Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
2.
Leukemia ; 18(2): 341-7, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14628073

ABSTRACT

Transplantation of immunodeficient mice with human hematopoietic cells has greatly facilitated studies of the earliest stages of human hematopoiesis. These include demonstration of the ability of injected 'human-specific' hematopoietic growth factors to enhance the production of human cells at multiple levels of differentiation. In contrast, the effects of continuous exposure to such molecules have not been well investigated. Here, we show that nonobese diabetic severe combined immunodeficiency mice genetically engineered to produce ng/ml serum levels of human interleukin-3 (IL-3), granulocyte/macrophage-stimulating factor (GM-CSF) and Steel factor (SF) display a complex phenotype when transplanted with primitive human bone marrow (BM) or fetal liver cells. This phenotype is characterized by an enhancement of terminal human myelopoiesis and a matched suppression of terminal human erythropoiesis, with a slight reduction in human B-lymphopoiesis in the BM of the engrafted mice. Human clonogenic progenitors are more prevalent in the blood of the transplanted growth factor-producing mice and this is accompanied by a very marked reduction of more primitive human cells in the BM. Our findings suggest that long-term exposure of primitive human hematopoietic cells to elevated levels of human IL-3, GM-CSF and SF in vivo may deleteriously affect the stem cell compartment, while expanding terminal myelopoiesis.


Subject(s)
Hematopoietic Cell Growth Factors/pharmacology , Hematopoietic Stem Cell Mobilization/methods , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/drug effects , Animals , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Hematopoiesis/drug effects , Hematopoietic Cell Growth Factors/genetics , Hematopoietic Stem Cells/physiology , Humans , Interleukin-3/genetics , Interleukin-3/pharmacology , Mice , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Regeneration , Stem Cell Factor/genetics , Stem Cell Factor/pharmacology , Transplantation, Heterologous
3.
Blood ; 96(12): 3979-81, 2000 Dec 01.
Article in English | MEDLINE | ID: mdl-11090087

ABSTRACT

Transplantable human hematopoietic stem cells (competitive repopulating units [CRU]) can be quantitated based on their ability to produce large populations of lymphoid and myeloid progeny within 6 weeks in the marrow of intravenously injected, sublethally irradiated NOD/SCID mice. It is shown that the proportions of total injected human fetal liver and cord blood CRU in the marrow of mice 24 hours after transplantation are 5% and 7%, respectively, as determined by limiting-dilution assays in other primary and secondary NOD/SCID mice. The similarity in these 2 seeding efficiency values suggests that mechanisms regulating the ability of human hematopoietic stem cells to enter the marrow from the blood, at least in this xenotransplant model, do not change between fetal life and birth. In addition, it appears that previously reported human stem cell frequencies and their in vivo self-renewal activity measured in NOD/SCID mice have been markedly underestimated. (Blood. 2000;96:3979-3981)


Subject(s)
Hematopoietic Stem Cells/cytology , Mice, Inbred NOD/blood , Mice, Inbred NOD/immunology , Mice, SCID/blood , Mice, SCID/immunology , Animals , Bone Marrow Cells/cytology , Fetal Blood/cytology , Fetal Tissue Transplantation/methods , Graft Survival , Hematopoietic Stem Cell Transplantation/methods , Humans , Liver/cytology , Mice , Transplantation, Heterologous
4.
Blood ; 94(11): 3722-9, 1999 Dec 01.
Article in English | MEDLINE | ID: mdl-10572085

ABSTRACT

Nonobese diabetic/severe combined immunodeficient (NOD/SCID) mice transplanted with human cord blood or adult marrow cells and injected 6 weeks posttransplant with 2 daily doses of transforming growth factor-beta(1) (TGF-beta(1)), monocyte chemoattractant protein-1 (MCP-1), or a nonaggregating form of macrophage inflammatory protein-1alpha (MIP-1alpha) showed unique patterns of inhibition of human progenitor proliferation 1 day later. TGF-beta(1) was active on long-term culture initiating cells (LTC-IC) and on primitive erythroid and granulopoietic colony-forming cells (HPP-CFC), but had no effect on mature CFC. MCP-1 inhibited the cycling of both types of HPP-CFC but not LTC-IC. MIP-1alpha did not inhibit either LTC-IC or granulopoietic HPP-CFC but was active on erythroid HPP-CFC and mature granulopoietic CFC. All of these responses were independent of the source of human cells transplanted. LTC-IC of either human cord blood or adult marrow origin continue to proliferate in NOD/SCID mice for many weeks, although the turnover of all types of human CFC in mice transplanted with adult human marrow (but not cord blood) is downregulated after 6 weeks. Interestingly, administration of either MIP-1beta, an antagonist of both MIP-1alpha and MCP-1 or MCP-1(9-76), an antagonist of MCP-1 (and MCP-2 and MCP-3), into mice in which human marrow-derived CFC had become quiescent, caused the rapid reactivation of these progenitors in vivo. These results provide the first definition of stage-specific inhibitors of human hematopoietic progenitor cell cycling in vivo. In addition they show that endogenous chemokines can contribute to late graft failure, which can be reversed by the administration of specific antagonists.


Subject(s)
Chemokine CCL2/pharmacology , Hematopoiesis , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Macrophage Inflammatory Proteins/pharmacology , Transforming Growth Factor beta/pharmacology , Adult , Animals , Cell Differentiation/drug effects , Cell Division/drug effects , Cell Lineage/drug effects , Cells, Cultured , Chemokine CCL3 , Chemokine CCL4 , Hematopoiesis/drug effects , Humans , Mice , Mice, Inbred NOD , Mice, SCID
5.
Blood ; 94(8): 2686-95, 1999 Oct 15.
Article in English | MEDLINE | ID: mdl-10515872

ABSTRACT

Comparative measurements of different types of hematopoietic progenitors present in human fetal liver, cord blood, and adult marrow showed a large (up to 250-fold), stage-specific, but lineage-unrestricted, amplification of the colony-forming cell (CFC) compartment in the fetal liver, with a higher ratio of all types of CFC to long-term culture-initiating cells (LTC-IC) and a lower ratio of total (mature) cells to CFC. Human fetal liver LTC-IC were also found to produce more CFC in LTC than cord blood or adult marrow LTC-IC, and more of the fetal liver LTC-IC-derived CFC were erythroid. Human fetal liver cells regenerated human multilineage hematopoiesis in NOD/SCID mice with the same kinetics as human cord blood and adult marrow cells, but sustained a high level of terminal erythropoiesis not seen in adult marrow-engrafted mice unless exogenous human erythropoietin (Epo) was injected. This may be due to a demonstrated 10-fold lower activity of murine versus human Epo on human cells, sufficient to distinguish between a differential Epo sensitivity of fetal and adult erythroid precursors. Examination of human LTC-IC, CFC, and erythroblasts generated either in NOD/SCID mice and/or in LTC showed the types of cells and hemoglobins produced also to reflect their ontological origin, regardless of the environment in which the erythroid precursors were generated. We suggest that ontogeny may affect the behavior of cells at many stages of hematopoietic cell differentiation through key changes in shared signaling pathways.


Subject(s)
Hematopoiesis , Hematopoietic Stem Cells/cytology , Liver/embryology , Adult , Animals , Bone Marrow Transplantation , Cell Differentiation , Cell Lineage , Cells, Cultured , Colony-Forming Units Assay , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/drug effects , Erythropoiesis/drug effects , Erythropoietin/pharmacology , Fetal Blood/cytology , Hematopoietic Stem Cell Transplantation , Humans , Liver/cytology , Mice , Mice, Inbred NOD , Mice, SCID , Species Specificity , Transplantation, Heterologous
6.
Blood ; 93(2): 481-7, 1999 Jan 15.
Article in English | MEDLINE | ID: mdl-9885209

ABSTRACT

Self-renewal is considered to be the essential defining property of a stem cell. Retroviral marking, in vitro amplification, and serial transplantation of human cells that can sustain long-term lymphomyelopoiesis in vivo have provided evidence that human hematopoietic stem cell self-renewal occurs both in vitro and in vivo. To investigate whether this process can be manipulated by cytokines, we administered two different combinations of human growth factors to sublethally irradiated nonobese diabetic/severe combined immunodeficient (SCID) mice transplanted with 10(7) light-density human cord blood cells and then performed secondary transplants to compare the number of transplantable human lymphomyeloid reconstituting cells present 4 to 6 weeks post-transplant. A 2-week course of Steel factor + interleukin (IL)-3 + granulocyte-macrophage colony-stimulating factor + erythropoietin (3 times per week just before sacrifice) specifically and significantly enhanced the numbers of transplantable human lymphomyeloid stem cells detectable in the primary mice (by a factor of 10). Steel factor + Flt3-ligand + IL-6 (using either the same schedule or administered daily until sacrifice 4 weeks post-transplant) gave a threefold enhancement of this population. These effects were obtained at a time when the regenerating human progenitor populations in such primary mice are known to be maximally cycling even in the absence of growth factor administration suggesting that the underlying mechanism may reflect an ability of these growth factors to alter the probability of differentiation of stem cells stimulated to proliferate in vivo.


Subject(s)
Growth Substances/pharmacology , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Animals , Cell Count , Cell Differentiation , Cell Division , Erythropoietin/administration & dosage , Fetal Blood/cytology , Flow Cytometry , Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Humans , Infant, Newborn , Interleukin-3/administration & dosage , Interleukin-6/administration & dosage , Membrane Proteins/administration & dosage , Mice , Mice, Inbred NOD , Mice, SCID , Stem Cell Factor/administration & dosage
7.
Blood ; 92(7): 2338-44, 1998 Oct 01.
Article in English | MEDLINE | ID: mdl-9746772

ABSTRACT

The long-term culture (LTC) system has been useful for analyzing mechanisms by which stromal cells regulate the proliferative activity of primitive normal, but not chronic myeloid leukemia (CML), hematopoietic progenitor cells. In previous studies, we identified two endogenous inhibitors in this system. One is transforming growth factor-beta (TGF-beta), which is equally active on primitive normal and CML progenitors. The other we now show to be monocyte chemoattractant protein-1 (MCP-1). Thus, MCP-1, when added to LTC, blocked the activation of primitive normal progenitors but did not arrest the cycling of primitive CML progenitors. Moreover, the endogenous inhibitory activity of LTC stromal layers could be overcome by the addition of neutralizing antibodies to MCP-1, but not to macrophage inflammatory protein-1alpha (MIP-1alpha). However, neither of these antibodies antagonized the inhibitory activity of NAc-Ser-Asp-Lys-Pro (AcSDKP) on primitive normal but not CML progenitor cycling in this system. Moreover, none of six other -C-C- or -C-X-C- chemokines, previously shown to inhibit primitive normal human CFC proliferation in semisolid assays, were found to act as negative regulators when added to normal LTC. These results provide further support for the concept that primitive CML progenitor cell proliferation is deregulated when these cells are exposed to limiting concentrations of multiple inhibitors, only some of which have differential actions on normal and Ph+/BCR-ABL+ cells.


Subject(s)
Bone Marrow/pathology , Chemokine CCL2/physiology , Hematopoiesis/physiology , Hematopoietic Stem Cells/drug effects , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Macrophage Inflammatory Proteins/physiology , Neoplastic Stem Cells/drug effects , Transforming Growth Factor beta/physiology , Cell Cycle , Cell Division/drug effects , Cells, Cultured , Chemokine CCL2/pharmacology , Chemokine CCL3 , Chemokine CCL4 , Cytokines/pharmacology , Fusion Proteins, bcr-abl/physiology , Hematopoiesis/drug effects , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Humans , Macrophage Inflammatory Proteins/pharmacology , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/physiology , Oligopeptides/pharmacology , Recombinant Proteins/pharmacology , Signal Transduction , Stromal Cells/physiology , Tumor Cells, Cultured
8.
Blood ; 91(7): 2406-14, 1998 Apr 01.
Article in English | MEDLINE | ID: mdl-9516140

ABSTRACT

We have previously shown that intravenously injected peripheral blood (PB) or bone marrow (BM) cells from newly diagnosed chronic myeloid leukemia (CML) patients can engraft the BM of sublethally irradiated severe combined immunodeficient (SCID) mice. We now report engraftment results for chronic phase CML cells in nonobese diabetic (NOD)/SCID recipients which show the superiority of this latter model. Transplantation of NOD/SCID mice with 7 to 10 x 10(7) patient PB or BM cells resulted in the continuing presence of human cells in the BM of the mice for up to 7 months, and primitive human CD34+ cells, including those detectable as colony-forming cells (CFC), as long-term culture-initiating cells, or by their coexpression of Thy-1, were found in a higher proportion of the NOD/SCID recipients analyzed, and at higher levels than were seen previously in SCID recipients. The human CFC and total human cells present in the BM of the NOD/SCID mice transplanted with CML cells also contained higher proportions of leukemic cells than were obtained in the SCID model, and NOD/SCID mice could be repopulated with transplants of enriched CD34+ cells from patients with CML. These results suggest that the NOD/SCID mouse may allow greater engraftment and amplification of both normal and leukemic (Ph+) cells sufficient for the quantitation and characterization of the normal and leukemic stem cells present in patients with CML. In addition, this model should make practical the investigation of mechanisms underlying progression of the disease and the development of more effective in vivo therapies.


Subject(s)
Graft Survival , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/pathology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Adult , Aged , Animals , Blood Cell Count , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Middle Aged , Neoplasm Transplantation , Transplantation, Homologous
9.
Stem Cells ; 16 Suppl 1: 77-83; discussion 89, 1998.
Article in English | MEDLINE | ID: mdl-11012149

ABSTRACT

Chronic myeloid leukemia (CML) is a clonal myeloproliferative disorder in which there is a deregulated amplification of CML progenitors at intermediate stages of their differentiation along the myeloid, erythroid and megakaryocyte pathways. Such cell populations are routinely quantified using standard in vitro colony-forming cell (CFC) assays. The excessive production of leukemic CFC that is seen in most CML patients at diagnosis may be explained at least in part by their increased proliferative activity. An anomalous cycling behavior in vivo has also been found to extend to more primitive CML progenitor populations detectable as long-term culture-initiating cells (LTC-IC). Although the molecular basis of these changes in CML progenitor regulation is not fully understood at the level of the primitive CFC compartment, a selective inability of CML progenitors to be inhibited by certain -C-C-type chemokines has been demonstrated. Failure of the CML stem cell compartment to expand in vivo at the same rate as later progenitor cell types may be explained by their unique additional possession of an intrinsically upregulated probability of differentiation. Such a mechanism would be consistent with the observed loss of LTC-IC activity by CML cells incubated in vitro under conditions that sustain or expand normal LTC-IC populations. Initial clinical studies undertaken at our center established the feasibility of exploiting the differential behavior of primitive normal and CML cells in vitro as a potential purging strategy for reducing the leukemic stem cell content of CML marrow autografts. The results of a larger, second trial now in progress on a group of unselected patients are encouraging. Future studies of nonobese diabetic/severe-combined immunodeficiency mice engrafted with CML cells should provide another useful preclinical model for evaluating treatments that may more effectively eradicate the neoplastic clone in vivo.


Subject(s)
Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Animals , Colony-Forming Units Assay , Hematopoietic Stem Cells/pathology , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/blood , Mice , Mice, Inbred NOD , Mice, SCID , Reference Values , Transplantation, Heterologous
10.
Blood ; 89(12): 4307-16, 1997 Jun 15.
Article in English | MEDLINE | ID: mdl-9192753

ABSTRACT

Based on initial observations of human CD34+ Thy-1+ cells and long-term culture-initiating cells (LTC-IC) in the bone marrow of some sublethally irradiated severe combined immunodeficient (SCID) mice transplanted intravenously with normal human marrow cells, and the subsequent finding that the NOD/LtSz-scid/scid (NOD/SCID) mouse supports higher levels of human cell engraftment, we undertook a series of time course experiments to examine posttransplant changes in the number, tissue distribution, cycling activity, and in vivo differentiation pattern of various human hematopoietic progenitor cell populations in this latter mouse model. These studies showed typical rapid posttransplant recovery curves for human CD34- CD19+ (B-lineage) cells, CD34+ granulopoietic, erythroid, and multilineage colony-forming cells (CFC), LTC-IC, and CD34+ Thy-1+ cells from a small initial population representing <0.1% of the original transplant. The most primitive human cell populations reached maximum values at 5 weeks posttransplant, after which they declined. More mature cell types peaked after another 5 weeks and then declined. A 2-week course of thrice weekly injections of human Steel factor, interleukin (IL)-3, granulocyte-macrophage colony-stimulating factor (GM-CSF), and erythropoietin (administered just before the mice were killed for analysis) did not alter the pace of regeneration of either primitive or mature human hematopoietic cells, or their predominantly granulopoietic and B-lymphoid pattern of differentiation, although a significant enhancing effect on the level of human cell engraftment sustained after 3 months was noted. Cycling studies showed the human CFC present at 4 to 5 weeks posttransplant to be rapidly proliferating even in mice not given human growth factors. However, by 10 weeks and thereafter, only quiescent human CFC were detected; interestingly, even in mice that were given the 2-week course of growth factor injections. These studies indicate the use of this model for future analysis of the properties and in vivo regulation of primitive human hematopoietic cells that possess in vivo repopulating ability.


Subject(s)
Bone Marrow Transplantation/pathology , Cell Lineage , Hematopoiesis , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Severe Combined Immunodeficiency/pathology , Transplantation, Heterologous/pathology , Animals , Bone Marrow/pathology , Cell Cycle , Cell Division , Cells, Cultured , Female , Graft Survival , Hematopoietic Cell Growth Factors/pharmacology , Humans , Male , Mice , Mice, Inbred NOD , Mice, Mutant Strains , Mice, SCID , Radiation Chimera , Species Specificity , Spleen/pathology
11.
Blood ; 87(4): 1539-48, 1996 Feb 15.
Article in English | MEDLINE | ID: mdl-8608245

ABSTRACT

Progress in understanding the abnormal regulation of hematopoiesis in chronic myelogenous leukemia (CML) would be facilitated if neoplastic cells, at all stages of the disease, could be studied in an animal model. In this report, we show that irradiated severe combined immunodeficient (SCID) mice can be transplanted with both normal (Philadelphia chromosome [Ph]-negative) and neoplastic (Ph+) cells from CML patients with either chronic or blast phase disease. Mice transplanted with peripheral blood (PB) or bone marrow (BM) cells from 9 of 12 chronic phase CML patients were well engrafted with human cells including multilineage colony-forming progenitors and CD34+ cells for at least 90 days posttransplantation. Repeated posttransplant injections of cytokines did not enhance the number of engrafted human cells. Interestingly, approximately 70% of the human progenitors found in the engrafted SCID BM were Ph-, suggesting that the growth of primitive normal cells is favored in this in vivo transplant model. A similar number of normal cells were found in mice transplanted with either PB or BM cells, suggesting that elevated numbers of primitive normal cells are present in CML PB. When cells from patients with CML in either myeloid or lymphoid blast crisis were transplanted into SCID mice, the BM of these mice was more rapidly repopulated and to a higher level than that observed with transplants of chronic phase cells. Moreover, all human colony-forming progenitors present in the BM of mice transplanted with blast crisis cells were Ph+, and the majority of cells showed the same morphological features of the blast crisis cells originally transplanted. These experiments provide a starting point for the creation of an animal model of CML and establish the feasibility of using this model for the future characterization of transplantable CML stem cells during disease progression.


Subject(s)
Bone Marrow/pathology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Neoplastic Stem Cells/pathology , Animals , Antigens, CD19/analysis , Antigens, CD34/analysis , Base Sequence , Blast Crisis , Chronic Disease , DNA Primers/chemistry , Fusion Proteins, bcr-abl/genetics , Genes, abl , Humans , Mice , Mice, SCID , Molecular Sequence Data , Neoplasm Transplantation , Radiation Chimera
12.
Blood ; 84(5): 1534-42, 1994 Sep 01.
Article in English | MEDLINE | ID: mdl-8068944

ABSTRACT

In the present study, we investigated the ability of the tetrapeptide NAc-Ser-Asp-Lys-Pro-OH (AcSDKP), a reported inhibitor of primitive hematopoietic cells, to influence the proliferative behavior of primitive normal and chronic myeloid leukemia (CML) progenitor cells in the adherent layer of long-term cultures (LTCs). Addition of > or = 50 ng/mL of AcSDKP to LTCs of normal cells at the time of the regular weekly half-medium change selectively and reversibly decreased the proportion of high proliferative potential erythroid and granulopoietic progenitors in the adherent layer that were in S-phase without changing their numbers, but had no effect on either the cycling activity or number of analogous (neoplastic) cells in the adherent layer of CML LTCs. Specificity of the effect of AcSDKP on primitive normal progenitors was demonstrated by the finding that a similar addition of either the control peptide, AcSDKE, or 100 ng/mL of tumor necrosis factor-alpha (TNF-alpha, which contains the SDKP sequence), or SDKP itself (at 300 ng/mL) did not inhibit the proliferation of primitive normal progenitors in LTC adherent layers. Incorporation of > or = 30 ng/mL of AcSDKP (but not the related control peptide, AcSDKE) directly into methylcellulose cultures of normal marrow cells resulted in a dose-dependent suppression of colony formation, which was not seen in similar studies with CML marrow or after removal of adherent cells from normal marrow. Additional experiments showed that the inhibitory effect of AcSDKP on primitive normal progenitor cycling in the LTC system could be overcome by the simultaneous addition of macrophage inflammatory protein-1 beta (MIP-1 beta); an antagonist of MIP-1 alpha. The apparent differential effect of AcSDKP on primitive normal and CML progenitors may thus be a secondary consequence of the differential responsiveness of these cells to MIP-1 alpha for another molecule antagonized by MIP-1 beta), whose production or release by adherent marrow cells is inducible by AcSDKP. Such a mechanism may offer a method for obtaining localized increases in vivo of cytokines like MIP-1 alpha, suggesting novel and perhaps less toxic strategies for protecting primitive normal progenitors during repeated treatments with cycle-active chemotherapeutic agents where escalating the dose of drug given would be desirable.


Subject(s)
Cell Cycle/drug effects , Hematopoietic Stem Cells/drug effects , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Oligopeptides/pharmacology , Amino Acid Sequence , Bone Marrow/pathology , Bone Marrow Cells , Cell Adhesion , Chemokine CCL4 , Colony-Forming Units Assay , Culture Techniques/methods , Cytokines/pharmacology , Dose-Response Relationship, Drug , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/pathology , Humans , Macrophage Inflammatory Proteins , Molecular Sequence Data , Monokines/pharmacology , Recombinant Proteins/pharmacology , Reference Values , S Phase/drug effects , Thymidine/metabolism , Time Factors
13.
Baillieres Clin Haematol ; 7(1): 49-63, 1994 Mar.
Article in English | MEDLINE | ID: mdl-8038499

ABSTRACT

In long-term cultures (LTC) of human haematopoietic cells, primitive progenitors termed LTC-initiating cells can be maintained for several months and will differentiate to produce clonogenic cells and mature granulocytes and macrophages when provided with a supportive feeder layer of adherent mesenchymal cells. Primitive haematopoietic cells become associated with this feeder layer and their proliferative status and differentiation are regulated by their interaction with these feeder cells and the growth factors they produce. Both positive and negative regulators are generated in LTC and the balance between these diverse factors is readily manipulated by both direct and indirect mechanisms which appear to operate in a localized fashion. These features parallel those believed to characterize the mechanisms that regulate haematopoiesis in the bone marrow microenvironment in vivo and suggest that further analysis of the LTC system will be useful in delineating the full mystery of this process.


Subject(s)
Cytokines/physiology , Hematopoiesis , Cell Division , Cells, Cultured , Hematopoietic Stem Cells/cytology , Humans
15.
Proc Natl Acad Sci U S A ; 90(24): 12015-9, 1993 Dec 15.
Article in English | MEDLINE | ID: mdl-8265663

ABSTRACT

Most primitive hematopoietic cells appear to be normally quiescent in vivo, whereas their leukemic counterparts in patients with chronic myeloid leukemia (CML) are maintained in a state of rapid turnover. This difference is also seen in the long-term culture system, where control of primitive hematopoietic progenitor proliferation is mediated by interactions of these cells with marrow-derived mesenchymal cells of the fibroblast lineage. We now show that exogenous addition of macrophage inflammatory protein 1 alpha (MIP-1 alpha) to normal long-term cultures can reversibly and specifically block the activation of "primitive" (high proliferative potential), but not "mature" (lower proliferative potential), progenitors in the adherent layer of these cultures. Moreover, addition of MIP-1 beta after primitive-progenitor activation can prevent the subsequent return of these cells to a quiescent state a few days later as shown previously in similar experiments using antibodies to transforming growth factor beta. This suggests that the level of MIP-1 alpha (or a related MIP-1 alpha agonist) produced in LTCs, like the level of transforming growth factor beta, may be necessary, but is not on its own sufficient, to mediate the inhibitory activity of the regulatory cells in the adherent layer. Addition of MIP-1 alpha to similar long-term cultures containing normal marrow adherent layers but supporting exclusively neoplastic (CML) hematopoiesis did not block the cycling of primitive neoplastic progenitors. A defect in the responsiveness of CML cells to MIP-1 alpha (or a similarly acting chemokine) would explain their deregulated proliferative behavior in this model and, by extrapolation to the in vivo setting, suggests a molecular mechanism whereby the leukemic clone may become amplified at the stem-cell level. In addition, these findings suggest approaches to the therapy of CML, using inhibitors such as MIP-1 alpha for the protection of primitive normal cells.


Subject(s)
Cytokines/pharmacology , Hematopoietic Stem Cells/cytology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Monokines/pharmacology , Recombinant Proteins/pharmacology , Animals , Bone Marrow Cells , CHO Cells , Cells, Cultured , Chemokine CCL3 , Chemokine CCL4 , Cricetinae , Cytokines/biosynthesis , DNA/biosynthesis , DNA/drug effects , DNA, Neoplasm/biosynthesis , DNA, Neoplasm/drug effects , Hematopoietic Stem Cells/drug effects , Kinetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/physiopathology , Macrophage Inflammatory Proteins , Monokines/biosynthesis , Recombinant Proteins/biosynthesis , Thymidine/metabolism , Time Factors , Transfection , Tumor Cells, Cultured
16.
Stem Cells ; 11 Suppl 3: 109-21, 1993 Oct.
Article in English | MEDLINE | ID: mdl-7905319

ABSTRACT

Recent developments have occurred in the identification and quantitation of a very primitive type of hematopoietic cell (referred to as a long-term culture initiating cell or LTC-IC) using defined long-term culture conditions. These have facilitated investigations of the numbers and properties of both normal and leukemic LTC-IC in patients with chronic myeloid leukemia (CML). Such studies have revealed that the marrow of many chronic phase patients contains a substantial population of normal LTC-IC that are functionally intact albeit suppressed in vivo. Leukemic LTC-IC are typically less numerous in the marrow of these patients but are found in elevated numbers in the peripheral blood which, on average, in total contains more leukemic LTC-IC than the marrow when the WBC count rises above 10(11) per liter. However, a very marked heterogeneity in all of these parameters exists among individual patients. Some of the properties of leukemic LTC-IC are indistinguishable from those of their normal counterparts. Others, particularly those typically associated with an activated state, are altered, although frequently in only 90% (or less) of the leukemic LTC-IC. A more marked disparity between primitive normal and leukemic LTC-IC is seen in terms of their relative abilities to maintain their numbers in vitro. At the level of primitive clonogenic cells, the leukemic population has been shown to exhibit an increased rate of turnover. This appears to be due to an inability of these cells to respond to the cytostatic effects of macrophage inflammatory protein-1 alpha (MIP-1 alpha). These findings provide new insights into the biology of CML and highlight the power of quantitative assays to guide the development of more generally applicable curative therapies.


Subject(s)
Hematopoietic Stem Cells/pathology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Hematopoietic Stem Cell Transplantation , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy , Leukocyte Count
17.
Leukemia ; 6(9): 886-92, 1992 Sep.
Article in English | MEDLINE | ID: mdl-1518302

ABSTRACT

Experiments were undertaken to investigate the molecular basis of primitive hematopoietic progenitor cell regulation in both the long-term culture system and in methylcellulose, particularly with a view to characterizing factors either able or unable to influence the behaviour of primitive leukemic cells from patients with chronic myeloid leukemia (CML). Long-term cultures of CML cells with or without irradiated normal marrow feeder layers were initiated from peripheral blood cells of CML patients with high white blood cell counts. Three weeks later the effect of exogenously added transforming growth factor-beta 1 (TGF-beta 1) on progenitor cycling status was examined. A single addition of 5 ng/ml TGF-beta 1 was able to reversibly arrest the otherwise uninterrupted turnover of primitive leukemic erythroid and granulopoietic progenitors for a period of up to 7 days both in the presence and absence of a normal adherent cell population. When TGF-beta 1 was incorporated into methylcellulose cultures, its ability to inhibit colony formation by CML progenitors showed the same differential activity on primitive cell types exhibited by normal progenitors. Dose-response curves for analogous populations of normal and leukemic cells were indistinguishable. Increasing the concentration of granulocyte-macrophage colony-stimulating factor (GM-CSF) in methylcellulose colony assays decreased the sensitivity displayed by normal clonogenic cells to TGF-beta 1 and no differences were detectable when CML cells were used in such regulator competition experiments. These findings support a general model of primitive hematopoietic cell regulation in which entry into S-phase is determined at the intracellular level by multiple convergent pathways that may deliver either positive or negative signals from activated cell surface receptors for distinct extracellular factors. The present study shows for the first time that primitive CML progenitors exposed to TGF-beta 1 in vitro can be transiently blocked in a noncycling state for several days without loss of viability and that the mechanisms responsible for the emergence and maintenance of a clonal population of CML cells in vivo do not appear to involve changes in their sensitivity to TGF-beta 1. It is thus unlikely that the heightened proliferative activity exhibited by primitive CML progenitors both in vivo and in long-term culture can be explained by an abnormality in the intracellular mechanisms normally activated by TGF-beta 1 receptor-ligand binding. We suggest that primitive CML cells are either defective in their ability to see (or activate) endogenously produced TGF-beta 1, or are defective in their responsiveness to another, undefined, regulator.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cells/pathology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Transforming Growth Factor beta/pharmacology , Cell Cycle , Cell Division , Cells, Cultured , Dose-Response Relationship, Drug , Drug Interactions , Hematopoietic Stem Cells/cytology , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/blood , Tumor Cells, Cultured/pathology
18.
Br J Haematol ; 82(1): 50-7, 1992 Sep.
Article in English | MEDLINE | ID: mdl-1419802

ABSTRACT

Essential thrombocytosis (ET) is currently diagnosed by histopathologic assessment of the marrow after exclusion of a secondary cause or another myeloproliferative disorder. To evaluate the potential of more direct diagnostic methods, we compared the frequency and association of several abnormal features characteristic of neoplastic precursors in 32 patients presenting with platelet counts > 500 x 10(9)/l. Assays for erythropoietin (Ep)-independent erythroid progenitors were performed on all patients, determination of the cycling status of circulating progenitors on 27, and assessment of granulocyte clonality on 15. In most, but not all, patients deregulated progenitor turnover. Ep-independent progenitors and clonal granulocytes were concordant findings. The presence of polyclonal granulocytes and lack of evidence of abnormalities in Ep-dependence or progenitor cycling were also concordant findings in most, but not all patients. Thus, normal (i.e. polyclonal) granulocytes may be produced in occasional patients in spite of the presence of a neoplastic clone. Interestingly, one third of patients thought to have ET on the basis of blood and marrow histopathology showed no abnormalities previously associated with neoplastic progenitors. These findings suggest variability in dominance of the neoplastic clone in some ET patients and the potential utility of a multifaceted laboratory approach to investigate the underlying pathology in patients with thrombocytosis.


Subject(s)
Bone Marrow/pathology , Neoplastic Stem Cells/pathology , Thrombocytosis/pathology , Adult , Aged , Aged, 80 and over , Cells, Cultured , Erythropoietin/physiology , Female , Granulocytes/pathology , Humans , Male , Middle Aged , Thrombocytosis/diagnosis
19.
Med J Aust ; 155(9): 618-23, 1991 Nov 04.
Article in English | MEDLINE | ID: mdl-1943961

ABSTRACT

OBJECTIVE: To determine the extent of use and mortality associated with peripheral intravenous injections of radiocontrast media. DESIGN: A retrospective study of injection data was made for the three and a half year period from January 1987 to June 1990 using the Health Insurance Commission database and the records of public hospital x-ray departments. Information about deaths associated with the injections was obtained from a survey of all radiologists and from other relevant sources. SETTING AND PARTICIPANTS: The study related to the entire population of New South Wales and the Australian Capital Territory, approximately 6 million people. INTERVENTIONS: Intravenous injections of radiographic contrast medium for computed tomographic scans, intravenous pyelograms and venograms. MAIN OUTCOME: A comprehensive record of intravenous contrast usage and associated mortality in a large community. RESULTS: Between January 1987 and June 1990, 613 581 intravenous injections of radiocontrast media were administered in New South Wales and the Australian Capital Territory. The overall annual incidence of use was estimated to be 2.9% and was markedly age dependent being more than 7% in subjects over 65 years. Eight deaths were documented, representing an overall mortality of 13 per million injections (95% confidence interval [CI], 5.6-25.7). Mortality appeared to be age related being 35 per million (95% CI, 12.7-75.6) in those over 65 years compared with 4.5 per million (95% CI, 0.6-16.4) in those under 65 years. Two of the deaths involved low osmolar contrast media. CONCLUSIONS: Death after injection of intravenous contrast medium is a rare event. There was no evidence that mortality was lower with the newer, low osmolar media than with the older, high osmolar media.


Subject(s)
Contrast Media/adverse effects , Drug Hypersensitivity/mortality , Adverse Drug Reaction Reporting Systems , Age Factors , Aged , Aged, 80 and over , Contrast Media/administration & dosage , Female , Hospitals, Public , Humans , Injections, Intravenous , Male , Middle Aged , New South Wales , Northern Territory , Osmolar Concentration , Phlebography , Private Practice , Retrospective Studies , Tomography, X-Ray Computed , Urography
20.
Blood ; 78(1): 110-7, 1991 Jul 01.
Article in English | MEDLINE | ID: mdl-1712638

ABSTRACT

Numerous factors that can influence the proliferation and differentiation in vitro of cells at various stages of hematopoiesis have been identified, but the mechanisms used by stromal cells to regulate the cycling status of the most primitive human hematopoietic cells are still poorly understood. Previous studies of long-term cultures (LTC) of human marrow have suggested that cytokine-induced variations in stromal cell production of one or more stimulators and inhibitors of hematopoiesis may be important. To identify the specific regulators involved, we performed Northern analyses on RNA extracted from human marrow LTC adherent layers, or stromal cell types derived from or related to those present in the adherent layer. These analyses showed marked increases in interleukin-1 beta (IL-1 beta), IL-6, and granulocyte colony-stimulating factor (G-CSF) mRNA levels within 8 hours after treatments that lead to the activation within 2 days of primitive hematopoietic progenitors in such cultures. Increases in granulocyte-macrophage (GM)-CSF and M-CSF mRNA were also sometimes seen. Bioassays using cell lines responsive to G-CSF, GM-CSF, and IL-6 showed significant elevation in growth factor levels 24 hours after IL-1 beta stimulation. Neither IL-3 nor IL-4 mRNA was detectable at any time. In contrast, transforming growth factor-beta (TGF-beta) mRNA and nanogram levels of TGF-beta bioactivity in the medium were detected at all times in established LTC, and these levels were not consistently altered by any of the manipulations that stimulated hematopoietic growth factor production and primitive progenitor cycling. We also found that addition of anti-TGF-beta antibody could prolong or reactivate primitive progenitor proliferation when added to previously stimulated or quiescent cultures, respectively. Together, these results indicate a dominant negative regulatory role of endogenously produced TGF-beta in unperturbed LTC, with activation of primitive hematopoietic cells being achieved by mechanisms that stimulate stromal cells to produce G-CSF, GM-CSF, and IL-6. Given the similarities between the LTC system and the marrow microenvironment, it seems likely that the control of human stem cell activation in vivo may involve similar variations in the production of these factors by stromal cells.


Subject(s)
Bone Marrow Cells , Cytokines/physiology , Hematopoietic System/cytology , Blotting, Northern , Bone Marrow/metabolism , Bone Marrow/physiology , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cell Cycle/drug effects , Cell Cycle/physiology , Cells, Cultured , Granulocyte Colony-Stimulating Factor/genetics , Granulocyte Colony-Stimulating Factor/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Hematopoietic System/drug effects , Hematopoietic System/metabolism , Humans , Interleukin-1/genetics , Interleukin-1/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Time Factors , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/immunology , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...