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










Publication year range
3.
Int J Oncol ; 17(4): 629-41, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10995872

ABSTRACT

The metastatic process requires changes in tumor cell adhesion properties, cell motility and remodeling of the extracellular matrix. The erbB2 proto-oncogene is overexpressed in approximately 30% of breast cancers and is a major prognostic parameter when present in invasive disease. A ligand for the erbB2 receptor has not yet been identified but it can be activated by heterodimerization with heregulin (HRG)-stimulated erbB3 and erbB4 receptors. The HRGs are a family of polypeptide growth factors that have been shown to play a role in embryogenesis, tumor formation, growth and differentiation of breast cancer cells. The erbB3 and erbB4 receptors are involved in transregulation of erbB2 signaling. The work presented here suggests biological roles for HRG including regulation of the actin cytoskeleton and induction of motility and invasion in breast cancer cells. HRG-expressing breast cancer cell lines are characterized by low erbB receptor levels and a high invasive and metastatic index, while those which overexpress erbB2 demonstrate minimal invasive potential in vitro and are non-tumorigenic in vivo. Treatment of the highly tumorigenic and metastatic HRG-expressing breast cancer cell line MDA-MB-231 with an HRG-neutralizing antibody significantly inhibited proliferation in culture and motility in the Boyden chamber assay. Addition of exogenous HRG to non-invasive erbB2 overexpressing cells (SKBr-3) at low concentrations induced formation of pseudopodia, enhanced phagocytic activity and increased chemomigration and invasion in the Boyden chamber assay. The specificity of the chemomigration response to HRG is demonstrated by inhibition with the anti-HRG neutralizing antibody. These results suggest that either HRG can act as an autocrine or paracrine ligand to promote the invasive behavior of breast cancer cells in vitro or thus may enhance the metastatic process in vivo.


Subject(s)
Actins/drug effects , Breast Neoplasms/pathology , Cytoskeleton/drug effects , Neoplasm Invasiveness/pathology , Neuregulin-1/pharmacology , Actins/metabolism , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Breast Neoplasms/genetics , Cell Differentiation/drug effects , Cell Division/drug effects , Cell Line , Cell Movement/drug effects , Chemotaxis/drug effects , Collagen , Cytoskeleton/metabolism , Dose-Response Relationship, Drug , Drug Combinations , Gene Expression Regulation, Neoplastic , Humans , Laminin , Neoplasm Invasiveness/genetics , Neuregulin-1/genetics , Neuregulin-1/immunology , Phagocytosis/drug effects , Phenotype , Proteoglycans , Proto-Oncogene Mas , Pseudopodia/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins/immunology , Recombinant Proteins/pharmacology , Tumor Cells, Cultured/cytology , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism
4.
Exp Cell Res ; 248(1): 260-71, 1999 Apr 10.
Article in English | MEDLINE | ID: mdl-10094832

ABSTRACT

Cellular adhesions to other cells and to the extracellular matrix play crucial roles in the malignant progression of cancer. In this study, we investigated the role of protein kinase C (PKC) in the regulation of cell-substratum adhesion by the breast adenocarcinoma cell line MCF-7. A PKC activator, 12-O-tetradecanoylphorbol-l, 3-acetate (TPA), stimulated cell adhesion to laminin and collagen I in a dose-dependent manner over a 1- to 4-h interval. This enhanced adhesion was mediated by alpha2beta1 integrin, since both anti-alpha2 and anti-beta1 blocking antibodies each completely abrogated the TPA-induced adhesion. FACS analysis determined that TPA treatment does not change the cell surface expression of alpha2beta1 integrin over a 4-h time interval. However, alpha2beta1 levels were increased after 24 h of TPA treatment. Thus, the enhanced avidity of alpha2beta1-dependent cellular adhesion preceded the induction of alpha2beta1 cell surface expression. Northern blot analysis revealed that mRNA levels of both alpha2 and beta1 subunits were increased after exposure to TPA for 4 h, indicating that the induction of alpha2beta1 mRNA preceded that of its cell surface expression. This further suggested that the TPA-induced avidity of alpha2beta1 was independent of increased expression of alpha2beta1. Pretreatment of cells with the PKC inhibitor calphostin C partially antagonized the TPA-induced increase in expression of alpha2beta1 integrin expression and of alpha2beta1-mediated cellular adhesion. To identify a possible mechanism by which TPA could be acting to promote the rapid induction of alpha2beta1 adhesion, we treated the cells with the Rho-GTPase inhibitor Clostridium botulinumexotoxin C3. C3 inhibited TPA-induced adhesion to laminin and collagen I in a dose-dependant manner, suggesting a likely role for Rho in TPA-induced adhesion. Together, these results suggest that PKC can modulate the alpha2beta1-dependent adhesion of MCF-7 cells by two distinct mechanisms: altering the gene expression of integrins alpha2 and beta1 and altering the avidity of the alpha2beta1 integrin by a Rho-dependant mechanism.


Subject(s)
Botulinum Toxins , Integrins/biosynthesis , Protein Kinase C/physiology , ADP Ribose Transferases/metabolism , ADP Ribose Transferases/pharmacology , Animals , Breast Neoplasms , Cell Adhesion/drug effects , Enzyme Activation , Enzyme Inhibitors/pharmacology , Female , Gene Expression Regulation/drug effects , Humans , Integrins/genetics , Mice , Naphthalenes/pharmacology , Protein Kinase C/antagonists & inhibitors , Rats , Receptors, Collagen , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Tetradecanoylphorbol Acetate/pharmacology , Tumor Cells, Cultured
5.
Clin Chim Acta ; 279(1-2): 77-96, 1999 Jan.
Article in English | MEDLINE | ID: mdl-10064120

ABSTRACT

Proton MR spectra and biochemical assays have been recorded on the sera of 40 patients and ten controls in order to document the correlation between spectroscopic and biochemical variations in selected pathologies (cancer, inflammatory and infectious diseases, diabetes). N-acetyl proton resonances are essentially generated by the N-acetyl residues of the glucidic moieties borne by the most abundant acute-phase proteins (alpha1-acid glycoprotein, alpha1-antitrypsin and haptoglobin). These resonances are not correlated to immunoglobulins A, G and M levels. Principal component analysis shows that variations in spectroscopic and biochemical data are independent markers of the inflammatory status of patients but no additional sensitivity or specificity is obtained when the two sets of data are combined.


Subject(s)
Acute-Phase Proteins/analysis , Acute-Phase Reaction/blood , Infections/blood , Acute-Phase Reaction/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Child , Child, Preschool , Female , Humans , Immunoglobulins/blood , Infections/pathology , Inflammation/blood , Inflammation/pathology , Male , Middle Aged , Nuclear Magnetic Resonance, Biomolecular , Statistics as Topic/methods
6.
Exp Cell Res ; 247(1): 105-13, 1999 Feb 25.
Article in English | MEDLINE | ID: mdl-10047452

ABSTRACT

We investigated a potentially central role of protein kinase C (PKC) in controlling multiple pathways in breast cancer cell invasiveness. To do this we evaluated the ability of pharmacologic agents that alter PKC activity to regulate the behavior of the poorly invasive human breast cancer cell line MCF-7. Treatment with 12-O-tetradecanoylphorbol-13-acetate (TPA) produced a dramatic induction of the invasiveness of these cells (18-fold), an effect that concurrent treatment with the PKC inhibitor Bryostatin-1 was able to block. To characterize alterations in the cellular properties that might be responsible for these effects we measured the impact of these two agents on a number of processes thought to be important for invasiveness. The motility of the cells was first examined; it was markedly increased by treatment with TPA (20-fold) and again, Bryostatin-1 inhibited this stimulation. We next examined the expression of MMP-1, 3, 9, 10, and 11 (matrix metalloproteinases), all of which have been shown to be PKC responsive in other systems. We found that the expression and secretion of MMP-9 were increased by at least 100-fold, though all of the enzyme secreted was in the latent form. Finally, the expression of both urokinase plasminogen activator (UPA) and its receptor (UPAR) were induced after TPA treatment by 8- and 7-fold, respectively. In conclusion, we have shown that stimulation of PKC activity markedly increases the invasiveness of MCF-7 cells, and that this change in behavior is correlated with a coordinated set of biochemical and cellular changes which are likely to contribute to this process. These data highlight the possible utility of PKC inhibitors such as Bryostatin-1 as anti-invasive and/or antimetastatic agents. Bryostatin-1 is currently in early clinical trials as an anticancer agent.


Subject(s)
Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cell Movement , Endopeptidases/biosynthesis , Protein Kinase C/physiology , Bryostatins , Cathepsin D/biosynthesis , Enzyme Activation/drug effects , Humans , Lactones/pharmacology , Macrolides , Metalloendopeptidases/biosynthesis , Neoplasm Invasiveness , Receptors, Cell Surface/biosynthesis , Receptors, Urokinase Plasminogen Activator , Tetradecanoylphorbol Acetate/pharmacology , Tumor Cells, Cultured , Urokinase-Type Plasminogen Activator/biosynthesis , Urokinase-Type Plasminogen Activator/metabolism
7.
Oncogene ; 16(21): 2755-66, 1998 May 28.
Article in English | MEDLINE | ID: mdl-9652742

ABSTRACT

We have examined defects in mammary development and tumorigenesis in a transgenic model expressing the c-myc gene under the MMTV-LTR promoter. The stochastic tumors which arise from hyperplastic ductal and lobular lesions in this model are characterized by high rates both of apoptosis and of chromosomal instability. Since the p53 gene product is thought to be central in the maintenance of genomic integrity, in part due to its ability to induce apoptosis in cells harboring DNA damage, we examined its expression and possible mutation. Initially, we observed that unmutated p53 is strongly expressed in premalignant mammary glands and in mammary tumors derived from the MMTV-c-myc strain. We then mated the MMTV-myc strain to a p53-deficient strain as a means of examining the effect of this lesion on mammary development and tumorigenesis in the context of c-myc overexpression. A lack of both p53 alleles in the presence of c-myc overexpression resulted in a dramatic hyerplastic alteration in mammary gland development. Specifically, in female bitransgenic MMTV-c-myc/p53 null mice (MMTV-myc/p53(-/-)), lobular hyperplasias were observed at almost every ductal end bud as early as 32 days of age. In contrast, only mild ductal and lobular hyperplasias were seen in MMTV-myc mice that contained both p53 alleles (MMTV-myc/p53(+/+)); an intermediate phenotype occurred in mice with a single intact (MMTV-myc/p53(+/-)) p53 allele. Mammary carcinomas arose with a high frequency in MMTV-myc/p53(+/-) mice; the tumors were comparable in frequency, histology and apoptotic index to the tumors in MMTV-myc/p53(+/+) mice. Also, as previously observed (Elson et al., 1995), lymphomas arose with extremely short latency in MMTV-myc/p53(-/-) mice, precluding study of the fate of their hyperplastic mammary lesions in situ. The frequency of p53 mutations in MMTV-myc/p53(+/+) and MMTV-myc/p53(+/-) mammary tumors and in cell lines derived from these tumors was examined by direct sequencing. No point mutations or deletions in p53 were observed in mammary tumors or cell lines from either genotype. Finally, a detailed chromosomal analysis using multicolor spectral karyotyping (SKY) revealed that there were multiple chromosomal alterations in the c-myc-overexpressing cells that contained either one or two unmutated p53 alleles. Variable ploidy changes, a common translocation of chromosome 11, and other chromosomal aberrations were observed. Our data thus support an interaction between c-Myc and p53 in mammary development, but suggest that loss of p53 is required neither for c-myc-dependent tumorigenesis nor for c-myc-dependent chromosomal instability.


Subject(s)
Genes, myc/genetics , Genes, p53/genetics , Mammary Neoplasms, Experimental/genetics , Animals , Apoptosis , Cells, Cultured , Epithelial Cells/cytology , Female , Gene Expression , Hyperplasia , Karyotyping , Mammary Glands, Animal/growth & development , Mammary Glands, Animal/pathology , Mammary Neoplasms, Experimental/pathology , Mammary Tumor Virus, Mouse/genetics , Mice , Mice, Transgenic , Sequence Analysis, DNA , Transgenes
8.
J Clin Oncol ; 16(6): 2150-6, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9626215

ABSTRACT

PURPOSE: This phase I study was performed to evaluate the safety and pharmacokinetics of escalating doses of Marimastat (British Biotech, Inc, Oxford, United Kingdom) in patients with advanced malignancies and to determine the phase II recommended dose to be used in subsequent studies. PATIENTS AND METHODS: A standard phase I design was used in this study, in which consecutive groups of three patients were treated with escalating doses of the study drug. Marimastat was administered orally at 25, 50, or 100 mg twice daily to consecutive groups of patients with advanced lung cancer. An additional three patients were added at the highest dose studied (100 mg orally twice daily) to assess whether the inflammatory polyarthitis observed at that dose level can be prevented by a concurrent administration of nonsteroidal antiinflammatory drugs (NSAIDS) and/or low-dose corticosteroids. Blood was drawn for safety monitoring, pharmacokinetic analysis, and plasma levels of metalloproteinase (MMP)-2 and MMP-9 (determined by zymography). A total of 12 patients were studied. RESULTS: The most significant toxicity at the highest dose studied (100 mg orally twice daily) was a symptomatic inflammatory polyarthritis that persisted for up to 8 weeks after discontinuation of the study drug and was dose-limiting. The estimated plasma elimination half-life of Marimastat was 4 to 5 hours. The mean maximum concentration (Cmax) at a reasonably well-tolerated dose (50 mg orally twice daily) was 196 ng/mL and was reached within 1 to 2 hours (Tmax) after administration. Areas under the curve (AUC) tended to correlate with the dose of Marimastat. Zymographic analysis of peripheral-blood ratios of activated proenzymatic forms of MMP-2 and -9 did not show any consistent patterns of change in MMP levels or in a degree of their activation during the course of treatment. CONCLUSION: Marimastat was well absorbed from the gastrointestinal tract, with high levels of the study drug detected in plasma within hours after drug administration. Plasma concentrations of Marimastat achieved at dose levels 2 and 3 (50 mg and 100 mg orally twice daily) were substantially higher than those required for MMP inhibition in vitro. The dose-limiting toxicity (DLT) was severe inflammatory polyarthritis, which seemed to be a cumulative toxicity.


Subject(s)
Enzyme Inhibitors/administration & dosage , Hydroxamic Acids/administration & dosage , Lung Neoplasms/drug therapy , Metalloendopeptidases/antagonists & inhibitors , Administration, Oral , Aged , Arthritis/chemically induced , Collagenases/blood , Enzyme Inhibitors/adverse effects , Enzyme Inhibitors/pharmacokinetics , Female , Gelatinases/blood , Humans , Hydroxamic Acids/adverse effects , Hydroxamic Acids/pharmacokinetics , Male , Matrix Metalloproteinase 2 , Matrix Metalloproteinase 9 , Metalloendopeptidases/adverse effects , Metalloendopeptidases/blood , Middle Aged , Treatment Outcome
9.
J Biol Chem ; 272(14): 9147-52, 1997 Apr 04.
Article in English | MEDLINE | ID: mdl-9083044

ABSTRACT

A major, apparently novel extracellular matrix-degrading protease was previously identified and partially isolated from hormone-dependent but not from hormone-independent human breast cancer cells (Shi, Y. E., Torri, J., Yieh, L., Wellstein, A., Lippman, M. E., and Dickson, R. B. (1993) Cancer Res. 53, 1409-1415). Although initially the 80-kDa protease was identified from breast cancer cell-conditioned medium, immunofluorescence staining of breast cancer cells with anti-80-kDa protease monoclonal antibody 21-9 showed that in addition to its detection in intracellular compartments, the protease was uniformly localized around periphery of the cells with more intensive staining on the pseudopodia and membrane ruffles. A surface biotinylation technique confirmed the plasma membrane localization of the protease. In addition, the 80-kDa protease could not be washed from the membrane fraction of homogenized breast cancer cells with high concentrations of salts or with EDTA. The 80-kDa protease may noncovalently associate with other protein(s) to form complexes, the 95- and 110-kDa proteases. Both complexes showed gelatinolytic activity and bore the epitopes recognized by monoclonal antibody 21-9. Furthermore, both complexes could be converted to 80-kDa forms by boiling in SDS in the absence of reducing agents. Expression of this novel, integral membrane gelatinase could allow breast cancer cells an alternative to other previously described matrix-degrading enzymes for degradation of the extracellular matrix in close proximity to their surfaces.


Subject(s)
Antibodies, Monoclonal/metabolism , Breast Neoplasms/enzymology , Endopeptidases/immunology , Extracellular Matrix/metabolism , Gelatinases/isolation & purification , Membrane Proteins/isolation & purification , Antibodies, Monoclonal/isolation & purification , Cell Membrane/enzymology , Chromatography, Affinity , Endopeptidases/analysis , Female , Fluorescent Antibody Technique, Direct , Gelatinases/chemistry , Gelatinases/metabolism , Humans , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Molecular Weight , Tumor Cells, Cultured
10.
Clin Exp Metastasis ; 12(3): 181-94, 1994 May.
Article in English | MEDLINE | ID: mdl-8194193

ABSTRACT

Expression of the intermediate filament protein vimentin, and loss of the cellular adhesion protein uvomorulin (E-cadherin) have been associated with increased invasiveness of established human breast cancer cell lines in vitro and in vivo. In the current study, we have further examined these relationships in oncogenically transformed human mammary epithelial cells. A normal human mammary epithelial strain, termed 184, was previously immortalized with benzo[a]pyrene, and two distinct sublines were derived (A1N4 and 184B5). These sublines were infected with retroviral vectors containing a single or two oncogenes of the nuclear, cytoplasmic, and plasma membrane-associated type (v-rasH, v-rasKi, v-mos, SV40T and c-myc). All infectants have been previously shown to exhibit some aspects of phenotypic transformation. In the current study, cellular invasiveness was determined in vitro using Matrigel, a reconstituted basement membrane extract. Lineage-specific differences were observed with respect to low constitutive invasiveness and invasive changes after infection with ras, despite similar ras-induced transformation of each line. Major effects on cellular invasiveness were observed after infection of the cells with two different oncogenes (v-rasH + SV40T and v-rasH + v-mos). In contrast, the effects of single oncogenes were only modest or negligible. All oncogenic infectants demonstrated increased attachment to laminin, but altered secretion of the 72 kDa and 92 kDa gelatinases was not associated with any aspect of malignant progression. Each of the two highly invasive double oncogene transformants were vimentin-positive and uvomorulin-negative, a phenotype indicative of the epithelial-mesenchymal transition (EMT) previously associated with invasiveness of established human breast cancer cell lines. Weakly invasive untransformed mammary epithelial cells in this study were positive for both vimentin and uvomorulin, suggesting that uvomorulin may over-ride the otherwise vimentin-associated invasiveness.


Subject(s)
Basement Membrane/pathology , Breast/cytology , Genes, ras , Neoplasm Invasiveness , Oncogenes , Animals , Cadherins/metabolism , Cell Adhesion , Cell Transformation, Neoplastic , Chemotaxis , Collagen , Drug Combinations , Epithelial Cells , Gelatinases/metabolism , Genes, mos , Genes, myc , Humans , Laminin , Mice , Mice, Nude , Proteoglycans , Vimentin/metabolism
11.
Breast Cancer Res Treat ; 31(2-3): 325-35, 1994.
Article in English | MEDLINE | ID: mdl-7881109

ABSTRACT

Eighteen breast cancer cell lines were examined for expression of markers of epithelial and fibroblastic differentiation: E-cadherin, desmoplakins, ZO-1, vimentin, keratin and beta 1 and beta 4 integrins. The cell lines were distributed along a spectrum of differentiation from epithelial to fibroblastic phenotypes. The most well-differentiated, epithelioid cell lines contained proteins characteristic of desmosomal, adherens and tight junctions, were adherent to one another on plastic and in the basement membrane matrix Matrigel and were keratin-positive and vimentin-negative. These cell lines were all weakly invasive in an in vitro chemoinvasion assay. The most poorly-differentiated, fibroblastic cell lines were E-cadherin-, desmoplakin- and ZO-1-negative and formed branching structures in Matrigel. They were vimentin-positive, contained only low levels of keratins and were highly invasive in the in vitro chemoinvasion assay. Of all of the markers analyzed, vimentin expression correlated best with in vitro invasive ability and fibroblastic differentiation. In a cell line with unstable expression of vimentin, T47DCO, the cells that were invasive were of the fibroblastic type. The differentiation markers described here may be useful for analysis of clinical specimens and could potentially provide a more precise measure of differentiation grade yielding more power for predicting prognosis.


Subject(s)
Breast Neoplasms/pathology , Neoplasm Invasiveness/pathology , Neoplasm Proteins/analysis , Tumor Cells, Cultured/pathology , 3T3 Cells , Animals , Biomarkers, Tumor/analysis , Breast Neoplasms/chemistry , Cell Differentiation , Chemotaxis , Collagen , Culture Media, Conditioned/pharmacology , Culture Techniques/methods , Drug Combinations , Epithelium/chemistry , Epithelium/pathology , Fibroblasts/chemistry , Fibroblasts/pathology , Humans , Intercellular Junctions/ultrastructure , Laminin , Mice , Proteoglycans , Tumor Cells, Cultured/chemistry , Vimentin/analysis
12.
Int J Oncol ; 4(1): 55-60, 1994 Jan.
Article in English | MEDLINE | ID: mdl-21566889

ABSTRACT

Laminin has been shown to promote the malignant phenotype and the level of the 32/67 Kd laminin receptor has been found to correlate with Dukes' staging of colon cancer. A biopsy of a Dukes' stage B2 human colon carcinoma formed a tumor in a nude mouse after coinjection with Matrigel. The parental tumor and the murine tumor appeared identical at the histological level. A cell line LCC-C1 was established from the murine tumor. The cell line appeared moderately differentiated although it did not produce mucin in vitro; however, the xenograft in vivo did produce low levels of mucin. Laminin adherent and non-adherent cell lines were selected. The parental and the laminin-selected cell subclones adhered equally well to plastic and to fibronectin and showed similar growth rates on plastic. When injected subcutaneously into nude mice, the laminin-adherent cells formed relatively undifferentiated tumors that were twice as large as the parental cell tumors whereas the laminin non adherent cells formed very small, but highly differentiated tumors. These data demonstrate that subpopulations of tumor cells which differ in their tumorigenic properties can be selected based on their adhesion to laminin and thus provide models for studying the mechanisms of tumor growth.

13.
Clin Exp Metastasis ; 11(3): 251-61, 1993 May.
Article in English | MEDLINE | ID: mdl-8472397

ABSTRACT

The level of 67 kDa laminin receptor (67LR) expression on breast and colon tumor cell surfaces was previously shown to be correlated with the capacity of tumor cells to metastasize. In the present work we investigate the effects of progestins and estrogen on the expression of 67LR in two sublines of the T47D human breast cancer cells: weakly tumorigenic, poorly invasive parental T47D cells and a highly tumorigenic, more invasive T47Dco subclone. Immunoblotting with an affinity purified antibody directed against a synthetic peptide recognizes the 67LR in these cells. 67LR expression in the T47Dco subclone is 5.5-fold higher than in their parental T47D cells. Treatment of T47D cells with 1 nM of the synthetic progestin R5020 results in a 4-fold increase in 67LR protein expression. Estrogen also induced 67LR expression, but only by 1.5-fold. The progestin-stimulated expression of the 67LR correlates with a 4.3-fold increase in attachment of T47D cells to laminin. A monoclonal antibody, mAb 13, directed against beta 1 integrin, completely blocks the attachment of T47D cells to fibronectin, only partially inhibits the attachment of T47D cells to laminin, and appears not to affect the progestin-stimulated laminin attachment of T47D cells. A new antiprogestin, ZK 112.993, significantly inhibits both progestin-stimulated 67LR expression and the increased attachment to laminin. These results suggest a possible role for progestin in mediating one of the multiple events thought to be important in metastasis of steroid receptor positive human breast cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/metabolism , Promegestone/antagonists & inhibitors , Promegestone/pharmacology , Receptors, Laminin/biosynthesis , Analysis of Variance , Animals , Basement Membrane/metabolism , Blotting, Western , Breast Neoplasms/pathology , Cell Adhesion/drug effects , Female , Fibronectins/metabolism , Humans , Laminin/metabolism , Mice , Mice, Nude , Mifepristone/analogs & derivatives , Mifepristone/pharmacology , Neoplasm Metastasis , Receptors, Laminin/drug effects , Reproducibility of Results , Tumor Cells, Cultured , Up-Regulation
14.
Cancer Res ; 53(6): 1409-15, 1993 Mar 15.
Article in English | MEDLINE | ID: mdl-8383010

ABSTRACT

A novel matrix-degrading enzyme was identified from human breast cancer cells. This enzyme appears as major gelatinase in hormone-dependent breast cancer cell lines and has as an apparent molecular mass of 80 kDa on gelatin zymography. The 80-kDa enzyme has a unique metal ion specificity. In addition to calcium ions, the gelatinolytic activity can be supported by manganese and/or magnesium. Unlike 92- and 72-kDa gelatinases and other known members of the metalloproteinase family, the 80-kDa protease is not activated by p-aminophenylmercuric acetate and its gelatinolytic activity is not inhibited by tissue inhibitor of metalloproteinase 2. It is active over the pH range 7.5-9.5 with an optimum at pH 8.5. The enzyme degrades gelatin and type IV collagen. The proteolytic activity of the enzyme is inhibited by EDTA and leupeptin. These unique features clearly distinguish the 80-kDa protease from the known 92-and 72-kDa gelatinases. The expression of 80-kDa enzyme can be detected in hormone-dependent human breast cancer cell lines in vitro and in tumors grown from these cells in athymic nude mice.


Subject(s)
Breast Neoplasms/enzymology , Endopeptidases/metabolism , Neoplasms, Hormone-Dependent/enzymology , Pepsin A/metabolism , Amino Acid Sequence , Edetic Acid/pharmacology , Endopeptidases/chemistry , Endopeptidases/isolation & purification , Enzyme Activation , Female , Gelatin/metabolism , Gelatinases , Humans , Hydrogen-Ion Concentration , Molecular Sequence Data , Neoplasm Proteins/pharmacology , Pepsin A/chemistry , Pepsin A/isolation & purification , Phenylmercuric Acetate/analogs & derivatives , Phenylmercuric Acetate/pharmacology , Substrate Specificity , Tissue Inhibitor of Metalloproteinase-2 , Tumor Cells, Cultured
15.
Cancer Res ; 53(4): 873-7, 1993 Feb 15.
Article in English | MEDLINE | ID: mdl-8428368

ABSTRACT

The aspartyl protease cathepsin D has been shown to be a marker of poor prognosis when found at high levels in primary breast tumors. It has been suggested that this is because the production of cathepsin D increases the invasive potential of the tumor cells, thus increasing the probability of metastasis. We have therefore conducted experiments to determine if secreted cathepsin D makes a significant contribution to the invasive phenotype of breast cancer cells in the Boyden chamber assay of invasion, which measures the ability of a cell to invade through an artificial basement membrane. Cathepsin D secretion and Boyden chamber invasiveness were measured in nine clones of the breast cancer cell line MCF-7, and no correlation was found between cathepsin secretion and invasive behavior. Invasion assays were also conducted in the presence of the aspartyl protease inhibitor pepstatin A, and no inhibition of the invasive behavior of cells was seen. Since low-pH environments are required for both the activation of pro-cathepsin D and the activity of the mature enzyme, assays were also conducted in the presence of chloroquine to neutralize the pH in the acidic compartments of the cells. This treatment did not inhibit invasiveness. Cathepsin D secretion by the breast cancer cell lines MDA-MB-231, MDA-MB-435, MDA-MB-435s, MDA-MB-468, SK-Br-3, and MCF-7-ADRr was also measured. Again, there was no correlation with invasion. In fact, cathepsin D levels were inversely correlated with aggressive behavior in vivo and in vitro in previously reported studies. These data suggest that cathepsin D secretion by tumor cells is not an important determinant of the invasiveness of the tumor cells per se. These data also reinforce the view that the poor prognosis in clinical breast cancer linked to high tumor levels of cathepsin D is probably due to high levels of cathepsin D in the stromal components of the tumor such as infiltrating inflammatory cells.


Subject(s)
Breast Neoplasms/pathology , Cathepsin D/metabolism , Breast Neoplasms/metabolism , Cathepsin D/chemistry , Chloroquine/pharmacology , Diffusion Chambers, Culture , Female , Humans , Molecular Weight , Neoplasm Invasiveness , Pepstatins/pharmacology
16.
Clin Exp Metastasis ; 11(1): 15-26, 1993 Jan.
Article in English | MEDLINE | ID: mdl-8380760

ABSTRACT

We have previously isolated a series of MCF-7 human breast cancer cell variants which no longer require estrogen-supplementation for tumor growth in nude mice (Clarke et al. Proc Natl Acad Sci USA 86: 3649-3653, 1989). We now report that these hormone-independent and hormone-responsive variants (MIII, MCF7/LCC1) can invade locally from solid mammary fat pad tumors, and produce primary extensions on the surface of intraperitoneal structures including liver, pancreas, and diaphragm. Both lymphatic and hematogenous dissemination are observed, resulting in the establishing of pulmonary, bone, and renal metastases. The pattern of metastasis by MIII and MCF7/LCC1 cells closely resembles that frequently observed in breast cancer patients, and provides the first evidence of metastasis from MCF-7 cells growing in vivo without supplementary estrogen. The interexperimental incidence of metastases, and the time from cell inoculation to the appearance of metastatic disease are variable. The increased metastatic potential is not associated with an increase in either the level of laminin attachment, laminin receptor mRNA expression, or secreted type IV collagenolytic activity. We also did not detect a significant decrease in the steady-state mRNA levels of the metastasis inhibitor nm23 gene. However, when growing without estrogen in vitro, MCF7/LCC1 cells produce elevated levels of the estrogen-inducible cathepsin D enzyme.


Subject(s)
Breast Neoplasms/pathology , Monomeric GTP-Binding Proteins , Nucleoside-Diphosphate Kinase , Transcription Factors , Animals , Breast Neoplasms/metabolism , Cathepsin D/metabolism , Collagenases/metabolism , Estradiol/pharmacology , Estradiol/physiology , Female , Gelatinases , Humans , Matrix Metalloproteinase 9 , Mice , Mice, Nude , NM23 Nucleoside Diphosphate Kinases , Neoplasm Invasiveness/physiopathology , Neoplasm Metastasis/physiopathology , Neoplasm Proteins/biosynthesis , Neoplasm Transplantation , Pepsin A/metabolism , Phenotype , Protein Biosynthesis , Receptors, Laminin/biosynthesis , Tumor Cells, Cultured
17.
Breast Cancer Res Treat ; 24(3): 241-55, 1993.
Article in English | MEDLINE | ID: mdl-8435479

ABSTRACT

In vitro analyses of basement membrane invasiveness employing Matrigel (a murine tumor extract rich in basement membrane components) have been performed on human breast cancer model systems. Constitutive invasiveness of different human breast cancer (HBC) cell lines has been examined as well as regulation by steroid hormones, growth factors, and oncogenes. Carcinoma cells exhibiting a mesenchymal-like phenotype (vimentin expression, lack of cell border associated uvomorulin) show dramatically increased motility, invasiveness, and metastatic potential in nude mice. These findings support the hypothesis that epithelial to mesenchymal transition (EMT)-like events may be instrumental in the metastatic progression of human breast cancer. The MCF-7 subline MCF-7ADR appears to have undergone such a transition. The importance of such a transition may be reflected in the emergence of vimentin expression as an indicator of poor prognosis in HBC. Matrix degradation and laminin recognition are highlighted as potential targets for antimetastatic therapy, and analyses of laminin attachment and the matrix metalloproteinase (MMP) family in HBC cell lines are summarized. Matrigel-based assays have proved useful in the study of the molecular mechanisms of basement membrane invasiveness, their regulation in HBC cells, and their potential as targets for antimetastatic therapy.


Subject(s)
Breast Neoplasms/pathology , Laminin/metabolism , Metalloendopeptidases/metabolism , Receptors, Estrogen/analysis , Vimentin/analysis , Animals , Basement Membrane/pathology , Breast Neoplasms/chemistry , Breast Neoplasms/enzymology , Breast Neoplasms/metabolism , Collagen , Collagenases/metabolism , Drug Combinations , Epithelium/pathology , Female , Humans , Mesoderm/pathology , Mice , Mice, Nude , Models, Biological , Neoplasm Invasiveness , Proteoglycans , Tumor Cells, Cultured
18.
Cancer Res ; 52(19): 5190-7, 1992 Oct 01.
Article in English | MEDLINE | ID: mdl-1382837

ABSTRACT

We have previously observed that breast cancer cell lines could exhibit either epithelial or fibroblastic phenotypes as reflected by their morphologies and intermediate filament protein expression (C. L. Sommers, D. Walker-Jones, S. E. Heckford, P. Worland, E. Valverius, R. Clark, M. Stampfer, and E. P. Gelmann, Cancer Res., 49:4258-4263, 1989). Fibroblastoid, vimentin-expressing breast cancer cell lines are more invasive in vitro and in vivo (E. W. Thompson, S. Paik, N. Brunner, C. L. Sommers, G. Zugmaier, R. Clarke, T. B. Shima, J. Torri, S. Donahue, M. E. Lippman, G. R. Martin, and R. B. Dickson, J. Cell. Physiol., 150: 534-544, 1992). We hypothesized that a breast cancer cell with an epithelial phenotype could undergo a transition to a fibroblastic phenotype, possibly resulting in more invasive capacity. We now show that two Adriamycin-resistant MCF-7 cell lines and a vinblastine-resistant ZR-75-B cell line have undergone such a transition. Adriamycin-resistant MCF-7 cells express vimentin, have diminished keratin 19 expression, have lost cell adhesion molecule uvomorulin expression, and have reduced formation of desmosomes and tight junctions as determined by reduced immunodetection of their components desmoplakins I and II and zonula occludens (ZO)-1. Other MCF-7 cell lines selected for resistance to vinblastine and to Adriamycin and verapamil did not have these characteristics, indicating that drug selection does not invariably cause these phenotypic changes. In addition, to determine if vimentin expression in MCF-7 cells alone could manifest a fibroblastic phenotype, we transfected the full-length human vimentin complementary DNA into MCF-7 cells. Although vimentin expression was achieved in MCF-7 cells, it did not affect the phenotype of the cells in terms of the distribution of keratins, desmoplakins I and II, ZO-1, or uvomorulin or in terms of in vitro invasiveness. We conclude that vimentin expression is a marker for a fibroblastic and invasive phenotype in breast cancer cells but does not by itself give rise to this phenotype.


Subject(s)
Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Doxorubicin/pharmacology , Vimentin/genetics , Vinblastine/pharmacology , Antibodies , Breast Neoplasms/pathology , Cell Adhesion Molecules/genetics , Cytoskeletal Proteins/analysis , Desmoplakins , Drug Resistance , Epithelium/pathology , Epithelium/physiology , Fibroblasts/pathology , Fibroblasts/physiology , Humans , Intercellular Junctions/physiology , Intermediate Filaments/chemistry , Intermediate Filaments/pathology , Keratins/genetics , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Phenotype , Staining and Labeling/methods , Transfection , Tumor Cells, Cultured , Verapamil/pharmacology
19.
Biochimie ; 74(9-10): 801-7, 1992.
Article in English | MEDLINE | ID: mdl-1467339

ABSTRACT

High resolution NMR spectroscopy of physiological fluids provides quantitative, qualitative and dynamic information on the metabolic status of the interstitial and plasma compartments under a variety of pathophysiological conditions. The simultaneous detection and quantitation by NMR spectroscopy of numerous compounds of the intermediary metabolism offers a new insight in the understanding of the milieu intérieur. NMR spectroscopy of physiological fluids offers a unique way to define and monitor the global metabolic homeostasis in humans. The development of this analytical approach is still limited by the scarcity of pluridisciplinary teams able to fully exploit the wealth of information present on the NMR spectrum of a fluid. While application in pharmacology and toxicology is already established, the main areas of current development are cancer, hereditary metabolic disorders, organ transplantation and neurological diseases.


Subject(s)
Body Fluids/metabolism , Magnetic Resonance Spectroscopy , Body Fluids/physiology , Cerebrospinal Fluid/metabolism , Glycosylation , Homeostasis/physiology , Humans
20.
J Cell Physiol ; 150(3): 534-44, 1992 Mar.
Article in English | MEDLINE | ID: mdl-1537883

ABSTRACT

Lack of estrogen receptor (ER) and presence of vimentin (VIM) associate with poor prognosis in human breast cancer. We have explored the relationships between ER, VIM, and invasiveness in human breast cancer cell lines. In the matrigel outgrowth assay, ER+/VIM- (MCF-7, T47D, ZR-75-1), and ER-/VIM- (MDA-MB-468, SK-Br-3) cell lines were uninvasive, while ER-/VIM+ (BT549, MDA-MB-231, MDA-MB-435, MDA-MB-436, Hs578T) lines formed invasive, penetrating colonies. Similarly, ER-/VIM+ cell lines were significantly more invasive than either the ER+/VIM- or ER-/VIM- cell lines in the Boyden chamber chemoinvasion assay. Invasive activity in nude mice was only seen with ER-/VIM+ cell lines MDA-MB-231, MDA-MB-435 and MDA-MB-436. Hs578T cells (ER-/VIM+) showed hematogenous dissemination to the lungs in one of five mice, but lacked local invasion. The ER-/VIM+ MCF-7ADR subline was significantly more active than the MCF-7 cells in vitro, but resembled the wild-type MCF-7 parent in in vivo activity. Data from these cell lines suggest that human breast cancer progression results first in the loss of ER, and subsequently in VIM acquisition, the latter being associated with increased metastatic potential through enhanced invasiveness. The MCF-7ADR data provide evidence that this transition can occur in human breast cancer cells. Vimentin expression may provide useful insights into mechanisms of invasion and/or breast cancer cell progression.


Subject(s)
Basement Membrane/ultrastructure , Breast Neoplasms/pathology , Receptors, Estrogen/metabolism , Vimentin/genetics , Animals , Basement Membrane/physiology , Blotting, Northern , Breast Neoplasms/metabolism , Cell Movement , Cells, Cultured , Chemotaxis , Collagen , Drug Combinations , Fluorescent Antibody Technique , Gene Expression , Humans , Laminin , Mice , Mice, Nude , Neoplasm Invasiveness , Proteoglycans , Tumor Cells, Cultured , Vimentin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...