal activity of U937 cells was increased by ubenimex treatment, and was further enhanced by treatment with macrophage colony-stimulating factor (M-CSF). Furthermore, ubenimex augmented the expression of M-CSF receptors by U937 cells and enhanced the tyrosine kinase activity of cellular pp60c-src. These findings indicated that ubenimex inhibited the proliferation of U937 cells and induced morphological, cytochemical and functional differentiation into monocyte/macrophages.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Leucine/analogs & derivatives , Lymphoma, Large B-Cell, Diffuse/pathology , Aminopeptidases/antagonists & inhibitors , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , CD13 Antigens/metabolism , Cell Differentiation/drug effects , Cell Division/drug effects , Cytotoxicity, Immunologic , Humans , Leucine/pharmacology , Lymphoma, Large B-Cell, Diffuse/immunology , Lymphoma, Large B-Cell, Diffuse/metabolism , Macrophage Colony-Stimulating Factor/pharmacology , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Monocytes/immunology , Monocytes/metabolism , Monocytes/pathology , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins pp60(c-src)/metabolism , Receptors, Granulocyte Colony-Stimulating Factor/metabolism , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/immunology , Tumor Cells, Cultured/metabolism , Tumor Cells, Cultured/pathology
14.
Leukemia ; 8(11): 1838-41, 1994 Nov.
Article in English | MEDLINE | ID: mdl-7526087

ABSTRACT

Granulocyte colony-stimulating factor (G-CSF) shortens the duration of chemotherapy-induced granulocytopenia in acute leukemia. G-CSF is administered by 30-min intravenous infusion at a dose of 200 micrograms/m2/day or 5 micrograms/kg/day in most studies. In this study, the efficacy of a reduced dose (33 micrograms/m2/day) of continuous subcutaneous infusion of G-CSF was compared with the effects achieved by 30-min intravenous infusion of the standard dose (200 micrograms/m2/day) in neutropenia after identical chemotherapy in seven patients with acute myelogenous leukemia who were in remission. The duration of granulocytopenia (< 0.5 x 10(9)/l), thrombocytopenia (< 50 x 10(9)/l); G-CSF administration and fever (> 38 degrees C) were 10.1 +/- 5.0 days, 16.5 +/- 9.3 days, 16.6 +/- 7.4 days and 3.1 +/- 5.4 days for 33 micrograms/m2/day continuous subcutaneous infusion, and 10.7 +/- 6.8 days, 16.7 +/- 9.9 days, 16.1 +/- 7.6 days and 2.0 +/- 2.5 days for 30-min intravenous infusion of the standard dose of G-CSF. In each parameter studied, there was no statistical difference between the two methods of G-CSF administration by paired t-test. The costs for G-CSF could be substantially reduced. In most patients, plasma G-CSF concentration rose to the highest level of 1.8-3.7 ng/ml 48-72 h after starting 33 micrograms/m2/day continuous subcutaneous infusion, and gradually decreased as the peripheral granulocyte count recovered, suggesting binding of G-CSF molecules to the specific receptors on the cells of granulocytic lineage.


Subject(s)
Granulocyte Colony-Stimulating Factor/administration & dosage , Leukemia, Myeloid, Acute/drug therapy , Neutropenia/drug therapy , Adult , Antineoplastic Agents/adverse effects , Female , Granulocyte Colony-Stimulating Factor/pharmacokinetics , Humans , Injections, Intravenous , Injections, Subcutaneous , Insulin Infusion Systems , Leukocyte Count , Male , Middle Aged , Neutropenia/chemically induced , Recombinant Proteins
15.
Int J Hematol ; 60(3): 197-205, 1994 Oct.
Article in English | MEDLINE | ID: mdl-7858239

ABSTRACT

Molecularly defined specific chromosomal translocation in leukemia allows detection of minimal residual leukemia cells by the reverse transcription-polymerase chain reaction (RT-PCR). However, the positivity of the specific fusion transcripts in chronic myelogenous leukemia and acute myelogenous leukemia with t(8;21) is reportedly not directly correlated with the predictability of relapse. We analyzed seven patients with acute promyelocytic leukemia (APL) in hematological remission for PML-retinoic acid receptor alpha (PML-RAR alpha) fusion transcripts by RT-PCR with the sensitivity level of one APL cell in 10(5) bone marrow mononuclear cells. Two of the four patients with chemotherapy-induced remission had detectable PML-RAR alpha only before treatment. In the other two patients with chemotherapy-induced remission, the PML-RAR alpha was detectable when their remission was first confirmed and became negative after consolidation chemotherapy. Two patients were resistant to chemotherapy and achieved remission by all-trans-retinoic acid; PML-RAR alpha was detectable in them for a few months during consolidation chemotherapy. Two patients whose PML-RAR alpha had become continuously positive had relapse 2 and 8 months later, but the other five patients with continuously negative or only transiently positive PML-RAR alpha remained in remission during follow-up for 11 to 35 months. These findings suggest the relevance of detectable PML-RAR alpha by RT-PCR to the predictability of relapse in acute promyelocytic leukemia.


Subject(s)
Leukemia, Promyelocytic, Acute/genetics , Receptors, Retinoic Acid/genetics , Transcription Factors/genetics , Adolescent , Adult , Base Sequence , Female , Humans , Leukemia, Promyelocytic, Acute/drug therapy , Male , Middle Aged , Molecular Sequence Data , Polymerase Chain Reaction , Prognosis , Remission Induction , Transcription, Genetic
16.
Biochem Mol Biol Int ; 34(1): 85-92, 1994 Aug.
Article in English | MEDLINE | ID: mdl-7849628

ABSTRACT

In order to elucidate the role of c-myb gene in erythroid differentiation of K562 cell induced by hemin (Hm) and erythropoietin (Epo), we constructed recombinant plasmid that could produce antisense myb RNA after induction with dexamethasone. During treatment with Hm, K562 cells constitutively expressed c-myb mRNA, and 50% of them began to synthesize hemoglobin (Hb). Expression of antisense myb RNA reduced the amount of c-myb mRNA, and the percentage of Hb-synthesizing cells was decreased to 20%. In the presence of Epo, c-myb mRNA declined and 20% of K562 cells synthesized Hb regardless of antisense myb RNA expression. It is suggested that constitutive expression of c-myb mRNA is necessary for Hm-induced differentiation, and that a decrease in the amount of c-myb mRNA induced by antisense myb RNA expression suppresses Hm-induced differentiation. The amount of c-myb mRNA in K562 cells was reduced during the differentiation induced by Epo. Expression of GATA-1 mRNA was almost constant during Hm-induced differentiation, but increased during Epo treatment. It is supposed that the mechanism of Hm-induced differentiation is distinguished from that of Epo-induced differentiation in K562 cells.


Subject(s)
Erythroid Precursor Cells/drug effects , Erythropoietin/pharmacology , Gene Expression Regulation, Neoplastic/genetics , Hemin/pharmacology , Analysis of Variance , Cell Differentiation/drug effects , Cell Differentiation/genetics , DNA-Binding Proteins/genetics , Dexamethasone/pharmacology , Electrophoresis, Polyacrylamide Gel , Erythroid Precursor Cells/cytology , Erythroid-Specific DNA-Binding Factors , GATA1 Transcription Factor , Globins/genetics , Glycophorins/biosynthesis , Hemoglobins/biosynthesis , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-myb , RNA, Antisense/biosynthesis , RNA, Antisense/genetics , RNA, Messenger/biosynthesis , Transcription Factors/genetics , Tumor Cells, Cultured
17.
Biochem Biophys Res Commun ; 201(3): 1534-40, 1994 Jun 30.
Article in English | MEDLINE | ID: mdl-7517673

ABSTRACT

We constructed a recombinant plasmid which expresses antisense src RNA in human cells and used it as a tool for investigating the role of pp60c-src in proliferation and differentiation of K562 human leukemia cells. In erythropoietin (EPO)-responsive cells, EPO induces rapid tyrosine phosphorylation of several cellular proteins including EPO receptor (EPOR) although EPOR has no tyrosine kinase domain. Here we show that antisense src RNA expression suppresses pp60c-src synthesis in the recombinant plasmid-transfected K562 cells, reduces the proliferation and inhibits hemoglobin synthesis and glycophorin A expression promoted by EPO in K562 cells. These findings suggest that pp60c-src plays crucial roles in the proliferation and EPO-induced erythroid differentiation of K562 cells.


Subject(s)
Erythropoiesis , Genes, src , Proto-Oncogene Proteins pp60(c-src)/genetics , Cell Differentiation , Cell Division , Erythropoietin/pharmacology , Glycophorins/biosynthesis , Hemoglobins/biosynthesis , Humans , Leukemia, Erythroblastic, Acute/pathology , RNA, Antisense , Tumor Cells, Cultured
18.
Biochem Biophys Res Commun ; 201(2): 1001-7, 1994 Jun 15.
Article in English | MEDLINE | ID: mdl-7516155

ABSTRACT

To elucidate the role of pp60c-src in U937 human monoblastoid leukemia cell proliferation, recombinant plasmids containing the src gene or myb gene, which could produce antisense src or antisense myb RNA after dexamethasone treatment, were constructed and transfected into U937 cells (U937-ASRC, U937-AMYB). pp60c-src synthesis in U937-ASRC was diminished by the third day after induction of antisense src RNA and the cell proliferation was reduced, furthermore, the amount of p75c-myb was significantly decreased by the third day. p75c-myb synthesis in U937-AMYB was diminished by the second day after induction of antisense myb RNA and the cell growth was significantly inhibited but the amount of pp60c-src in U937-AMYB was not reduced. These results suggest that a decrease in the amount of pp60c-src leads to an inhibition of p75c-myb expression and subsequent reduction in the U937 cell proliferation.


Subject(s)
Genes, src , Proto-Oncogene Proteins pp60(c-src)/metabolism , RNA, Antisense/metabolism , Cell Division/drug effects , Cell Line , DNA-Binding Proteins/biosynthesis , Dexamethasone/pharmacology , Humans , Kinetics , Leukemia , Lymphoma, Large B-Cell, Diffuse , Oncogenes , Plasmids , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins c-myb , Proto-Oncogene Proteins pp60(c-src)/biosynthesis , Restriction Mapping , Time Factors , Transfection , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL