Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Oncogene ; 29(25): 3665-76, 2010 Jun 24.
Article in English | MEDLINE | ID: mdl-20453882

ABSTRACT

We hypothesized that specific activation of a single retinoic acid receptor-alpha (RARalpha), without direct and concurrent activation of RARbeta and gamma, will inhibit mammary tumor oncogenesis in murine models relevant to human cancer. A total of 50 uniparous mouse mammary tumor virus (MMTV)-neu and 50 nuliparous MMTV-wnt1 transgenic mice were treated with RARalpha agonist (retinobenzoic acid, Am580) that was added to the diet for 40 (neu) and 35 weeks (wnt1), respectively. Among the shared antitumor effects was the inhibition of epithelial hyperplasia, a significant increase (P<0.05) in tumor-free survival and a reduction in tumor incidence and in the growth of established tumors. In both models, the mechanisms responsible for these effects involved inhibition of proliferation and survival pathways, and induction of apoptosis. The treatment was more effective in the MMTV-wnt1 model in which Am580 also induced differentiation, in both in vivo and three-dimensional (3D) cultures. In these tumors Am580 inhibited the wnt pathway, measured by loss of nuclear beta-catenin, suggesting partial oncogene dependence of therapy. Am580 treatment increased RARbeta and lowered the level of RARgamma, an isotype whose expression we linked with tumor proliferation. The anticancer effect of RARalpha, together with the newly discovered pro-proliferative role of RARgamma, suggests that specific activation of RARalpha and inhibition of RARgamma might be effective in breast cancer therapy.


Subject(s)
Benzoates/pharmacology , Mammary Neoplasms, Experimental/pathology , Mammary Tumor Virus, Mouse/genetics , Mammary Tumor Virus, Mouse/physiology , Receptor, ErbB-2/genetics , Receptors, Retinoic Acid/agonists , Tetrahydronaphthalenes/pharmacology , Wnt1 Protein/genetics , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Benzoates/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Transformation, Viral/genetics , Female , Humans , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/virology , Mice , Mice, Transgenic , Oncogenes/genetics , Retinoic Acid Receptor alpha , Tetrahydronaphthalenes/therapeutic use
3.
Oncogene ; 18(33): 4718-25, 1999 Aug 19.
Article in English | MEDLINE | ID: mdl-10467419

ABSTRACT

Overproduction of urokinase-type plasminogen activator (uPA) and metalloproteases (MMPs) is strongly correlated with tumorigenicity and with invasive and metastatic phenotypes of human and experimental tumors. We demonstrated previously that overproduction of uPA in tumor cells is mediated by a phospholipase D (PLD)- and protein kinase C-dependent mechanism. The oncogenic stimulus of v-Src and v-Ras results in the activation of PLD, which is dependent upon the monomeric GTPase RalA. We have therefore investigated whether RalA plays a role in uPA and MMP overproduction that is observed in response to oncogenic signals. We report here that NIH3T3 cells transformed by both v-Src and v-Ras, constitutively overproduce uPA and that expression of a dominant negative RalA mutant (S28N) blocks overproduction of uPA in both the v-Src-and v-Ras-transformed cells. v-Src and v-Ras also induced an upregulation of the activity of MMP-2 and MMP-9 as detected by zymograms, however only the v-Src induction correlated with MMP protein levels detected by Western blot analysis. The dominant negative RalA mutant blocked increased MMP-2 and 9 overproduction induced by v-Src, but not the increased activity of MMP-2 and 9 induced by v-Ras. And, consistent with a role for the RalA/PLD pathway in mitogenesis and tumor development, the dominant negative RalA mutant completely blocked tumor formation by v-Src- and v-Ras-transformed NIH3T3 cells injected subcutaneously in syngeneic mice. The data presented here implicate RalA and PLD as signaling mediators for tumor formation and protease production by transformed cells.


Subject(s)
Cell Transformation, Neoplastic/genetics , GTP Phosphohydrolases/genetics , Genes, ras , Genes, src , Metalloendopeptidases/biosynthesis , Urokinase-Type Plasminogen Activator/biosynthesis , ral GTP-Binding Proteins , 3T3 Cells , Animals , Collagenases/biosynthesis , Gelatinases/biosynthesis , Gene Expression Regulation, Neoplastic , Matrix Metalloproteinase 2 , Matrix Metalloproteinase 9 , Mice , Mice, Inbred BALB C , Mutation , Up-Regulation
4.
Eur J Biochem ; 263(2): 295-304, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10406935

ABSTRACT

We review the evidence in support of the notion that, upon experimental oncogenic transformation or in spontaneous human cancers, mitogenesis and expression of urokinase (uPA) and its receptor (uPAR) are activated through common signaling complexes and pathways. It is well documented that uPA, uPAR or metalloproteinases (MMPs) are overexpressed in tumor cells of mesenchymal or epithelial origin and these molecules are required for tumor invasion and metastasis. Furthermore, oncogenic stimuli, which may render the transformed cells tumorigenic and metastatic in vivo, activate, in a constitutive fashion, the extracellular-regulated kinases (Erk 1 and 2) classical mitogenic pathway and others such as the NH(2)-Jun-kinase (Jnk). Cells from human tumors or oncogene-transformed cells overexpress uPA and uPAR, and also show a sustained activation of the above-mentioned signaling modules. In this paper we show that the classical mitogenic pathway involving Ras-Erk, PKC-Erk or Rac-JNK, among others, is activated by growth factors or endogenously by oncogenes, and constitutively activates uPA and uPAR expression. All the data obtained from human tumors or experimental systems, incorporated into a general model, indicate that oncogenic stimuli lead to the constitutive activation of mitogenesis and uPA and its receptor expression, through the activation of the same classical and nonclassical signaling complexes and pathways that regulate cell proliferation. We also discuss contrasting points of view. For instance, what governs the differential regulation of mitogenesis and the signal that leads to protease overexpression in a way that allows normal cells during physiological events to respond to growth factors, and proliferate without overexpressing extracellular matrix (ECM) proteases? Or how can cells remodel their microenvironment without proliferating? What restrains benign tumors from overexpressing tumor-associated proteases when they certainly have the mitogenic signal fully activated? This may occur by the differential regulation of transcriptional programs and recent reports reviewed in this paper may provide an insight into how this occurs at the signaling and transcriptional levels.


Subject(s)
Signal Transduction , Urokinase-Type Plasminogen Activator/biosynthesis , Urokinase-Type Plasminogen Activator/genetics , Cell Transformation, Neoplastic , Gene Expression Regulation, Neoplastic , Humans , Models, Biological , Tumor Suppressor Protein p53/metabolism
5.
Breast Cancer Res Treat ; 57(3): 271-5, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10617303

ABSTRACT

We have investigated the effects of desmopressin (DDAVP), a synthetic analog of the natural hormone vasopressin, on experimental lung colonization of mammary tumor cells using a syngeneic BALB/c mouse model. Coinjection of DDAVP (1-2 microg/kg body weight) at the time of i.v. inoculation of F3II carcinoma cells or LM3 adenocarcinoma cells significantly inhibited the formation of experimental lung metastases. In both cases, the number of pulmonary nodules was reduced about 70%. Inhibition of metastasis was also obtained with i.v. administration of DDAVP 24 h after tumor cell inoculation. Interestingly, the inhibition of lung metastasis was not due to direct cytotoxic effects of DDAVP on mammary tumor cells. The in vitro formation of multicellular aggregates in the presence of citrated plasma from control and DDAVP-treated mice was also examined. Control plasma rapidly induced a significant tumor cell aggregation. In contrast, in the presence of plasma from DDAVP-treated mice, tumor cells remained as a single cell suspension. DDAVP may help to dissolve the protective fibrin shield of circulating tumor cells. Our data suggest, for the first time, that adjuvant DDAVP therapy may impair successful implantation of circulating mammary tumor cells.


Subject(s)
Deamino Arginine Vasopressin/pharmacology , Hemostatics/pharmacology , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/pathology , Animals , Cell Aggregation/drug effects , Female , Mice , Mice, Inbred BALB C
6.
Int J Cancer ; 78(6): 727-34, 1998 Dec 09.
Article in English | MEDLINE | ID: mdl-9833766

ABSTRACT

The invasion and metastasis process involves degradation of the extracellular matrix mediated by tumor- and host-produced proteolytic enzymes. The main enzymes involved in this process are urokinase-type plasminogen activator (uPA) and the matrix metalloproteinases (MMPs). Calcium is a main co-factor in the signaling pathways that regulate cell proliferation and protease production. We have studied here the effect of verapamil, a calcium channel blocker widely used to treat hypertensive diseases, on local tumor growth, spontaneous and experimental metastasis development, tumor-associated protease production and circulating MMP activity in tumor-bearing mice. BALB/c mice treated for 45 days with verapamil showed no toxic effects. Oral administration of verapamil to mice injected with F311 tumor cells, either pre-treated or not with verapamil, showed a significant decrease of local tumor invasion and both spontaneous and experimental metastasis development (51.3% inhibition of metastasis in both cases, p < 0.01). uPA and MMP-9 production by tumor cells in vitro was significantly inhibited by verapamil in a dose-dependent manner, showing a long-term inhibition after removal of the drug. Verapamil also exhibited a marked cytostatic effect on F311 cell proliferation in vitro. In addition, circulating MMP activity, usually enhanced in tumor-bearing mice, diminished significantly with all verapamil treatments. Our results suggest that modulation of the calcium-dependent signaling pathways that regulate tumor- or host-dependent production of proteases and tumor cell proliferation could contribute to the inhibition of metastasis development. Finally, we describe the inhibitory effects of a commonly used hypotensor in humans, verapamil, on the invasive and metastatic capacity of mammary tumor cells.


Subject(s)
Calcium Channel Blockers/pharmacology , Endopeptidases/metabolism , Verapamil/pharmacology , Animals , Cell Division , Dose-Response Relationship, Drug , Lung Neoplasms/secondary , Male , Metalloendopeptidases/metabolism , Mice , Mice, Inbred BALB C , Neoplasm Invasiveness , Neoplasm Metastasis , Tumor Cells, Cultured , Urokinase-Type Plasminogen Activator/metabolism
7.
Int J Cancer ; 76(3): 362-7, 1998 May 04.
Article in English | MEDLINE | ID: mdl-9579573

ABSTRACT

Urokinase-type plasminogen activator (uPA) is a key serine protease involved in invasion and metastasis. We had shown that overproduction of uPA in tumor cells is controlled by a phospholipase D-protein kinase C-dependent pathway. Now we studied whether other signaling pathways participate in the regulation of constitutive uPA and metalloproteinase (MMP) overproduction in tumor cells. Staurosporine, a protein kinase inhibitor, stimulated uPA and MMP-9 secretion as measured by radial caseinolysis, zymography and Western blotting. Genistein, a specific tyrosine kinase inhibitor, reduced the constitutive and staurosporine-induced uPA and MMP-9 secretion. Interestingly, the phosphatase inhibitor vanadate stimulated uPA secretion. Verapamil, a calcium channel blocker, inhibited both endogenous and PMA-stimulated secretion of uPA but was unable to inhibit staurosporine-induced secretion. The alcohol n-butanol, a phospholipase D and protein kinase C inhibitor, besides inhibiting constitutive uPA secretion, blocked staurosporine-induced secretion. Our results suggest that constitutive and staurosporine-induced uPA and MMP-9 secretion by LM3 murine mammary tumor cells is controlled by an endogenous tyrosine kinase pathway and probably involves protein phosphatases. In addition, the staurosporine-induced signal regulating urokinase secretion is independent of extracellular calcium but dependent on phospholipase D.


Subject(s)
Enzyme Inhibitors/pharmacology , Mammary Neoplasms, Animal/enzymology , Metalloendopeptidases/drug effects , Protein-Tyrosine Kinases/metabolism , Staurosporine/pharmacology , Urokinase-Type Plasminogen Activator/drug effects , Animals , Calcium/metabolism , Female , Metalloendopeptidases/metabolism , Mice , Mice, Inbred BALB C , Plasminogen Activators/metabolism , Tumor Cells, Cultured/drug effects , Urokinase-Type Plasminogen Activator/metabolism
8.
Anticancer Res ; 18(6A): 4499-504, 1998.
Article in English | MEDLINE | ID: mdl-9891516

ABSTRACT

We have analyzed the anti-invasive properties of the selective synthetic urokinase inhibitor 4-iodo benzo[b]thiopene-2-carboxamidine (B428) in the mouse mammary carcinoma model F3II. At non-cytotoxic concentrations (10-20 microM), B428 blocked secreted and cell-associated tumor-derived urokinase activity as well as whole cell plasminogen-dependent casein degradation. Pretreatment of F3II monolayers with B428 enhanced membrane bound uPA, suggesting that the compound may modify urokinase receptor mobilization and urokinase-dependent cell signaling. B428 exerted a dose-dependent inhibition of Matrigel invasion by F3II cells and also reduced tumor cell adhesion and migration using the same doses. Our data indicate that uPA and its cell surface receptor are involved in attachment, migration, and invasion of mammary tumor cells, and that the three processes can be blocked by a synthetic urokinase inhibitor.


Subject(s)
Amidines/pharmacology , Antineoplastic Agents/pharmacology , Cell Adhesion/drug effects , Enzyme Inhibitors/pharmacology , Mammary Neoplasms, Experimental/physiopathology , Neoplasm Invasiveness/prevention & control , Thiophenes/pharmacology , Urokinase-Type Plasminogen Activator/antagonists & inhibitors , Animals , Cell Division/drug effects , Cell Movement/drug effects , Culture Media, Conditioned , Female , Fibronectins , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Tumor Cells, Cultured , Urokinase-Type Plasminogen Activator/biosynthesis
9.
Blood Coagul Fibrinolysis ; 8(7): 403-10, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9391721

ABSTRACT

The aim of this work was to clarify the role of urokinase-type plasminogen activator (uPA) on the profibrinolytic activity of heparin, chemically modified heparins [partially: N-desulfated (N-des), N-desulfated N-acetylated (N-des N-ac), O-desulfated (O-des), O/N-desulfated N-acetylated (O/N-des N-ac)] and heparan sulfate. Binding competition assays of plasminogen and uPA to heparin-sepharose demonstrated that heparin bound to both enzymes. Moreover, in the presence of increasing amounts of heparin, plasminogen activation mediated by uPA occurred as a bell-shaped curve, suggesting the formation of a ternary complex. In contrast, all chemically-modified heparins lacked this cofactor activity, although N-des and heparan sulfate partially retained the uPA binding capacity, and O-des partially bound to both plasminogen and uPA. Plasmatic euglobulins from mice treated with heparin, as well as with modified heparins with uPA binding capacity, presented a 2-fold enhancement of 47 kDa lytic band, as assessed by zymographic analysis. Western blotting analysis anti-uPA (47 kDa) showed that the enhanced uPA activity correlated with a true increase in uPA protein levels. These results suggest that the profibrinolytic activity of heparin mediated by uPA could be caused by an increase in uPA protein levels rather than by a cofactor activity mediated by a formation of ternary complexes.


Subject(s)
Fibrinolysis , Heparin/metabolism , Plasminogen/metabolism , Urokinase-Type Plasminogen Activator/metabolism , Animals , Heparin/chemistry , Heparitin Sulfate/metabolism , Male , Mice , Mice, Inbred BALB C
10.
Int J Cancer ; 71(5): 881-90, 1997 May 29.
Article in English | MEDLINE | ID: mdl-9180160

ABSTRACT

Spreading is a critical process involved in motility and growth of tumor cells during the metastatic cascade. Focal adhesion kinase, src-proteins and PKC have been reported to participate in the regulation of cytoskeleton organization in both normal and transformed cells during spreading. The role of other signaling enzymes such as PLD and PAP has not been studied during spreading in tumor cells. We now show that the spreading of murine mammary adenocarcinoma LM3 cells was significantly reduced by n-butanol, a PLD and PKC inhibitor, with a maximal inhibition of 54% (p < 0.001) in both the presence and absence of serum, as measured by phase-contrast microscopy. PMA only stimulated cell spreading over the control in the absence of serum and n-butanol inhibition was completely reversed by PMA treatment in both conditions. PA, the product of PLD activity, stimulated LM3 cell spreading and the same effect was observed with staurosporine. Spreading was enhanced when cells were seeded on collagen-IV- or fibronectin-coated surfaces and n-butanol could inhibit both integrin-derived signals. Cell spreading inhibition correlated with the absence of f-actin bundles and fewer beta1-integrin point contacts as determined by double immunofluorescence microscopy. In addition, n-butanol inhibited the proliferation of LM3 cells in the presence of serum (p < 0.01). These results suggest that beta1-integrin and f-actin/point contact assembly, involved in spreading and proliferation, require the participation of PLD-PKC regulatory pathways in LM3 cells.


Subject(s)
Actins/analysis , Adenocarcinoma/pathology , Integrin beta1/analysis , Mammary Neoplasms, Experimental/pathology , Phospholipase D/antagonists & inhibitors , Protein Kinase C/antagonists & inhibitors , 1-Butanol , Animals , Butanols/pharmacology , Cell Adhesion , Cell Division/drug effects , Enzyme Inhibitors/pharmacology , Genistein , Isoflavones/pharmacology , Mice , Microscopy, Fluorescence , Phosphatidic Acids/pharmacology , Signal Transduction , Staurosporine/pharmacology , Tetradecanoylphorbol Acetate/pharmacology , Tumor Cells, Cultured
11.
Biochim Biophys Acta ; 1356(2): 171-84, 1997 Apr 24.
Article in English | MEDLINE | ID: mdl-9150275

ABSTRACT

Urokinase-type plasminogen activator (uPA) initiates a proteolytic cascade with which invasive cells eliminate barriers to movement. The signaling pathways regulating uPA production in tumor cells remain unclear. We first studied the effects of n-butanol, a phospholipase D (PLD) and protein kinase C (PKC) inhibitor, on the production of uPA in murine mammary adenocarcinoma cells. Tumor cell monolayers treated during 24 h with 0.3% v/v n-butanol, secreted 45-50% less uPA to the culture medium than control monolayers (P < 0.001) as determined by radial caseinolysis, zymography and western blot. This inhibition occurred also with 5-h treatments and remained up to 5 h after the removal of the alcohol. Treatment with the phorbol ester PMA or with EGF, strongly increased uPA production (P < 0.001). Interestingly, a mild inhibition of uPA production was observed when PMA stimulation was assayed in cotreatments with n-butanol. In contrast EGF was unable to reverse the inhibition induced by n-butanol. H7 significantly inhibited uPA activity (P < 0.001) secreted to the culture media. Furthermore, phosphatidic acid significantly stimulated uPA production meanwhile propranolol, which blocks phosphatidic acid availability, reduced it, suggesting a main regulatory role for this intermediary metabolite. These results suggest for the first time that uPA production is regulated by PLD and PKC signal transduction pathways in murine mammary adenocarcinoma cells.


Subject(s)
Adenocarcinoma/metabolism , Mammary Neoplasms, Experimental/metabolism , Phospholipase D/metabolism , Protein Kinase C/metabolism , Urokinase-Type Plasminogen Activator/biosynthesis , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , 1-Butanol , Animals , Butanols/pharmacology , Epidermal Growth Factor/pharmacology , Female , Mice , Phosphatidic Acids/pharmacology , Phospholipase D/antagonists & inhibitors , Protein Kinase C/antagonists & inhibitors , Signal Transduction , Temperature , Tetradecanoylphorbol Acetate/pharmacology , Time Factors , Tumor Cells, Cultured/metabolism , Up-Regulation/drug effects
12.
J Surg Oncol ; 62(4): 288-97, 1996 Aug.
Article in English | MEDLINE | ID: mdl-8691844

ABSTRACT

We characterized a new mammary tumor cell line, F3II, previously established in vitro from a clonal subpopulation of the BALB/c transplantable mammary adenocarcinoma M3, moderately metastatic to lung. The F3II cell line has been passaged > 50 times. It has grown as elongated cells adherent to the bottom of the flask. Cytogenetic studies showed that F3II cultures were nearly triploid. Tumor cells expressed fibronectin and showed high levels of cell-surface urokinase, a key protease in invasion and metastasis. F3II cells grew as poorly differentiated, spindle-cell carcinoma tumors (sarcomatoid carcinomas) with a prominent local invasiveness, a high angiogenic response, and a 90-100% incidence of lung metastases when inoculated s.c. into syngeneic mice. Ultrastructural and immunocytochemical analysis revealed characteristic features of carcinomas. Our data suggest that F3II is less differentiated and more aggressive than the original tumor line, supporting the notion that mammary carcinomas are heterogeneous neoplasms and contain subpopulations with diverse biologic behavior. The F3II mouse mammary sarcomatoid carcinoma line is a suitable model to examine antiinvasive, antiangiogenic, and antimetastatic agents.


Subject(s)
Adenocarcinoma/pathology , Carcinosarcoma/pathology , Mammary Neoplasms, Experimental/pathology , Animals , Carcinosarcoma/chemistry , Cell Division , Female , Fibronectins/analysis , Immunohistochemistry , Male , Mammary Neoplasms, Experimental/chemistry , Mice , Mice, Inbred BALB C , Tumor Cells, Cultured , Urokinase-Type Plasminogen Activator/metabolism
13.
J Neurol Sci ; 139(1): 83-8, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8836977

ABSTRACT

We examined the activity of the serine protease urokinase-type plasminogen activator (uPA) present in the euglobulin fraction of plasma from 17 demented patients with probable Alzheimer's disease (AD), 12 patients with vascular dementia (VD) and 10 healthy controls. Euglobulin protein fractions were separated by electrophoresis and gels were incubated at the surface of plasminogen-rich casein-agarose underlays. The degradative activity of uPA in this system was measured by densitometric analysis. In 8/17 (47%) patients with AD we observed an excessive uPA activity (> 200 mIU/ml). In contrast, only 2/12 (16%) patients with VD and 1/10 (10%) control subjects revealed a comparable increase in circulating uPA activity. Further evaluation of dementia stage in patients with AD allow us to associate high levels of uPA activity with severity of disease. uPA levels were significantly elevated (2.8-fold increase) in AD patients with severe cognitive and memory impairments (Alzheimer Disease Assessment Scale) with respect to controls, VD patients or AD patients with moderate cognitive and memory impairments (P < 0.001, ANOVA). Our data suggest that the anormalities in circulating fibrinolytic enzymes could be correlated with the severity of dementia. In light of this findings, the free uPA activity in euglobulin plasma fraction should be considered a marker of serious damage in patients with AD.


Subject(s)
Alzheimer Disease/blood , Serum Globulins/analysis , Aged , Aged, 80 and over , Alzheimer Disease/enzymology , Analysis of Variance , Female , Humans , Male , Middle Aged , Reference Values , Urokinase-Type Plasminogen Activator/blood
14.
Breast Cancer Res Treat ; 40(3): 209-23, 1996.
Article in English | MEDLINE | ID: mdl-8883963

ABSTRACT

Urokinase-type plasminogen activator (uPA) initiates an extracellular proteolytic cascade with which invasive cells eliminate barriers to movement. We have evaluated the antiinvasive and antimetastatic properties of two recently developed synthetic uPA inhibitors, B428 and B623, in a BALB/c mouse mammary carcinoma model. We used the F3II and M3 tumor cell lines, previously described by our laboratory. In vitro, noncytotoxic concentrations of B428 or B623 inhibited secreted and cell-associated uPA activity produced by tumor cells and blocked uPA-mediated whole tumor cell degradation of fibronectin, allowing deposition of extracellular fibronectin fibrils. In vivo, administration of compounds was not associated with overt toxic effects. Daily i.p. treatment with B428 (20 mg/kg/day) or B623 (7.5 mg/kg/day) for 2 weeks, beginning after tumor take, markedly blocked the invasion of the muscle and adipose layers of the subcutis and dermis in mice bearing highly invasive F3II tumors. However, these compounds neither inhibited tumor-induced angiogenesis nor reduced the incidence of spontaneous lung metastasis. Moreover, B623 enhanced the formation of experimental lung metastasis. Our results suggest that synthetic uPA inhibitors act as potent antiinvasiveness agents in vivo but may be unable to control progression of the metastatic disease in the present mammary tumor model.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Lung Neoplasms/drug therapy , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/pathology , Urokinase-Type Plasminogen Activator/antagonists & inhibitors , Amidines/pharmacology , Amiloride/pharmacology , Animals , Cell Division/drug effects , Diuretics/pharmacology , Female , Fibrinolysis/drug effects , Fibronectins/metabolism , Mammary Neoplasms, Experimental/blood supply , Mice , Mice, Inbred BALB C , Neoplasm Invasiveness , Neoplasm Metastasis , Neovascularization, Pathologic/drug therapy , Thiophenes/pharmacology , Triamterene/pharmacology , Tumor Cells, Cultured
15.
Cancer Lett ; 70(3): 181-7, 1993 Jul 16.
Article in English | MEDLINE | ID: mdl-8353814

ABSTRACT

The fibrinolytic activity present in the euglobulin (EU) fraction of BALB/c mice before and during the growth of M3 and MM3 murine mammary adenocarcinomas was characterized. The main plasminogen activator (PA) form contained in EUs from control mice was defined as murine urokinase-type PA (uPA). Overall fibrinolytic activity decreased significantly during tumor development. Zymographies showed that this fall was associated with a reduction in the free uPA band (47 kDa) and to the detection of a tissue-type PA (tPA) complexed band (117 kDa). Western blotting showed free tPA protein (68 kDa) in control mice, that disappeared in M3 tumor-bearing mice. In this model, high subcutaneous tumor burden induces a severe impairment in the circulating fibrinolytic system.


Subject(s)
Adenocarcinoma/blood , Fibrinolysis/physiology , Mammary Neoplasms, Experimental/blood , Plasminogen Activators/blood , Adenocarcinoma/enzymology , Adenocarcinoma/pathology , Adenocarcinoma/secondary , Animals , Blotting, Western , Electrophoresis, Polyacrylamide Gel , Female , Gene Expression Regulation, Neoplastic , Lung Neoplasms/enzymology , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/enzymology , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Plasminogen Inactivators/blood , Tissue Plasminogen Activator/blood , Urokinase-Type Plasminogen Activator/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...