Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Br J Haematol ; 108(3): 621-8, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10759722

ABSTRACT

The objective of this study was to evaluate the cycling status of cord blood (CB)-derived colony-forming cells (CFC) and long-term culture-initiating cells (LTC-IC), and their recruitment into the S-phase of the cell cycle. By using the cytosine arabinoside (Ara-C) suicide approach, we found that only small proportions of both CFC and LTC-IC were in the S-phase of the cell cycle. These estimates were confirmed by flow cytometric DNA analysis, which showed that 96 +/- 2% of CB-derived CD34+ cells were in G0/G1 and only 1.6 +/- 0.4% in the S-phase. Staining of CD34+ cells with an antistatin monoclonal antibody, a marker of the G0 phase, indicated that among CD34+ cells with a flow cytometric DNA content typical of the G0/G1 phase 68 +/- 7% of cells were in the G0 phase of the cell cycle. Incubation (24 h) with interleukin 3 (IL-3), recombinant human stem cell factor (SCF) and granulocyte colony-stimulating factor (G-CSF) significantly increased the proportion of cells in the S-phase for both CFC and LTC-IC without inducing any loss in numbers. Flow cytometric DNA analysis also showed an increase in CD34+ cells in the S-phase upon continuous exposure to these cytokines. Our findings indicate that: (i) very few CB-derived CFC or LTC-IC were in the S-phase of the cell cycle; (ii) a substantial amount of CD34+ cells with a flow cytometric DNA content typical of the G0/G1 fraction was cycling, as found in the G1 phase of the cell cycle; and (iii) 24-h incubation with IL-3, SCF and G-CSF could drive a proportion of progenitor cells into the S-phase without reducing their number. These data might be useful for gene transfer protocols and the ex vivo expansion of CB-derived progenitor cells.


Subject(s)
Fetal Blood/cytology , Hematopoietic Stem Cells/cytology , S Phase , Antigens, CD34/immunology , Cell Cycle/drug effects , Cell Cycle Proteins , Cells, Cultured , Fetal Blood/immunology , Flow Cytometry , Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cells/immunology , Humans , Interleukin-3/pharmacology , Peptide Elongation Factor 1 , Proteins/analysis , Recombinant Proteins , Stem Cell Factor/pharmacology
3.
Br J Haematol ; 102(4): 996-1003, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9734650

ABSTRACT

We studied X-chromosome inactivation patterns in blood cells from normal females in three age groups: neonates (umbilical cord blood), 25-32 years old (young women group) and >75 years old (elderly women). Using PCR, the differential allele methylation status was evaluated on active and inactive X chromosomes at the human androgen receptor (HUMARA) and phosphoglycerate kinase (PGK) loci. A cleavage ratio (CR) > or = 3.0 was adopted as a cut-off to discriminate between balanced and unbalanced X-chromosome inactivation. In adult women this analysis was also performed on hair bulbs. The frequency of skewed X-inactivation in polymorphonuclear (PMN) cells increased with age: CR > or = 3.0 was found in 3/36 cord blood samples, 5/30 young women and 14/31 elderly women. Mathematical analysis of patterns found in neonates indicated that X-chromosome inactivation probably occurs when the total number of haemopoietic stem cell precursors is 14-16. The inactivation patterns found in T lymphocytes were significantly related to those observed in PMNs in both young (P < 0.001) and elderly women (P < 0.01). However, the use of T lymphocytes as a control tissue for distinguishing between skewed inactivation and clonal proliferation proved to be reliable in young females, but not in elderly women, where overestimation of the frequency of clonal myelopoiesis may appear.


Subject(s)
Dosage Compensation, Genetic , Hematopoietic Stem Cells/cytology , X Chromosome , Adult , Aged , Aging/genetics , Bone Marrow Transplantation , Clone Cells/cytology , Female , Fetal Blood/cytology , Hair/cytology , Humans , Infant, Newborn , Leukemia/therapy , Neutrophils/cytology , Polymerase Chain Reaction , T-Lymphocytes/cytology
4.
Oncol Rep ; 5(2): 427-9, 1998.
Article in English | MEDLINE | ID: mdl-9468573

ABSTRACT

The feasibility and safety of the administration of multiple cycles of dose-intensive chemotherapy (CT) supported with repeated reinfusions of circulating progenitor cells (CPCs) were evaluated in a prospective study of adjuvant initial therapy of poor-prognosis breast cancer. Eighteen patients with resectable breast cancer involving >/= 10 axillary nodes or >/= 5 axillary nodes and negativity of the estrogen receptor status received a cycle of standard FEC regimen (5-FU 600 mg/m2, epirubicin 60 mg/m2, CTX 600 mg/m2, i.v. on day 1) followed by G-CSF as CPC mobilization technique. Collected CPCs were fractionated and reinfused, with G-CSF, after each of the 4 subsequent cycles of high-dose FEC (HD-FEC) (5-FU 750 mg/m2, epirubicin 120 mg/m2, CTX 3 g/m2, i.v.) planned at 21 day intervals. The median numbers of CD34+ cells and CFU-GM collected (with one or two leukaphereses per patient) were 9.7x10(6)/kg (range: 2.5-22.9) and 9.9x10(4)/kg (range: 1.9-27.3), respectively, and day 9 was the median first day of procedure (range: 8-12) after FEC. All patients received the 4 courses of HD-FEC (for a total of 72 cycles). Hemopoietic recovery was rapid after each cycle and there was no treatment-related delays in CT administration. Mucositis was the major non-hematological toxicity. There were 2, 3, 7 and 9 episodes of WHO grade 3/4 mucositis in cycles 1, 2, 3 and 4, respectively. These severe episodes lasted a median of 4 days (range: 2-6) but no patient required parenteral nutrition. The mean +/- SD total hospital stay lasted 10 +/- 2 days. The delivery of 4 cycles of dose-intensive FEC CT supported by CPCs (mobilized with a single course of standard-dose FEC + G-CSF) is feasible and safe. It could represent an effective alternative strategy to other more aggressive programs for the adjuvant therapy of high-risk early breast cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Breast Neoplasms/therapy , Granulocyte Colony-Stimulating Factor/administration & dosage , Hematopoietic Stem Cell Transplantation , Adult , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cyclophosphamide/administration & dosage , Cyclophosphamide/adverse effects , Epirubicin/administration & dosage , Epirubicin/adverse effects , Feasibility Studies , Female , Fluorouracil/administration & dosage , Fluorouracil/adverse effects , Granulocyte Colony-Stimulating Factor/adverse effects , Hematopoiesis/drug effects , Humans , Leukapheresis , Lymphatic Metastasis , Middle Aged , Prognosis , Prospective Studies , Safety
5.
Br J Haematol ; 98(3): 621-6, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9332317

ABSTRACT

Although devoid of proliferative capacity, polymorphonuclear neutrophils (PMN) express receptors for haemopoietic growth factors and need growth factors for survival and functional stimulation. This study showed that in vitro treatment of human PMN with GM-CSF for up to 48 h increases cell surface expression of the beta 2-integrin molecules CD11b/CD18 and CD11c/CD18 and of the receptor for the chemotactic peptide fMLP. Such modifications are usually expression of PMN activation. PMN treated with GM-CSF also displayed increased phagocytosis of latex particles and enhanced oxidative burst and superoxide anion release. Since integrins mediate PMN adhesion to endothelium, homotypic adhesion, chemotaxis/phagocytosis and the triggering of respiratory burst, our results suggested that functional stimulation of PMN persisted following prolonged exposure of PMN to growth factors and that it was not a temporary phenomenon which lasted only for the first 12-24 h of treatment. We also used oligonucleotides antisense to the Bcr gene mRNA to inhibit expression of the gene and evaluate its function in PMN, following the recent observation that PMN from Bcr-null mutant mice produced increased amounts of reactive oxygen metabolites upon activation. The antisense oligonucleotides had no effect on the parameters investigated. This may indicate that increased production of O2 by neutrophils in which the Bcr gene is not expressed requires either that gene expression is absent in the earlier stages of myeloid differentiation/maturation, so that when inhibition occurs in the terminally differentiated neutrophils their functional status is no longer influenced, or that the residual low-level expression of the gene which may be present in the antisense-treated cells is sufficient to provide a normal response to stimulation.


Subject(s)
CD18 Antigens/metabolism , Cell Adhesion Molecules/metabolism , Fusion Proteins, bcr-abl/physiology , Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Neutrophils/physiology , Antibodies, Monoclonal/physiology , Cells, Cultured , Humans , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Polymerase Chain Reaction
6.
Haematologica ; 81(5): 434-41, 1996.
Article in English | MEDLINE | ID: mdl-8952157

ABSTRACT

BACKGROUND: Since only a portion of anemic patients outside the uremia setting benefit from erythropoietin treatment, a reliable means of predicting potential responders and nonresponders would be very useful. MATERIALS AND METHODS: We retrospectively reviewed the clinical records of 58 patients with refractory anemia associated with various malignant disorders who had been treated with subcutaneous rHuEpo. The starting rHuEpo dose was 375 U/kg/week for 4 weeks, and was increased to 750 U/kg/week for another 4 weeks if no response was observed. Response was defined as a Hb increase > or = 2 g/dL with no need for blood transfusion. We examined the value of various laboratory parameters (baseline levels, 2-week and 4-week changes) as predictors of response. Endogenous erythropoietin production was evaluated by its serum level and erythroid activity was assessed through reticulocyte count and circulating transferrin receptor. RESULTS: Forty-eight individuals were evaluable, 58% of whom responded to rHuEpo within 8 weeks. Multiple regression analysis showed that 53% of the variation in the 8-week Hb concentration was explained by variations in baseline serum erythropoietin and the 2-week change in serum transferrin receptor (p < 0.001). Based on these two parameters, response prediction in individual patients would have resulted in a sensitivity of 96%, a specificity of 79% and an overall accuracy of 88%. In addition, 58% of the variation in the 8-week Hb was explained by variations in the 4-week changes in Hb and reticulocyte count (p < 0.001). Utilizing these latter parameters and baseline serum erythropoietin, response prediction in individual patients would have resulted in a sensitivity of 92%, a specificity of 82% and an overall accuracy of 88%. CONCLUSIONS: This retrospective analysis suggests that response to rHuEpo can be reasonably predicted by pretreatment serum erythropoietin together with early changes in simple laboratory parameters.


Subject(s)
Anemia/drug therapy , Erythropoietin/therapeutic use , Neoplasms/complications , Anemia/etiology , Humans , Predictive Value of Tests , Recombinant Proteins/therapeutic use
7.
Br J Haematol ; 93(4): 789-94, 1996 Jun.
Article in English | MEDLINE | ID: mdl-8703804

ABSTRACT

The plant toxin saporin is a ribosome-inactivating protein which inhibits protein synthesis and growth of both normal and tumour cells. Its cytotoxic activity can be increased by coupling with antibodies recognizing cell surface antigens. In this work we performed experiments to test the hypothesis that saporin induces cell death via apoptosis. Exposure to saporin induced apoptosis in different cellular models, such as human peripheral blood B lymphocytes and neutrophils, in the Daudi B-cell line, and in the haemopoietic cell lines HL-60 and TF-1. This was indicated by: (a) the appearance of typical morphological features such as chromatin condensation, nuclear fragmentation and blebbing of plasma membranes: (b) DNA degradation into oligonucleosomal fragments: (c) the appearance of apoptotic cells on DNA flow cytometry as a cell population with reduced DNA content (A0 region). The fraction of cells showing features of apoptosis ranged from 19 +/- 5% for TF-1 cells to 35 +/- 8% for neutrophils. In experiments with normal peripheral blood B lymphocytes or with Daudi cells, we compared the activity of native saporin with that of an immunotoxin hybrid molecule obtained by binding the toxin to two bispecific antibodies recognizing both saporin and the B lymphocyte-specific antigen CD22 (Sap/BsAb complexes). Saporin bound to the antibodies was 2-3 logs more effective than native saporin in inducing apoptosis, with maximal inhibitions being observed at concentrations of 10(-6) M for native saporin and 10(-9)-10(-8) M for the hybrid molecules. These findings indicate that treatment with saporin results in apoptosis of target cells and suggest that this may be relevant to the therapeutic use of saporin-containing immunotoxins. In fact, if used in vivo as an immunotoxin, its cytotoxic activity could be devoid of more extensive and non-specific tissue damaging effects as would be the case if saporin induced necrosis of target cells.


Subject(s)
Apoptosis/drug effects , B-Lymphocytes/drug effects , Immunotoxins/pharmacology , N-Glycosyl Hydrolases , Neutrophils/drug effects , Plant Proteins/pharmacology , Antibodies, Monoclonal , B-Lymphocytes/cytology , Cell Division/drug effects , Cell Line , Humans , Neutrophils/cytology , Ribosome Inactivating Proteins, Type 1 , Saporins
8.
Int J Oncol ; 9(5): 971-6, 1996 Nov.
Article in English | MEDLINE | ID: mdl-21541603

ABSTRACT

The biological mechanisms by which the association of interleukin-3 (IL-3) and granulocyte-macrophage colony-stimulating factor (GM-CSF) is expected to effectively reduce the hematological toxicity associated with chemotherapy (CT) are not completely elucidated. We exploited the cell kinetic changes of the bone marrow CD34(+) cell subset after CT followed by the IL-3+GM-CSF together with the clinical effects of this association. Eighteen patients with advanced cancers and normal hematopoiesis were treated with an intensified CT course (mg/m(2): CTX 1100, epirubicin 100, VP-16 200; iv day 1). Six cycles were planned at 14-day intervals with the support of IL-3 (5 mu g/kg/day; from day 2 to 6) sequenced with GM-CSF (same dose; from day 7 to 11). DNA content and bromodeoxyuridine incorporation were evaluated using flow cytometry on immunomagnetically-sorted bone marrow CD34(+) cells, at baseline and at different times (days 5, 6, 7, 8, 11 and 14) after CT followed by IL-3+GM-CSF. Treatment with IL-3 induced a marked increase in the % of myeloid precursors with respect to the baseline and in the % of CD34(+) cells in S-phase. However, while the first parameter remained elevated until day 14, the enhanced proliferative activity of the CD34(+) cell subset decreased after IL-3 was stopped and remained significantly low during GM-CSF administration. These data suggest a negative rebound effect on CD34(+) cell proliferation after IL-3 discontinuation which is maintained during GMCSF, that led to kinetic refractoriness of the hyperplastic marrow. In the 99 courses completed a rapid neutrophil and platelet recovery was obtained without cumulative multilineage toxicity. The modifications of CD34(+) cell cycling after CT followed by IL-3+GM-CSF could provide additional myeloprotection during multicyclic, dose-intensive programs.

9.
Blood ; 86(9): 3387-93, 1995 Nov 01.
Article in English | MEDLINE | ID: mdl-7579442

ABSTRACT

A number of experimental observations suggest that the proto-oncogene c-abl participates in the regulation of hematopoietic cell growth. We used an antisense strategy to study the relationship between c-abl expression and hematopoietic cell proliferation and differentiation. Purified normal human bone marrow-derived CD34+ cells were obtained by immunomagnetic selection and incubated with 18-base-unmodified antisense oligodeoxynucleotides complementary to the first six codons of the two alternative first exons of c-abl, la and lb. At the end of incubation, an aliquot of cells was assayed for clonogenic growth and the remainder was used for flow cytometric analyses. Cell kinetics were evaluated by means of both single parameter DNA and bivariate DNA/bromodeoxyuridine (BrdU) flow cytometry. Apoptosis was routinely studied by DNA flow cytometric analysis and, in some cases, also through DNA agarose gel electrophoresis for detection of oligonucleosomal DNA fragments. Expression of differentiation markers was studied by flow cytometry. Exposure to antisense oligonucleotides specifically inhibited the accumulation of c-abl mRNA in CD34+ cells. Preincubation with the c-abl antisense oligomers reduced the proportion of cells in S-phase from 19% +/- 5% (mean +/- SD) to 7% +/- 4% (P < .05), and BrdU labeling from 13% +/- 6% to 6% +/- 3% (P < .05). Flow cytometry and DNA agarose gel electrophoresis showed that treated CD34+ cells accumulated in the G0/G1 region of the DNA histogram with no evidence of either differentiation or apoptosis. By contrast, both growth factor deprivation and exposure of CD34+ cells to the tyrosine kinase inhibitor tyrphostin AG82 clearly induced apoptosis. When cells were preincubated with antisense oligonucleotides and then plated for evaluation of colony formation, this resulted in a significant inhibition of colony forming unit granulocyte-macrophage growth (from 44 +/- 15 to 22 +/- 9; P < .01) but had no effect on burst-forming unit erythroid growth (24 +/- 11 v 21 +/- 11; P < .05). These results suggest that c-abl expression is critical for entry of human CD34+ hematopoietic cells into S-phase and for their differentiation to granulocyte-macrophage progenitors. They also indicate that other tyrosine kinases besides p145c-alb are active in the prevention of apoptosis, so that inhibition of c-abl RNA accumulation arrests CD34+ cells in G0/G1 without activating programmed death.


Subject(s)
Antigens, CD34 , Genes, abl , Granulocytes , Hematopoiesis/drug effects , Hematopoietic Stem Cells/drug effects , Macrophages , Oligonucleotides, Antisense/pharmacology , Proto-Oncogene Proteins c-abl/physiology , S Phase/genetics , Apoptosis/drug effects , Base Sequence , Cell Differentiation/drug effects , Cell Division/drug effects , Cells, Cultured , Colony-Forming Units Assay , DNA Damage , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Immunophenotyping , Molecular Sequence Data , Polymerase Chain Reaction , Proto-Oncogene Mas
12.
Leukemia ; 7(12): 2012-8, 1993 Dec.
Article in English | MEDLINE | ID: mdl-8255101

ABSTRACT

Experimental evidence suggests that hematopoietic growth factors promote cell survival by suppressing apoptosis or programmed cell death. Since interleukin 3 (IL-3) and granulocyte-macrophage colony stimulating factor (GM-CSF) induce tyrosine phosphorylation of a common set of proteins in the factor-dependent cell line M07e, we have investigated whether growth-factor-induced tyrosine phosphorylation is involved in the promotion of cell survival and suppression of apoptosis. Experiments were carried out with the leukemic cell lines HL-60 and M07e and the tyrosine kinase inhibitors genistein and tyrphostin AG82. Both the tyrosine kinase inhibitors induced apoptosis of HL-60 and M07e cells. This was indicated by the appearance of DNA degradation and morphologic evidence of nuclear condensation and fragmentation. It was also confirmed by flow cytometry of DNA, which showed apoptotic cells as a fraction of cells characterized by a diminished DNA stainability, represented on the DNA frequency histograms as a distinct peak below the G0/G1 population. Kinase inhibitors also reduced the fraction of cells in the S phase of the cell cycle. That tyrphostin specifically inhibited tyrosine kinases was further suggested by the prevention of its effects by the tyrosine phosphatase inhibitor sodium orthovanadate (vanadate), at least during the first 18-24 h of treatment. The incomplete prevention of genistein effects by vanadate suggests that genistein is a less specific inhibitor of tyrosine kinases than tyrphostin, and may also act as an inhibitor of topoisomerase II. Vanadate also prevented apoptosis and reduction of the S phase in M07e cells cultured for 24 h in the absence of growth factors. These results suggest that tyrosine phosphorylation is an essential step in IL-3 and GM-CSF signal transduction. Since in our experimental model the effects of tyrosine kinase inhibition and growth factor deprivation could be reversed by concomitant inhibition of tyrosine phosphatases, it is suggested that a balance between tyrosine kinases and tyrosine phosphatases establishes whether a cell will survive or undergo apoptosis.


Subject(s)
Catechols/pharmacology , Isoflavones/pharmacology , Leukemia, Megakaryoblastic, Acute/pathology , Leukemia, Promyelocytic, Acute/pathology , Nitriles/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Tyrosine/metabolism , Tyrphostins , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Division/drug effects , Flow Cytometry , Genistein , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Interleukin-3/pharmacology , Leukemia, Megakaryoblastic, Acute/enzymology , Leukemia, Megakaryoblastic, Acute/metabolism , Leukemia, Promyelocytic, Acute/enzymology , Leukemia, Promyelocytic, Acute/metabolism , Phosphorylation , Protein Tyrosine Phosphatases/physiology , Protein-Tyrosine Kinases/physiology , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism , Tumor Cells, Cultured/pathology , Vanadates/pharmacology
14.
Haematologica ; 78(4): 208-12, 1993.
Article in English | MEDLINE | ID: mdl-8294051

ABSTRACT

BACKGROUND: Several clinical studies have shown that recombinant human erythropoietin (rHuEpo) can ameliorate the anemia associated with hematologic malignancies and solid tumors. On the other hand, only a few studies have been performed to investigate whether rHuEpo can affect or modulate the growth of malignant cells. MATERIALS AND METHODS: We studied the effects of rHuEpo (0.5 to 10 IU/mL) on clonogenic growth and cell kinetics in ten cell lines derived from both hematologic malignancies and solid tumors. Clonogenic assays were performed by plating 5 x 10(3) cells in agar, while the percentage of cells in S phase was assessed by DNA flow cytometry. RESULTS: rHuEpo did not affect either in vitro colony formation or S phase percentage in the human erythroid cell lines K-562 and HEL expressing erythropoietin receptors (< 40 receptors per cell). No effect of rHuEpo was observed in the remaining hematopoietic cell lines or in five solid tumor cell lines. CONCLUSIONS: These findings indicate that rHuEpo, even at very high concentrations, does not affect either clonogenic growth or DNA synthesis in the cell lines tested. Available evidence suggests that rHuEpo can be safely employed in all malignancies except acute myeloid leukemia.


Subject(s)
Erythropoietin/pharmacology , Hematopoietic Stem Cells/drug effects , Leukemia, Erythroblastic, Acute/pathology , Neoplastic Stem Cells/drug effects , Recombinant Proteins/pharmacology , Cell Division , Clone Cells/drug effects , DNA, Neoplasm/analysis , Humans , Leukemia, Myeloid, Acute/pathology , Neoplasms/pathology , Tumor Cells, Cultured/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...