Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Br J Haematol ; 140(1): 90-8, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17908183

ABSTRACT

Ex vivo expansion of haematopoietic stem and progenitor cells in cytokine combinations is effective in promoting differentiation and proliferation of multilineage progenitor cells, but often results in reduction of self-renewable stem cells. This study investigated the effect of a mannose-binding lectin, NTL, purified from Narcissus tazetta var. chinensis, on prolonged maintenance and expansion of cord blood CD34+ cells. Our results showed that the presence of NTL or Flt-3 ligand (FL) significantly preserved a population of early stem/progenitor cells in a serum- and cytokine-free culture for 35 d. The effect of NTL on the ex vivo expansion of CD34+ cells in the presence of stem cell factor, thrombopoietin (TPO) and FL was also investigated. NTL-enhanced expansion of early progenitors (CD34+, CD34+CD38-, mixed colony-forming units and CFU-GEMM) and committed progenitor cells (granulocyte CFU, erythroid burst-forming units/CFU and megakayocyte CFU) after 8 and 12 d of culture. Six weeks after transplanting 12 d-expanded cells to non-obese diabetic severe combined immunodeficient mice, increased engraftment of human CD45+ cells was observed in the bone marrow of animals that received NTL-treated cells. The dual functions of NTL on long-term preservation and expansion of early stem/multilineage progenitor cells could be developed for applications in various cell therapy strategies, such as the clinical expansion of CD34+ cells for transplantation.


Subject(s)
Blood Preservation/methods , Fetal Blood/drug effects , Hematopoietic Stem Cells/drug effects , Mannose-Binding Lectin/pharmacology , Plant Lectins/pharmacology , Animals , Antigens, CD34 , Cell Culture Techniques , Cell Proliferation , Culture Media, Serum-Free , Fetal Blood/cytology , Graft Survival , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Plant Leaves
2.
Stem Cells ; 25(7): 1800-6, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17446559

ABSTRACT

Serotonin is a monoamine neurotransmitter that has multiple extraneuronal functions. We previously reported that serotonin exerted mitogenic stimulation on megakaryocytopoiesis mediated by 5-hydroxytryptamine (5-HT)2 receptors. In this study, we investigated effects of serotonin on ex vivo expansion of human cord blood CD34+ cells, bone marrow (BM) stromal cell colony-forming unit-fibroblast (CFU-F) formation, and antiapoptosis of megakaryoblastic M-07e cells. Our results showed that serotonin at 200 nM significantly enhanced the expansion of CD34+ cells to early stem/progenitors (CD34+ cells, colony-forming unit-mixed [CFU-GEMM]) and multilineage committed progenitors (burst-forming unit/colony-forming unit-erythroid [BFU/CFU-E], colony-forming unit-granulocyte macrophage, colony-forming unit-megakaryocyte, CD61+ CD41+ cells). Serotonin also increased nonobese diabetic/severe combined immunodeficient repopulating cells in the expansion culture in terms of human CD45+, CD33+, CD14+ cells, BFU/CFU-E, and CFU-GEMM engraftment in BM of animals 6 weeks post-transplantation. Serotonin alone or in addition to fibroblast growth factor, platelet-derived growth factor, or vascular endothelial growth factor stimulated BM CFU-F formation. In M-07e cells, serotonin exerted antiapoptotic effects (annexin V, caspase-3, and propidium iodide staining) and reduced mitochondria membrane potential damage. The addition of ketanserin, a competitive antagonist of 5-HT2 receptor, nullified the antiapoptotic effects of serotonin. Our data suggest the involvement of serotonin in promoting hematopoietic stem cells and the BM microenvironment. Serotonin could be developed for clinical ex vivo expansion of hematopoietic stem cells for transplantation. Disclosure of potential conflicts of interest is found at the end of this article.


Subject(s)
Antigens, CD34/metabolism , Apoptosis/drug effects , Fetal Blood/cytology , Hematopoiesis/drug effects , Serotonin/pharmacology , Stem Cells/drug effects , Stromal Cells/cytology , Animals , Annexin A5/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Caspase 3/metabolism , Cell Proliferation/drug effects , Fetal Blood/drug effects , Fetal Blood/enzymology , Hematopoietic Stem Cell Transplantation , Humans , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, SCID , Stem Cells/cytology , Stem Cells/enzymology , Stromal Cells/drug effects
3.
Stem Cells ; 24(1): 55-64, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16123381

ABSTRACT

The SDF-1/CXCR4 axis has been implicated in the chemotaxis, homing, mobilization, and expansion of hematopoietic stem and progenitor cells. We studied the effects of a SDF-1 peptide analogue CTCE-0214 on the survival of cord blood CD34+ cells in culture, expansion, and engraftment of expanded cells in the nonobese diabetic/severe combined immunodeficient (NOD/SCID) mouse model. Our results demonstrated that CTCE-0214 synergized with thrombopoietin (TPO), stem cell factor (SCF), or flt-3 ligand (FL) on the survival of stem and progenitor cells in culture. Adding CTCE-0214 at a low concentration (0.01 ng/ml) for 4 days together with TPO, SCF, and FL significantly enhanced ex vivo expansion of CD34+ cells to subsets of primitive (CD34+CD38- cells, colony-forming unit-mixed [CFU-GEMMs]), erythroid (CFU-Es), myeloid (CFU-GMs), and megakaryocytic (CD61+CD41+ cells, CFU-MKs) progenitors, as well as their multilineage engraftment in NOD/SCID mice. Interestingly, the short exposure of expanded cells to CTCE-0214 (100 and 500 ng/ml) for 4 hours did not increase the quantity of progenitor cells but enhanced their engraftment capacity. The proportion of CD34+ cells expressing surface CXCR4 was decreased, but the overall number of this population increased upon expansion. The small peptide analogue of SDF-1 could be developed for ex vivo expansion and improving engraftment of cord blood transplantation.


Subject(s)
Cell Survival/drug effects , Chemokines, CXC/physiology , Cytokines/pharmacology , Membrane Proteins/pharmacology , Stem Cell Factor/pharmacology , Thrombopoietin/pharmacology , Animals , Antigens, CD34/metabolism , Chemokine CXCL12 , Dose-Response Relationship, Drug , Drug Synergism , Fetal Blood/cytology , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells , Mice , Mice, SCID , Receptors, CXCR4/metabolism
4.
Int J Oncol ; 27(2): 537-45, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16010437

ABSTRACT

Arsenic trioxide (ATO) induces apoptosis in a range of solid tumors and leukemia cells, and has been clinically applied for the treatment of acute promyelocytic leukemia with confirmed efficacy. Acute megakaryocytic leukemia (AMKL) is an aggressive malignancy with poor prognosis, if bone marrow transplantation is not possible. In this study, we applied flow cytometry, Western blot analysis and microarray techniques to investigate the effects of ATO on apoptosis and the cell division cycle of AMKL cell lines CHRF-288-11 and MEG-01. Our data demonstrated that ATO is a potent agent against AMKL as indicated by apoptotic markers, Annexin V and caspase-3. ATO activated the intrinsic (mitochondrial) pathway of apoptosis, which involved disrupting mitochondrial membrane potential, increased Bax/Bcl-2 ratio and caspase-9 activation, as well as the extrinsic (death receptor) pathway mediated by Fas and caspase-8 activation. We provided the first evidence that ATO stimulated expressions of CD137 mRNA and protein, which might be relevant to the extrinsic mechanism. ATO induced delays of cell cycle progression at S phase and arrest at G2/M phase of AMKL cells, but caspase-3 expression appeared not to be phase-specific. The multiple-signaling mechanism of ATO warrants it a potential agent to incorporate in the treatment regimen of AMKL.


Subject(s)
Apoptosis/drug effects , Arsenicals/pharmacology , Cell Cycle/drug effects , Oxides/pharmacology , Antigens, CD/genetics , Antigens, CD/metabolism , Apoptosis/genetics , Arsenic Trioxide , Caspase 8 , Caspase 9 , Caspases/genetics , Caspases/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Flow Cytometry , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Humans , Intracellular Membranes/drug effects , Intracellular Membranes/physiology , Leukemia, Megakaryoblastic, Acute/genetics , Leukemia, Megakaryoblastic, Acute/metabolism , Leukemia, Megakaryoblastic, Acute/pathology , Membrane Potentials/drug effects , Mitochondria/drug effects , Mitochondria/physiology , Oligonucleotide Array Sequence Analysis/methods , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Receptors, Nerve Growth Factor/genetics , Receptors, Nerve Growth Factor/metabolism , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/metabolism , Signal Transduction/drug effects , Time Factors , Tumor Necrosis Factor Receptor Superfamily, Member 9 , fas Receptor/genetics , fas Receptor/metabolism
5.
Eur J Haematol ; 74(2): 128-35, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15654904

ABSTRACT

OBJECTIVES: Ex vivo expansion of granulocyte-colony stimulating factor (G-CSF)-mobilized peripheral blood stem cells (PBSC) is a promising approach for overcoming the developmental delay of bone marrow (BM) reconstitution after transplantation. This project investigated the effects of culture duration, serum-free media, cytokine combinations, and chemotherapy on the outcomes of expansion. METHODS: Enriched CD34+ cells were cultured for 8 or 10 d in serum-free media (QBSF-60 or X-Vivo 10) and four combinations of cytokines consisting of recombinant human pegylated-megakaryocyte growth and development factor, stem cell factor, flt-3 ligand, G-CSF, interleukin (IL)-6, platelet-derived growth factor (PDGF), and IL-1beta. RESULTS: Eight days of culture in QBSF-60 significantly supported efficient expansions of CD34+ cells, CD34+ CD38- cells, colony-forming units (CFU) of myeloid, erythroid, megakaryocytic, and mixed lineages to 3.76-, 14.4-, 28.3-, 24.0-, 38.1-, and 15.7-fold, respectively. Whilst PDGF or IL-6 enhanced the expansion of early, myeloid, and erythroid progenitors, IL-1beta specifically promoted the megakaryocytic lineage. Engraftment of human CD45+ cells were detectable in all non-obese diabetic/severe-combined immunodeficient mice transplanted with expanded PBSC from donor samples, being 5.80 +/- 3.34% of mouse BM cells. The expansion and engraftment capacity of CD34+ cells from subjects postchemotherapy were significantly compromised across the panel of progenitor cells. CONCLUSION: Our results provided an optimized protocol for PBSC expansion, applicable to ameliorating neutropenia and thrombocytopenia in post-BM transplant patients by the prompt provision of progenitor cells. For postchemotherapy patients, expansion products might provide committed progenitors for improving short-term engraftment, but not self-renewable stem cells.


Subject(s)
Cell Proliferation/drug effects , Culture Media, Serum-Free , Cytokines/pharmacology , Granulocyte Colony-Stimulating Factor/pharmacology , Myeloid Progenitor Cells , ADP-ribosyl Cyclase/metabolism , ADP-ribosyl Cyclase 1 , Adolescent , Adult , Antigens, CD/metabolism , Antigens, CD34/metabolism , Bone Marrow/pathology , Bone Marrow/physiology , Bone Marrow Transplantation , Cells, Cultured , Child , Humans , Membrane Glycoproteins , Middle Aged , Myeloid Progenitor Cells/metabolism , Neoplasms/pathology , Neoplasms/therapy , Stem Cells/drug effects
6.
J Lab Clin Med ; 143(2): 75-88, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14966463

ABSTRACT

The multifunctional cytokine interleukin-1beta (IL-1beta) plays a central role in the body's immune and inflammatory responses. The mechanism of IL-1beta on thrombocytosis and megakaryocytopoiesis has remained controversial. In previous reports, we have demonstrated the expression of IL-1 receptors (IL-1RI and IL-1RII) and enhancing effects of IL-1beta on primary human megakaryocytic (MK) cells. In this study, we investigated the possible direct effects of IL-1beta on the expression of thrombopoietin (TPO) and transcription factors c-Jun, c-Fos, GATA-1, and p45 nuclear factor-E2 (NF-E2) in MK cell lines CHRF and Meg-01. Our results demonstrated that IL-1beta up-regulated messenger RNA (mRNA) and protein expressions of these transcription factors in a dose- and time-dependent manner. In CHRF cells, mRNA: c-Jun [3.4-fold, peaked at 15 minutes], c-Fos [4.2-fold, 15 minutes], GATA-1 [4.0-fold, 60 minutes], NF-E2 [3.2-fold, 120 minutes] and protein expression: c-Jun [3.0-fold, 30 minutes], c-Fos [1.7-fold, 30 minutes], GATA-1 [11.5-fold, 60 minutes], NF-E2 [12.5-fold, 120 minutes] were evidently enhanced after treatment with IL-1beta. The response to IL-1beta was consistent in the total cell and nuclear extracts and was significantly reduced by pretreatment with actinomycin D or cycloheximide. An IL-1-receptor antagonist (IL-1RA) inhibited the stimulatory effects of IL-1beta on these transcription factors by as much as 78%. TPO expression was increased by more than 9.9-fold on stimulation with IL-1beta. A TPO-neutralizing antibody did not significantly reduce the effects of IL-1beta. We conclude that IL-1beta up-regulates the expression of TPO, c-Jun, c-Fos, GATA-1, and NF-E2 in MK cells. The mechanism might be mediated by IL-1beta receptors and require transcription or protein synthesis. The direct involvement of IL-1beta in the MK lineage may provide an explanation for the phenomenon of thrombocytosis during inflammatory responses.


Subject(s)
DNA-Binding Proteins/genetics , Gene Expression Regulation/drug effects , Interleukin-1/pharmacology , Megakaryocytes/physiology , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-jun/genetics , Thrombopoietin/genetics , Transcription Factors/genetics , Base Sequence , Blotting, Southern , Cell Line , Cycloheximide/pharmacology , DNA Primers , Dactinomycin/pharmacology , Erythroid-Specific DNA-Binding Factors , GATA1 Transcription Factor , Humans , Kinetics , Megakaryocytes/cytology , NF-E2 Transcription Factor , NF-E2 Transcription Factor, p45 Subunit
7.
Cytokine ; 21(2): 51-64, 2003 Jan 21.
Article in English | MEDLINE | ID: mdl-12670444

ABSTRACT

Platelet-derived growth factor (PDGF) is a platelet alpha-granule protein. In previous reports, we demonstrated the expression of PDGF receptors on platelets and megakaryocytic cells and that PDGF enhanced the proliferation of megakaryocytic progenitor cells. In this study, we investigated the effects of PDGF on mRNA and protein expressions of megakaryocyte-associated transcription factors, c-Fos, GATA-1, NF-E2 and PU.1, in two human megakaryocytic cell lines CHRF-288-11 and DAMI. RT-PCR/Southern blot analysis and Real-time PCR demonstrated that PDGF increased the mRNA expression of c-Fos, GATA-1 and NF-E2, but not PU.1 in a dose- and time-dependent manner. The activation was confirmed at the protein level by Western blot analysis of both total cell and nuclear lysates. The addition of increasing concentrations of Tyrphostin AG1295, an inhibitor of PDGF receptor kinase, blocked the stimulatory effect of PDGF on the mRNA and protein expressions of these transcription factors. The up-regulation of c-Fos, GATA-1 and NF-E2 protein by PDGF was inhibited by actinomycin D and cycloheximide, suggesting that mRNA and protein synthesis might be involved in the mechanism. Our data suggest a direct stimulatory effect of PDGF on c-Fos, GATA-1 and NF-E2 expressions and we speculate that these transcription factors might be involved in the signal transduction of PDGF on the regulation of megakaryocytopoiesis.


Subject(s)
DNA-Binding Proteins/biosynthesis , Megakaryocytes/metabolism , Platelet-Derived Growth Factor/physiology , Proto-Oncogene Proteins c-fos/biosynthesis , Transcription Factors/biosynthesis , Up-Regulation , Blotting, Southern , Blotting, Western , Cell Division , Cell Line , Cell Nucleus/metabolism , Cycloheximide/pharmacology , Dactinomycin/pharmacology , Dose-Response Relationship, Drug , Erythroid-Specific DNA-Binding Factors , GATA1 Transcription Factor , Humans , NF-E2 Transcription Factor , NF-E2 Transcription Factor, p45 Subunit , Platelet-Derived Growth Factor/metabolism , Protein Synthesis Inhibitors/pharmacology , Proto-Oncogene Proteins/biosynthesis , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Time Factors , Trans-Activators/biosynthesis , Tyrphostins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL