Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
2.
Ann Oncol ; 24(7): 1777-1785, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23510984

ABSTRACT

BACKGROUND: Targeted agents presently available for mutant KRAS metastatic colorectal cancer (mCRC) are bevacizumab and aflibercept. We evaluated the efficacy and safety of conatumumab (an agonistic monoclonal antibody against human death receptor 5) and ganitumab (a monoclonal antibody against the type 1 insulin-like growth factor receptor) combined with standard FOLFIRI chemotherapy as a second-line treatment in patients with mutant KRAS mCRC. PATIENTS AND METHODS: Patients with mutant KRAS metastatic adenocarcinoma of the colon or rectum refractory to fluoropyrimidine- and oxaliplatin-based chemotherapy were randomized 1 : 1 : 1 to receive intravenous FOLFIRI plus conatumumab 10 mg/kg (Arm A), ganitumab 12 mg/kg (Arm B), or placebo (Arm C) Q2W. The primary end point was progression-free survival (PFS). RESULTS: In total, 155 patients were randomized. Median PFS in Arms A, B, and C was 6.5 months (HR, 0.69; P = 0.147), 4.5 months (HR, 1.01; P = 0.998), and 4.6 months, respectively; median overall survival was 12.3 months (HR, 0.89; P = 0.650), 12.4 months (HR, 1.27; P = 0.357), and 12.0 months; and objective response rate was 14%, 8%, and 2%. The most common grade ≥3 adverse events in Arms A/B/C included neutropenia (30%/25%/18%) and diarrhea (18%/2%/10%). CONCLUSIONS: Conatumumab, but not ganitumab, plus FOLFIRI was associated with a trend toward improved PFS. Both combinations had acceptable toxicity.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colorectal Neoplasms/drug therapy , Liver Neoplasms/drug therapy , Proto-Oncogene Proteins/genetics , ras Proteins/genetics , Adenocarcinoma/blood , Adenocarcinoma/mortality , Adenocarcinoma/secondary , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal, Humanized , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Biomarkers, Tumor/blood , Camptothecin/administration & dosage , Camptothecin/analogs & derivatives , Colorectal Neoplasms/blood , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Disease-Free Survival , Double-Blind Method , Female , Fluorouracil/administration & dosage , Genotype , Humans , Insulin-Like Growth Factor Binding Proteins/blood , Irinotecan , Kaplan-Meier Estimate , Leucovorin/administration & dosage , Liver Neoplasms/blood , Liver Neoplasms/mortality , Liver Neoplasms/secondary , Male , Middle Aged , Proportional Hazards Models , Proto-Oncogene Proteins p21(ras) , Receptors, IgG/genetics , Treatment Outcome
3.
Oncogene ; 32(11): 1428-40, 2013 Mar 14.
Article in English | MEDLINE | ID: mdl-22562252

ABSTRACT

Basal-like breast cancer is an aggressive subtype of mammary carcinoma. Despite expressing basal markers, typical of mammary stem cells, this tumor has been proposed to originate from luminal progenitors, which are downstream of stem cells along the mammary epithelial hierarchy. This suggests that committed luminal progenitors may reacquire basal, stem-like characteristics, but the mechanisms that regulate this transition remain unclear. Using mouse models, we found that luminal progenitors express high levels of the Met receptor for hepatocyte growth factor (HGF), as compared with the other mammary epithelial sub-populations. Constitutive activation of Met led luminal progenitors to attain stem cell properties, including enhanced clonogenic activity in vitro and de novo ability to reconstitute mammary glands in repopulation assays in vivo. Moreover, in response to Met signaling, luminal progenitors gave rise to hyperplastic ductal morphogenesis and preferentially underwent basal lineage commitment at the expense of luminal cell-fate specification. Opposite and symmetric results were produced by systemic pharmacological inhibition of Met. Hence, Met signaling targets luminal progenitors for expansion, impairs their differentiation toward the mature luminal phenotype and enables their commitment toward the basal lineage. These results emphasize a critical role for Met in promoting deregulated proliferation and basal plasticity of normal luminal progenitors in the mammary gland, a complex of events that may be required for sustaining the functional and phenotypic properties of basal-like breast tumors.


Subject(s)
Breast Neoplasms/pathology , Cell Differentiation/genetics , Cell Proliferation , Epithelial Cells/physiology , Mammary Glands, Animal/physiology , Neoplasms, Basal Cell/pathology , Proto-Oncogene Proteins c-met/physiology , Animals , Breast Neoplasms/genetics , Cell Lineage/genetics , Cells, Cultured , Epithelial Cells/metabolism , Female , Mammary Glands, Animal/cytology , Mammary Glands, Animal/metabolism , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mice , Neoplasms, Basal Cell/genetics , Phenotype , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , Signal Transduction/genetics , Stem Cells/metabolism , Stem Cells/physiology
4.
Ann Oncol ; 23(11): 2834-2842, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22700995

ABSTRACT

BACKGROUND: We evaluated the efficacy and safety of ganitumab (a mAb antagonist of insulin-like growth factor 1 receptor) or conatumumab (a mAb agonist of human death receptor 5) combined with gemcitabine in a randomized phase 2 trial in patients with metastatic pancreatic cancer. PATIENTS AND METHODS: Patients with a previously untreated metastatic pancreatic adenocarcinoma and an Eastern Cooperative Oncology Group (ECOG) performance status ≤1 were randomized 1 : 1 : 1 to i.v. gemcitabine 1000 mg/m(2) (days 1, 8, and 15 of each 28-day cycle) combined with open-label ganitumab (12 mg/kg every 2 weeks [Q2W]), double-blind conatumumab (10 mg/kg Q2W), or double-blind placebo Q2W. The primary end point was 6-month survival rate. Results In total, 125 patients were randomized. The 6-month survival rates were 57% (95% CI 41-70) in the ganitumab arm, 59% (42-73) in the conatumumab arm, and 50% (33-64) in the placebo arm. The grade ≥3 adverse events in the ganitumab, conatumumab, and placebo arms, respectively, included neutropenia (18/22/13%), thrombocytopenia (15/17/8%), fatigue (13/12/5%), alanine aminotransferase increase (15/5/8%), and hyperglycemia (18/2/3%). CONCLUSIONS: Ganitumab combined with gemcitabine had tolerable toxicity and showed trends toward an improved 6-month survival rate and overall survival. Additional investigation into this combination is warranted. Conatumumab combined with gemcitabine showed some evidence of activity as assessed by the 6-month survival rate.


Subject(s)
Adenocarcinoma/drug therapy , Antibodies, Monoclonal/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Deoxycytidine/analogs & derivatives , Pancreatic Neoplasms/drug therapy , Adenocarcinoma/mortality , Adenocarcinoma/secondary , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal, Humanized , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Deoxycytidine/administration & dosage , Deoxycytidine/adverse effects , Female , Humans , Male , Middle Aged , Neoplasm Metastasis/drug therapy , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Placebos , Receptor, IGF Type 1/antagonists & inhibitors , Receptor, IGF Type 1/immunology , Survival Rate , Treatment Outcome , Gemcitabine
5.
Am J Transplant ; 6(11): 2601-11, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16981910

ABSTRACT

Donor intra-islet endothelial cells contribute to neovascularization after transplantation. Several factors may interfere with this process and ultimately influence islet engraftment. Rapamycin, a central immunosuppressant in islet transplantation, is an mTOR inhibitor that has been shown to inhibit cancer angiogenesis. The aim of this study was to evaluate the effects of rapamycin on islet endothelium. Rapamycin inhibited the outgrowth of endothelial cells from freshly purified human islets and the formation of capillary-like structures in vitro and in vivo after subcutaneous injection within Matrigel plugs into SCID mice. Rapamycin decreased migration, proliferation and angiogenic properties of human and mouse islet-derived endothelial cell lines with appearance of apoptosis. The expression of angiogenesis-related factors VEGF, alphaVbeta3 integrin and thrombospondin-1 on islet endothelium was altered in the presence of rapamycin. On the other hand, rapamycin decreased the surface expression of molecules involved in immune processes such as ICAM-1 and CD40 and reduced the adhesion of T cells to islet endothelium. Our results suggest that rapamycin exerts dual effects on islet endothelium inducing a simultaneous inhibition of angiogenesis and a down-regulation of receptors involved in lymphocyte adhesion and activation.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Endothelium, Vascular/physiology , Immunologic Factors/pharmacology , Islets of Langerhans Transplantation/pathology , Islets of Langerhans/cytology , Sirolimus/pharmacology , Animals , Cell Division/drug effects , Cell Movement/drug effects , Cell Survival/drug effects , Collagen , Drug Combinations , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Humans , Islets of Langerhans/blood supply , Laminin , Mice , Mice, SCID , Neovascularization, Pathologic/prevention & control , Proteoglycans , Transplantation, Heterologous
7.
J Immunol ; 166(2): 1241-7, 2001 Jan 15.
Article in English | MEDLINE | ID: mdl-11145707

ABSTRACT

Hepatocyte growth factor (HGF) is a potent paracrine mediator of stromal/epithelial interactions, which is secreted as a matrix-associated inactive precursor (pro-HGF) and locally activated by tightly controlled urokinase cleavage. It induces proliferation and motility in epithelial and endothelial cells, and plays a role in physiological and pathological processes involving invasive cell growth, such as angiogenesis and parenchymal regeneration. We now report that HGF induces directional migration and cytokine secretion in human monocytes. Monocyte activation by endotoxin and IL-1beta results in the up-regulation of the HGF receptor expression and in the induction of cell-associated pro-HGF convertase activity, thus enhancing cell responsiveness to the factor. Furthermore, we provide evidence for the secretion of biologically active HGF by activated monocytes, implying an autocrine stimulation. Altogether, these data indicate that monocyte function is modulated by HGF in a paracrine/autocrine manner, and provide a new link between stromal environment and mononuclear phagocytes.


Subject(s)
Hepatocyte Growth Factor/physiology , Macrophages/immunology , Monocytes/immunology , Cell Adhesion/immunology , Cells, Cultured , Chemotaxis, Leukocyte/immunology , Culture Media/metabolism , Granulocyte Colony-Stimulating Factor/biosynthesis , Granulocyte Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Hepatocyte Growth Factor/biosynthesis , Hepatocyte Growth Factor/genetics , Hepatocyte Growth Factor/metabolism , Humans , Interleukin-6/biosynthesis , Interleukin-6/genetics , Macrophage Activation/immunology , Macrophages/metabolism , Monocytes/metabolism , Platelet Activating Factor/pharmacology , Protein Precursors/metabolism , Proto-Oncogene Proteins c-met/biosynthesis , Proto-Oncogene Proteins c-met/blood , RNA, Messenger/biosynthesis , Recombinant Proteins/pharmacology , Transcription, Genetic/immunology
8.
Proc Natl Acad Sci U S A ; 93(15): 7644-8, 1996 Jul 23.
Article in English | MEDLINE | ID: mdl-8755529

ABSTRACT

Hepatocyte growth factor (HGF), also known as scatter factor, is a powerful motogen, mitogen, and morphogen produced by cells of mesodermal origin, acting on epithelial and endothelial cells. Its receptor is the tyrosine kinase encoded by the c-MET protooncogene. We show that the HGF receptor is expressed by human primary osteoclasts, by osteoclast-like cell lines, and by osteoblasts. In both cell lineages, HGF stimulation triggers the receptor kinase activity and autophosphorylation. In osteoclasts, HGF receptor activation is followed by increase in intracellular Ca2+ concentration and by activation of the pp60c-Src kinase. HGF induces changes in osteoclast shape and stimulates chemotactic migration and DNA replication. Osteoblasts respond to HGF by entering the cell cycle, as indicated by stimulation of DNA synthesis. Interestingly, osteoclasts were found to synthesize and secrete biologically active HGF. These data strongly suggest the possibility of an autocrine regulation of the osteoclast by HGF and a paracrine regulation of the osteoblast by the HGF produced by the osteoclast.


Subject(s)
Hepatocyte Growth Factor/pharmacology , Osteoblasts/physiology , Osteoclasts/physiology , Receptor Protein-Tyrosine Kinases/physiology , Bone Neoplasms , Calcium/metabolism , Cell Communication , Cell Movement/drug effects , Cells, Cultured , Coculture Techniques , DNA Replication/drug effects , Enzyme Activation , Giant Cell Tumors , Hepatocyte Growth Factor/metabolism , Humans , Kinetics , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoclasts/cytology , Osteoclasts/drug effects , Proto-Oncogene Proteins c-met , Proto-Oncogene Proteins pp60(c-src)/metabolism , Receptor Protein-Tyrosine Kinases/biosynthesis , Time Factors
9.
J Biol Chem ; 271(12): 6590-3, 1996 Mar 22.
Article in English | MEDLINE | ID: mdl-8636073

ABSTRACT

nef is a human immunodeficiency virus (HIV) gene encoding a 27-kDa myristoylated protein with structural features of a signal transducing molecule, but whose functions are largely unknown. We studied the interactions of Nef with the signal transduction pathways triggered by the platelet-derived growth factor (PDGF) receptor. The association of phosphatidylinositol (PI) 3-kinase with the activated receptor was severely impaired by nef expression. Conversely, PDGF-induced receptor tyrosine phosphorylation, binding to phospholipase C-gamma and to Ras-GAP were not modified. Microtubule-associated protein kinase activation and intracellular calcium influx in response to PDGF were either unaffected or only slightly enhanced. Nef significantly reduced the proliferative response to the growth factor, while the chemotactic response was unchanged. These data show that Nef affects selectively the PI 3-kinase signaling pathway and suggest that this interference results in some of the HIV adverse effects on host cell functions.


Subject(s)
Gene Products, nef/physiology , HIV-1/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , 3T3 Cells , Animals , Becaplermin , Calcium Channels/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Division , Enzyme Activation , Gene Expression Regulation , Gene Products, nef/genetics , Genes, nef , Mice , Phosphatidylinositol 3-Kinases , Phosphorylation , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Platelet-Derived Growth Factor/antagonists & inhibitors , Platelet-Derived Growth Factor/pharmacology , Proto-Oncogene Proteins c-sis , Receptors, Platelet-Derived Growth Factor/metabolism , Signal Transduction , nef Gene Products, Human Immunodeficiency Virus
10.
J Biol Chem ; 270(2): 603-11, 1995 Jan 13.
Article in English | MEDLINE | ID: mdl-7822285

ABSTRACT

Hepatocyte growth factor (HGF) is a paracrine inducer of morphogenesis and invasive growth in epithelial and endothelial cells. HGF is secreted by mesenchymal cells as an inactive precursor (pro-HGF). The crucial step for HGF activation is the extracellular hydrolysis of the Arg494-Val495 bond, which converts pro-HGF into alpha beta-HGF, the high-affinity ligand for the Met receptor. We previously reported that the urokinase-type plasminogen activator (uPA) activates pro-HGF in vitro. We now show that this is a stoichiometric reaction, and provide evidence for its occurrence in tissue culture. Activation involves the formation of a stable complex between pro-HGF and uPA. This complex was isolated from the in vitro reaction of pure uPA with recombinant pro-HGF, as well as from the membrane of target cells, after sequential addition of uPA and pro-HGF. On the cell membrane, the uPA-HGF complex was bound to the Met receptor. Monocytic cell lines, and primary monocytes after adhesion, activated efficiently pro-HGF both on their surface and in the culture medium. This activation was inhibited by anti-catalytic anti-uPA antibodies, and occurred by a stoichiometric reaction. The stoichiometry of the activation reaction suggests that the biological effects of HGF can be titrated in vivo by the level of uPA activity. Adequate amounts of uPA can be locally provided by the macrophages, which would condition the tissue microenvironment by rendering HGF bioavailable to its target cells.


Subject(s)
Hepatocyte Growth Factor/metabolism , Protein Precursors/metabolism , Urokinase-Type Plasminogen Activator/physiology , Cells, Cultured , Humans , Kinetics , Protein Processing, Post-Translational , Structure-Activity Relationship
11.
J Cell Biol ; 127(6 Pt 1): 1743-54, 1994 Dec.
Article in English | MEDLINE | ID: mdl-7528222

ABSTRACT

Hepatocyte growth factor (HGF) is a mesenchymal derived growth factor known to induce proliferation and "scattering" of epithelial and endothelial cells. Its receptor is the tyrosine kinase encoded by the c-MET protooncogene. Here we show that highly purified recombinant HGF stimulates hemopoietic progenitors to form colonies in vitro. In the presence of erythropoietin, picomolar concentrations of HGF induced the formation of erythroid burst-forming unit colonies from CD34-positive cells purified from human bone marrow, peripheral blood, or umbilical cord blood. The growth stimulatory activity was restricted to the erythroid lineage. HGF also stimulated the formation of multipotent CFU-GEMM colonies. This effect is synergized by stem cell factor, the ligand of the tyrosine kinase receptor encoded by the c-KIT protooncogene, which is active on early hemopoietic progenitors. By flow cytometry analysis, the receptor for HGF was found to be expressed on the cell surface in a fraction of CD34+ progenitors. Moreover, in situ hybridization experiments showed that HGF receptor mRNA is highly expressed in embryonic erythroid cells (megaloblasts). HGF mRNA was also found to be produced in the embryonal liver. These data show that HGF plays a direct role in the control of proliferation and differentiation of erythroid progenitors, and they suggest that it may be one of the long-sought mediators of paracrine interactions between stromal and hemopoietic cells within the hemopoietic microenvironment.


Subject(s)
Erythroid Precursor Cells/drug effects , Hepatocyte Growth Factor/pharmacology , Animals , Antigens, CD , Antigens, CD34 , Baculoviridae/genetics , Bone Marrow/embryology , Cell Differentiation/drug effects , Cell Division/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Drug Synergism , Hematopoietic Cell Growth Factors/pharmacology , Hematopoietic Stem Cells/drug effects , Hepatocyte Growth Factor/genetics , Humans , In Situ Hybridization , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-kit , Proto-Oncogene Proteins c-met , RNA, Messenger/analysis , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Colony-Stimulating Factor/metabolism , Recombinant Proteins/pharmacology , Spodoptera/cytology , Stem Cell Factor
12.
Liver ; 14(4): 213-9, 1994 Aug.
Article in English | MEDLINE | ID: mdl-7968281

ABSTRACT

Recent evidence suggests that anti-androgen therapy may be useful in patients with androgen receptor (AR)-positive hepatocellular carcinomas (HCC), as determined by a steroid binding assay. To evaluate the AR expression of HCC, in both histological and cytological material, we developed a non-radioisotopic in situ hybridisation (NISH) assay specific for the human AR mRNA. A synthetic oligonucleotide complementary to positions 661-695 of the human AR coding sequence was end-labelled with digoxigenin-dUTP and revealed by an alkaline phosphatase-conjugated anti-digoxigenin antibody. We analysed 22 formalin-fixed, paraffin-embedded HCC, obtained at surgery, together with the corresponding non-neoplastic liver tissues (19 cases). In six cases, cell blocks obtained by fine-needle aspiration (FNA) prior to surgery were also available. Positive controls included seminal vesicles and prostate tissues. Sixteen HCCs (73%) expressed a variable amount of AR mRNA, with the proportion of positive cells ranging from very few to more than 90%. Normal hepatocytes were stained weakly and focally in eight cases (42%). Appropriate controls, inclusive of immunohistochemical detection of the AR protein in selected cases, established the specificity of the assay. Data obtained on FNA specimens were predictive of the results on histologic material. However, in two cases the NISH assay was negative on the cytological specimen but stained rare hepatocytes within the surgically resected tumor. In conclusion, NISH is a novel procedure for rapid and specific assessment of the expression of AR in HCC tissue. Its clinical significance, in terms of predictivity of response to anti-androgen treatment, needs to be assessed in large correlative studies.


Subject(s)
Carcinoma, Hepatocellular/chemistry , Liver Neoplasms/chemistry , Liver/chemistry , RNA, Messenger/analysis , Receptors, Androgen/analysis , Adult , Aged , Carcinoma, Hepatocellular/genetics , Female , Gene Expression , Humans , In Situ Hybridization , Liver Neoplasms/genetics , Male , Middle Aged , RNA Probes , Receptors, Androgen/genetics
13.
J Biol Chem ; 269(17): 12846-51, 1994 Apr 29.
Article in English | MEDLINE | ID: mdl-8175699

ABSTRACT

The c-MET proto-oncogene encodes the tyrosine kinase receptor for hepatocyte growth factor (HGF), also known as scatter factor, a potent mitogen and motogen for epithelial cells. The level of the HGF receptor expressed by epithelial cells varies in different growth conditions, being lower in growth arrested confluent monolayers and higher in growing sparse cells. The amount of HGF receptor mRNA increases from 3- to 5-fold after stimulation of confluent monolayers by serum and up to 10-fold after stimulation of protein kinase C by 12-O-tetradecanoylphorbol-13-acetate (TPA). An increased level of the receptor mRNA was also observed after cell stimulation with nanomolar concentration of HGF itself. The effect was transient, dose, and time-dependent. Transcription of a reporter gene under control of the cloned 297 base pair c-MET promoter was also stimulated by serum, TPA, or HGF. The accumulation of specific mRNA is followed by appearance of the HGF receptor precursor protein, which is further processed to the receptor mature form. After HGF stimulation, HGF receptor expression follows c-FOS and c-JUN induction with a peak approximately 4 h. Pretreatment with the protein synthesis inhibitor puromycin strongly reduced the response to HGF, while cycloheximide alone increased the level of the receptor mRNA. These data show that c-MET behaves as a delayed early-response gene and suggest that the HGF response is autoamplified by inducing the specific receptor.


Subject(s)
Hepatocyte Growth Factor/pharmacology , Receptor Protein-Tyrosine Kinases/biosynthesis , Blood , Cell Division , Epithelium/metabolism , Genes, Immediate-Early , Humans , Promoter Regions, Genetic , Protein Biosynthesis , Protein Processing, Post-Translational , Proto-Oncogene Mas , Proto-Oncogene Proteins c-met , RNA, Messenger/metabolism , Receptor Protein-Tyrosine Kinases/drug effects , Receptor Protein-Tyrosine Kinases/genetics , Tetradecanoylphorbol Acetate/pharmacology , Tumor Cells, Cultured
14.
Stem Cells ; 11 Suppl 2: 22-30, 1993 Jul.
Article in English | MEDLINE | ID: mdl-8401259

ABSTRACT

Hepatocyte growth factor/scatter factor (HGF/SF) is secreted by cells of mesodermal origin and shows powerful mitogenic, motogenic and morphogenic activities on epithelial and endothelial cells. It is a heparin-binding polypeptide with an alpha/beta heterodimeric structure, showing structural homologies with enzymes of the blood clotting cascade. HGF binds with high affinity to the receptor encoded by the MET protooncogene (p190MET). The MET receptor is a heterodimer of two disulfide-linked subunits (alpha and beta); the alpha subunit is extracellular, while the beta is transmembrane and endowed with tyrosine kinase activity. The HGF-triggered signalling is mediated by different cytoplasmic effectors, including phosphatidylinositol 3-kinase, phospholipase C-gamma, and Src-related tyrosine kinases. p190MET is expressed in several normal epithelial tissues (e.g., liver, gastrointestinal tract, kidney) and is often overexpressed in neoplastic cells. p190MET expression has been reported also in central nervous system microglia, a monocyte-derived cell population. We recently found that p190MET is expressed in selected peripheral blood cell populations, such as macrophages. The amount of both mRNA and protein is barely detectable, while it is dramatically increased upon activation. These findings suggest that HGF may play a role in hemopoietic cell signaling, during activation and differentiation of blood cell lineages.


Subject(s)
Hepatocyte Growth Factor/physiology , Protein-Tyrosine Kinases/physiology , Proto-Oncogene Proteins/physiology , Receptor Protein-Tyrosine Kinases/physiology , Receptors, Cell Surface/physiology , Animals , Carcinoma/metabolism , Cell Division , Cell Movement , Epithelial Cells , Epithelium/metabolism , Gene Expression Regulation , Genes , Hepatocyte Growth Factor/genetics , Hepatocyte Growth Factor/metabolism , Humans , Macrophages/metabolism , Mice , Mitosis , Monocytes/metabolism , Neoplasm Proteins/metabolism , Neuroglia/metabolism , Organ Specificity , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-met , Proto-Oncogenes , Rabbits , Rats , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Signal Transduction
15.
Proc Natl Acad Sci U S A ; 90(2): 649-53, 1993 Jan 15.
Article in English | MEDLINE | ID: mdl-8380644

ABSTRACT

The MET protooncogene encodes p190MET, a tyrosine kinase which is the receptor for a molecule known as scatter factor or hepatocyte growth factor (SF/HGF). This molecule has different biological activities, including stimulation of cell motility, promotion of matrix invasion and, in some cells, mitogenesis. We have cloned the full-length MET cDNA and transfected it into NIH 3T3 fibroblasts. Stable transfectants expressed the p190MET receptor together with two previously described truncated forms of 140 and 130 kDa lacking the tyrosine kinase domain. All three forms bound radiolabeled SF/HGF. The factor stimulated tyrosine kinase activity of the transfected p190MET and induced changes in cell shape, migration in Boyden chambers, and invasion of collagen matrices in vitro. The motile and invasive phenotype was transient and strictly dependent on the presence of SF/HGF. The factor did not stimulate either cell growth or thymidine incorporation in transfected cells, while it promoted colony formation in soft agar in the presence of 5% fetal calf serum. These data show that, in the presence of its ligand, the MET receptor expressed in fibroblasts induces cells to pursue a motogenic-invasive rather than a proliferative program.


Subject(s)
Cell Movement/genetics , Hepatocyte Growth Factor/metabolism , Proto-Oncogene Proteins/genetics , Receptors, Cell Surface/genetics , 3T3 Cells/drug effects , Animals , Cell Division/drug effects , Chemotaxis/genetics , Cloning, Molecular , Hepatocyte Growth Factor/pharmacology , Humans , Mice , Protein Kinases/metabolism , Proto-Oncogene Proteins c-met , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...