Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Horm Cancer ; 9(4): 240-253, 2018 08.
Article in English | MEDLINE | ID: mdl-29687205

ABSTRACT

This retrospective case series study, using data obtained through questionnaires and histopathological diagnoses from 656 patients enrolled in the Department of Defense (DoD) Clinical Breast Care Project (CBCP), evaluated associations between hormonal contraceptive use and breast cancer pathology including benign breast pathologies. Three combination hormonal contraceptive agents (COCs) Lo Ovral (LO), Ortho Novum (ON), and Ortho Tri-Cyclen (OTC) were evaluated as they represented the most commonly used hormonal contraceptives in our cohort. The results of this study suggest that the ever use of LO + ON + OTC does not influence the overall incidence of benign breast condition or malignant disease compared to other COCs; however, patients that have used OTC had an association with a diagnosis of benign or luminal A pathologies whereas ON was associated with a diagnosis of benign and DCIS; LO showed no association with any diagnosis-benign or malignant. Patients that have used LO or ON were more likely to be diagnosed with breast cancer at age ≥ 40 years whereas patients that had ever used OTC were likely to be diagnosed before the age of 40. Caucasians were less likely to have used OTC and more likely to have used ON; however, use of either hormonal agent positively correlated with premenopausal status at diagnosis and having a benign condition. Age at diagnosis, ethnicity, BMI, family history, menstruation status, and duration of use were all independent predictors of different histopathological subtypes. We conclude that patient-specific variables should be considered when deciding on which type of hormonal contraceptive to use to minimize the risk of developing breast cancer or a breast-related pathology.


Subject(s)
Breast Neoplasms/epidemiology , Contraceptives, Oral, Combined/adverse effects , Adult , Female , Humans , Incidence , Middle Aged , Retrospective Studies
2.
J Transl Med ; 14: 48, 2016 Feb 12.
Article in English | MEDLINE | ID: mdl-26867596

ABSTRACT

BACKGROUND: Previous studies suggest that certain transition metal complexes, such as cisplatin, are efficacious for treating various cancer types, including ovarian, lung, and breast. METHODS: In order to further evaluate ruthenium (Ru) complexes as potential anti-cancer agents, we synthesized and evaluated Ru-arene complexes. Two complexes with the general formula [Ru (η (6)-p-cym) (N-N) Cl](+) were tested for their abilities to inhibit cancer cells. RESULTS: The complex with o-phenylenediamine as the N-N ligand (o-PDA) significantly inhibited growth of breast (MDA-MB-231, MCF-7, SKBR-3, and SUM149), lymphoma (Raji), melanoma (Bowes), and osteosarcoma (HT1080); however, the complex with o-benzoquinonediimine (o-BQDI) was ineffective except for SUM149. In contrast, o-PDA failed to inhibit growth of human breast epithelial cells, MCF-10A. Treatment of MDA-MBA-231 cells with o-PDA resulted in a significant reduction of productions of PDGF-AA, GM-CSF, and VEGF-A proteins at the transcriptional levels. Finally, we demonstrated that o-PDA synergistically inhibited MDA-MB-231 cell growth with cyclophosphamide but not doxorubicin or paclitaxel. CONCLUSION: These results suggest that Ru-arene complexes are promising anti-cancer drugs that inhibit progression and metastasis by blocking multiple processes for breast and other types of cancer.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Coordination Complexes/therapeutic use , Ruthenium/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , Female , Humans , Intercellular Signaling Peptides and Proteins/biosynthesis , Phenylenediamines/pharmacology , Ruthenium/chemistry , Ruthenium/pharmacology
3.
Genetics ; 201(4): 1341-8, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26510790

ABSTRACT

Molecular heterogeneity in human breast cancer has challenged diagnosis, prognosis, and clinical treatment. It is well known that molecular subtypes of breast tumors are associated with significant differences in prognosis and survival. Assuming that the differences are attributed to subtype-specific pathways, we then suspect that there might be gene regulatory mechanisms that modulate the behavior of the pathways and their interactions. In this study, we proposed an integrated methodology, including machine learning and information theory, to explore the mechanisms. Using existing data from three large cohorts of human breast cancer populations, we have identified an ensemble of 16 master regulator genes (or MR16) that can discriminate breast tumor samples into four major subtypes. Evidence from gene expression across the three cohorts has consistently indicated that the MR16 can be divided into two groups that demonstrate subtype-specific gene expression patterns. For example, group 1 MRs, including ESR1, FOXA1, and GATA3, are overexpressed in luminal A and luminal B subtypes, but lowly expressed in HER2-enriched and basal-like subtypes. In contrast, group 2 MRs, including FOXM1, EZH2, MYBL2, and ZNF695, display an opposite pattern. Furthermore, evidence from mutual information modeling has congruently indicated that the two groups of MRs either up- or down-regulate cancer driver-related genes in opposite directions. Furthermore, integration of somatic mutations with pathway changes leads to identification of canonical genomic alternations in a subtype-specific fashion. Taken together, these studies have implicated a gene regulatory program for breast tumor progression.


Subject(s)
Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Genes, Regulator , Adult , Cohort Studies , Disease Progression , Disease-Free Survival , Female , Gene Expression Profiling , Genes, Neoplasm , Humans , Machine Learning , Middle Aged , Mutation
4.
Exp Cell Res ; 330(2): 358-370, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25445787

ABSTRACT

There are lines of evidence demonstrating that NEDD9 (Cas-L, HEF-1) plays a key role in the development, progression, and metastasis of breast cancer cells. We previously reported that NEDD9 plays a critical role for promoting migration and growth of MDA-MB-231. In order to further characterize the mechanisms of NEDD9-mediated cancer migration and growth, stable cells overexpressing NEDD9 were generated using HCC38 as a parental cell line which expresses low level of endogenous NEDD9. Microarray studies demonstrated that core proteins of CD44 and Serglycin were markedly upregulated in HCC38(NEDD9) cells compared to HCC38(Vector) cells, while those of Syndecan-1, Syndecan-2, and Versican were downregulated in HCC38(NEDD9). Importantly, enzymes generating chondroitin sulfate glycosaminoglycans (CS) such as CHST11, CHST15, and CSGALNACT1 were upregulated in HCC38(NEDD9) compared to HCC38(Vector). Immunofluorescence studies using specific antibody, GD3G7, confirmed the enhanced expression of CS-E subunit in HCC38(NEDD9). Immunoprecipitation and western blotting analysis demonstrated that CS-E was attached to CD44 core protein. We demonstrated that removing CS by chondroitinase ABC significantly inhibited anchorage-independent colony formation of HCC38(NEDD9) in methylcellulose. Importantly, the fact that GD3G7 significantly inhibited colony formation of HCC38(NEDD9) cells suggests that CS-E subunit plays a key role in this process. Furthermore, treatment of HCC38(NEDD9) cells with chondroitinase ABC or GD3G7 significantly inhibited mammosphere formation. Exogenous addition of CS-E enhanced colony formation and mammosphere formation of HCC38 parental and HCC38(Vector) cells. These results suggest that NEDD9 regulates the synthesis and expression of tumor associated glycocalyx structures including CS-E, which plays a key role in promoting and regulating breast cancer progression and metastasis and possibly stem cell phenotypes.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Breast Neoplasms/pathology , Chondroitin Sulfates/biosynthesis , Phosphoproteins/metabolism , Spheroids, Cellular/pathology , Adaptor Proteins, Signal Transducing/biosynthesis , Antibodies, Monoclonal/immunology , Antigens/biosynthesis , Antigens/metabolism , Cell Movement , Cell Proliferation , Chondroitin ABC Lyase/metabolism , Chondroitin ABC Lyase/pharmacology , Down-Regulation , Female , Fluorescent Antibody Technique , Humans , Hyaluronan Receptors/biosynthesis , Membrane Glycoproteins/biosynthesis , N-Acetylgalactosaminyltransferases/biosynthesis , Neoplasm Metastasis/pathology , Phosphoproteins/biosynthesis , Proteoglycans/biosynthesis , Proteoglycans/metabolism , Sulfotransferases/biosynthesis , Syndecan-1/biosynthesis , Syndecan-2/biosynthesis , Tumor Cells, Cultured , Up-Regulation , Versicans/biosynthesis , Vesicular Transport Proteins/biosynthesis
5.
PLoS One ; 9(2): e88712, 2014.
Article in English | MEDLINE | ID: mdl-24586375

ABSTRACT

CD44 adhesion molecules are expressed in many breast cancer cells and have been demonstrated to play a key role in regulating malignant phenotypes such as growth, migration, and invasion. CD44 is an integral transmembrane protein encoded by a single 20-exon gene. The diversity of the biological functions of CD44 is the result of the various splicing variants of these exons. Previous studies suggest that exon v10 of CD44 plays a key role in promoting cancer invasion and metastasis, however, the molecular mechanisms are not clear. Given the fact that exon v10 is in the ectodomain of CD44, we hypothesized that CD44 forms a molecular complex with other cell surface molecules through exon v10 in order to promote migration of breast cancer cells. In order to test this hypothesis, we selected DNA aptamers that specifically bound to CD44 exon v10 using Systematic Evolution of Ligands by Exponential Enrichment (SELEX). We selected aptamers that inhibited migration of breast cancer cells. Co-immunoprecipitation studies demonstrated that EphA2 was co-precipitated with CD44. Pull-down studies demonstrated that recombinant CD44 exon v10 bound to EphA2 and more importantly aptamers that inhibited migration also prevented the binding of EphA2 to exon v10. These results suggest that CD44 forms a molecular complex with EphA2 on the breast cancer cell surface and this complex plays a key role in enhancing breast cancer migration. These results provide insight not only for characterizing mechanisms of breast cancer migration but also for developing target-specific therapy for breast cancers and possibly other cancer types expressing CD44 exon v10.


Subject(s)
Aptamers, Nucleotide/metabolism , Breast Neoplasms/pathology , Cell Movement , Exons , Hyaluronan Receptors/genetics , Aptamers, Nucleotide/genetics , Base Sequence , Cell Line, Tumor , Disease Progression , Humans , Peptides/metabolism , Receptor, EphA2/metabolism , SELEX Aptamer Technique , Substrate Specificity
6.
PLoS One ; 7(9): e44418, 2012.
Article in English | MEDLINE | ID: mdl-22984505

ABSTRACT

There is substantial evidence indicating that the WNT signaling pathway is activated in various cancer cell types including breast cancer. Previous studies reported that FH535, a small molecule inhibitor of the WNT signaling pathway, decreased growth of cancer cells but not normal fibroblasts, suggesting this pathway plays a role in tumor progression and metastasis. In this study, we tested FH535 as a potential inhibitor for malignant phenotypes of breast cancer cells including migration, invasion, and growth. FH535 significantly inhibited growth, migration, and invasion of triple negative (TN) breast cancer cell lines (MDA-MB231 and HCC38) in vitro. We demonstrate that FH535 was a potent growth inhibitor for TN breast cancer cell lines (HCC38 and MDA-MB-231) but not for other, non-TN breast cancer cell lines (MCF-7, T47D or SK-Br3) when cultured in three dimensional (3D) type I collagen gels. Western blotting analyses suggest that treatment of MDA-MB-231 cells with FH535 markedly inhibited the expression of NEDD9 but not activations of FAK, Src, or downstream targets such as p38 and Erk1/2. We demonstrated that NEDD9 was specifically associated with CSPG4 but not with ß1 integrin or CD44 in MDA-MB-231 cells. Analyses of gene expression profiles in breast cancer tissues suggest that CSPG4 expression is higher in Basal-type breast cancers, many of which are TN, than any other subtypes. These results suggest not only a mechanism for migration and invasion involving the canonical WNT-signaling pathways but also novel strategies for treating patients who develop TN breast cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Cell Movement/drug effects , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor/methods , Gene Expression Regulation, Neoplastic , Sulfonamides/pharmacology , Adaptor Proteins, Signal Transducing/metabolism , Cell Adhesion , Cell Line, Tumor , Chondroitin Sulfate Proteoglycans/metabolism , Female , Fibroblasts/metabolism , Gene Expression Profiling , Humans , Membrane Proteins/metabolism , Neoplasm Invasiveness , Phenotype , Phosphoproteins/metabolism , Wnt Proteins/metabolism
7.
J Periodontol ; 80(10): 1631-7, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19792853

ABSTRACT

BACKGROUND: Enamel matrix derivative (EMD) protein has been clinically used for periodontal regeneration, but the molecular mechanisms are not clear. Previous studies suggested that the activation of phosphoinositide 3-kinase (PI 3-kinase) plays a key role in facilitating cell migration. Given that the migration of osteoblasts is one of the key steps in the wound-healing processes, we hypothesized that EMD protein would stimulate osteoblast migration by activating PI 3-kinase. In this study, we tested this hypothesis using MG-63 cells as model systems to evaluate mechanisms of migration by stimulation with EMD protein. METHODS: Confluent MG-63 cells were mechanically scratched using a sterilized 1-mm pipette tip that removed the cells within a circular area. The wells were incubated for 24 hours in various stimulation conditions (25, 50, or 100 microg/ml EMD protein) with or without the PI 3-kinase inhibitor wortmannin (1, 10, and 100 nM) or LY294002 (1, 10, and 100 microM). Migrated cells in the wound section were counted by randomly selecting three areas from one well. The activation of PI 3-kinase by EMD protein was evaluated by the phosphorylation of Akt using Western blot analysis. RESULTS: Although EMD protein did not affect proliferation, it enhanced migration into wounds on MG-63 cells. We showed that EMD protein enhanced the phosphorylation of Akt in a dose-dependent manner. We demonstrated that the PI 3-kinase inhibitors wortmannin and LY294002 blocked migration into wounds and the phosphorylation of Akt enhanced by EMD protein in MG-63 cells. CONCLUSION: These results demonstrated that the activation of PI 3-kinase plays a key role in the EMD protein-stimulated migration of osteoblasts.


Subject(s)
Dental Enamel Proteins/pharmacology , Osteoblasts/drug effects , Phosphatidylinositol 3-Kinases/drug effects , Androstadienes/pharmacology , Animals , Blotting, Western , Cell Count , Cell Line , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Chromones/pharmacology , Dose-Response Relationship, Drug , Enzyme Activation , Enzyme Inhibitors/pharmacology , Humans , Morpholines/pharmacology , Osteoblasts/enzymology , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/drug effects , Wortmannin , Wound Healing/drug effects
8.
Surgery ; 145(3): 322-9, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19231585

ABSTRACT

INTRODUCTION: Hyaluronan (HA) and its biosynthetic enzymes hyaluronan synthases (HAS2 and HAS3) mediate Matrigel invasion by SW620 colon carcinoma cells. Because matrix metalloproteinases (MMPs) have been implicated in cancer invasion, we hypothesized that changes in HAS expression would alter MMP expression and activity in these cells. METHODS: To determine whether an MMP was involved in invasion, Matrigel invasion assays with SW620 cells were performed in the presence or absence of the MMP inhibitors GM6001 or TIMP2. HAS isozymes were inhibited by stably transfecting SW620 cells with vectors that contained antisense HAS2 and/or -3 cDNA; transfection with an empty vector served as a control. MMP-7 transcription was assessed by quantitative reverse transcription polymerase chain reaction (RT-PCR). MMP-7 protein was detected by enzyme-linked immunosorbent assay (ELISA) and enzymatic activity compared using zymography. RESULTS: GM6001 and TIMP2 decreased Matrigel invasion, which confirms that an MMP played a key role in this process. MMP-7 expression was then detected in SW620 cells. Finally, MMP-7 expression, protein, and enzymatic activity were significantly lower in antisense HAS tranfectants than in SW620 or vector control cells. CONCLUSION: We have demonstrated previously that inhibition of HAS expression and HA production in SW620 colon carcinoma cells inhibits Matrigel invasion. In the studies presented here, we have demonstrated that SW620 cells express high levels of MMP-7 and that inhibition of HAS isozymes dramatically decreases MMP-7 expression, protein, and enzymatic activity. Taken together, these findings suggest that HAS and HA may mediate cellular invasion via changes in MMP-7 expression.


Subject(s)
Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Glucuronosyltransferase/antagonists & inhibitors , Hyaluronic Acid/metabolism , Matrix Metalloproteinase 7/metabolism , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Collagen , DNA, Antisense/genetics , Dipeptides/pharmacology , Disease Progression , Drug Combinations , Glucuronosyltransferase/genetics , Glucuronosyltransferase/metabolism , Humans , Hyaluronan Synthases , Laminin , Proteoglycans , Tissue Inhibitor of Metalloproteinase-2/pharmacology , Transfection
9.
Melanoma Res ; 17(4): 205-13, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17625450

ABSTRACT

Tumor cell invasion through basement membranes and into stromal tissue are key steps for promoting growth and metastasis. Tumor cells express various extracellular-matrix-degrading enzymes such as matrix metalloproteinases (MMPs) to degrade extracellular matrix components to facilitate tumor migration and invasion. Histological and clinical studies suggest a role for MMP-1 (collagenase-1) in malignant melanoma invasion. In this study, we evaluated MMP-1 in regulating malignant phenotypes of human melanoma cells by generating human melanoma cells stably transfected with pro-MMP-1 cDNA. The transfectants expressed the active form of MMP-1 associated with cells and showed enhanced invasive and growth abilities in type I collagen gel. Furthermore, MMP-1 expression promoted anchorage-independent growth, which was inhibited in the presence of type II transforming growth factor (TGF)-beta receptor:Fc fusion protein that scavenges TGF-beta receptors. Finally, we demonstrated that MMP-1 directly generated active TGF-beta from its latent form. Thus, these results suggest that MMP-1 produced from melanoma cells would play a role in tumor progression by both degrading matrix proteins and generating active growth factors such as TGF-beta in vivo.


Subject(s)
Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Matrix Metalloproteinase 1/biosynthesis , Matrix Metalloproteinase 1/chemistry , Transforming Growth Factor beta/metabolism , Animals , Cell Line, Tumor , Collagen Type I/chemistry , Epithelial Cells/cytology , Humans , Lung/cytology , Melanoma/enzymology , Melanoma/metabolism , Mink , Models, Biological , Neoplasm Metastasis , Transfection
10.
Biochem J ; 403(3): 553-63, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17217338

ABSTRACT

We previously reported that CS (chondroitin sulfate) GAG (glycosaminoglycan), expressed on MCSP (melanoma-specific CS proteoglycan), is important for regulating MT3-MMP [membrane-type 3 MMP (matrix metalloproteinase)]-mediated human melanoma invasion and gelatinolytic activity in vitro. In the present study, we sought to determine if CS can directly enhance MT3-MMP-mediated activation of pro-MMP-2. Co-immunoprecipitation studies suggest that MCSP forms a complex with MT3-MMP and MMP-2 on melanoma cell surface. When melanoma cells were treated with betaDX (p-nitro-beta-D-xylopyranoside) to inhibit coupling of CS on the core protein, both active form and proform of MMP-2 were no longer co-immunoprecipitated with either MCSP or MT3-MMP, suggesting a model in which CS directly binds to MMP-2 and presents the gelatinase to MT3-MMP to be activated. By using recombinant proteins, we determined that MT3-MMP directly activates pro-MMP-2 and that this activation requires the interaction of the C-terminal domain of pro-MMP-2 with MT3-MMP. Activation of pro-MMP-2 by suboptimal concentrations of MT3-MMP is also significantly enhanced in the presence of excess C4S (chondroitin 4-sulfate), whereas C6S (chondroitin 6-sulfate) or low-molecular-mass hyaluronan was ineffective. Affinity chromatography studies using CS isolated from aggrecan indicate that the catalytic domain of MT3-MMP and the C-terminal domain of MMP-2 directly bind to the GAG. Thus the direct binding of pro-MMP-2 with CS through the C-domain would present the catalytic domain of pro-MMP-2 to MT3-MMP, which facilitates the generation of the active form of MMP-2. These results suggest that C4S, which is expressed on tumour cell surface, can function to bind to pro-MMP-2 and facilitate its activation by MT3-MMP-expressing tumour cells to enhance invasion and metastasis.


Subject(s)
Chondroitin Sulfate Proteoglycans/metabolism , Chondroitin Sulfates/physiology , Enzyme Precursors/metabolism , Gelatinases/metabolism , Matrix Metalloproteinase 16/physiology , Melanoma/metabolism , Membrane Proteins/metabolism , Metalloendopeptidases/metabolism , Humans , Matrix Metalloproteinase Inhibitors , Protein Structure, Tertiary , Tissue Inhibitor of Metalloproteinase-2/pharmacology , Tumor Cells, Cultured
11.
Am J Pathol ; 169(2): 445-58, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16877347

ABSTRACT

Natural killer (NK) cells play a key role in inflammation and tumor regression through their ability to migrate into tissues. CXCL12 is a chemokine that promotes lymphocyte invasion and migration into tissues; however, the mechanism for this process remains incompletely understood. In this study, we show that CXCL12 significantly enhanced CD16(+)CD56(+) human peripheral NK-cell invasion into type I collagen by the catalytic activity of matrix metalloproteinase-1 (MMP-1). Confocal immunofluorescence and co-immunoprecipitation studies suggest that MMP-1 colocalized with alpha(2)beta(1) integrin on CXCL-12-stimulated NK-cell surface. The binding of pro-MMP-1 with alpha(2)beta(1) integrin required activation of G(i)-coupled pathway. However, the production of MMP-1 from CXCL12-stimulated NK cells was mediated by p38 and mitogen-activated or extracellular signal-regulation protein kinase kinase 1/2 in a manner independent of the G(i)-coupled pathway. These results suggest that CXCL12/CXCR4 interaction transduces the two signaling pathways to promote NK-cell invasion, which stimulates pericellular degradation of extracellular matrix proteins by membrane-associated MMP-1. The mechanisms would thus play a role in facilitating lymphocyte trafficking and accumulation in tissues during physiological and pathological processes.


Subject(s)
Chemokines, CXC/pharmacology , Killer Cells, Natural/drug effects , Killer Cells, Natural/enzymology , Matrix Metalloproteinase 1/biosynthesis , Matrix Metalloproteinase 1/metabolism , CD56 Antigen/immunology , Chemokine CXCL12 , Collagen Type I/metabolism , Culture Media, Conditioned , Humans , Integrin alpha2beta1/metabolism , Killer Cells, Natural/cytology , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 2/antagonists & inhibitors , Pertussis Toxin/pharmacology , Receptors, IgG/immunology , Tissue Inhibitor of Metalloproteinase-1/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
12.
J Transl Med ; 3: 42, 2005 Nov 30.
Article in English | MEDLINE | ID: mdl-16318634

ABSTRACT

BACKGROUND: The tetraspan protein CD63, originally described as a stage-specific melanoma antigen but also present in a number of normal cells, regulates melanoma cell growth in nude mice, motility in serum containing media, and adhesion to several extracellular matrix proteins. CD63 has been reported to associate with beta1 and beta2 integrins, but the mechanism of signal transduction by CD63 is not clear. This study examined whether CD63 is associated with protein kinase and can transmit signals in melanoma cells. METHODS: Immunoprecipitation and radiolabeling were used to test for association of protein kinase activity with CD63. Adhesion of cells to monoclonal antibodies immobilized to microtiter plates was used to examine the ability of CD63 to transmit signals. RESULTS: CD63 was capable of transmitting a signal in melanoma cells that required extracellular calcium. In the absence of extracellular calcium at the time of binding to the CD63 mAb, the cell was no longer responsive to stimulation by CD63. Immunoprecipitation studies demonstrated protein kinase activity associated with CD63, and phosphoamino acid analysis revealed that most of this protein kinase activity was due to serine kinase activity. CONCLUSION: The current study suggests that serine protein kinase activity associated with CD63 may play a role in signaling by CD63 in melanoma cells.

13.
J Surg Res ; 122(1): 70-4, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15522317

ABSTRACT

BACKGROUND: Hyaluronan (HA) is a cell-surface glycosaminoglycan that has been implicated in cancer progression. Cells isolated from metastatic colon carcinoma (SW620) produce greater amounts of pericellular HA than cells isolated from a primary tumor (SW480). Inhibition of hyaluronan synthases (HAS) by transfection with antisense cDNA decreases HA production. Because adhesion to the extracellular matrix (ECM) is required for invasion and metastasis, we hypothesized that pericellular HA mediates adhesion to ECM proteins such as laminin, collagen, and fibronectin and that inhibition of HA production or removal of HA by digestion with hyaluronidase would impair adhesion. MATERIALS AND METHODS: SW480, SW620, and antisense transfectants (SW620 cells transfected with vector alone, antisense HAS2, antisense HAS3, and both antisense HAS2 and HAS3) were assessed for adhesion to laminin, Type 1 collagen, or fibronectin-coated plates. To confirm that adhesion was mediated by HA, cells were treated with or without hyaluronidase prior to the assays. RESULTS: Metastatic SW620 cells adhered well to laminin; SW480 cells demonstrated 46% less adhesion (P < 0.05; Student's t test). SW620 cell adhesion to Type 1 collagen and fibronectin was >50% less than adhesion to laminin. Inhibition of HAS2 and/or HAS3 or pretreatment with hyaluronidase significantly decreased adhesion of SW620 cells to laminin (P < 0.05), suggesting that adhesion was dependent upon pericellular HA. CONCLUSIONS: Metastatic SW620 cells that produce large amounts of pericellular HA adhered well to laminin. Inhibition of HAS2 and/or HAS3 expression, or hyaluronidase digestion of pericellular HA significantly inhibited adhesion. These data suggest that HA promotes adhesion to laminin and may thereby facilitate invasion of the basement membrane and metastasis in colon carcinoma.


Subject(s)
Carcinoma/physiopathology , Carcinoma/secondary , Colonic Neoplasms/physiopathology , Hyaluronic Acid/metabolism , Lymphatic Metastasis , Carcinoma/metabolism , Cell Adhesion/drug effects , Cell Line, Tumor , Collagen Type I , Colonic Neoplasms/metabolism , Fibronectins , Glucuronosyltransferase/metabolism , Humans , Hyaluronan Synthases , Hyaluronic Acid/antagonists & inhibitors , Hyaluronoglucosaminidase/pharmacology , Laminin , Oligonucleotides, Antisense/pharmacology , Transferases/genetics
14.
Cancer Res ; 64(13): 4569-76, 2004 Jul 01.
Article in English | MEDLINE | ID: mdl-15231668

ABSTRACT

Hyaluronan (HA) and its biosynthetic enzymes, HA synthases (HAS1, 2, and 3) are thought to participate in cancer progression. We have shown previously that HA production and HAS3 expression are increased in metastatic colon carcinoma cells (SW620) when compared with cells isolated from a primary tumor (SW480). Because invasion of the extracellular matrix is a fundamental event in tumor growth and metastasis, we hypothesized that SW620 cells would show greater invasive capability than SW480 cells, that invasion is HA dependent, and that HA mediates invasion via interaction with a cell-surface receptor. Invasion into artificial basement membrane (Matrigel) was assessed in vitro. To assess HA functionality, HAS expression was inhibited in SW620 cells by transfection with antisense HAS constructs. Decreased HA secretion and retention in the transfectants were confirmed using competitive binding and particle exclusion assays. SW620 cells demonstrated greater invasion through Matrigel than did SW480 cells. Antisense transfection decreased Matrigel invasion by SW620 cells by >60%; addition of exogenous HA restored invasion. Because the cell-surface HA receptor CD44 has been implicated in cancer progression, HA-CD44 interaction was then inhibited by incubation with an anti-CD44 antibody. Anti-CD44 antibody impaired invasion into Matrigel by 95%. Taken together, these data suggest that pericellular HA is critical for colon carcinoma cell invasion and that this invasive capability is dependent on interaction with CD44.


Subject(s)
Colonic Neoplasms/pathology , Hyaluronan Receptors/physiology , Hyaluronic Acid/physiology , Binding, Competitive , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , DNA, Antisense/administration & dosage , DNA, Antisense/genetics , Humans , Hyaluronan Receptors/metabolism , Hyaluronic Acid/antagonists & inhibitors , Hyaluronic Acid/genetics , Hyaluronic Acid/metabolism , Neoplasm Invasiveness , Transfection
15.
J Cell Biol ; 165(6): 881-91, 2004 Jun 21.
Article in English | MEDLINE | ID: mdl-15210734

ABSTRACT

Melanoma chondroitin sulfate proteoglycan (MCSP) is an early cell surface melanoma progression marker implicated in stimulating tumor cell proliferation, migration, and invasion. Focal adhesion kinase (FAK) plays a pivotal role in integrating growth factor and adhesion-related signaling pathways, facilitating cell spreading and migration. Extracellular signal-regulated kinase (ERK) 1 and 2, implicated in tumor growth and survival, has also been linked to clinical melanoma progression. We have cloned the MCSP core protein and expressed it in the MCSP-negative melanoma cell line WM1552C. Expression of MCSP enhances integrin-mediated cell spreading, FAK phosphorylation, and activation of ERK1/2. MCSP transfectants exhibit extensive MCSP-rich microspikes on adherent cells, where it also colocalizes with alpha4 integrin. Enhanced activation of FAK and ERK1/2 by MCSP appears to involve independent mechanisms because inhibition of FAK activation had no effect on ERK1/2 phosphorylation. These results indicate that MCSP may facilitate primary melanoma progression by enhancing the activation of key signaling pathways important for tumor invasion and growth.


Subject(s)
Chondroitin Sulfate Proteoglycans/pharmacology , Membrane Proteins/pharmacology , Mitogen-Activated Protein Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Amino Acid Sequence , Cell Line, Tumor , Chondroitin Sulfate Proteoglycans/chemistry , Chondroitin Sulfate Proteoglycans/genetics , Enzyme Activation/drug effects , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , Humans , Melanoma , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Molecular Sequence Data , Sequence Alignment , Sequence Homology, Amino Acid
16.
J Invest Dermatol ; 122(1): 167-76, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14962105

ABSTRACT

Membrane type-I metalloproteinase (MT1-MMP) is a transmembrane metalloproteinase that is critical for tumor cell invasion. MT1-MMP can degrade extracellular matrix (ECM) proteins directly and/or indirectly by activating soluble MMPs such as pro-MMP-2. Although MT1-MMP is upregulated in malignant melanoma, the biological consequences of elevated MT1-MMP expression for tumor progression are not entirely understood. In the current study, we have utilized the Bowes melanoma line for evaluating MT1-MMP in invasion and growth. Our studies extend the earlier observations to demonstrate that MT1-MMP expression in Bowes melanoma cells promotes selective invasion into matrigel but not matrices consisting of type-I collagen. Furthermore, MT1-MMP expressing melanoma cells exhibit increased migration in response to laminin 1 but not to type-I or type-IV collagen. MT1-MMP expression results in enhanced 3 dimensional growth in agarose gels and in long-term cultures within matrigel. The hydroxymate inhibitor BB94 inhibits MT1-MMP enhanced invasion and growth in 3 dimensional culture systems, but had no effect on increased motility. We demonstrated that MT1-MMP expression significantly facilitated tumorigenicity and growth by intradermal injection. The results suggest a more general role for elevated MT1-MMP in promoting both the selective invasion and increased growth of malignant melanoma in vivo.


Subject(s)
Melanoma/pathology , Metalloendopeptidases/metabolism , Skin Neoplasms/pathology , Animals , Biocompatible Materials , Cell Division , Cell Line, Tumor , Collagen , Collagen Type I , Collagen Type IV , Drug Combinations , Enzyme Precursors/genetics , Female , Fibrosarcoma , Gelatinases/genetics , Humans , Injections, Intradermal , Laminin , Matrix Metalloproteinase 14 , Matrix Metalloproteinases, Membrane-Associated , Metalloendopeptidases/genetics , Mice , Mice, Inbred NOD , Neoplasm Invasiveness , Neoplasm Transplantation , Proteoglycans , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...