Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters










Publication year range
1.
Neuroscience ; 157(4): 833-43, 2008 Dec 10.
Article in English | MEDLINE | ID: mdl-18951956

ABSTRACT

We report that somatostatin receptor subtype 1 (sst1) associates in vivo and in vitro with synapse-associated protein SAP) 97, a membrane-associated guanylate kinase homolog implicated as a scaffolding protein in the structural organization of specialized membrane complexes in various tissues, including the CNS. SAP97 and sst1 were coimmuno-precipitated from rodent brain and from transfected human embryonic kidney (HEK) 293 cells, and pull-down experiments demonstrated that the interaction is dependent on the class I PDZ binding motif in sst1 carboxyterminus. Calorimetric titration indicated that the postsynaptic density-95/discs large/zona occludens-1 (PDZ) 2 domain of SAP97 provides the main contribution to the interaction. We noticed substantial sst1 immunoreactivity in differentiating cortical neurons in culture which declined as the cultures matured. The sst1 immunoreactivity extended, together with SAP97 to neuronal growth cones. Somatostatin (1 microM) triggered retraction of the filopodia and lamellipodia in the growth cones. This growth cone collapse was enhanced by overexpression of green fluorescent protein-tagged sst1, whereas sst1 mutant lacking the PDZ binding motif had no effect. These findings suggest a role for somatostatin signaling in the regulation of growth cone stability, which may involve PDZ domain proteins interacting with sst1 and/or other somatostatin receptors. Consistent with a developmental role, sst1 immunoreactivity was present transiently in the developing mouse cortex, peaking at postnatal day 5 and declining thereafter to low levels in the adult cortex.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Growth Cones/physiology , Membrane Proteins/metabolism , Neurons/cytology , PDZ Domains/physiology , Receptors, Somatostatin/metabolism , Adaptor Proteins, Signal Transducing/genetics , Amino Acid Sequence , Animals , Animals, Newborn , Cells, Cultured , Chlorocebus aethiops , Discs Large Homolog 1 Protein , Embryo, Mammalian , Green Fluorescent Proteins/genetics , Humans , Immunoprecipitation/methods , Ligands , Membrane Proteins/genetics , Mice , Microtubule-Associated Proteins/metabolism , Mutation , Prosencephalon/cytology , Receptors, Somatostatin/genetics , Transfection
2.
Biochemistry ; 40(40): 12157-68, 2001 Oct 09.
Article in English | MEDLINE | ID: mdl-11580291

ABSTRACT

The high-affinity interaction between urokinase-type plasminogen activator (uPA) and its glycolipid-anchored receptor (uPAR) plays an important role in pericellular plasminogen activation. Since proteolytic degradation of the extracellular matrix has an established role in tumor invasion and metastasis, the uPA-uPAR interaction represents a potential target for therapeutic intervention. By affinity maturation using combinatorial chemistry we have now developed and characterized a 9-mer, linear peptide antagonist of the uPA-uPAR interaction demonstrating specific, high-affinity binding to human uPAR (K(d) approximately 0.4 nM). Studies by surface plasmon resonance reveal that the off-rate for this receptor-peptide complex is comparable to that measured for the natural protein ligand, uPA. The functional epitope on human uPAR for this antagonist has been delineated by site-directed mutagenesis, and its assignment to loop 3 of uPAR domain III (Met(246), His(249), His(251), and Phe(256)) corroborates data previously obtained by photoaffinity labeling and provides a molecular explanation for the extreme selectivity observed for the antagonist toward human compared to mouse, monkey, and hamster uPAR. When human HEp-3 cancer cells were inoculated in the presence of this peptide antagonist, a specific inhibition of cancer cell intravasation was observed in a chicken chorioallantoic membrane assay. These data imply that design of small organic molecules mimicking the binding determinants of this 9-mer peptide antagonist may have a potential application in combination therapy for certain types of cancer.


Subject(s)
Combinatorial Chemistry Techniques , Epitopes/chemistry , Neoplasms/pathology , Receptors, Cell Surface/antagonists & inhibitors , Animals , Base Sequence , Cell Line , Cricetinae , DNA Primers , Humans , Mice , Oligopeptides/chemistry , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/immunology , Receptors, Urokinase Plasminogen Activator , Recombinant Proteins/chemistry , Recombinant Proteins/immunology
3.
Cancer Res ; 61(20): 7544-51, 2001 Oct 15.
Article in English | MEDLINE | ID: mdl-11606392

ABSTRACT

An androgen-independent (AI) prostate cancer cell line, derived recently from an LNCaP cell line maintained in androgen-poor conditions, has properties resembling a subgroup of advanced prostate cancers in that it has an overexpressed androgen receptor (AR), undetectable levels of p21WAF1 and prostate-specific antigen, and is resistant to apoptosis. The loss of prostate-specific antigen expression but not the p21WAF1 is attributable to gene silencing by hypermethylation. The high AR and undetectable p21WAF1 of AI cells, and lower AR but highly expressed p21WAF1 of androgen-dependent parental LNCaP cells, suggest a possibility of a functional link between these two proteins. Therefore, we examined the impact the modulation of AR will have on the expression of p21WAF1. Treatment of androgen-dependent cells with an androgen agonist, R1881, increased the AR protein level, whereas it simultaneously reduced the endogenous p21WAF1-protein 8-fold and the activity of a transiently transfected p21-promoter-reporter 10-fold. The down-regulation of p21WAF1 promoter appeared to be ARE mediated, dependent on AR, and not cell-type specific. Furthermore, a reduction of the AR level in AI cells by AR-antisense oligonucleotide increased the p21WAF1 promoter-reporter activity by approximately 4-fold, confirming a functional link between these two proteins. A strong, direct induction of p21WAF1 expression achieved by treatment of AI cells with trichostatin A produced a partial reversion of the AI phenotype evidenced by increased sensitivity of these cells to paclitaxel-induced apoptosis. Moreover, a reduction of AR level by antisense treatment, which also increased p21WAF1 expression, partially restored the androgen dependence of AI cells for growth. The functional link between AR dosage and p21WAF1 expression suggests that therapeutic reduction of AR protein in advanced prostate cancers with elevated AR levels may re-establish their hormone dependence and improve therapeutic response to repeated hormonal ablation and/or induction of apoptosis.


Subject(s)
Apoptosis/physiology , Cyclins/genetics , Gene Silencing , Neoplasms, Hormone-Dependent/metabolism , Prostatic Neoplasms/metabolism , Receptors, Androgen/biosynthesis , Cell Nucleus/metabolism , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/biosynthesis , Cyclins/physiology , Gene Expression Regulation, Neoplastic , Humans , Male , Neoplasms, Hormone-Dependent/genetics , Neoplasms, Hormone-Dependent/pathology , Prostate-Specific Antigen/biosynthesis , Prostate-Specific Antigen/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Receptors, Androgen/genetics
4.
Mol Biol Cell ; 12(4): 863-79, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11294892

ABSTRACT

We discovered that a shift between the state of tumorigenicity and dormancy in human carcinoma (HEp3) is attained through regulation of the balance between two classical mitogen-activated protein kinase (MAPK)-signaling pathways, the mitogenic extracellular regulated kinase (ERK) and the apoptotic/growth suppressive stress-activated protein kinase 2 (p38(MAPK)), and that urokinase plasminogen activator receptor (uPAR) is an important regulator of these events. This is a novel function for uPAR whereby, when expressed at high level, it enters into frequent, activating interactions with the alpha5beta1-integrin, which facilitates the formation of insoluble fibronectin (FN) fibrils. Activation of alpha5beta1-integrin by uPAR generates persistently high level of active ERK necessary for tumor growth in vivo. Our results show that ERK activation is generated through a convergence of two pathways: a positive signal through uPAR-activated alpha5beta1, which activates ERK, and a signal generated by the presence of FN fibrils that suppresses p38 activity. When fibrils are removed or their assembly is blocked, p38 activity increases. Low uPAR derivatives of HEp3 cells, which are growth arrested (dormant) in vivo, have a high p38/ERK activity ratio, but in spite of a similar level of alpha5beta1-integrin, they do not assemble FN fibrils. However, when p38 activity is inhibited by pharmacological (SB203580) or genetic (dominant negative-p38) approaches, their ERK becomes activated, uPAR is overexpressed, alpha5beta1-integrins are activated, and dormancy is interrupted. Restoration of these properties in dormant cells can be mimicked by a direct re-expression of uPAR through transfection with a uPAR-coding plasmid. We conclude that overexpression of uPAR and its interaction with the integrin are responsible for generating two feedback loops; one increases the ERK activity that feeds back by increasing the expression of uPAR. The second loop, through the presence of FN fibrils, suppresses p38 activity, further increasing ERK activity. Together these results indicate that uPAR and its interaction with the integrin should be considered important targets for induction of tumor dormancy.


Subject(s)
Fibronectins/metabolism , Mitogen-Activated Protein Kinases/metabolism , Receptors, Cell Surface/physiology , Signal Transduction/physiology , Cell Division , Humans , Integrin beta1/metabolism , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Receptors, Fibronectin/metabolism , Receptors, Urokinase Plasminogen Activator , Tumor Cells, Cultured , p38 Mitogen-Activated Protein Kinases
5.
Curr Opin Cell Biol ; 12(5): 613-20, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10978898

ABSTRACT

Urokinase-type plasminogen activator receptor (uPAR) binds the urokinase-type plasminogen activator (uPA) and facilitates a proteolytic cascade focused at the cell surface. More recently, uPAR was recognized as a multifunctional protein that, through its interactions with integrins, initiates signaling events that alter cell adhesion, migration and proliferation. Results obtained recently have led to new insights into the structural aspects of uPAR interaction with integrins, provided a more detailed description of the signaling pathway they induce, and determined that uPAR signaling plays a role in cell migration and tumorigenicity.


Subject(s)
Integrins/metabolism , Receptors, Cell Surface/metabolism , Animals , Cell Adhesion/physiology , Cell Division/physiology , Cell Movement/physiology , Humans , Protein Binding/physiology , Receptors, Urokinase Plasminogen Activator , Signal Transduction
6.
J Cell Biol ; 147(1): 89-104, 1999 Oct 04.
Article in English | MEDLINE | ID: mdl-10508858

ABSTRACT

Mechanisms that regulate the transition of metastases from clinically undetectable and dormant to progressively growing are the least understood aspects of cancer biology. Here, we show that a large ( approximately 70%) reduction in the urokinase plasminogen activator receptor (uPAR) level in human carcinoma HEp3 cells, while not affecting their in vitro growth, induced a protracted state of tumor dormancy in vivo, with G(0)/G(1) arrest. We have now identified the mechanism responsible for the induction of dormancy. We found that uPA/uPAR proteins were physically associated with alpha5beta1, and that in cells with low uPAR the frequency of this association was significantly reduced, leading to a reduced avidity of alpha5beta1 and a lower adhesion of cells to the fibronectin (FN). Adhesion to FN resulted in a robust and persistent ERK1/2 activation and serum-independent growth stimulation of only uPAR-rich cells. Compared with uPAR-rich tumorigenic cells, the basal level of active extracellular regulated kinase (ERK) was four to sixfold reduced in uPAR-poor dormant cells and its stimulation by single chain uPA (scuPA) was weak and showed slow kinetics. The high basal level of active ERK in uPAR-rich cells could be strongly and rapidly stimulated by scuPA. Disruption of uPAR-alpha5beta1 complexes in uPAR-rich cells with antibodies or a peptide that disrupts uPAR-beta1 interactions, reduced the FN-dependent ERK1/2 activation. These results indicate that dormancy of low uPAR cells may be the consequence of insufficient uPA/uPAR/alpha5beta1 complexes, which cannot induce ERK1/2 activity above a threshold needed to sustain tumor growth in vivo. In support of this conclusion we found that treatment of uPAR-rich cells, which maintain high ERK activity in vivo, with reagents interfering with the uPAR/beta1 signal to ERK activation, mimic the in vivo dormancy induced by downregulation of uPAR.


Subject(s)
Down-Regulation , Integrins/metabolism , MAP Kinase Signaling System , Neoplasms/pathology , Protein Serine-Threonine Kinases , Receptors, Cell Surface/metabolism , Animals , Cell Adhesion , Cell Adhesion Molecules/metabolism , Cell Division , Chick Embryo , Enzyme Activation , Fibronectins/metabolism , Humans , Kinetics , MAP Kinase Kinase 1 , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Neoplasms/enzymology , Neoplasms/metabolism , Phosphorylation , Precipitin Tests , Protein Binding , Receptors, Cell Surface/genetics , Receptors, Fibronectin/metabolism , Receptors, Urokinase Plasminogen Activator , Tumor Cells, Cultured , Urokinase-Type Plasminogen Activator/chemistry , Urokinase-Type Plasminogen Activator/metabolism
7.
J Cell Physiol ; 179(3): 336-46, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10228952

ABSTRACT

Androgen ablation-induced prostate cancer regression is transient and ends with the regrowth of androgen-independent (AI) tumors. To mimic this evolution in culture, we chronically deprived an androgen-dependent (AD) prostate cancer cell line (LNCaP) of androgen, generating an AI derivative which retained limited hormone proliferative responsiveness and a barely detectable prostate-specific antigen (PSA) mRNA level. While the cytokeratin 8 (CK8) level was low, the androgen receptor (AR) protein in AI cells was on average tenfold greater than in AD cells. When challenged for susceptibility to undergo apoptosis, the AI cells were more resistant than AD cells to all-trans retinoic acid (tRA) and two chemotherapeutic agents, Taxol and Adriamycin, requiring higher doses and longer periods of treatment to achieve similar effects. Compared to AD cells, the partially apoptosis-resistant AI cells expressed four times more Bcl-2 protein and undetectable levels of p21/WAF1. Induction of apoptosis by tRA in both cell types did not affect their expression but was preceded by the activation of Rb and a pronounced reduction of AR protein level. The kinetics of the Rb activation and AR downmodulation in both cell types matched their tRA sensitivity, suggesting that these events may be required for tRA-induced apoptosis. The results show that the apoptotic pathway in AI cells, although more difficult to induce, is not irrevocably lost and that targeted reduction of the AR protein level with retinoids in combination with androgen ablation therapy may prolong remissions in advanced prostate cancer patients.


Subject(s)
Antigens, Surface , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Dihydrotestosterone/pharmacology , Receptors, Androgen/genetics , Retinoblastoma Protein/genetics , Tretinoin/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/genetics , Blotting, Western , Carboxypeptidases/analysis , Carboxypeptidases/genetics , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Division/drug effects , Cell Division/genetics , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Doxorubicin/pharmacology , Flow Cytometry , Gene Expression Regulation, Neoplastic/drug effects , Glutamate Carboxypeptidase II , Growth Inhibitors/pharmacology , Humans , Keratins/analysis , Keratins/genetics , Male , Paclitaxel/pharmacology , Prostate-Specific Antigen/analysis , Prostate-Specific Antigen/genetics , Prostatic Neoplasms , Receptors, Androgen/analysis , Tumor Cells, Cultured/chemistry , Tumor Cells, Cultured/cytology , Tumor Cells, Cultured/metabolism
8.
Medicina (B Aires) ; 59(5 Pt 2): 547-52, 1999.
Article in English | MEDLINE | ID: mdl-10684157

ABSTRACT

Urokinase plasminogen activator receptor (uPAR) has been identified some 15 years ago and the anticipation was that its presence on the cell surface will provide a focus for anchoring uPA and possibly protect the enzyme from native inhibitors. The studies of the last decade have shown that uPA localized to the surface of cells by uPAR is indeed an important factor in the process of cancer cell invasion and metastasis. We developed a chick embryo model in which we showed that uPAR is crucial in invasion of stroma and in intravasation (breaching of the blood vessels walls). More recently and unexpectedly, uPAR--a protein anchored in the outer leaf-let of the plasma membrane, has been shown to initiate signal transduction events and affect cell migration. We have shown that uPAR co-associates with fibronectin binding integrin, alpha 5 beta 1, activates them and that this interaction leads to a greatly increased level of active ERK. When the association between uPAR and integrin or integrin and fibronectin are interrupted either by reduction of surface uPAR expression, or by other means, human carcinoma cells enter a state of protracted dormancy. We show that very high levels of active ERK are required to keep cancer cells proliferating in vivo.


Subject(s)
Neoplasm Invasiveness , Neoplasms/physiopathology , Plasminogen Activators/physiology , Receptors, Cell Surface/physiology , Urokinase-Type Plasminogen Activator/physiology , Animals , Chick Embryo , Humans , Neoplasm Metastasis , Neoplasms/metabolism , Signal Transduction , Time Factors
9.
Cell ; 94(3): 353-62, 1998 Aug 07.
Article in English | MEDLINE | ID: mdl-9708737

ABSTRACT

Proteases are crucial for cancer metastasis, but due to lack of assays, their role in intravasation has not yet been tested. We have developed a human Alu sequence PCR-based assay to quantitate intravasated cells in an in vivo model. We demonstrated that metalloproteinases (MMPs), and most likely MMP-9, are required for intravasation by showing that marimastat, an inhibitor of MMPs, reduced intravasation by more than 90%, and that only tumor cell lines expressing MMP-9 intravasated. Cells with low surface urokinase plasminogen activator (uPA) and uPA receptor (uPAR) were also incapable of intravasation, despite the presence of high levels of MMP-9. We concluded that breaching of the vascular wall is a rate-limiting step for intravasation, and consequently for metastasis, and that cooperation between uPA/uPAR and MMP-9 is required to complete this step.


Subject(s)
Endopeptidases/physiology , Neoplasm Invasiveness/pathology , Neoplasm Metastasis/pathology , Polymerase Chain Reaction/methods , Allantois , Animals , Cell Membrane/enzymology , Cell Membrane/pathology , Cell Transformation, Neoplastic/pathology , Chick Embryo , Chorion , Female , Humans , Hydrolysis , Male , Neoplasm Invasiveness/genetics , Neoplasm Metastasis/genetics , Repetitive Sequences, Nucleic Acid/genetics , Sensitivity and Specificity , Tumor Cells, Cultured , Vascular Neoplasms/enzymology , Vascular Neoplasms/pathology
10.
Exp Cell Res ; 232(2): 420-9, 1997 May 01.
Article in English | MEDLINE | ID: mdl-9168821

ABSTRACT

Cell migration involves the integrins, their extracellular matrix ligands, and pericellular proteolytic enzyme systems. We have studied the role of plasminogen activator inhibitor-1 (PAI-1) in cell migration, using human amnion WISH cells and human epidermoid carcinoma HEp-2 cells in an assay measuring migration from microcarrier beads and a modified Boyden-chamber assay. Active, but not latent or reactive center-cleaved, PAI-1 inhibited migration. A PAI-1 mutant without ability to inhibit plasminogen activation was as active as wild-type PAI-1 as a migration inhibitor, showing that inhibition of plasminogen activation was not involved. PAI-1 specifically interfered with intergrin- and vitronectin-mediated migration: Migration onto vitronectin-coated but not onto fibronectin-coated surfaces was inhibited by PAI-1, a cyclic RGD peptide inhibited migration, and both cell lines expressed vitronectin-binding alpha v-integrins. In addition, active PAI-1, but not latent or reactive center-cleaved PAI-1, inhibited vitronectin binding to integrins in an in vitro binding assay, without affecting binding of fibronectin. Monoclonal antibodies against the urokinase receptor, another vitronectin binding protein, did not affect cell migration in the beads assay, while some inhibitory effect was observed in the Boyden-chamber assay. We conclude that PAI-1, independently of its role as a proteinase inhibitor, inhibits cell migration by competing for vitronectin binding to integrins, while the interference of PAI-1 with binding of vitronectin to the urokinase receptor may play a secondary role. These data define a novel function for the serpin PAI-1, enabling it to regulate cell migration over vitronectin-rich extracellular matrix in the body.


Subject(s)
Cell Movement/drug effects , Integrins/physiology , Plasminogen Activator Inhibitor 1/physiology , Vitronectin/physiology , Amnion/cytology , Carcinoma, Squamous Cell/pathology , Cell Line , Culture Media, Serum-Free , Enzyme Activation/drug effects , Extracellular Matrix/metabolism , Humans , Neoplasm Proteins/physiology , Oligopeptides/pharmacology , Peptides, Cyclic/pharmacology , Plasminogen/metabolism , Plasminogen Activator Inhibitor 1/chemistry , Plasminogen Activator Inhibitor 1/pharmacology , Protein Conformation , Receptors, Cell Surface/physiology , Receptors, Urokinase Plasminogen Activator , Tumor Cells, Cultured , Urokinase-Type Plasminogen Activator/physiology
11.
J Cell Biol ; 137(3): 767-77, 1997 May 05.
Article in English | MEDLINE | ID: mdl-9151680

ABSTRACT

Considerable evidence links urokinase plasminogen activator (uPA) bound to its surface receptor (uPAR) with enhanced invasiveness of cancer cells. By blocking uPAR expression in human epidermoid carcinoma cells (HEp3), we have now identified an additional and novel in vivo function for this receptor by showing that receptor-deficient cells enter a state of dormancy reminiscent of that observed in human cancer metastasis. Its main characteristic is survival without signs of progressive growth. Five clones transfected with a vector expressing uPAR antisense RNA under the beta-actin promoter were isolated and shown to have uPAR (at the mRNA and protein levels) reduced by 50 to 80%; four clones, transfected with vector alone and having uPAR levels similar to those of parental cells, served as controls. In confirmation of our previous results, reduced uPAR always coincided with a significantly reduced invasiveness. Each of the control clones produced rapidly growing, highly metastatic tumors within 2 wk of inoculation on chorioallantoic membranes (CAMs) of chick embryos. In contrast, each of the clones with low surface uPAR, whose proliferation rate in culture was indistinguishable from controls, remained dormant for up to 5 mo when inoculated on CAMs. Thus, the reduction in uPAR altered the phenotype of HEp3 tumor cells from tumorigenic to dormant. Although protracted, tumor dormancy was not permanent since in spite of maintaining low uPAR levels, each of the in vivo-passaged antisense clones eventually reemerged from dormancy to initiate progressive growth and to form metastases at a level of 20 to 90% of that of fully malignant control. This observation suggested that other factors, whose expression is dependent on cumulative and prolonged in vivo effects, can compensate for the lack of a full complement of surface uPAR required for the expression of malignant properties. These "reemerged," uPAR-deficient clones were easily distinguishable from the vector-transfected controls by the fact that after only 1 wk in culture, the invasion of CAM by all five clones and tumorigenicity of four of the five clones were reduced back to the values observed before in vivo maintenance. In contrast, dissociated and in vitro-grown cells of control tumors were fully invasive and produced large, metastatic tumors when reinoculated on CAMs. Quantitation of the percent of apoptotic and S-phase cells in vivo, in the control and uPAR-deficient, dormant clones, showed that the mechanism responsible for the dormancy was a diminished proliferation.


Subject(s)
Receptors, Cell Surface/physiology , Apoptosis , Cell Division , DNA, Neoplasm/biosynthesis , Down-Regulation , Humans , Necrosis , Neoplasm Invasiveness , Neoplasm Metastasis , RNA, Antisense , Receptors, Urokinase Plasminogen Activator , Tumor Cells, Cultured , Urokinase-Type Plasminogen Activator/metabolism
12.
Mol Endocrinol ; 11(13): 2004-15, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9415404

ABSTRACT

Comparison of mRNA ratios of a non-DNA-binding estrogen receptor (ER(alpha)) isoform, missing exon 3 (ER(alpha)delta3), to the full-length ER(alpha), in normal breast epithelium to that in primary breast cancers and breast cancer cell lines revealed a 30-fold reduction of this ratio in cancer cells (P < 0.0001). To test what functions may have been affected by the loss of ER(alpha)delta3, stable clones of MCF-7 cells expressing ectopic ER(alpha)delta3 protein, at the range of physiological ER(alpha), were generated. In vector-transfected controls the ER(alpha)delta3-mRNA and protein were less than 10% while in the ER(alpha)delta3-expressing clones, ER(alpha)delta3-mRNA and protein ranged from 36-76% of the total ER(alpha). Estrogen (E2) stimulated the expression of pS2-mRNA in pMV7 vector control cells, but the stimulation was reduced by up to 93% in ER(alpha)delta3-expressing clones. In addition, several properties associated with the transformed phenotype were also strongly affected when ER(alpha)delta3 protein was reexpressed. Compared with vector-transfected control cells, the saturation density of the ER(alpha)delta3-expressing clones was reduced by 50-68%, while their exponential growth rate was only slightly (14.5 +/- 5%) lower. The in vivo invasiveness of the ER(alpha)delta3-expressing cells was significantly reduced (P = 0.007) by up to 79%. E2 stimulated anchorage-independent growth of the pMV7 vector control cells, but reduced it to below baseline levels in ER(alpha)delta3 clones. The reduction of the pS2 response to E2 in the ER(alpha)delta3-expressing clones and the E2 block of anchorage-independent growth to below baseline were more pronounced than expected from the dominant negative function of ER(alpha)delta3. These observations suggest that E2 may activate an additional ER(alpha)delta3-dependent inhibitory pathway. The drastic reduction of ER(alpha)delta3 to ER(alpha) ratio in breast cancer, and the fact that when present in breast cancer cells this isoform leads to a suppression, rather than enhancement, of the transformed phenotype by E2 suggests that the regulation of ER(alpha)-mRNA splicing may need to be altered for the breast carcinogenesis to proceed.


Subject(s)
Breast Neoplasms/genetics , Cell Transformation, Neoplastic/genetics , Estrogens/physiology , Gene Deletion , Gene Expression Regulation, Neoplastic , Receptors, Estrogen/genetics , Adult , Aged , Aged, 80 and over , Breast/cytology , Breast/metabolism , Breast Neoplasms/pathology , Cell Count/drug effects , Cell Transformation, Neoplastic/drug effects , Estrogen Receptor alpha , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Isomerism , Middle Aged , Phenotype , Protein Binding/drug effects , RNA, Messenger/analysis , RNA, Messenger/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Transfection , Transgenes , Tumor Cells, Cultured
13.
Braz J Med Biol Res ; 29(9): 1099-103, 1996 Sep.
Article in English | MEDLINE | ID: mdl-9181051

ABSTRACT

This paper discusses the feasibility of extrapolating to human cancer results obtained concerning the role of proteases in experimental models of invasion and metastasis. It is customary to view metastasis as a series of individual steps, which must be completed in sequence for the process to be successful. The role of proteases in this process is usually studied by testing the blocking effects of various inhibitors on tumor cell invasion of extracellular matrices (ECM), or in assays of "experimental metastasis" (tail vein injections). The tests of invasion, which utilize established tumor cell lines, provide an account of the type of proteases that potentially may be produced by cancer cells, but, as indicated by recent evidence, they do not reflect the much more restricted repertoire of proteases detected by in situ techniques in human cancer cells. It is much more difficult to find in the progression of human cancer the counterpart to the widely used tail vein injection, which purports to test "late" stages of metastasis, since intravasation is most likely a slow, selective process and not a massive flux of a huge number of cells injected under substantial pressure resulting in non-physiological responses. As a rule, therefore, once it is established that an isolated cancer cell can produce a protease, or degrade and invade ECM, the question is whether this process actually occurs according to a model most reflective of the disease studied (such as orthotopic models) and whether this activity is actually present in cancer tissue sections.


Subject(s)
Disease Models, Animal , Endopeptidases/metabolism , Neoplasm Invasiveness/pathology , Neoplasm Metastasis/pathology , Neoplasms/enzymology , Neoplasms/pathology , Animals , Humans , Mice , Neoplasm Transplantation
16.
Carcinogenesis ; 16(9): 2259-62, 1995 Sep.
Article in English | MEDLINE | ID: mdl-7554087

ABSTRACT

nm23H1 has properties of a metastasis suppressor gene. Although its mechanism of action is unknown, nm23 has been implicated in transforming growth factor beta 1 (TGF beta 1) signal transduction. In an earlier study we decreased nm23 mRNA levels 2- to 8-fold by antisense phosphorothiolated oligonucleotides in two HT29 colon carcinoma sublines at different stages in tumor progression with different responses to TGF beta 1: the HD3 subline, which shows TGF beta 1-induced growth arrest and differentiation; and the more tumorigenic U9 subline, whose growth and invasion are stimulated by TGF beta 1. Only TGF beta 1-mediated responses in HD3 cells were inhibited by nm23 antisense oligos, suggesting that nm23 functions in only one TGF beta 1 signaling pathway. In the current report we have extended this study to cell motility. HD3 motility was increased by nm23 phosphorothiolated antisense oligos which decrease nm23 mRNA levels, while HD3 cell motility was conversely decreased by TGF beta 1 which increases nm23 mRNA levels. HD3 motility was not increased by basic FGF, TGF beta 1 or TGF alpha, while the 13-fold higher basal motility of U9 cells was stimulated 3-fold by basic FGF, 4-fold by TGF beta 1 and 5-fold by TGF alpha, but not by scatter factor. Differences in motility and response to motility factors could not be ascribed to differences in either basal levels of proteases or modulation of their levels by TGF beta 1. Both HD3 and U9 cells displayed equal levels of urokinase activity and mRNA, equal expression of the metalloproteinase inhibitor TIMP-1, and no detectable collagenases by zymography. No differential response to TGF beta 1 was seen in any of these assays. Thus limited cell motility and lack of response to motility factors in HD3 colon cancer cells could be correlated with expression of nm23 active in signal transduction.


Subject(s)
Cell Movement/drug effects , Colonic Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Growth Substances/pharmacology , Monomeric GTP-Binding Proteins , Nucleoside-Diphosphate Kinase , Transcription Factors/genetics , Base Sequence , Cell Movement/physiology , Colonic Neoplasms/genetics , Genes, Tumor Suppressor , Growth Substances/physiology , HT29 Cells , Humans , Molecular Sequence Data , NM23 Nucleoside Diphosphate Kinases , Oligonucleotides, Antisense/pharmacology , Signal Transduction/physiology
17.
EMBO J ; 13(17): 3983-91, 1994 Sep 01.
Article in English | MEDLINE | ID: mdl-8076594

ABSTRACT

Concomitant expression of urokinase type plasminogen activator (uPA) and its surface receptor (uPAR) has been shown to correlate strongly with a more invasive tumor cell phenotype. A highly malignant human epidermoid carcinoma cell line (HEp3) was transfected with a vector capable of expressing an antisense transcript complementary to 300 bases of the 5' end of uPAR, including the ATG codon. Six stably transfected antisense (AS-2, 3, 5, 9, 10, 12) and eight control clones were characterized. All clones produced high levels of uPA activity. Examination of collagenase production and doubling time showed that all of the clones tested produced similar activities. The antisense clones showed a 20-74% reduction in the uPAR sites; the uPAR mRNA level was also reduced. A test of the invasive ability of all clones in a modified chorioallantoic membrane (CAM) showed that invasiveness of the antisense-inhibited clones was directly proportional to the density of surface uPAR. The AS-2 clone, which expressed the lowest number of uPARs showed a significantly reduced level of invasion. The invasiveness of additional AS-inhibited clones was also reduced. Seven control and four AS-inhibited clones were tested for tumorigenicity on CAMs of chick embryos. Inoculation of control cells produced large tumors, while the As clones were non-tumorigenic. AS-2 did not produce tumors even if kept in vivo for up to 10 weeks.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Carcinoma, Squamous Cell/pathology , Plasminogen Activators/genetics , RNA, Antisense/pharmacology , Receptors, Cell Surface/genetics , Allantois , Animals , Carcinoma, Squamous Cell/genetics , Chick Embryo , Chorion , Clone Cells , Humans , Mice , Mice, Nude , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Transplantation , Protein Binding , Receptors, Urokinase Plasminogen Activator , Transfection , Urokinase-Type Plasminogen Activator/metabolism
18.
J Cell Biol ; 126(1): 259-70, 1994 Jul.
Article in English | MEDLINE | ID: mdl-7517943

ABSTRACT

A human epithelial cell line, WISH, and a mouse cell line, LB6-uPAR, transfected with the human urokinase receptor (uPAR), both expressed high affinity uPAR but undetectable levels of urokinase (uPA). In two independent assays, binding of exogenous pro-uPA produced an up to threefold enhancement of migration. The migration was time and concentration dependent and did not involve extracellular proteolysis. This biologic response suggested that uPAR can trigger an intracellular signal. Since this receptor is a glycosyl-phosphatidylinositol-linked protein, we postulated that it must do so by interacting with other proteins, among which, by analogy to other systems, would be a kinase. To test this hypothesis, we carried out a solid phase capture of uPAR from WISH cell lysates using either antibodies against uPAR or pro-uPA adsorbed to plastic wells, followed by in vitro phosphorylation of the immobilized proteins. SDS-PAGE and autoradiography revealed two phosphorylated protein bands of 47 and 55 kD. Both proteins were phosphorylated on serine residues. Partial sequence of the two proteins showed a 100% homology to cytokeratin 18 (CK18) and 8 (CK8), respectively. A similar pattern of phosphorylation was obtained with lysates from A459 cells, a lung carcinoma, but not HL60, LB6-uPAR or HEp3 cell lysates, suggesting that the identified multiprotein uPAR-complex may be specific for simple epithelia. Moreover, immunocapture with antibody to another glycosyl-phosphatidylinositol-linked protein, CD55, which is highly expressed in WISH cells, was ineffective. The kinase was tentatively identified as protein kinase C, because it was inhibited by an analogue of staurosporine more specific for PKC and not by a PKA or tyrosine kinase inhibitors. The kinase was tentatively identified as PKC epsilon because of its resistance to PMA down-modulation, independence of Ca2+ for activity, and reaction with a specific anti-PKC epsilon antibody in Western blots. Cell fractionation into cytosolic and particulate fractions revealed that all four proteins, the kinase, uPAR, CK18, and CK8, were present in the particulate fraction. In vivo, CK8, and to a lesser degree CK18, were found to be phosphorylated on serine residues. Occupation of uPAR elicited a time-dependent increase in the phosphorylation intensity of CK8, a cell shape change and a redistribution of the cytokeratin filaments. These results strongly suggest that uPAR serves not only as an anchor for uPA but participates in a signal transduction pathway resulting in a pronounced biological response.


Subject(s)
Cell Movement/physiology , Plasminogen Activators/metabolism , Receptors, Cell Surface/metabolism , Signal Transduction/physiology , Urokinase-Type Plasminogen Activator/metabolism , Amino Acid Sequence , Cell Line , Cell Size/physiology , Enzyme Precursors/metabolism , Epithelial Cells , Epithelium/enzymology , Humans , Keratins/metabolism , Macromolecular Substances , Molecular Sequence Data , Multiprotein Complexes , Phosphorylation , Protein-Tyrosine Kinases/metabolism , Receptors, Urokinase Plasminogen Activator , Recombinant Proteins/metabolism
19.
Cancer Res ; 52(24): 6754-60, 1992 Dec 15.
Article in English | MEDLINE | ID: mdl-1333882

ABSTRACT

We have screened six human squamous carcinoma cell lines for their ability to invade connective tissue by using the experimentally modified chorioallantoic membrane of a chick embryo as an in vivo model of invasion. In confirmation of our earlier studies, all the invasive cell lines expressed high levels of surface-bound urokinase type plasminogen activator (uPA). However, some cell lines expressing this activity were not invasive, suggesting that surface uPA, although necessary, was not sufficient. Since in addition to fibronectin, that can be degraded by uPA or plasmin, chorioallantoic membrane connective tissue contains collagen, we examined the profile of collagenases secreted by the various cell lines in search for an activity that would coincide with the invasive phenotype. We found, using gelatin substrate gels, that type IV gelatinase was produced by all six cell types tested, three cell types produced the M(r) 92,000 gelatinase, and three a lower-molecular-weight activity, which we identified by immunoprecipitation with specific antibodies, and by a direct assay of activity, as interstitial collagenase. Only the latter cells were found to be highly invasive. We showed previously that continuous culture in vitro of one of the carcinoma cell lines, HEp3, led to a gradual extinction of their malignant phenotype. To confirm the correlation between invasion and the production of interstitial collagenase, we examined these two functions in cells freshly isolated from a HEp3 tumor and intermittently during passage in vitro. We found that, although the surface uPA activity was slightly diminished in the in vitro grown cultures, it was still within the range of values found in highly malignant cells, suggesting that it is not the reason for the decrease in invasiveness. In contrast, the reduction in interstitial collagenase closely followed the loss of the invasive phenotype; after 30 in vitro passages the cells were almost completely devoid of interstitial collagenase and unable to invade. The decrease in collagenase activity was not the result of an increased tissue inhibitor of metalloproteinases production.


Subject(s)
Carcinoma, Squamous Cell/pathology , Collagenases/physiology , Connective Tissue/pathology , Neoplasm Invasiveness , Receptors, Cell Surface/physiology , Urokinase-Type Plasminogen Activator/physiology , Animals , Chick Embryo , Collagenases/analysis , Glycoproteins/physiology , Humans , Molecular Weight , Receptors, Urokinase Plasminogen Activator , Tissue Inhibitor of Metalloproteinases , Tumor Cells, Cultured
20.
J Cell Biol ; 115(4): 1107-12, 1991 Nov.
Article in English | MEDLINE | ID: mdl-1659573

ABSTRACT

Numerous studies have linked the production of increased levels of urokinase type plasminogen activator (uPA) with the malignant phenotype. It has also been shown that a specific cell surface receptor can bind uPA through a domain distinct and distant from the proteolytic domain. In an in vivo model of invasion, consisting of experimentally modified chorioallantoic membrane (CAM) of a chick embryo, only cells that concurrently expressed both uPA and a receptor for uPA, and in which the receptor was saturated with uPA, were efficient in invasion. To test whether uPA produced by one cell can, in a paracrine fashion, affect the invasive capacity of a receptor-expressing cell, we transfected LB6 mouse cells with human uPA (LB6[uPA]), or human uPA-receptor cDNA (LB6[uPAR]). LB6(uPA) cells released into the medium 1-2 Ploug units of human uPA per 10(6) cells in 24 h. The LB6(uPAR) cells expressed on their surface approximately 12,000 high affinity (Kd 1.7 x 10(-10) M uPA binding sites per cell. Unlabeled LB6(uPA) and 125-IUdR-labeled LB6(uPAR) cells were coinoculated onto experimentally wounded and resealed CAMs and their invasion was compared to that of homologous mixtures of labeled and unlabeled LB6(uPAR) or LB6(uPA) cells. Concurrent presence of both cell types in the CAMs resulted in a 1.8-fold increase of invasion of the uPA-receptor expressing cells. A four-fold stimulation of invasion was observed when cells were cocultured in vitro, prior to in vivo inoculation. Enhancement of invasion was prevented in both sets of experiments by treatment with specific antihuman uPA antibodies, indicating that uPA was the main mediator of the invasion-enhancing, paracrine effect on the receptor-expressing cells.


Subject(s)
Neoplasm Invasiveness , Plasminogen Activators/metabolism , Receptors, Cell Surface/metabolism , Urokinase-Type Plasminogen Activator/metabolism , Animals , Cell Communication , Chick Embryo , Disease Models, Animal , Humans , Kinetics , Mice , Receptors, Urokinase Plasminogen Activator , Transfection , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...