Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67
Filter
1.
Leukemia ; 24(10): 1667-75, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20703253

ABSTRACT

Hematopoietic stem and progenitor cells (HSPCs), as well as other types of stem cells, circulate under steady-state conditions at detectable levels in peripheral blood (PB), with their numbers increasing in response to stress, inflammation and tissue/organ injury. This mobilization process may be envisioned as a danger-sensing response mechanism triggered by hypoxia or mechanical or infection-induced tissue damage that recruits into PB different types of stem cells that have a role in immune surveillance and organ/tissue regeneration. Mobilization is also significantly enhanced by the administration of pharmacological agents, which has been exploited in hematological transplantology as a means to obtain HSPCs for hematopoietic reconstitution. In this review we will present mounting evidence that innate immunity orchestrates this evolutionarily conserved mechanism of HSPC mobilization.


Subject(s)
Hematologic Diseases/immunology , Hematopoietic Stem Cell Mobilization , Immunity, Innate , Stem Cell Transplantation , Stem Cells/immunology , Humans , Stem Cells/cytology
2.
Leukemia ; 24(5): 976-85, 2010 May.
Article in English | MEDLINE | ID: mdl-20357827

ABSTRACT

The complement cascade (CC) becomes activated and its cleavage fragments play a crucial role in the mobilization of hematopoietic stem/progenitor cells (HSPCs). Here, we sought to determine which major chemoattractant present in peripheral blood (PB) is responsible for the egress of HSPCs from the bone marrow (BM). We noticed that normal and mobilized plasma strongly chemoattracts HSPCs in a stromal-derived factor-1 (SDF-1)-independent manner because (i) plasma SDF-1 level does not correlate with mobilization efficiency; (ii) the chemotactic plasma gradient is not affected in the presence of AMD3100 and (iii) it is resistant to denaturation by heat. Surprisingly, the observed loss of plasma chemotactic activity after charcoal stripping suggested the involvement of bioactive lipids and we focused on sphingosine-1-phosphate (S1P), a known chemoattracant of HSPCs. We found that S1P (i) creates in plasma a continuously present gradient for BM-residing HSPCs; (ii) is at physiologically relevant concentrations a chemoattractant several magnitudes stronger than SDF-1 and (iii) its plasma level increases during mobilization due to CC activation and interaction of the membrane attack complex (MAC) with erythrocytes that are a major reservoir of S1P. We conclude and propose a new paradigm that S1P is a crucial chemoattractant for BM-residing HSPCs and that CC through MAC induces the release of S1P from erythrocytes for optimal egress/mobilization of HSPCs.


Subject(s)
Bone Marrow/metabolism , Cell Movement , Complement C5/physiology , Complement Membrane Attack Complex/metabolism , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cells/physiology , Lysophospholipids/metabolism , Sphingosine/analogs & derivatives , Animals , Anti-HIV Agents/pharmacology , Benzylamines , Chemokine CXCL12/blood , Colony-Forming Units Assay , Complement Activation , Cyclams , Enzyme-Linked Immunosorbent Assay , Erythrocytes/cytology , Erythrocytes/metabolism , Heterocyclic Compounds/pharmacology , Hot Temperature , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, CXCR4/antagonists & inhibitors , Sphingosine/metabolism
3.
Cytotherapy ; 10(2): 182-92, 2008.
Article in English | MEDLINE | ID: mdl-18368597

ABSTRACT

BACKGROUND: In patients transplanted with cord blood (CB), prolonged thrombocytopenia is a major complication. However, this could be alleviated by supplementing the CB graft with ex vivo-expanded megakaryocytic progenitors (CFU-Meg), provided that the homing properties of these cells are not affected negatively by expansion. METHODS AND RESULTS: We assessed the in vitro homing potential of CFU-Meg progenitors expanded from CB and showed that the combination of thrombopoietin (TPO) with interleukin-3 (IL-3) used for expansion not only results in optimal proliferation of CFU-Meg but also protects these cells from apoptosis. Moreover, we found that ex vivo-expanded CFU-Meg maintained expression of the CXCR4 receptor throughout a 9-day culture and were chemoattracted towards a stromal cell-derived factor-1 (SDF-1) gradient. They also expressed matrix metalloproteinase-9 (MMP-9) and membrane-type (MT) 1-MMP, and transmigrated across the reconstituted basement membrane Matrigel. Finally, we observed that SDF-1 up-regulated the expression of both MMP-9 and MT1-MMP in CB CD34(+) cells and ex vivo-expanded CFU-Meg. DISCUSSION: We suggest that CB-expanded CFU-Meg, in particular those from day 3 of expansion, when their proliferation and in vitro homing potential are maximal, could be employed to supplement CB grafts and speed up platelet recovery in transplant recipients.


Subject(s)
Colony-Forming Units Assay , Fetal Blood/cytology , Fetal Blood/enzymology , Matrix Metalloproteinase 14/metabolism , Matrix Metalloproteinase 9/metabolism , Megakaryocytes/cytology , Stem Cells/cytology , Antigens, CD34/metabolism , Catechin/analogs & derivatives , Catechin/pharmacology , Cell Proliferation/drug effects , Cell Survival/drug effects , Chemokine CXCL12/metabolism , Chemotactic Factors/pharmacology , Chemotaxis/drug effects , Collagen/metabolism , Drug Combinations , Fetal Blood/drug effects , Humans , Interleukin-3/pharmacology , Kinetics , Laminin/metabolism , Matrix Metalloproteinase 14/genetics , Matrix Metalloproteinase 9/genetics , Megakaryocytes/drug effects , Megakaryocytes/enzymology , Platelet Membrane Glycoprotein IIb/metabolism , Proteoglycans/metabolism , Receptors, CXCR4/metabolism , Stem Cells/drug effects , Thrombopoietin/pharmacology , Up-Regulation/drug effects
4.
Leukemia ; 20(9): 1487-95, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16791265

ABSTRACT

Normal and malignant cells shed from their surface membranes as well as secrete from the endosomal membrane compartment circular membrane fragments called microvesicles (MV). MV that are released from viable cells are usually smaller in size compared to the apoptotic bodies derived from damaged cells and unlike them do not contain fragmented DNA. Growing experimental evidence indicates that MV are an underappreciated component of the cell environment and play an important pleiotropic role in many biological processes. Generally, MV are enriched in various bioactive molecules and may (i) directly stimulate cells as a kind of 'signaling complex', (ii) transfer membrane receptors, proteins, mRNA and organelles (e.g., mitochondria) between cells and finally (iii) deliver infectious agents into cells (e.g., human immuno deficiency virus, prions). In this review, we discuss the pleiotropic effects of MV that are important for communication between cells, as well as the role of MV in carcinogenesis, coagulation, immune responses and modulation of susceptibility/infectability of cells to retroviruses or prions.


Subject(s)
Cell Communication , Cell Membrane/physiology , Organelles/physiology , Disease Progression , Humans , Neoplasms/pathology
5.
Leukemia ; 18(1): 29-40, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14586476

ABSTRACT

It has been suggested that bone marrow (BM)-derived hematopoietic stem cells transdifferentiate into tissue-specific stem cells (the so-called phenomenon of stem cell plasticity), but the possibility of committed tissue-specific stem cells pre-existing in BM has not been given sufficient consideration. We hypothesized that (i) tissue-committed stem cells circulate at a low level in the peripheral blood (PB) under normal steady-state conditions, maintaining a pool of stem cells in peripheral tissues, and their levels increase in PB during stress/tissue injury, and (ii) they could be chemoattracted to the BM where they find a supportive environment and that the SDF-1-CXCR4 axis plays a prominent role in the homing/retention of these cells to BM niches. We performed all experiments using freshly isolated cells to exclude the potential for 'transdifferentiation' of hematopoietic stem or mesenchymal cells associated with in vitro culture systems. We detected mRNA for various early markers for muscle (Myf-5, Myo-D), neural (GFAP, nestin) and liver (CK19, fetoprotein) cells in circulating (adherent cell-depleted) PB mononuclear cells (MNC) and increased levels of expression of these markers in PB after mobilization by G-CSF (as measured using real-time RT-PCR). Furthermore, SDF-1 chemotaxis combined with real-time RT-PCR analysis revealed that (i) these early tissue-specific cells reside in normal murine BM, (ii) express CXCR4 on their surface and (iii) can be enriched (up to 60 x) after chemotaxis to an SDF-1 gradient. These cells were also highly enriched within purified populations of murine Sca-1(+) BM MNC as well as of human CD34(+)-, AC133(+)- and CXCR4-positive cells. We also found that the expression of mRNA for SDF-1 is upregulated in damaged heart, kidney and liver. Hence our data provide a new perspective on BM not only as a home for hematopoietic stem cells but also a 'hideout' for already differentiated CXCR4-positive tissue-committed stem/progenitor cells that follow an SDF-1 gradient, could be mobilized into PB, and subsequently take part in organ/tissue regeneration.


Subject(s)
Bone Marrow/metabolism , DNA-Binding Proteins , Hematopoietic Stem Cells/metabolism , Liver/metabolism , Muscle, Skeletal/metabolism , Nerve Tissue Proteins , Neurons/metabolism , RNA, Messenger/metabolism , Receptors, CXCR4/metabolism , Trans-Activators , Animals , Antigens, CD34/metabolism , Biomarkers/analysis , Biomarkers/blood , Blood Cells/cytology , Blood Cells/metabolism , Cell Line , Chemokine CXCL12 , Chemokines, CXC/genetics , Chemokines, CXC/metabolism , Female , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cells/drug effects , Humans , Intermediate Filament Proteins/genetics , Intermediate Filament Proteins/metabolism , Keratins/genetics , Keratins/metabolism , Mice , Mice, Inbred BALB C , Muscle Proteins/genetics , Muscle Proteins/metabolism , MyoD Protein/genetics , MyoD Protein/metabolism , Myogenic Regulatory Factor 5 , Nestin , RNA, Messenger/genetics , alpha-Fetoproteins/genetics , alpha-Fetoproteins/metabolism
6.
Leukemia ; 16(6): 1160-6, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12040448

ABSTRACT

To further elucidate the role of angiogenesis in the pathogenesis of chronic myelogenous leukemia (CML) we evaluated the effects of the bcr-abl translocation on the secretion of the angiogenic factors VEGF, FGF-2, HGF, IL-8 and matrix metalloproteinases (MMPs) as well as on the angiogenic potential in vivo of bcr-abl+ cells. First, we examined murine FL5.12 cells transfected with the bcr-abl constructs p185, p210 and p230 and found that the transfected cells secreted as much as four-fold more VEGF (p185 > p210 >p230) than wild-type (wt) cells, as well as MMP-9 and MMP-2. When Matrigel fragments containing these bcr-abl+ cells were implanted subcutaneously in SCID or Balb-C mice they became significantly more vascularized and hemoglobinized than implants containing normal or wt cells (p185 > p210 > p230). Similarly, we found that myeloblasts expanded from bone marrow (BM) CD34+ cells derived from Philadelphia-positive CML patients secreted up to 10 times more VEGF, FGF-2, HGF and IL-8 compared to myeloblasts derived from normal donors' BM CD34+ cells and that BM mononuclear cells (MNC) isolated from CML patients induced vascularization of Matrigel implants in mice. Moreover, we found that peripheral blood MNC expressed MMP-2 and membrane-type (MT)1-MMP in about 50% of CML patients studied, and MMP-9 in all of them. Furthermore, VEGF stimulated the secretion of MMP-9 in these primary CML cells. We conclude that stimulation of angiogenesis by angiogenic factors, including MMPs, could play an important role in the pathogenesis of CML, suggesting that therapies targeting the newly formed endothelium could be developed for CML.


Subject(s)
Angiogenesis Inducing Agents/biosynthesis , Fusion Proteins, bcr-abl/analysis , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Matrix Metalloproteinases/biosynthesis , Neovascularization, Pathologic , Adult , Aged , Angiogenesis Inducing Agents/metabolism , Animals , Cell Line, Transformed , Cells, Cultured , Collagen/administration & dosage , Drug Combinations , Endothelial Growth Factors/metabolism , Female , Fusion Proteins, bcr-abl/genetics , Humans , Laminin/administration & dosage , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Lymphokines/metabolism , Male , Matrix Metalloproteinases/genetics , Matrix Metalloproteinases/metabolism , Mice , Mice, Inbred BALB C , Mice, SCID , Middle Aged , Proteoglycans/administration & dosage , RNA, Neoplasm/biosynthesis , Tissue Inhibitor of Metalloproteinases/biosynthesis , Tissue Inhibitor of Metalloproteinases/genetics , Tissue Inhibitor of Metalloproteinases/metabolism , Transfection , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
7.
Br J Haematol ; 115(3): 595-604, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11736941

ABSTRACT

Matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinases (TIMPs) regulate the turnover of the extracellular matrix and may modulate the biology of haematopoietic cells. We investigated whether MMPs and TIMPs are produced in long-term marrow cultures (LTMCs) established from normal donors and acute myelogenous leukaemia (AML) patients, and by fibroblast- (F), granulocyte macrophage- (GM) and megakaryocyte- (Meg) colony-forming unit (CFU) and erythroid burst-forming unit (BFU-E)-derived precursor cells. ProMMP-9 levels were highest (> 400 ng/ml) at week 1 of normal LTMC, whereas proMMP-2, TIMP-1, TIMP-2 and TIMP-3 levels peaked (up to 1000 ng/ml) after the establishment of the adherent layer. In LTMC from AML patients, these patterns of secretion were reversed. Moreover, we found that after a 24 h incubation in serum-free media, normal CFU-GM-, BFU-E- and CFU-Meg-derived cells secreted proMMP-9 and CFU-F-derived cells proMMP-2, in contrast to cells from LTMC adherent layer which secreted both active and latent forms of MMP-2 and MMP-9 under serum-free conditions. However, when these adherent cells were incubated in 12.5% fetal calf or horse serum or complete LTMC growth media, active forms of MMP-2 and MMP-9 were no longer detectable, and TIMP levels increased. Hence, we concluded that (i) MMPs/TIMPs are secreted by normal human bone marrow haematopoietic and stromal cells and may play an important role in intercellular cross-talk in haematopoiesis; and (ii) only latent forms of MMPs are present under LTMC conditions, indicating that the specific media used for weekly re-feeding of LTMC can block activation of MMP-2 and MMP-9, maintaining the integrity of the stromal layer and supporting haematopoiesis in vitro.


Subject(s)
Hematopoietic Stem Cells/enzymology , Leukemia, Myeloid, Acute/enzymology , Matrix Metalloproteinases/metabolism , Tissue Inhibitor of Metalloproteinases/metabolism , Bone Marrow Cells/enzymology , Cell Culture Techniques , Cell Division , Cells, Cultured , Electrophoresis, Polyacrylamide Gel/methods , Humans , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tissue Inhibitor of Metalloproteinase-2/metabolism , Tissue Inhibitor of Metalloproteinase-3/metabolism , Tissue Inhibitor of Metalloproteinases/pharmacology
8.
Blood ; 98(10): 3143-9, 2001 Nov 15.
Article in English | MEDLINE | ID: mdl-11698303

ABSTRACT

Because human CD34+ and murine Sca-1+ hematopoietic stem-progenitor cells (HSPCs) express platelet-binding sialomucin P-selectin (CD162) and integrin Mac-1 (CD11b-CD18) antigen, it was inferred that these cells might interact with platelets. As a result of this interaction, microparticles derived from platelets (PMPs) may transfer many platelet antigens (CD41, CD61, CD62, CXCR4, PAR-1) to the surfaces of HSPCs. To determine the biologic significance of the presence of PMPs on human CD34+ and murine Sca-1+ cells, their expressions on mobilized peripheral blood (mPB) and on nonmobilized PB- and bone marrow (BM)-derived CD34+ cells were compared. In addition, the effects of PMPs on the proliferation of CD34+ and Sca-1+ cells and on adhesion of HSPCs to endothelium and immobilized SDF-1 were studied. Finally, the hematopoietic reconstitution of lethally irradiated mice receiving transplanted BM mononuclear cells covered or not covered with PMPs was examined. It was found that PMPs are more numerous on mPB than on BM CD34+ cells, do not affect the clonogenicity of human and murine HSPCs, and increase adhesion of these cells to endothelium and immobilized SDF-1. Moreover, murine BM cells covered with PMPs engrafted lethally irradiated mice significantly faster than those not covered, indicating that PMPs play an important role in the homing of HSPCs. This could explain why in a clinical setting human mPB HSPCs (densely covered with PMPs) engraft more rapidly than BM HSPCs (covered with fewer PMPs). These findings indicate a new role for PMPs in stem cell transplantation and may have clinical implications for the optimization of transplantations.


Subject(s)
Blood Platelets/metabolism , Graft Survival , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/metabolism , Animals , Antigens, CD34/analysis , Antigens, Human Platelet/metabolism , Antigens, Ly/analysis , Blood Platelets/ultrastructure , Bone Marrow Cells/metabolism , Cell Adhesion , Cell Division , Cell Membrane/metabolism , Cells, Cultured , Chemokine CXCL12 , Chemokines, CXC/metabolism , Colony-Forming Units Assay , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Female , HL-60 Cells , Hematopoietic Stem Cell Mobilization , Humans , Membrane Glycoproteins/analysis , Membrane Proteins/analysis , Mice , Mice, Inbred C57BL , Platelet Activation , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Radiation Chimera , Time Factors , Umbilical Veins
9.
Br J Haematol ; 115(1): 195-204, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11722433

ABSTRACT

The aim of this study was to identify signal transduction pathways activated by erythropoietin (EpO) and erythropoietin co-stimulatory factors (kit ligand), insulin-like growth factor, thrombopoietin, interleukin 3 and granulocyte-macrophage colony-stimulating factor) in normal human bone marrow CD34(+) cells and d 11 erythroid burst forming unit derived glycophorin+ cells. The activation of these signal transduction pathways was further correlated with various biological effects such as (i) cell proliferation, (ii) inhibition of apoptosis, (iii) activation of adhesion and (iv) secretion of the matrix metalloproteinases (MMPs) MMP-9 and MMP-2, and vascular endothelial growth factor (VEGF). We found that in human CD34(+) cells and erythroblasts erythropoietic factors may activate similar but different signalling pathways, and that activation of each of the JAK-STAT, MAPK p42/44 or PI-3K-AKT axes alone is not sufficient either to stimulate cell proliferation or inhibit apoptosis, suggesting that these processes are regulated by orchestrated activation of multiple signalling cascades. Accordingly, we found that although cell proliferation was more related to simultaneous activation of JAK-STAT and MAPK p42/44, the effect on cell survival correlated with activation of PI-3K-AKT, MAPK p42/44 and JAK-STAT proteins. We also demonstrated that differentiating normal human erythroid cells lose their adhesive properties and secrete angiopoietic factors such as MMP-9, MMP-2 and VEGF, and we postulate that this secretion by early erythroid cells may play a role in their maturation and egress from the haematopoietic niches of the bone marrow.


Subject(s)
DNA-Binding Proteins/metabolism , Erythroid Precursor Cells/metabolism , MAP Kinase Signaling System , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/metabolism , Signal Transduction/physiology , Trans-Activators/metabolism , Antigens, CD34 , Apoptosis/drug effects , Cell Adhesion/drug effects , Cell Division/drug effects , Cells, Cultured , Endothelial Growth Factors/metabolism , Enzyme Activation , Erythroid Precursor Cells/immunology , Erythropoietin/pharmacology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Insulin-Like Growth Factor I/pharmacology , Interleukin-3/pharmacology , Lymphokines/metabolism , Matrix Metalloproteinase 2/metabolism , Proto-Oncogene Proteins c-akt , STAT1 Transcription Factor , Stem Cell Factor/pharmacology , Thrombopoietin/pharmacology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
10.
Stem Cells ; 19(5): 453-66, 2001.
Article in English | MEDLINE | ID: mdl-11553854

ABSTRACT

To better define the role HIV-related chemokine receptor-chemokine axes play in human hematopoiesis, we investigated the function of the CXCR4 and CCR5 receptors in human myeloid, T- and B-lymphoid cell lines selected for the expression of these receptors (CXCR4(+), CXCR4(+) CCR5(+), and CCR5(+) cell lines). We evaluated the phosphorylation of MAPK p42/44, AKT, and STAT proteins and examined the ability of the ligands for these receptors (stromal-derived factor-1 [SDF-1] and macrophage inflammatory protein-1beta [MIP-1beta]) to influence cell growth, apoptosis, adhesion, and production of vascular endothelial growth factors (VEGF), matrix metalloproteinases (MMPs) and their tissue inhibitors (TIMPs) in these cell lines. We found that A) SDF-1, after binding to CXCR4, activates multiple signaling pathways and that in comparison with the MIP-1beta-CCR5 axis, plays a privileged role in hematopoiesis; B) SDF-1 activation of the MAPK p42/44 pathway and the PI-3K-AKT axis does not affect proliferation and apoptosis but modulates integrin-mediated adhesion to fibronectin, and C) SDF-1 induces secretion of VEGF, but not of MMPs or TIMPs. Thus the role of SDF-1 relates primarily to the interaction of lymphohematopoietic cells with their microenvironment and does not directly influence their proliferation or survival. We conclude that perturbation of the SDF-1-CXCR4 axis during HIV infection may affect interactions of hematopoietic cells with the hematopoietic microenvironment.


Subject(s)
Chemokines, CXC/metabolism , Endothelial Growth Factors/biosynthesis , Hematopoietic Stem Cells/cytology , Integrins/metabolism , Lymphokines/biosynthesis , Receptors, CXCR4/metabolism , Apoptosis , Blotting, Western , Cell Adhesion , Cell Division , Cell Line , Cell Survival , Chemokine CXCL12 , Coloring Agents/pharmacology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , HL-60 Cells , Humans , Jurkat Cells , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Protein Binding , Reverse Transcriptase Polymerase Chain Reaction , Tetrazolium Salts/pharmacology , Thiazoles/pharmacology , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tissue Inhibitor of Metalloproteinase-2/metabolism , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
11.
Blood ; 97(10): 3075-85, 2001 May 15.
Article in English | MEDLINE | ID: mdl-11342433

ABSTRACT

The aim of this study was to explore further the hypothesis that early stages of normal human hematopoiesis might be coregulated by autocrine/paracrine regulatory loops and by cross-talk among early hematopoietic cells. Highly purified normal human CD34(+) cells and ex vivo expanded early colony-forming unit-granulocyte-macrophage (CFU-GM)-derived, burst forming unit-erythroid (BFU-E)-derived, and CFU-megakaryocyte (CFU-Meg)-derived cells were phenotyped for messenger RNA expression and protein secretion of various growth factors, cytokines, and chemokines to determine the biological significance of this secretion. Transcripts were found for numerous growth factors (kit ligand [KL], FLT3 ligand, fibroblast growth factor-2 [FGF-2], vascular endothelial growth factor [VEGF], hepatocyte growth factor [HGF], insulinlike growth factor-1 [IGF-1], and thrombopoietin [TPO]); cytokines (tumor necrosis factor-alpha, Fas ligand, interferon alpha, interleukin 1 [IL-1], and IL-16); and chemokines (macrophage inflammatory protein-1alpha [MIP-1alpha], MIP-1beta, regulated upon activation, normal T cell expressed and secreted [RANTES], monocyte chemotactic protein-3 [MCP-3], MCP-4, IL-8, interferon-inducible protein-10, macrophage-derived chemokine [MDC], and platelet factor-4 [PF-4]) to be expressed by CD34(+) cells. More importantly, the regulatory proteins VEGF, HGF, FGF-2, KL, FLT3 ligand, TPO, IL-16, IGF-1, transforming growth factor-beta1 (TGF-beta1), TGF-beta2, RANTES, MIP-1alpha, MIP-1beta, IL-8, and PF-4 were identified in media conditioned by these cells. Moreover, media conditioned by CD34(+) cells were found to inhibit apoptosis and slightly stimulate the proliferation of other freshly isolated CD34(+) cells; chemo-attract CFU-GM- and CFU-Meg-derived cells as well as other CD34(+) cells; and, finally, stimulate the proliferation of human endothelial cells. It was also demonstrated that these various hematopoietic growth factors, cytokines, and chemokines are expressed and secreted by CFU-GM-, CFU-Meg-, and BFU-E-derived cells. It is concluded that normal human CD34(+) cells and hematopoietic precursors secrete numerous regulatory molecules that form the basis of intercellular cross-talk networks and regulate in an autocrine and/or a paracrine manner the various stages of normal human hematopoiesis.


Subject(s)
Chemokines/metabolism , Cytokines/metabolism , Growth Substances/metabolism , Hematopoiesis , Hematopoietic Stem Cells/physiology , Homeostasis , Antigens, CD34/analysis , Cell Division , Cell Separation , Cell Survival , Cells, Cultured , Chemokines/genetics , Chemotaxis , Culture Media, Conditioned , Cytokines/genetics , Erythroblasts/physiology , Flow Cytometry , Gene Expression , Granulocytes/physiology , Growth Substances/genetics , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , Humans , Megakaryocytes/physiology , RNA, Messenger/analysis , Rh-Hr Blood-Group System/physiology
12.
Stem Cells ; 19(2): 99-107, 2001.
Article in English | MEDLINE | ID: mdl-11239164

ABSTRACT

Autocrine/paracrine regulatory mechanisms are believed to play a role in the pathophysiology of several hematologic malignancies. Evidence is accumulating that various growth factors, cytokines, and chemokines are expressed and secreted by normal early and differentiated hematopoietic cells and thus could also regulate normal hematopoiesis in an autocrine/paracrine manner. In this review we summarize recent advances in identification and understanding of the role of autocrine/paracrine axes in the growth of both malignant and normal human hematopoietic cells. Better understanding of intercellular crosstalk operating in normal and pathological states and the mechanisms regulating synthesis of these endogenously produced factors (potential targets for various pharmacological approaches) may allow us to improve antileukemia treatments, undertake more efficient ex vivo stem cell expansion, and develop other therapeutic strategies.


Subject(s)
Autocrine Communication/physiology , Hematopoiesis/physiology , Paracrine Communication/physiology , Animals , Chemokines/genetics , Cytokines/genetics , Gene Expression , Growth Substances/genetics , Hematopoietic Stem Cells , Humans , Mice , RNA, Messenger
13.
Blood ; 96(13): 4142-51, 2000 Dec 15.
Article in English | MEDLINE | ID: mdl-11110685

ABSTRACT

The role of the chemokine binding stromal-derived factor 1 (SDF-1) in normal human megakaryopoiesis at the cellular and molecular levels and its comparison with that of thrombopoietin (TPO) have not been determined. In this study it was found that SDF-1, unlike TPO, does not stimulate alpha(IIb)beta(3)(+) cell proliferation or differentiation or have an antiapoptotic effect. However, it does induce chemotaxis, trans-Matrigel migration, and secretion of matrix metalloproteinase 9 (MMP-9) and vascular endothelial growth factor (VEGF) by these cells, and both SDF-1 and TPO increase the adhesion of alpha(IIb)beta(3)(+) cells to fibrinogen and vitronectin. Investigating the intracellular signaling pathways induced by SDF-1 and TPO revealed some overlapping patterns of protein phosphorylation/activation (mitogen-activated protein kinase [MAPK] p42/44, MAPK p38, and AKT [protein kinase B]) and some that were distinct for TPO (eg, JAK-STAT) and for SDF-1 (eg, NF-kappa B). It was also found that though inhibition of phosphatidyl-inositol 3-kinase (PI-3K) by LY294002 in alpha(IIb)beta(3)(+) cells induced apoptosis and inhibited chemotaxis adhesion and the secretion of MMP-9 and VEGF, the inhibition of MAPK p42/44 (by the MEK inhibitor U0126) had no effect on the survival, proliferation, and migration of these cells. Hence, it is suggested that the proliferative effect of TPO is more related to activation of the JAK-STAT pathway (unique to TPO), and the PI-3K-AKT axis is differentially involved in TPO- and SDF-1-dependent signaling. Accordingly, PI-3K is involved in TPO-mediated inhibition of apoptosis, TPO- and SDF-1-regulated adhesion to fibrinogen and vitronectin, and SDF-1-mediated migration. This study expands the understanding of the role of SDF-1 and TPO in normal human megakaryopoiesis and indicates the molecular basis of the observed differences in cellular responses. (Blood. 2000;96:4142-4151)


Subject(s)
Chemokines, CXC/physiology , Megakaryocytes/cytology , Thrombopoietin/physiology , Adult , Apoptosis/drug effects , Blood Platelets/drug effects , Blood Platelets/metabolism , Calcium/metabolism , Calcium Signaling/drug effects , Caspase 3 , Caspases/metabolism , Cell Adhesion/drug effects , Cell Cycle/drug effects , Chemokine CXCL12 , Chemokines, CXC/pharmacology , Chemotaxis/drug effects , Chromones/pharmacology , Collagen , Drug Combinations , Endothelial Growth Factors/biosynthesis , Endothelial Growth Factors/genetics , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Fibrinogen/metabolism , Gene Expression Regulation/drug effects , Humans , Ion Transport/drug effects , Laminin , Lymphokines/biosynthesis , Lymphokines/genetics , MAP Kinase Signaling System/drug effects , Megakaryocytes/drug effects , Metalloendopeptidases/biosynthesis , Metalloendopeptidases/genetics , Morpholines/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Platelet Glycoprotein GPIIb-IIIa Complex/physiology , Poly(ADP-ribose) Polymerases/metabolism , Protein Kinases/metabolism , Protein Processing, Post-Translational/drug effects , Proteoglycans , Recombinant Proteins/pharmacology , Thrombopoietin/pharmacology , Transcription Factors/metabolism , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , Vitronectin/metabolism
14.
Exp Hematol ; 28(11): 1274-85, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11063876

ABSTRACT

As stromal cell-derived factor-1 (SDF-1), macrophage inflammatory protein-1alpha (MIP-1alpha), and interleukin-8 (IL-8) are implicated in the homing and mobilization of human hematopoietic progenitors (HPC), we hypothesized that these chemokines mediate the migration of HPC across subendothelial basement membranes by regulating production of matrix metalloproteinases (MMPs) and their natural tissue inhibitors (TIMPs). Assays for migration across reconstituted basement membrane (Matrigel) and chemotaxis were carried out using CD34(+) cells derived from normal human bone marrow (BM) and mobilized peripheral blood (PB). Secretion of MMPs and TIMPs was evaluated by zymography and reverse zymography and gene expression by RT-PCR. We found that an SDF-1 gradient increases the chemotaxis of BM and PB CD34(+) cells across Matrigel (BM > PB), which is blocked by inhibitors of MMPs (o-phenanthroline, rhTIMP-1, rhTIMP-2, and anti-MMP-9 and anti-MMP-2 antibodies) but enhanced by tumor necrosis factor-alpha (TNF-alpha), a strong stimulator of MMPs. Preincubation of these cells with SDF-1 stimulated the secretion of MMP-2 and MMP-9 in BM and PB CD34(+) cells but of TIMP-1 and TIMP-2 only in PB CD34(+) cells. Preincubation with MIP-1alpha and IL-8 also stimulated the secretion of MMP-9 and MMP-2 (BM > PB), but with respect to TIMPs, the effect was reversed (PB > BM), resulting in trans-Matrigel migration of BM but not of PB CD34(+) cells. We therefore propose that MMPs and TIMPs are involved in 1) SDF-1-induced chemotaxis of human HPC across subendothelial basement membranes, and 2) MIP-1alpha- and IL-8-stimulated migration of HPC.


Subject(s)
Chemokines/pharmacology , Hematopoietic Stem Cells/metabolism , Matrix Metalloproteinases/biosynthesis , Tissue Inhibitor of Metalloproteinases/biosynthesis , Antigens, CD34 , Blood Cells/cytology , Blood Cells/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Differentiation , Cells, Cultured , Hematopoietic Stem Cells/cytology , Humans , Organ Specificity
15.
Exp Hematol ; 28(12): 1334-42, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11146155

ABSTRACT

The resistance of human bone marrow (BM) CD34(+) cells to human immunodeficiency virus (HIV) infection is at this point not fully understood. Recently we reported that the chemokines MIP-1alpha, MIP-1beta, and RANTES secreted by BM-derived CD34(+) cells may compete with the macrophagotropic HIV (R5 HIV) strain for the CCR5 coreceptor.In this study we extended our previous observations and examined various lympho-hematopoietic CD34(+) cells isolated from thymus (Th), cord blood (CB), mobilized peripheral blood (mPB), and BM for the expression of beta-chemokines binding to CCR5, i.e., MIP-1alpha, MIP-1beta, RANTES, MCP-2, MCP-3, and MCP-4, and the alpha chemokine SDF-1 (binding to CXCR4) as these chemokines may compete with the R5 and X4 HIV strains, respectively, for entry into cells. We found that Th-, CB-, mPB-, and BM-derived CD34(+) cells express mRNA transcripts for all the beta-chemokines tested but not for SDF-1. Using sensitive ELISA assays we found that although MIP-1alpha and MIP-1beta proteins were secreted by all the lympho-hematopoietic CD34(+) cells tested, RANTES was detectable only in media conditioned by BM- and CB-derived CD34(+) cells and not Th-derived cells. However, media conditioned by BM-, mPB- and Th-derived CD34(+) cells protected the T lymphocytic cell line (PB-1) from infection by the R5 but not the X4 HIV strain. Hence this study demonstrates that beta-chemokines are secreted by lympho-hematopoietic CD34(+) cells originating from various sources and that these endogenously secreted chemokines may limit entry of the R5 HIV strain into the cells by competing for the CCR5 coreceptor.


Subject(s)
Bone Marrow Cells/virology , Chemokines/biosynthesis , Cytokines , Fetal Blood/cytology , HIV/pathogenicity , Hematopoietic Stem Cells/virology , Thymus Gland/cytology , Antigens, CD34/analysis , Bone Marrow Cells/immunology , Chemokine CCL3 , Chemokine CCL4 , Chemokine CCL5/genetics , Chemokine CCL5/physiology , Chemokine CCL7 , Chemokine CCL8 , Chemokines/genetics , Culture Media, Conditioned , Gene Expression , Hematopoietic Stem Cells/immunology , Humans , Macrophage Inflammatory Proteins/genetics , Macrophage Inflammatory Proteins/physiology , Monocyte Chemoattractant Proteins/genetics , Monocyte Chemoattractant Proteins/physiology , RNA, Messenger/analysis , Receptors, CCR5/metabolism , Reverse Transcriptase Polymerase Chain Reaction
16.
Leuk Lymphoma ; 39(5-6): 485-93, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11342332

ABSTRACT

Human malignant non-Hodgkin's lymphomas (NHL) represent a heterogeneous group of neoplasms, which vary in their clinical behavior and pathophysiology. Matrix metalloproteinases (MMPs) and their tissue inhibitors (TIMPs) have been shown to play a role in the pathophysiology and clinical aggressiveness of human NHL. In this setting, MMP-9 and TIMP-1 appear to be the most important members of the MMP and TIMP families, and overexpression of both correlates with a poor clinical outcome of patients with NHL. MMP-9 and TIMP-1, however, act through different mechanisms and are produced by different cell types. Expression of both is upregulated by interleukin-6 (IL-6), a cytokine that is known as one of the factors involved in the pathophysiology of human NHL. In this review we summarize the complex regulation of MMP and TIMP expression in human NHL and propose a mechanism by which MMP-9, TIMP-1 and IL-6 may influence the biology of these tumors.


Subject(s)
Lymphoma, Non-Hodgkin/metabolism , Matrix Metalloproteinases/metabolism , Tissue Inhibitor of Metalloproteinases/metabolism , Cytokines/pharmacology , Gene Expression Regulation/drug effects , Humans , Lymphoma, Non-Hodgkin/etiology
17.
Blood ; 94(6): 2080-9, 1999 Sep 15.
Article in English | MEDLINE | ID: mdl-10477738

ABSTRACT

We showed previously that human malignant non-Hodgkin's lymphomas (NHL) degrade extracellular matrix (ECM) components through the action of metalloproteinases and that elevated expression of matrix metalloproteinase-9 (MMP-9) and tissue inhibitor of metalloproteinase-1 (TIMP-1) correlated with a poor clinical outcome in patients with NHL. In the present study we sought to investigate whether there is any correlation between the expression of gelatinases (MMP-2 and MMP-9), TIMP-1, and the expression of cytokines and growth factors such as interleukin-1beta (IL-1beta), IL-6, IL-10, tumor necrosis factor alpha (TNF-alpha), transforming growth factor beta (TGFbeta), and basic fibroblast growth factor (bFGF) in human NHL. In lymphoma tissues obtained from 32 patients, elevated expression of IL-6 correlated significantly with elevated messenger RNA (mRNA) levels of MMP-9, MMP-2, and TIMP-1. Moreover, in human lymphoid cell lines of B- and T-cell origin (Raji, Jurkat, and NC-37), IL-6 stimulated production of MMP-9 and MMP-2 but not TIMP-1. In the Matrigel invasion assay IL-6 significantly upregulated transmigration of Raji and Jurkat cells, which in turn was inhibited by recombinant human TIMP-1 and anti-MMP-9 and MMP-2 antibodies. We postulate that IL-6 may play a role in the clinical aggressiveness of human NHL by stimulating MMP production.


Subject(s)
Collagenases/genetics , Cytokines/genetics , Gelatinases/genetics , Gene Expression Regulation, Neoplastic/physiology , Growth Substances/genetics , Interleukin-6/genetics , Lymphoma, Non-Hodgkin/genetics , Metalloendopeptidases/genetics , Tissue Inhibitor of Metalloproteinase-1/genetics , Culture Media, Conditioned , DNA Primers , Gene Expression Regulation, Enzymologic/immunology , Gene Expression Regulation, Enzymologic/physiology , Gene Expression Regulation, Neoplastic/immunology , Humans , Lymphoma, Non-Hodgkin/enzymology , Lymphoma, Non-Hodgkin/pathology , Matrix Metalloproteinase 2 , Matrix Metalloproteinase 9 , Neoplasm Invasiveness , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
18.
Br J Haematol ; 105(2): 402-11, 1999 May.
Article in English | MEDLINE | ID: mdl-10233411

ABSTRACT

We compared the expression of matrix metalloproteinases (MMP-2 and MMP-9) and tissue inhibitors of metalloproteinases (TIMP-1 and TIMP-2) in bone marrow acute myelogenous leukaemia (AML) blasts and leukaemic cell lines (HEL, HL-60, K-562 and KG-1) with their expression in normal bone marrow cells. All AML samples and leukaemic cell lines tested expressed MMP-9 and/or MMP-2 mRNA and, accordingly, these gelatinases were secreted into media. Moreover, TIMP-1 and TIMP-2 mRNA and secreted proteins were demonstrated in all the AML samples. Although all the leukaemic cell lines expressed TIMP-1, the HL-60 cells also expressed TIMP-2. In contrast, normal steady-state bone marrow immature progenitor cells (CD34+ cells) did not express or secrete either MMP-2 or MMP-9, but more mature mononuclear cells from normal bone marrow expressed and secreted MMP-9. Also, normal bone marrow CD34+ cells and mononuclear cells expressed TIMP-1 and TIMP-2 mRNA, but these proteins were not detectable by reverse zymography. Furthermore, whereas bone marrow fibroblasts and endothelial cells secreted only latent MMP-2, the activated form of this enzyme was found in media conditioned by cells obtained from long-term cultures of normal and AML bone marrow adherent layers. Our finding of up-regulated production of gelatinases, TIMP-1 and TIMP-2 by leukaemic cells suggests that these proteins may be implicated in the invasive phenotype of AML.


Subject(s)
Bone Marrow Cells/metabolism , Collagenases/metabolism , Gelatinases/metabolism , Leukemia, Myeloid, Acute/metabolism , Metalloendopeptidases/metabolism , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tissue Inhibitor of Metalloproteinase-2/metabolism , Adolescent , Adult , Aged , Female , Hematopoietic Stem Cells/metabolism , Humans , Male , Matrix Metalloproteinase 2 , Matrix Metalloproteinase 9 , Middle Aged , Reverse Transcriptase Polymerase Chain Reaction/methods , Tumor Cells, Cultured
19.
Blood ; 93(10): 3379-90, 1999 May 15.
Article in English | MEDLINE | ID: mdl-10233890

ABSTRACT

The mechanism(s) underlying the release of stem/progenitor cells from bone marrow into the circulation is poorly understood. We hypothesized that matrix metalloproteinases (MMPs), especially gelatinases, which are believed to participate in the proteolysis of basement membranes and in the migration of leukocytes, may facilitate this process. First, we investigated whether CD34(+) stem/progenitor cells express gelatinases A (MMP-2) and/or B (MMP-9) and whether growth factors and cytokines (granulocyte colony-stimulating factor [G-CSF], granulocyte-macrophage colony-stimulating factor [GM-CSF], stem cell factor [SCF], macrophage colony-stimulating factor [M-CSF], interleukin-3 [IL-3], IL-6, IL-8, and tumor necrosis factor-alpha [TNF-alpha]) are able to modulate their expression. Next, we examined the transmigration of these stem/progenitor cells through reconstituted basement membrane (Matrigel) and its modulation by growth factors and cytokines. CD34(+) cells were obtained from steady-state bone marrow and peripheral blood (from leukapheresis products collected either in steady-state hematopoiesis or after mobilization with G-CSF plus chemotherapy or G-CSF alone). We found that peripheral blood CD34(+) cells, regardless of whether they were mobilized or not, strongly expressed both gelatinases (MMP-2 and MMP-9) in contrast to steady-state bone marrow CD34(+) cells, which did not. However, all the growth factors and cytokines tested could induce MMP-2 and MMP-9 secretion by the latter cells. Moreover, the stimulatory effects of G-CSF and SCF on both MMP-2 and MMP-9 secretion were found to be significantly higher in CD34(+) cells isolated from bone marrow than in those from peripheral blood. In addition TNF-alpha, GM-CSF, and IL-6 increased the secretion of a partially active form of MMP-2. Basal transmigration of bone marrow CD34(+) cells through Matrigel was lower than that of peripheral blood CD34(+) cells (P <.0001), but growth factors and cytokines increased it by 50% to 150%. Positive correlations were established between expression of gelatinases and CD34(+) cell migration (r >.9). The stimulatory effect of G-CSF was significantly greater on the migration of CD34(+) cells from bone marrow than on those from peripheral blood (P =.004). Moreover, CD34(+) cell migration was reduced to approximately 50% by antibodies to MMP-2 and MMP-9, tissue inhibitors of metalloproteinases (rhTIMP-1 and -2), and o-phenanthroline. TNF-alpha-induced gelatinase secretion and migration of CD34(+) cells and of clonogenic progenitors (colony-forming unit-granulocyte-macrophage [CFU-GM], burst-forming unit-erythroid [BFU-E], colony-forming unit granulocyte, erythroid, monocyte, megakaryocyte [CFU-GEMM], and colony-forming unit-megakaryocyte [CFU-MK]) were dose-dependent. Therefore, this study demonstrated that CD34(+) cells that are circulating in peripheral blood express both MMP-2 and MMP-9 and transmigrate through Matrigel. In contrast, CD34(+) cells from steady-state bone marrow acquire similar properties after exposure to growth factors and cytokines, which upregulate expression of gelatinases and transmigration of these cells when they enter the bloodstream. Hence, we suggest that growth factors and cytokines induce release of stem/progenitor cells from bone marrow into peripheral blood during mobilization, as well as during steady-state hematopoiesis, by signaling through gelatinase pathways.


Subject(s)
Bone Marrow Cells/physiology , Cytokines/pharmacology , Gelatinases/genetics , Gene Expression Regulation, Enzymologic/physiology , Growth Substances/pharmacology , Hematopoietic Stem Cells/physiology , Antigens, CD34 , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Basement Membrane/physiology , Bone Marrow Cells/cytology , Bone Marrow Cells/pathology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cells, Cultured , Chemotaxis , Collagenases/genetics , Colony-Forming Units Assay , Culture Media, Conditioned , Female , Filgrastim , Gene Expression Regulation, Enzymologic/drug effects , Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoiesis , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Humans , Matrix Metalloproteinase 2 , Matrix Metalloproteinase 9 , Metalloendopeptidases/genetics , Recombinant Proteins/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/pharmacology
20.
Semin Oncol ; 26(1 Suppl 3): 10-6, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10203265

ABSTRACT

Considering the efficacy of docetaxel (Taxotere, Rhône-Poulenc Rorer, Antony, France) and doxorubicin in advanced breast cancer and their potential noncross-resistance, two pilot studies of docetaxel/doxorubicin (TA)-based combinations were conducted, one being a phase I dose-finding study of TA and the second a phase II study of docetaxel/doxorubicin/cyclophosphamide (TAC). The only significant toxicity, seen in both trials, was neutropenia and its consequences such as febrile neutropenia without significant documented infections. Extrahematologic and particularly docetaxel-specific side effects (fluid retention) were mild. Particularly noteworthy was the absence of significant cardiac toxicity; overall, there was only one case of congestive heart failure (1%). In terms of efficacy, response rates in excess of 70% and 80% were noted in both studies, even for patients with visceral metastases. Several phase III randomized trials using TA or TAC are presently being performed in first-line metastatic breast cancer and most importantly in the adjuvant setting to assess whether TA-based combinations will change the natural history of breast cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Paclitaxel/analogs & derivatives , Taxoids , Breast Neoplasms/pathology , Chemotherapy, Adjuvant , Clinical Trials as Topic , Cyclophosphamide/administration & dosage , Docetaxel , Doxorubicin/administration & dosage , Humans , Neoplasm Metastasis , Paclitaxel/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...