Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
J Transl Med ; 15(1): 47, 2017 02 23.
Article in English | MEDLINE | ID: mdl-28231806

ABSTRACT

BACKGROUND: Imatinib causes increased turnover of stromal collagen, reduces collagen fibril diameter, enhances extracellular fluid turnover and lowers interstitial fluid pressure (IFP) in the human colonic carcinoma KAT-4/HT-29 (KAT-4) xenograft model. METHODS: We compared the effects of imatinib on oxygen levels, vascular morphology and IFP in three experimental tumor models differing in their content of a collagenous extracellular matrix. RESULTS: Neither the KAT4 and CT-26 colonic carcinoma models, nor B16BB melanoma expressed PDGF ß-receptors in the malignant cells. KAT-4 tumors exhibited a well-developed ECM in contrast to the other two model systems. The collagen content was substantially higher in KAT-4 than in CT-26, while collagen was not detectable in B16BB tumors. The pO2 was on average 5.4, 13.9 and 19.3 mmHg in KAT-4, CT-26 and B16BB tumors, respectively. Treatment with imatinib resulted in similar pO2-levels in all three tumor models but only in KAT-4 tumors did the increase reach statistical significance. It is likely that after imatinib treatment the increase in pO2 in KAT-4 tumors is caused by increased blood flow due to reduced vascular resistance. This notion is supported by the significant reduction observed in IFP in KAT-4 tumors after imatinib treatment. Vessel area varied between 4.5 and 7% in the three tumor models and was not affected by imatinib treatment. Imatinib had no effect on the fraction of proliferating cells, whereas the fraction of apoptotic cells increased to a similar degree in all three tumor models. CONCLUSION: Our data suggest that the effects of imatinib on pO2-levels depend on a well-developed ECM and provide further support to the suggestion that imatinib acts by causing interstitial stroma cells to produce a less dense ECM, which would in turn allow for an increased blood flow. The potential of imatinib treatment to render solid tumors more accessible to conventional treatments would therefore depend on the degree of tumor desmoplasia.


Subject(s)
Colonic Neoplasms/metabolism , Extracellular Matrix/metabolism , Imatinib Mesylate/pharmacology , Neoplasms, Experimental/metabolism , Oxygen/pharmacology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Collagen/metabolism , Colonic Neoplasms/blood supply , Colonic Neoplasms/pathology , Extracellular Fluid/drug effects , Extracellular Fluid/metabolism , Extracellular Matrix/drug effects , Mice, SCID , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/pathology , Pressure , Receptor, Platelet-Derived Growth Factor beta/metabolism , Stromal Cells/metabolism , Tumor Burden/drug effects , Water
2.
J Cell Sci ; 130(3): 577-589, 2017 02 01.
Article in English | MEDLINE | ID: mdl-27980069

ABSTRACT

Platelet-derived growth factor receptor ß (PDGFRß) is a receptor tyrosine kinase which upon activation by PDGF-BB stimulates cell proliferation, migration and angiogenesis. Ligand binding induces intracellular signaling cascades but also internalization of the receptor, eventually resulting in its lysosomal degradation. However, endocytic trafficking of receptors often modulates their downstream signaling. We previously reported that internalization of PDGFRß occurs via dynamin-dependent and -independent pathways but their further molecular determinants remained unknown. Here we show that, in human fibroblasts expressing endogenous PDGFRß and stimulated with 50 ng/ml PDGF-BB, ligand-receptor uptake proceeds via the parallel routes of clathrin-mediated endocytosis (CME) and clathrin-independent endocytosis (CIE). CME involves the canonical AP2 complex as a clathrin adaptor, while CIE requires RhoA-ROCK, Cdc42 and galectin-3, the latter indicating lectin-mediated internalization via clathrin-independent carriers (CLICs). Although different uptake routes appear to be partly interdependent, they cannot fully substitute for each other. Strikingly, inhibition of any internalization mechanism impaired activation of STAT3 but not of other downstream effectors of PDGFRß. Our data indicate that multiple routes of internalization of PDGFRß contribute to a transcriptional and mitogenic response of cells to PDGF.


Subject(s)
Endocytosis/drug effects , Platelet-Derived Growth Factor/pharmacology , Receptor, Platelet-Derived Growth Factor beta/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Adaptor Protein Complex 2/metabolism , Clathrin/metabolism , DNA/biosynthesis , Dynamins/metabolism , Endocytosis/genetics , Galectin 3/metabolism , Gene Expression Regulation/drug effects , Humans , Hyaluronan Receptors/metabolism , Male , Signal Transduction/genetics , Transcription, Genetic/drug effects , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/metabolism , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism
3.
J Cell Sci ; 129(15): 2962-71, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27352860

ABSTRACT

Focal adhesions are complex multi-molecular structures that link the actin cytoskeleton to the extracellular matrix through integrin adhesion receptors and play a key role in regulation of many cellular functions. LAR (also known as PTPRF) is a receptor protein tyrosine phosphatase that regulates PDGF signalling and localises to focal adhesions. We have observed that loss of LAR phosphatase activity in mouse embryonic fibroblasts results in reduced numbers of focal adhesions and decreased adhesion to fibronectin. To understand how LAR regulates cell adhesion we used phosphoproteomic data, comparing global phosphorylation events in wild-type and LAR phosphatase-deficient cells, to analyse differential kinase activity. Kinase prediction analysis of LAR-regulated phosphosites identified a node of cytoskeleton- and adhesion-related proteins centred on cyclin-dependent kinase-1 (CDK1). We found that loss of LAR activity resulted in reduced activity of CDK1, and that CDK1 activity was required for LAR-mediated focal adhesion complex formation. We also established that LAR regulates CDK1 activity through c-Abl and Akt family proteins. In summary, we have identified a new role for a receptor protein tyrosine phosphatase in regulating CDK1 activity and hence cell adhesion to the extracellular matrix.


Subject(s)
CDC2 Protein Kinase/metabolism , Focal Adhesions/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 2/metabolism , Animals , Cell Adhesion/drug effects , Fibronectins/pharmacology , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Focal Adhesions/drug effects , Mice , Models, Biological , Phosphorylation/drug effects , Proto-Oncogene Proteins c-abl/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects
4.
Mol Cell Proteomics ; 15(6): 1823-36, 2016 06.
Article in English | MEDLINE | ID: mdl-27074791

ABSTRACT

Intracellular signaling pathways are reliant on protein phosphorylation events that are controlled by a balance of kinase and phosphatase activity. Although kinases have been extensively studied, the role of phosphatases in controlling specific cell signaling pathways has been less so. Leukocyte common antigen-related protein (LAR) is a member of the LAR subfamily of receptor-like protein tyrosine phosphatases (RPTPs). LAR is known to regulate the activity of a number of receptor tyrosine kinases, including platelet-derived growth factor receptor (PDGFR). To gain insight into the signaling pathways regulated by LAR, including those that are PDGF-dependent, we have carried out the first systematic analysis of LAR-regulated signal transduction using SILAC-based quantitative proteomic and phosphoproteomic techniques. We haveanalyzed differential phosphorylation between wild-type mouse embryo fibroblasts (MEFs) and MEFs in which the LAR cytoplasmic phosphatase domains had been deleted (LARΔP), and found a significant change in abundance of phosphorylation on 270 phosphosites from 205 proteins because of the absence of the phosphatase domains of LAR. Further investigation of specific LAR-dependent phosphorylation sites and enriched biological processes reveal that LAR phosphatase activity impacts on a variety of cellular processes, most notably regulation of the actin cytoskeleton. Analysis of putative upstream kinases that may play an intermediary role between LAR and the identified LAR-dependent phosphorylation events has revealed a role for LAR in regulating mTOR and JNK signaling.


Subject(s)
MAP Kinase Signaling System , Platelet-Derived Growth Factor/metabolism , Proteomics/methods , Receptor-Like Protein Tyrosine Phosphatases, Class 2/genetics , TOR Serine-Threonine Kinases/metabolism , Animals , Cells, Cultured , Isotope Labeling , Mass Spectrometry , Mice , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Phosphorylation , Receptor-Like Protein Tyrosine Phosphatases, Class 2/metabolism , Sequence Deletion , Signal Transduction
5.
Methods Enzymol ; 535: 167-77, 2014.
Article in English | MEDLINE | ID: mdl-24377924

ABSTRACT

Microscopical analyses of endocytic trafficking require tools for efficient detection of internalized cargo. Due to the lack of suitable reagents and limitations related to its biological properties, visualization of platelet-derived growth factor (PDGF) by microscopy remained a challenge. To overcome these restrictions, we generated a biologically active PDGF labeled with up to five biotins on cleavable linkers. Subsequently, we stimulated cells with such ligand followed by removal of extracellular biotins. PDGF captured in endocytic vesicles was successfully detected with antibiotin antibodies with parallel detection of PDGF receptor, as well as other markers of endocytic compartments. Labeled PDGF was successfully validated and can be utilized in various microscopical techniques.


Subject(s)
Endocytosis , Platelet-Derived Growth Factor/metabolism , Biotinylation , Cell Line , Humans , Microscopy, Fluorescence , Platelet-Derived Growth Factor/chemistry , Receptors, Platelet-Derived Growth Factor/metabolism , Staining and Labeling
6.
Proc Natl Acad Sci U S A ; 110(33): 13398-403, 2013 Aug 13.
Article in English | MEDLINE | ID: mdl-23901112

ABSTRACT

The inhibitory reversible oxidation of protein tyrosine phosphatases (PTPs) is an important regulatory mechanism in growth factor signaling. Studies on PTP oxidation have focused on pathways that increase or decrease reactive oxygen species levels and thereby affect PTP oxidation. The processes involved in reactivation of oxidized PTPs remain largely unknown. Here the role of the thioredoxin (Trx) system in reactivation of oxidized PTPs was analyzed using a combination of in vitro and cell-based assays. Cells lacking the major Trx reductase TrxR1 (Txnrd1(-/-)) displayed increased oxidation of PTP1B, whereas SHP2 oxidation was unchanged. Furthermore, in vivo-oxidized PTP1B was reduced by exogenously added Trx system components, whereas SHP2 oxidation remained unchanged. Trx1 reduced oxidized PTP1B in vitro but failed to reactivate oxidized SHP2. Interestingly, the alternative TrxR1 substrate TRP14 also reactivated oxidized PTP1B, but not SHP2. Txnrd1-depleted cells displayed increased phosphorylation of PDGF-ß receptor, and an enhanced mitogenic response, after PDGF-BB stimulation. The TrxR inhibitor auranofin also increased PDGF-ß receptor phosphorylation. This effect was not observed in cells specifically lacking PTP1B. Together these results demonstrate that the Trx system, including both Trx1 and TRP14, impacts differentially on the oxidation of individual PTPs, with a preference of PTP1B over SHP2 activation. The studies demonstrate a previously unrecognized pathway for selective redox-regulated control of receptor tyrosine kinase signaling.


Subject(s)
Enzyme Activation/physiology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Protein-Tyrosine Kinases/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , Signal Transduction/physiology , Thioredoxins/pharmacology , Animals , Enzyme Activation/drug effects , Fibroblasts , Gene Knockout Techniques , Gentian Violet , Mice , Oxidation-Reduction , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Reactive Oxygen Species/metabolism , Thioredoxin Reductase 1/deficiency
7.
J Biol Chem ; 288(22): 15736-44, 2013 May 31.
Article in English | MEDLINE | ID: mdl-23589302

ABSTRACT

Fer is a cytoplasmic tyrosine kinase that is activated in response to platelet-derived growth factor (PDGF) stimulation. In the present report, we show that Fer associates with the activated PDGF ß-receptor (PDGFRß) through multiple autophosphorylation sites, i.e. Tyr-579, Tyr-581, Tyr-740, and Tyr-1021. Using low molecular weight inhibitors, we found that PDGF-BB-induced Fer activation is dependent on PDGFRß kinase activity, but not on the enzymatic activity of Src or Jak kinases. In cells in which Fer was down-regulated using siRNA, PDGF-BB was unable to induce phosphorylation of STAT3, whereas phosphorylations of STAT5, ERK1/2, and Akt were unaffected. PDGF-BB-induced activation of STAT3 occurred also in cells expressing kinase-dead Fer, suggesting a kinase-independent adaptor role of Fer. Expression of Fer was dispensable for PDGF-BB-induced proliferation and migration but essential for colony formation in soft agar. Tumor growth in vivo was delayed in cells depleted of Fer expression. Our data suggest a critical role of Fer in PDGF-BB-induced STAT3 activation and cell transformation.


Subject(s)
Angiogenesis Inducing Agents/pharmacology , Cell Movement/drug effects , Cell Proliferation/drug effects , Neoplasms/metabolism , Protein-Tyrosine Kinases/biosynthesis , Proto-Oncogene Proteins c-sis/pharmacology , STAT3 Transcription Factor/metabolism , Animals , Becaplermin , Cell Line, Tumor , Cell Movement/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Enzyme Activation/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Humans , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , Mice , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , NIH 3T3 Cells , Neoplasms/genetics , Neoplasms/pathology , Phosphorylation/drug effects , Phosphorylation/genetics , Protein-Tyrosine Kinases/genetics , Receptor, Platelet-Derived Growth Factor beta/genetics , Receptor, Platelet-Derived Growth Factor beta/metabolism , STAT3 Transcription Factor/genetics , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/metabolism , src-Family Kinases/genetics , src-Family Kinases/metabolism
8.
Nat Commun ; 4: 1672, 2013.
Article in English | MEDLINE | ID: mdl-23575676

ABSTRACT

Vascular endothelial growth factor (VEGF) guides the path of new vessel sprouts by inducing VEGF receptor-2 activity in the sprout tip. In the stalk cells of the sprout, VEGF receptor-2 activity is downregulated. Here, we show that VEGF receptor-2 in stalk cells is dephosphorylated by the endothelium-specific vascular endothelial-phosphotyrosine phosphatase (VE-PTP). VE-PTP acts on VEGF receptor-2 located in endothelial junctions indirectly, via the Angiopoietin-1 receptor Tie2. VE-PTP inactivation in mouse embryoid bodies leads to excess VEGF receptor-2 activity in stalk cells, increased tyrosine phosphorylation of VE-cadherin and loss of cell polarity and lumen formation. Vessels in ve-ptp(-/-) teratomas also show increased VEGF receptor-2 activity and loss of endothelial polarization. Moreover, the zebrafish VE-PTP orthologue ptp-rb is essential for polarization and lumen formation in intersomitic vessels. We conclude that the role of Tie2 in maintenance of vascular quiescence involves VE-PTP-dependent dephosphorylation of VEGF receptor-2, and that VEGF receptor-2 activity regulates VE-cadherin tyrosine phosphorylation, endothelial cell polarity and lumen formation.


Subject(s)
Cell Polarity , Endothelium, Vascular/cytology , Receptor-Like Protein Tyrosine Phosphatases, Class 3/metabolism , Vascular Endothelial Growth Factor Receptor-2/physiology , Animals , Endothelium, Vascular/enzymology , Endothelium, Vascular/metabolism , Intercellular Junctions , Mice , Phosphorylation , Receptor, TIE-2/metabolism
9.
Traffic ; 14(6): 725-36, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23425318

ABSTRACT

Platelet-derived growth factor (PDGF) isoforms regulate cell proliferation, migration and differentiation both in embryonic development and adult tissue remodeling. At the cellular level, growth-factor signaling is often modulated by endocytosis. Despite important functions of PDGF, its endocytosis remains poorly studied, mainly for lack of tools to track internalized ligand by microscopy. Here, we developed such a tool and quantitatively analyzed internalization and endosomal trafficking of PDGF-BB in human fibroblasts. We further show that PDGF can be internalized in the presence of dynamin inhibitors, arguing that both dynamin-dependent and dynamin-independent pathways can mediate PDGF uptake. Although these routes operate with somewhat different kinetics, they both ultimately lead to lysosomal degradation of PDGF. Although acute inhibition of dynamin activity only moderately affects PDGF endocytosis, it specifically decreases downstream signaling of PDGF via signal transducer and activator of transcription 3 (STAT3). This correlates with reduced expression of MYC and impaired cell entry into S-phase, indicating that dynamin activity is required for PDGF-induced mitogenesis. Our data support a general view that the components governing endocytic trafficking may selectively regulate certain signaling effectors activated by a growth factor.


Subject(s)
Dynamins/antagonists & inhibitors , Endocytosis , MAP Kinase Signaling System , Platelet-Derived Growth Factor/metabolism , Cell Line , Fibroblasts/metabolism , Humans , Lysosomes/metabolism , Protein Transport , Proteolysis , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , S Phase/drug effects , STAT3 Transcription Factor/metabolism , Transcription, Genetic
10.
Cell Signal ; 23(6): 1050-6, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21300149

ABSTRACT

The receptor tyrosine phosphatase (RPTP) LAR negatively regulates the activity of several receptor tyrosine kinases. To investigate if LAR affects the platelet-derived growth factor (PDGF) receptor signaling, mouse embryonic fibroblasts (MEFs) from mice where the LAR phosphatase domains were deleted (LARΔP), and wt littermates, were stimulated with 20ng/ml PDGF-BB. In LAR phosphatase deficient MEFs, the phosphorylation of the PDGF ß-receptor was surprisingly reduced, an event that was rescued by re-expression of wt LAR. The decreased phosphorylation of the PDGF ß-receptor was observed independent of ligand concentration and occurred on all tyrosine residues, as determined by immunoblotting analysis using site-selective phosphotyrosine antibodies. This suggests that LAR is required for full PDGF ß-receptor kinase activation. Downstream of receptor activation, phosphorylation of Akt and PLCγ were decreased in LARΔP MEFs, whereas Src and Erk MAP kinase pathways were less affected. The proliferation of LARΔP MEFs in response to PDGF-BB was also reduced. The inhibitory effect on the PDGF ß-receptor in LARΔP cells was exerted via increased basal activity of c-Abl, since inhibition of c-Abl, by AG957 or siRNA, restored PDGF ß-receptor phosphorylation. These observations suggest that LAR reduces the basal c-Abl activity thereby allowing for PDGF ß-receptor kinase activation.


Subject(s)
Proto-Oncogene Proteins c-abl/antagonists & inhibitors , Receptor, Platelet-Derived Growth Factor beta/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 2/metabolism , Animals , Becaplermin , Cells, Cultured , Enzyme Activation , Fibroblasts , Gene Deletion , Humans , Mice , Phosphorylation , Platelet-Derived Growth Factor/physiology , Protein Structure, Tertiary , Proto-Oncogene Proteins c-abl/metabolism , Proto-Oncogene Proteins c-sis , Receptor-Like Protein Tyrosine Phosphatases, Class 2/genetics , Signal Transduction , src-Family Kinases/metabolism
11.
Melanoma Res ; 21(1): 57-65, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20975605

ABSTRACT

Melanomas respond poorly to chemotherapy. In this study, we investigated the sensitization of B16 mouse melanoma tumors to paclitaxel by a combination of two tyrosine kinase inhibitors: vatalanib, targeting vascular endothelial growth factor receptors, and imatinib, an inhibitor targeting for example, Abl/BCR-ABL, the platelet-derived growth factor receptor, and stem cell factor receptor c-Kit. C57Bl6/J mice carrying B16/PDGF-BB mouse melanoma tumors were treated daily with vatalanib (25 mg/kg), imatinib (100 mg/kg), or a combination of these drugs. Paclitaxel was given subcutaneously twice during the study. The effects of the drugs on tumor cell proliferation in vitro were determined by counting cells. B16/PDGF-BB mouse melanoma tumors were not sensitive to paclitaxel at doses of either 5 or 20 mg/kg. However, the tumor growth was significantly reduced by 58%, in response to paclitaxel (5 mg/kg) when administered with daily doses of both vatalanib and imatinib. Paclitaxel only inhibited the in-vitro growth of B16/PDGF-BB tumor cells when given in combination with imatinib. Imatinib presumably targets c-Kit, as the cells do not express platelet-derived growth factor receptor and as another c-Abl inhibitor was without effect. This was supported by data from three c-Kit-expressing human melanoma cell lines showing varying sensitization to paclitaxel by the kinase inhibitors. In addition, small interfering RNA knockdown of c-Kit sensitized the cells to paclitaxel. These data show that combination of two tyrosine kinase inhibitors, imatinib and vatalanib, increases the effects of paclitaxel on B16/PDGF-BB tumors, thus suggesting a novel strategy for the treatment of melanomas expressing c-Kit.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Benzamides/administration & dosage , Melanoma, Experimental/drug therapy , Paclitaxel/administration & dosage , Phthalazines/administration & dosage , Piperazines/administration & dosage , Pyridines/administration & dosage , Pyrimidines/administration & dosage , Angiogenesis Inhibitors/administration & dosage , Animals , Becaplermin , Cell Line, Tumor , Cell Proliferation , Drug Synergism , Imatinib Mesylate , Mice , Mice, Inbred C57BL , Protein-Tyrosine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins c-kit/metabolism , Proto-Oncogene Proteins c-sis/metabolism , RNA, Small Interfering/metabolism , Signal Transduction
12.
J Biol Chem ; 285(16): 12268-78, 2010 Apr 16.
Article in English | MEDLINE | ID: mdl-20164181

ABSTRACT

The fusion of TEL with platelet-derived growth factor receptor (PDGFR) beta (TPbeta) is found in a subset of patients with atypical myeloid neoplasms associated with eosinophilia and is the archetype of a larger group of hybrid receptors that are produced by rearrangements of PDGFR genes. TPbeta is activated by oligomerization mediated by the pointed domain of TEL/ETV6, leading to constitutive activation of the PDGFRbeta kinase domain. The receptor transmembrane (TM) domain is retained in TPbeta and in most of the described PDGFRbeta hybrids. Deletion of the TM domain (DeltaTM-TPbeta) strongly impaired the ability of TPbeta to sustain growth factor-independent cell proliferation. We confirmed that TPbeta resides in the cytosol, indicating that the PDGFRbeta TM domain does not act as a transmembrane domain in the context of the hybrid receptor but has a completely different function. The DeltaTM-TPbeta protein was expressed at a lower level because of increased degradation. It could form oligomers, was phosphorylated at a slightly higher level, co-immunoprecipitated with the p85 adaptor protein, but showed a much reduced capacity to activate STAT5 and ERK1/2 in Ba/F3 cells, compared with TPbeta. In an in vitro kinase assay, DeltaTM-TPbeta was more active than TPbeta and less sensitive to imatinib, a PDGFR inhibitor. In conclusion, we show that the TM domain is required for TPbeta-mediated signaling and proliferation, suggesting that the activation of the PDGFRbeta kinase domain is not enough for cell transformation.


Subject(s)
Oncogene Proteins, Fusion/chemistry , Oncogene Proteins, Fusion/metabolism , Amino Acid Sequence , Animals , Cell Line , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cytosol/metabolism , Humans , Leukemia, Myeloid/etiology , Leukemia, Myeloid/genetics , Leukemia, Myeloid/metabolism , Molecular Sequence Data , Myeloid Cells/metabolism , Myeloid Cells/pathology , Oncogene Proteins, Fusion/genetics , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Deletion , Signal Transduction , Swine
13.
Recent Results Cancer Res ; 180: 103-14, 2010.
Article in English | MEDLINE | ID: mdl-20033380

ABSTRACT

Pericytes are smooth muscle-like cells found in close contact with the endothelium in capillaries, where they regulate the morphology and function of the vessels. During vessel formation, platelet-derived growth factor-BB (PDGF-BB) is required for the recruitment and differentiation of pericytes. Tumor vessels display abnormal morphology and increased endothelial proliferation, resulting in leaky, tortuous vessels that are often poorly perfused. These vessels typically display decreased pericyte density, and the tumor-associated pericytes often express abnormal markers and show abnormal morphology. Anti-angiogenic therapy targeting pro-angiogenic growth factor pathways has been applied to a broad range of solid tumors with varying results. Studies utilizing mouse models indicate that the presence of pericytes protect endothelial cells against inhibition of vascular endothelial growth factor (VEGF) signaling. Simultaneous inhibition of PDGF receptors on pericytes therefore improves the effect of VEGF inhibitors on endothelial cells and enhances anti-angiogenic therapy.


Subject(s)
Neovascularization, Physiologic/physiology , Platelet-Derived Growth Factor/physiology , Angiogenesis Inhibitors/therapeutic use , Animals , Humans , Neoplasms/blood supply , Neoplasms/drug therapy , Pericytes/drug effects , Pericytes/physiology , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors , Receptors, Platelet-Derived Growth Factor/physiology
14.
PLoS One ; 4(12): e8149, 2009 Dec 04.
Article in English | MEDLINE | ID: mdl-19997591

ABSTRACT

Elevation of the interstitial fluid pressure (IFP) of carcinoma is an obstacle in treatment of tumors by chemotherapy and correlates with poor drug uptake. Previous studies have shown that treatment with inhibitors of platelet-derived growth factor (PDGF) or vascular endothelial growth factor (VEGF) signaling lowers the IFP of tumors and improve chemotherapy. In this study, we investigated whether the combination of PDGFR and VEGFR inhibitors could further reduce the IFP of KAT-4 human carcinoma tumors. The tumor IFP was measured using the wick-in-needle technique. The combination of STI571 and PTK/ZK gave an additive effect on the lowering of the IFP of KAT-4 tumors, but the timing of the treatment was crucial. The lowering of IFP following combination therapy was accompanied by vascular remodeling and decreased vascular leakiness. The effects of the inhibitors on the therapeutic efficiency of Taxol were investigated. Whereas the anti-PDGF and anti-VEGF treatment did not significantly inhibit tumor growth, the inhibitors enhanced the effect of chemotherapy. Despite having an additive effect in decreasing tumor IFP, the combination therapy did not further enhance the effect of chemotherapy. Simultaneous targeting of VEGFR and PDGFR kinase activity may be a useful strategy to decrease tumor IFP, but the timing of the inhibitors should be carefully determined.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Extracellular Fluid/drug effects , Neoplasms, Experimental/drug therapy , Pressure , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Animals , Apoptosis/drug effects , Blood Volume/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Synergism , Humans , Mice , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/pathology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Pericytes/drug effects , Pericytes/pathology , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use
15.
Mol Biol Cell ; 20(12): 2856-63, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19369415

ABSTRACT

Previous studies showed that loss of the T-cell protein tyrosine phosphatase (TC-PTP) induces Rab4a-dependent recycling of the platelet-derived growth factor (PDGF) beta-receptor in mouse embryonic fibroblasts (MEFs). Here we identify protein kinase C (PKC) alpha as the critical signaling component that regulates the sorting of the PDGF beta-receptor at the early endosomes. Down-regulation of PKC abrogated receptor recycling by preventing the sorting of the activated receptor into EGFP-Rab4a positive domains on the early endosomes. This effect was mimicked by inhibition of PKCalpha, using myristoylated inhibitory peptides or by knockdown of PKCalpha with shRNAi. In wt MEFs, short-term preactivation of PKC by PMA caused a ligand-induced PDGF beta-receptor recycling that was dependent on Rab4a function. Together, these observations demonstrate that PKC activity is necessary for recycling of ligand-stimulated PDGF beta-receptor to occur. The sorting also required Rab4a function as it was prevented by expression of EGFP-Rab4aS22N. Preventing receptor sorting into recycling endosomes increased the rate of receptor degradation, indicating that the sorting of activated receptors at early endosomes directly regulates the duration of receptor signaling. Activation of PKC through the LPA receptor also induced PDGF beta-receptor recycling and potentiated the chemotactic response to PDGF-BB. Taken together, our present findings indicate that sorting of PDGF beta-receptors on early endosomes is regulated by sequential activation of PKCalpha and Rab4a and that this sorting step could constitute a point of cross-talk with other receptors.


Subject(s)
Endocytosis , Protein Kinase C-alpha/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , rab4 GTP-Binding Proteins/metabolism , Animals , Becaplermin , Cell Movement/drug effects , Endocytosis/drug effects , Endosomes/drug effects , Endosomes/enzymology , Enzyme Activation/drug effects , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/enzymology , HeLa Cells , Humans , Lysophospholipids/pharmacology , Mice , Microscopy, Confocal , Platelet-Derived Growth Factor/pharmacology , Protein Processing, Post-Translational/drug effects , Protein Transport/drug effects , Proto-Oncogene Proteins c-sis , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism
16.
Int J Cancer ; 121(12): 2606-14, 2007 Dec 15.
Article in English | MEDLINE | ID: mdl-17691110

ABSTRACT

The aim of our study was to further explore the use of anti-angiogenic therapy targeting the vascular endothelial growth factor receptor (VEGFR) on endothelial cells while simultaneously targeting platelet-derived growth factor receptors (PDGFRs) on adjacent pericytes. B16 mouse melanoma tumors exogenously expressing PDGF-BB (B16/PDGF-BB) display higher pericyte coverage on the vasculature compared to the parental B16 tumors (B16/mock). These models were used to investigate the effects of combination therapy targeting VEGFR and PDGFR signaling on size-matched tumors. Combination therapy using 25 mg/kg/day of the VEGFR inhibitor PTK787 and 100 mg/kg/day of the PDGFR inhibitor STI571 decreased the tumor growth rate of both tumor types, but the inhibition was only significant in the B16/PDGF-BB tumors. Combination therapy induced vessel remodeling, primarily by reducing the vessel density in B16/mock tumors, and by reducing the vessel size in B16/PDGF-BB tumors. When analyzing the effects of combination therapy on tumor vessel pericytes, it was found to primarily reduce the subpopulation of alpha-smooth muscle actin and PDGFRbeta-positive pericytes partly detached from the tumor vessels, without affecting the number of pericytes closely attached to the endothelium, which also express desmin. Taken together, these data demonstrate an increased benefit of targeting both VEGFR and PDGFR pathways in B16/PDGF-BB tumors, and demonstrates that the increased tumor growth inhibition in this model is accompanied by a reduction in a specific subset of pericytes, characterized by being loosely attached to endothelial cells and negative for the pericyte marker desmin.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Melanoma, Experimental/drug therapy , Melanoma, Experimental/metabolism , Pericytes/drug effects , Platelet-Derived Growth Factor/metabolism , Signal Transduction/drug effects , Vascular Endothelial Growth Factor A/metabolism , Angiogenesis Inducing Agents/metabolism , Animals , Becaplermin , Benzamides , Imatinib Mesylate , Immunohistochemistry , Mice , Pericytes/metabolism , Phthalazines/pharmacology , Piperazines/pharmacology , Platelet-Derived Growth Factor/drug effects , Proto-Oncogene Proteins c-sis , Pyridines/pharmacology , Pyrimidines/pharmacology , Receptor, Platelet-Derived Growth Factor beta/metabolism , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A/drug effects
17.
Int J Dev Biol ; 51(5): 361-70, 2007.
Article in English | MEDLINE | ID: mdl-17616925

ABSTRACT

An oncogenic D842V mutation in the platelet-derived growth factor (PDGF) alpha-receptor (Pdgfra) has recently been described in patients with gastrointestinal stromal tumors. In order to test if the same mutation would confer oncogenic properties to the homologous PDGF beta-receptor (Pdgfrb), the corresponding aspartic acid residue at position 849 of Pdgfrb was changed into valine (D849V) using a knock-in strategy. This mutation turned out to be dominantly lethal and caused death even in chimeras (from 345 transferred chimeric blastocysts, no living coat chimeras were detected). Experiments employing mouse embryonic fibroblasts (MEFs) indicated hyperactivity of the mutant receptor. The mutant receptor was phosphorylated in a ligand-independent manner and, in contrast to wild-type MEFs, mutant cells proliferated even in the absence of ligand. Knockout experiments have previously indicated a role for Pdgfrb in placental development. We therefore analyzed wild-type and Pdgfrb D849V chimeric placentas from different gestational stages. No differences were detected at embryonic days 11.5 and 13.5 (n=4). At embryonic day 17.5, however, chimeric placentas (n=3/4) displayed abnormalities both in the labyrinth and in the chorionic plate. The changes included hyper-proliferation of alpha-smooth muscle actin and platelet/endothelial cell adhesion molecule-1 positive cells in the labyrinth and cells in the chorionic plate. In addition, the fetal blood vessel compartment of the labyrinth was completely disorganized.


Subject(s)
Placenta/abnormalities , Placenta/enzymology , Receptor, Platelet-Derived Growth Factor beta/genetics , Receptor, Platelet-Derived Growth Factor beta/metabolism , Animals , Aspartic Acid/genetics , Aspartic Acid/metabolism , Cell Proliferation , Cell Shape , Cells, Cultured , Embryo Loss , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Female , Fibroblasts , Ligands , Mice , Mice, Transgenic , Mutant Chimeric Proteins/genetics , Mutant Chimeric Proteins/metabolism , Mutation/genetics , Phosphorylation , Placenta/blood supply , Pregnancy
18.
FASEB J ; 21(2): 523-34, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17158785

ABSTRACT

Growth factor-dependent tissue remodeling, such as restenosis, is believed to be predominantly regulated by changes in expression of receptor-tyrosine-kinases (RTKs) and their ligands. As endogenous antagonists of RTKs, protein-tyrosine-phosphatases (PTPs) are additional candidate regulators of these processes. Using laser-capture-microdissection and quantitative RT-polymerase chain reaction (qRT-PCR), we investigated the layer-specific expression of the four platelet-derived growth factor (PDGF) isoforms, the PDGF-alpha and beta receptors, and five PTPs implied in control of PDGF-receptor signaling 8 and 14 days after balloon injury of the rat carotid. Results were correlated with analyses of PDGF-beta receptor phosphorylation and vascular smooth muscle cell (VSMC) proliferation in vivo. The expression levels of all components, as well as receptor activation and VSMC proliferation, showed specific changes, which varied between media and neointima. Interestingly, PTP expression--particularly, DEP-1 levels--appeared to be the dominating factor determining receptor-phosphorylation and VSMC proliferation. In support of these findings, cultured DEP-1(-/-) cells displayed increased PDGF-dependent cell signaling. Hyperactivation of PDGF-induced signaling was also observed after siRNA-down-regulation of DEP-1 in VSMCs. The results indicate a previously unrecognized role of PDGF-receptor-targeting PTPs in controlling neointima formation. In more general terms, the observations indicate transcriptional regulation of PTPs as an important mechanism for controlling onset and termination of RTK-dependent tissue remodeling.


Subject(s)
Muscle, Smooth, Vascular/metabolism , Protein Tyrosine Phosphatases/genetics , Receptors, Platelet-Derived Growth Factor/metabolism , Tunica Intima/metabolism , Animals , Carotid Artery Injuries/physiopathology , Cell Proliferation , Chemotaxis , Enzyme-Linked Immunosorbent Assay , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression , Immunoblotting , Immunohistochemistry , Immunoprecipitation , Muscle, Smooth, Vascular/cytology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Tyrosine Phosphatases/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors , Reverse Transcriptase Polymerase Chain Reaction/methods , Time Factors , Tunica Intima/pathology
19.
Mol Biol Cell ; 17(11): 4846-55, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16971512

ABSTRACT

We have previously shown that the T-cell protein tyrosine phosphatase (TC-PTP) dephosphorylates the platelet-derived growth factor (PDGF) beta-receptor. Here, we show that the increased PDGF beta-receptor phosphorylation in TC-PTP knockout (ko) mouse embryonic fibroblasts (MEFs) occurs primarily on the cell surface. The increased phosphorylation is accompanied by a TC-PTP-dependent, monensin-sensitive delay in clearance of cell surface PDGF beta-receptors and delayed receptor degradation, suggesting PDGF beta-receptor recycling. Recycled receptors could also be directly detected on the cell surface of TC-PTP ko MEFs. The effect of TC-PTP depletion was specific for the PDGF beta-receptor, because PDGF alpha-receptor homodimers were cleared from the cell surface at the same rate in TC-PTP ko MEFs as in wild-type MEFs. Interestingly, PDGF alphabeta-receptor heterodimers were recycling. Analysis by confocal microscopy revealed that, in TC-PTP ko MEFs, activated PDGF beta-receptors colocalized with Rab4a, a marker for rapid recycling. In accordance with this, transient expression of a dominant-negative Rab4a construct increased the rate of clearance of cell surface receptors on TC-PTP ko MEFs. Thus, loss of TC-PTP specifically redirects the PDGF beta-receptor toward rapid recycling, which is the first evidence of differential trafficking of PDGF receptor family members.


Subject(s)
Protein Tyrosine Phosphatases/deficiency , Protein Tyrosine Phosphatases/metabolism , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , Animals , Cytoplasmic Vesicles/metabolism , Dimerization , Fibroblasts/cytology , Ligands , Mice , Mice, Knockout , Phosphorylation , Protein Processing, Post-Translational , Protein Transport , Protein Tyrosine Phosphatase, Non-Receptor Type 2 , Recombinant Fusion Proteins/metabolism , rab4 GTP-Binding Proteins/metabolism
20.
Nat Rev Cancer ; 6(4): 307-20, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16557282

ABSTRACT

Tyrosine phosphorylation is an important signalling mechanism in eukaryotic cells. In cancer, oncogenic activation of tyrosine kinases is a common feature, and novel anticancer drugs have been introduced that target these enzymes. Tyrosine phosphorylation is also controlled by protein-tyrosine phosphatases (PTPs). Recent evidence has shown that PTPs can function as tumour suppressors. In addition, some PTPs, including SHP2, positively regulate the signalling of growth-factor receptors, and can be oncogenic. An improved understanding of how these enzymes function and how they are regulated might aid the development of new anticancer agents.


Subject(s)
Neoplasms/enzymology , Protein Tyrosine Phosphatases/metabolism , Animals , ErbB Receptors/metabolism , Humans , Neoplasms/therapy , Phosphorylation , Protein Tyrosine Phosphatases/genetics , Tyrosine
SELECTION OF CITATIONS
SEARCH DETAIL
...