Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Clin Exp Metastasis ; 30(4): 369-79, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23076771

ABSTRACT

Osteosarcoma (OS) is the most common malignant bone tumor and the prognosis depends on pulmonary metastases, which arise from multi-step progression of malignant tumors. We herein aimed to clarify the critical step of pulmonary metastasis using the syngeneic mouse spontaneous highly metastatic OS LM8 and parental Dunn cell lines, to identify new candidate molecules to suppress pulmonary metastasis. We first investigated the chronological detection of circulating tumor cells (CTCs) from mice with either cell line. LM8 CTCs appeared faster, at a higher rate and with a greater number compared to Dunn CTCs. Cultured cells from CTCs of LM8 showed higher proliferative ability than cells from the primary site in suspension culture, which mimicked the environment of the bloodstream for CTCs. The proliferative ability of LM8 cells was also higher than that of Dunn cells in 3D collagen culture with low stiffness (-150 Pa; close to conditions in the lung). We next focused on the extravasation step. LM8 showed higher migration ability compared to Dunn with transendothelial migration assay. We also found a disruption in endothelial barrier function throughout co-culture with LM8 using time-lapse imaging. In addition, LM8 secreted high levels of vascular endothelial growth factor (VEGF), while VEGF signal inhibition with a small molecule tyrosine kinase inhibitor (pazopanib) decreased disruption of the vascular barrier and transendothelial migration of LM8. Finally, daily oral administration of pazopanib reduced the rate and size of pulmonary metastasis in vivo. Collectively, these results show anti-VEGF therapy as a candidate for pulmonary metastasis of OS.


Subject(s)
Bone Neoplasms/pathology , Lung Neoplasms/secondary , Osteosarcoma/pathology , Pyrimidines/pharmacology , Sulfonamides/pharmacology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Angiogenesis Inhibitors/pharmacology , Animals , Apoptosis/drug effects , Blotting, Western , Bone Neoplasms/drug therapy , Bone Neoplasms/metabolism , Cell Movement/drug effects , Cell Proliferation/drug effects , Coculture Techniques , Immunoenzyme Techniques , Indazoles , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Mice , Mice, Inbred C3H , Osteosarcoma/drug therapy , Osteosarcoma/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
2.
Int J Oncol ; 41(4): 1520-30, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22859339

ABSTRACT

Gefitinib is known to suppress the activation of EGFR signaling, which is required for cell survival and proliferation in non-small cell lung cancer (NSCLC) cell lines. We previously demonstrated that the gefitinib-sensitive NSCLC cell line PC9 shows efficient ligand-induced endocytosis of phosphorylated EGFR (pEGFR). In contrast, the gefitinib-resistant NSCLC cell lines QG56 and A549 showed internalized pEGFR accumulation in the aggregated early endosomes, and this was associated with SNX1, a protein that interacts with and enhances the degradation of EGFR upon EGF stimulation. In the present study, to investigate the role of SNX1 on EGF-stimulated EGFR/pEGFR endocytosis via the endocytic pathway, we examined the effect of depletion of SNX1 expression by siRNA in human NSCLC cell lines. Using immunofluorescence, we demonstrated that transfection of SNX1 siRNA into gefitinib-resistant NSCLC cells resulted in the disappearance of a large amounts of SNX1 staining. In addition, upon 15 min of EGF stimulation, we observed an efficient EGFR phosphorylation and a rapid endocytic delivery of pEGFR from early endosomes to late endosomes. Further, western blot analysis revealed that silencing of SNX1 expression by siRNA in the gefitinib-resistant cells leads to an accelerated degradation of EGFR along with a dramatic increase in the amounts of pEGFR after EGF stimulation. Based on these findings, we suggest that SNX1 is involved in the negative regulation of ligand-induced EGFR phosphorylation and mediates EGFR/pEGFR trafficking out of early endosomes for targeting to late endosomes/lysosomes via the early/late endocytic pathway in human lung cancer cells.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , ErbB Receptors/genetics , Quinazolines/administration & dosage , Sorting Nexins/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Resistance, Neoplasm/genetics , Endocytosis/drug effects , Endosomes/drug effects , Endosomes/metabolism , ErbB Receptors/biosynthesis , Gefitinib , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lysosomes/drug effects , Lysosomes/metabolism , Phosphorylation/drug effects , RNA, Small Interfering/genetics , Signal Transduction/drug effects , Sorting Nexins/antagonists & inhibitors , Sorting Nexins/biosynthesis
3.
FEBS Lett ; 585(24): 4018-24, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22100295

ABSTRACT

We showed that the migration, morphology and adhesiveness of undifferentiated mesenchymal cells dramatically changed during osteogenic differentiation. The migration of these cells was transiently upregulated early in osteogenic differentiation. At a later stage, migration was decreased but adhesiveness was increased. Furthermore, Cdc42 and Rac1 Rho-family small GTPases were activated at early stages of differentiation and the phosphorylation level of FAK decreased as differentiation progressed. We also showed cell migration was promoted by inhibition of the Rho-ROCK-myosin signaling. Finally, using a mouse model of ectopic bone formation, we confirmed that treatment with ROCK inhibitor, Y-27632 increased cell movement into bone formation sites, resulting in enhanced osteogenesis. These results provide a new insight into the link between cell migration and osteogenic differentiation.


Subject(s)
Cell Differentiation , Cell Movement , Mesenchymal Stem Cells/cytology , Osteogenesis , Animals , Bone Marrow Cells/cytology , Cell Differentiation/drug effects , Cell Line , Cell Movement/drug effects , Fracture Healing/drug effects , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/enzymology , Mice , Osteogenesis/drug effects , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/metabolism
4.
J Mol Histol ; 42(5): 427-42, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21847509

ABSTRACT

The small GTPase RhoA and its downstream effectors, the Rho-associated kinase (Rho-kinase) family, are known to regulate cell morphology, motility, and tumor progression via the regulation of actin cytoskeleton rearrangement. In the present study, we evaluated the role of Rho-kinase in the intracellular endocytic trafficking of ligand-induced phosphorylated epidermal growth factor receptor (pEGFR). We investigated the time course of the internalization fate of EGF-induced pEGFR via the early/late endocytic pathway in human fibrosarcoma cell line HT1080 cells using Y-27632, a selective Rho-kinase inhibitor. We found, using confocal immunofluorescence microscopy and Western blot analysis, a large accumulation of pEGFR in the nuclei of HT1080 cells. In contrast, we observed decreased amounts of the pEGFR-positive staining in the nuclei along with an accumulation of cytosolic pEGFR staining when the cells were incubated for 15-30 min in the presence of Y-27632, implying that an aberrant endocytic trafficking mechanism of pEGFR occurs in HT1080 cells whereby pEGFR might be selectively translocated into the nucleus. Moreover, we demonstrated that after 15-min of stimulation with Texas Red-EGF, increasing numbers of pEGFR-positive staining that had colocalized with Texas Red-EGF-positive punctate staining were seen in the cytoplasm of HT1080 cells but after 30-min of stimulation, most of this staining had disappeared from the cytoplasm and a large accumulation of pEGFR-positive staining appeared in the nucleus. Thus, nuclear accumulation of pEGFR appears to occur in an EGF-dependent manner. In contrast, such nuclear pEGFR-positive staining was not seen in the Y-27632-treated cells. Furthermore, silencing of RhoA or Rho-kinases I/II by sequence specific siRNAs considerably inhibited the EGF-dependent nuclear accumulation of pEGFR. Collectively, these results provide the first evidence that Rho-kinase signaling pathway plays a suppressive role in the intracellular vesicle trafficking of pEGFR via the endocytic pathway and that an increased Rho-kinase activity leads to the attenuation of the normal endocytic vesicular traffic of pEGFR via the early/late endocytic pathway, instead causing pEGFR to be trafficked out of the endocytic vesicles into the nucleus.


Subject(s)
Endocytosis , ErbB Receptors/metabolism , Fibrosarcoma/enzymology , Fibrosarcoma/pathology , Signal Transduction , rho-Associated Kinases/metabolism , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/metabolism , Amides/pharmacology , Blotting, Western , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Endocytosis/drug effects , Epidermal Growth Factor/pharmacology , Gene Silencing/drug effects , Humans , Ligands , Phosphorylation/drug effects , Protein Transport/drug effects , Pyridines/pharmacology , RNA, Small Interfering/metabolism , Signal Transduction/drug effects , rac1 GTP-Binding Protein/antagonists & inhibitors , rac1 GTP-Binding Protein/metabolism , rho-Associated Kinases/antagonists & inhibitors
5.
Cancer Cell ; 19(1): 125-37, 2011 Jan 18.
Article in English | MEDLINE | ID: mdl-21251616

ABSTRACT

Metastasis is responsible for most cancer deaths. Here, we show that Aes (or Grg5) gene functions as an endogenous metastasis suppressor. Expression of Aes was decreased in liver metastases compared with primary colon tumors in both mice and humans. Aes inhibited Notch signaling by converting active Rbpj transcription complexes into repression complexes on insoluble nuclear matrix. In tumor cells, Notch signaling was triggered by ligands on adjoining blood vessels, and stimulated transendothelial migration. Genetic depletion of Aes in Apc(Δ716) intestinal polyposis mice caused marked tumor invasion and intravasation that were suppressed by Notch signaling inhibition. These results suggest that inhibition of Notch signaling can be a promising strategy for prevention and treatment of colon cancer metastasis.


Subject(s)
Colonic Neoplasms/pathology , Receptors, Notch/metabolism , Signal Transduction/physiology , Transcription Factors/metabolism , Animals , Benzodiazepinones/pharmacology , Benzodiazepinones/therapeutic use , Cell Line, Tumor , Co-Repressor Proteins , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Down-Regulation/genetics , Gene Expression/genetics , Gene Silencing/physiology , HCT116 Cells , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Intestinal Polyposis/drug therapy , Intestinal Polyposis/metabolism , Intestinal Polyposis/pathology , Ligands , Liver Neoplasms/pathology , Liver Neoplasms/secondary , Lung Neoplasms/pathology , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Mutant Strains , Mice, Nude , Mice, Transgenic , Models, Biological , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasm Invasiveness/prevention & control , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology , Neoplasm Metastasis/prevention & control , Nuclear Matrix/metabolism , Receptor, Notch1/metabolism , Receptors, Notch/antagonists & inhibitors , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction/drug effects , Stromal Cells/metabolism , Transcription Factors/genetics , Transendothelial and Transepithelial Migration/physiology
6.
Clin Exp Metastasis ; 27(8): 619-30, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20872237

ABSTRACT

The outcomes of osteosarcoma patients still remain poor because of intractable pulmonary metastasis. We previously established a highly metastatic osteosarcoma cell line, LM8 from Dunn mouse osteosarcoma by in vivo selection. We herein aimed to clarify the characteristic biological features related with high metastatic potential and new target molecules to suppress pulmonary metastasis of osteosarcoma, using this syngeneic spontaneous metastatic model. LM8 cells acquired fibroblastic morphology with striking filopodia on the cell surface. Immunostaining showed faint stress fiber formation and peripherally localized integrin ß1, and biochemical analyses showed the activated Cdc42 and autophosphorylation of focal adhesion kinase (FAK) in LM8 cells when compared to Dunn cells. LM8 cells had activated motility in single cell migration mode. LM8 migration was increased by a Rho-associated kinase (ROCK) inhibitor, Y-27632, while decreased by Cdc42 silencing using RNA interference system. We found that a clinically approved camptothecin analog, irinotecan suppressed the migration, Cdc42 activity, and autophosphorylation of FAK, and attenuated integrin ß1 distribution selectively in LM8 cells. Daily oral administration of irinotecan significantly reduced the rate and size of pulmonary metastasis in syngeneic C3H mice. The fibroblastic morphology and activated cell migration with the dependency on Cdc42 but not Rho-ROCK signaling pathway argued that LM8 moved in mesenchymal mode of cell migration. This activated mesenchymal migration was a key component of the pulmonary metastasis of LM8 cells. The inhibition of mesenchymal migration by irinotecan, in addition to its cytotoxic effects, might be effective in preventing pulmonary metastasis of osteosarcoma.


Subject(s)
Cell Movement , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Mesoderm/pathology , Osteosarcoma/pathology , Animals , Camptothecin/analogs & derivatives , Camptothecin/blood , Camptothecin/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation , Disease Models, Animal , Irinotecan , Lung Neoplasms/blood , Mesoderm/drug effects , Mice , Mice, Inbred C3H , Osteosarcoma/blood , Signal Transduction , cdc42 GTP-Binding Protein/metabolism
7.
Stem Cells ; 28(7): 1119-31, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20518020

ABSTRACT

Synovial sarcoma (SS) is a malignant soft tissue tumor characterized by its unique t(X;18)(p11;q11) chromosomal translocation leading to the formation of the SS18-SSX fusion gene. The resulting fusion protein product is considered to play as an aberrant transcription factor and transform target cells by perturbing their gene expression program. However, the cellular origin of SS is highly debated. We herein established two novel human SS cell lines, named Yamato-SS and Aska-SS, and investigated their biological properties. We found the self-renewal ability of these cells to generate sarcospheres, to form tumors in serial xenotransplantation and reconstitute the tumor phenotypes without fractionation by any surface markers. Both SS cells as well as clinical tissue specimens from 15 patients expressed the marker genes-associated stem cell identity, Oct3/4, Nanog, and Sox2. We also found that both SS cells displayed limited differentiation potentials for mesenchymal lineages into osteocytes and chondrocytes albeit with the expression of early mesenchymal and hematopoietic lineage genes. Upon SS18-SSX silencing with sequence-specific siRNAs, these SS cells exhibited morphological transition from spherical growth in suspension to adherent growth in monolayer, additional expression of later mesenchymal and hematopoietic lineage genes, and broader differentiation potentials into osteocytes, chondrocytes, adipocytes, and macrophages in appropriate differentiation cocktails. Collectively, these data suggest that a human multipotent mesenchymal stem cell can serve as a cell of origin for SS and SS is a stem cell malignancy resulting from dysregulation of self-renewal and differentiation capacities driven by SS18-SSX fusion protein.


Subject(s)
Neoplastic Stem Cells/metabolism , Sarcoma, Synovial/metabolism , Animals , Cell Culture Techniques , Cell Differentiation , Cell Lineage , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Nude , Neoplastic Stem Cells/cytology , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , RNA, Small Interfering/genetics , Sarcoma, Synovial/genetics , Sarcoma, Synovial/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
Int J Oncol ; 36(4): 823-31, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20198325

ABSTRACT

Synovial sarcoma (SS) is an aggressive soft-tissue malignancy characterized by a unique t(X;18) translocation resulting in expression of SS18-SSX fusion protein. In order to investigate the biological function of this fusion protein and to develop a novel therapeutic option, we examined downregulation of SS18-SSX1 expression by small interfering RNA targeting SS18-SSX1 in three human SS cell lines. Microarray analysis comparing SS18-SSX1-silenced cells with control cells in three SS cell lines showed that SS18-SSX1 mainly affected the focal adhesion pathway. In accord with the array data, silencing of SS18-SSX1 enhances adhesion to the extracellular matrix through the induction of expression of myosin light-chain kinase. Furthermore, the silencing of SS18-SSX1 inhibits anchorage-independent growth in vitro and systemic delivery of siRNA against SS18-SSX1 using a nanoparticle system inhibited tumor growth in a nude mouse xenograft model. Our results demonstrate that siRNA targeting of SS18-SSX1 has therapeutic potential for the treatment of SS.


Subject(s)
Cell Proliferation , Focal Adhesions/genetics , Genetic Therapy/methods , Neoplasm Proteins/genetics , Oncogene Proteins, Fusion/genetics , Proto-Oncogene Proteins/genetics , RNA Interference , Repressor Proteins/genetics , Sarcoma, Synovial/therapy , Adult , Animals , Calcium-Binding Proteins/metabolism , Cell Adhesion/genetics , Cell Line, Tumor , Cell Shape , Down-Regulation , Focal Adhesions/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Mice, Nude , Myosin-Light-Chain Kinase/metabolism , Neoplasm Proteins/metabolism , Oncogene Proteins, Fusion/metabolism , Proto-Oncogene Proteins/metabolism , Recombinant Fusion Proteins/metabolism , Repressor Proteins/metabolism , Sarcoma, Synovial/genetics , Sarcoma, Synovial/metabolism , Sarcoma, Synovial/pathology , Time Factors , Transcription, Genetic , Transfection , Tumor Burden , Xenograft Model Antitumor Assays
9.
Clin Orthop Relat Res ; 467(12): 3087-95, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19609629

ABSTRACT

The small GTPase Rho and Rho-associated protein kinase (Rho kinase, ROCK) signal participates in a variety of biological functions including vascular contraction, tumor invasion, and penile erection. Evidence also suggests Rho-ROCK is involved in signaling for mesenchymal cellular differentiation. However, whether it is involved in osteoblastic differentiation is unknown. We therefore asked whether Rho-ROCK signaling participates in recombinant human bone morphogenetic protein (rhBMP-2)-induced osteogenesis both in vitro and in vivo. Continuous delivery of a specific ROCK inhibitor (Y-27632) enhanced ectopic bone formation induced by rhBMP-2 impregnated into an atelocollagen carrier in mice without affecting systemic bone metabolism. Treatment with Y-27632 also enhanced the osteoblastic differentiation of cultured murine neonatal calvarial cells. These effects were associated with increased expression of BMP-4 gene. Expression of a dominant negative mutant of ROCK in ST2 cells promoted osteoblastic differentiation, while a constitutively active mutant of ROCK attenuated osteoblastic differentiation and the ROCK inhibitor reversed this phenotype. Thus, ROCK inhibits osteogenesis, and a ROCK inhibitor in combination with the local delivery of rhBMP/collagen composite may be clinically applicable for stimulating bone formation.


Subject(s)
Amides/administration & dosage , Bone Morphogenetic Protein 2/administration & dosage , Ossification, Heterotopic/chemically induced , Osteoblasts/drug effects , Osteogenesis/drug effects , Protein Kinase Inhibitors/administration & dosage , Pyridines/administration & dosage , rho-Associated Kinases/antagonists & inhibitors , Animals , Animals, Newborn , Bone Morphogenetic Protein 4/genetics , Cell Differentiation/drug effects , Cell Movement/drug effects , Cells, Cultured , Collagen , Dose-Response Relationship, Drug , Drug Carriers , Humans , Male , Mice , Mice, Inbred ICR , Mutation , Ossification, Heterotopic/enzymology , Ossification, Heterotopic/physiopathology , Osteoblasts/enzymology , Pilot Projects , RNA, Messenger/metabolism , Recombinant Proteins/administration & dosage , Skull/drug effects , Skull/enzymology , Stromal Cells/drug effects , Stromal Cells/enzymology , Transfection , rho-Associated Kinases/genetics , rho-Associated Kinases/metabolism
10.
Bone ; 43(5): 869-79, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18718566

ABSTRACT

Osteoblast recruitment to the site of future bone formation is essential for skeletal development, bone remodeling and fracture healing. A number of factors associated with bone tissue have been reported to induce directional migration of osteoblasts but the mechanism remains to be clarified. In this study, to explore a major chemotactic factor(s) for osteoblasts, we examined the serum-free medium conditioned by MC3T3-E1 osteoblast-like cells for its ability to induce osteoblast migration. Employing sequential chromatography and tandem mass spectrometry analysis, we purified and identified IGF-I as a potent chemotactic factor from the conditioned medium. IGF-I induced cell migration of both MC3T3-E1 cells and primary mouse osteoblasts, and checkerboard analysis revealed that IGF-I markedly induced directional migration (chemotaxis) of osteoblasts. Neutralization of mouse IGF-I with monoclonal antibodies resulted in delayed osteoblast monolayer wound healing and cellular polarization but addition of human IGF-I reversed these effects. IGF-I also promoted cell spreading on fibronectin in an integrin beta1-dependent manner. IGF-I induced Akt and Rac activation and localized accumulation of phosphatidylinositol 3,4,5-triphosphate (PtdIns (3,4,5)P3) at the membrane in osteoblasts. The phosphatidyl inositol 3 kinase (PI3K) inhibitor LY294002 inhibited IGF-I-induced cell migration and wound healing. Together, the results suggest that IGF-I secreted from osteoblasts in the bone tissue is a potent chemotactic factor that may play a major role in recruitment of osteoblasts during bone formation.


Subject(s)
Chemotactic Factors/metabolism , Chemotaxis/physiology , Insulin-Like Growth Factor I/metabolism , Osteoblasts/metabolism , 3T3 Cells , Animals , Culture Media, Conditioned , Culture Media, Serum-Free , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Insulin-Like Growth Factor I/genetics , MAP Kinase Kinase Kinases/metabolism , Mice , Phosphatidylinositol 3-Kinases/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction/physiology
11.
Mol Cancer ; 7: 42, 2008 May 21.
Article in English | MEDLINE | ID: mdl-18492291

ABSTRACT

Gefitinib (Iressa)-a specific inhibitor of epidermal growth factor receptor (EGFR) tyrosine kinase-has been shown to suppress the activation of EGFR signaling required for cell survival and proliferation in non-small cell lung cancer (NSCLC) cell lines. We recently provided novel evidence that gefitinib-sensitive PC9 cells show normal endocytosis of EGFR: internalized EGF-EGFR complexes were transported to late endosomes/lysosomes 15 min after EGF stimulation, and then degraded within the lysosomes. However, gefitinib-resistant QG56 cells showed internalized EGFR accumulation in early endosomes after 60 min of internalization, instead of its trafficking to lysosomes, indicating an aberration in some steps of EGF-EGFR trafficking from the early endosomes to late endosomes/lysosomes. Therefore, we postulate that impairment in some steps of EGF-EGFR trafficking from early endosomes to late endosomes/lysosomes might confer gefitinib-resistance in NSCLC cell lines. To further substantiate the detailed internalization mechanism of gefitinib-sensitive and gefitinib-resistant cells, using confocal immunofluorescence microscopy, we examined the endocytic trafficking of phosphorylated EGFR (pEGFR) in the absence or presence of gefitinib. In PC9 and QG56 cells without EGF stimulation, a large number of pEGFR-positive small vesicular structures not colocalized with late endosomes/lysosomes were spread throughout the cytoplasm, and some pEGFR staining was distributed in the nucleus. This implies a novel intracellular trafficking pathway for pEGFR from cytoplasmic vesicles to the nucleus. Furthermore, an aggregated vesicular structure of early endosomes was observed in the perinuclear region of QG56 cells; it was revealed to be associated with SNX1, originally identified as a protein that interacts with EGFR. Therefore, we confirmed our previous data that an aberration in some steps of EGF-EGFR trafficking from the early endosomes to late endosomes/lysosomes occurs in QG56 cells. Furthermore, in PC9 cells, efficient phosphorylation of EGFR and rapid internalization of pEGFR was observed at 3 min after EGF stimulation; these internalized pEGFR-positive vesicles were trafficked to late endosomes at 15 min, indicating rapid trafficking of EGF-pEGFR complexes from early to late endosomes in PC9 cells. Gefitinib treatment strongly reduced the phosphorylation level of EGFR, and subsequent endocytosis of EGFR was significantly suppressed in PC9 cells. In contrast, in QG56 cells, EGFR trafficking via the early endocytic pathway was basically impaired; therefore, gefitinib appeared to slightly suppress the internalization of pEGFR. Collectively, our data provide novel evidence that extensive impairment in pEGFR endocytosis via the early endocytic pathway might confer gefitinib-resistance in QG56 cells.


Subject(s)
Endocytosis/drug effects , ErbB Receptors/metabolism , Quinazolines/pharmacology , Biological Transport/drug effects , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Endosomes/metabolism , Epidermal Growth Factor/pharmacology , Gefitinib , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lysosomes/metabolism , Microscopy, Fluorescence , Phosphorylation/drug effects , Signal Transduction/drug effects , Sorting Nexins , Vesicular Transport Proteins/metabolism
12.
Exp Cell Res ; 312(19): 3927-38, 2006 Nov 15.
Article in English | MEDLINE | ID: mdl-17022972

ABSTRACT

Cell migration is essential for both organogenesis and tumor progression. Bone morphogenetic proteins (BMPs) are reported to be critical for not only bone formation but also tumor invasion. Here, we found that treatment with recombinant human BMP-2 (rhBMP-2) enhanced the haptotactic response of murine osteoblastic MC3T3-E1 and osteosarcoma Dunn cells to various extracellular matrix (ECM) components, including fibronectin, type I collagen, and laminin-1. Function-blocking antibody against integrin alpha5beta1 partially inhibited haptotaxis to fibronectin, suggesting that the response was propagated via these integrins. rhBMP-2 slightly increased the expression level of integrin beta1, and enhanced the speed of cell spreading on fibronectin, focal adhesion formation and phosphorylation of focal adhesion kinase (FAK) at Tyr397. By means of sucrose gradient flotation, incorporation of integrin beta1 in fractions of detergent (CHAPS) resistant membrane was increased when the cells were treated with rhBMP-2. Further, treatment with methyl-beta-cyclodextrin to deplete membrane cholesterol abrogated the effect of rhBMP-2 on haptotaxis, and exogenously added cholesterol reversed this inhibitory effect. Collectively, these results provide insights into the mechanism by which BMP signaling enhances cell migration by modulating fibronectin-integrin beta1 signaling via cholesterol enriched membrane microdomains, lipid rafts.


Subject(s)
Bone Morphogenetic Proteins/pharmacology , Integrin beta1/metabolism , Osteoblasts/drug effects , Transforming Growth Factor beta/pharmacology , 3T3 Cells , Animals , Bone Morphogenetic Protein 2 , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Focal Adhesions/drug effects , Humans , Membrane Microdomains/drug effects , Membrane Microdomains/metabolism , Mice , Osteoblasts/cytology , Osteoblasts/metabolism , Osteosarcoma/metabolism , Osteosarcoma/pathology , Recombinant Proteins/pharmacology , Signal Transduction
13.
Histochem Cell Biol ; 126(5): 627-38, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16763828

ABSTRACT

We have previously shown that overexpression of LIM kinase1 (LIMK1) resulted in a marked retardation of the internalization of the receptor-mediated endocytic tracer, Texas red-labeled epidermal growth factor (EGF) in low-invasive human breast cancer cell MCF-7. We thereby postulate that LIMK1 signaling plays an important role in the regulation of ligand-induced endocytosis of EGF receptor (EGFR) in tumor cells by reorganizing and influencing actin-filament dynamics. In the present study, we further assessed the effect of wild-type LIMK1, a kinase-deficient dominant negative mutant of LIMK1 (DN-LIMK1) and an active, unphosphorylatable cofilin mutant (S3A cofilin) on internalization of EGF-EGFR in MDA-MB-231, a highly invasive human breast cancer cell line. We demonstrate here that a marked delay in the receptor-mediated internalization of Texas red-labeled EGF was observed in the wild-type LIMK1 transfectants, and that most of the internalized EGF staining were accumulated within transferrin receptor-positive early endosomes even after 30 min internalization. In contrast, the expression of dominant-negative LIMK1 mutant rescued the efficient endocytosis of Texas red-EGF, and large amounts of Texas red-EGF staining already reached LIMPII-positive late endosomes/lysosomal vacuoles after 15 min internalization. We further analyzed the effect of S3A cofilin mutant on EGFR trafficking, and found an efficient delivery of Texas red-EGF into late endosomes/lysosomes at 15-30 min after internalization. Taken together, our novel findings presented in this paper implicate that LIMK1 signaling indeed plays a pivotal role in the regulation of EGFR trafficking through the endocytic pathway in invasive tumor cells.


Subject(s)
Actin Depolymerizing Factors/physiology , Breast Neoplasms/metabolism , Endocytosis/physiology , ErbB Receptors/metabolism , Protein Kinases/physiology , Actin Depolymerizing Factors/genetics , Cathepsin D/metabolism , Female , Humans , Immunoblotting , Lim Kinases , Lysosomal Membrane Proteins/metabolism , Protein Transport , Receptors, Scavenger/metabolism , Signal Transduction , Transfection , Tumor Cells, Cultured
14.
Breast Cancer Res Treat ; 93(2): 159-68, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16187236

ABSTRACT

In order to display the full metastatic phenotype, the cancer cell must acquire the ability to migrate. In breast cancer, we have previously shown that insulin-like growth factor I (IGF-I) enhances cell motility in the highly metastatic MDA-231BO cell line by activating the type I IGF receptor (IGF1R). This motility response requires activation of IRS-2 and integrin ligation. In order to identify the key molecules downstream of IRS-2, we examined several signaling pathways known to be involved in cell motility. Focal adhesion kinase (FAK) was not activated by IGF-I, but IGF-I caused redistribution of FAK away from focal adhesion plaques. IGF-I treatment of MDA-231BO cells activated RhoA and inhibition of Rho-kinase (ROCK) inhibited the IGF-mediated motility response. The mitogen activated protein kinase (MAPK), p38, was also activated by IGF-I and inhibition of p38 by SB203580 blocked IGF-I induced cell motility. ROCK inhibition with Y-27632 also inhibited p38 phosphorylation suggesting that p38 lies downstream of ROCK. Both Erk1,2 and phosphatidyl-3 kinase (PI3K) were required for IGF-I stimulated cell motility, but only PI3K appeared to be directly downstream of IGF-I. Thus, IGF-I activation of its receptor coordinates multiple signaling pathways required for cell motility. Defining the key molecules downstream of the type I IGF receptor may provide a basis for optimizing therapies directed at this target.


Subject(s)
Cell Movement/drug effects , Insulin-Like Growth Factor I/pharmacology , Signal Transduction/drug effects , Amides/pharmacology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Adhesion/drug effects , Cell Line, Tumor , Enzyme Inhibitors/pharmacology , Female , Focal Adhesion Kinase 1/metabolism , Humans , Imidazoles/pharmacology , Immunoblotting , Microscopy, Confocal , Mitogen-Activated Protein Kinase Kinases/metabolism , Pyridines/pharmacology , rho GTP-Binding Proteins/metabolism
15.
Mol Cancer Res ; 3(8): 425-33, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16123138

ABSTRACT

The small GTP-binding protein Rho and its target Rho-associated kinase trigger an intracellular signaling cascade that controls actin cytoskeleton and plays an essential role in cell motility and adhesion. A specific Rho-associated kinase inhibitor, Y-27632, has been reported to inhibit cancer invasion. Clinically, disseminated tumor cells in the cerebrospinal fluid invade the intraparenchymal region, damaging the brain and nerves, resulting in fatal brain stem dysfunction, despite intrathecal chemotherapy. To expand therapeutic options for this devastating neoplastic meningitis, we evaluated the potential use of intrathecal Y-27632 administration by employing Walker 256 cells, a rat mammary cancer cell line. Y-27632 dose-dependently inhibited chemotactic and invasive activity of Walker 256 cells. Y-27632 also inhibited the phosphorylation level of regulatory myosin light chain in vitro, but the effect was temporary and was considerably diminished within 16 hours. Y-27632 induced striking morphologic changes in Walker 256 cells, as evidenced by decreased cell-matrix adhesion in culture dishes and three-dimensional collagen I gels, and slightly inhibited colony formation in soft agar. Nevertheless, this drug treatment did not affect Walker 256 cell growth rate. We were able to administer continuous delivery of this inhibitor using an osmotic pump and maintaining drug concentration of 10 mumol/L within the brain. Importantly, this concentration of Y-27632 showed minimal neurotoxicity both in vitro and in vivo. We found that an intrathecal therapy, combining 5-fluoro-2'-deoxyuridine with Y-27632, significantly increased the survival time of rats bearing meningeal carcinomatosis in comparison with animals treated with 5-fluoro-2'-deoxyuridine alone. Taken together, our findings indicate that continuous intrathecal administration of Y-27632 could be a promising therapeutic method when combined with chemotherapy for treating human neoplastic meningitis.


Subject(s)
Amides/pharmacology , Enzyme Inhibitors/pharmacology , Meningeal Neoplasms/drug therapy , Meningitis/drug therapy , Pyridines/pharmacology , Actins/metabolism , Agar/chemistry , Amides/administration & dosage , Animals , Brain/metabolism , Cell Adhesion , Cell Line, Tumor , Cell Movement , Cells, Cultured , Chemotaxis , Cytoskeleton/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/administration & dosage , Female , Floxuridine/pharmacology , GTP-Binding Proteins/metabolism , Injections, Spinal , Mammary Neoplasms, Animal/metabolism , Mice , Microscopy, Fluorescence , Myosin Light Chains/chemistry , Neoplasm Invasiveness , Neoplasm Transplantation , Neurons/metabolism , Osmosis , Phosphorylation , Pyridines/administration & dosage , Rats , Rats, Wistar , Signal Transduction , Time Factors , rho GTP-Binding Proteins/chemistry
16.
J Mol Diagn ; 7(2): 187-97, 2005 May.
Article in English | MEDLINE | ID: mdl-15858142

ABSTRACT

The SSX family proteins have been considered new members of the cancer/testis antigens because of the restricted expression in testis among normal tissues and the activation in a wide range of cancers. Thus, they would be potential molecular targets for immunotherapeutic strategies. We have developed a competitive nucleic acid sequence-based amplification (NASBA) assay to analyze SSX mRNA expression in 211 bone and soft tissue tumors. The copy numbers of SSX mRNA per mug of total RNA in tumor tissues were widely distributed, ranging logarithmically from 0.6 to 6.6. We found that malignant tumors showed significantly higher expression of SSX mRNA than benign tumors (P < 0.0001). Further, SSX mRNA expression in stage III tumors was significantly higher than that in stage I or II tumors (P < 0.005). This NASBA assay was also more sensitive compared to immunohistochemistry using newly affinity-purified polyclonal antibody against SSX. Collectively, these results suggest that the SSX quantitative NASBA assay could provide useful information to select eligible patients for SSX-specific cancer vaccines.


Subject(s)
Bone Neoplasms/genetics , Neoplasm Proteins/genetics , RNA, Messenger/analysis , Repressor Proteins/genetics , Self-Sustained Sequence Replication , Soft Tissue Neoplasms/genetics , Adolescent , Adult , Aged , Animals , Bone Neoplasms/immunology , COS Cells , Child , Female , Gene Expression , Humans , Male , Middle Aged , Neoplasm Proteins/analysis , RNA, Messenger/metabolism , RNA, Neoplasm/analysis , RNA, Neoplasm/metabolism , Repressor Proteins/analysis , Soft Tissue Neoplasms/immunology
17.
Eur J Cell Biol ; 83(7): 369-80, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15503860

ABSTRACT

LIM kinase (LIMK) plays a critical role in stimulus-induced remodeling of the actin cytoskeleton by linking signals from the Rho family GTPases to changes in cofilin activity. Recent studies have shown an important role for LIMK1 signaling in tumor cell invasion through regulating actin dynamics. In this study, we investigate the role of LIMK1 in intracellular vesicle trafficking of lysosomes/endosomes. We analyzed by confocal immunofluorescence microscopy the cellular distribution of lysosomal proteins and the endocytosis of an endocytic tracer, epidermal growth factor (EGF), in LIMK1-transfected cells. We found in these cells an abnormal dispersed translocation of lysosomes stained for LIMPII and cathepsin D throughout the cytoplasm. The small punctate structures that stained for these lysosomal proteins were redistributed to the periphery of the cell. Computational 3D-image analysis of confocal immunofluorescence micrographs further demonstrated that these vesicles did not colocalize with the transferrin receptor, an early endosomal marker. Furthermore, LIMPII-positive lysosomes did not colocalize with early endosomes labeled with endocytosed Texas red-transferrin. These results indicate that there is no mixing between dispersed lysosomes and early endosomes in the LIMK1-transfected cells. Moreover, LIMK1 overexpression resulted in a marked retardation in the receptor-mediated internalization of Texas red-labeled EGF in comparison with mock-transfected cells. At 30 min after internalization, most of the Texas red-EGF staining overlapped with LIMPII-positive late endosomes/lysosomes in mock-transfected cells, whereas in LIMK1 transfectants only a small fraction of internalized EGF colocalized with LIMPII-positive structures in the perinuclear region. Taken together, the findings presented in this paper suggest that LIMK1 has a role in regulating vesicle trafficking of lysosomes and endosomes in invasive tumor cells.


Subject(s)
Actins/metabolism , Breast Neoplasms/enzymology , Cytoskeleton/metabolism , Endosomes/metabolism , Lysosomes/metabolism , Protein Kinases/metabolism , Biological Transport, Active , Breast Neoplasms/pathology , Cathepsin D/metabolism , Cell Line, Tumor , Endocytosis/physiology , Endosomes/pathology , Epidermal Growth Factor/pharmacology , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lim Kinases , Lysosomal Membrane Proteins , Lysosomes/pathology , Membrane Proteins/metabolism , Microscopy, Confocal , Microscopy, Fluorescence , Neoplasm Metastasis/physiopathology , Proteins/metabolism , Receptors, Scavenger , Sialoglycoproteins/metabolism , Signal Transduction/drug effects
18.
Am J Pathol ; 165(2): 471-80, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15277221

ABSTRACT

Pleomorphic malignant fibrous histiocytoma (MFH) is occasionally associated with inflammatory paraneoplastic syndrome (PNS). Recently, we reported that interleukin (IL)-6, one of the candidate cytokines, which induces such systemic inflammatory reaction, may be a tumor-associated factor involved in the pathogenesis and its clinical manifestations of MFH. In the local microenvironment, tumor-induced inflammatory reaction may play a role favoring tumor progression. To clarify the biological relevance of IL-6 in MFH, we established a human MFH cell line, named MIPS-2, derived from a resected specimen of a patient presenting with PNS. In this patient, the serum IL-6 level ran parallel to the disease course: elevated serum IL-6 concentration normalized immediately after radical surgery, and re-elevation occurred on tumor recurrence. MIPS-2 presented pleomorphic appearance, severe nuclear abnormalities with prominent nucleoli, and tumorigenesis in nude mice. MIPS-2 expressed IL-6, IL-6 receptor (IL-6R), and glycoprotein 130 (gp130) but lacked the soluble form of IL-6R (sIL-6R), as determined by flow cytometry and reverse transcriptase-polymerase chain reaction analyses. Stimulation of MIPS-2 with IL-6 combined with exogenous sIL-6R induced phosphorylation of both signal transducer and activator of transcription 3 (STAT3) and mitogen-activated protein kinase (MAPK), decreased cell proliferation, attenuated invasion, and induced morphological changes. Collectively, these data suggested that the IL-6/sIL-6R signaling pathway plays a pivotal role for proliferation, invasion, and morphology of MFH via STAT3 and MAPK pathway as autocrine and/or paracrine manner, and proposed the therapeutic potential for the use of both anti-growth factor and proinflammatory cytokine-targeting strategies to combat devastating MFH.


Subject(s)
Histiocytoma, Benign Fibrous/pathology , Interleukin-6/metabolism , Neoplasm Invasiveness/pathology , Paraneoplastic Syndromes/pathology , Receptors, Interleukin-6/metabolism , Signal Transduction , Adult , Animals , Antigens, CD/metabolism , Apoptosis , Cell Division/drug effects , Cytokine Receptor gp130 , DNA-Binding Proteins/metabolism , Flow Cytometry , Histiocytoma, Benign Fibrous/metabolism , Humans , Male , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred BALB C , Mitogen-Activated Protein Kinase Kinases/metabolism , Neoplasm Recurrence, Local/pathology , Paraneoplastic Syndromes/etiology , Paraneoplastic Syndromes/metabolism , Phosphorylation , Reverse Transcriptase Polymerase Chain Reaction , STAT3 Transcription Factor , Trans-Activators/metabolism , Tumor Cells, Cultured
19.
Pathol Oncol Res ; 9(2): 83-95, 2003.
Article in English | MEDLINE | ID: mdl-12858212

ABSTRACT

Small GTPase Rho and its downstream effectors, ROCK family of Rho-associated serine-threonine kinases, are thought to participate in cell morphology, motility, and tumor progression through regulating the rearrangement of actin cytoskeleton. Here we present evidence that transfection of human breast cancer cells with cDNA encoding a dominant active mutant of ROCK causes dispersal of lysosomal vesicles throughout the cytoplasm without perturbing the machinery of the endocytic pathway. The intracellular distribution of lysosomes and endocytosed transferrin, an early endosomal marker, were further assessed by confocal immunofluorescence microscopy. In the active ROCK transfected cells the lysosomal proteins, cathepsin D, LIMPII, and LAMP1, were found throughout the cytoplasm in dispersed small vesicles, which were accessible to the endocytosed Texas Red-labeled transferrin. 3D-image analysis of lysosomal distribution in the active ROCK transfectants revealed abundant punctate signals in the peripheral region of the basal plasma membrane. Cells expressing vector alone did not exhibit these alterations. Wortmannin, a phosphatidylinositol 3-kinase inhibitor, induced LIMPII-positive/ transferrin negative large vacuoles in the perinuclear region, and disappearence of the dispersed small vesicular structures. To our knowledge, this is the first evidence that increasing ROCK expression contributes to selective cellular dispersion of lysosomes in invasive breast cancer cells.


Subject(s)
Breast Neoplasms/enzymology , Intracellular Membranes/enzymology , Lysosomes/enzymology , Membrane Proteins , Protein Serine-Threonine Kinases/metabolism , Protein Transport , Sialoglycoproteins , Androstadienes/pharmacology , Antigens, CD/metabolism , CD36 Antigens/metabolism , Cathepsin D/metabolism , Endocytosis/physiology , Endosomes/drug effects , Endosomes/enzymology , Endosomes/ultrastructure , Enzyme Inhibitors/pharmacology , Female , Humans , Intracellular Signaling Peptides and Proteins , Lysosomal-Associated Membrane Protein 1 , Lysosomal Membrane Proteins , Lysosomes/ultrastructure , Microscopy, Confocal , Microscopy, Fluorescence , Phosphoinositide-3 Kinase Inhibitors , Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptors, Scavenger , Receptors, Transferrin/immunology , Signal Transduction , Transfection , Transferrin/metabolism , Tumor Cells, Cultured , Up-Regulation , Wortmannin , rho-Associated Kinases
20.
Proc Natl Acad Sci U S A ; 100(12): 7247-52, 2003 Jun 10.
Article in English | MEDLINE | ID: mdl-12777619

ABSTRACT

In this study, we show that LIM kinase 1 (LIMK1), a critical regulator of actin dynamics, plays a regulatory role in tumor cell invasion. We found that the level and activity of endogenous LIMK1 is increased in invasive breast and prostate cancer cell lines in comparison with less invasive cells. Overexpression of LIMK1 in MCF-7 and in MDA-MB-231 human breast cancer cell lines increased their motility, whereas the specific ROCK and Rho inhibitors Y-27632 and C3, respectively, attenuated this effect. In addition, inhibition of LIMK1 activity in the MDA-MB-231 cells by expression of dominant-negative LIMK1 resulted in decreased motility and formation of osteolytic bone lesions in an animal model of tumor invasion. This study shows an important role for LIMK1 signaling in invasion of cancer, demonstrating its potential as a therapeutic molecular target to decrease metastasis.


Subject(s)
Neoplasm Invasiveness/physiopathology , Protein Kinases/physiology , Animals , Bone Neoplasms/enzymology , Bone Neoplasms/genetics , Bone Neoplasms/secondary , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Female , Gene Expression , Humans , Lim Kinases , Male , Mice , Mice, Nude , Mutation , Neoplasm Transplantation , Osteolysis/enzymology , Osteolysis/etiology , Osteolysis/pathology , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Protein Kinase Inhibitors , Protein Kinases/genetics , Signal Transduction , Transfection , Transplantation, Heterologous , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...