Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters











Publication year range
3.
Cell Cycle ; 14(19): 3146-54, 2015.
Article in English | MEDLINE | ID: mdl-26291130

ABSTRACT

We previously showed that incubation of chronic myeloid leukemia (CML) cells in very low oxygen selects a cell subset where the oncogenetic BCR/Abl protein is suppressed and which is thereby refractory to tyrosine kinase inhibitors used for CML therapy. In this study, salarin C, an anticancer macrolide extracted from the Fascaplysinopsis sponge, was tested as for its activity on CML cells, especially after their incubation in atmosphere at 0.1% oxygen. Salarin C induced mitotic cycle arrest, apoptosis and DNA damage. Salarin C also concentration-dependently inhibited the maintenance of stem cell potential in cultures in low oxygen of either CML cell lines or primary cells. Surprisingly, the drug also concentration-dependently enforced the maintenance of BCR/Abl signaling in low oxygen, an effect which was paralleled by the rescue of sensitivity of stem cell potential to IM. These results suggest a potential use of salarin C for the suppression of CML cells refractory to tyrosine kinase inhibitors.


Subject(s)
Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Macrolides/pharmacology , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/drug effects , Apoptosis/drug effects , Blotting, Western , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Damage/drug effects , Fusion Proteins, bcr-abl/metabolism , Humans , Oxygen/pharmacology
4.
Oncogene ; 33(34): 4359-64, 2014 Aug 21.
Article in English | MEDLINE | ID: mdl-24362524

ABSTRACT

The colony-stimulating factor-1 (CSF-1) and its receptor CSF-1R physiologically regulate the monocyte/macrophage system, trophoblast implantation and breast development. An abnormal CSF-1R expression has been documented in several human epithelial tumors, including breast carcinomas. We recently demonstrated that CSF-1/CSF-1R signaling drives proliferation of breast cancer cells via 'classical' receptor tyrosine kinase signaling, including activation of the extracellular signal-regulated kinase 1/2. In this paper, we show that CSF-1R can also localize within the nucleus of breast cancer cells, either cell lines or tissue specimens, irrespectively of their intrinsic molecular subtype. We found that the majority of nuclear CSF-1R is located in the chromatin-bound subcellular compartment. Chromatin immunoprecipitation revealed that CSF-1R, once in the nucleus, binds to the promoters of the proliferation-related genes CCND1, c-JUN and c-MYC. CSF-1R also binds the promoter of its ligand CSF-1 and positively regulates CSF-1 expression. The existence of such a receptor/ligand regulatory loop is a novel aspect of CSF-1R signaling. Moreover, our results provided the first evidence of a novel localization site of CSF-1R in breast cancer cells, suggesting that CSF-1R could act as a transcriptional regulator on proliferation-related genes.


Subject(s)
Breast Neoplasms/metabolism , Cell Proliferation , Chromatin/metabolism , Promoter Regions, Genetic , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Animals , Base Sequence , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Nucleus/metabolism , Feedback, Physiological , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , NIH 3T3 Cells , Protein Binding , Protein Transport , Signal Transduction , Solubility , Transcription, Genetic
5.
Cell Death Dis ; 4: e536, 2013 Mar 14.
Article in English | MEDLINE | ID: mdl-23492767

ABSTRACT

We determined the effects of severe hypoxia (∼0.1% O2) on acute myeloid leukemia cells expressing the AML1/ETO oncogene. Incubation of Kasumi-1 cells in hypoxia induced growth arrest, apoptosis and reduction of AML1/ETO protein expression. The conditional expression of AML1/ETO in U937-A/E cells showed that hypoxia induces marked apoptosis in AML1/ETO-expressing cells only, pointing to AML1/ETO as a factor predisposing cells to hypoxia-induced apoptosis. In AML1/ETO-expressing cells, hypoxia enhanced TRAIL expression and its proapoptotic effects. AML1/ETO was found to bind TRAIL promoter and induce TRAIL transcription, although TRAIL expression was restrained by a concomitant relative transcription block. In hypoxia, such a TRAIL repression was removed and an increase of TRAIL expression was induced. Finally, blocking anti-TRAIL antibodies markedly reduced (Kasumi-1 cells) or completely inhibited (U937-A/E cells) hypoxia-induced apoptosis. Taken together, these results indicated that hypoxia induces apoptosis in AML1/ETO-expressing cells via a TRAIL/caspase 8-dependent autocrine loop and that TRAIL is a key regulator of hypoxia-induced apoptosis in these cells.


Subject(s)
Cell Hypoxia/genetics , Core Binding Factor Alpha 2 Subunit/genetics , Gene Expression Regulation, Leukemic/drug effects , Leukemia, Myeloid, Acute/genetics , Proto-Oncogene Proteins/genetics , TNF-Related Apoptosis-Inducing Ligand/genetics , Transcription Factors/genetics , Antibodies/pharmacology , Apoptosis/drug effects , Base Sequence , Caspase 8/genetics , Caspase 8/metabolism , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Core Binding Factor Alpha 2 Subunit/metabolism , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Molecular Sequence Data , Oxygen/pharmacology , Promoter Regions, Genetic , Protein Binding , Proto-Oncogene Proteins/metabolism , RUNX1 Translocation Partner 1 Protein , Signal Transduction/drug effects , TNF-Related Apoptosis-Inducing Ligand/antagonists & inhibitors , TNF-Related Apoptosis-Inducing Ligand/metabolism , Transcription Factors/metabolism , Transcription, Genetic/drug effects
6.
Oncogene ; 27(12): 1767-78, 2008 Mar 13.
Article in English | MEDLINE | ID: mdl-17891169

ABSTRACT

We analysed the in vitro effects of a new hydroxamate derivative, ITF2357, on AML cells. ITF2357 potently induced histone acetylation. ITF2357 0.1 microM blocked proliferation and induced apoptosis in AML1/ETO-positive Kasumi-1 cells, while AML1/ETO-negative HL60, THP1 and NB4 cell lines were sensitive only to 1 microM ITF2357. Apoptosis was induced by 0.1 microM ITF2357 in AML1/ETO-positive primary blasts and U937-A/E cells induced to express AML1/ETO, but not in U937-A/E cells non-expressing AML1/ETO. In Kasumi-1 cells 0.1 microM ITF2357 induced AML1/ETO degradation through a caspase-dependent mechanism. ITF2357 0.1 microM also determined DNMT1 efflux from, and p300 influx to, the nucleus. Moreover, 0.1 microM ITF2357 determined local H4 acetylation and release of DNMT1, HDAC1 and AML1/ETO, paralleled by recruitment of p300 to the IL-3 gene promoter. ITF2357 treatment, however, did not induce re-expression of IL-3 gene. Accordingly, the methylation level of IL-3 promoter, as well as of several other genes, was unmodified. In conclusion, ITF2357 emerged as an anti-leukaemic agent very potent on AML cells, and on AML1/ETO-positive cells in particular. More relevantly, clearly emerged from our results that ITF2357 could be an ideal agent to treat AML subtypes presenting AML1/ETO fusion protein which determine HDAC involvement in leukaemogenesis.


Subject(s)
Antineoplastic Agents/pharmacology , Core Binding Factor Alpha 2 Subunit/biosynthesis , DNA-Binding Proteins/biosynthesis , Histone Deacetylase Inhibitors , Hydroxamic Acids/pharmacology , Leukemia/drug therapy , Proto-Oncogene Proteins/biosynthesis , Transcription Factors/biosynthesis , Acetylation , Cell Line, Tumor , Dose-Response Relationship, Drug , HL-60 Cells , Humans , Leukemia/enzymology , Leukemia/pathology , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/enzymology , RUNX1 Translocation Partner 1 Protein , U937 Cells
8.
Gut ; 55(7): 961-8, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16368781

ABSTRACT

BACKGROUND: We previously demonstrated the existence of two distinct subsets of T cell receptor (TCR)alphabeta+CD8alphabeta+ single positive (SP) cells in human postnatal thymus which express the chemokine receptor CCR7 or CXCR3 and migrate in vitro in response to their specific ligands. AIM: To investigate whether these two CD8+ thymocyte subsets had distinct peripheral colonisation. METHODS: TCRalphabeta+CD8+ SP cells were obtained from normal postnatal thymus, mesenteric lymph node (LNs), small bowel, and peripheral blood (PB) specimens. Cells were then evaluated for expression of surface molecules, cytolytic potential, telomere length, and profile of cytokine production. RESULTS: CD8+CCR7+CXCR3- thymocytes exhibited CD62L, in common with those which localise to LNs. In contrast, CD8+CCR7-CXCR3+ thymocytes lacked CD62L but exhibited CD103, similar to intraepithelial lymphocytes (IELs) present in the gut mucosa where the CXCR3 ligand, CXCL10, and the CD103 ligand, E-cadherin, are highly and consistently expressed. In addition, thymocytes and gut CD8+CXCR3+CD103+ cells showed comparable telomere length, which was higher than that of PB CXCR3+CD8+ T cells. However, both of these populations contained perforin and granzyme A, and displayed the ability to produce interferon gamma and interleukin 2. Of note, CXCR3 deficient, in comparison with wild-type C57Black/6, mice showed decreased proportions of CD3+CD8alphabeta+ and increased proportions of CD3+CD8alphaalpha+ lymphocytes at gut level. Moreover, adoptive transfer of CD3+CD8alphabeta+ thymocytes from wild-type into CXCR3 deficient mice resulted in a significant increase in CD3+CD8alphabeta+ T cells in the gut mucosa but not in other tissues. CONCLUSIONS: The results of this study demonstrate the existence of a previously unrecognised subset of TCRalphabeta+CD8alphabeta+ SP CXCR3+CD103+ thymocytes which share phenotypic and functional features with CD8+ IELs, thus suggesting the possibility of their direct colonisation of the gut mucosa.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Integrins/analysis , Intestinal Mucosa/immunology , Receptors, Chemokine/analysis , Adoptive Transfer , Adult , Analysis of Variance , Animals , CD8-Positive T-Lymphocytes/chemistry , CD8-Positive T-Lymphocytes/ultrastructure , Cell Separation/methods , Child, Preschool , Flow Cytometry , Humans , Immunohistochemistry/methods , Infant , Infant, Newborn , Interleukins/biosynthesis , Male , Mice , Mice, Knockout , Mice, Mutant Strains , Receptors, Antigen, T-Cell, alpha-beta/analysis , Receptors, CCR7 , Receptors, CXCR3 , Receptors, Chemokine/genetics , Telomere/ultrastructure
9.
J Biol Chem ; 276(42): 39027-36, 2001 Oct 19.
Article in English | MEDLINE | ID: mdl-11495898

ABSTRACT

Survival of memory B lymphocytes is tightly linked to the integrity of the Bcl-2 protein and is regulated by a nerve growth factor (NGF) autocrine circuit. In factor-starved memory B cells, the addition of exogenous NGF promptly induced p38 mitogen-activated protein kinase (MAPK), but not c-Jun N-terminal kinase (JNK), dephosphorylation. Conversely, withdrawal of endogenous NGF was followed by p38 MAPK activation and translocation onto mitochondria, whereby it combined with and phosphorylated Bcl-2, as assessed by co-immunoprecipitation and kinase assays in vivo and in vitro. Mitochondria isolated from human memory B cells, then exposed to recombinant p38 MAPK, released cytochrome c, as did mitochondria from Bcl-2-negative MDCK cells loaded with recombinant Bcl-2. Apoptosis induced by NGF neutralization could be blocked by the specific p38 MAPK inhibitor SB203580 or by Bcl-2 mutations in Ser-87 or Thr-56. These data demonstrate that the molecular mechanisms underlying the survival factor function of NGF critically rely upon the continuous inactivation of p38 MAPK, a Bcl-2-modifying enzyme.


Subject(s)
Apoptosis , B-Lymphocytes/pathology , Cytochrome c Group/metabolism , JNK Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Nerve Growth Factor/metabolism , Nerve Growth Factor/physiology , Proto-Oncogene Proteins c-bcl-2/metabolism , Animals , Cell Nucleus/metabolism , Cells, Cultured , Cytosol/metabolism , DNA Fragmentation , Enzyme Inhibitors/pharmacology , Humans , Imidazoles/pharmacology , Immunologic Memory , MAP Kinase Kinase 4 , Microscopy, Fluorescence , Mitochondria/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Precipitin Tests , Protein Binding , Protein Transport , Pyridines/pharmacology , Rats , Recombinant Proteins/metabolism , Serine/chemistry , Threonine/chemistry , Time Factors , p38 Mitogen-Activated Protein Kinases
10.
J Immunol ; 166(3): 1583-9, 2001 Feb 01.
Article in English | MEDLINE | ID: mdl-11160199

ABSTRACT

We previously reported that macrophage activators such as LPS, IL-2, and IL-4 down-modulate the M-CSFR via a mechanism involving protein kinase C and phospholipase C. In this study, we showed that M-CSFR is shed from macrophage surface and identified the protease responsible for M-CSFR cleavage and down-modulation. The shedding of M-CSFR elicited by phorbol esters (tetradecanoylphorbol myristate acetate (TPA)) or LPS in murine BAC.1-2F5 macrophages was prevented by cation chelators, as well as hydroxamate-based competitive inhibitors of metalloproteases. We found that the protease cleaving M-CSFR is a transmembrane enzyme and that its expression is controlled by furin-like serine endoproteases, which selectively process transmembrane metalloproteases. M-CSFR down-modulation was inhibited by treating cells in vivo, before TPA stimulation, with an Ab raised against the extracellular, catalytic domain of proTNF-converting enzyme (TACE). TACE expression was confirmed in BAC.1-2F5 cells and found inhibited after blocking furin-dependent processing. Using TACE-negative murine Dexter-ras-myc cell monocytes, we found that in these cells TPA is unable to down-modulate M-CSFR expression. These data indicated that TACE is required for the TPA-induced M-CSFR cleavage. The possibility that the cleavage is indirectly driven by TACE via the release of TNF was excluded by treating cells in vivo with anti-TNF Ab. Thus, we concluded that TACE is the protease responsible for M-CSFR shedding and down-modulation in mononuclear phagocytes undergoing activation. The possible physiological relevance of this mechanism is discussed.


Subject(s)
Macrophage Activation/immunology , Macrophages/enzymology , Macrophages/immunology , Metalloendopeptidases/metabolism , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Tumor Necrosis Factor-alpha/metabolism , ADAM Proteins , ADAM17 Protein , Animals , Antibodies, Monoclonal/pharmacology , Catalytic Domain/immunology , Cell Line , Cell Line, Transformed , Down-Regulation/drug effects , Down-Regulation/immunology , Enzyme Activation/drug effects , Enzyme Activation/immunology , Humans , Hydrolysis , Lipopolysaccharides/pharmacology , Macrophage Activation/drug effects , Macrophages/drug effects , Macrophages/metabolism , Metalloendopeptidases/immunology , Metalloendopeptidases/isolation & purification , Mice , Mice, Inbred A , Mice, Inbred BALB C , Mice, Inbred C57BL , Monocytes/drug effects , Monocytes/enzymology , Monocytes/immunology , Receptor, Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Tetradecanoylphorbol Acetate/pharmacology
11.
Methods Mol Med ; 55: 323-38, 2001.
Article in English | MEDLINE | ID: mdl-21312115

ABSTRACT

Apoptosis, or programmed cell death, represents in cell biology a functional program as important as cell growth or differentiation. Programmed cell death is of basic importance for the development of multicellular organisms and its basic mechanisms are conserved during the evolution of metazoa. Mammalian cells exhibit several different apoptotic pathways that converge to a common endpoint. Each pathway is triggered by a different stimulus: growth factor default, irradiation, induction of the p53 oncosuppressor protein, glucocorticoid hormones (in lymphocytes), ligand binding to Fas/APO (CD95), or tumor necrosis factor receptor (TNF-R), perforin secreted by cytotoxic T cells (reviewed by Hale et al. [1]). As opposed to necrosis, apoptosis is a "clean" process: as the cell shrinks, the cell membrane turns into the "apoptotic shell," the nucleus is condensed and reduced in volume, and eventually the cell disappears from the tissue, due to phagocytosis by neighboring cells or professional phagocytes, such as macrophages.

12.
J Leukoc Biol ; 68(2): 260-6, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10947071

ABSTRACT

We evaluated the synthesis of nitric oxide (NO) and of the neurotoxic kynurenine metabolites 3OH-kynurenine and quinolinic acid (QUIN) in interferon-gamma (IFN-gamma)-activated macrophages of the murine BACl.2F5 cell line with the aim of investigating the roles of mononuclear phagocytes in inflammatory neurological disorders. IFN-gamma induced indoleamine 2,3-dioxygenase (IDO) and NO synthase (NOS) and increased the synthesis of 3OH-kynurenine, QUIN, and NO that accumulated in the incubation medium where they reached neurotoxic levels. Macrophage exposure to norharmane, an IDO inhibitor, resulted in a decreased formation of not only the kynurenine metabolites but also NO. The inhibition of NO synthesis could not be ascribed to reduced NADPH availability or decreased NOS induction. Norharmane inhibited NOS activity also in coronary vascular endothelial cells and in isolated aortic rings. Our findings suggest that activated macrophages release large amounts of neurotoxic molecules and that norharmane may represent a prototype compound to study macrophage involvement in inflammatory brain damage.


Subject(s)
Interferon-gamma/pharmacology , Kynurenine/biosynthesis , Macrophage Activation , Macrophages/physiology , Nitric Oxide Synthase/physiology , Quinolinic Acid/metabolism , Tryptophan Oxygenase/physiology , Animals , Cell Line , Indoleamine-Pyrrole 2,3,-Dioxygenase , Macrophage Activation/drug effects , Mice , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase Type II , Tryptophan Oxygenase/antagonists & inhibitors
14.
Blood ; 95(12): 3959-63, 2000 Jun 15.
Article in English | MEDLINE | ID: mdl-10845934

ABSTRACT

Fes is a nonreceptor tyrosine kinase expressed at the highest level in macrophages. We previously showed that the overexpression of c-fes in murine macrophages of the BAC-1.2F5 cell line renders these cells independent of macrophage colony-stimulating factor (MCSF) for survival and proliferation, although no direct relationship could be established between tyrosine-phosphorylated substrates of Fes- and MCSF receptor-dependent signaling and mitogenesis. In this study, we investigated whether the mitogen-activated protein kinase (MAPK) pathway is involved in the growth factor-independent growth of v-fes-overexpressing macrophages. We found a constitutively increased phosphorylation of extracellularly regulated kinase (ERK) in v-fes-overexpressing macrophages as compared with mock-infected cells. This finding was associated with activation of mitogen/extracellular signal-regulated kinase (MEK) and with constitutive localization of ERK in the nucleus. Treatment of v-fes-overexpressing cells with the MEK-specific inhibitor PD98059 markedly reduced cell growth, hyperphosphorylation, and nuclear localization of ERK, indicating that the MAPK pathway mediates the mitogenic effect of v-fes. (Blood. 2000;95:3959-3963)


Subject(s)
Fusion Proteins, gag-onc/physiology , Macrophages/cytology , Macrophages/physiology , Mitogen-Activated Protein Kinases/metabolism , Receptor, Macrophage Colony-Stimulating Factor/physiology , Animals , Cell Division/physiology , Cell Line , Fusion Proteins, gag-onc/genetics , Macrophage Colony-Stimulating Factor/pharmacology , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Recombinant Fusion Proteins/metabolism , Signal Transduction
15.
Leukemia ; 14(4): 735-9, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10764163

ABSTRACT

In liquid cultures of murine bone marrow cells stimulated with interleukin-3 and granulocyte/macrophage colony-stimulating factor, hypoxia (1% oxygen) induced a reversible block of hematopoiesis, maintaining the progenitors' expansion potential unreduced. Progenitors repopulating day-14 hypoxic cultures with cells or granulocyte/macrophage colony-forming units (CFU-GM) were found, on the basis of their maintenance in hypoxia (12% and 76%, respectively), to belong to different subsets, the latter being much more efficiently maintained. The maintenance in hypoxic cultures of progenitors detectable by marrow-repopulating ability (MRA) assay was 18% for MRAcell progenitors and 69% for MRACFU progenitors. Thus, the repopulation of hypoxic cultures with cells or CFU-GM closely reflected the presence of progenitors capable of repopulating, with cells or CFU-GM, the bone marrow of lethally irradiated syngeneic animals. Progenitors repopulating hypoxic cultures were, like MRA progenitors, significantly resistant to 5-fluorouracil, progenitors repopulating cultures with CFU-GM being two-fold more resistant than those repopulating cultures with cells. We concluded that the repopulation of day-14 hypoxic cultures occurring after their transfer to air is to be considered an indicator of the maintenance of MRA progenitors in hypoxia. The relevance of these results to stem cell biology and their potential practical applications are discussed.


Subject(s)
Bone Marrow Cells/cytology , Cell Hypoxia , Animals , Antimetabolites/pharmacology , Cells, Cultured , Colony-Forming Units Assay , Drug Resistance , Fluorouracil/pharmacology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Mice , Mice, Inbred CBA , Radiation Chimera
16.
Br J Haematol ; 108(2): 424-9, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10691876

ABSTRACT

We developed previously a hypoxic culture system in which progenitors endowed with marrow-repopulating ability (MRA), unlike committed progenitors, were selected and maintained better than in air. We report here an improvement to this system targeted at combining the maintenance of progenitors sustaining MRA with the numerical expansion of multipotent and committed progenitors. Murine bone marrow cells were incubated at 1% oxygen in liquid medium supplemented with stem cell factor, granulocyte colony-stimulating factor, interleukin-6 and interleukin-3. In day 8 hypoxic cultures, the numbers of high proliferative potential and granulocyte/macrophage colony-forming cells (HPP-CFC and CFU-GM) were increased with respect to time zero. Colonies generated by HPP-CFC derived from hypoxic cultures exhibited a high replating ability, whereas colonies generated by HPP-CFC derived from control cultures exhibited a low replating ability. MRA was fully maintained in hypoxia and markedly reduced in air. Thus, severe hypoxia is able to ensure a full maintenance of progenitors sustaining MRA, together with a significant expansion of in vitro-detectable clonogenic progenitors, including those endowed with replating ability. This system could contribute to the improvement of current techniques for the in vitro treatment of human haematopoietic cell populations before transplantation.


Subject(s)
Bone Marrow Cells/cytology , Hematopoietic Stem Cells/cytology , Oxygen , Animals , Cell Division , Cell Survival , Cells, Cultured , Female , Male , Mice , Mice, Inbred CBA
17.
Transfusion ; 40(12): 1482-8, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11134568

ABSTRACT

BACKGROUND: The liquid culture of murine bone marrow cells at 1-percent oxygen maintains the balance between primative progenitor cell renewal and clonogenic progenitor expansion better than that at 20-percent oxygen. These results are of potential interest for the ex vivo expansion of human progenitor cells, as low O(2) tension could preserve the engraftment potential of cultured apheresis products. STUDY DESIGN AND METHODS: G-CSF-mobilized blood cells collected by apheresis, now the main source of progenitor cells for autologous transplantation, were cultured at 1-percent and 20-percent O(2) for 7 days in serum-free liquid cultures in the presence of IL-3 and SCF (5 ng/mL). The growth of the clonogenic progenitors (CFU-GM, BFU-E, CFU-Mix) and of the more primitive human HPCs that are capable of generating clongenic progenitors in secondary liquid culture, as well as the proliferation and differentiation of total and CD34+ cells, was analyzed. RESULTS: The expansion of CD34+ cells and of clonogenic progenitors was significantly lower in liquid cultures at 1-percent O(2) than at 20-percent O(2). On the contrary, the primitive human HPCs were better maintained and expanded at 1-percent O(2), although the number of CD34+ cells remaining quiescent was lower. After 7 days of liquid culture at 1-percent or 20-percent O(2) the percentage of CD34+ cells was similar. However, the CD34+ cells that divided more than four times (PKH2 staining) were more numerous in liquid cultures incubated at 1-percent O(2). CONCLUSION: When cultured at 1-percent O(2) for 7 days in presence of IL-3 and SCF, the CD34+ cells present in apheresis components underwent more cell divisions and better maintained their primitive progenitor cell potential. As suggested by previous results in mice, our data on human cells emphasize the potential interest of cultures at low O(2) tension (1%) for cell therapy protocols aimed at expanding primitive HPCs in autografts.


Subject(s)
Hematopoietic Stem Cells/cytology , Oxygen/analysis , Cell Count , Cell Cycle/physiology , Cell Differentiation/physiology , Cell Division/drug effects , Cells, Cultured , Culture Media, Serum-Free/chemistry , Humans , Lymphoma/blood , Oxygen/pharmacology
18.
FEBS Lett ; 459(2): 191-4, 1999 Oct 08.
Article in English | MEDLINE | ID: mdl-10518016

ABSTRACT

Fibroblast growth factor receptor (class IV) shares a certain degree of similarity with class III members like platelet-derived growth factor and macrophage-colony-stimulating factor receptors, which, once activated, are substrates of low M(r) phosphotyrosine protein phosphatase. Up until now no phosphotyrosine phosphatase has been shown to act on this receptor in vivo. Here we demonstrate that low M(r) phosphotyrosine protein phosphatase is able to reduce receptor tyrosine phosphorylation and cell proliferation in response to basic fibroblast growth factor. Contrary to what was previously observed for platelet-derived growth factor, during cell stimulation with basic fibroblast growth factor, no enzyme redistribution among cellular compartments is observed.


Subject(s)
Isoenzymes/metabolism , Protein Tyrosine Phosphatases/metabolism , Proto-Oncogene Proteins , Receptors, Fibroblast Growth Factor/metabolism , 3T3 Cells , Animals , Biological Transport , Fibroblast Growth Factors/metabolism , Macrophage Colony-Stimulating Factor/metabolism , Mice , Molecular Weight , Phosphorylation , Signal Transduction
19.
Biochem Biophys Res Commun ; 253(2): 300-4, 1998 Dec 18.
Article in English | MEDLINE | ID: mdl-9878532

ABSTRACT

The interference of low-molecular-weight phosphotyrosine protein phosphatase with the macrophage response to macrophage colony-stimulating factor was investigated. This paper shows that this phosphatase, already known to be involved in platelet-derived growth factor receptor signaling, is physiologically expressed in murine macrophages and dephosphorylates in vitro macrophage colony-stimulating factor receptor molecules immunoprecipitated from macrophage colony-stimulating factor-stimulated macrophages. We obtained the first demonstration that a phosphotyrosine-specific protein phosphatase dephosphorylates the macrophage colony-stimulating factor receptor in vivo and reduces the mitogenic response to macrophage colony-stimulating factor. The data indicate that low-molecular-weight phosphotyrosine protein phosphatase is a negative regulator of macrophage colony-stimulating factor receptor signaling.


Subject(s)
Macrophage Colony-Stimulating Factor/physiology , Mitogens/physiology , Protein Tyrosine Phosphatases/biosynthesis , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Receptor, Macrophage Colony-Stimulating Factor/physiology , Tyrosine/metabolism , 3T3 Cells , Animals , Cell Count , Cell Line , DNA/metabolism , Humans , Macrophages/cytology , Macrophages/enzymology , Macrophages/metabolism , Mice , Mitogens/antagonists & inhibitors , Molecular Weight , Phosphorylation/drug effects , Protein Tyrosine Phosphatases/physiology , Receptor, Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Signal Transduction/drug effects , Thymidine/metabolism , Transfection , Tritium/metabolism , Tyrosine/antagonists & inhibitors
20.
Exp Hematol ; 25(11): 1187-94, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9328456

ABSTRACT

Incubation in severe hypoxia (1% oxygen) increased the number of erythroid bursts generated from full-term CD34+, or premature mononucleated, human cord blood (CB) cells, in semisolid cultures containing stem cell factor (SCF), interleukin (IL)-3 and erythropoietin (EPO). Severe hypoxia also enhanced the maintenance of erythroid burst-forming units (BFU-E) in CB cell liquid cultures. These positive effects of hypoxia on the maintenance and cloning efficiency of BFU-E did not extend to the other progenitors assayed. Hypoxia, on the other hand, markedly reduced the size and level of hemoglobinization of bursts and, in liquid cultures, suppressed the growth factor-stimulated numerical increase in BFU-E and inhibited the expression of CD36, a marker of erythroid colony-forming units and maturing erythroid precursors. However, when transferred to clonal assays incubated in air, cells from liquid cultures incubated in hypoxia or in air generated fully expanded and hemoglobinized bursts, suggesting that in hypoxia the clonogenic potential of BFU-E was maintained and the development of erythroid clones reversibly inhibited. These results indicate that hypoxia inversely regulates two subsequent phases of erythropoiesis, i.e., it enhances the maintenance of BFU-E and the early development of erythroid clones but inhibits the terminal expansion and maturation of these clones. The cloning of CB cells selected for CD34 positivity, when compared with that of the total population of mononucleated CB cells, revealed that the early development of erythroid bursts was either hypoxia-enhanced or hypoxia-insensitive, reflecting the existence of two different types of BFU-E. Hypoxia-enhanced BFU-E are relatively immature, are maintained in hypoxia but not in air, and account for a large part of CD34+ BFU-E and for a high percentage of the BFU-E in premature CB. Hypoxia-insensitive BFU-E are mostly CD34- and are largely predominant in full-term CB, and most probably correspond to a more mature type of BFU-E.


Subject(s)
Cell Hypoxia , Growth Substances/pharmacology , Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Antigens, CD/analysis , Antigens, CD34/analysis , CD36 Antigens/analysis , Clone Cells , Colony-Forming Units Assay , Erythropoiesis/drug effects , Erythropoiesis/physiology , Erythropoietin/pharmacology , Fetal Blood/cytology , Hematopoiesis/drug effects , Hematopoietic Stem Cells/drug effects , Humans , Immunophenotyping , Infant, Newborn , Infant, Premature , Interleukin-3/pharmacology , Recombinant Proteins/pharmacology , Stem Cell Factor/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL