Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
1.
Leukemia ; 21(4): 714-24, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17375123

ABSTRACT

Type I interferon (IFN) is shown to control the reversible quiescence of a primitive human bone marrow mesenchymal stem cell (MSC) subpopulation. A 24 h pre-treatment of Stro1+/GlycoA- or CD45-/GlycoA- subpopulations with a monoclonal antibody (mAb) against the IFNAR1 chain of the human type I IFN receptor (64G12), or with a polyclonal anti-IFNalpha antibody, resulted in a marked increase in the number of very large colonies (CFU-F >3000 cells) obtained in the presence of low, but necessary, concentrations of bFGF. Over a 2-month culture period, this short activation promoted a faster and greater amplification of mesenchymal progenitors for adipocytes and osteoblasts. Activation correlated with inhibition of STAT1 and STAT2 phosphorylation and of STAT1 nuclear translocation. A non-neutralizing anti-IFNAR1 mAb was ineffective. We demonstrate that control and activated MSCs express ST3GAL3, a sialyltransferase necessary to produce the embryonic antigens SSEA-3 and -4. Interestingly, activated MSC progeny expressed SSEA-3 and -4 at a higher level than control cultures, but this was not correlated with a significant expression of other embryonic markers. As MSCs represent an essential tool in tissue regeneration, the use of 64G12, which rapidly recruits a higher number of primitive cells, might increase amplification safety for cell therapy.


Subject(s)
Interferon-alpha/physiology , Interferon-beta/physiology , Mesenchymal Stem Cells/cytology , Bone Marrow Cells/cytology , Cell Culture Techniques , Cell Differentiation , Cell Division/drug effects , Colony-Forming Units Assay , DNA Primers , Extracellular Matrix/physiology , Humans , Immunophenotyping , Kinetics , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/immunology , Polymerase Chain Reaction , Transforming Growth Factor beta1/pharmacology
2.
Pathol Biol (Paris) ; 54(2): 94-9, 2006 Mar.
Article in French | MEDLINE | ID: mdl-16140469

ABSTRACT

Human embryonic stem cells (hESCs) are derived from pre-implantation embryos given to research with the informed consent of the parents. These cells cannot give rise to a human being: they are not totipotent. They have an unlimited self-renewal capacity and they can generate the three embryonic germ layers, their respective derivatives and the extra-embryonic tissues: they are pluripotent. These cells represent an outstanding material for understanding functional genomics of not yet characterized human genes. They will be an important tool for pharmaceutical and clinical research.


Subject(s)
Blastocyst/cytology , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/physiology , Stem Cell Transplantation , Cell Division , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Humans , Research
3.
Rev Med Brux ; 25(3): 153-9, 2004 Jun.
Article in French | MEDLINE | ID: mdl-15291447

ABSTRACT

During the last decade, new insights in cellular and molecular biology have opened new avenues in cancer immunotherapy. Two distinct modalities have been developed: adoptive immunotherapy and anti-tumoral vaccination (active immunotherapy). We will first describe the main strategies of adoptive immunotherapy and then elaborate on the protocols of anti-tumoral vaccination against tumor associated antigens (TAA). In that context, we will pay peculiar attention on the pivotal role of dendritic cells (DC) as natural adjuvant.


Subject(s)
Cancer Vaccines/therapeutic use , Melanoma/immunology , Melanoma/therapy , Dendritic Cells/immunology , Humans , Immunotherapy/methods , Melanoma/genetics
4.
Eur Cytokine Netw ; 12(1): 172-6, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11282562

ABSTRACT

We have previously demonstrated that purified human fibrinogen (Fg), a major plasma component removed during serum preparation, shows mitogenic properties towards lymphoma cells and normal human hematopoietic progenitors. Indeed, adding Fg with IL-3 to a serum-containing medium stimulates growth of human CD34+ progenitors. In this report, we show in serum-free medium, that this stimulating effect only occurs in the presence of IL-6. To clarify the cooperative effect between Fg and IL-6, the kinetics of IL-6 receptor (IL-6R) mRNA expression in CD34+ cells have been analyzed by semi-quantitative in situ hybridization. In the presence of both IL-3 and Fg, more cells express IL-6R mRNA, and this expression per cell is significantly greater than with each factor added separately. These results suggest that Fg does not promote the growth of normal cells by itself, but sensitizes the cells to cytokines. Fg behaves not as a "progression" factor but as a typical "competence" factor, which induces a faster and greater IL-6R expression in early human hematopoietic progenitors by cooperating with other cytokines.


Subject(s)
Antigens, CD34/immunology , Fibrinogen/physiology , Gene Expression Regulation/physiology , Hematopoietic Stem Cells/metabolism , Interleukin-6/physiology , RNA, Messenger/genetics , Receptors, Interleukin-6/genetics , Hematopoietic Stem Cells/immunology , Humans , In Situ Hybridization
5.
Gene Ther ; 7(20): 1790-4, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11083502

ABSTRACT

Various protocols have been described to optimize gene transfer into hematopoietic cells. However, most of these methods do not specify whether they are associated with an improved transduction of the more primitive stem/progenitor cells, the best candidates for long-term engraftment. The majority of these primitive cells remains in quiescence because of the negative control of TGF-beta1, effective on these cells at low concentrations (10 pg/ml). In this study, CD34- cells were activated by a 10 h pretreatment with anti-TGF-beta1 followed by four successive retroviral supernatant incubations of 6 h each. After 12 h (two incubations), a significant increase in TGF-beta1 mRNA in CD34+ cells was observed. We wondered whether neo-synthesized autocrine TGF-beta1 could induce reversion to quiescence of the more primitive CD34+ cells transduced after one cell cycle. This would prevent their subsequent detection in a classic clonal assay. Using the HPP-Q assay comparing a rapid mixed colony assay with or without anti-TGF-beta1, we indeed observed, that in clonal growth conditions the more primitive transduced cells were activated and detectable only with anti-TGF-beta1. Therefore, this assay represents not only a rapid means to detect quiescent multipotent stem/progenitor cells but also a necessary step for the detection of the more primitive transduced cells which have returned to quiescence after retroviral induction of TGF-beta1 secretion.


Subject(s)
Gene Transfer Techniques , Hematopoietic Stem Cells , Membrane Glycoproteins , Transforming Growth Factor beta/metabolism , Antigens, CD/metabolism , CD24 Antigen , Cell Culture Techniques , Cell Division , Gene Expression , Humans , RNA, Messenger/genetics , Transduction, Genetic , Transforming Growth Factor beta/genetics
6.
Blood ; 96(6): 2022-36, 2000 Sep 15.
Article in English | MEDLINE | ID: mdl-10979943

ABSTRACT

Hematopoiesis is a remarkable cell-renewal process that leads to the continuous generation of large numbers of multiple mature cell types, starting from a relatively small stem cell compartment. A highly complex but efficient regulatory network is necessary to tightly control this production and to maintain the hematopoietic tissue in homeostasis. During the last 3 decades, constantly growing numbers of molecules involved in this regulation have been identified. They include soluble cytokines and growth factors, cell-cell interaction molecules, and extracellular matrix components, which provide a multifunctional scaffolding specific for each tissue. The cloning of numerous growth factors and their mass production have led to their possible use for both fundamental research and clinical application.


Subject(s)
Hematopoiesis , Transforming Growth Factor beta/physiology , Animals , Humans , Signal Transduction/physiology
7.
J Cell Physiol ; 184(1): 80-5, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10825236

ABSTRACT

Transforming growth factor-beta1 (TGF-beta1) has been described as an efficient growth inhibitor that maintains the CD34(+) hematopoietic progenitor cells in quiescence. The concept of high proliferative potential-quiescent cells or HPP-Q cells has been introduced as a working model to study the effect of TGF-beta1 in maintaining the reversible quiescence of the more primitive hematopoietic stem cell compartment. HPP-Q cells are primitive quiescent stem/progenitor cells on which TGF-beta1 has downmodulated the cytokine receptors. These cells can be released from quiescence by neutralization of autocrine or endogenous TGF-beta1 with a TGF-beta1 blocking antibody or a TGF-beta1 antisense oligonucleotide. In nonhematopoietic systems, TGF-beta1 cooperates with the cyclin-dependent kinase inhibitor, p21(cip1), to induce cell cycle arrest. We therefore analyzed whether endogenous TGF-beta1 controls the expression of the p21(cip1) in the CD34(+) undifferentiated cells using a sensitive in situ hybridization method. We observed that addition of anti-TGF-beta1 is followed by a rapid decrease in the level of p21(cip1) mRNA whereas TGF-beta1 enhances p21(cip1) mRNA expression concurrently with an inhibitory effect on progenitor cell proliferation. These results suggest the involvement of p21(cip1) in the cell cycle control of early human hematopoietic quiescent stem/progenitors and not only in the differentiation of more mature myeloid cells as previously described. The modulation of p21(cip1) observed in response to TGF-beta1 allows us to further precise the working model of high proliferative potential-quiescent cells.


Subject(s)
Cyclins/genetics , Hematopoietic Stem Cells/metabolism , Transcription, Genetic/drug effects , Transforming Growth Factor beta/physiology , Antigens, CD34/analysis , Cell Cycle/physiology , Cell Division/drug effects , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21 , Enzyme Inhibitors/metabolism , Fetal Blood/cytology , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Humans , Infant, Newborn , Kinetics , Oligodeoxyribonucleotides, Antisense/pharmacology , Recombinant Proteins/pharmacology , Transforming Growth Factor beta/pharmacology
8.
Stem Cells ; 18(2): 102-11, 2000.
Article in English | MEDLINE | ID: mdl-10742382

ABSTRACT

Genetic alterations of the signaling cascade of transforming growth factor-beta (TGF-beta) are often associated with neoplastic transformation of primitive cells. This demonstrates the key role for this pleiotropic factor in the control of quiescence and cell proliferation in vivo. In the high proliferative potential-quiescent cell (HPP-Q) in vitro assay, the use of TGF-beta1 blocking antibodies (anti-TGF-beta1) allows the detection within two to three weeks of primitive hematopoietic cells called HPP-Q, which otherwise would not grow. However, the possibility of triggering cell proliferation by blocking the cell-surface TGF-beta receptors has not been investigated until now. We have tested here the efficiency of a blocking antibody against TGF-betaRII (anti-TGF-betaRII) on CD34(+)CD38(-) hematopoietic cells, a subpopulation enriched in primitive stem/progenitor cells, and compared its effect with that of anti-TGF-beta1. About twice as many HPP colony-forming cells were detected in the presence of anti-TGF-beta1 or anti-TGF-betaRII, compared to the control (p < 0.02). Moreover, anti-TGF-betaRII was as efficient as anti-TGF-beta1 for activating multipotent HPP-granulocyte erythroid macrophage megakaryocyte and HPP-Mix, bipotent HPP-granulocyte-macrophage (GM) and unipotent HPP-G, HPP-M and HPP-BFU-E. We therefore propose the use of anti-TGF-betaRII to release primitive cells from quiescence in the HPP-Q assay. This strategy could be extended to nonhematopoietic tissues, as TGF-beta1 may be a pleiotropic regulator of somatic stem cell quiescence.


Subject(s)
Activin Receptors, Type I , Antigens, CD34 , Antigens, CD , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Receptors, Transforming Growth Factor beta/metabolism , ADP-ribosyl Cyclase , ADP-ribosyl Cyclase 1 , Antigens, Differentiation , Cell Differentiation , Cell Division , Cloning, Molecular , Humans , Membrane Glycoproteins , NAD+ Nucleosidase , Protein Serine-Threonine Kinases/genetics , RNA, Messenger , Receptor, Transforming Growth Factor-beta Type I , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/immunology , Time Factors , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/immunology
9.
J Cell Sci ; 113 ( Pt 3): 383-90, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10639326

ABSTRACT

Somatic stem cells are largely quiescent in spite of their considerable proliferative potential. Transforming growth factor-(beta)1 (TGF-(beta)1) appears to be a good candidate for controlling this quiescence. Indeed, various mutations in the TGF-beta signalling pathway are responsible for neoplasic proliferation of primitive stem/progenitor cells in human tissues of various origins. In hemopoietic single cell culture assays, blocking autocrine and endogeneous TGF-(beta)1 triggers the cell cycling of high proliferative potential undifferenciated stem/progenitor cells. However, it has never been demonstrated whether TGF-(beta)1 has an apoptotic effect or a differentiating effect on these primitive cells, as already described for more mature cells. Using single cell experiments both in liquid or semi-solid culture assays and dye tracking experiments by flow cytometry, we demonstrate that low, physiological concentrations of TGF-(beta)1, which specifically maintain primitive human hemopoietic stem/progenitor cells in quiescence, have a reversible effect and do not induce apoptosis. We moreover demonstrate that these low concentrations prevent the rapid loss of the mucin-like protein CD34, a most common marker of immature hematopoietic stem/progenitor cells, which is progressively lost during differentiation. TGF-(beta)1 not only up-modulated the CD34 antigen before S phase entry but also maintained a high level of CD34 expression on cells which had escaped cell cycle inhibition, suggesting that proliferation inhibition and differentiation control by TGF-(beta)1 may be independent. These data provide additional evidence that TGF-(beta)1 acts as a key physiological factor ensuring the maintenance of a stem cell reserve.


Subject(s)
Antigens, CD34/biosynthesis , Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Organic Chemicals , Transforming Growth Factor beta/physiology , Antigens, CD34/genetics , Biomarkers , Cell Cycle/drug effects , Cell Cycle/physiology , Cell Differentiation/drug effects , Cell Division , Cells, Cultured , Coloring Agents/analysis , Dose-Response Relationship, Drug , Fetal Blood/cytology , Flow Cytometry , Fluorescent Dyes/analysis , Hematopoietic Cell Growth Factors/pharmacology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Humans , Interleukin-6/pharmacology , Recombinant Proteins/pharmacology , Transforming Growth Factor beta/pharmacology
10.
Hematol J ; 1(2): 126-35, 2000.
Article in English | MEDLINE | ID: mdl-11920180

ABSTRACT

INTRODUCTION: Transforming Growth Factor-beta1 is known to maintain primitive human hematopoietic stem/progenitor cells in a quiescent state. However, its specific role in the control of distinct progenitor cell types needs to be further elucidated. In this study, we have investigated the dose-response effect of TGF-beta1 on progenitors ranging from primitive high proliferative potential (HPP)-Mix, -GM or -BFU-E to later BFU-E, CFU-G or CFU-M. MATERIALS AND METHODS: A clonal semi-solid assay has been used to analyze the effects of a TGF-beta1 blocking antibody (anti-TGF-beta1) or that of active TGF-beta1 added to the medium at concentrations from 10-3000 pg/ml, on these different hematopoietic stem/progenitor cell types. RESULTS AND CONCLUSION: A preferential growth inhibitory effect on the earlier progenitors was observed when low concentrations of TGF-beta1 (10-300 pg/ml) were used. Concentrations of 10-30 pg/ml TGF-beta1 were sufficient to inhibit 90% of the primitive multipotent HPP-Mix, while 100-300 pg/ml TGF-beta1 were required to inhibit 70% of the bipotent HPP-GM and early HPP-BFU-E. TGF-beta1 did not significantly inhibit or activate the growth of later CFU-G and CFU-M, even when added at concentrations 10-100 fold higher. In contrast, a significant growth-inducing effect of very low TGF-beta1 concentrations (< or =30 pg/ml) on a subset of later BFU-E was observed and cannot be explained by a switch of early into later BFU-E. These results emphasize the polyfunctional role of TGF-beta1 in the regulation of hematopoiesis and the need for low, physiological concentrations of TGF-beta1, when studying both the stem cell compartment and more mature progenitor cell subpopulations.


Subject(s)
Fetal Blood/cytology , Hematopoietic Stem Cells/drug effects , Transforming Growth Factor beta/pharmacology , Cell Culture Techniques , Cell Cycle/drug effects , Colony-Forming Units Assay , Delivery, Obstetric , Female , Growth Substances/pharmacology , Hematopoietic Stem Cells/cytology , Humans , Infant, Newborn , Recombinant Proteins/pharmacology , Transforming Growth Factor beta1
11.
J Cell Sci ; 111 ( Pt 13): 1867-75, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9625749

ABSTRACT

Human adult hematopoietic stem cells are mostly quiescent or slow cycling. We have previously demonstrated that blocking of transforming growth factor-beta1 (TGF-beta1) is able to activate, in the presence of cytokines, primitive quiescent hematopoietic multipotent progenitors which could not grow in a two week semi-solid culture assay (short term culture). We have also shown that anti-TGF-beta1 can up-modulate c-KIT, the receptor of the stem cell factor (steel factor). To elucidate whether TGF-beta1 plays a central role in controlling the quiescence of hematopoietic primitive cells, it was necessary to demonstrate, as detailed in this study, that: (1) whatever the cytokine combination tested, addition of anti-TGF-beta1 releases from quiescence multipotent progenitors with a significantly higher hematopoietic potential than those activated by cytokines alone. (2) Other important cytokine receptors controlling the most primitive hematopoietic cells such as FLT3 and the IL6 receptor (IL6-R) are down-modulated by TGF-beta1 but rapidly up-modulated by anti-TGF-beta1. (3) Anti-TGF-beta1-sensitive multipotent and high proliferative potential progenitors express these cytokine receptors at a low level (FLT3(low) and IL6-Rlow). According to these results, we propose the working model of 'High Proliferative Potential-Quiescent cells' to refer to these primitive hematopoietic multipotent progenitors that are highly sensitive to the growth inhibitory effect of TGF-beta1. This model could be valid not only to study the human hematopoietic quiescent progenitors but also for other somatic stem cell systems.


Subject(s)
Hematopoietic Stem Cells/metabolism , Models, Biological , Antibodies/pharmacology , Antigens, CD34/analysis , Antigens, CD34/biosynthesis , Cell Division/drug effects , Cells, Cultured , Fetal Blood , Fibroblast Growth Factor 2/pharmacology , Hematopoietic Stem Cells/drug effects , Humans , Immunophenotyping , Interphase/drug effects , Membrane Proteins/biosynthesis , Membrane Proteins/pharmacology , Receptors, Interleukin-6/analysis , Receptors, Interleukin-6/biosynthesis , Receptors, Interleukin-6/metabolism , Transforming Growth Factor beta/immunology , Transforming Growth Factor beta/pharmacology
12.
Biochem Biophys Res Commun ; 246(1): 231-7, 1998 May 08.
Article in English | MEDLINE | ID: mdl-9600098

ABSTRACT

In addition to its well-known functions in blood clotting and cell adhesion, fibrinogen has been reported to be a mitogen for lymphoid cell lines and for human hematopoietic progenitors. Two specific receptors, the mitogenic fibrinogen receptor (MFR) and intercellular adhesion molecule-1 (ICAM-1/CD54), have been identified as possible candidates for the mediation of the mitogenic effect of fibrinogen. However, it has been questioned whether the MFR and ICAM-1 are truly distinct molecules. Using an antiserum specific for the MFR, we demonstrate that the MFR is a cell surface molecule clearly distinct from ICAM-1. Both receptors can be expressed separately or coexpressed on different cell types. Moreover, they are regulated differently: ICAM-1 is calcium-dependent whereas the MFR is not and the MFR is down-regulated by fibrinogen whereas ICAM-1 is not. The inhibition by an anti-MFR serum of the mitogenic effect of fibrinogen confirms the mitogenic function of the MFR.


Subject(s)
Fibrinogen/pharmacology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Intercellular Adhesion Molecule-1/metabolism , Mitogens/pharmacology , Receptors, Peptide/metabolism , Antibodies/pharmacology , Cell Division/drug effects , Cell Line , Fibrinogen/metabolism , Hematopoiesis/drug effects , Hematopoietic Stem Cells/cytology , Humans , In Vitro Techniques , Mitogens/metabolism , Receptors, Peptide/antagonists & inhibitors
13.
J Cell Sci ; 109 ( Pt 7): 1795-801, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8832402

ABSTRACT

We have previously shown that a low concentration of CSF-1 (1 U/ml) can trigger human immature monocytic progenitor proliferation in the presence of low concentrations of IL3 (1.7 U/ml). No c-fms down-regulation was observed during this early cell activation. In contrast, 20 U/ml of CSF-1, active on late monocytic cell growth, down-regulated c-fms mRNA expression in immature progenitors and monocytes derived from bone marrow CD34+ cells in culture. We have now extended this study to include the effects of various concentrations of GM-CSF, IL3 and G-CSF on c-fms expression. We observed that high doses of GM-CSF or IL3 down-modulated c-fms mRNA, whereas low doses of GM-CSF or IL3, which were active on early monocytic growth, had no such effect. Similar results were observed at the protein level. In contrast, whatever the concentration, G-CSF had no effect on c-fms mRNA or protein levels. We further observed that the more immature the c-fms expressing progenitors, the faster the down-modulation of this receptor. This was observed within less than 1 hour for immature bone marrow cells, 6 hours for peripheral blood monocytes and even longer for transformed monocytic cells. These results suggest that oncogene expression can be regulated much more rapidly in immature progenitors than was previously observed in mature cells or transformed cell lines.


Subject(s)
Gene Expression Regulation/drug effects , Genes, fms , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Interleukin-3/pharmacology , Macrophage Colony-Stimulating Factor/pharmacology , Monocytes/metabolism , RNA, Messenger/biosynthesis , Cell Differentiation , HL-60 Cells , Hematopoiesis/genetics , Humans , Monocytes/cytology , RNA, Messenger/genetics
14.
Hum Gene Ther ; 7(2): 207-13, 1996 Jan 20.
Article in English | MEDLINE | ID: mdl-8788171

ABSTRACT

It has been previously demonstrated that prestimulation with cytokines could improve gene transfer in hematopoietic progenitors. However, we have shown that no combination of cytokines so far tested is able to release rapidly in vitro the stem cell compartment from quiescence unless an autocrine transforming growth factor-beta 1 (TGF-beta 1) is blocked by specific oligonucleotide antisense or antiserum (Hatzfeld et al., 1991, J. Exp. Med., 174, 925). We now report that a 10-hr cytokine prestimulation of SBA-CD34high human umbilical cord blood progenitors increases retrovirally mediated transfer of the nls-lacZ reporter gene from 1% to 23.8% and addition of anti-TGF-beta serum doubles this increase (47.3%). Interestingly, the effect of anti-TGF-beta preincubation on gene transfer is most effective on the most immature progenitors, which develop into high proliferative potential mixed colonies with 1-2 x 10(5) cells. Anti-TGF-beta serum pretreatment increases gene transfer in these early colony-forming units granulocyte-erythroid-megakaryocyte-macrophage (CFU-GEMM) from 54.1% to 93.3%. It augments significantly the stability of gene expression in all subpopulations of mixed colonies. Colonies obtained after pretreatment with anti-TGF-beta serum are larger and the expression of the stably integrated recombinant provirus does not reduce their size. This prestimulation method provides a substantial improvement for gene transfer efficiency within the quiescent stem cell compartment that is responsible for long-term engraftment.


Subject(s)
Gene Transfer Techniques , Hematopoietic Stem Cells/physiology , Retroviridae/genetics , Antigens, CD34/analysis , Fetal Blood , Gene Expression , Genes, Reporter/genetics , Hematopoietic Cell Growth Factors/pharmacology , Humans , Immune Sera , Interleukin-6/pharmacology , Transforming Growth Factor beta/physiology , beta-Galactosidase/biosynthesis , beta-Galactosidase/genetics
15.
Wien Med Wochenschr ; 146(24): 619-23, 1996.
Article in German | MEDLINE | ID: mdl-9123949

ABSTRACT

At the Ulm University Hospital 43 type-1-diabetes patients took part in a structured in-patient diabetes education program during a 12-day hospitalization period. 27 of the patients received an additional motivational support program which addressed psychological and social impediments related to the topics of the structured diabetes education program. 16 patients underwent the structured diabetes education program only. Motivational support aimed at reducing the specifically addressed sociopsychological barriers. Thus, the effects of the structured diabetes education program should be stabilized and an optimal outcome insured. All patients were asked to complete a questionnaire before, right after and 3 months after the program. Glycosylated hemoglobin (HbAlc) was evaluated before and 3 months after the program. Patients who underwent the motivational support program still felt 3 months after completion of the program that metabolic control was important, whereas patients without motivational support did not. Metabolic control--as indicated by measurement of glycosylated hemoglobin--could be maintained in the patient group with motivational support. Although their blood glucose levels had been in the normal range to start with. These encouraging results suggest implementation of a motivational support program into standard diabetes education programs for type-I-diabetes patients.


Subject(s)
Diabetes Mellitus, Type 1/rehabilitation , Motivation , Patient Education as Topic , Adaptation, Psychological , Adolescent , Adult , Combined Modality Therapy , Diabetes Mellitus, Type 1/psychology , Female , Glycated Hemoglobin/metabolism , Humans , Male , Middle Aged , Patient Admission , Patient Compliance/psychology , Quality of Life , Sick Role , Treatment Outcome
16.
Blood ; 86(5): 1729-35, 1995 Sep 01.
Article in English | MEDLINE | ID: mdl-7544639

ABSTRACT

We have previously shown that early human CD34high hematopoietic progenitors are maintained quiescent in part through autocrine transforming growth factor-beta 1 (TGF-beta 1). We also demonstrated that, in the presence of interleukin-3, interleukin-6, granulocyte colony-stimulating factor, and erythropoietin, TGF-beta 1 antisense oligonucleotides or anti-TGF-beta serum have an additive effect with KIT ligand (Steel factor [SF]), which suggests that they control different pathways of regulation in these conditions. This finding also suggests that autocrine TGF-beta 1 might suppress c-kit expression in primitive human hematopoietic progenitors. We have now distinguished two subpopulations of CD34high cells. One subpopulation expresses a c-kit mRNA that can be downmodulated by exogenous TGF-beta 1 within 6 hours. Another subpopulation of early CD34high cells expresses a low or undetectable level of c-kit mRNA, but its expression can be upmodulated within 6 hours by anti-TGF-beta. These effects disappear 48 hours after induction and cannot be maintained longer than 72 hours, even if TGF-beta 1 or anti-TGF-beta serum are added every day. Similar kinetics, although delayed, are observed with KIT protein expression. On the contrary, no specific effect of TGF-beta 1 was observed on c-fms, GAPDH, and transferrin receptor gene expression in these early progenitors. These results clarify the complex interaction between TGF-beta 1 and SF in normal early hematopoietic progenitors. SF does not switch off the TGF-beta 1 inhibitory pathway. Autocrine TGF-beta 1 appears to maintain these cells in a quiescent state, suppressing cell division by downmodulating the receptor of SF, a key cytokine costimulator of early progenitors.


Subject(s)
Antigens, CD/analysis , Hematopoietic Cell Growth Factors/pharmacology , Hematopoietic Stem Cells/cytology , Proto-Oncogene Proteins/analysis , Receptor Protein-Tyrosine Kinases/analysis , Receptors, Colony-Stimulating Factor/analysis , Transforming Growth Factor beta/pharmacology , Antibodies, Monoclonal/pharmacology , Antigens, CD34 , Biomarkers, Tumor/analysis , Cell Separation/methods , Erythropoietin/pharmacology , Fetal Blood/cytology , Gene Expression , Glyceraldehyde-3-Phosphate Dehydrogenases/biosynthesis , Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Humans , Infant, Newborn , Interleukin-3/pharmacology , Interleukin-6/pharmacology , Kinetics , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins c-kit , Proto-Oncogenes , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptor, Macrophage Colony-Stimulating Factor/biosynthesis , Receptors, Colony-Stimulating Factor/biosynthesis , Receptors, Transferrin/biosynthesis , Recombinant Proteins/pharmacology , Transforming Growth Factor beta/immunology , Transforming Growth Factor beta/physiology
17.
Exp Hematol ; 23(5): 407-12, 1995 May.
Article in English | MEDLINE | ID: mdl-7536682

ABSTRACT

The human hematopoietic pluripotent repopulating "stem cell" is thought to be present within a minor subpopulation of CD34+ cells. This has not been definitively shown, although the more primitive CD34+ cell subset contains precursors for all lymphoid and nonlymphoid cell lineages. When purifying CD34+ cells, it is important to recover these early progenitors, which are more strongly immunoadsorbent to the separation devices. Using a commercialized panning system (AIS CELLector flasks), we observed that a high degree of purity requires a thorough washing procedure so that cells not binding or weakly binding to CD34 antibodies are removed. High recoveries can be obtained if the adherent cells are then efficiently detached by a 2-hour incubation in culture medium without added cytokines. In this way, we can routinely obtain 93.5 +/- 3.4% purity of CD34+ cells with a 74% yield of the multipotent colony-forming units (CFU-GEMM). Complete recovery of the putative "stem cell," or at least the early progenitor cell compartment (CD34+ CD38low/- CD34+ Thy-1+ cells), is also obtained. More than 30% of these cells can generate day-14 colonies in vitro. Comparable results were obtained when the separation was scaled up for clinical application using appropriate large-scale devices. The various incubation times of the procedure can be easily adjusted to the work schedule. This renders the procedure easy to handle, efficient, safe, and, because the cells can be observed under light microscopy, easy to control.


Subject(s)
Antigens, CD , Bone Marrow Cells , Cell Separation/methods , Hematopoietic Stem Cells , Immunosorbent Techniques , ADP-ribosyl Cyclase , ADP-ribosyl Cyclase 1 , ATP Binding Cassette Transporter, Subfamily B, Member 1/analysis , Antigens, CD34 , Antigens, Differentiation/analysis , Colony-Forming Units Assay , Colony-Stimulating Factors/pharmacology , Erythropoietin/pharmacology , HLA-DR Antigens/analysis , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/immunology , Humans , Immunophenotyping , Interleukin-3/pharmacology , Interleukin-6/pharmacology , Membrane Glycoproteins , Proto-Oncogene Proteins/analysis , Proto-Oncogene Proteins c-kit , Receptor Protein-Tyrosine Kinases/analysis , Receptors, Colony-Stimulating Factor/analysis , Recombinant Proteins/pharmacology , Thy-1 Antigens/analysis
18.
Leukemia ; 8(3): 441-5, 1994 Mar.
Article in English | MEDLINE | ID: mdl-7510356

ABSTRACT

We previously demonstrated that TGF-beta 1 antisense oligodeoxynucleotides can release early CD34+ bone marrow progenitors from quiescence, and increase the numbers of mixed and large erythroid colonies. As Steel Factor (SF) has a similar effect on colony formation by CD34+ cells, we tested whether this factor acts by blocking the inhibitory effects of TGF-beta. That this is not generally the case was demonstrated by the finding that the combination of TGF-beta 1 antisense with SF in cultures of CD34+ bone marrow cells yielded enhanced colony formation that was more than additive when compared to cultures containing the single agents. This combination also yielded enhanced colony formation by CD34+ umbilical cord blood cells, but in this case, the effect was slightly less than additive. Thus in cord blood, some, but not all, of the progenitors that are maintained in quiescence by TGF-beta can be triggered into cycle by SF. However, the absolute number of CFU-GEMMs found in the antisense TGF-beta plus SF cultures of cord blood was 4-fold higher than that found in comparable bone marrow cultures. These data correlate well with our previous observations that umbilical cord blood contains 4-fold more CD34+ CD38- cells, the population found to respond to TGF-beta 1 antisense oligodeoxynucleotides.


Subject(s)
Hematopoietic Cell Growth Factors/pharmacology , Hematopoietic Stem Cells/drug effects , Oligonucleotides, Antisense/pharmacology , Transforming Growth Factor beta/pharmacology , Antigens, CD , Antigens, CD34 , Drug Synergism , Fetal Blood/cytology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , Humans , Phenotype , Proto-Oncogene Proteins/analysis , Proto-Oncogene Proteins c-kit , Receptor Protein-Tyrosine Kinases/analysis , Receptors, Colony-Stimulating Factor/analysis , Stem Cell Factor
19.
Blood Cells ; 20(2-3): 430-4; discussion 434-5, 1994.
Article in English | MEDLINE | ID: mdl-7538349

ABSTRACT

Steel factor (SF) increases the frequency of colony formation by CD34+ CD38- cycling cells, but it does not reverse the effect of an autocrine production of transforming growth factor (TGF)-beta 1 by early progenitors of the stem cell compartment. We have used optimal culture conditions supplemented with SF and anti-TGF-beta serum to estimate the proliferative capacity and ability to generate early progenitors in long-term cultures of bone marrow and umbilical cord blood cells. We estimate that the CD34+ CD38- cells from a typical umbilical cord blood sample produce equivalent numbers of granulocyte erythrocyte macrophage megakaryocyte colony-forming units (CFU), twice as many granulocyte-macrophage (GM) CFU, and three times as many erythroid burst-forming units as the same population from an average bone marrow sample used in adult transplantation. These results suggest that umbilical cord blood is a suitable source of cells for adult transplantation.


Subject(s)
Fetal Blood/cytology , Hematopoietic Cell Growth Factors/pharmacology , Hematopoietic Stem Cells , ADP-ribosyl Cyclase , ADP-ribosyl Cyclase 1 , Adult , Antigens, CD/analysis , Antigens, CD34 , Antigens, Differentiation/analysis , Bone Marrow Cells , Cell Count , Cell Division/drug effects , Cells, Cultured , Colony-Forming Units Assay , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Humans , Immune Sera/pharmacology , Membrane Glycoproteins , Stem Cell Factor , Transforming Growth Factor beta/antagonists & inhibitors
20.
Blood ; 82(9): 2649-63, 1993 Nov 01.
Article in English | MEDLINE | ID: mdl-7693043

ABSTRACT

The platelet-endothelial cell adhesion molecule-1 (PE-CAM-1), defined by the CD31 monoclonal antibody (MoAb), was initially described as a cell-cell adhesion molecule mediating both homotypic and heterotypic adhesion. In this report, we show that enriched CD34+ human hematopoietic progenitor cell populations, containing early myeloid, erythroid, and multipotential progenitor cells, are CD31+. Analyses of CD34+ cell lines representing early myeloid, multipotential, and pre-pre-B-lymphoid progenitors indicate that precursors of both myeloid and B-lymphoid cells express PECAM-1 at high levels. Three-color flow-cytometric analyses also show that normal human bone marrow CD31+ CD34+ subsets coexpress myeloid (CD33) or B-lymphoid (CD19, CD10) markers. Except for the monocytic cell line, U937, all CD34- cell lines tested, which represent more mature stages of the myeloid, erythroid, and lymphoid lineages, expressed substantially lower or negligible levels of PECAM-1. Western blotting studies indicated that the CD31 MoAb, JC/70A, detected molecules in the 120- to 140-kD molecular weight range on the monocytic CD34- CD33+ CD31+ cell line, U937; on the CD34+ CD31+ CD33+ CD19- multipotential/lymphomyeloid precursor cell lines, KG1 and KG1B; on the CD34+ CD31+ CD19+ CD10+ CD33- precursor pre-pre-B-cell line, MIK-ALL; and on a CD34(+)-enriched precursor cell population from normal human bone marrow. A single molecular weight species was generally observed with enriched membrane preparations, whereas two PECAM-1 molecules were present in whole-cell lysates of cell lines and the CD34+ bone marrow cell subset. Preliminary studies show that a proportion of the PECAM-1 molecules on the lymphomyeloid/multipotential progenitor cell line, KG1, and on the monocytic cell line, U937, binds to heparin-sepharose. A soluble form of PECAM-1 also binds heparin-sepharose. The high level of expression of PECAM-1 on CD34+ cells suggests that this glycoprotein may function as a heterotypic adhesion molecule, possibly mediating multipotential, myeloid, and early-B-lymphoid precursor cell interactions with stromal cells and extracellular matrix molecules via heparan sulfate proteoglycans. It may also act as a homotypic adhesion molecule by interacting with PECAM-1 on bone marrow stromal macrophage-like cells and endothelial cells or on endothelial cells during stem/progenitor cell migration. Thus, this molecule has the potential importance of directing both lineage commitment and trafficking of early hematopoietic progenitor cells.


Subject(s)
Antigens, CD/analysis , Antigens, Differentiation, Myelomonocytic/analysis , B-Lymphocytes/chemistry , Cell Adhesion Molecules/analysis , Hematopoietic Stem Cells/chemistry , Adult , Animals , Antigens, CD34 , Antigens, Differentiation, Myelomonocytic/physiology , Base Sequence , Bone Marrow/chemistry , Bone Marrow Cells , Cell Line , Heparitin Sulfate/physiology , Humans , Hyaluronan Receptors , Mice , Molecular Sequence Data , Phenotype , Platelet Endothelial Cell Adhesion Molecule-1 , Receptors, Lymphocyte Homing/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...