Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Arch Dermatol Res ; 293(3): 147-58, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11357229

ABSTRACT

Dendritic cells (DC) are highly specialized antigen-presenting cells located in many nonlymphoid tissues, and Langerhans cells (LC), a specialized form of DC, are found in the skin. LC as antigen-presenting cells play a critical role in the induction of allergic contact dermatitis. LC research is difficult because few LCs can be isolated from human skin, so efforts have focused on obtaining DCs from alternative sources. Mononuclear cells from peripheral blood and CD34+ stem cells from human cord blood and marrow can be induced to form phenotypic and functional DCs, but experiments of this type are expensive and the DC yield is low. We report here the induction of the myeloid leukemia cell line (KG-1) to a DC morphology and phenotype by culturing the cells in a defined cytokine cocktail. Morphologically, the KG-1-derived DCs are large irregularly shaped cells with prominent dendritic processes and hair-like cytoplasmic projections. Phenotypically, the KG-1-derived DCs lack lineage-specific markers, and express MHC class II, costimulatory molecules CD80 and CD86, and CD83. Functionally, KG-1-derived DCs are capable of phagocytosing latex microspheres and are able to induce a potent allogeneic T-cell response. Within the KG-1-derived DCs, a subpopulation maintains the DC phenotype and morphology described above but further develops CD1a+ marker expression similar to that of resident skin-derived LCs. These findings illustrate that phenotypic, morphologic and functional DCs can be derived from the KG-1 cell line.


Subject(s)
Antigens, CD1/analysis , Cytokines/pharmacology , Dendritic Cells/immunology , Leukemia, Myeloid, Acute/pathology , Dendritic Cells/physiology , Dendritic Cells/ultrastructure , Humans , Phenotype , Tumor Cells, Cultured
2.
Toxicol In Vitro ; 15(1): 43-55, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11259869

ABSTRACT

Allergic contact dermatitis (ACD) is a cell-mediated immune response to small molecular weight chemicals that contact and penetrate the skin. There are a variety of characteristics that determine whether a chemical can function as a contact sensitizer (or allergen) including the ability to penetrate into the skin, react with protein and be recognized as antigenic by immune cells. The ultimate challenge for developing non-animal test methods for skin sensitization testing will be applying our mechanistic understanding of ACD to the design of predictive in vitro alternative test methods. Specifically, the in vitro approach should be designed so that a chemical's potential to penetrate the skin, react with protein/peptide (biotransformation may be required) and initiate an antigen-specific immune response is incorporated in the test methods developed. In this review, we have focused on cellular-based assays that have been developed or proposed for assessing a chemical's skin sensitization potential in vitro. All of the promising leads to date are based on observations made from in vivo studies conducted in animals and humans, and therefore have a strong mechanistic foundation. However, it remains to be demonstrated whether a single in vitro test, or several in vitro tests in combination, which model the critical steps in sensitization, can replace animal experiments for predicting contact allergic reactions in humans. Regardless, the future looks promising with continued development of our understanding of the chemical and biological aspects of allergic contact dermatitis, and most importantly, with the application of genomics/proteomics to this field on the immediate horizon.


Subject(s)
Allergens/immunology , Animal Testing Alternatives , Dendritic Cells/immunology , Dermatitis, Contact/immunology , Langerhans Cells/immunology , Animals , Antibody Formation , Biological Assay/methods , Cell Culture Techniques/methods , Humans , Keratinocytes/immunology , Skin/immunology
3.
Eur J Cancer ; 29A(16): 2279-83, 1993.
Article in English | MEDLINE | ID: mdl-7509165

ABSTRACT

Cells from patients with acute myeloid leukaemia (AML) or chronic myeloid leukaemia (CML) were separated into CD34-enriched and CD34-depleted subpopulations. The clonogenic capacities of these two subpopulations were then compared to each other and to the original unseparated cell population. In every study, the CD34-enriched subpopulation demonstrated a substantial increase in clonogenicity in vitro in comparison with the original cell population, while the reverse was the case for the CD34-depleted subpopulations. For reasons not clear at present, the enrichment for clonogenic cells far exceeded the enrichment for cells expressing the CD34 antigen. Additionally, the clonogenic potential was found to be unrelated to the level of myc expression in the various cell populations.


Subject(s)
Antigens, CD/analysis , Leukemia, Myeloid/pathology , Antigens, CD34 , Cell Division , Cell Separation , Gene Expression , Genes, myc , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology , Leukemia, Myeloid/genetics , Leukemia, Myeloid/immunology , Tumor Stem Cell Assay
4.
Leuk Res ; 16(10): 1003-11, 1992 Oct.
Article in English | MEDLINE | ID: mdl-1405703

ABSTRACT

Monoclonal antibodies and flow cytometry were used to detect the expression of c-myc and c-myb in the bone marrow (BM) and peripheral blood (PB) cells of patients with acute myelogenous leukemia (AML). The expression of neither gene was correlated with the percent blast cells in the BM or PB nor was there a correlation between c-myc and c-myb expression. A wide range of expression of each gene was found within each FAB type of AML. Patients who had a high proportion of leukemia cells expressing c-myb were less likely to respond to remission induction therapy than patients in whom a low proportion of cells expressed c-myb. This association appears to reflect an inverse relationship between the proportion of cells expressing c-myb and the sensitivity of leukemia cells to the killing effects of chemotherapy in vivo. Treatment outcome was unrelated to c-myc expression.


Subject(s)
Leukemia, Myeloid, Acute/genetics , Proto-Oncogene Proteins c-myc/analysis , Proto-Oncogene Proteins/analysis , Bone Marrow/ultrastructure , Flow Cytometry , Gene Expression , Humans , Proto-Oncogene Proteins c-myb
5.
J Leukoc Biol ; 51(4): 336-42, 1992 Apr.
Article in English | MEDLINE | ID: mdl-1314278

ABSTRACT

The antiviral drug used in the treatment of acquired immunodeficiency syndrome, zidovudine, has proved effective in ameliorating the morbidity and mortality associated with human immunodeficiency virus infection. However, associated with zidovudine is the development of severe bone marrow toxicity manifested by anemia, neutropenia, and occasionally thrombocytopenia. We report the results of studies that demonstrate the ability of basic fibroblast growth factor (B-FGF) to reduce zidovudine toxicity to several classes of hematopoietic progenitors (granulocyte-macrophage, CFU-GM; megakaryocyte. CFU-Meg; and erythroid, BFU-E) from normal murine, human, and murine retrovirus-infected bone marrow cells when cocultured with zidovudine in vitro. Optimal response to B-FGF was observed at a dose concentration of 10 ng/ml. The specificity of B-FGF was demonstrated in the presence of protamine sulfate, an effective inhibitor of B-FGF mitogenic activity. In addition, synergistic activity of B-FGF on zidovudine-induced hematopoietic stem cell toxicity was observed in the presence of interleukin 1 (IL-1) (30 ng/ml). These studies demonstrate that B-FGF is capable of reducing the hematopoietic toxicity associated with zidovudine and that such an effect can be amplified in the presence of IL-1.


Subject(s)
Fibroblast Growth Factor 2/administration & dosage , Hematopoietic Stem Cells/drug effects , Interleukin-1/administration & dosage , Zidovudine/toxicity , Animals , Cells, Cultured , Drug Synergism , Humans , In Vitro Techniques , Mice , Mice, Inbred BALB C , Protamines/pharmacology , Rauscher Virus , Retroviridae Infections/pathology
6.
J Med ; 23(3-4): 195-216, 1992.
Article in English | MEDLINE | ID: mdl-1479300

ABSTRACT

Bone marrow transplantation (BMT) has become a widely used procedure in the treatment of numerous hematological and non-hematological clinical disorders. The preparative regimen used for marrow recipients is not without risk as the immunodeficient recipient is susceptible to life-threatening infections due to the inability of the marrow to engraft properly. The monovalent cation lithium has been demonstrated to influence regenerating hematopoiesis following the use of several agents known to suppress hematopoiesis. The following report summarizes our studies which have been designed to determine the rate of hematopoietic reconstitution in lethally irradiated mice that were transplanted with bone marrow cells, harvested from either syngeneic or allogeneic donor animals, treated with lithium or phosphate buffered saline (PBS). Transplanted recipients receiving marrow cells from lithium treated donors were evaluated for their survival, peripheral blood indices and several classes of hematopoietic progenitors (granulocyte, erythroid, and megakaryocyte). Transplanted animals that received marrow cells from either syngeneic or allogeneic donors treated with lithium demonstrated greater survival, increased recovery of peripheral indices and hematopoietic progenitors compared to PBS-treated controls. These results indicate that the use of lithium to treat the donor may be an effective procedure to enhance hematopoietic recovery and engraftment in the transplanted recipient. Because of its wide-ranging effects, the use of lithium as a single agent, may be more efficacious than administering several hematopoietic growth factors in order to achieve a similar response.


Subject(s)
Bone Marrow Transplantation , Hematopoiesis/drug effects , Lithium Carbonate/therapeutic use , Animals , Bone Marrow Transplantation/pathology , Colony-Forming Units Assay , Female , Male , Mice , Mice, Inbred AKR , Mice, Inbred C3H , Tissue Donors , Transplantation, Homologous , Transplantation, Isogeneic
7.
Cytometry ; 13(6): 653-8, 1992.
Article in English | MEDLINE | ID: mdl-1451597

ABSTRACT

This report describes a method for preserving the light scatter patterns of cells in which myc and myb expression are being measured. Exposure of cells to 1% paraformaldehyde for 72 h prior to antibody staining for myc and myb proteins preserved the light scatter patterns. Using this method, myc and myb expression was found to be highest in lymphocytes and monocytes and lowest in granulocytes. The measurement of differences in the level of expression of these genes in subpopulations of leukemia cells obtained from individual patients is possible as is assessment of the levels of expression amongst normal and leukemia cells present in the same patient.


Subject(s)
Gene Expression Regulation, Leukemic , Genes, myc , Leukemia, Myeloid, Acute/genetics , Oncogenes , Bone Marrow/pathology , Flow Cytometry , Humans , Leukemia, Myeloid, Acute/blood , Leukemia, Myeloid, Acute/pathology , Leukocytes, Mononuclear/chemistry , Lymphocytes/chemistry , Neoplastic Stem Cells/chemistry , Nephelometry and Turbidimetry , Neutrophils/chemistry , Proto-Oncogene Proteins/analysis , Proto-Oncogene Proteins c-myb , Proto-Oncogene Proteins c-myc/analysis
8.
Med Oncol Tumor Pharmacother ; 9(3): 139-47, 1992.
Article in English | MEDLINE | ID: mdl-1341325

ABSTRACT

To study the regulation of expression of the myc protooncogene, cells from normal individuals and patients with acute myelogenous leukemia (AML), and chronic phase and blastic crisis of chronic myeloid leukemia (CML) cells were put in overnight culture in the presence or absence of fetal calf serum. Myc expression in normal marrow cells and chronic phase CML cells fell after culture in vitro. In contrast, myc expression was maintained or increased in a majority of the AML and blastic crisis CML specimens. These data demonstrate that the regulation of myc expression is disordered in many AML and blastic crisis specimens but not in chronic phase CML cells.


Subject(s)
Blast Crisis/genetics , Gene Expression Regulation, Leukemic/genetics , Genes, myc/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Leukemia, Myeloid, Acute/genetics , Base Sequence , Blotting, Southern , Bone Marrow Cells , Cell Count , Gene Expression Regulation, Leukemic/drug effects , Genes, myc/drug effects , Genes, ras/genetics , Humans , Interferon alpha-2 , Interferon-alpha/pharmacology , Molecular Sequence Data , Mutation/genetics , RNA, Neoplasm/genetics , Recombinant Proteins , Tretinoin/pharmacology , Tumor Cells, Cultured
9.
J Leukoc Biol ; 50(6): 580-6, 1991 Dec.
Article in English | MEDLINE | ID: mdl-1940611

ABSTRACT

The drug 3'-azido-3'-deoxythymidine (AZT), a synthetic thymidine analogue, has been used clinically in the management of acquired immune deficiency syndrome (AIDS). The drug is an effective antiviral agent due to its ability to block reverse transcriptase activity. This action of AZT was demonstrated in the Rauscher leukemia virus (RLV)-induced murine erythroleukemia model system. Unfortunately, associated with AZT has been the development of hematopoietic toxicity manifested by anemia, neutropenia, and overall bone marrow suppression. Hematopoietic growth factors (GM-CSF, erythropoietin), cytokines (interleukin-1), and agents known to potentiate hematopoiesis (lithium) have been demonstrated to modulate drug and/or radiation-induced hematopoietic toxicity. We report the results of further studies designed to investigate the ability of GM-CSF, erythropoietin, interleukin-1, and lithium to modulate AZT toxicity on murine hematopoietic granulocyte-macrophage (CFU-GM), megakaryocytic (CFU-Meg), and erythroid (BFU-E) progenitors cultured from bone marrow and spleen cells from mice infected with RLV. Hematopoietic progenitors from either normal or RLV-infected animals when exposed to AZT demonstrated concentration-dependent toxicity and differed for each progenitor with BFU-E being the most sensitive (ID50 concentration, 5 x 10(-9) M) and CFU-GM the least sensitive (ID50 concentration, 5 x 10(-5) M). As has been previously demonstrated using normal murine hematopoietic progenitors, when cultured with RLV-infected marrow or spleen cells, addition of GM-CSF, Meg-CSF or erythropoietin failed to inhibit AZT toxicity in vitro on CFU-GM, CFU-Meg, and BFU-E, respectively. However, in the presence of interleukin-1 (recombinant human IL-1 alpha, 30 ngm) or lithium chloride (ultra-pure, 1.0 mM), AZT toxicity CFU-GM, CFU-Meg, and BFU-E cultured from RLV-infected marrow or spleen cells was reduced. These results further demonstrate interleukin-1 and lithium are effective in modulating the toxic action of AZT on hematopoietic progenitors and that RLV-infected animals serve as a useful viral model system to study the effect of agents capable of modulating hematopoiesis in the presence of the anti-viral drug AZT.


Subject(s)
Erythropoietin/pharmacology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Hematopoiesis/drug effects , Hematopoietic Stem Cells/drug effects , Interleukin-1/pharmacology , Lithium/pharmacology , Zidovudine/toxicity , Animals , Bone Marrow Cells , Colony-Forming Units Assay , Erythroid Precursor Cells/drug effects , Female , In Vitro Techniques , Leukemia, Experimental/pathology , Mice , Mice, Inbred BALB C , Rauscher Virus , Spleen/cytology
10.
Int J Cell Cloning ; 9(5): 474-90, 1991 Sep.
Article in English | MEDLINE | ID: mdl-1720155

ABSTRACT

A modified version of the original reported panning technique was used to separate CD34+ cells from the peripheral blood of patients with chronic myeloid leukemia (CML). In 13 out of 23 separations, populations of cells were obtained in which CD34+ cells constituted greater than 50% of the cells present. The best recovery and enrichment of the CD34+ cells was achieved when cells were obtained from patients in the accelerated phase of CML, when the cells were processed on the same day they were obtained from patients, and when adherence to soybean agglutinin flasks was used as a pre-enrichment step. In suspension culture, the CD34+ cells were capable of extensive proliferation and differentiation. In semi-solid culture, the number of colony-forming units (CFUs) directly correlated with CD34 positivity. The number of clonogenic cells/CD34+ cells was highest at the time of initial diagnosis of CML, fell during the chronic phase (CP) of the disease, and rose at the time of disease acceleration. This observation suggests that therapy during the CP of the disease produces a greater reduction in clonogenic cells than in the number of CD34+ cells. This effect disappears at the time of disease acceleration, presumably because of the development of drug resistance in the clonogenic cells.


Subject(s)
Antigens, CD/isolation & purification , Cell Separation/methods , Hematopoietic Stem Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/blood , Adult , Antigens, CD/metabolism , Antigens, CD34 , Culture Media, Serum-Free , Female , Flow Cytometry , Hematopoietic Stem Cells/immunology , Humans , Male , Middle Aged , Tumor Cells, Cultured
11.
Int J Cell Cloning ; 9(3): 220-32, 1991 May.
Article in English | MEDLINE | ID: mdl-2061622

ABSTRACT

Basic fibroblastic growth factor (B-FGF) is a hormone-like protein which belongs to a class of heparin-binding growth factors. B-FGF is synthesized and released to circulate in the blood where it can be recognized by target cells through specific high-affinity plasma membrane receptors. B-FGF is known to be a potent mitogen for a number of specific cell types. We report data which demonstrates B-FGF can influence noncommited and specific lineage-derived hematopoietic progenitors when incubated in vitro. When combined with adherent cell-depleted normal murine marrow cells, B-FGF increased the number of both day 9 and day 12 spleen colony-forming units (CFU-s) from lethally irradiated animals. However, day 12-derived CFU-s were more sensitive to B-FGF, since optimal CFU-s production was observed at 10 ng/ml vs. 100 ng/ml for day 9 CFU-s (p less than 0.05). In adherent cell-depleted murine and human marrow cultures, the addition of B-FGF possessed synergistic activity in combination with the optimal concentration of GM-CSF for CFU-gm at a dose of 10 ng/ml which was inhibited in the presence of protamine sulfate (LD50 dose, 100 mu gm/ml), an inhibitor of B-FGF mitogenic activity, or in the presence of heparin (LD50 dose, 100 U/ml), an effective B-FGF binding agent. B-FGF also expressed synergistic activity in the presence of optimal concentrations of erythropoietin and Meg-CSF for murine and human BFU-e, and murine CFU-meg. No in vitro colony formation was observed when cells were cultured in the presence of B-FGF, but in the absence of the specific hematopoietic growth factor. Finally, B-FGF was also shown to be an effective radioprotective agent in vitro. Murine and human CFU-gm exposed to increasing doses of radiation (0.5 to 5 Gy) combined with GM-CSF and increasing doses of B-FGF (0.1 to 100 ng/ml) produced less radiation-induced toxicity compared to cultures containing GM-CSF alone. This data demonstrates B-FGF influences early- and late-stage hematopoietic progenitors, possesses synergistic activity with hematopoietic growth factors, and is a radioprotective agent in vitro. These results suggest B-FGF must be considered as a member of the family of molecules capable of influencing hematopoiesis in vitro.


Subject(s)
Erythropoietin/pharmacology , Fibroblast Growth Factor 2/pharmacology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cells/drug effects , Proteins/pharmacology , Radiation-Protective Agents , Animals , Bone Marrow Cells , Cells, Cultured , Cesium Radioisotopes , Drug Synergism , GPI-Linked Proteins , Heparin/pharmacology , Humans , Male , Membrane Glycoproteins , Mesothelin , Mice , Mice, Inbred C3H , Protamines/pharmacology , Radiation-Protective Agents/pharmacology , Spleen/cytology
12.
Leuk Res ; 15(7): 603-8, 1991.
Article in English | MEDLINE | ID: mdl-1713638

ABSTRACT

The expression of progenitor cell associated antigen CD34 was investigated in cells from 28 patients with chronic myeloid leukemia (CML). The CD34 positivity varied from 0-26% in patients with chronic phases CML (n = 17); from 6-64% in patients with accelerated phase CML (n = 4); and from 27-97% in the patients with blastic crisis of CML (n = 8). The difference in CD34 positivity between chronic (mean 10.1 +/- 2.3%), accelerated (37.7 +/- 13.3%) and blastic (58.0 +/- 7.3%) phases of CML is statistically significant (p less than 0.05), however, the number of patients studied, especially in accelerated and blastic phases is very small. There was no difference in the CD34 positivity of the cells in the peripheral blood and in the bone marrow. CD34 positivity was higher in patients with chronic phase CML at diagnosis (untreated patients) than in those who were studied during treatment. The possible importance of serially studying CD34 positivity in patients with CML is discussed in the paper.


Subject(s)
Antigens, CD/physiology , Hematopoietic Stem Cells/immunology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology , Adolescent , Adult , Aged , Aged, 80 and over , Antigens, CD34 , Blast Crisis/immunology , Bone Marrow/immunology , Bone Marrow Cells , Female , Humans , Leukemia, Myeloid, Accelerated Phase/immunology , Male , Middle Aged , Prognosis
13.
Proc Soc Exp Biol Med ; 192(2): 201-4, 1989 Nov.
Article in English | MEDLINE | ID: mdl-2813453

ABSTRACT

3'-Azido-3'-deoxythymidine (AZT) has attained wide clinical utility in the treatment of acquired immunodeficiency syndrome (AIDS). Unfortunately, associated with AZT use, is the development of severe hematopoietic toxicity as manifested by anemia, neutropenia and overall bone marrow suppression. Interleukin-1 (IL-1), a cytokine, primarily produced by activated macrophages, has been involved in the control of hematopoiesis by acting synergistically with other hematopoietic growth factors, and has been demonstrated to be an effective agent in reducing the myelosuppression associated with the therapy for malignant disease. We report here the ability of recombinant human IL-1 alpha to protect normal murine hematopoietic progenitors (CFU-GM, BFU-E, and CFU-Meg) from the toxic effects of AZT. Following the determination of the LD50 dose for each progenitor, IL-1 was added in co-culture studies (10-1000 units; 0.001-1.0 micrograms/ml protein) with adherent cell depleted marrow. Marrow progenitors expressed differences in AZT sensitivity, e.g., BFU-E, LD50 5 x 10(-9)M; CFU-Meg, LD50 10(-7) M; CFU-GM, 5 x 10(-5) M respectively. IL-1 inhibited AZT induced toxicity. The maximum IL-1 dose effect was observed for CFU-GM and CFU-Meg at 300 units, 0.3 micrograms protein; however BFU-E required a dose of 600 units, 0.6 micrograms/ml protein to reverse the effects of AZT. These results demonstrate marrow progenitors respond differently to AZT and identifies the potential efficacy of IL-1 to minimize the hematopoietic toxicity associated with AZT treatment.


Subject(s)
Cell Survival/drug effects , Hematopoietic Stem Cells/cytology , Interleukin-1/pharmacology , Zidovudine/antagonists & inhibitors , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Erythropoietin/pharmacology , Female , Hematopoietic Stem Cells/drug effects , Kinetics , Mice , Mice, Inbred C3H , Recombinant Proteins/pharmacology , Zidovudine/pharmacology
14.
J Biol Response Mod ; 8(5): 479-87, 1989 Oct.
Article in English | MEDLINE | ID: mdl-2795093

ABSTRACT

Radiation exposure to various systems can result in the development of severe toxicity, known as the acute radiation syndrome, with hematopoietic tissues being acutely susceptible to radiation-induced injury. Usually it is the degree of hematopoietic toxicity that determines the feasibility of further tumor control doses of therapy. Therefore the development of agents capable of protecting hematopoietic tissues could have important clinical applications. The cytokine interleukin-1 (IL-1) has been demonstrated to be an effective agent capable of protecting hematopoietic tissues in vivo from the toxicity associated with radiation exposure. We report here the results of studies designed to further investigate the capability of various cytokines (IL-1 and interleukins-2 and -3) to protect bone marrow-derived hematopoietic progenitors [granulocyte-macrophage and megakaryocyte colony-forming units (CFU-GM and CFU-Meg)] from radiation exposure in vitro. Only IL-1 was effective in protecting CFU-GM and CFU-Meg from radiation-induced toxicity in the range of 1-10 U/ml; however, no protection was observed when the radiation dose was greater than 300 rad. The ability of IL-1 to block radiation-induced toxicity was negated in the presence of an antibody to IL-1. These studies provide further information on the effectiveness of IL-1 in protecting in vitro hematopoietic tissues from toxicity associated with radiation exposure.


Subject(s)
Interleukins/pharmacology , Radiation-Protective Agents , Stem Cells/drug effects , Animals , Bone Marrow/radiation effects , Bone Marrow Cells , Cells, Cultured , Colony-Forming Units Assay , Female , Interleukin-1/pharmacology , Interleukin-2/pharmacology , Interleukin-3/pharmacology , Mice , Mice, Inbred C3H , Stem Cells/radiation effects
15.
J Biol Response Mod ; 8(4): 422-39, 1989 Aug.
Article in English | MEDLINE | ID: mdl-2666587

ABSTRACT

Interleukin-1 (IL-1), a cytokine, primarily produced by monocytes, is the molecule involved in mediating many of the body's responses associated with infection and inflammation. More recently, IL-1 has been shown to sustain elevated levels of circulating granulocytes, stimulate the production of granulocyte-macrophage colony stimulating factors (CSFs) in vitro, increase plasma levels of CSF, and act synergistically with CSFs to increase the number of granulocyte-macrophage progenitors (colony-forming units) (CFU-GM) in vitro. The purpose of this study was to investigate the effect of murine IL-1 on steady-state hematopoiesis in vivo. C3H/HeJ or its normal littermate C3H/HeN male mice were administered either murine recombinant IL-1 at 45, 50, 200, 225, or 900 units (0.0125-0.25 micrograms)/animal, or 200 units (0.05 micrograms) of semipurified IL-1 derived from P388D1 cell culture supernatants. Because one of the responses to IL-1 is increased prostaglandin (PG) production and with the known activity of PGs on hematopoiesis, additional studies incorporated the cyclooxygenase inhibitor indomethacin (IM) (10 mg/kg body weight). In order to study the effect of IL-1 in vivo on pluripotential progenitors (CFU-S), IL-1 was compared with recombinant murine GM-CSF (50, 200, and 900 units; 0.0125, 0.05, and 0.25 micrograms). Control groups consisted of animals receiving either lipopolysaccharide (0.5 mg/kg body weight) or phosphate-buffered saline where appropriate. After 24 h, animals were sacrificed, and their peripheral blood indices and stem cell content of both bone marrow and spleen were evaluated for various committed hematopoietic progenitors: CFU-GM, CFU-Meg, CFU-E, BFU-E, and CFU-DG. Circulating neutrophils were increased following IL-1; however, this increase was reduced following IM. IL-1 marrow-derived CFU-GM, CFU-E, BFU-E, and CFU-Meg were below controls. In contrast, splenic CFU-GM and CFU-Meg were significantly elevated with increasing IL-1 concentrations. Erythroid progenitors were increased following low IL-1 concentrations and reduced in animals receiving IM, thus indicating a role for prostaglandins in the mechanism of IL-1 for influencing hematopoiesis. CFU-DG were increased, however, only when animals were pretreated with IL-1 and their cells implanted into normal hosts, not when normal cells were implanted into animals pretreated with IL-1, indicating a potential target cell effect rather than an indirect, factor-related response.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
Hematopoiesis/drug effects , Interleukin-1/pharmacology , Animals , Blood Platelets/drug effects , Bone Marrow Cells , Colony-Stimulating Factors/pharmacology , Diffusion Chambers, Culture , Granulocyte-Macrophage Colony-Stimulating Factor , Granulocytes/drug effects , Growth Substances/pharmacology , Hematocrit , Hematopoietic Stem Cells/drug effects , Leukocytes/drug effects , Macrophages/drug effects , Male , Megakaryocytes/drug effects , Mice , Recombinant Proteins/pharmacology , Spleen/cytology
16.
J Biol Response Mod ; 8(2): 116-21, 1989 Apr.
Article in English | MEDLINE | ID: mdl-2732751

ABSTRACT

Coumarin (1,2-benzopyrone), in combination with cimetidine, has been subjected to separate clinical trials for the treatment of advanced renal cell carcinoma, malignant melanoma, and non-small cell lung cancer. While objective tumor regressions were observed only in renal carcinoma, no symptomatic or organ dysfunction toxicity was observed in any of the trials. The purpose of the present in vitro study was to determine the concentrations of coumarin and 7-hydroxycoumarin (7-HC) that would be toxic to human peripheral blood mononuclear cells (PB-MNC) and human and murine bone marrow (GM) progenitor stem cells. Coumarin was nontoxic for PB-MNC in concentrations up to 100 micrograms/ml. Concentrations of coumarin or 7-HC greater than or equal to 200 micrograms/ml produced significant suppression of human marrow GM stem cell activity. Coumarin greater than or equal to 25 micrograms/ml produced suppression of murine marrow GM stem cell activity. Differences in human and murine marrow sensitivity probably reflect interspecies differences in metabolism of coumarin. Correlations between toxic concentrations in vitro and maximally achievable serum concentration in vivo in humans await the results of further clinical trials.


Subject(s)
Bone Marrow/drug effects , Coumarins/toxicity , Leukocytes, Mononuclear/drug effects , Stem Cells/drug effects , Umbelliferones/toxicity , Animals , Bone Marrow Cells , Cells, Cultured , Humans , Male , Mice
17.
Proc Soc Exp Biol Med ; 190(1): 109-16, 1989 Jan.
Article in English | MEDLINE | ID: mdl-2536179

ABSTRACT

Lithium (Li) is a known agent capable of producing leukocytosis, first observed in manic depressive patients receiving Li as therapy; however, a certain percentage of cases are nonresponsive to Li therapy. These patients are responsive to carbamazepine (CBZ); however, severe hematopoietic side effects have been associated with CBZ treatment such as leukopenia. We report here the results of dose-response studies (0.1-100 micrograms/ml) that demonstrate CBZ treatment inhibits both murine and human bone marrow-derived granulocyte-macrophage (CFU-GM), erythroid (BFU-E), and megakaryocyte progenitor (CFU-Meg) cells. The addition of Li prior to and simultaneously with CBZ to marrow cultures was effective in reversing the CBZ-induced toxicity only in the presence of an optimal Li dose (1.0 mM) known to stimulate bone marrow function. However, when the addition of Li was delayed 24 hr to CBZ-treated cultures no protective effect was observed for any marrow progenitor. Thus, the time of Li-CBZ exposure is critical to observe the protective effect of Li. These results demonstrate that the leukopenia associated with CBZ treatment may be due to the ability of CBZ to inhibit marrow progenitor cells and suggest Li may be an effective agent to reverse the marrow toxic effects of CBZ.


Subject(s)
Carbamazepine/pharmacology , Chlorides/pharmacology , Erythrocytes/cytology , Granulocytes/cytology , Hematopoietic Stem Cells/cytology , Lithium/pharmacology , Macrophages/cytology , Megakaryocytes/cytology , Animals , Bone Marrow/drug effects , Bone Marrow Cells , Cells, Cultured , Erythrocytes/drug effects , Granulocytes/drug effects , Hematopoietic Stem Cells/drug effects , Humans , Lithium Chloride , Macrophages/drug effects , Male , Megakaryocytes/drug effects , Mice , Mice, Inbred C3H , Rats , Reference Values
SELECTION OF CITATIONS
SEARCH DETAIL
...