Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Int Immunopharmacol ; 123: 110688, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37499396

ABSTRACT

Antibiotic treatment may lead to side effects that require mechanistic explanation. We investigated the effect of azithromycin (AZM) treatment on bone marrow-derived macrophage (Mφ) generation, their functional output, and the subsequent effect on bacterial clearance in a mouse model of S. flexneri infection. To our fascination, AZM increased PU.1, C/EBPß, CSF-1R/pCSF-1R expressions leading to M2-skewed in vitro BMDM generation. Altered Mφ-functions like- phagocytosis, oxidative stress generation, inflammasome-activation, cytokine release, and phenotype (pro-inflammatory-M1, anti-inflammatory-M2) even in the presence of infection were observed with AZM treatment. AZM increased CD206, egr2, arg1 (M2-marker) expression and activity while reducing CD68, inducible nitric oxide (iNOS) expression, and activity (M1-marker) in Mφs during infection. Pro-inflammatory cytokines (TNF-α, IL-12, IL-1ß) were reduced and anti-inflammatory IL-10 release was augmented by AZM-treated-iMφs (aiMφs) along with decreased asc, nlrp3, aim2, nlrp1a, caspase1 expressions, and caspase3 activity signifying that aMφs/aiMφs were primed towards an anti-inflammatory phenotype. Interestingly, CSF-1R blockade increased NO, IL-12, TNF-α, IL-1ß, decreased TGF-ß release, and CD206 expression in aiMφs. T-cell co-stimulatory molecule cd40, cd86, and cd80 expressions were decreased in ai/aM1-Mφs and co-cultured CD8+, CD4+ T-cells had decreased proliferation, t-bet, IFN-γ, IL-17, IL-2 but increased foxp3, TGF-ß, IL-4 which were rescued with CSF-1R blockade. Thus AZM affected Mφ-functions and subsequent T-cell responses independent of its antibacterial actions. This was validated in the balb/c model of S. flexneri infection. We conclude that AZM skewed BMDM generation to anti-inflammatory M2-like via increased CSF-1R expression. This warrants further investigation of AZM-induced altered-Mφ-generation during intracellular infections.


Subject(s)
Azithromycin , Colony-Stimulating Factors , Receptor, Macrophage Colony-Stimulating Factor , Animals , Mice , Anti-Bacterial Agents/pharmacology , Azithromycin/pharmacology , Cytokines/metabolism , Interleukin-12/metabolism , Macrophages , Transforming Growth Factor beta/metabolism , Tumor Necrosis Factor-alpha/metabolism , Receptor, Macrophage Colony-Stimulating Factor/drug effects
2.
Arthritis Rheumatol ; 66(11): 2989-3000, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24623505

ABSTRACT

OBJECTIVE: Colony-stimulating factor 1 receptor (CSF-1R) essentially modulates monocyte proliferation, migration, and activation, which are considered important for the pathogenesis of rheumatoid arthritis (RA). We undertook this study to determine CSF-1R expression in human RA as well as the efficacy of a specific anti-CSF-1R monoclonal antibody (AFS98) in 2 different animal models of RA. METHODS: CSF-1R expression was examined in blood, synovium, and bone samples from RA patients, osteoarthritis (OA) patients, and healthy subjects. The efficacy of AFS98 was examined by clinical assessment, histology, and bone histomorphometry in collagen-induced arthritis (CIA) and serum-transfer arthritis. RESULTS: CSF-1R expression was increased in the synovium of RA patients compared to OA patients and healthy controls in fibroblast-like synoviocytes, follicular dendritic cells, macrophages, and osteoclasts. Circulating RA monocytes and neutrophils but not lymphocytes were CSF-1R+. In mice, blockade of CSF-1R abrogated cartilage damage, bone erosion, and systemic bone loss, and this was associated with the depletion of osteoclasts in both models. While blockade of CSF-1R did not affect inflammation in passive serum-transfer arthritis, it significantly reduced inflammation in CIA, and this was associated with the absence of synovial macrophages and reduced splenic CD11b+Gr-1- monocytes. CONCLUSION: CSF-1R was broadly expressed in human RA. Blockade of CSF-1R protected against bone and cartilage destruction in both mouse models and also showed significant antiinflammatory effects in the CIA model. These data provide evidence for CSF-1R as a therapeutic target in RA.


Subject(s)
Antibodies, Monoclonal/pharmacology , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/pathology , Bone and Bones/pathology , Cartilage/pathology , Osteoarthritis/pathology , Receptor, Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Arthritis, Experimental/drug therapy , Arthritis, Experimental/metabolism , Arthritis, Rheumatoid/metabolism , Bone and Bones/drug effects , Bone and Bones/metabolism , Cartilage/drug effects , Cartilage/metabolism , Case-Control Studies , Dendritic Cells/metabolism , Dendritic Cells/pathology , Disease Models, Animal , Female , Humans , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Inbred DBA , Middle Aged , Monocytes/metabolism , Monocytes/pathology , Osteoarthritis/metabolism , Osteoclasts/metabolism , Osteoclasts/pathology , Receptor, Macrophage Colony-Stimulating Factor/drug effects , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Synovial Membrane/drug effects , Synovial Membrane/metabolism , Synovial Membrane/pathology
3.
Arthritis Res Ther ; 12(1): R32, 2010.
Article in English | MEDLINE | ID: mdl-20181277

ABSTRACT

INTRODUCTION: Tyrosine kinases are key mediators of multiple signaling pathways implicated in rheumatoid arthritis (RA). We previously demonstrated that imatinib mesylate--a Food and Drug Administration (FDA)-approved, antineoplastic drug that potently inhibits the tyrosine kinases Abl, c-Kit, platelet-derived growth factor receptor (PDGFR), and c-Fms--ameliorates murine autoimmune arthritis. However, which of the imatinib-targeted kinases is the principal culprit in disease pathogenesis remains unknown. Here we examine the role of c-Fms in autoimmune arthritis. METHODS: We tested the therapeutic efficacy of orally administered imatinib or GW2580, a small molecule that specifically inhibits c-Fms, in three mouse models of RA: collagen-induced arthritis (CIA), anti-collagen antibody-induced arthritis (CAIA), and K/BxN serum transfer-induced arthritis (K/BxN). Efficacy was evaluated by visual scoring of arthritis severity, paw thickness measurements, and histological analysis. We assessed the in vivo effects of imatinib and GW2580 on macrophage infiltration of synovial joints in CIA, and their in vitro effects on macrophage and osteoclast differentiation, and on osteoclast-mediated bone resorption. Further, we determined the effects of imatinib and GW2580 on the ability of macrophage colony-stimulating factor (M-CSF; the ligand for c-Fms) to prime bone marrow-derived macrophages to produce tumor necrosis factor (TNF) upon subsequent Fc receptor ligation. Finally, we measured M-CSF levels in synovial fluid from patients with RA, osteoarthritis (OA), or psoriatic arthritis (PsA), and levels of total and phosphorylated c-Fms in synovial tissue from patients with RA. RESULTS: GW2580 was as efficacious as imatinib in reducing arthritis severity in CIA, CAIA, and K/BxN models of RA. Specific inhibition of c-Fms abrogated (i) infiltration of macrophages into synovial joints of arthritic mice; (ii) differentiation of monocytes into macrophages and osteoclasts; (iii) osteoclast-mediated bone resorption; and (iv) priming of macrophages to produce TNF upon Fc receptor stimulation, an important trigger of synovitis in RA. Expression and activation of c-Fms in RA synovium were high, and levels of M-CSF were higher in RA synovial fluid than in OA or PsA synovial fluid. CONCLUSIONS: These results suggest that c-Fms plays a central role in the pathogenesis of RA by mediating the differentiation and priming of monocyte lineage cells. Therapeutic targeting of c-Fms could provide benefit in RA.


Subject(s)
Antirheumatic Agents/pharmacology , Arthritis, Experimental/immunology , Arthritis, Rheumatoid/immunology , Cell Differentiation/immunology , Genes, fms/immunology , Monocytes/cytology , Animals , Anisoles/pharmacology , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Benzamides , Bone Resorption/immunology , Bone Resorption/metabolism , Cell Differentiation/drug effects , Cell Lineage/drug effects , Cell Lineage/immunology , Humans , Imatinib Mesylate , Immunohistochemistry , Inflammation/immunology , Macrophage Activation/drug effects , Macrophage Activation/immunology , Macrophage Colony-Stimulating Factor , Macrophages/cytology , Macrophages/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Monocytes/immunology , Osteoclasts/drug effects , Piperazines/pharmacology , Pyrimidines/pharmacology , Receptor, Macrophage Colony-Stimulating Factor/drug effects , Receptor, Macrophage Colony-Stimulating Factor/immunology , Receptor, Macrophage Colony-Stimulating Factor/metabolism
4.
J Leukoc Biol ; 87(1): 127-35, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19801501

ABSTRACT

M-CSF regulates the production, survival, and function of monocytes and macrophages. The MAPKs ERK1/2 are key elements for signal integration downstream of the M-CSFR, and their sustained activation is essential for macrophage differentiation. In this study, we sought to isolate genes whose induction by M-CSF is dependent on persistent MAPK activation, thereby being possibly involved in the commitment of myeloid progenitors to macrophage differentiation. Following SSH between cDNA libraries from FD-Fms cells stimulated by M-CSF for 8 h in the presence or the absence of the MEK inhibitor U0126, we isolated DUSP5. DUSP5 expression is induced by M-CSF in various myeloid cells and acts as a specific negative-feedback regulator of ERK1/2. In FD-Fms cells that proliferate and differentiate toward macrophages in response to M-CSF, overexpression of DUSP5 increased M-CSF-dependent proliferation and strongly decreased differentiation. Similarly, overexpression of DUSP5 in the multipotent EGER-Fms cells not only significantly increased M-CSF-induced proliferation and prevented macrophage differentiation but also favored granulocytic differentiation. Altogether, experiments demonstrated that DUSP5 is implicated in M-CSF signaling and suggested that it may influence myeloid cell fate.


Subject(s)
Dual-Specificity Phosphatases/physiology , Macrophage Colony-Stimulating Factor/pharmacology , Macrophages/cytology , Multipotent Stem Cells/drug effects , Myelopoiesis/drug effects , ADAM Proteins/biosynthesis , ADAM Proteins/genetics , Animals , Antigens, CD/biosynthesis , Antigens, CD/genetics , Butadienes/pharmacology , Cells, Cultured/cytology , Cells, Cultured/drug effects , Dual-Specificity Phosphatases/biosynthesis , Dual-Specificity Phosphatases/genetics , Gene Expression Regulation/drug effects , Humans , MAP Kinase Signaling System/drug effects , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Myelopoiesis/genetics , Nitriles/pharmacology , Osteopontin/biosynthesis , Osteopontin/genetics , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Receptor, Macrophage Colony-Stimulating Factor/drug effects , Receptor, Macrophage Colony-Stimulating Factor/genetics , Recombinant Fusion Proteins/drug effects , Recombinant Fusion Proteins/physiology
5.
Neoplasia ; 11(2): 136-44, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19177198

ABSTRACT

Coexpression of the macrophage colony-stimulating factor (CSF-1) and its receptor (CSF-1R) in metastatic ovarian cancer specimens is a predictor of poor outcome in epithelial ovarian cancer. This suggests that an autocrine loop is produced by which ovarian tumors can secrete CSF-1 stimulating the CSF-1R resulting in a more aggressive phenotype. Our current work sought to validate this autocrine stimulation model using stable transfection of a 4-kb CSF-1 construct into otherwise nonvirulent Bix3 ovarian cancer cells. A representative clone, Bix3T8.2, produced a 72-fold increase in CSF-1 gene transcription rate (by nuclear run-off assays) and a 57-fold increase in secreted CSF-1 protein (by sandwich ELISA), compared to parent cells. Comparison of Bix3T8.2 invasion, adhesion, and motility in vitro and metastasis in vivo were made to parental and transfectant controls. Up to 12-fold higher invasiveness was seen with Bix3T8.2 and 2- and 6-fold higher adhesion and motility, respectively, over controls in vitro. In nude mice, i.p. injection of Bix3T8.2 produced a wide array of visceral, nodal, and distant metastasis with a degree of enhanced tumor burden not seen in any of the 10 mice inoculated with transfectant control cells. Complete absence of tumor take distinguished 40% of mice implanted with transfectant control cells. Disruption of this autocrine loop using antisense oligomer therapy against CSF-1R and 3' untranslated region knockdown of CSF-1 protein resulted in reversal of in vitro and in vivo tumor phenotypes. This CSF-1 feedback loop offers a model by which novel biologic therapies can potentially target multiple levels of this pathway.


Subject(s)
Cell Transformation, Neoplastic , Macrophage Colony-Stimulating Factor/genetics , Macrophage Colony-Stimulating Factor/metabolism , Neoplasm Metastasis/pathology , Ovarian Neoplasms/pathology , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Adhesion , Cell Movement , Cell Transformation, Neoplastic/genetics , Female , Humans , Mice , Mice, Nude , Neoplasm Invasiveness , Neoplasm Transplantation , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Oligonucleotides, Antisense/therapeutic use , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Phenotype , Receptor, Macrophage Colony-Stimulating Factor/drug effects , Receptor, Macrophage Colony-Stimulating Factor/genetics , Tumor Cells, Cultured
6.
Atherosclerosis ; 196(2): 598-607, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17675037

ABSTRACT

Monocyte-to-macrophage differentiation and LDL oxidation play a pivotal role in early atherogenesis. We thus questioned possible mechanisms for oxidized LDL (Ox-LDL)-induced monocyte-to-macrophage differentiation in vivo. Mouse peritoneal mononuclear cells, that were isolated 1, 2, or 3 days after Ox-LDL intraperitoneal injection, gradually exhibited the characteristic macrophage morphology, along with the expression of the cell-surface antigen CD11b. Molecular mechanisms involved in Ox-LDL-induced differentiation were further investigated in vitro using the THP-1 monocytic cell line. THP-1 cells incubated with Ox-LDL in the presence of as low as 1 ng/ml of PMA differentiated into macrophages, as evidenced by morphologic, phenotypic, and functional properties. Stimulation of monocyte-to-macrophage differentiation was selective to Ox-LDL (and not native LDL), was dependent on the extent of LDL oxidation, and required Ox-LDL internalization by the cells. These effects of Ox-LDL could be attributed to its major oxysterols, 7-ketocholesterol and 7beta-hydroxycholesterol. Finally, the stimulation of monocyte differentiation to macrophages by Ox-LDL was shown to require the M-CSF-receptor, since blocking the binding to the receptor abolished Ox-LDL/7beta-hydroxycholesterol-induced differentiation. Furthermore, Ox-LDL/7beta-hydroxycholesterol elicited tyrosine phosphorylation and activation of the M-CSF-R. We thus conclude that Ox-LDL induces monocyte differentiation to macrophages in vivo and this phenomenon involves activation of the M-CSF-receptor.


Subject(s)
Cell Differentiation/drug effects , Lipoproteins, LDL/pharmacology , Macrophages, Peritoneal/cytology , Monocytes/cytology , Receptor, Macrophage Colony-Stimulating Factor/physiology , Animals , Cell Line, Tumor , Humans , Hydroxycholesterols/pharmacology , Lipoproteins, LDL/metabolism , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred BALB C , Receptor, Macrophage Colony-Stimulating Factor/drug effects
7.
Immunobiology ; 210(2-4): 97-107, 2005.
Article in English | MEDLINE | ID: mdl-16164016

ABSTRACT

We previously reported that bacterial products such as LPS and CpG DNA down-modulated cell surface levels of the Colony Stimulating Factor (CSF)-1 receptor (CSF-1R) on primary murine macrophages in an all-or-nothing manner. Here we show that the ability of bacterial products to down-modulate the CSF-1R rendered bone marrow-derived macrophages (BMM) unresponsive to CSF-1 as assessed by Akt and ERK1/2 phosphorylation. Using toll-like receptor (tlr)9 as a model CSF-1-repressed gene, we show that LPS induced tlr9 expression in BMM only when CSF-1 was present, suggesting that LPS relieves CSF-1-mediated inhibition to induce gene expression. Using cDNA microarrays, we identified a cluster of similarly CSF-1 repressed genes in BMM. By real time PCR we confirmed that the expression of a selection of these genes, including integral membrane protein 2B (itm2b), receptor activity-modifying protein 2 (ramp2) and macrophage-specific gene 1 (mpg-1), were repressed by CSF-1 and were induced by LPS only in the presence of CSF-1. This pattern of gene regulation was also apparent in thioglycollate-elicited peritoneal macrophages (TEPM). LPS also counteracted CSF-1 action to induce mRNA expression of a number of transcription factors including interferon consensus sequence binding protein 1 (Icsbp1), suggesting that this mechanism leads to transcriptional reprogramming in macrophages. Since the majority of in vitro studies on macrophage biology do not include CSF-1, these genes represent a set of previously uncharacterised LPS-inducible genes. This study identifies a new mechanism of macrophage activation, in which LPS (and other toll-like receptor agonists) regulate gene expression by switching off the CSF-1R signal. This finding also provides a biological relevance to the well-documented ability of macrophage activators to down-modulate surface expression of the CSF-1R.


Subject(s)
Gene Expression Regulation/drug effects , Lipopolysaccharides/pharmacology , Macrophage Activation/drug effects , Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Macrophages/drug effects , Animals , DNA-Binding Proteins/drug effects , DNA-Binding Proteins/immunology , DNA-Binding Proteins/metabolism , Gene Expression Regulation/immunology , Lipopolysaccharides/immunology , Lipopolysaccharides/metabolism , Macrophage Activation/immunology , Macrophage Colony-Stimulating Factor/immunology , Macrophages/immunology , Mice , Oligonucleotide Array Sequence Analysis , RNA, Messenger/analysis , Receptor, Macrophage Colony-Stimulating Factor/drug effects , Receptor, Macrophage Colony-Stimulating Factor/immunology , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Receptors, Cell Surface/drug effects , Receptors, Cell Surface/immunology , Receptors, Cell Surface/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Toll-Like Receptor 9
8.
Biochem J ; 380(Pt 1): 243-53, 2004 May 15.
Article in English | MEDLINE | ID: mdl-14969583

ABSTRACT

Macrophage colony-stimulating factor (M-CSF or CSF-1) controls the development of macrophage lineage cells via activation of its tyrosine kinase receptor, c-Fms. After adding CSF-1 to M1 myeloid cells expressing CSF-1R (CSF-1 receptor), tyrosine phosphorylation of many cellular proteins occurs, which might be linked to subsequent macrophage differentiation. The biological significance and characterization of such proteins were explored by a dual strategy comprising two-dimensional SDS/PAGE analysis of cell lysates of CSF-1-treated M1 cells expressing the wild-type or a mutated receptor, together with an enrichment strategy involving a tyrosine-phosphorylated receptor construct. In the present study, we report the identification by MS of a novel, low-abundance, 110 kDa form of myosin XVIIIA (MysPDZ, myosin containing PDZ domain), which appears to be preferentially tyrosine-phosphorylated after CSF-1R activation when compared with other known isoforms. Receptor mutation studies indicate that CSF-1R-dependent tyrosine phosphorylation of p110myosin XVIIIA requires Tyr-559 in the cytoplasmic domain of the receptor and is therefore Src-family kinase-dependent. Gelsolin, Erp61 protein disulphide-isomerase and possibly non-muscle myosin IIA were also tyrosine-phosphorylated under similar conditions. Similar to the more abundant p190 isoform, p110 myosin XVIIIA lacks a PDZ domain and, in addition, it may lack motor activity. The phosphorylation of p110 myosin XVIIIA by CSF-1 may alter its cellular localization or target its association with other proteins.


Subject(s)
Macrophage Colony-Stimulating Factor/pharmacology , Myosins/metabolism , Protein Processing, Post-Translational , Receptor, Macrophage Colony-Stimulating Factor/physiology , Signal Transduction , Amino Acid Sequence , Animals , Cell Differentiation , Cell Line/drug effects , Cell Line/metabolism , Electrophoresis, Gel, Two-Dimensional , Gelsolin/metabolism , Genes, fms , Heat-Shock Proteins/metabolism , Isomerases/metabolism , Macrophages/drug effects , Mice , Molecular Sequence Data , Myeloid Cells/metabolism , Myosins/chemistry , Myosins/isolation & purification , Nonmuscle Myosin Type IIA/metabolism , Phosphorylation/drug effects , Phosphotyrosine/analysis , Protein Disulfide-Isomerases , Protein Processing, Post-Translational/drug effects , Receptor, Macrophage Colony-Stimulating Factor/drug effects , Recombinant Fusion Proteins/physiology , Transfection , src-Family Kinases/metabolism
9.
Neuroscience ; 112(4): 889-900, 2002.
Article in English | MEDLINE | ID: mdl-12088748

ABSTRACT

Prenatal exposure to teratogen agents is linked to the pathogenesis of neurodevelopment disorders, but the mechanisms leading to the neurodevelopmental disturbance are poorly understood. To elucidate this, an in vitro model of microglial activation induced by neuronal injury has been characterized. In this connection, exposure of primary microglial cells to the conditioned medium from the neuronal damage induced by teratogen, cyclophosphamide, is accompanied by a reactive microgliosis as assessed by reverse transcription-polymerase chain reaction, enzyme-linked immunosorbent assay, lectin histochemistry, double labeling immunohistochemistry and in situ hybridization. Our results showed that reactive microglia were capable of releasing various cytokines such as tumor necrosis factor-alpha, interleukin-1, interleukin-6, transforming growth factor-beta and nitric oxide. Also, we have shown that macrophage colony-stimulating factor (M-CSF) was in fact produced by the reactive microglia. Concomitant to this was the increased expression of M-CSF receptor in these cells following the teratogen-induced neuronal injury. The up-regulation of M-CSF receptor suggests that the cells are capable of responding to self-derived M-CSF in an autocrine fashion. Results with antibody neutralization further suggest that microglial proinflammatory response, as manifested by cytokine expression in culture, is mediated by M-CSF, which acts as a molecular signal that initiates a microglial reaction. We therefore suggest that microglial activation following cyclophosphamide treatment is not only a response to the neuronal damage, but is also a cause of the damage during pathogenesis of neurodevelopment disorders. To this end, the increased expression of M-CSF and its receptor on microglia would be directly linked to the active cell proliferation and proinflammatory response in the teratogen-induced injury.


Subject(s)
Cerebral Cortex/drug effects , Cyclophosphamide/toxicity , Macrophage Colony-Stimulating Factor/metabolism , Microglia/metabolism , Neurons/drug effects , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Teratogens/toxicity , Animals , Cell Culture Techniques , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Immunohistochemistry , In Situ Hybridization , Interleukin-1/metabolism , Interleukin-6/metabolism , Macrophage Colony-Stimulating Factor/drug effects , Microglia/drug effects , Microglia/immunology , Microscopy, Confocal , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Macrophage Colony-Stimulating Factor/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Transforming Growth Factor beta/metabolism , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation
10.
J Soc Gynecol Investig ; 8(2): 114-6, 2001.
Article in English | MEDLINE | ID: mdl-11336883

ABSTRACT

OBJECTIVE: We present the results of the application of organ culture techniques previously described in this journal to the study of steroid hormone responsiveness of primary breast carcinoma specimens. METHODS AND RESULTS: Nearly all breast carcinomas that express macrophage colony-stimulating factor-1R (CSF-1R) at tissue harvest (15 of 18) had levels of CSF-1R expression lowered after incubation in steroid-free media. The decrease in CSF-1R expression was reversed by treatment with glucocorticoids; this glucocorticoid-induced increase in CSF-1R expression can be blocked by mifepristone (RU-486), a competitive inhibitor of glucocorticoid action. CONCLUSION: These results demonstrate that steroid hormone responsiveness of primary breast carcinomas can be assayed in vitro, a result which can not only be employed to better predict the responsiveness of breast carcinomas to therapies with steroid hormone agonists and antagonists, but also suggests that the therapeutic utility of mifepristone in breast cancer deserves further study.


Subject(s)
Breast Neoplasms/chemistry , Glucocorticoids/pharmacology , Hormone Antagonists/pharmacology , Mifepristone/pharmacology , Receptor, Macrophage Colony-Stimulating Factor/analysis , Dexamethasone/pharmacology , Glucocorticoids/antagonists & inhibitors , Humans , Immunohistochemistry , Organ Culture Techniques , Receptor, Macrophage Colony-Stimulating Factor/drug effects
11.
J Cell Biochem ; 72(1): 119-34, 1999 Jan 01.
Article in English | MEDLINE | ID: mdl-10025673

ABSTRACT

Addition of colony stimulating factor-1 (CSF-1) to macrophages stimulates the rapid, transient tyrosine phosphorylation, membrane association and multiubiquitination of Cbl (Wang et al. [1996] J. Biol. Chem. 271:17-20). Kinetic analysis reveals that the tyrosine phosphorylation of Cbl is coincident with its plasma membrane translocation and association with the activated tyrosine phosphorylated CSF-1 R, p85, Grb2, and tyrosine phosphorylated p58Shc and that these events precede the simultaneous multiubiquitination of Cbl and the CSF-1 R. Tyrosine phosphorylation and multiubiquitination of the cell surface CSF-1 R are stoichiometric and the multiubiquitinated CSF-1 R is degraded. Similarly, the membrane associated Cbl is almost stoichiometrically ubiquitinated, but the ubiquitinated Cbl is not degraded, being recovered, deubiquitinated, in the cytosol 3-10 min after stimulation at 37 degrees C. In the membrane fraction of cells stimulated at 4 degrees C, the association of p58Shc and Grb2 with Cbl is stable, whereas its association with Sos and p85 is transient and their dissociation occurs at the time CSF-1 R and Cbl multiubiquitination commence. The membrane translocation and the pattern of association of Sos with the CSF-1R, p85, Grb2, and p58Shc resemble those of Cbl but Sos is not tyrosine phosphorylated, nor multiubiquitinated and the coprecipitation of these proteins, other than Grb2, with Sos is much less. Complexes formed by Sos and Cbl are largely independent and membrane complexes of Cbl with other tyrosine phosphorylated proteins, p85 and Grb2 also contain CSF-1 R. These data raise the possibility that the predicted negative regulatory role of Cbl in macrophages is its enhancement of ligand-induced CSF-1 R internalization/degradation.


Subject(s)
Adaptor Proteins, Signal Transducing , Macrophage Colony-Stimulating Factor/pharmacology , Macrophages/metabolism , Proto-Oncogene Proteins/metabolism , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Tyrosine/metabolism , Ubiquitin-Protein Ligases , Ubiquitins/metabolism , Amino Acid Sequence , Animals , Cell Line , Cell Membrane/metabolism , Fluorescent Antibody Technique , GRB2 Adaptor Protein , Mice , Molecular Sequence Data , Phosphorylation , Proteins/metabolism , Proto-Oncogene Proteins c-cbl , Receptor, Macrophage Colony-Stimulating Factor/drug effects , Signal Transduction/physiology
12.
Mol Reprod Dev ; 46(1): 96-103, 1997 Jan.
Article in English | MEDLINE | ID: mdl-8981370

ABSTRACT

The normal proto-oncogene c-fms encodes the macrophage growth factor (M-CSF) receptor involved in growth, survival, and differentiation along the monocyte-macrophage lineage of hematopoietic cell development. A major portion of our research concerns unraveling the temporal, molecular, and structural features that determine and regulate these events. Previous results indicated that c-fms can transmit a growth signal as well as a signal for differentiation in the appropriate cells. To investigate the role of the Fms tyrosine autophosphorylation sites in proliferation vs. differentiation signaling, four of these sites were disrupted and the mutant receptors expressed in a clone derived from the myeloid FDC-P1 cell line. These analyses revealed that: (1) none of the four autophosphorylation sites studied (Y697, Y706, Y721, and Y807) are essential for M-CSF-dependent proliferation of the FDC-P1 clone; (2) Y697, Y706, and Y721 sites, located in the kinase insert region of Fms, are not necessary for differentiation but their presence augments this process; and (3) the Y807 site is essential for the Fms differentiation signal: its mutation totally abrogates the differentiation of the FDC-P1 clone and conversely increases the rate of M-CSF-dependent proliferation. This suggests that the Y807 site may control a switch between growth and differentiation. The assignment of Y807 as a critical site for the reciprocal regulation of growth and differentiation may provide a paradigm for Fms involvement in leukemogenesis, and we are currently investigating the downstream signals transmitted by the tyrosine-phosphorylated 807 site. In Fms-expressing FDC-P1 cells, M-CSF stimulation results in the rapid (30 sec) tyrosine phosphorylation of Fms on the five cytoplasmic tyrosine autophosphorylation sites, and subsequent tyrosine phosphorylation of several host cell proteins occurs within 1-2 min. Complexes are formed between Fms and other signal transduction proteins such as Grb2, Shc, Sos1, and p85. In addition, a new signal transduction protein of 150 kDa is detectable in the FDC-P1 cells. The p150 is phosphorylated on tyrosine, and forms a complex with Shc and Grb2. The interaction with Shc occurs via a protein tyrosine binding (PTB) domain at the N-terminus of Shc. The p150 is not detectable in Fms signaling within fibroblasts, yet the PDGF receptor induces the tyrosine phosphorylation of a similarly sized protein. In hematopoietic cells, this protein is involved in signaling by receptors for GM-CSF, IL-3, KL, MPO, and EPO. We have now cloned a cDNA for this protein and found at least one related family member. The related family member is a Fanconia Anemia gene product, and this suggests potential ways the p150 protein may function in Fms signaling.


Subject(s)
Cell Differentiation/physiology , Cell Division/physiology , Macrophage Colony-Stimulating Factor/physiology , Receptor, Macrophage Colony-Stimulating Factor/physiology , Animals , Cell Differentiation/drug effects , Cell Division/drug effects , Cloning, Molecular , DNA, Complementary/genetics , Humans , Macrophage Colony-Stimulating Factor/pharmacology , Mice , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/physiology , Phosphorylation , Protein Conformation , Protein Kinases/metabolism , Protein Processing, Post-Translational , Proto-Oncogene Mas , Receptor, Macrophage Colony-Stimulating Factor/chemistry , Receptor, Macrophage Colony-Stimulating Factor/deficiency , Receptor, Macrophage Colony-Stimulating Factor/drug effects , Receptor, Macrophage Colony-Stimulating Factor/genetics , src Homology Domains
13.
Oncogene ; 13(12): 2607-13, 1996 Dec 19.
Article in English | MEDLINE | ID: mdl-9000134

ABSTRACT

Using FDC-P1 derived cell lines which ectopically express either the wild type or mutant forms of the murine CSF-1 receptor in which individual tyrosine residues have been replaced with phenylalanine, we analysed the requirement for tyrosine residues of the receptor for the activation of STAT proteins in response to CSF-1. We found Y706 to be required for efficient activation of STAT1. The activation of STAT3 was not affected by the mutation of Y706 to phenylalanine. The addition of phosphopeptides spanning Y708 of the human CSF-1 receptor (identical with the sequence surrounding Y706 of the murine receptor) to electrophoretic mobility shift assays led to competition of the formation of STAT1 containing complexes, SIF-B and SIF-C with the DNA probe. These phosphopeptides did, however, not affect the formation of the STAT3 containing complex, SIF-A, with the probe. Replacement of Y807 with phenylalanine led to a complete block of activation of all STAT proteins in response to CSF-1, however, this phosphotyrosine does not appear to represent a STAT binding site of the receptor as a phosphopeptide spanning Y809 of the human CSF-1 receptor could not compete any STAT/DNA complex formation in electrophoretic mobility shift assays.


Subject(s)
DNA-Binding Proteins/metabolism , Macrophage Colony-Stimulating Factor/pharmacology , Protein-Tyrosine Kinases , Receptor, Macrophage Colony-Stimulating Factor/drug effects , Trans-Activators/metabolism , Animals , Binding Sites , Cell Line , Humans , Mice , Phosphorylation , Proteins/metabolism , Receptor, Macrophage Colony-Stimulating Factor/metabolism , STAT1 Transcription Factor , STAT3 Transcription Factor , Signal Transduction , TYK2 Kinase
14.
Leuk Res ; 20(8): 665-76, 1996 Aug.
Article in English | MEDLINE | ID: mdl-8913320

ABSTRACT

Retinoic acid (RA) and 1,25-dihydroxyvitamin D3 (D3) are well known for inducing differentiation in many leukemic cell lines. The nuclear signalling pathways of RA and D3 are mediated through their cognate receptors, the retinoic acid receptor (RAR) and vitamin D3 receptor (VDR), respectively. Retinoid X receptor (RXR) is an auxiliary factor that forms a heterodimer with RAR and VDR, enabling their efficient transcriptional activation. 9-cis RA, a high-affinity ligand for RXR, greatly enhanced D3-induced CD14 expression in U937 cells, while RA alone did not induce CD14 expression. 9-cis RA also resulted in morphological changes of U937 cells to macrophage-like cells when combined with D3, while RA alone resulted in granulocyte-like cells. RA and D3 together enhanced c-fms expression, phagocytic activity, and acted synergistically to promote nitroblue tetrazolium reduction activity and inhibit proliferation. Northern analysis showed that U937 cells constitutively expressed RAR-alpha, VDR and RXR-alpha mRNAs. RA or D3 alone or in combination did not affect RAR-alpha and VDR expression, while 9-cis RA and 9-cis RA plus all-trans RA significantly reduced RXR-alpha expression. Interestingly, D3 could restore the down-regulation of RXR-alpha mRNA by 9-cis RA. These findings suggest that there is crossover of the nuclear signalling pathways of RA and D3. This may have clinical implications in that RA and D3 may be used in combination for differentiation-inducing therapy in acute myelogenous leukemia and myelodysplastic syndrome.


Subject(s)
Antineoplastic Agents/pharmacology , Calcitriol/pharmacology , Cell Differentiation/drug effects , Monocytes/drug effects , Tretinoin/pharmacology , Cell Line , Granulocytes/cytology , Humans , Lipopolysaccharide Receptors/drug effects , Lipopolysaccharide Receptors/metabolism , Monocytes/cytology , RNA, Messenger/metabolism , Receptor, Macrophage Colony-Stimulating Factor/drug effects , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Stereoisomerism , Tretinoin/analogs & derivatives
15.
Bone ; 18(4): 355-60, 1996 Apr.
Article in English | MEDLINE | ID: mdl-8726394

ABSTRACT

Macrophage colony-stimulating factor (M-CSF) receptor has been previously reported to be present in osteoclasts both at mRNA and protein levels. However, the biochemical interactions between M-CSF and its receptor on osteoclasts are less well characterized than in mononuclear phagocytes. In this study, we show that (1) 125I-labeled M-CSF ligand specifically binds to the M-CSF receptor on osteoclasts by autoradiography; (2) binding of M-CSF to the receptor stimulates protein tyrosine phosphorylation in osteoclasts by immunostaining; (3) oxygen-derived free radicals produced by calvarial osteoclasts are increased by M-CSF stimulation (1.37 +/- 0.08, n = 10, P < 0.01); and (4) bone resorption in calvarial explants is enhanced by M-CSF (1.153 +/- 0.09, n = 10, p < 0.001). Thus, our data provide multiple lines of evidences that mouse calvarial osteoclasts are activated by M-CSF. These data suggest that under the conditions present in the calvarial model, M-CSF activates osteoclastic bone resorption.


Subject(s)
Macrophage Colony-Stimulating Factor/metabolism , Osteoclasts/metabolism , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Animals , Autoradiography , Binding, Competitive , Bone Resorption/chemically induced , Free Radicals/metabolism , Humans , In Situ Hybridization , Iodine Radioisotopes , Isotope Labeling , Macrophage Colony-Stimulating Factor/toxicity , Mice , Mice, Inbred C57BL , Osteoclasts/cytology , Osteoclasts/drug effects , Phosphorylation , Radioligand Assay , Receptor, Macrophage Colony-Stimulating Factor/drug effects , Receptor, Macrophage Colony-Stimulating Factor/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/toxicity , Tyrosine/metabolism
16.
Endocrinology ; 136(12): 5643-50, 1995 Dec.
Article in English | MEDLINE | ID: mdl-7588319

ABSTRACT

1,25-Dihydroxyvitamin D3 [1,25-(OH)2D3] and macrophage colony-stimulating factor (M-CSF) both accelerate differentiation of marrow macrophages from which osteoclasts are derived. Previously, we showed that the steroid's effect on early macrophage precursors may be mediated through M-CSF, as the steroid enhances cytokine receptor expression. In contrast, 1,25-(OH)2D3 blunts M-CSF receptor expression on more mature, yet still pluripotential, hematopoietic precursors. Extending these observations to marrow cells committed to macrophage differentiation, we found that 1,25-(OH)2D3 causes a marked decrease in cellular proliferation despite a 2- to 3-fold increase in [125I]M-CSF binding in a similar dose-dependent metabolite-specific manner. Scatchard analysis demonstrated that increased binding reflects increased receptor capacity without an alteration in affinity. Steroid-induced M-CSF receptor enhancement reflects acceleration of protein appearance rather than overexpression, as treated and untreated cells ultimately exhibit equivalent binding. Increased M-CSF receptor expression is mirrored by increased c-fms messenger RNA levels, and actinomycin D or cycloheximide experiments indicate that new receptor synthesis, rather than mobilization of intracellular pools, is required. Thus, 1,25-(OH)2D3 differentially impacts on M-CSF receptor expression throughout the spectrum of bone marrow macrophage differentiation.


Subject(s)
Calcitriol/pharmacology , Macrophages/drug effects , Stem Cells/drug effects , Animals , Bone Marrow Cells , Cell Differentiation/drug effects , Cell Division/drug effects , Cells, Cultured , Mice , Receptor, Macrophage Colony-Stimulating Factor/analysis , Receptor, Macrophage Colony-Stimulating Factor/biosynthesis , Receptor, Macrophage Colony-Stimulating Factor/drug effects , Receptor, Macrophage Colony-Stimulating Factor/genetics
17.
Blood ; 86(8): 2948-56, 1995 Oct 15.
Article in English | MEDLINE | ID: mdl-7579387

ABSTRACT

Colony-stimulating factor 1 (CSF-1) causes the activation of STAT1 and STAT3 transcription factors in bone marrow macrophages (BMM), in the macrophage cell line BAC1.2F5, and in fibroblasts that express the wild-type receptor for CSF-1. Fibroblasts expressing a mutant receptor in which the tyrosine 809 is replaced with phenylalanine do not activate STAT proteins in response to CSF-1. The activation of the STAT proteins in BMM is accompanied by tyrosine phosphorylation of Tyk2. In fibroblasts, the activation of the STAT proteins is accompanied by tyrosine phosphorylation of Tyk2 and JAK1. We propose that these JAK kinases are subjected to very rapid phosphorylation in response to CSF-1, followed by rapid dephosphorylation. Furthermore, we propose that kinases other than JAK kinase may be involved in the phosphorylation of the STAT proteins in response to CSF-1.


Subject(s)
3T3 Cells/drug effects , Bone Marrow/drug effects , DNA-Binding Proteins/metabolism , Macrophage Colony-Stimulating Factor/pharmacology , Macrophages/drug effects , Protein Processing, Post-Translational/drug effects , Protein-Tyrosine Kinases/metabolism , Proteins/metabolism , Trans-Activators/metabolism , 3T3 Cells/metabolism , Amino Acid Sequence , Animals , Base Sequence , Bone Marrow Cells , Enzyme Activation/drug effects , Interferon-beta/pharmacology , Interferon-gamma/pharmacology , Janus Kinase 1 , Macrophages/metabolism , Mice , Molecular Sequence Data , Phosphorylation/drug effects , Platelet-Derived Growth Factor/pharmacology , Receptor, Macrophage Colony-Stimulating Factor/drug effects , Receptor, Macrophage Colony-Stimulating Factor/genetics , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Recombinant Proteins , STAT1 Transcription Factor , STAT3 Transcription Factor , TYK2 Kinase
18.
Cell Growth Differ ; 6(6): 631-45, 1995 Jun.
Article in English | MEDLINE | ID: mdl-7545432

ABSTRACT

The macrophage colony-stimulating factor (M-CSF) regulates proliferation and differentiation of cells belonging to the monocytic lineage. We have investigated the nature and origin of the proliferation and differentiation signals derived from the M-CSF receptor (Fms) by mutating Fms at the four tyrosine autophosphorylation sites and examining their biological effects in an FDC-P1 clone. Wild-type Fms stimulated both growth and differentiation of FDC-P1 cells in response to M-CSF stimulation. In contrast, both proliferation and differentiation were differentially disrupted by mutations affecting the four tyrosine autophosphorylation sites. These analyses revealed that: (a) none of the four autophosphorylation sites studied (Y697, Y706, Y721, and Y807) were essential for M-CSF-dependent proliferation of the FDC-P1 clone; (b) Y697, Y706, and Y721 sites, located in the kinase insert region of Fms, were not necessary for differentiation, but their presence augmented this process; (c) mutation of the Y807 site totally abrogated the differentiation of the FDC-P1 clone and simultaneously increased the rate of M-CSF-dependent proliferation; and (d) conversely, increasing the intracellular cAMP level blocked the growth signal in the FDC-P1 clone but had no effect on differentiation. These results suggest that autophosphorylation of Fms at the Y807 site controls the balance between signals for growth and differentiation.


Subject(s)
Cell Differentiation/drug effects , Cell Division/drug effects , Hematopoietic Stem Cells/drug effects , Macrophage Colony-Stimulating Factor/pharmacology , Protein Processing, Post-Translational , Receptor, Macrophage Colony-Stimulating Factor/physiology , Animals , Colforsin/pharmacology , Cyclic AMP/physiology , Dinoprostone/pharmacology , Hematopoietic Stem Cells/cytology , Mice , Mutagenesis, Site-Directed , Phosphorylation , Phosphotyrosine , Receptor, Macrophage Colony-Stimulating Factor/drug effects , Receptor, Macrophage Colony-Stimulating Factor/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction , Transfection , Tyrosine/analogs & derivatives , Tyrosine/chemistry , Tyrosine/physiology
19.
Bone ; 16(6): 611-8, 1995 Jun.
Article in English | MEDLINE | ID: mdl-7669437

ABSTRACT

The toothless (tl) osteopetrotic mutation in the rat is characterized by generalized skeletal sclerosis, a severe reduction in the numbers of osteoclasts, monocytes, and macrophages, and absence of tooth eruption. Studies examining gene expression in bone-derived cells of tl rats and their normal littermates have shown that genes related to osteoblast function are aberrantly expressed in tl rats compared to normal littemates. We have previously shown that exogenous administration of colony stimulating factor-1 (CSF-1) to tl rats results in a dramatic reduction of the skeletal sclerosis and significant increases in the number of osteoclasts. Thus, we examined the effects of CSF-1 on osteoblast and osteoclast gene expression in tl rats as demonstrated by Northern blot analysis. While osteoblast-related gene expression as reflected by mRNA levels of alkaline phosphatase, osteocalcin, osteopontin, and type I collagen was normalized, osteoclast-related gene expression, as reflected by mRNA levels of carbonic anhydrase II and tartrate-resistant adenosine triphosphatase, remained significantly lower in CSF-1-treated tl rats compared to untreated normal littermates. Since previous studies have not demonstrated the CSF-1 receptor on osteoblasts, these results suggest that osteoblast abnormalities in tl rats are an effect of the osteopetrotic condition rather than the cause of the disease.


Subject(s)
Gene Expression Regulation/drug effects , Macrophage Colony-Stimulating Factor/pharmacology , Osteoblasts/drug effects , Osteoclasts/drug effects , Osteopetrosis/genetics , Adenosine Diphosphate/metabolism , Alkaline Phosphatase/genetics , Alkaline Phosphatase/metabolism , Animals , Blotting, Northern , Bone and Bones/diagnostic imaging , Bone and Bones/drug effects , Carbonic Anhydrases/genetics , Carbonic Anhydrases/metabolism , Collagen/genetics , Collagen/metabolism , Disease Models, Animal , Gene Expression Regulation/genetics , Histones/genetics , Histones/metabolism , Macrophage Colony-Stimulating Factor/administration & dosage , Macrophage Colony-Stimulating Factor/therapeutic use , Macrophages/cytology , Macrophages/drug effects , Monocytes/cytology , Monocytes/drug effects , Osteoblasts/cytology , Osteocalcin/genetics , Osteocalcin/metabolism , Osteoclasts/cytology , Osteopetrosis/pathology , Osteopetrosis/physiopathology , Osteopontin , RNA, Messenger/metabolism , Radiography , Rats , Receptor, Macrophage Colony-Stimulating Factor/drug effects , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Sialoglycoproteins/genetics , Sialoglycoproteins/metabolism , Tibia/diagnostic imaging , Tibia/drug effects
20.
Mol Cell Biol ; 14(7): 4606-15, 1994 Jul.
Article in English | MEDLINE | ID: mdl-8007965

ABSTRACT

c-fps/fes encodes a 92-kDa protein-tyrosine kinase (NCP92) that is expressed at the highest levels in macrophages. To determine if c-fps/fes can mediate the action of the colony-stimulating factor 1 (CSF-1) receptor (CSF-1R) and to identify potential targets of c-fps/fes in macrophages, we have overexpressed c-fps/fes in a CSF-1-dependent macrophage cell line. A 30- to 50-fold overexpression of c-fps/fes partially released these cells from their factor dependence by a nonautocrine mechanism, and this correlated with the tyrosine phosphorylation of two proteins of 130 and 75 kDa (P130 and P75). c-fps/fes did not cause tyrosine phosphorylation or activation of CSF-1 dependent targets, including CSF-1R, Shc, and phosphatidylinositol 3-kinase, and conversely, CSF-1 did not induce tyrosine phosphorylation of P130 and P75. P75 appears to be a novel phosphotyrosyl protein, whereas P130 cross-reacts with a known substrate of v-src. P130 and P75 may be direct substrates of c-fps/fes: P130 was tightly associated with NCP92, and the src homology 2 domain of NCP92 specifically bound phosphorylated P130 and P75 but not the CSF-1-induced phosphotyrosyl proteins, consistent with the possibility that P130 and P75 are physiological targets of c-fps/fes. We conclude that although c-fps/fes can functionally substitute for CSF-1R to a certain extent, these tyrosine kinases act largely independently of each other and that P130 and P75 are novel targets whose mechanisms of action may be unrelated to the signalling pathways utilized by receptor tyrosine kinases.


Subject(s)
Macrophage Colony-Stimulating Factor/pharmacology , Macrophages/metabolism , Phosphoproteins/metabolism , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Animals , Cell Line , Cell Membrane/enzymology , Cytoplasm/enzymology , GTPase-Activating Proteins , Humans , Kinetics , Oncogene Protein pp60(v-src)/metabolism , Phosphoproteins/isolation & purification , Proteins/metabolism , Receptor, Macrophage Colony-Stimulating Factor/drug effects , Substrate Specificity , Thermodynamics , Transfection , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL