Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 9 de 9
Filtrer
1.
Br J Haematol ; 102(3): 759-67, 1998 Aug.
Article de Anglais | MEDLINE | ID: mdl-9722304

RÉSUMÉ

To investigate the mechanisms behind the leukaemic expansion of chronic myelogenous leukaemia (CML), we examined the cell cycle status and activation kinetics of purified subpopulations of CD34+ cells from normal and CML bone marrow (BM). Propidium iodide staining was used to assess cell cycle status of fresh cells or those stimulated with cytokines. Although the cell cycle status of fresh low-density cells from CML and normal BM was similar, a larger percentage of CML CD34+ cells were cycling than those from normal BM. The HLA-DR compartment of CML CD34+ cells, a fraction enriched for normal, non-leukaemic progenitors, contained a higher percentage of quiescent cells than the CD34+ HLA-DR+ fraction. When the activation of CD34+ cells was examined in response to SCF or IL-3 alone, or SCF+IL-3+IL-6, CML CD34+ cells exited GO/G1 more rapidly than normal CD34+ cells. Interestingly, although normal BM CD34+ cells failed to cycle in response to IL-6 alone, or in the absence of exogenous cytokines, 30% of CML cells cycled under these conditions. No differences in the degree of apoptosis were documented among CML and normal CD34+ cells in these cultures. These data suggest that enhanced cell cycle activation of CML CD34+ cells, by either autocrine stimuli or via enhanced sensitivity to exogenous stimuli, may be partially responsible for the pronounced cellular expansion characteristic of CML.


Sujet(s)
Antigènes CD34/métabolisme , Cellules de la moelle osseuse/anatomopathologie , Phase G1/physiologie , Leucémie myéloïde en phase chronique/anatomopathologie , Phase G0/physiologie , Adulte , Apoptose/physiologie , Division cellulaire/physiologie , Humains , Interleukine-3/pharmacologie , Interleukine-6/pharmacologie , Adulte d'âge moyen , Facteur de croissance des cellules souches/pharmacologie , Cellules cancéreuses en culture
2.
Exp Hematol ; 26(7): 562-70, 1998 Jul.
Article de Anglais | MEDLINE | ID: mdl-9657130

RÉSUMÉ

Exit of primitive hematopoietic progenitor cells (HPCs) from the G0 phase of the cell cycle in response to in vitro cytokine stimulation is a limiting step in successful ex vivo expansion. Simultaneous DNA/RNA staining with Hoechst 33342 and pyronin Y was used to separate human bone marrow CD34+ cells residing in G0 (G0CD34+) from those cycling in G1 and S/G2+M. Compared with CD34+ cells isolated in G1, G0CD34+ cells were characterized by a delayed response to cytokine stimulation and were enriched for long-term hematopoietic culture-initiating cells. We next compared the activation kinetics of individually sorted G0CD34+ cells stimulated with stem cell factor (SCF), flt3-ligand (FL), or interleukin-3 (IL-3) as single factors. In a novel clonal proliferation assay, the functional status of cells that had remained quiescent after an initial 7-day period and of those that had completed successive division cycles under each of these three factors was evaluated by assessment of subsequent proliferative capacity and maintenance of colony-forming cell precursor (pre-CFC) activity. All three cytokines were equally able to support the survival of primitive HPCs in the absence of cell division. Cells that did not respond to any cytokine stimulation for 7 days retained higher proliferative and pre-CFC activities than dividing cells. The hematopoietic function of cells that divided in response to SCF, FL, or IL-3 decreased after each division cycle. However, G0CD34+ cells displayed a heterogeneous response pattern to cytokine stimulation whereby SCF appeared to have a superior ability to promote the cycling of cells with high proliferative and pre-CFC activities. These results indicate that HPCs reside in opposing hierarchies of hematopoietic potential and responsiveness to cytokine stimulation. The data also begin to indicate relationships between cellular division in response to different stimuli and maintenance of hematopoietic function.


Sujet(s)
Antigènes CD34/analyse , Cellules de la moelle osseuse/immunologie , Phase G0 , Adulte , Division cellulaire/immunologie , Séparation cellulaire , Clones cellulaires , Test clonogénique , Études d'évaluation comme sujet , Humains , Mitose/immunologie , Valeurs de référence
3.
Blood ; 91(10): 3693-701, 1998 May 15.
Article de Anglais | MEDLINE | ID: mdl-9573006

RÉSUMÉ

Primitive hematopoietic progenitor cells (HPCs) are potential targets for treatment of numerous hematopoietic diseases using retroviral-mediated gene transfer (RMGT). To achieve high efficiency of gene transfer into primitive HPCs, a delicate balance between cellular activation and proliferation and maintenance of hematopoietic potential must be established. We have demonstrated that a subpopulation of human bone marrow (BM) CD34(+) cells, highly enriched for primitive HPCs, persists in culture in a mitotically quiescent state due to their cytokine-nonresponsive (CNR) nature, a characteristic that may prevent efficient RMGT of these cells. To evaluate and possibly circumvent this, we designed a two-step transduction protocol using neoR-containing vectors coupled with flow cytometric cell sorting to isolate and examine transduction efficiency in different fractions of cultured CD34(+) cells. BM CD34(+) cells stained on day 0 (d0) with the membrane dye PKH2 were prestimulated for 24 hours with stem cell factor (SCF), interleukin-3 (IL-3), and IL-6, and then transduced on fibronectin with the retroviral vector LNL6 on d1. On d5, half of the cultured cells were transduced with the retroviral vector G1Na and sorted on d6 into cytokine-responsive (d6 CR) cells (detected via their loss of PKH2 fluorescence relative to d0 sample) and d6 CNR cells that had not divided since d0. The other half of the cultured cells were first sorted on d5 into d5 CR and d5 CNR cells and then infected separately with G1Na. Both sets of d5 and d6 CR and CNR cells were cultured in secondary long-term cultures (LTCs) and assayed weekly for transduced progenitor cells. Significantly higher numbers of G418-resistant colonies were produced in cultures initiated with d5 and d6 CNR cells compared with respective CR fractions (P < .05). At week 2, transduction efficiency was comparable between d5 and d6 transduced CR and CNR cells (P > .05). However, at weeks 3 and 4, d5 and d6 CNR fractions generated significantly higher numbers of neoR progenitor cells relative to the respective CR fractions (P < .05), while no difference in transduction efficiency between d5 and d6 CNR cells could be demonstrated. Polymerase chain reaction (PCR) analysis of the origin of transduced neoR gene in clonogenic cells demonstrated that mature progenitors (CR fractions) contained predominantly LNL6 sequences, while more primitive progenitor cells (CNR fractions) were transduced with G1Na. These results demonstrate that prolonged stimulation of primitive HPCs is essential for achieving efficient RMGT into cells capable of sustaining long-term in vitro hematopoiesis. These findings may have significant implications for the development of clinical gene therapy protocols.


Sujet(s)
Ciblage de gène/méthodes , Vecteurs génétiques/génétique , Cellules souches hématopoïétiques/virologie , Kanamycin kinase/biosynthèse , Protéines de fusion recombinantes/biosynthèse , Retroviridae/génétique , Transfection , Adulte , Animaux , Antigènes CD34/analyse , Division cellulaire , Séparation cellulaire , Test clonogénique , Résistance microbienne aux médicaments , Fibronectines , Cytométrie en flux , Expression des gènes , Cellules souches hématopoïétiques/effets des médicaments et des substances chimiques , Cellules souches hématopoïétiques/métabolisme , Humains , Séparation immunomagnétique , Interleukine-3/pharmacologie , Interleukine-6/pharmacologie , Kanamycin kinase/génétique , Réaction de polymérisation en chaîne , Protéines de fusion recombinantes/génétique , Facteur de croissance des cellules souches/pharmacologie
4.
Exp Hematol ; 26(1): 53-62, 1998 Jan.
Article de Anglais | MEDLINE | ID: mdl-9430514

RÉSUMÉ

We examined the decline in hematopoietic potential observed when human CD34+ cells are cultured in vitro by evaluating the association between proliferation history and the fate of long-term hematopoietic culture-initiating cells (LTHC-ICs) as well as the onset of programmed cell death. The membrane dye PKH2 was used to track ex vivo expanded human CD34+ cells from bone marrow, cord blood, and mobilized peripheral blood, and to identify and isolate CD34+ cells that had divided once, twice, three, or four times or more, as well as cells that had remained cytokine nonresponsive and therefore failed to proliferate. These isolated groups of cells were assayed for their hematopoietic potential, cell cycle status, and percentage of apoptotic cells. A gradual decline in the content of LTHC-ICs, as well as in their ability to initiate and sustain in vitro hematopoiesis, was found to correlate with the number of in vitro cellular divisions, such that the hematopoietic potential of CD34+ cells dividing four or more times was nearly depleted. DNA analysis revealed that cells dividing more than three times resided predominantly in G0/G1 phases of the cell cycle. In addition, the percentage of CD34+ cells undergoing apoptosis was found to increase concomitantly with the number of in vitro cellular divisions; less than 10% of cells dividing once were apoptotic, whereas more than 25% of CD34+ cells dividing four or more times underwent programmed cell death. Together, these data suggest that a proliferation-associated, and possibly activation-induced, loss of hematopoietic potential among dividing CD34+ cells may result from an increase in programmed cell death among dividing primitive hematopoietic progenitor cells.


Sujet(s)
Apoptose , Hématopoïèse , Cellules souches hématopoïétiques/anatomopathologie , Antigènes CD34 , Hémogramme , Division cellulaire , Cellules cultivées , Cellules souches hématopoïétiques/immunologie , Humains
5.
Blood ; 90(11): 4384-93, 1997 Dec 01.
Article de Anglais | MEDLINE | ID: mdl-9373249

RÉSUMÉ

Using simultaneous Hoechst 33342 (Hst) and Pyronin Y (PY) staining for determination of DNA and RNA content, respectively, human CD34(+) cells were isolated in subcompartments of the G0 /G1 phase of the cell cycle by flow cytometric cell sorting. In both bone marrow (BM) and mobilized peripheral blood (MPB) CD34(+) cells, primitive long-term hematopoietic culture-initiating cell (LTHC-IC) activity was higher in CD34(+) cells isolated in G0 (G0CD34(+) cells) than in those residing in G1 (G1CD34(+) cells). However, as MPB CD34(+) cells displayed a more homogeneous cell-cycle status within the G0 /G1 phase and a relative absence of cells in late G1 , DNA/RNA fractionation was less effective in segregating LTHC-IC in MPB than in BM. BM CD34(+) cells belonging to four subcompartments of increasing RNA content within the G0 /G1 phase were evaluated in functional assays. The persistence of CD34 expression in suspension culture was inversely correlated with the initial RNA content of test cells. Multipotential progenitors were present in G0 or early G1 subcompartments, while lineage-restricted granulomonocytic progenitors were more abundant in late G1 . In vitro hematopoiesis was maintained for up to 6 weeks with G0CD34(+) cells, whereas production of clonogenic progenitors was more limited in cultures initiated with G1CD34(+) cells. To test the hypothesis that primitive LTHC-ICs would reenter a state of relative quiescence after in vitro division, BM CD34(+) cells proliferating in ex vivo cultures were identified from their quiescent counterparts by a relative loss of membrane intercalating dye PKH2, and were further fractionated with Hst and PY. The same functional hierarchy was documented within the PKH2(dim) population whereby LTHC-IC frequency was higher for CD34(+) cells reselected in G0 after in vitro division than for CD34(+) cells reisolated in G1 or in S/G2 + M. However, the highest LTHC-IC frequency was found in quiescent PKH2(bright) CD34(+) cells. Together, these results support the concept that cells with distinct hematopoietic capabilities follow different pathways during the G0 /G1 phase of the cell cycle both in vivo and during ex vivo culture.


Sujet(s)
Antigènes CD34/analyse , Cycle cellulaire , Cellules souches hématopoïétiques/composition chimique , Séparation cellulaire , Cellules cultivées , Cytométrie en flux , Phase G1 , Cellules souches hématopoïétiques/cytologie , Humains , Phase G0
6.
J Biol Chem ; 272(34): 21334-40, 1997 Aug 22.
Article de Anglais | MEDLINE | ID: mdl-9261146

RÉSUMÉ

Interleukin-9 receptor (IL-9R) complex consists of a ligand-specific alpha chain and IL-2R gamma chain. In this study, two regions in the cytoplasmic domain of human IL-9Ralpha were found to be important for IL-9-mediated cell growth. A membrane-proximal region that contains the BOX1 consensus sequence is required for IL-9-induced cell proliferation and tyrosine phosphorylation of Janus kinases (JAKs). Deletion of this region or internal deletion of the BOX1 motif abrogated IL-9-induced cell proliferation and signal transduction. However, substitution of the Pro-X-Pro in the BOX1 motif with Ala-X-Ala failed to abolish IL-9-induced cell proliferation but decreased IL-9-mediated tyrosine phosphorylation of JAK kinases, insulin receptor substrate-2, and signal transducer and activator of transcription 3 (STAT3) and expression of c-myc and junB. Another important region is downstream of the BOX1 motif and contains a STAT3 binding motif YLPQ. Deletion of this region significantly impaired IL-9-induced cell growth, activation of JAK kinases, insulin receptor substrate-2, and STAT3 and expression of early response genes. A point mutation changing YLPQ into YLPA greatly reduced IL-9-induced activation of STAT3 and expression of c-myc but did not affect cell proliferation. These results suggest that cooperation or cross-talk of signaling molecules associated with different domains of IL-9Ralpha other than STAT3 is essential for IL-9-mediated cell growth.


Sujet(s)
Interleukine-9/physiologie , Récepteurs aux interleukines/composition chimique , Animaux , Division cellulaire , Cellules cultivées , Cytoplasme/composition chimique , Protéines de liaison à l'ADN/métabolisme , Gènes myc , Humains , Substrats du récepteur à l'insuline , Protéines et peptides de signalisation intracellulaire , Janus kinase 1 , Janus kinase 3 , Souris , Phosphoprotéines/métabolisme , Phosphotyrosine/métabolisme , Mutation ponctuelle , Protein-tyrosine kinases/métabolisme , Protéines proto-oncogènes c-jun/génétique , Récepteurs aux interleukines/physiologie , Récepteur à l'interleukine-9 , Protéines recombinantes , Facteur de transcription STAT-3 , Délétion de séquence , Transduction du signal , Relation structure-activité , Lymphocytes T , Transactivateurs/métabolisme , Transfection
7.
Blood ; 90(2): 658-68, 1997 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-9226166

RÉSUMÉ

Bone marrow (BM) CD34+ cells residing in the G0 phase of cell cycle may be the most suited candidates for the examination of cell cycle activation and proliferation of primitive hematopoietic progenitor cells (HPCs). We designed a double simultaneous labeling technique using both DNA and RNA staining with Hoechst 33342 and Pyronin Y, respectively, to isolate CD34+ cells residing in G0(G0CD34+). Using long-term BM cultures and limiting dilution analysis, G0CD34+ cells were found to be enriched for primitive HPCs. In vitro proliferation of G0CD34+ cells in response to sequential cytokine stimulation was examined in a two-step assay. In the first step, cells received a primary stimulation consisting of either stem cell factor (SCF), Flt3-ligand (FL), interleukin-3 (IL-3), or IL-6 for 7 days. In the second step, cells from each group were washed and split into four or more groups, each of which was cultured again for another week with one of the four primary cytokines individually, or in combination. Tracking of progeny cells was accomplished by staining cells with PKH2 on day 0 and with PKH26 on day 7. Overall examination of proliferation patterns over 2 weeks showed that cells could progress into four phases of proliferation. Phase I contained cytokine nonresponsive cells that failed to proliferate. Phase II contained cells dividing up to three times within the first 7 days. Phases III and IV consisted of cells dividing up to five divisions and greater than six divisions, respectively, by the end of the 14-day period. Regardless of the cytokine used for primary stimulation, G0CD34+ cells moved only to phase II by day 7, whereas a substantial percentage of cells incubated with SCF or FL remained in phase I. Cells cultured in SCF or FL for the entire 14-day period did not progress beyond phase III but proliferated into phase IV (with <20% of cells remaining in phases I and II) if IL-3, but not IL-6, was substituted for either cytokine on day 7. G0CD34+ cells incubated with IL-3 for 14 days proliferated the most and progressed into phase IV; however, when SCF was substituted on day 7, cells failed to proliferate into phase IV. Most intriguing was a group of cells, many of which were CD34+, detected in cultures initially stimulated with IL-3, which remained as a distinct population, mostly in G0/G1, unable to progress out of phase II regardless of the nature of the second stimulus received on day 7. A small percentage of these cells expressed cyclin E, suggesting that their proliferation arrest may have been mediated by a cyclin-related disruption in cell cycle. These results suggest that a programmed response to sequential cytokine stimulation may be part of a control mechanism required for maintenance of proliferation of primitive HPCs and that unscheduled stimulation of CD34+ cells residing in G0 may result in disruption of cell-cycle regulation.


Sujet(s)
Cellules de la moelle osseuse , Cytokines/pharmacologie , Cellules souches hématopoïétiques/effets des médicaments et des substances chimiques , Adulte , Antigènes CD34/analyse , Cycle cellulaire/effets des médicaments et des substances chimiques , Division cellulaire/effets des médicaments et des substances chimiques , Cellules cultivées , ADN/analyse , Cellules souches hématopoïétiques/cytologie , Cellules souches hématopoïétiques/physiologie , Humains , Interleukine-3/pharmacologie , Interleukine-6/pharmacologie , ARN/analyse , Phase G0 , Facteur de croissance des cellules souches/pharmacologie , Facteurs temps
8.
Bone Marrow Transplant ; 19(12): 1213-21, 1997 Jun.
Article de Anglais | MEDLINE | ID: mdl-9208115

RÉSUMÉ

It is believed that long-term cultures of CML marrow cells favor the outgrowth of BCR/ABL negative hematopoietic progenitor cells (HPC) and that this phenomenon may be enhanced with negative hematopoietic regulators which can maintain primitive HPC in a quiescent state. Proliferation of CML marrow CD34+ cells in primary short-term cultures, maintained in the presence or absence of macrophage inhibitory protein-1 alpha (MIP-1 alpha), was tracked with the membrane dye PKH2. After 7 to 10 days it was possible to distinguish between cytokine responsive (CR) CD34+ cells (cells which had divided thus becoming PKH2dim) and cytokine nonresponsive (CNR) CD34+ cells (cells which had not divided and had therefore remained PKH2bright). CR and CNR CD34+ cells were isolated by flow cytometric cell sorting, seeded in secondary long-term cultures, and their progeny cells assayed weekly for their clonogenic progenitor cell content and expression of BCR/ABL by reverse transcriptase polymerase chain reaction (RT-PCR). Whereas CNR cells isolated from control primary cultures (control/CNR) sustained in vitro hematopoiesis, similar cells from cultures treated with MIP-1 alpha (MIP-1 alpha/CNR) supported a higher and, in some patients, a more extended production of clonogenic HPC, indicating that MIP-1 alpha was able to maintain primitive HPC in a quiescent state. Predominance of BCR/ABL negative progenitors in vitro was more evident in secondary cultures initiated with CNR cells than in those initiated with CR cells, especially those established with MIP-1 alpha/CNR cells. Of interest is the observed decline in the percentage of BCR/ABL+ progenitors in these cultures with time. Whereas up to 100% of progenitors were BCR/ABL+ on day 0, by day 14, only 46% of progenitors in MIP-1 alpha/CNR secondary cultures were BCR/ABL+ and by day 28 and beyond, the percentage of BCR/ABL+ progenitors dropped to below 20%. These results suggest that the quiescent nature of normal HPC present in CML marrow may favor their identification via cell tracking and, subsequently, their isolation from the more actively cycling leukemic cells. These studies also confirm the feasibility of employing negative hematopoietic regulators to augment the sequestration of normal HPC among the cytokine nonresponsive fraction of CD34+ cells, an approach that may be clinically feasible for autotransplantation.


Sujet(s)
Transplantation de cellules souches hématopoïétiques , Cellules souches hématopoïétiques/immunologie , Cellules souches hématopoïétiques/anatomopathologie , Leucémie myéloïde chronique BCR-ABL positive/anatomopathologie , Leucémie myéloïde chronique BCR-ABL positive/thérapie , Adulte , Antigènes CD34/métabolisme , Séquence nucléotidique , Purge médullaire , Division cellulaire , Cellules cultivées , Chimiokine CCL4 , Cytokines/pharmacologie , Amorces ADN/génétique , Protéines de fusion bcr-abl/génétique , Protéines de fusion bcr-abl/métabolisme , Cellules souches hématopoïétiques/effets des médicaments et des substances chimiques , Humains , Techniques in vitro , Leucémie myéloïde chronique BCR-ABL positive/immunologie , Protéines inflammatoires des macrophages/pharmacologie , Adulte d'âge moyen , Réaction de polymérisation en chaîne , Transplantation autologue
9.
J Hematother ; 5(2): 171-7, 1996 Apr.
Article de Anglais | MEDLINE | ID: mdl-8723796

RÉSUMÉ

Interleukin 12 (IL-12), a heterodimeric cytokine with potent biologic activity, was evaluated for effects on retroviral-mediated gene transduction into human myeloid progenitor cells in vitro. Cord blood CD34 cells were prestimulated with Steel factor (SLF), IL-3, GM-CSF, and erythropoietin (Epo) in the presence and absence of 5-80 ng/ml IL-12 for 40 hr in suspension culture prior to gene transduction using viral supernatant collected from a packaging cell line containing the pLNL6 vector encoding Neo sequences. After gene transduction, cells were assayed for colony formation stimulated by Epo, GM-CSF, IL-3, and SLF, and gene transduction efficiency was determined by the percentage of G418 resistant (R) colonies and confirmed by PCR analysis. IL-12 dose-dependently inhibited retroviral-mediated gene transduction into human cord blood CD34 granulocyte-macrophage (CFU-GM) and erythroid (BFU-E) progenitors. These suppressive effects could be neutralized by incubation of IL-12 with polyclonal antihuman IL-12. IL-12 had no inhibitory effects directly on colony formation. To understand the possible mechanisms for this suppression, ELISA assays were used to detect the release of interferon (IFN)-gamma and tumor necrosis factor (TNF)-alpha, which could potentially have been induced by IL-12 from CD34 cells. TNF-alpha protein release was significantly increased in CD34 cells incubated with IL-12. No detectable levels of IFN-gamma were noted. Anti-TNF-alpha, but not anti-IFN-gamma, blocked the inhibitory effects of IL-12 on gene transduction. Moreover, TNF-alpha, but not IFN-gamma, suppressed gene transfer to the same degree as IL-12. No change of amphotropic receptor mRNA expression was noted by Northern blot analysis in cells treated with or without IL-12. The results suggest that the suppressive effects of IL-12 on retroviral gene transduction are, at least in part, mediated by IL-12 induction of the release of TNF-alpha.


Sujet(s)
Antigènes CD34/sang , Antigènes CD/sang , Sang foetal , Substances de croissance/pharmacologie , Cellules souches hématopoïétiques/cytologie , Interleukine-12/pharmacologie , Retroviridae , Transfection/effets des médicaments et des substances chimiques , Facteur de nécrose tumorale alpha/pharmacologie , Séquence nucléotidique , Amorces ADN , Gènes rapporteurs , Vecteurs génétiques , Cellules souches hématopoïétiques/effets des médicaments et des substances chimiques , Cellules souches hématopoïétiques/immunologie , Humains , Nouveau-né , Kanamycin kinase , Données de séquences moléculaires , Phosphotransferases (Alcohol Group Acceptor)/biosynthèse , Réaction de polymérisation en chaîne , Facteur de nécrose tumorale alpha/antagonistes et inhibiteurs
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...