Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Mol Ther ; 22(1): 149-59, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23985698

ABSTRACT

Despite the progress in our understanding of genes essential for stem cell regulation and development, little is known about the factors secreted by stem cells and their effect on tissue regeneration. In particular, the factors secreted by human CD34+ cells remain to be elucidated. We have approached this challenge by performing a cytokine/growth factor microarray analysis of secreted soluble factors in medium conditioned by adherent human CD34+ cells. Thirty-two abundantly secreted factors have been identified, all of which are associated with cell proliferation, survival, tissue repair, and wound healing. The cultured CD34+ cells expressed known stem cell genes such as Nanog, Oct4, Sox2, c-kit, and HoxB4. The conditioned medium containing the secreted factors prevented cell death in liver cells exposed to liver toxin in vitro via inhibition of the caspase-3 signaling pathway. More importantly, in vivo studies using animal models of liver damage demonstrated that injection of the conditioned medium could repair damaged liver tissue (significant reduction in the necroinflammatory activity), as well as enable the animals to survive. Thus, we demonstrate that medium conditioned by human CD34+ cells has the potential for therapeutic repair of damaged tissue in vivo.


Subject(s)
Antigens, CD34/metabolism , Culture Media, Conditioned/pharmacology , Hematopoietic Stem Cells/metabolism , Regeneration/drug effects , Wound Healing/drug effects , Animals , Biomarkers/metabolism , Cell Death/drug effects , Cell Line , Culture Media, Serum-Free , Cytokines/genetics , Cytokines/metabolism , Humans , Liver Regeneration/drug effects , Male , Primary Cell Culture , Protein Interaction Mapping , Protein Interaction Maps , Rats , Transcriptome
2.
Int J Cardiol ; 166(3): 664-71, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-22192286

ABSTRACT

AIMS: Anaemia is a prevalent and adverse comorbidity in chronic heart failure (CHF) but its origins are frequently elusive. Diffuse inflammation is also prominent in CHF and a potent inhibitor of erythrocyte production. We tested the hypothesis that unexplained anaemia in CHF might be subsequent to diminished erythropoiesis as a result of an immune-mediated suppression of erythroid colony formation. METHODS: We studied 61 CHF patients and 20 healthy control subjects. Circulating primitive haematopoietic (CD34(+)) and erythroid precursor cells were quantified by flow cytometry. Circulating erythroid progenitors (BFU-E) were cultured in methylcellulose in the presence and absence of monocytes and sera, and with anti-TNFα neutralising antibodies. RESULTS: Despite higher erythropoietin levels, anaemic patients exhibited lower absolute reticulocyte counts and reticulocyte production indices (P<0.001) than non-anaemic patients and healthy controls. Diminished erythropoiesis was paralleled by attenuated circulating CD34(+), erythroid progenitor and precursor cells in anaemic patients (all P<0.01). Depletion of monocytes from cultures derived only from anaemic patients enhanced BFU-E growth (P=0.03). Only the addition of monocytes and sera from anaemic patients suppressed autologous or allogeneic BFU-E colony formation (P=0.02). Serum TNFα levels were highest in anaemic patients and anti-TNFα neutralising antibodies partly abrogated the inhibitory effects of anaemic sera on erythroid colony growth (P=0.03). CONCLUSION: Unexplained anaemia in patients with CHF results partly from suppressed erythropoiesis and monocytes, via a direct effect of TNFα on erythroid cells, orchestrate a degree of this suppression.


Subject(s)
Anemia/diagnosis , Anemia/immunology , Erythropoiesis/immunology , Heart Failure/diagnosis , Heart Failure/immunology , Monocytes/immunology , Aged , Anemia/epidemiology , Cells, Cultured , Chronic Disease , Female , Heart Failure/epidemiology , Humans , Male , Middle Aged
3.
Int J Cardiol ; 164(3): 359-64, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-21821297

ABSTRACT

BACKGROUND: Chronic heart failure (CHF) patients are frequently anaemic despite elevated endogenous erythropoietin (Epo) levels. We tested the hypothesis that this might be due to Epo resistance and investigated whether any defects apparent were due to Epo receptor (EpoR) downregulation and/or impaired Epo-induced signal transduction. METHODS: We studied 28 CHF patients (age 64 ± 10 yrs, LVEF 29 ± 9%, 89% male) and 12 healthy controls (65 ± 11 yrs, 75% male). Circulating erythroid progenitors (BFU-E) were cultured with 0, 1, 3 and 9 U/mL Epo. Circulating erythroblast surface EpoR and intracellular phosphorylated Signal Transducer and Activator of Transcription (phosphoSTAT)-5 expression were determined by flow cytometry. RESULTS: Whilst BFU-E from control and non-anaemic subjects required only 3 U/mL Epo to significantly increase their numbers from baseline (1 U/mL), those from anaemic patients required 9 U/mL Epo. Lower Epo sensitivities related to higher interleukin-6 (r=-0.41, P=0.01) and soluble tumour necrosis factor receptor 2 (r=-0.38, P=0.02) levels. EpoR-positive cells were more abundant in anaemic patients (P<0.001). Although erythroblasts from anaemic patients exhibited higher baseline EpoR and phosphoSTAT5 expression (all P<0.05), Epo stimulation triggered significant increases in phosphoSTAT5 levels only in erythroblasts from control subjects and not in those from anaemic patients. CONCLUSION: The responsiveness of erythroid cells to Epo is diminished in anaemic CHF patients. This is not due to EpoR downregulation but relates to a profound blunting of Epo-induced JAK-STAT signalling. Whilst residual Epo sensitivity can be exploited clinically with erythropoietic agents, targeting the mechanisms underlying Epo resistance in CHF may provide greater efficacy.


Subject(s)
Anemia/drug therapy , Drug Resistance/physiology , Erythroid Precursor Cells/drug effects , Erythropoietin/therapeutic use , Heart Failure/complications , Signal Transduction/drug effects , Aged , Anemia/etiology , Anemia/metabolism , Cells, Cultured , Chronic Disease , Down-Regulation/physiology , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/metabolism , Female , Flow Cytometry , Humans , Janus Kinases/metabolism , Male , Middle Aged , Receptors, Erythropoietin/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction/physiology
4.
Br J Haematol ; 146(6): 637-51, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19656152

ABSTRACT

The regulation of myeloid progenitor cell (granulocyte-macrophage colony-forming units, CFU-GM) proliferation/differentiation by the Wnt and phosphatidylinositol-3 kinase (PI-3K) pathways was investigated using a colony-replating assay. The PI-3K pathway promoted differentiation of interleukin-3 (IL-3)-stimulated myelopoiesis via Akt, because inhibition of the PI-3K/Akt pathway with LY294002 or SH-5 increased proliferation. The involvement of canonical and non-canonical Wnt pathways was investigated using Wnt3a and Wnt5a respectively. Addition of the recombinant Wnts to IL-3 increased CFU-GM proliferation. Dkk-1, when combined with the Wnt proteins, abrogated the effects of Wnt3a but not Wnt5a. Surprisingly, the addition of Dkk-1 to LY294002 or SH-5 blocked their proliferative effects. We hypothesized that increased proliferation induced by PI-3K/Akt inhibitors was not mediated by downstream activation of the Wnt pathway but by induced endogenous production/release of Wnt proteins. The addition of SH-5 to IL-3 created an autocrine Wnt loop in CD34(+) cells, resulting in the phosphorylation of lipoprotein-receptor-related-protein 6. Furthermore, the addition of medium conditioned by CD34(+) cells cultured in IL-3 + SH-5 to IL-3 increased CFU-GM proliferation. This effect was abrogated by Dkk-1, suggesting that a Wnt in the conditioned medium increased proliferation. In summary, IL-3 via the PI-3K pathway promoted differentiation of myeloid progenitor cells through a decrease of endogenous Wnt production/release.


Subject(s)
Cell Differentiation/drug effects , Cell Proliferation/drug effects , Granulocyte-Macrophage Progenitor Cells/cytology , Intercellular Signaling Peptides and Proteins/pharmacology , Interleukin-3/pharmacology , Myeloid Progenitor Cells/metabolism , Signal Transduction/drug effects , Antigens, CD34 , Cells, Cultured , Humans , Myelopoiesis/drug effects , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins/pharmacology , Wnt Proteins/pharmacology , Wnt-5a Protein , Wnt3 Protein , Wnt3A Protein
5.
Am J Gastroenterol ; 103(8): 1952-8, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18637092

ABSTRACT

OBJECTIVES: Recent advances in regenerative medicine, including hematopoietic stem cell (HSC) transplantation, have brought hope for patients with severe alcoholic liver cirrhosis (ALC). The aim of this study was to assess the safety and efficacy of administering autologous expanded mobilized adult progenitor CD34+ cells into the hepatic artery of ALC patients and the potential improvement in the liver function. METHODS: Nine patients with biopsy-proven ALC, who had abstained from alcohol for at least 6 months, were recruited into the study. Following granulocyte colony-stimulating factor (G-CSF) mobilization and leukapheresis, the autologous CD34+ cells were expanded in vitro and injected into the hepatic artery. All patients were monitored for side effects, toxicities, and changes in the clinical, hematological, and biochemical parameters. RESULTS: On average, a five-fold expansion in cell number was achieved in vitro, with a mean total nucleated cell count (TNCC) of 2.3 x 10(8) pre infusion. All patients tolerated the procedure well, and there were no treatment-related side effects or toxicities observed. There were significant decreases in serum bilirubin (P < 0.05) 4, 8, and 12 wk post infusion. The levels of alanine transaminase (ALT) and aspartate transaminase (AST) showed improvement through the study period and were significant (P < 0.05) 1 wk post infusion. The Child-Pugh score improved in 7 out of 9 patients, while 5 patients had improvement in ascites on imaging. CONCLUSION: It is safe to mobilize, expand, and reinfuse autologous CD34+ cells in patients with ALC. The clinical and biochemical improvement in the study group is encouraging and warrants further clinical trials.


Subject(s)
Antigens, CD34/physiology , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cell Transplantation/methods , Liver Cirrhosis, Alcoholic/therapy , Adult Stem Cells/transplantation , Cell Culture Techniques , Cohort Studies , Female , Hepatic Artery , Humans , Infusions, Intra-Arterial , Male , Middle Aged , Treatment Outcome
6.
Hum Reprod ; 23(4): 928-33, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18238907

ABSTRACT

BACKGROUND: During pregnancy, fetal cells enter the maternal bloodstream resulting in fetal cell microchimerism. The fetal cells persist in the mother for decades and colonize a variety of maternal organs. They are associated with maternal autoimmune diseases and may also participate in tissue repair. The identity of the microchimeric cells is not certain but they must be able to persist long-term and have potential for multitissue differentiation. METHODS AND RESULTS: Here we tested the hypothesis that the fetal microchimeric cells are primitive stem cells, represented by CD34+ adherent cells, which have a wide potential for differentiation. We isolated these stem cells from the blood of pregnant females (n = 25) and detected fetal cells of the correct gender, using fluorescence in situ hybridization, in a high proportion (71% male fetuses and 90% female fetuses; false positive rate 11%, false negative rate 29%) of cases. By RT-PCR, we demonstrated that the cells express Oct-4, Nanog and Rex-1. No fetal cells were detected in the mononuclear or total CD34+ cell populations but high frequencies (mean 11.8%) of fetal cells were detected in the adherent CD34+ cell population. CONCLUSIONS: These results identify adherent CD34+ stem cells as candidate fetal microchimeric cells, which are capable of sustaining the fetal cell population in the long term and have the ability to colonize multiple tissues and organs.


Subject(s)
Chimerism , Fetus/cytology , Maternal-Fetal Exchange/physiology , Stem Cells/cytology , Adult , Antigens, CD34 , Blood , Case-Control Studies , Child, Preschool , Female , Fluorescence , Humans , Infant , Male , Pregnancy , Reverse Transcriptase Polymerase Chain Reaction
8.
Stem Cells ; 25(4): 844-51, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17185613

ABSTRACT

The control of symmetric and asymmetric division in the hematopoietic stem/progenitor cell population is critically important for the regulation of blood cell production. Asymmetric divisions depend on cell polarization, which may be conferred by location and/or interaction with neighboring cells. In this study, we sought evidence for polarization in CD34+ cells, which interact by binding to one another. In these cells, surface molecules became redistributed by mechanisms that included transport by lipid rafts, and the interacting cells were able to communicate via gap junctions. These changes were accompanied by modulation of cell cycle regulating proteins (p16(Ink4a), p27(kip1), cyclins D, and the retinoblastoma pathway proteins) and a reduction in progenitor cell proliferation in vitro. These results are consistent with an increase in asymmetric cell division kinetics. Accordingly, we found that interaction between CD34+ cells influenced the plane of cell division in a way that suggests unequal sharing of Notch-1 between daughter cell progeny. We conclude that interaction between CD34+ cells may coordinate cell function and participate in the control of hematopoietic stem/progenitor cell division kinetics.


Subject(s)
Antigens, CD34/physiology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Antigens, CD/physiology , Cell Aggregation , Cell Cycle , Cell Division , Gap Junctions/physiology , Humans , Kinetics , Lymphocyte Function-Associated Antigen-1/physiology , Membrane Microdomains/physiology , RNA, Small Interfering/genetics , Tissue Donors
9.
Clin Cancer Res ; 12(22): 6853-62, 2006 Nov 15.
Article in English | MEDLINE | ID: mdl-17121907

ABSTRACT

PURPOSE: Oncolytic herpes simplex virus type 1 (HSV-1) vectors show considerable promise as agents for cancer therapy. We have developed a novel recombinant HSV-1 virus (JS1/34.5-/47-) for purging of occult breast cancer cells from bone marrow of patients. Here, we evaluate the therapeutic efficacy of this oncolytic virus. EXPERIMENTAL DESIGN: Electron microscopy was used to determine whether human breast cancer and bone marrow cells are permissive for JS1/34.5-/47- infection. Subsequently, the biological effects of JS1/34.5-/47- infection on human breast cancer cells and bone marrow were established using cell proliferation and colony formation assays, and the efficiency of cell kill was evaluated. Finally, the efficiency of JS1/34.5-/47- purging of breast cancer cells was examined in cocultures of breast cancer cells with bone marrow as well as bone marrow samples from high-risk breast cancer patients. RESULTS: We show effective killing of human breast cancer cell lines with the JS1/34.5-/47- virus. Furthermore, we show that treatment with JS1/34.5-/47- can significantly inhibit the growth of breast cancer cell lines without affecting cocultured mononuclear hematopoietic cells. Finally, we have found that the virus is effective in destroying disseminated tumors cells in bone marrow taken from breast cancer patients, without affecting the hematopoietic contents in these samples. CONCLUSION: Collectively, our data show that the JS1/34.5-/47- virus can selectively target breast cancer cells while sparing hematopoietic cells, suggesting that JS1/34.5-/47- can be used to purge contaminating breast cancer cells from human bone marrow in the setting of autologous hematopoietic cell transplantation.


Subject(s)
Adenocarcinoma/pathology , Bone Marrow/pathology , Breast Neoplasms/pathology , Herpesvirus 1, Human , Oncolytic Viruses/physiology , Adenocarcinoma/virology , Biomarkers, Tumor/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/virology , Breast Neoplasms/therapy , Cell Death , Cell Line, Tumor , Cell Survival , Flow Cytometry , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/pathogenicity , Herpesvirus 1, Human/physiology , Humans , Keratin-19/metabolism , Mammary Glands, Human/pathology , Mammary Glands, Human/virology , Oncolytic Virotherapy/adverse effects
10.
Genes Chromosomes Cancer ; 45(12): 1121-9, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16955467

ABSTRACT

Chronic myeloid leukemia (CML) arises as a consequence of the expression of a chimeric fusion protein, p210BCR-ABL1, which is localized to the cytoplasm and has constitutive protein tyrosine kinase activity. Extensive publications report that p210BCR-ABL1 complexed with multiple cytoplasmic proteins can modulate several cell signaling pathways. However, while altered signaling states can be demonstrated in primary CML material, most of the reported analytical work on complexed proteins has been done in cell lines expressing p210BCR-ABL1. This has been necessary because primary hemopoietic cell lysates contain a degradative activity which rapidly and permanently destroys p210BCR-ABL1, precluding accurate p210BCR-ABL1 quantification by Western blotting or investigation of coimmunoprecipitating proteins in primary cells. This degradative activity has proven intractable to inhibition by conventional protease inhibitors. We show here that the degradative activity in primary cells is associated with cell lysosomes and is most likely to be an acid-dependent hydrolase. By lysing primary hemopoietic cells at high pH, we have demonstrated substantial inhibition of the p210BCR-ABL1-degradative activity and now report, to the best of our knowledge, the first published demonstration by coimmunoprecipitation of the association between p210BCR-ABL1 and cytoplasmic effector proteins in primary CML material.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Fusion Proteins, bcr-abl/metabolism , GRB2 Adaptor Protein/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Nuclear Proteins/metabolism , Proto-Oncogene Proteins c-cbl/metabolism , Caspases/metabolism , Cell Line, Tumor , Chloroquine/pharmacology , Humans , Hydrogen-Ion Concentration , Immunoprecipitation/methods , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukocytes, Mononuclear/chemistry , Leukocytes, Mononuclear/metabolism , Lysosomes/enzymology , Phosphorylation/drug effects , Signal Transduction , Tumor Cells, Cultured
11.
Stem Cells ; 24(7): 1822-30, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16556705

ABSTRACT

A phase I study was performed to determine the safety and tolerability of injecting autologous CD34(+) cells into five patients with liver insufficiency. The study was based on the hypothesis that the CD34(+) cell population in granulocyte colony-stimulating factor (G-CSF)-mobilized blood contains a subpopulation of cells with the potential for regenerating damaged tissue. We separated a candidate CD34(+) stem cell population from the majority of the CD34(+) cells (99%) by adherence to tissue culture plastic. The adherent and nonadherent CD34(+) cells were distinct in morphology, immunophenotype, and gene expression profile. Reverse transcription-polymerase chain reaction-based gene expression analysis indicated that the adherent CD34(+) cells had the potential to express determinants consistent with liver, pancreas, heart, muscle, and nerve cell differentiation as well as hematopoiesis. Overall, the characteristics of the adherent CD34(+) cells identify them as a separate putative stem/progenitor cell population. In culture, they produced a population of cells exhibiting diverse morphologies and expressing genes corresponding to multiple tissue types. Encouraged by this evidence that the CD34(+) cell population contains cells with the potential to form hepatocyte-like cells, we gave G-CSF to five patients with liver insufficiency to mobilize their stem cells for collection by leukapheresis. Between 1 x 10(6) and 2 x 10(8) CD34(+) cells were injected into the portal vein (three patients) or hepatic artery (two patients). No complications or specific side effects related to the procedure were observed. Three of the five patients showed improvement in serum bilirubin and four of five in serum albumin. These observations warrant further clinical trials.


Subject(s)
Antigens, CD34/metabolism , Granulocyte Colony-Stimulating Factor/blood , Granulocyte Colony-Stimulating Factor/metabolism , Hematopoietic Stem Cell Mobilization , Antigens, CD34/blood , Cell Adhesion/physiology , Cell Differentiation , Cells, Cultured , Female , Gene Expression , Hematopoietic Stem Cell Transplantation , Humans , Liver Diseases, Alcoholic/therapy , Liver Diseases, Parasitic/therapy , Male , Middle Aged , Multipotent Stem Cells/metabolism
12.
Clin Sci (Lond) ; 109(1): 13-25, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15966868

ABSTRACT

The biology of CML (chronic myeloid leukaemia) has been extensively investigated as the disease is a paradigm of neoplasms induced when a translocation results in expression of a novel fusion protein, in this instance p210(BCR-ABL). Although CML manifests itself principally as unregulated expansion of the myeloid lineage, the lesion is present in the stem cell population and it has long been assumed that disregulated stem cell kinetics must underlie the basic pathology of the disease. In this review, we present evidence that, in normal haemopoiesis, less primitive precursor cells retain considerable flexibility in their capacity to undergo self-renewal, allowing them to maintain lineage-specific homoeostasis without inflicting proliferative stress upon the stem cell population. This mechanism is dysregulated in CML and we have developed a self-renewal assay for CFU-GM (colony-forming unit-granulocyte/macrophage) which demonstrates that, in CML, the PI (proliferative index) of the myeloid progenitor cell population is increased. The ability to measure the PI as an endpoint of p210(BCR-ABL) expression gives considerable versatility to the in vitro investigation of putative therapeutic regimes in CML.


Subject(s)
Hematopoietic Stem Cells/pathology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/etiology , Benzamides , Cell Proliferation , Fusion Proteins, bcr-abl/metabolism , Humans , Imatinib Mesylate , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Philadelphia Chromosome , Piperazines/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Pyrimidines/therapeutic use , Signal Transduction , Translocation, Genetic
13.
Br J Haematol ; 129(3): 377-80, 2005 May.
Article in English | MEDLINE | ID: mdl-15842662

ABSTRACT

We have investigated functional outcome of challenging primary chronic myeloid leukaemia (CML) cells with Bcr-Abl fusion sequence-directed RNA interference (RNAi). We targeted the Bcr-Abl b3a2 variant, by RNAi, in primary chronic phase CML cells, and detected strikingly reduced proliferation of myeloid precursor cells expressing this variant. Lack of an effect in cells expressing a distinct Bcr-Abl variant confirmed the specificity of the response. Through the functional targeting of an oncogene in primary human tumour cells, we have demonstrated that Bcr-Abl enhances CML progenitor cell amplification, and that RNAi may be suitable for development as a specific anti-leukaemia treatment.


Subject(s)
Fusion Proteins, bcr-abl/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , RNA Interference , RNA, Neoplasm/genetics , Cell Proliferation , Humans , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , RNA, Small Interfering/genetics , Transfection
14.
Exp Hematol ; 32(1): 36-44, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14725899

ABSTRACT

OBJECTIVE: To investigate 1) the effects of lineage-specific cytokines (G-CSF and EPO) combined with ligands for different classes of cytokine receptors (common beta chain, gp130, and tyrosine kinase) on proliferation by human myeloid and erythroid progenitor cells; and 2) the signal transduction pathways associated with combinatorial cytokine actions. PATIENTS AND METHODS: CFU-GM and BFU-E were cloned in vitro. Secondary colony formation by replated CFU-GM and subcolony formation by BFU-E provided measures of progenitor cell proliferation. Studies were performed in the presence of cytokine combinations with and without signal transduction inhibitors. RESULTS: Proliferation by CFU-GM and BFU-E was enhanced synergistically when common beta chain receptor cytokines (IL-3 or GM-CSF) were combined with G-CSF or EPO, but not with gp130 receptor cytokines (LIF or IL-6) or tyrosine kinase receptor cytokines (SCF, HGF, Flt-3 ligand, or PDGF). Delayed addition studies with G-CSF+IL-3 and EPO+IL-3 demonstrated that synergy required the presence of both cytokines from the initiation of the culture. The Jak2-specific inhibitor, AG490, abrogated the effect of combining IL-3 with EPO but had no effect on the enhanced CFU-GM proliferation stimulated by IL-3+G-CSF. The PI3 kinase inhibitors LY294002 and wortmannin substituted for G-CSF in combination with IL-3 since proliferation in the presence of LY294002/wortmannin+IL-3 was enhanced to the same extent as in the presence of G-CSF+IL-3. In contrast, LY294002 and wortmannin inhibited proliferation in the presence of EPO and in the presence of EPO+IL-3. CONCLUSION: 1) IL-3 may activate different signal transduction pathways when combined with G-CSF and when combined with EPO; 2) different signal transducing intermediates regulate erythroid and myeloid progenitor cell proliferation; and 3) inhibition of the PI3 kinase pathway suppresses myeloid progenitor cell differentiation and thereby increases proliferation.


Subject(s)
Erythroid Precursor Cells/physiology , Hematopoietic Stem Cells/physiology , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins , Cell Differentiation/drug effects , Cell Division/drug effects , Cells, Cultured , Chromones/pharmacology , Enzyme Activation , Erythroid Precursor Cells/drug effects , Erythropoietin/pharmacology , Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cells/drug effects , Humans , Interleukin-3/pharmacology , Janus Kinase 2 , Mitogen-Activated Protein Kinases/physiology , Morpholines/pharmacology , Protein-Tyrosine Kinases/physiology , Signal Transduction
15.
Br J Haematol ; 122(3): 424-9, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12877669

ABSTRACT

The molecular basis of chronic myeloid leukaemia (CML) is well defined and highly consistent, yet prognosis varies considerably. This could reflect the biological diversity occurring in normal populations. We used a colony replating assay to measure the proliferative capacity of progenitor cells from 211 CML patients and 86 normal persons. Results were expressed as the frequency distributions of the proliferation index (PI) for individual cases. Normal PI values varied among individuals but were reproducible in individuals. The PIs for CML patients were moderately but significantly greater (P = 0.004) than normal values, consistent with increased progenitor cell proliferation in CML. Exposure of CML progenitor cells to the Abl-kinase inhibitor imatinib shifted their PI towards the normal range, implicating p210BCR-ABL. as a cause of the increased PI. The PIs of CML patients were higher than those of their human leucocyte antigen (HLA)-matched siblings PI (P = 0.003) and patient PI increased exponentially with sibling PI (r = 0.77; P = 0.001), but not with the PI values of HLA-matched unrelated individuals (P = 0.66). Finally, patients with high-risk prognostic scores (according to the Sokal or Hasford systems) had a significantly higher PI than those with low risk scores (P = 0.01 and 0.03 respectively). We conclude that heterogeneity in the CML patient population is analogous to the constitutional diversity in normal subjects.


Subject(s)
Fusion Proteins, bcr-abl/metabolism , Hematopoietic Stem Cells/pathology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Adult , Benzamides , Case-Control Studies , Cell Division , Female , Genetic Variation , Hematopoietic Stem Cells/drug effects , Humans , Imatinib Mesylate , Male , Middle Aged , Piperazines/pharmacology , Prognosis , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrimidines/pharmacology
16.
Br J Haematol ; 121(4): 643-8, 2003 May.
Article in English | MEDLINE | ID: mdl-12752107

ABSTRACT

Self-renewal is the most fundamental property of haemopoietic stem and progenitor cells. However, because of the need to produce differentiated cells, not all cell divisions involve self-renewal. We have used a colony replating assay to follow the fates of individual haemopoietic progenitor cell clones. For this, human myeloid colony-forming cells (CFCs) were cultured by standard methodology. Onset of proliferation and growth rates were established by a video recording method. Individual colonies were replated several times to document the rate of clonal extinction, and the numbers of secondary, tertiary and quaternary CFCs. The clonogenic population exhibited similar kinetics in terms of onset of proliferation and growth rate. Clonal extinction was progressive so that only 30 +/- 7% (mean +/- standard error of the mean; n = 4) of the original primary colonies formed quaternary colonies after the third replating step. However, individual primary CFCs that produced colonies throughout the experiment generated, on average, 40 +/- 8 secondary and tertiary CFCs overall. The values obtained in standard culture conditions were modified when granulocyte colony-stimulating factor (G-CSF) or G-CSF plus interleukin 3 were used to stimulate colony growth, showing that the kinetics of colony formation respond to extrinsic regulation. Examination of the replating potential of individual secondary colonies in the clones demonstrated that they generated different numbers of tertiary colonies. The data best fit a stochastic model of haemopoietic cell development where event probabilities can be modified by extracellular factors.


Subject(s)
Hematopoietic Stem Cells/cytology , Cell Division/physiology , Cell Survival , Cells, Cultured , Clone Cells/cytology , Colony-Forming Units Assay , Humans
17.
Exp Hematol ; 30(12): 1428-35, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12482505

ABSTRACT

Bone marrow from wild-type mice and mice with mutated Fas (lpr) or mutated Fas ligand (gld) was used to investigate the role of the Fas/FasL system in the regulation of myeloid progenitor cell kinetics.Granulocyte-macrophage colony-forming cells (CFU-GM) were measured by a standard colony assay and the proliferative activity of CFU-GM was measured by replating primary colonies and observing secondary colony formation. Fas expression was restored to lpr mouse bone marrow cells by retrovirus-mediated gene transfer and gld mouse marrow cells were treated with soluble FasL. Wild-type marrow cells were treated with YVAD (a caspase inhibitor) or anti-Fas monoclonal antibodies. There were greater frequencies of myeloid progenitor cells (CFU-GM) in lpr and gld mouse marrow compared to wild-type (WT) marrow (p = 0.0008). The proliferative capacity of CFU-GM was also significantly greater for lpr and gld CFU-GM compared to WT CFU-GM (p = 0.0003 and 0.0001, respectively). Retrovirus-mediated restoration of Fas into lpr marrow, and provision of soluble FasL (sFasL) to gld CFU-GM reduced CFU-GM proliferation to WT levels. Treatment of WT CFU-GM with YVAD or anti-FasL monoclonal antibody increased CFU-GM proliferation to the levels found in lpr and gld CFU-GM. YVAD significantly increased and anti-Fas significantly reduced the proliferative capacity of human CFU-GM (p = 0.015 and 0.04, respectively).Fas, FasL, and caspase activation may play an important role in regulating myeloid progenitor cell kinetics.


Subject(s)
Apoptosis , Membrane Glycoproteins/metabolism , Myeloid Progenitor Cells/cytology , fas Receptor/metabolism , Animals , Bone Marrow Cells/cytology , Caspases/metabolism , Cell Division/drug effects , Fas Ligand Protein , Humans , Kinetics , Membrane Glycoproteins/genetics , Membrane Glycoproteins/pharmacology , Mice , Mice, Knockout , Myeloid Progenitor Cells/metabolism , Transduction, Genetic , fas Receptor/genetics
19.
Br J Haematol ; 116(1): 162-5, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11841411

ABSTRACT

Combination of STI571, a tyrosine kinase inhibitor, with other drugs may be beneficial in the treatment of chronic myeloid leukaemia (CML). We measured the effects of STI571, AG490, farnesyltransferase inhibitor (FTI), interferon alpha (IFN-alpha), cytosine arabinoside (Ara-C) and all-trans retinoic acid (ATRA), singly and in combination, on clonogenic leukaemic cell proliferation. STI571, IFN-alpha and ATRA each reduced proliferation by 50-60%; AG490, FTI and Ara-C had less effect. Comparing the observed and expected (i.e. additive) effects of drug combinations showed STI571 + FTI, STI571 + AG490 and IFN-alpha + ATRA were additive; STI571 + IFN-alpha, IFN-alpha + Ara-C and STI571 + AG490 + FTI were less than additive. Thus, STI571 + FTI, STI571 + AG490 and IFN-alpha + ATRA may be better combination therapies for CML than STI571 + IFN-alpha, IFN-alpha + Ara-C or STI571 + AG490 + FTI.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Stem Cells/pathology , Alkyl and Aryl Transferases/antagonists & inhibitors , Area Under Curve , Benzamides , Cell Division/drug effects , Cytarabine/therapeutic use , Enzyme Inhibitors/therapeutic use , Farnesyltranstransferase , Humans , Imatinib Mesylate , Interferon-alpha , Piperazines , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrimidines/therapeutic use , Stem Cells/drug effects , Tretinoin/therapeutic use , Tyrphostins/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...