Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Perfusion ; 30(4): 302-4, 2015 May.
Article in English | MEDLINE | ID: mdl-25106413

ABSTRACT

Electrical storm refers to a state of cardiac electrical instability characterized by multiple episodes of ventricular tachycardia (VT) or ventricular fibrillation (VF) within a relatively short period of time and is associated with increased mortality and morbidity. The management of electrical storm involves a variety of strategies, including sedation, anti-arrhythmic and electrolyte replacement as well as revascularization and electrical ablation. However, the management strategy in patients with refractory storm is less clear and may require more invasive approaches. We present a case of severe ventricular tachycardia storm refractory to conservative management that was managed with a HeartMate II left ventricular assist device.


Subject(s)
Heart-Assist Devices , Tachycardia, Ventricular/physiopathology , Tachycardia, Ventricular/surgery , Ventricular Fibrillation/physiopathology , Ventricular Fibrillation/surgery , Humans , Male , Middle Aged
2.
Bone Marrow Transplant ; 25 Suppl 2: S105-9, 2000 May.
Article in English | MEDLINE | ID: mdl-10933201

ABSTRACT

This report demonstrates that expression of the P140A O6-methylguanine DNA methyl transferase (MGMT) mutant via retrovirus-mediated gene transfer leads to significant, but modest, resistance of cells to both 6-benzylguanine (6-BG) depletion and treatment with 1,3-bis (2-chloroethyl)-1-nitrosourea (BCNU). Expression of the P140A/G156A double mutant appeared to be associated with reduced or unstable protein in hematopoietic cells.


Subject(s)
Carmustine/adverse effects , Genetic Therapy/methods , Guanine/analogs & derivatives , Hematopoietic Stem Cells/drug effects , O(6)-Methylguanine-DNA Methyltransferase/genetics , Animals , Antineoplastic Agents/adverse effects , DNA Repair/genetics , Drug Resistance/genetics , Gene Expression , Guanine/adverse effects , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/metabolism , Leukemia L1210/genetics , Leukemia L1210/metabolism , Leukemia L1210/therapy , Mice , Mutation , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Retroviridae/genetics , Transduction, Genetic
3.
J Pharmacol Exp Ther ; 290(3): 1467-74, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10454526

ABSTRACT

O(6)-Benzylguanine (6-BG) inactivates mammalian O(6)-methylguanine DNA methyltransferase (MGMT), an important DNA repair protein that protects cells against chloroethylnitrosourea (CENU) cytotoxicity. 6-BG is being tested as an approach to treat CENU-resistant tumors that overexpress endogenous MGMT. However, in addition to restoring CENU tumor cell sensitivity, 6-BG also increases the cytotoxic effects of CENUs on hematopoietic cells. Several 6-BG-resistant human MGMT mutants have been characterized in Escherichia coli and are predicted to protect mammalian cells against the combination of 6-BG and CENU treatment in vivo. Two mutants, P140A and P140A/G156A, demonstrated 20- and 1200-fold more resistance to 6-BG depletion of MGMT activity compared with wild-type MGMT (WTMGMT). Here, we analyzed retroviral vectors that express either WTMGMT, the P140A or P140A/G156A mutant forms of MGMT. Retroviral-infected L1210 hematopoietic cells demonstrated similar levels of RNA in all transduced clones. However, the amount of MGMT protein and DNA repair activity was reduced in clones expressing the P140A/G156A mutant compared with those expressing WTMGMT or P140A. Expression of P140A was associated with a 4- to 8-fold increase in resistance to 6-BG depletion of MGMT in transduced L1210 clones and a 1, 3-bis(2-chloroethyl)-1-nitrosourea IC(50) of 50 microM (compared with 27.5 microM for WTMGMT) in primary murine hematopoietic cells. These results demonstrate the utility of screening 6-BG-resistant MGMT proteins in hematopoietic cells and provide evidence that the P140A mutant form of MGMT generates 6-BG- and CENU-resistant hematopoietic cells. Retrovirus vectors expressing this mutant may be useful in future human gene therapy trials.


Subject(s)
Antineoplastic Agents/toxicity , Ethylnitrosourea/analogs & derivatives , Guanine/analogs & derivatives , Hematopoietic Stem Cells/enzymology , O(6)-Methylguanine-DNA Methyltransferase/biosynthesis , Animals , Carcinoma , Ethylnitrosourea/toxicity , Female , Genetic Vectors , Guanine/pharmacology , Hematopoietic Stem Cells/drug effects , Humans , Leukemia L1210 , Melanoma, Experimental , Mice , Mice, Inbred C57BL , Mutation , O(6)-Methylguanine-DNA Methyltransferase/genetics , O(6)-Methylguanine-DNA Methyltransferase/metabolism , O(6)-Methylguanine-DNA Methyltransferase/physiology , Retroviridae/genetics , Retroviridae/metabolism , Tumor Cells, Cultured
4.
Mol Med Today ; 3(8): 350-8, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9269688

ABSTRACT

An attractive approach to circumvent chemotherapy-induced myelosuppression is the use of gene-transfer technology to introduce new genetic material into hematopoietic cells. Several pre-clinical studies have demonstrated that increasing the expression of genes encoding proteins that modulate drug resistance in hematopoietic cells provides significant protection against chemotherapy-induced myelosuppression both in vitro and in vivo. Most work in this area has focused on the use of recombinant retroviruses as vectors for the delivery of DNA sequences into hematopoietic stem cells and progenitor cells. Based on these studies, clinical trials are now under way to evaluate the potential use of two gene sequences-multidrug resistance gene 1 and O6-methylguanine DNA methyltransferase. Reducing chemotherapy-induced myelosuppression by increasing the expression of genes that modulate drug resistance via gene transfer into bone marrow cells might allow dose-intensification of chemotherapy, which might result in an improvement in the clinical outcome of patients with high-risk tumors.


Subject(s)
Drug Resistance, Neoplasm/genetics , Hematopoietic Stem Cells/drug effects , Aldehyde Dehydrogenase/genetics , Animals , Drug Resistance, Multiple/genetics , Drug Resistance, Neoplasm/physiology , Fibronectins/genetics , Gene Transfer Techniques/trends , Genetic Therapy/adverse effects , Genetic Therapy/methods , Glutathione Transferase/genetics , Humans , Methyltransferases/genetics , Mice , Tetrahydrofolate Dehydrogenase/genetics
5.
J Pediatr Surg ; 32(2): 312-5, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9044143

ABSTRACT

PURPOSE: Major burns are associated with a high mortality, an increased rate of bacterial translocation, and bone marrow suppression. This study evaluates the effect of interleukin-11 (IL-11), a bone marrow-derived growth factor on survival, intestinal cytoarchitecture, bacterial translocation, and bone marrow suppression in a highly lethal murine burn model with a lethal dose greater than 50. METHODS: C3H/HeJ 8 to 10-week-old mice underwent a standardized 32% total body surface area (TBSA) scald burn using a burn template. Mice were divided equally between groups receiving IL-11 (125 micrograms/kg, twice daily, subcutaneously [SC]) and 0.1% same-volume Bovine Serum Albumin (BSA) (0.2 mL, twice daily, sc). Animals were evaluated for mesenteric lymph node bacterial counts, intestinal mucosal villus height, number of mucosal crypt cell mitoses per 100 crypts, and peripheral platelet and total lymphocyte counts. Survival was calculated to 7 days postburn. RESULTS: At 24 hours postburn, IL-11-treated mice had significantly less enteric bacteria cultured from mesenteric lymph nodes (P < .001), increased intestinal crypt cell mitoses (P = .002) and intestinal villus height (P = .002), increased peripheral platelet (P = .002) and lymphocyte counts (P = .004), and an improved survival compared with BSA controls (P = .003). CONCLUSION: These data show that IL-11 improves survival, intestinal cytoarchitecture, reduces bacterial translocation, and reduces bone marrow suppression after a 32% TBSA burn in mice. These data imply that IL-11 cytokine therapy may be a useful adjunct in extensive burn injury.


Subject(s)
Bacterial Translocation/drug effects , Bone Marrow/drug effects , Burns/therapy , Interleukin-11/therapeutic use , Animals , Burns/blood , Burns/mortality , Disease Models, Animal , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Leukocyte Count , Lymph Nodes/microbiology , Male , Mice , Mice, Inbred C3H , Mice, Inbred Strains , Platelet Count
6.
J Immunol ; 158(2): 1006-13, 1997 Jan 15.
Article in English | MEDLINE | ID: mdl-8993023

ABSTRACT

Bone marrow toxicity is a dose-limiting side effect of chloroethylnitrosourea (CNU) chemotherapeutic alkylating agents. A major determinant of CNU cytotoxicity is the methylation of guanine at the O6-position and the subsequent formation of interstrand DNA cross-links. O6-Methylguanine DNA methyltransferase (MGMT) removes alkyl groups from the O6 position of guanine and has been shown to repair CNU-induced DNA damage. We have previously demonstrated that transplantation of murine bone marrow cells transduced with a recombinant retroviral vector expressing MGMT via the human phosphoglycerate kinase promoter (PGK-MGMT) protects animals in vivo from acute myelotoxicity associated with CNU treatment. In the present study, we examined the effects of 1,3-bis(2-chloroethyl)-1-nitrosourea (BCNU), a commonly used CNU, on long term recovery of the lymphoid compartment, including thymus reconstitution, peripheral T and B cell populations, and lymphocyte mitogen responses in mice reconstituted with PGK-MGMT-transduced hemopoietic cells. Mice transplanted with either mock-infected control or PGK-MGMT-transduced stem cells were treated with five weekly doses of BCNU. Analysis of the lymphoid compartment demonstrated significant damage 3 mo after the last BCNU dose in control animals. In contrast, the profound deficiency in CD4+CD8+ double-positive thymocytes and mature lymphocytes observed in control mice surviving BCNU treatment was completely reversed in mice transplanted with PGK-MGMT-transduced bone marrow and was associated with molecular evidence of in vivo selection of transduced cells in the lymphoid compartment. Thus, long term immunodeficiency following CNU therapy may be prevented by genetic modification of murine hemopoietic stem cells with MGMT, leading to significant improvement in post-transplant immune function.


Subject(s)
Carmustine/toxicity , DNA, Complementary/therapeutic use , Hematopoietic Stem Cells/enzymology , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/therapy , Methyltransferases/therapeutic use , Animals , Bone Marrow Transplantation , Immunologic Deficiency Syndromes/chemically induced , Mice , Mice, Inbred C57BL , Mice, Transgenic , O(6)-Methylguanine-DNA Methyltransferase , Transfection
7.
Proc Natl Acad Sci U S A ; 93(1): 206-10, 1996 Jan 09.
Article in English | MEDLINE | ID: mdl-8552605

ABSTRACT

The chloroethylnitrosourea (CNU) alkylating agents are commonly used for cancer chemotherapy, but their usefulness is limited by severe bone marrow toxicity that causes the cumulative depletion of all hematopoietic lineages (pancytopenia). Bone marrow CNU sensitivity is probably due to the inefficient repair of CNU-induced DNA damage; relative to other tissues, bone marrow cells express extremely low levels of the O6-methylguanine DNA methyltransferase (MGMT) protein that repairs cytotoxic O6-chloroethylguanine DNA lesions. Using a simplified recombinant retroviral vector expressing the human MGMT gene under control of the phosphoglycerate kinase promoter (PGK-MGMT) we increased the capacity of murine bone marrow-derived cells to repair CNU-induced DNA damage. Stable reconstitution of mouse bone marrow with genetically modified, MGMT-expressing hematopoietic stem cells conferred considerable resistance to the cytotoxic effects of 1,3-bis(2-chloroethyl)-1-nitrosourea (BCNU), a CNU commonly used for chemotherapy. Bone marrow harvested from mice transplanted with PGK-MGMT-transduced cells showed extensive in vitro BCNU resistance. Moreover, MGMT expression in mouse bone marrow conferred in vivo resistance to BCNU-induced pancytopenia and significantly reduced BCNU-induced mortality due to bone marrow hypoplasia. These data demonstrate that increased DNA alkylation repair in primitive hematopoietic stem cells confers multilineage protection from the myelosuppressive effects of BCNU and suggest a possible approach to protecting cancer patients from CNU chemotherapy-related toxicity.


Subject(s)
Carmustine/toxicity , DNA Repair , Methyltransferases/administration & dosage , Animals , Bone Marrow/enzymology , Bone Marrow Cells , Bone Marrow Transplantation , Drug Resistance , Female , Gene Transfer Techniques , Humans , Methyltransferases/genetics , Mice , Mice, Inbred C57BL , O(6)-Methylguanine-DNA Methyltransferase , Survival Analysis
8.
Cancer Res ; 54(18): 4947-51, 1994 Sep 15.
Article in English | MEDLINE | ID: mdl-8069861

ABSTRACT

The chloroethylnitrosoureas, such as 1,3-bis(2-chloroethyl)-1-nitrosourea, are alkylating agents which are thought to exert antitumor activity by initiating lethal DNA interstrand cross-links. Although nitrosoureas are among the most active agents against childhood and adult gliomas, the utility of this class of agents has been limited by severe and cumulative myelosuppression, which can be fatal. Nitrosourea-induced myelosuppression in humans is delayed and may continue after withdrawal of the agent. We have developed a murine model which mimics the delayed and cumulative myelosuppression seen in humans receiving nitrosoureas. In this model, we demonstrate that interleukin-11, a stromal-derived hematopoietic growth factor with pleiotropic effects in a number of preclinical ablation models, markedly diminishes nitrosourea-induced pancytopenia and leads to a significant reduction in chemotherapy-related mortality. These data suggest that interleukin-11 could allow significant dose intensification in the treatment of tumors which are nitrosourea sensitive.


Subject(s)
Bone Marrow/drug effects , Carmustine/adverse effects , Hematopoietic Stem Cells/drug effects , Interleukin-11/pharmacology , Animals , Carmustine/antagonists & inhibitors , Female , Granulocytes/drug effects , Macrophages/drug effects , Mice , Mice, Inbred C57BL
9.
Blood ; 82(3): 1016-22, 1993 Aug 01.
Article in English | MEDLINE | ID: mdl-7687888

ABSTRACT

Molecular analysis of the hematopoietic microenvironment (HM) has led to the characterization and molecular cloning of two unique growth factors produced by stromal cells. Interleukin (IL)-11 and stem-cell factor (SCF; steel factor [SF]) have been shown in a variety of in vitro culture systems to stimulate distinct populations of stem, progenitor, and more differentiated cell types. We have analyzed and compared the effects of each growth factor administered to mice undergoing bone marrow transplantation (BMT) after total body irradiation (TBI). We report that IL-11 stimulates platelet and neutrophil recovery, while the main effect of SCF is on erythroid cell recovery in this model. Mice treated with the combination of IL-11 and SCF show increases in all three lineages compared with control mice, without obvious toxicity. In addition, both the type of progenitor- and stem-cell populations stimulated and the anatomic localization of effects seen with each growth factor are distinct. These data in mice suggest that the combination of IL-11 and SCF might be useful in humans undergoing myeloablative therapies.


Subject(s)
Bone Marrow Transplantation/methods , Hematopoiesis/drug effects , Hematopoietic Cell Growth Factors/therapeutic use , Interleukin-11/therapeutic use , Animals , Bone Marrow Cells , Hematopoietic Stem Cells/cytology , Leukocyte Count/drug effects , Male , Mice , Mice, Inbred C3H , Recombinant Proteins , Spleen/cytology , Stem Cell Factor , Time Factors
10.
Am J Trop Med Hyg ; 48(5): 716-25, 1993 May.
Article in English | MEDLINE | ID: mdl-8517491

ABSTRACT

Compartmental analysis of Strongloides stercoralis burdens in experimentally infected, serially necropsied dogs was used to test an autoinfective burst hypothesis. The hypothesis states that in well-established, active infections and in chronic infections as well, the rate of larval development is down-regulated so that most larvae do not attain infectivity internally. The majority pass in the feces as preinfective, rhabditiform larvae, but a few (those with the most rapid developmental rate) attain infectivity internally, and therefore are positioned for autoinfectivity. In contrast, in immunologically naive hosts, larval development proceeds without host hindrance and many larvae, proceeding at the most rapid rate of a spectrum of normal intrinsic developmental rates, attain infectivity internally. For a brief period, hyperinfection occurs, during which the adult worm population increases sharply. Gut-level resistance soon occurs, larval development is retarded, and an increasing proportion of larvae are discharged as preinfective rhabditiform larvae. With fewer larvae developing to infectivity internally, recruitment into the adult population decreases, with an attendant increase in the mean age and a gradual decrease in the size of the adult population. The data and the attendant model strongly support this autoinfective burst hypothesis.


Subject(s)
Strongyloides stercoralis/physiology , Strongyloidiasis/parasitology , Animals , Dogs , Down-Regulation , Feces/parasitology , Host-Parasite Interactions , Larva/physiology , Larva/ultrastructure , Models, Biological , Muscles/parasitology , Skin/parasitology , Strongyloides stercoralis/isolation & purification
11.
Exp Hematol ; 21(4): 502-7, 1993 Apr.
Article in English | MEDLINE | ID: mdl-8462659

ABSTRACT

The regulation of hematopoiesis has been suggested to take place in close association with various cell types found in the bone marrow (BM) microenvironment. In the present study the role of fibroblasts, adipocytes and cell surface heparan sulfate in regulating hematopoiesis in an in vitro mouse system was examined. Mouse BM cells were allowed to adhere to a mouse embryo fibroblast cell line (C3H 10T1/2) or a clonally derived adipogenically determined derivative (Clone D) of the 10T1/2 cell line. Nonadherent cells were removed, cultures were overlaid with semisolid media supplemented with growth factors and colony formation by granulocyte-macrophage (CFU-GM), erythroid (BFU-E) and multipotential (CFU-GEMM) progenitor cells was quantitated. Adherence and co-culture of BM cells with the fibroblast cell line resulted in increased numbers of total CFU-GM and CFU-GEMM colonies. In contrast, adherence and co-culture of BM cells with the adipocytic cell line resulted in an increase only in CFU-GEMM colonies. Morphological analysis revealed a preferential adherence/growth of granulocyte and macrophage progenitors at the expense of bipotent granulocyte-macrophage progenitors to the fibroblastic cell line and an increase in the adherence/growth of granulocyte progenitors to the adipogenic cell line. Progenitor cell adherence was abolished when the fibroblastic or adipocytic cell lines were pretreated with heparitinase. These results demonstrate enhanced proliferation/differentiation of hematopoietic progenitor cells when there is direct contact between hematopoietic progenitors and cell types characteristic of those found in the microenvironment and that heparan sulfate and different types of stromal cells appear to play different roles in this interaction.


Subject(s)
Adipose Tissue/cytology , Fibroblasts/cytology , Hematopoiesis/drug effects , Hematopoietic Stem Cells/cytology , Animals , Bone Marrow Cells , Cell Adhesion , Cell Differentiation/drug effects , Cell Division/drug effects , Cell Line , Colony-Forming Units Assay , Extracellular Matrix/physiology , Female , Heparitin Sulfate/pharmacology , Mice , Mice, Inbred C3H
12.
J Immunol ; 150(8 Pt 1): 3448-58, 1993 Apr 15.
Article in English | MEDLINE | ID: mdl-7682242

ABSTRACT

Macrophage inflammatory protein (MIP)-1 alpha, part of a family termed chemokines, has been implicated in suppression of hemopoietic stem and progenitor cell proliferation. The chemokine family has been organized into two subgroups with MIP-1 alpha, MIP-1 beta, macrophage chemotactic and activating factor (MCAF) and RANTES belonging to one subgroup, and GRO-alpha, MIP-2 alpha (GRO-beta), MIP-2 beta (GRO-gamma), platelet factor 4 (PF4), IL-8, and neutrophil activating peptide (NAP)-2 belonging to the other. These molecules were evaluated for effects on colony formation by human bone marrow multipotential (CFU-GEMM), erythroid (BFU-E) and granulocyte-macrophage (CFU-GM) progenitor cells. None of the chemokines stimulated colony formation in the absence of CSF, or influenced colony formation stimulated by a single growth factor such as granulocyte-macrophage-CSF or erythropoietin. However, MIP-1 alpha, MIP-2 alpha, PF4, IL-8, and MCAF suppressed in dose-response fashion colony formation of immature subsets of myeloid progenitor cells stimulated by GM-CSF plus steel factor. Effects were apparent on low density and CD34 HLA-DR(+)-sorted marrow cells in which up to 88.4% of the cells were composed of progenitor cells, suggesting direct effects on the progenitors themselves. Up to 2500-fold less of each chemokine could be used to demonstrate synergistic suppression when any two of these five chemokines were used together at low concentrations, effects also apparently directly on the progenitors. In contrast, MIP-1 beta, MIP-2 beta, GRO-alpha, NAP-2, and RANTES were not suppressive nor did they synergize with MIP-1 alpha, MIP-2 alpha, PF4, IL-8, or MCAF to suppress. However, a fivefold excess of MIP-1 beta blocked the suppressive effects of MIP-1 alpha. Similarly, a fivefold excess of either MIP-2 beta or GRO-alpha blocked the suppressive effects of IL-8 and PF4. These suppressing, synergizing and blocking effects may be of relevance to blood cell regulation.


Subject(s)
Bone Marrow/drug effects , Cytokines/pharmacology , Hematopoietic Stem Cells/drug effects , Monokines/pharmacology , Animals , Cell Division/drug effects , Cells, Cultured , Chemokine CCL4 , Chemokine CCL5 , Chemokine CXCL2 , Chemotactic Factors/pharmacology , Drug Interactions , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cells/cytology , Humans , Interleukin-8/pharmacology , Lymphokines/pharmacology , Macrophage Inflammatory Proteins , Mice , Monocyte Chemoattractant Proteins , Platelet Factor 4/pharmacology , Recombinant Proteins/pharmacology
13.
Exp Hematol ; 21(4): 545-51, 1993 Apr.
Article in English | MEDLINE | ID: mdl-7681784

ABSTRACT

Steel factor (SLF, c-kit ligand), a potent costimulating cytokine in vitro for myeloid progenitor cells from normal donors, is currently being evaluated in clinical trials for effects on hematopoiesis. Based on a preliminary observation that colony-stimulating factor (CSF)-responsive myeloid progenitor cells (CFU-GM) from a few patients with acute myeloid leukemia (AML) did not respond to the costimulating effects of SLF, we evaluated responsiveness of bone marrow or blood CFU-GM from 26 patients with either AML, chronic myeloid leukemia (CML) or myelodysplastic syndrome (MDS) to the effects in vitro of SLF and/or granulocyte-macrophage CSF (GM-CSF). Cells from all 26 patients responded to the stimulating effects of GM-CSF, but marked heterogeneity was detected in each disease category to the costimulating effects of SLF. Nine of 13 patients with AML, 2 of 6 patients with CML and 4 of 7 patients with MDS had clonogenic cells that did not respond significantly to the costimulating effects of SLF. In a more limited study of cells from patients with MDS, it was noted that if the CFU-GM of that patient did not respond to SLF enhancement of CSF-induced colony formation, neither did the erythropoietin (Epo)-dependent erythroid (BFU-E) or multipotential (CFU-GEMM) cells of that patient (3 cases of refractory anemia [RA] evaluating bone marrow and in 1 case blood progenitors as well). If CFU-GM responded, BFU-E and CFU-GEMM responded (bone marrow from 1 patient with chronic myelomonocytic leukemia [CMMol]). Clinical criteria did not readily distinguish between patients who had SLF-responsive vs. -nonresponsive clonogenic cells. While the mechanistic reason for this heterogeneity in responsiveness is not clear, these differences should be carefully considered for possible clinical trials with SLF in patients with acute and chronic myeloid leukemia and MDS.


Subject(s)
Hematopoiesis/drug effects , Hematopoietic Cell Growth Factors/pharmacology , Hematopoietic Stem Cells/drug effects , Leukemia, Myeloid/pathology , Myelodysplastic Syndromes/pathology , Adult , Aged , Cells, Cultured , Colony-Forming Units Assay , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , In Vitro Techniques , Male , Middle Aged , Recombinant Proteins/pharmacology , Stem Cell Factor
14.
J Immunol ; 149(3): 1004-9, 1992 Aug 01.
Article in English | MEDLINE | ID: mdl-1634758

ABSTRACT

Purified recombinant murine macrophage inflammatory protein-1 alpha (rmuMIP-1 alpha), a cytokine with myelopoietic activity in vitro, was assessed in vivo by injection into C3H/HeJ mice for effects on proliferation (percentage of cells in S phase DNA synthesis of the cell cycle) and absolute numbers of granulocyte-macrophage, erythroid, and multipotential progenitor cells in the femur and spleen, and on nucleated cellularity in the bone marrow, spleen, and blood. rmuMIP-1 alpha rapidly decreased cycling rates (at 2 to 10 micrograms/mouse i.v.) and absolute numbers (at 5 to 10 micrograms/mouse i.v.) of myeloid progenitor cells in the marrow and spleen. These effects were dose- and time-dependent and reversible. Suppressive effects were noted within 3 to 24 h for cell cycling and absolute numbers of progenitor cells in the marrow and spleen, and by 48 h for circulating neutrophils. A study comparing the effects of i.v. injection of rmuMIP-1 alpha versus rmuMIP-1 beta, a biochemically similar molecule but with no myelosuppressive effects in vitro, demonstrated myelosuppression in vivo by rmuMIP-1 alpha, but not by rmuMIP-1 beta. The results suggest that rmuMIP-1 alpha has myelosuppressive activity in vivo and offers the possibility that it may be a useful adjunct to treatments involving cytotoxic drugs because of its reversible suppressive effects on normal progenitor cell cycling.


Subject(s)
Cytokines/pharmacology , Hematopoiesis/drug effects , Monokines/pharmacology , Animals , Bone Marrow Cells , Cell Cycle/drug effects , Chemokine CCL4 , Cytokines/chemistry , Dose-Response Relationship, Drug , Hematopoietic Stem Cells/drug effects , Macromolecular Substances , Macrophage Inflammatory Proteins , Mice , Mice, Inbred C3H , Monokines/chemistry , Recombinant Proteins , Spleen/cytology
15.
Cancer Cells ; 3(12): 480-7, 1991 Dec.
Article in English | MEDLINE | ID: mdl-1726456

ABSTRACT

Mouse strains carrying mutations at the Dominant White Spotting (W) locus or the Steel (Sl) locus are anemic and display defects in pigmentation and gametogenesis. In W mutants the anemia is due to a deficiency of hemopoietic stem cells and, in Sl mutants, to a deficiency of supporting stromal cells in the bone marrow. The W locus encodes the c-kit proto-oncogene product, a cell surface receptor with protein-tyrosine kinase activity, and the Sl locus encodes its ligand, a hemopoietic cytokine known variously as Steel factor (SLF), mast cell growth factor, stem cell factor, and Kit ligand. SLF can synergize with a number of other cytokines to stimulate growth of hemopoietic progenitors in vitro and stimulates blood cell production in vivo in animals. Here we review the biological activities of SLF, with particular emphasis on its effects on hemopoietic stem and progenitor cells. We also discuss present knowledge of the molecules involved in SLF-triggered signal transduction, and speculate on potential therapeutic applications for SLF in human disease.


Subject(s)
Hematopoiesis/physiology , Hematopoietic Cell Growth Factors/physiology , Proto-Oncogene Proteins/physiology , Anemia/drug therapy , Anemia/genetics , Animals , Bone Marrow/embryology , Cell Differentiation , Cell Movement , Drug Evaluation, Preclinical , Gene Expression Regulation , Hematopoietic Cell Growth Factors/genetics , Hematopoietic Cell Growth Factors/isolation & purification , Hematopoietic Cell Growth Factors/pharmacology , Hematopoietic Cell Growth Factors/therapeutic use , Hematopoietic Stem Cells/drug effects , Humans , Leukemia/pathology , Melanocytes/cytology , Mice , Mice, Mutant Strains/embryology , Mice, Mutant Strains/genetics , Mice, Mutant Strains/physiology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Mas , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-kit , Proto-Oncogenes , Rats , Signal Transduction , Stem Cell Factor
16.
Leuk Res ; 15(8): 733-44, 1991.
Article in English | MEDLINE | ID: mdl-1895754

ABSTRACT

A B-lymphoblastoid cell line ESKOL, composed of differentiated cells resembling hairy-cell leukemia (HCL) has been established from the peripheral blood (PB) of a HCL patient. Morphologically, ESKOL cells share several features with HCL B cells. Flow cytometric analysis revealed that ESKOL cells express HC2, CD21, PCA-1, CD24, FMC7, and CD25. Analysis by Northern-blot hybridization indicated that cultured cells expressed the oncogenes c-myc, H-ras and c-fos. RNA from 3T3 cells transfected with ESKOL DNA hybridized with H-ras and c-fos DNA probes. The ESKOL cells cultured in the presence of increasing concentrations, of alpha interferon demonstrated a decrease in the rate of cellular growth and an increase in the expression of CD21, CD25, FMC7 and PCA-1. Scanning electron microscopy revealed that cells incubated in the presence of alpha interferon underwent membranous changes with a loss of villosity. These observations suggest that IFN tends to drive HC out of their developmental arrest towards maturation.


Subject(s)
B-Lymphocytes/pathology , Gene Expression Regulation, Leukemic/physiology , Leukemia, Hairy Cell/pathology , Aged , Animals , Antigens, Neoplasm/analysis , Antigens, Surface/analysis , Antigens, Viral/analysis , B-Lymphocytes/physiology , Cell Differentiation/drug effects , Cell Transformation, Neoplastic/genetics , DNA, Neoplasm/genetics , Epstein-Barr Virus Nuclear Antigens , Humans , Interferon alpha-2 , Interferon-alpha/pharmacology , Leukemia, Hairy Cell/drug therapy , Leukemia, Hairy Cell/physiopathology , Male , Recombinant Proteins , Tumor Cells, Cultured/immunology
SELECTION OF CITATIONS
SEARCH DETAIL