Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Protein Sci ; 9(10): 2047-53, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11106181

ABSTRACT

A sequence variant of human MIP-1alpha, in which Asp26 has been replaced by Al alpha, has been chemically modified by the addition of 13C-labeled methyl groups at each of the lysine residues and the N-terminus. The sites of methylation have been verified by a combination of MALDI-TOF mass spectrometric experiments and tryptic digestion followed by N-terminal mapping. The effect of the modification on the structure and activity of the protein have been determined by analytical ultra-centrifugation, 13C NMR spectroscopy and receptor binding studies. The results of these experiments suggest that huMIP-alpha D26A (BB10010), when present as a dimer, adopts a globular structure, like MCP-3, rather than the elongated or cylindrical structure determined for dimers of huMIP-1beta and RANTES.


Subject(s)
Macrophage Inflammatory Proteins/chemistry , Macrophage Inflammatory Proteins/genetics , Alanine , Amino Acid Substitution , Aspartic Acid , Binding Sites , Chemokine CCL3 , Chemokine CCL4 , Chemokine CCL5/chemistry , Dimerization , Genetic Variation , Humans , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular , Peptide Fragments/chemistry , Protein Conformation , Receptors, Chemokine/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Trypsin , Ultracentrifugation
2.
J Hematother Stem Cell Res ; 9(5): 703-9, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11091494

ABSTRACT

A macrophage inflammatory protein-1alpha (MIP-1alpha) receptor has been cloned from rat (Rattus norvegicus) genomic DNA by PCR using oligonucleotides based on the mouse CCR1 DNA sequence and expressed in HEK293 cells to enable its characterization. The receptor was cloned three times in independent experiments to generate a consensus sequence. The rat ccrl receptor amino acid sequence is 92% identical to mouse and 80% identical to human CCR1. A consensus clone was transfected into HEK293 cells using the expression vector pIRES, and stable receptor expressing cell lines were isolated. In competitive receptor binding assays using iodinated human MIP-1alpha, rat ccrl binds hMIP-1alpha, hMIP-1beta, and hMCP-3, but not hRANTES or human interleukin-8 (hIL-8). We have been unable to demonstrate calcium mobilization by rat ccrl in HEK293 cells using human chemokines as ligands. Therefore, we have adopted lowercase nomenclature for the receptor until signaling is observed. This receptor and cell line may be of use in the preclinical development of CCR1 antagonists.


Subject(s)
Macrophage Inflammatory Proteins/metabolism , Receptors, Chemokine/physiology , Amino Acid Sequence , Animals , Base Sequence , Calcium Signaling , Cell Line , Cell Membrane/physiology , Chemokine CCL3 , Chemokine CCL4 , Chemokines/pharmacology , Chemokines/physiology , Consensus Sequence , DNA/blood , Humans , Mice , Molecular Sequence Data , Polymerase Chain Reaction/methods , Rats , Rats, Inbred Lew , Receptors, CCR1 , Receptors, Chemokine/chemistry , Receptors, Chemokine/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Substrate Specificity , Transfection
3.
Int Immunol ; 12(8): 1173-82, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10917892

ABSTRACT

RANTES (regulated upon activation, normal T cell expressed and secreted) is released by cytotoxic T lymphocytes (CTL), and is a potent chemoattractant factor for monocytes and T cells, also known for its ability to suppress HIV infection. At micromolar concentration, RANTES is able to activate leukocytes, and, paradoxically, to enhance HIV infection in vitro. These latter properties are dependent on its ability to self-aggregate. In order to understand further the mechanism of RANTES-induced activation, the effects of both aggregated and disaggregated RANTES on antigen-specific CD8(+) clones were studied in comparison with the effects of specific antigens and in the presence of specific inhibitors of RANTES-mediated activation. We observed large amounts of RANTES aggregated on the cell surface, which led to cell activation, including up-regulation of cell surface markers, and secretion of IFN-gamma and macrophage inflammatory protein (MIP)-1beta. Specific inhibitors of RANTES-induced activation, such as soluble glycosaminoglycans, MIP-1alpha and MIP-1beta, acted by preventing the binding of RANTES on the cell surface. These studies suggest that RANTES acted more like a mitogen than an antigen-independent activator.


Subject(s)
Chemokine CCL5/pharmacology , Cytotoxicity, Immunologic/drug effects , Lymphocyte Activation/drug effects , T-Lymphocytes, Cytotoxic/drug effects , Animals , Antigens, Surface/biosynthesis , Biopolymers , Blood Proteins/pharmacology , Cattle , Chemokine CCL3 , Chemokine CCL4 , Chemokine CCL5/antagonists & inhibitors , Chemokine CCL5/chemistry , Clone Cells , Fetal Blood , Glycosaminoglycans/pharmacology , Humans , Interferon-gamma/metabolism , Macromolecular Substances , Macrophage Inflammatory Proteins/metabolism , Macrophage Inflammatory Proteins/pharmacology , Protein Binding/drug effects , Receptors, Chemokine/drug effects , Receptors, Chemokine/metabolism , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism
4.
J Biol Chem ; 274(39): 27505-12, 1999 Sep 24.
Article in English | MEDLINE | ID: mdl-10488085

ABSTRACT

The biology of RANTES (regulated on activation normal T cell expressed) aggregation has been investigated using RANTES and disaggregated variants, enabling comparison of aggregated, tetrameric, and dimeric RANTES forms. Disaggregated variants retain their G(i)-type G protein-coupled receptor-mediated biological activities. A correlation between RANTES aggregation and cellular activation has been demonstrated. Aggregated RANTES, but not disaggregated RANTES, activates human T cells, monocytes, and neutrophils. Dimeric RANTES has lost its cellular activating activity, rendering it noninflammatory. Macrophage inflammatory protein 1alpha, macrophage inflammatory protein-1beta, and erythrocytes are potent natural antagonists of aggregated RANTES-induced cellular activation. There is a clear difference in the signaling properties of aggregated and disaggregated RANTES forms, separating the dual signaling pathways of RANTES and the enhancing and suppressive effects of RANTES on human immunodeficiency virus infection. Disaggregated RANTES will be a valuable tool to explore the biology of RANTES action in human immunodeficiency virus infection and in inflammatory disease.


Subject(s)
Calcium/metabolism , Chemokine CCL5/physiology , Chemotaxis, Leukocyte/physiology , Inflammation/immunology , Amino Acid Substitution , Cell Division/drug effects , Cell Line , Chemokine CCL4 , Chemokine CCL5/genetics , Chemokine CCL5/pharmacology , Chemotaxis, Leukocyte/drug effects , Erythrocytes/physiology , Genetic Variation , Humans , Jurkat Cells , Macromolecular Substances , Macrophage Inflammatory Proteins/pharmacology , Monocytes/physiology , Mutagenesis, Site-Directed , Neutrophils/physiology , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
5.
J Intraven Nurs ; 22(2): 75-80, 1999.
Article in English | MEDLINE | ID: mdl-10418410

ABSTRACT

Marketing an existing or new venture is a vital part of business. For the nurse entrepreneur, marketing involves applying previously learned skills to new situations. The methods used to market a service may mean the difference between success and failure. Unfortunately many entrepreneurs think that because they have a great idea, clients will beat a path to their door. Marketing requires planning, creativity, time, and money. It is an ongoing process that must be evaluated regularly. When marketing achieves results, clients commit to using the entrepreneur's services and profits are realized. Basic marketing concepts are considered, and strategies for developing a workable marketing plan are presented.


Subject(s)
Entrepreneurship , Marketing of Health Services/methods , Professional Competence/standards , Specialties, Nursing/organization & administration , Advertising , Career Mobility , Humans , Needs Assessment , Organizational Objectives , Referral and Consultation , Specialties, Nursing/education
6.
J Virol ; 73(8): 6370-9, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10400729

ABSTRACT

We have studied the mechanisms by which the CC-chemokine RANTES can enhance the infectivities of human immunodeficiency virus type 1 (HIV-1) and other enveloped viruses, when present at concentrations in excess of 500 ng/ml in vitro. Understanding the underlying mechanisms might throw light on fundamental processes of viral infection, in particular for HIV-1. Our principal findings are twofold: firstly, that oligomers of RANTES can cross-link enveloped viruses, including HIV-1, to cells via glycosaminoglycans (GAGs) present on the membranes of both virions and cells; secondly, that oligomers of RANTES interact with cell-surface GAGs to transduce a herbimycin A-sensitive signal which, over a period of several hours, renders the cells more permissive to infection by several viruses, including HIV-1. The enhancement mechanisms require that RANTES oligomerize either in solution or following binding to GAGs, since no viral infectivity enhancement is observed with a mutant form of the RANTES molecule that contains a single-amino-acid change (glutamic acid to serine at position 66) which abrogates oligomerization.


Subject(s)
Chemokine CCL5/metabolism , Glycosaminoglycans/metabolism , HIV-1/metabolism , Receptors, HIV/metabolism , Signal Transduction , Animals , CHO Cells , Cell Fusion , Cricetinae , HeLa Cells , Humans , Structure-Activity Relationship , Virion
7.
J Biol Chem ; 274(23): 16077-84, 1999 Jun 04.
Article in English | MEDLINE | ID: mdl-10347159

ABSTRACT

Human CC chemokines macrophage inflammatory protein (MIP)-1alpha, MIP-1beta, and RANTES (regulated on activation normal T cell expressed) self-associate to form high-molecular mass aggregates. To explore the biological significance of chemokine aggregation, nonaggregating variants were sought. The phenotypes of 105 hMIP-1alpha variants generated by systematic mutagenesis and expression in yeast were determined. hMIP-1alpha residues Asp26 and Glu66 were critical to the self-association process. Substitution at either residue resulted in the formation of essentially homogenous tetramers at 0.5 mg/ml. Substitution of identical or analogous residues in homologous positions in both hMIP-1beta and RANTES demonstrated that they were also critical to aggregation. Our analysis suggests that a single charged residue at either position 26 or 66 is insufficient to support extensive aggregation and that two charged residues must be present. Solution of the three-dimensional NMR structure of hMIP-1alpha has enabled comparison of these residues in hMIP-1beta and RANTES. Aggregated and disaggregated forms of hMIP-1alpha, hMIP-1beta, and RANTES generally have equivalent G-protein-coupled receptor-mediated biological potencies. We have therefore generated novel reagents to evaluate the role of hMIP-1alpha, hMIP-1beta, and RANTES aggregation in vitro and in vivo. The disaggregated chemokines retained their human immunodeficiency virus (HIV) inhibitory activities. Surprisingly, high concentrations of RANTES, but not disaggregated RANTES variants, enhanced infection of cells by both M- and T-tropic HIV isolates/strains. This observation has important implications for potential therapeutic uses of chemokines implying that disaggregated forms may be necessary for safe clinical investigation.


Subject(s)
Amino Acids/analysis , Chemokine CCL5/chemistry , Macrophage Inflammatory Proteins/chemistry , Amino Acid Sequence , Cell Line , Chemokine CCL3 , Chemokine CCL4 , Chemokine CCL5/genetics , HIV Infections/metabolism , HIV-1 , Humans , Macrophage Inflammatory Proteins/genetics , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Peptide Library , Protein Conformation , Structure-Activity Relationship
8.
Eur J Cancer ; 34(7): 1023-9, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9849450

ABSTRACT

BB-10010 is a genetically engineered variant of human macrophage inflammatory protein-1 alpha (hMIP-1 alpha) with improved pharmaceutical formulation properties. Although initially described as a pro-inflammatory cytokine, it is now recognised that hMIP-1 alpha has additional effects on haemopoietic stem cell cycling and on human immunodeficiency virus uptake by macrophages. In view of the potential clinical utility of the molecule, we have embarked on a clinical trials programme to evaluate the safety, tolerability and haematological effects of BB-10010. We now report the results of two phase I clinical studies in which 49 subjects (9 patients with advanced breast carcinoma and 40 normal healthy volunteers) received escalating doses of BB-10010, from 0.1 to 300 micrograms/kg using the subcutaneous (s.c.) or intravenous route (i.v.) of administration. Treatment was associated with a dose-related increase in monocyte count which peaked at 200% of steady-state levels and was preceded by an acute, short-lived, monocytopenia, 50-100% of baseline. no measurable effects were noted on other leucocyte subsets or on circulating progenitor cell numbers. In all cases, BB-10010 was extremely well tolerated with no significant toxicity observed at any dose level and a maximum tolerated dose was not defined. Pharmacokinetic analysis revealed that serum concentrations of BB-10010 were detectable using doses of > or = 10 micrograms/kg i.v. or > or = 30 micrograms/kg s.c., and that a single s.c. injection resulted in sustained plasma levels over a 24 h period. These preliminary studies have confirmed the safety and tolerability of BB-10010 using a dose range up to 300 micrograms/kg. Further clinical studies are ongoing to determine the biological effects and to investigate the potential myeloprotective properties using a variable dose range and schedule of BB-10010 in combination with cytotoxic chemotherapy.


Subject(s)
Breast Neoplasms/therapy , Macrophage Inflammatory Proteins/administration & dosage , Adult , Aged , Chemokine CCL3 , Chemokine CCL4 , Dose-Response Relationship, Drug , Female , Hematopoietic Stem Cells/physiology , Humans , Infusions, Intravenous , Injections , Leukocyte Count , Leukocytes/physiology , Macrophage Inflammatory Proteins/adverse effects , Macrophage Inflammatory Proteins/pharmacokinetics , Male , Middle Aged
9.
Blood ; 92(5): 1532-40, 1998 Sep 01.
Article in English | MEDLINE | ID: mdl-9716580

ABSTRACT

BB-10010 is a variant of the human form of macrophage inflammatory protein-1alpha (MIP-1alpha), which has been shown in mice to block the entry of hematopoietic stem cells into S-phase and to increase their self-renewal capacity during recovery from cytotoxic damage. Its use may constitute a novel approach for protecting the quality of the stem cell population and its capacity to regenerate after periods of cytotoxic treatment. Thirty patients with locally advanced or metastatic breast cancer were entered into the first randomized, parallel group controlled phase II study. This was designed to evaluate the potential myeloprotective effects of a 7-day regimen of BB-10010 administered to patients receiving six cycles of 5-fluorouracil (5-FU), adriamycin, and cyclophosphamide (FAC) chemotherapy. Patients were randomized, 10 receiving 100 microgram/kg BB-10010, 11 receiving 30 microgram/kg BB-10010, and nine control patients receiving no BB-10010. BB-10010 was well-tolerated in all patients with no severe adverse events related to the drug. Episodes of febrile neutropenia complicated only 4% of the treatment cycles and there was no difference in incidence between the treated and nontreated groups. Studies to assess the generation of progenitor cells in long-term bone marrow cultures were performed immediately preceding chemotherapy and at the end of six dosing cycles in 18 patients. Circulating neutrophils, platelets, CD 34(+) cells, and granulocyte/macrophage colony-forming cell (GM-CFC) levels were determined at serial time points in cycles 1, 3, and 6. The results showed similar hemoglobin and platelet kinetics in all three groups. On completion of the six treatment cycles, the average pretreatment neutrophil levels were reduced from 5.3 to 1.7 x 10(9)/L in the control patients and from 4.3 to 1.9 and 4.5 to 2.5 x 10(9)/L in the 30/100 microgram/kg BB-10010 groups, respectively. Relative to their pretreatment values, 50% of the patients receiving BB-10010 completed the treatment with neutrophil values significantly higher than any of the controls (P = .02). Mobilization of GM-CFC was enhanced by BB-10010 with an additional fivefold increase over that generated by chemotherapy alone, giving a maximal 25-fold increase over pretreatment values. Bone marrow progenitor assays before and after this standard regimen of chemotherapy indicated little long-term cumulative impairment to recovery from chemotherapy. Despite the limited cumulative damage to the bone marrow, which may have minimized the protective value of BB-10010 during this regimen of chemotherapy, better recovery of neutrophils in the later treatment cycles with BB-10010 was indicated in a number of patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Macrophage Inflammatory Proteins/therapeutic use , Adult , Aged , Bone Marrow Cells/cytology , Breast Neoplasms/pathology , Cell Count , Cells, Cultured , Chemokine CCL3 , Chemokine CCL4 , Cyclophosphamide/administration & dosage , Doxorubicin/administration & dosage , Female , Fluorouracil/administration & dosage , Hematopoiesis , Hematopoietic Stem Cells/cytology , Humans , Leukocyte Count , Macrophage Inflammatory Proteins/adverse effects , Macrophage Inflammatory Proteins/pharmacokinetics , Middle Aged , Neoplasm Metastasis , Neutrophils
10.
Leuk Lymphoma ; 30(1-2): 41-53, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9669675

ABSTRACT

The control of primitive haemopoietic progenitor cell proliferation in vitro can be achieved with combinations of growth stimulatory cytokines. Acting in apparent opposition to these growth stimulators are growth inhibitory substances, including prostaglandins, cytokines and chemokines which bind to specific cognate cell surface receptors and promote signal transduction events that interfere with cellular proliferation. Within the bone marrow microenvironment, significant quantities of both growth inhibitors and growth promoters can be detected. The ratio of their concentrations within microenvironmental niches of the marrow may regulate primitive blood cell production. The potential exists, therefore, for the disregulation of haemopoiesis via the disruption of the balance between positive and negative regulators of haemopoietic progenitor proliferation. In one particular disease, chronic myeloid leukaemia (CML), there is a lack of response of leukaemic cells to the chemokine growth inhibitor, Macrophage Inflammatory Protein-1alpha (MIP-1alpha). The role of MIP-1alpha in regulation of haemopoiesis, the response of CML progenitor cells and other myeloid leukaemic cells to this chemokine, and the reasons for lack of response to MIP-1alpha in leukaemic cells are reviewed.


Subject(s)
Growth Inhibitors/therapeutic use , Hematopoiesis/drug effects , Leukemia, Myeloid/drug therapy , Macrophage Inflammatory Proteins/therapeutic use , Receptors, Chemokine/drug effects , Chemokine CCL3 , Chemokine CCL4 , Clinical Trials as Topic , Drug Resistance, Neoplasm , Hematopoietic Stem Cell Mobilization , Humans
11.
Oncogene ; 16(10): 1319-24, 1998 Mar 12.
Article in English | MEDLINE | ID: mdl-9546433

ABSTRACT

The clonogenic cells of chronic myeloid leukaemia (CML), unlike normal haemopoietic progenitor cells, are resistant to the growth inhibitory effects of the chemokine macrophage inflammatory protein-1 alpha (MIP-1alpha). CML is also relatively resistant to chemotherapy and the disease is difficult to cure using conventional therapeutic routes. CML is associated with increased abl oncogene protein tyrosine kinase (PTK) activity. Here, we have tested the hypothesis that these aberrant responses to MIP-1alpha and the relative resistance to chemotherapy are directly related to this increased abl PTK activity in primitive haemopoietic cells. To do this we have expressed a temperature sensitive abl PTK in a growth factor dependent, multipotent stem cell line (FDCP-Mix) in which growth is normally suppressed by MIP-1alpha. In FDCP-Mix cells expressing the ts v-abl PTK and grown at the restrictive temperature for PTK activity the cells were relatively sensitive to cytotoxic agents such as cytosine arabinoside and 5-fluorouracil but MIP-1alpha could induce growth inhibition and confer some degree of protection from these agents. At the permissive temperature for abl PTK, the cells were relatively resistant to cytotoxic drugs and MIP-1alpha treatment neither induced growth inhibition nor protected the cells from cytotoxic drug induced cell death. This lack of response to MIP-1alpha was not due to receptor down modulation as neither the affinity nor the number of 125I-MIP-1alpha binding sites was altered by activating Abl PTK. However, MIP-1alpha mediated increases in cytosolic Ca2+ levels were abrogated by switching cells to the permissive temperature for Abl PTK activity. These data suggest that the relative resistance of CML progenitor cells to therapeutic drugs and the lack of response to MIP-1alpha occurs as a direct consequence of abl PTK activity and involves desensitisation of signal transduction events stimulated by MIP-1alpha receptors. Thus one contributory mechanism to transformation of primitive haemopoietic cells is abrogation of response to a growth inhibitor.


Subject(s)
Growth Inhibitors/pharmacology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Macrophage Inflammatory Proteins/pharmacology , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins c-abl/metabolism , Calcium/metabolism , Chemokine CCL3 , Chemokine CCL4 , DNA Replication/drug effects , Hematopoietic Stem Cells/drug effects , Humans , Interleukin-3/pharmacology , Kinetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Protein-Tyrosine Kinases/biosynthesis , Proto-Oncogene Proteins c-abl/biosynthesis , Recombinant Proteins/biosynthesis , Recombinant Proteins/metabolism , Thymidine/metabolism , Transfection
12.
Cytokines Cell Mol Ther ; 3(1): 41-50, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9287243

ABSTRACT

BB-10010 is a genetically modified form of human macrophage inflammatory protein-1 alpha (MIP-1 alpha) with a single amino acid substitution of Asp26 by alanine, which inhibits aggregation of the native molecule. BB-10010 has stem cell protective properties, and has entered clinical trials for this purpose. The aim of this study was to determine the effects of BB-10010 on human phagocyte function and compare them with the native molecule. MIP-1 alpha and BB-10010 had identical dose-response curves in assays of calcium mobilization; however, neutrophils were less sensitive than monocytes to either MIP-1 alpha form, suggesting differences in receptor expression. Neither MIP-1 alpha type directly stimulated phagocyte superoxide production, nor did it have any priming effect on agonist-induced superoxide production. Both MIP-1 alpha and BB-10010 enhanced monocyte migration; however, cells were more sensitive to the native molecule, with optimal effects seen at 1 ng/ml compared with 100 ng/ml BB-10010. Concomitant alteration of CD11b, CD18, and CD49d (VLA-alpha 4) cell adhesion molecule expression was not seen with either MIP-1 alpha type. With the exception of the difference in monocyte sensitivity in chemotaxis assays, BB-10010 reproduces the effects of the native molecule on phagocytes. BB-10010 does not have proinflammatory effects on neutrophil activation, and this bodes well for its clinical use.


Subject(s)
Macrophage Inflammatory Proteins/genetics , Macrophage Inflammatory Proteins/pharmacology , Phagocytes/drug effects , Calcium/metabolism , Cell Adhesion Molecules/metabolism , Cell Line , Chemokine CCL3 , Chemokine CCL4 , Genetic Variation , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , In Vitro Techniques , Intracellular Fluid/metabolism , Monocytes/drug effects , Monocytes/physiology , NADPH Oxidases/metabolism , Neutrophils/drug effects , Neutrophils/physiology , Phagocytes/physiology , Recombinant Proteins , Respiratory Burst/drug effects , Tumor Necrosis Factor-alpha/pharmacology
14.
Br J Haematol ; 95(1): 77-84, 1996 Oct.
Article in English | MEDLINE | ID: mdl-8857942

ABSTRACT

The effects of recombinant macrophage inflammatory protein 1 alpha (rhMIP-1 alpha) on the proliferation of leukaemic blast cells from patients with acute myeloid leukaemia was assessed. Using the previously described [3H]thymidine incorporation index assay, the response of autonomous and growth factor responsive AML blast cells to the chemokine rhMIP-1 alpha was measured. In the case of autonomous proliferators, rhMIP-1 alpha had no inhibitory effect on [3H]thymidine incorporation and in 4/6 cases [3H]-thymidine incorporation was stimulated by rhMIP-1 alpha. In the presence of stem cell factor (SCF), a majority (8/9) of the samples which responded to this growth factor were inhibited when rhMIP-1 alpha was included in the assay medium. Similar results were obtained with GM-CSF-responsive samples; however, when these two cytokines were combined, only 3/14 were significantly inhibited. In the presence of human placental conditioned medium (HPCM), rhMIP-1 alpha significantly inhibited [3H]thymidine incorporation in only 2/10 of HPCM-responsive samples. In methylcellulose assays rhMIP-1 alpha had no consistent effect on colony/cluster formation in the presence of either GM-CSF + SCF or HPCM. Similar results were obtained with BB-10010, a mutant of rhMIP-1 alpha which has defined aggregation properties in solution. These data suggest that autonomously proliferating AML cells, and also some AML samples which require cytokines to proliferate, are non-responsive to the growth inhibitors rhMIP-1 alpha and BB-10010 in the presence of multiple growth factors.


Subject(s)
Leukemia, Myeloid/pathology , Macrophage Inflammatory Proteins/pharmacology , Acute Disease , Cell Division/drug effects , Chemokine CCL3 , Chemokine CCL4 , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Leukocytes, Mononuclear/pathology , Stem Cell Factor/pharmacology , Tumor Cells, Cultured
15.
Blood ; 86(12): 4400-8, 1995 Dec 15.
Article in English | MEDLINE | ID: mdl-8541527

ABSTRACT

The stem cell inhibitor, macrophage inflammatory protein-1 alpha (MIP-1 alpha) or LD78, protects multipotent hematopoietic progenitors in murine models from the cytotoxic effects of chemotherapy. Clinical use of human MIP-1 alpha during chemotherapy could therefore lead to faster hematologic recovery and may allow dose intensification. We have also shown that human MIP-1 alpha causes the rapid mobilization of hematopoietic cells, suggesting an additional clinical use in peripheral blood stem cell transplantation. However, the clinical evaluation of human MIP-1 alpha is complicated by its tendency to associate and form high molecular weight polymers. We have produced a variant of rhMIP-1 alpha, BB-10010, carrying a single amino acid substitution of Asp26 > Ala, with a reduced tendency to form large polymers at physiologic pH and ionic strength. This greatly increases its solubility, facilitating its production and clinical formulation. We confirmed the potency of BB-10010 as a human MIP-1 alpha-like agonist in receptor binding, calcium mobilization, inhibition of colony formation, and thymidine suicide assays. The myeloprotective activity of BB-10010 was shown in a murine model of repeated chemotherapy using hydroxyurea. BB-10010 is therefore an ideal variant with which to evaluate the therapeutic potential of recombinant human MIP-1 alpha.


Subject(s)
Monokines/pharmacokinetics , Receptors, Chemokine , Recombinant Fusion Proteins/pharmacokinetics , Amino Acid Sequence , Animals , Biopolymers , Bone Marrow/drug effects , Bone Marrow Diseases/chemically induced , Bone Marrow Diseases/drug therapy , Calcium/metabolism , Cell Cycle/drug effects , Chemokine CCL3 , Chemokine CCL4 , Colony-Forming Units Assay , Growth Inhibitors/pharmacology , Hematopoietic Stem Cells/drug effects , Humans , Hydroxyurea/toxicity , Macrophage Inflammatory Proteins , Mice , Molecular Sequence Data , Monokines/chemistry , Monokines/genetics , Monokines/pharmacology , Mutagenesis, Site-Directed , Point Mutation , Radiation Injuries, Experimental/drug therapy , Receptors, Cytokine/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Sequence Alignment , Sequence Homology, Amino Acid , Solubility , Structure-Activity Relationship
16.
Blood ; 85(12): 3412-5, 1995 Jun 15.
Article in English | MEDLINE | ID: mdl-7540061

ABSTRACT

BB-10010 is a genetically engineered variant of human macrophage inflammatory protein-1 alpha with improved solution properties. We show here that it mobilizes stem cells into the peripheral blood. We investigated the mobilizing effects of BB-10010 on the numbers of circulating 8-day spleen colony-forming units (CFU-S8), CFU-S12, and progenitors with marrow repopulating ability (MRA). A single subcutaneous dose of BB-10010 caused a twofold increase in circulating numbers of CFU-S8, CFU-S12, and MRA 30 minutes after dosing. We also investigated the effects of granulocyte colony-stimulating factor (G-CSF) and the combination of G-CSF with BB-10010 on progenitor mobilization. Two days of G-CSF treatment increased circulating CFU-S8, CFU-S12, and MRA progenitors by 25.7-, 19.8-, and 27.7-fold. A single administration of BB-10010 after 2 days of G-CSF treatment increased circulating CFU-S8, CFU-S12, and MRA even further to 38-, 33-, and 100-fold. Splenectomy resulted in increased circulating progenitor numbers but did not change the pattern of mobilization. Two days of treatment with G-CSF then increased circulating CFU-S8, CFU-S12, and MRA by 64-, 69-, and 32-fold. A single BB-10010 administration after G-CSF treatment further increased them to 85-, 117-, and 140-fold, respectively, compared with control. We conclude that BB-10010 causes a rapid increase in the number of circulating hematopoietic progenitors and further enhances the numbers induced by pretreatment with G-CSF. BB-10010 preferentially mobilized the more primitive progenitors with marrow repopulating activity, releasing four times the number achieved with G-CSF alone. Translated into a clinical setting, this improvement in progenitor cell mobilization may enhance the efficiency of harvest and the quality of grafts for peripheral blood stem cell transplantation.


Subject(s)
Cytokines/metabolism , Hematopoietic Stem Cells/drug effects , Monokines/metabolism , Animals , Cell Count/drug effects , Chemokine CCL3 , Chemokine CCL4 , Colony-Forming Units Assay , Cytokines/genetics , Granulocyte Colony-Stimulating Factor/pharmacology , Humans , Macrophage Inflammatory Proteins , Mice , Monokines/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology
17.
Mol Microbiol ; 6(2): 267-75, 1992 Jan.
Article in English | MEDLINE | ID: mdl-1312212

ABSTRACT

The arginine-dependent repressor-activator from Bacillus subtilis, AhrC, has been overexpressed in Escherichia coli and purified to homogeneity. AhrC, expressed in E. coli, is able to repress a Bacillus promoter (argCp), which lies upstream of the argC gene. The purified protein is a hexamer with a subunit molecular mass of 16.7 kDa. Its ability to recognize DNA has been examined in vitro using argCp in both DNase I and hydroxyl radical protection assays. AhrC binds at two distinct sites within the argCp fragment. One site, argCo1, with the highest affinity for protein, is located within the 5' promoter sequences, whilst the other, argCo2, is within the coding region of argC. The data are consistent with the binding of a single hexamer of AhrC to argCo1 via four of its subunits, possibly allowing the remaining two subunits to bind at argCo2 in vivo forming a repression loop similar to those observed for the E. coli Lac repressor.


Subject(s)
Arginine/metabolism , Bacillus subtilis/genetics , Bacterial Proteins , Cyclic AMP Receptor Protein/metabolism , DNA, Bacterial/metabolism , Escherichia coli Proteins , Repressor Proteins/metabolism , Amino Acid Sequence , Bacillus subtilis/metabolism , Base Sequence , Cyclic AMP Receptor Protein/chemistry , Cyclic AMP Receptor Protein/genetics , Cyclic AMP Receptor Protein/isolation & purification , Escherichia coli/genetics , Escherichia coli/metabolism , Genes, Bacterial , Molecular Sequence Data , Molecular Weight , Promoter Regions, Genetic , Repressor Proteins/chemistry , Repressor Proteins/genetics , Repressor Proteins/isolation & purification
18.
J Mol Biol ; 213(2): 227-8, 1990 May 20.
Article in English | MEDLINE | ID: mdl-2111409

ABSTRACT

The arginine-dependent repressor/activator AhrC from Bacillus subtilis has been crystallized in space group C222(1), with unit cell dimensions a = 229.8 A, b = 72.8 A, c = 137.7 A and one aporepressor hexamer per asymmetric unit. Preliminary X-ray photographs show measurable intensities beyond 3.0 A.


Subject(s)
Bacterial Proteins , Repressor Proteins , Transcription Factors , Arginine/metabolism , Bacillus subtilis , Crystallization , X-Ray Diffraction
19.
Mol Microbiol ; 3(1): 23-8, 1989 Jan.
Article in English | MEDLINE | ID: mdl-2497296

ABSTRACT

The region required for regulation of a previously characterized arginine-regulatable promoter upstream from the argC gene in the argCAEBD-cpa-argF cluster of Bacillus subtilis was defined by integration of argC-lacZ translational fusions into the chromosome at a site distant from the arginine loci. Some sequence similarity was detected between the argC regulatory region and the well-characterized Escherichia coli arginine operators (ARG boxes). This similarity was shown to be functional in vivo in that the B. subtilis repressor regulated the E. coli arginine genes, but the E. coli repressor, even when encoded by a multicopy plasmid, could not repress the B. subtilis argC promoter. In vitro binding studies using purified repressors on DNA fragments encoding operators from both E. coli and B. subtilis demonstrated interactions by both proteins.


Subject(s)
Arginine/genetics , Bacillus subtilis/genetics , Escherichia coli/genetics , Promoter Regions, Genetic , Regulatory Sequences, Nucleic Acid , Arginine/biosynthesis , Base Sequence , DNA, Bacterial , Operator Regions, Genetic , Repressor Proteins/genetics , Transformation, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...