Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Res ; 69(6): 2260-8, 2009 Mar 15.
Article in English | MEDLINE | ID: mdl-19276390

ABSTRACT

The external pH of solid tumors is acidic as a consequence of increased metabolism of glucose and poor perfusion. Acid pH has been shown to stimulate tumor cell invasion and metastasis in vitro and in cells before tail vein injection in vivo. The present study investigates whether inhibition of this tumor acidity will reduce the incidence of in vivo metastases. Here, we show that oral NaHCO(3) selectively increased the pH of tumors and reduced the formation of spontaneous metastases in mouse models of metastatic breast cancer. This treatment regimen was shown to significantly increase the extracellular pH, but not the intracellular pH, of tumors by (31)P magnetic resonance spectroscopy and the export of acid from growing tumors by fluorescence microscopy of tumors grown in window chambers. NaHCO(3) therapy also reduced the rate of lymph node involvement, yet did not affect the levels of circulating tumor cells, suggesting that reduced organ metastases were not due to increased intravasation. In contrast, NaHCO(3) therapy significantly reduced the formation of hepatic metastases following intrasplenic injection, suggesting that it did inhibit extravasation and colonization. In tail vein injections of alternative cancer models, bicarbonate had mixed results, inhibiting the formation of metastases from PC3M prostate cancer cells, but not those of B16 melanoma. Although the mechanism of this therapy is not known with certainty, low pH was shown to increase the release of active cathepsin B, an important matrix remodeling protease.


Subject(s)
Breast Neoplasms/drug therapy , Melanoma, Experimental/drug therapy , Prostatic Neoplasms/drug therapy , Sodium Bicarbonate/pharmacology , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cathepsin B/antagonists & inhibitors , Cathepsin B/metabolism , Cell Line, Tumor , Female , Humans , Hydrogen-Ion Concentration , Liver Neoplasms/metabolism , Liver Neoplasms/prevention & control , Liver Neoplasms/secondary , Lung Neoplasms/metabolism , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Male , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Nude , Mice, SCID , Neoplasm Invasiveness , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology
2.
Clin Exp Metastasis ; 26(4): 299-309, 2009.
Article in English | MEDLINE | ID: mdl-19082919

ABSTRACT

The roles of proteases in cancer are dynamic. Furthermore, the roles or functions of any one protease may differ from one stage of cancer to another. Proteases from tumor-associated cells (e.g., fibroblasts, inflammatory cells, endothelial cells) as well as from tumor cells make important contributions to 'tumor proteolysis'. Many tumors exhibit increases in expression of proteases at the level of transcripts and protein; however, whether those proteases play causal roles in malignant progression is known for only a handful of proteases. What the critical substrate or substrates that are cleaved in vivo by any given protease is also known for only a few proteases. Therefore, the recent development of techniques and reagents for live cell imaging of protease activity, in conjunction with informed knowledge of critical natural substrates, should help to define protease functions. Here we describe live cell assays for imaging proteolysis, protocols for quantifying proteolysis and the use of such assays to follow the dynamics of proteolysis by tumor cells alone and tumor cells interacting with other cells found in the tumor microenvironment. In addition, we describe an in vitro model that recapitulates the architecture of the mammary gland, a model designed to determine the effects of dynamic interactions with the surrounding microenvironment on 'tumor proteolysis' and the respective contributions of various cell types to 'tumor proteolysis'. The assays and models described here could serve as screening platforms for the identification of proteolytic pathways that are potential therapeutic targets and for further development of technologies and imaging probes for in vivo use.


Subject(s)
Diagnostic Imaging , Neoplasms/diagnosis , Neoplasms/metabolism , Peptide Hydrolases/metabolism , Animals , Humans
3.
Mol Imaging ; 7(5): 199-213, 2008.
Article in English | MEDLINE | ID: mdl-19123990

ABSTRACT

The ability of tumor cells to adhere to, migrate on, and remodel extracellular matrices is mediated by cell surface receptors such as beta1-integrins. Here we conducted functional live-cell imaging in real time to investigate the effects of modulating beta1-integrin expression and function on proteolytic remodeling of the extracellular matrix. Human breast and prostate cancer cells were grown on reconstituted basement membrane containing a quenched fluorescent form of collagen IV. Generation of cleavage products and the resulting increases in fluorescence were imaged and quantified. Decreases in the expression and activity of beta1-integrin reduced digestion of quenched fluorescent-collagen IV by the breast and prostate cancer cells and correspondingly their invasion through and migration on reconstituted basement membrane. Decreased extracellular matrix degradation also was associated with changes in the constituents of proteolytic pathways: decreases in secretion of the cysteine protease cathepsin B, the matrix metalloproteinase (MMP)-13, and tissue inhibitors of metalloproteinases (TIMP)-1 and 2; a decrease in expression of MMP-14 or membrane type 1 MMP; and an increase in secretion of TIMP-3. This is the first study to demonstrate through functional live-cell imaging that downregulation of beta1-integrin expression and function reduces proteolysis of collagen IV by breast and prostate cancer cells.


Subject(s)
Breast Neoplasms/metabolism , Collagen Type IV/metabolism , Diagnostic Imaging , Integrin beta1/metabolism , Prostatic Neoplasms/metabolism , Breast Neoplasms/pathology , Cathepsin B/analysis , Cathepsin B/metabolism , Cell Adhesion/physiology , Cell Line, Tumor , Cell Movement/physiology , Culture Media, Conditioned/metabolism , DNA/metabolism , Female , Humans , Immunohistochemistry , Male , Neoplasm Invasiveness/physiopathology , Prostatic Neoplasms/pathology
4.
Protein Expr Purif ; 52(2): 463-71, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17208452

ABSTRACT

Recombinant human cysteine protease inhibitor, stefin A, was expressed in both Escherichia coli and BS-C-1 monkey kidney cells utilizing pET and recombinant vaccinia virus systems, respectively. The expressed protein was purified and analyzed by SDS-PAGE and Western blot analysis utilizing a polyclonal antibody against rat cystatin alpha. In both cases the purified protein appeared as a single band corresponding to the molecular weight of stefin A ( approximately 10kDa). Viability of the expressed stefin A was determined by the inhibition of the plant cysteine protease, papain. Recombinant human stefin A expressed in both E. coli and BS-C-1 cells, was shown to almost completely inhibit papain. The expression of a fully functional recombinant human stefin A in the bacterial system provides a highly efficient tool for the production of large quantities of the protein. This can be an important tool in kinetic studies as well as in production of antibodies for other analytical studies (immunoblot, immunohistochemical studies, etc.). Expression in the mammalian cells, on the other hand, can provide a significant research tool to study the functional roles of stefin A in mammalian systems such as regulation of cysteine proteases.


Subject(s)
Cystatins/metabolism , Cysteine Proteinase Inhibitors/metabolism , Amino Acid Sequence , Animals , Cells, Cultured , Cystatin B , Cystatins/genetics , Cystatins/pharmacology , Cysteine Proteinase Inhibitors/genetics , Cysteine Proteinase Inhibitors/pharmacology , Escherichia coli/genetics , Gene Expression , Haplorhini , Humans , Molecular Sequence Data , Rats , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Sequence Homology, Amino Acid
5.
J Cell Sci ; 118(Pt 7): 1493-503, 2005 Apr 01.
Article in English | MEDLINE | ID: mdl-15769846

ABSTRACT

Cathepsin B and pro-urokinase plasminogen activator (pro-uPA) localize to the caveolae of HCT 116 human colorectal carcinoma cells, an association mediated by active K-RAS. In this study, we established a stable HCT 116 cell line with a gene encoding antisense caveolin-1 (AS-cav-1) to examine the effects of caveolin-1, the main structural protein of caveolae, on the expression and localization of cathepsin B and pro-uPA, and their cell-surface receptors p11 and uPA receptor (uPAR), respectively. AS-cav-1 HCT 116 cells secreted less procathepsin B than control (empty vector) cells as measured by immunoblotting and pepsin activation of the proenzyme. Expression and secretion of pro-uPA was also downregulated in AS-cav-1 HCT 116 cells. Localization of cathepsin B and pro-uPA to caveolae was reduced in AS-cav-1 HCT 116 cells, and these cells expressed less total and caveolae-associated p11 and uPAR compared with control cells. Previous studies have shown that uPAR forms a complex with caveolin-1 and beta1-integrin, and we here show that downregulation of caveolin-1 also suppressed the localization of beta1-integrin to caveolae of these cells. Finally, downregulation of caveolin-1 in HCT 116 cells inhibited degradation of the extracellular matrix protein collagen IV and the invasion of these cells through Matrigel. Based on these results, we hypothesize that caveolin-1 affects the expression and localization of cathepsin B and pro-uPA, and their receptors, thereby mediating cell-surface proteolytic events associated with invasion of colon cancer cells.


Subject(s)
Cathepsin B/metabolism , Caveolins/metabolism , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Receptors, Cell Surface/metabolism , Urokinase-Type Plasminogen Activator/metabolism , Animals , Cathepsin B/genetics , Caveolin 1 , Caveolins/genetics , Cell Line, Tumor , Collagen Type IV/metabolism , Colonic Neoplasms/enzymology , HCT116 Cells , Humans , Mice , NIH 3T3 Cells , Receptors, Cell Surface/genetics , Receptors, Urokinase Plasminogen Activator , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Urokinase-Type Plasminogen Activator/genetics
6.
Semin Cancer Biol ; 15(2): 149-57, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15652460

ABSTRACT

Tumor-stromal interactions induce expression of matrix metalloproteinases and serine proteases and, as shown recently, the cysteine protease cathepsin B. We speculate that such interactions upregulate the transcription factor Ets1, resulting in increased cathepsin B expression. This would be consistent with the observed concomitant upregulation of matrix metalloproteinases and serine proteases as well as with the ability of extracellular matrices and their binding partners to alter cathepsin B expression and secretion. Using a confocal assay to analyze the contribution of tumor-stromal interactions to proteolysis, we have been able to confirm enhanced degradation of extracellular matrices by all three classes of proteases.


Subject(s)
Cathepsin B/metabolism , Neoplasms/enzymology , Animals , Cathepsin B/analysis , Cell Communication/physiology , Humans , Mice , Neoplasms/pathology , Stromal Cells/physiology
7.
Mol Imaging ; 2(3): 159-75, 2003 Jul.
Article in English | MEDLINE | ID: mdl-14649059

ABSTRACT

The underlying basement membrane is degraded during progression of breast and colon carcinoma. Thus, we imaged degradation of a quenched fluorescent derivative of basement membrane type IV collagen (DQ-collagen IV) by living human breast and colon tumor spheroids. Proteolysis of DQ-collagen IV by HCT 116 and HKh-2 human colon tumor spheroids was both intracellular and pericellular. In contrast, proteolysis of DQ-collagen IV by BT20 human breast tumor spheroids was pericellular. As stromal elements can contribute to proteolytic activities associated with tumors, we also examined degradation of DQ-collagen IV by human monocytes/macrophages and colon and breast fibroblasts. Fibroblasts themselves exhibited a modest amount of pericellular degradation. Degradation was increased 4-17-fold in cocultures of fibroblasts and tumor cells as compared to either cell type alone. Inhibitors of matrix metalloproteinases, plasmin, and the cysteine protease, cathepsin B, all reduced degradation in the cocultures. Monocytes did not degrade DQ-collagen IV; however, macrophages degraded DQ-collagen IV intracellularly. In coculture of tumor cells, fibroblasts, and macrophages, degradation of DQ-collagen IV was further increased. Imaging of living tumor and stromal cells has, thus, allowed us to establish that tumor proteolysis occurs pericellularly and intracellularly and that tumor, stromal, and inflammatory cells all contribute to degradative processes.


Subject(s)
Endopeptidases/metabolism , Image Processing, Computer-Assisted , Macrophages/metabolism , Neoplasms/metabolism , Spheroids, Cellular/metabolism , Stromal Cells/enzymology , Breast Neoplasms/pathology , Carcinoma/pathology , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Movement , Cell Survival , Coculture Techniques , Collagen/metabolism , Collagen Type IV/metabolism , Colonic Neoplasms/pathology , Drug Combinations , Endopeptidases/drug effects , Humans , Hydroxamic Acids/pharmacology , Immunohistochemistry , Laminin/metabolism , Macrophages/cytology , Monocytes/drug effects , Neoplasms/enzymology , Neoplasms/pathology , Proteoglycans/metabolism , Spheroids, Cellular/enzymology , Stromal Cells/cytology , Stromal Cells/immunology , Tetradecanoylphorbol Acetate/pharmacology , U937 Cells
8.
Neoplasia ; 5(6): 507-19, 2003.
Article in English | MEDLINE | ID: mdl-14965444

ABSTRACT

Cathepsin B protein and activity are known to localize to the basal plasma membrane of colon carcinoma cells following the appearance of K-ras mutations. Using immunofluorescence and subcellular fractionation techniques and two human colon carcinoma cell lines - one with a mutated K-ras allele (HCT 116) and a daughter line in which the mutated allele has been disrupted (HKh-2)-we demonstrate that the localization of cathepsin B to caveolae on the surface of these carcinoma cells is regulated by mutant K-ras. In HCT 116 cells, a greater percentage of cathepsin B was distributed to the caveolae, and the secretion of cathepsin B and pericellular (membrane-associated and secreted) cathepsin B activity were greater than observed in HKh-2 cells. Previous studies established the light chain of annexin II tetramer, p11, as a binding site for cathepsin B on the surface of tumor cells. The deletion of active K-ras in HKh-2 cells reduced the steady-state levels of p11 and caveolin-1 and the distribution of p11 to caveolae. Based upon these results, we speculate that cathepsin B, a protease implicated in tumor progression, plays a functional role in initiating proteolytic cascades in caveolae as downstream components of this cascade (e.g., urokinase plasminogen activator and urokinase plasminogen activator receptor) are also present in HCT 116 caveolae.


Subject(s)
Annexin A2/metabolism , Cathepsin B/metabolism , Cell Membrane/metabolism , Colorectal Neoplasms/metabolism , Genes, ras/genetics , S100 Proteins/metabolism , Annexin A2/genetics , Caveolae/metabolism , Caveolin 1 , Caveolins/genetics , Caveolins/metabolism , Cell Line, Tumor , Colorectal Neoplasms/genetics , Electrophoresis, Polyacrylamide Gel , Fluorescent Antibody Technique , Humans , Immunoblotting , Immunohistochemistry , Microscopy, Confocal , Mutation , Protein Transport/physiology , Receptors, Cell Surface/metabolism , Receptors, Urokinase Plasminogen Activator , S100 Proteins/genetics , Urokinase-Type Plasminogen Activator/metabolism
9.
J Biol Chem ; 277(35): 32220-7, 2002 Aug 30.
Article in English | MEDLINE | ID: mdl-12072442

ABSTRACT

Interactions of stromal and tumor cells with the extracellular matrix may regulate expression of proteases including the lysosomal proteases cathepsins B and D. In the present study, we determined whether the expression of these two proteases in human breast fibroblasts was modulated by interactions with the extracellular matrix component, collagen I. Breast fibroblasts were isolated from non-malignant breast tissue as well as from tissue surrounding malignant human breast tumors. Growth of these fibroblasts on collagen I gels affected cell morphology, but not the intracellular localization of vesicles staining for cathepsin B or D. Cathepsins B and D levels (mRNA or intracellular protein) were not affected in fibroblasts growing on collagen I gels or plastic, nor was cathepsin D secreted from these cells. In contrast, protein expression and secretion of cathepsin B, primarily procathepsin B, was induced by growth on collagen I gels. The induced secretion appeared to be mediated by integrins binding to collagen I, as inhibitory antibodies against alpha(1), alpha(2), and beta(1) integrin subunits prevented procathepsin B secretion from fibroblasts grown on collagen. In addition, procathepsin B secretion was induced when cells were plated on beta(1) integrin antibodies. To our knowledge, this is the first examination of cathepsin B and D expression and localization in human breast fibroblasts and their regulation by a matrix protein. Secretion of the cysteine protease procathepsin B from breast fibroblasts may have physiological and pathological consequences, as proteases are required for normal development and for lactation of the mammary gland, yet can also initiate and accelerate the progression of breast cancer.


Subject(s)
Breast Neoplasms/enzymology , Breast/cytology , Cathepsin B/genetics , Cathepsin B/metabolism , Collagen Type I/physiology , Enzyme Precursors/metabolism , Fibroblasts/enzymology , Breast Neoplasms/pathology , Cell Division , Cells, Cultured , Culture Media, Conditioned , Female , Fibroblasts/cytology , Gene Expression Regulation, Enzymologic , Humans , Integrins/biosynthesis , Transcription, Genetic , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...