Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
Technol Cancer Res Treat ; 6(6): 641-50, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17994795

ABSTRACT

The growth of solid tumors dependent on the process of angiogenesis in which growth factors secreted by tumor and stromal cells promote endothelial cell proliferation, migration, and maturation. This process generates a tumor-specific vascular supply and enables small or dormant tumors to grow rapidly with exponential increases in tumor volume. Determination of tumor oxygenation at the microvascular level will provide important insight into tumor growth, angiogenesis, necrosis, and therapeutic response, and will facilitate to develop protocols for studying tumor behavior. A non-invasive multi-modality approach based on near infrared spectroscopy (NIRS) technique, namely: Steady State Diffuse Optical Spectroscopy (SSDOS) along with Magnetic Resonance Imaging (MRI) is applied for monitoring the concentration of oxyhemoglobin, deoxyhemoglobin and water within tumor region and for studying the vascular status of tumor and the patho-physiological changes that occur during angiogenesis. Since, the growth of solid tumors depends on the formation of new blood vessels, an association between intramural microvessel density (MVD) and tumor oxygenation is also investigated. The relative decrease in oxygenation value with tumor growth indicates that though blood vessels infiltrate and proliferate the tumor region, a hypoxic trend is clearly present.


Subject(s)
Brain Neoplasms/blood supply , Glioblastoma/blood supply , Magnetic Resonance Imaging , Neovascularization, Pathologic/diagnosis , Spectroscopy, Near-Infrared/methods , Algorithms , Animals , Brain Neoplasms/metabolism , Cell Line, Tumor , Female , Glioblastoma/metabolism , Humans , Mice , Mice, Nude , Models, Theoretical , Optics and Photonics , Oxyhemoglobins/analysis , Water/analysis
2.
Biotechniques ; 34(6): 1184-8, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12813886

ABSTRACT

In vivo bioluminescence imaging is becoming increasingly popular. Quantification of bioluminescence signals requires knowledge of the variability and reproducibility of this technique. The objective of this study was to analyze the time course of luminescent signal emitted from firefly luciferase-expressing tumors in two locations, following luciferin injection and at different times after tumor cell implantation. Knowledge of the kinetics of the bioluminescent signals is required for the reliable quantification and comparison of signal during longitudinal studies. The kinetics of bioluminescence was evaluated in orthotopic and heterotopic brain tumors in mice using a human brain tumor cell line constitutively expressing luciferase. Tumor cells were implanted in the brains and flanks of the animals, and whole-body images revealing tumor location were obtained. Tumor burden was monitored over time by the quantitation of photon emission. The magnitude of bioluminescence measured in vivo varied with time after the injection of luciferin, as well as with dose, which necessitated that the comparison of the quantitative results take into consideration the time after injection. Heterotopic and orthotopic tumors exhibited significantly different time courses; however, time after implantation as characterized by kinetic studies performed on days 4 and 14 after cell implantation revealed no significant differences in orthotopic tumors. Future quantitative longitudinal studies must take into account the differences in the kinetics of different models.


Subject(s)
Brain Neoplasms/enzymology , Glioblastoma/enzymology , Animals , Biotechnology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Kinetics , Luciferases/genetics , Luciferases/metabolism , Luminescent Measurements , Mice , Mice, Nude , Neoplasm Transplantation , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transplantation, Heterologous , Transplantation, Heterotopic
3.
J Surg Res ; 107(1): 1-6, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12384057

ABSTRACT

The mechanisms by which surgery increases metastatic proliferation remain poorly characterized, although endotoxin and immunocytes play a role. Recent evidence suggests that endothelial adherence of tumor cells may be important in the formation of metastases. Soluble receptors of interleukin-6 (sIL-6R) shed by activated neutrophils exert IL-6 effects on endothelial cells, which are unresponsive under normal circumstances. This study examined the hypothesis that sIL-6R released by surgical stress increases tumor cell adherence to the endothelium. Neutrophils (PMN) were stimulated with lipopolysaccharide, C-reactive protein (CRP), and tumor necrosis factor-alpha. Soluble IL-6R release was measured by enzyme-linked immunosorbent assay. Colonic tumor cells transfected with green fluorescent protein and endothelial cells were exposed to sIL-6R, and tumor cell adherence and transmigration were measured by fluorescence microscopy. Basal release of sIL-6R from PMN was 44.7 +/- 8.2 pg/ml at 60 min. This was significantly increased by endotoxin and CRP (131 +/- 16.8 and 84.1 +/- 5.3, respectively; both P < 0.05). However, tumor necrosis factor-alpha did not significantly alter sIL-6R release. Endothelial and tumor cell exposure to sIL-6R increased tumor cell adherence by 71.3% within 2 h but did not significantly increase transmigration, even at 6 h. Mediators of surgical stress induce neutrophil release of a soluble receptor for IL-6 that enhances colon cancer cell endothelial adherence. Since adherence to the endothelium is now considered to be a key event in metastatic genesis, these findings have important implications for colon cancer treatment strategies.


Subject(s)
Colonic Neoplasms/physiopathology , Endothelium, Vascular/physiopathology , Receptors, Interleukin-6/metabolism , Cell Adhesion/physiology , Cell Movement , Cells, Cultured , Colonic Neoplasms/secondary , Humans , Intercellular Adhesion Molecule-1/metabolism , Neoplasm Metastasis/physiopathology , Solubility
4.
Cancer Res ; 61(14): 5587-94, 2001 Jul 15.
Article in English | MEDLINE | ID: mdl-11454712

ABSTRACT

Increased expression of plasminogen activator inhibitor-1 (PAI-1) in cancer patients is associated with unfavorable outcome, and the reason for this paradox has been poorly understood. We have previously reported elevated levels of PAI-1 in primary tumors of advanced neuroblastomas (Y. Sugiura et al., Cancer Res., 59: 1327-1336, 1999). Here we demonstrate that PAI-1 is coexpressed with the angiogenesis marker alpha(v)beta3 integrin in blood vessels of primary neuroblastoma tumors, suggesting that PAI-1 plays a role in angiogenesis. Using human brain microvascular endothelial cells (HBMECs), we found that PAI-1 inhibits alpha(v)beta3 integrin-mediated cell adhesion to vitronectin but promotes alpha5beta1-mediated migration from vitronectin toward fibronectin. Inhibition of vitronectin adhesion by PAI-1 did not induce HBMEC apoptosis. PAI-1 also inhibited endothelial tube formation on Matrigel in the presence of vitronectin but had a stimulatory effect in the presence of fibronectin. This effect of PAI-1 on microvascular endothelial cells is primarily related to the ability of PAI-1 to bind to vitronectin via its NH2-terminal domain and to interfere with cell adhesion to vitronectin. We propose that PAI-1 acts as a positive switch for angiogenesis by promoting endothelial cell migration away from their vitronectin-containing perivascular space toward fibronectin-rich tumor tissue. These observations provide a novel explanation for the enhancing effect of PAI-1 in cancer progression.


Subject(s)
Cell Movement/drug effects , Endothelium, Vascular/drug effects , Fibronectins/pharmacology , Neovascularization, Pathologic/pathology , Plasminogen Activator Inhibitor 1/pharmacology , Apoptosis/drug effects , Cell Adhesion/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Endothelium, Vascular/chemistry , Endothelium, Vascular/cytology , Humans , Immunohistochemistry , Integrins/physiology , Neovascularization, Pathologic/metabolism , Neovascularization, Physiologic/drug effects , Plasminogen Activator Inhibitor 1/biosynthesis , Plasminogen Activator Inhibitor 1/physiology , Recombinant Proteins/pharmacology , Vitronectin/pharmacology
5.
J Surg Res ; 97(1): 14-9, 2001 May 01.
Article in English | MEDLINE | ID: mdl-11319874

ABSTRACT

BACKGROUND: Recent studies have demonstrated that metastatic disease develops from tumor cells that adhere to endothelial cells and proliferate intravascularly. The beta-1 integrin family and its ligand laminin have been shown to be important in tumor-to-endothelial cell adhesion. Lipopolysaccharide (LPS) has been implicated in the increased metastatic tumor growth that is seen postoperatively. We postulated that LPS increases tumor cell expression of beta-1 integrins and that this leads to increased adhesion. METHODS: The human metastatic colon cancer cell line LS174T was labeled with an enhanced green fluorescent protein (eGFP) using retroviral transfection. Cell cultures were treated with LPS for 1, 2, and 4 h (n = 6 each) and were subsequently cocultured for 30 or 120 min with confluent human umbilical vein endothelial cells (HUVECs), to allow adherence. Adherent tumor cells were counted using fluorescence microscopy. These experiments were carried out in the presence or absence of a functional blocking beta-1 integrin monoclonal antibody (4B4). Expression of beta-1 integrin and laminin on tumor and HUVECs was assessed using flow cytometric analysis. Tumor cell NF-kappaB activation after incubation with LPS was measured. RESULTS: Tumor cell and HUVEC beta-1 integrin expression and HUVEC expression of laminin were significantly (P < 0.05) enhanced after incubation with LPS. Tumor cell adhesion to HUVECs was significantly increased. Addition of the beta-1 integrin blocking antibody reduced tumor cell adhesion to control levels. LPS increased tumor cell NF-kappaB activation. CONCLUSIONS: Exposure to LPS increases tumor cell adhesion to the endothelium through a beta-1 integrin-mediated pathway that is NF-kappaB dependent. This may provide a target for immunotherapy directed at reducing postoperative metastatic tumor growth.


Subject(s)
Cell Adhesion/drug effects , Endothelium, Vascular/metabolism , Integrin beta1/biosynthesis , Lipopolysaccharides/pharmacology , Neoplasms/metabolism , Cells, Cultured , Endothelium, Vascular/drug effects , Humans , Kinetics , Laminin/biosynthesis , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Neoplasm Metastasis , Neoplasms/pathology , Peptides/pharmacology , Tumor Cells, Cultured , Up-Regulation
6.
Neurosurgery ; 48(1): 151-7, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11152340

ABSTRACT

OBJECTIVE: Brain tumors are highly angiogenic, and their growth and spread depend on the generation of new blood vessels. We examined the effect of the cyclic peptide antagonist pentapeptide EMD 121974, an antiangiogenic agent, on orthotopic and heterotopic brain tumor growth. METHODS: The human brain tumor cell lines DAOY (medulloblastoma) and U87 MG (glioblastoma) were injected into either the forebrain (orthotopic) or the subcutis (heterotopic) of nude mice, and daily systemic treatment with the active peptide was initiated after tumors were established. RESULTS: All control animals with orthotopic brain tumors and that received the inactive peptide EMD 135981 daily died as a result of tumor progression within 4 to 6 weeks; tumors measured 3 to 5 mm in diameter. In contrast, mice with orthotopic tumors that were treated daily with the active peptide survived for more than 16 weeks, and histological examination of the brains after 4, 8, and 12 weeks showed either no tumors or microscopic residual tumors. The growth of these brain tumor cells injected simultaneously or separately into the subcutis of nude mice (heterotopic model) was not affected by the active peptide, suggesting that the brain environment is a critical determinant of brain tumor susceptibility to growth inhibition by this pentapeptide. CONCLUSION: The cyclic pentapeptide EMD 121974 may become a treatment option specific to brain tumors. Because of its antiangiogenic effect, its use may be especially indicated after tumors are removed surgically.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Brain Neoplasms/drug therapy , Peptides, Cyclic/therapeutic use , Allantois/blood supply , Animals , Blood Vessels/drug effects , Brain Neoplasms/pathology , Chick Embryo , Chorion/blood supply , Drug Resistance , Humans , Integrins/antagonists & inhibitors , Mice , Mice, Nude , Neoplasm Transplantation , Skin Neoplasms/drug therapy , Snake Venoms , Transplantation, Heterologous
7.
Cancer Res ; 59(6): 1327-36, 1999 Mar 15.
Article in English | MEDLINE | ID: mdl-10096567

ABSTRACT

Proteases of the plasminogen-plasminogen activator (PA) system play an important role in cancer metastasis. We have examined the expression of these proteases and their cell surface receptors and inhibitors in neuroblastoma, a tumor that originates in cells of the neural crest and is the second most common solid tumor in children. This analysis was performed in seven established human cell lines and 20 primary tumor specimens. Urokinase PA and, in particular, tissue-type PA were expressed in cell lines and in tumor tissues; however, their levels of expression did not correlate with clinical stage. There was little evidence suggesting that neuroblastoma cells concentrate PA activity at their cell surface because urokinase-type PA receptor mRNA was detected in two cell lines and in 5 of 20 tumor samples by reverse transcription-PCR only. PA inhibitor (PAI)-2 was absent in all cell lines and tumor tissue samples examined. However, PAI-1, which was not expressed by the cell lines, was expressed by stromal cells and, specifically, endothelial cells in tumor tissue. By extending the analysis of PAI-1 expression in 64 primary tumor specimens, we found that high PAI-1 expression paradoxically correlated with metastatic stage and tumor recurrence. In vitro experiments indicated that the expression of PAI-1 by human microvascular endothelial cells was stimulated in the presence of SK-N-BE(2) human neuroblastoma cells and neuroblastoma culture medium. Recombinant PAI-1 also promoted SK-N-BE(2) cell detachment from vitronectin and migration from vitronectin toward fibronectin. From these data, we conclude that the up-regulation of PAI-1 expression in endothelial cells may promote rather than inhibit metastasis in neuroblastoma.


Subject(s)
Neoplasm Metastasis , Neuroblastoma/secondary , Plasminogen Activator Inhibitor 1/physiology , Plasminogen Activators/physiology , Plasminogen/physiology , Cell Adhesion/drug effects , Cell Movement/drug effects , Endothelium/metabolism , Fibronectins/physiology , Humans , Neoplasm Recurrence, Local , Neuroblastoma/metabolism , Neuroblastoma/pathology , Plasminogen/biosynthesis , Plasminogen Activator Inhibitor 1/biosynthesis , Plasminogen Activators/biosynthesis , RNA, Messenger/biosynthesis , Stromal Cells/metabolism , Tumor Cells, Cultured , Vitronectin/physiology
8.
Neurosurgery ; 43(6): 1437-42; discussion 1442-3, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9848858

ABSTRACT

OBJECTIVE: To determine whether fluorescence from human brain tumor cells transfected with the enhanced green fluorescent protein (EGFP) gene in vitro and xenotransplanted into the brain of nude mice would permit the detection of brain tumor invasion and metastasis in vivo. METHODS: Daoy medulloblastoma cells were transfected with a long terminal repeat-based retroviral vector containing the EGFP gene. Stable EGFP-expressing clones were isolated and stereotactically injected into the frontal cortex of nude mice. Four weeks later, whole brain sections were examined using fluorescence microscopy, immunohistochemistry, and routine hematoxylin and eosin staining for the visualization and detection of tumor cell invasion and metastasis. RESULTS: We demonstrate that EGFP-transduced Daoy cells maintain stable high-level EGFP expression in the central nervous system during their growth in vivo. EGFP fluorescence clearly demarcated the primary tumor margins and readily allowed for the visualization of distant micrometastases and local invasion on the single-cell level. Small metastatic and locally invasive foci, including those immediately adjacent to the tumor's leading invasive edge, were virtually undetectable by routine hematoxylin and eosin staining and immunohistochemistry. EGFP expression also persisted in vitro after cell reculture from brain tissue extracts. CONCLUSION: We show, for the first time, that EGFP-transduced human brain tumor cells can be visualized by fluorescence microscopy after intracerebral implantation. This method is superior to routine hematoxylin and eosin staining and immunohistochemistry for the detection and study of physiologically relevant patterns of brain tumor invasion and metastasis in vivo.


Subject(s)
Brain Neoplasms/pathology , Genes, Reporter , Luminescent Proteins/analysis , Medulloblastoma/pathology , Neoplasm Invasiveness/diagnosis , Neoplasm Metastasis/diagnosis , Recombinant Fusion Proteins/analysis , Animals , Brain Neoplasms/chemistry , DNA, Complementary/genetics , Eosine Yellowish-(YS) , Female , Gene Expression Regulation, Neoplastic , Green Fluorescent Proteins , Hematoxylin , Humans , Immunoenzyme Techniques , Luminescent Proteins/biosynthesis , Luminescent Proteins/genetics , Medulloblastoma/chemistry , Mice , Mice, Nude , Microscopy, Fluorescence , Moloney murine leukemia virus/genetics , Neoplasm Proteins/analysis , Neoplasm Transplantation , Receptors, Cell Surface/analysis , Receptors, Urokinase Plasminogen Activator , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Staining and Labeling/methods , Stereotaxic Techniques , Terminal Repeat Sequences , Transfection , Transplantation, Heterologous , Tumor Cells, Cultured/transplantation
9.
Cancer Res ; 58(10): 2209-16, 1998 May 15.
Article in English | MEDLINE | ID: mdl-9605768

ABSTRACT

Neuroblastoma, the second most common solid childhood tumor, can be a highly invasive and metastatic form of cancer. To assess the role of matrix-degrading proteases in this cancer, we have examined the expression of matrix metalloproteinases (MMPs) and their corresponding tissue inhibitors of metalloproteinases (TIMPs) in 7 human neuroblastoma cell lines and 24 primary untreated tumors. MMP-2 (gelatinase A) and MMP-9 (gelatinase B) were the only two MMPs expressed. MMP-2 was detected predominantly in an inactive proform in all tumor cell lines and tumor tissue extracts. The lack of MMP-2 activation in cell lines was attributed to the absence of expression of a membrane-type MMP (MT1-MMP), which activates proMMP-2, and to the abundant expression of TIMPs, particularly TIMP-2. Immunohistochemical analysis of tumor tissue samples indicated that MMP-2 was present in both tumor cells and stromal cells. In contrast, MMP-9 was not expressed by neuroblastoma cell lines but was present in inactive and active forms in extracts from tumor tissues. Immunohistochemical analysis of positive specimens indicated that MMP-9 was predominantly present in stromal, vascular, and perivascular cells surrounding nests of tumor cells. There was no correlation between the levels of these MMPs and the MYCN copy number or the histopathological phenotype. However, there were higher levels of MMP-2 and MMP-9 in stage IV (metastatic) disease when compared with stages I and II (noninvasive and nonmetastatic) or IV-S (P < 0.05).


Subject(s)
Collagenases/metabolism , Gelatinases/metabolism , Metalloendopeptidases/metabolism , Neoplasm Proteins/metabolism , Neuroblastoma/enzymology , Stromal Cells/enzymology , Tissue Inhibitor of Metalloproteinases/metabolism , Enzyme Activation , Humans , Matrix Metalloproteinase 2 , Matrix Metalloproteinase 9 , Neuroblastoma/secondary , Tumor Cells, Cultured
10.
J Neurooncol ; 40(3): 215-26, 1998 Dec.
Article in English | MEDLINE | ID: mdl-10066093

ABSTRACT

The plasminogen activation (PA) system plays an important role in tumor invasion by initiating pericellular proteolysis of the extracellular matrix (ECM) and inducing cell migration. Malignant brain tumors overexpress PA members and characteristically invade by migrating on ECM-producing white matter tracts and blood vessel walls. To determine whether urokinase-type plasminogen activator (uPA) and its receptor (uPAR) directly modulate the migration of brain tumor cells, we examined six human brain tumor cell lines, 2 astrocytomas (SW1088, SW1783), 2 medullobastomas (Daoy, D341Med), and 2 glioblastomas (U87MG, U118MG), for their surface uPAR expression, endogenous PA activity, and functional proteolytic activity by an ECM-degradation assay. Migration on Transwell membranes and invasion of Matrigel was then tested by pre-incubating the cells with increasing concentrations of either uPA, the proteolytically inactive amino-terminal fragment (ATF) of uPA, or the uPAR cleaving enzyme, phosphatidylinositol-specific phospholipase C (PI-PLC). All of the cell lines, except D341Med, express surface uPAR protein and uPA activity. High levels of uPAR and uPA activity correlated with cellular degradation of ECM, cell migration, and Matrigel invasion. Cell migration and invasion were enhanced by uPA or ATF in a dose dependent manner, while PI-PLC treatment abolished the uPA effect and inhibited migration and invasion. We conclude that ligation of uPAR by uPA directly induces brain tumor cell migration, independent of uPA-mediated proteolysis; and in concert with ECM degradation, markedly enhances invasion. Conversely, removing membrane bound uPAR from the surface of the cells studied inhibited their ability to migrate and invade even in the presence of proteolytically active uPA.


Subject(s)
Brain Neoplasms/pathology , Brain Neoplasms/physiopathology , Neoplasm Invasiveness/physiopathology , Receptors, Cell Surface/physiology , Urokinase-Type Plasminogen Activator/pharmacology , Brain Neoplasms/metabolism , Cell Movement/drug effects , Cell Movement/physiology , Collagen , Drug Combinations , Extracellular Matrix/metabolism , Humans , Laminin , Proteoglycans , Receptors, Urokinase Plasminogen Activator , Tumor Cells, Cultured
11.
Adv Exp Med Biol ; 425: 89-97, 1997.
Article in English | MEDLINE | ID: mdl-9433492

ABSTRACT

Our understanding of the role of matrix degrading proteases in cancer has dramatically expanded over the last two decades. From correlative observations linking proteases to cancer progression, we have accumulated evidence supporting a causal role for proteases in various steps of tumor progression and have become increasingly aware of the complex interactions that exist among proteases. Specific natural inhibitors of these proteases have also been identified and their role as potent cytostatic agents in cancer has been suggested. In this article some of the concepts on the role of proteases in cancer are discussed and examples of cooperation between matrix metalloproteinases and the plasmin/plasminogen activators system are presented. The role of protease inhibitors such as tissue inhibitor of metalloproteinases-2 (TIMP-2) and plasminogen activator inhibitor-2 (PAI-2) as inhibitors of tumor growth, invasion and metastasis is discussed.


Subject(s)
Endopeptidases/metabolism , Neoplasms/pathology , Neoplasms/physiopathology , Protease Inhibitors/metabolism , Amino Acid Sequence , Animals , Cell Division/drug effects , Disease Progression , Humans , Metalloendopeptidases/chemistry , Metalloendopeptidases/metabolism , Molecular Sequence Data , Neoplasm Invasiveness , Neoplasm Metastasis , Plasminogen Activator Inhibitor 2/pharmacology , Sequence Alignment , Serine Endopeptidases/chemistry , Serine Endopeptidases/metabolism , Tissue Inhibitor of Metalloproteinase-2/pharmacology
12.
J Biol Chem ; 271(43): 26892-9, 1996 Oct 25.
Article in English | MEDLINE | ID: mdl-8900173

ABSTRACT

We have used a combination of biochemical and immunological methods to probe for proteins that interact with the cytoplasmic form of plasminogen activator inhibitor 2 (PAI-2) and to identify the structure in PAI-2 that mediates the binding. By affinity chromatography on immobilized PAI-2, we purified a collection of PAI-2-binding proteins. These proteins bound 125I-labeled PAI-2 in vitro (IC50, approximately 10-100 nM) in a calcium-independent reaction that did not abrogate the proteinase inhibitory function of PAI-2. Annexin I was identified among the eluted proteins, and purified annexins I, II, IV, and V, but not III and VI, possessed 125I-labeled PAI-2 binding activity. Immune precipitation by anti-PAI-2 monoclonal and polyclonal antibodies of metabolically labeled melanoma cells treated with a cleavable cross-linker prior to analysis revealed three prominent proteins with apparent masses of 100, 70, and 50 kDa. We localized the protein binding domain in PAI-2 between amino acid residues 66 and 98, as determined by using a PAI-2 mutant lacking this domain and a synthetic peptide spanning this region. This region of PAI-2 corresponds to exon 3 of the gene sequence thought to be critical for PAI-2 functions.


Subject(s)
Exons , Plasminogen Activator Inhibitor 2/genetics , Serine Proteinase Inhibitors/genetics , Annexins/isolation & purification , Annexins/metabolism , Binding Sites , Chromatography, Affinity , Humans , Plasminogen Activator Inhibitor 2/chemistry , Plasminogen Activator Inhibitor 2/metabolism , Precipitin Tests , Protein Binding , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Serine Proteinase Inhibitors/metabolism
13.
J Cell Physiol ; 167(2): 333-40, 1996 May.
Article in English | MEDLINE | ID: mdl-8613475

ABSTRACT

Serine proteases and matrix metalloproteinases have been shown to often cooperate in multiple physiological and pathological processes associated with changes in the extracellular matrix (ECM). We have examined the interaction between the plasminogen activator (PA)-plasmin system and matrix metalloproteinases (MMPs) in HT1080 human fibrosarcoma cells treated with 12-O-tetradecanoyl-phorbol-13-acetate (TPA). While TPA treatment evoked a temporary increased expression of urokinase type PA (uPA), the production of both types of human plasminogen activator inhibitors (PAI) was induced and sustained over 12 h by TPA treatment shifting the protease-protease inhibitors balance in favor of the inhibitors. TPA treatment of HT1080 cells induced the expression of interstitial collagenase (MMP-1) and increased the expression of gelatinase B (MMP-9), tissue inhibitor of metalloproteinases-1 (TIMP-1), and MT-MMP, a membrane-bound activator of progelatinase A (proMMP-2), while MMP-2 and TIMP-2 expression were decreased. Increased MT-MMP expression by TPA treatment was associated with increased activation of proMMP-2. These data show that the regulation of PA-plasmin and metalloproteinase and their specific inhibitors is uncoordinated. In addition, inhibition of the PA-plasmin system by PAI-2 or aprotinin did not prevent the activation of proMMP-2 by TPA, suggesting that plasmin is not involved in MT-MMP-mediated activation of proMMP-2.


Subject(s)
Fibrosarcoma/enzymology , Metalloendopeptidases/biosynthesis , Plasminogen Activators/biosynthesis , Aprotinin/pharmacology , Blotting, Northern , Carcinogens/pharmacology , Collagenases/metabolism , Enzyme Precursors/metabolism , Fibrinolysin/pharmacology , Fibrinolytic Agents/pharmacology , Gelatinases/metabolism , Humans , Matrix Metalloproteinase 1 , Matrix Metalloproteinase 2 , Matrix Metalloproteinase 9 , Matrix Metalloproteinases, Membrane-Associated , Metalloendopeptidases/genetics , Metalloendopeptidases/metabolism , Plasminogen Activator Inhibitor 2/pharmacology , Plasminogen Activators/genetics , Protease Inhibitors/pharmacology , Proteins/pharmacology , RNA, Messenger/analysis , Serine Proteinase Inhibitors/pharmacology , Tetradecanoylphorbol Acetate/pharmacology , Time Factors , Tissue Inhibitor of Metalloproteinase-2 , Tumor Cells, Cultured/enzymology
14.
J Invest Dermatol ; 106(5): 1007-11, 1996 May.
Article in English | MEDLINE | ID: mdl-8618030

ABSTRACT

Using a 3-dimensional fibrin gel model system simulating fibroplasia of wound repair, we investigated the interaction between keloid fibroblasts and fibrin matrix and compared it with that of normal fibroblasts. Normal skin fibroblasts caused fibrin gel degradation under serum-free conditions, whereas keloid fibroblasts did not cause microscopically detectable gel degradation. Fibrin gel degradation occurred through plasmin-mediated fibrinolysis, which was initiated by fibroblasts exhibited high uPA but low plasminogen activator inhibitor-1 (PAI-1) activities, and transforming growth factor-beta 1 prevented fibrinolysis of normal fibroblasts by upregulating PAI-1 while downregulating uPA activities. In contrast, keloid fibroblasts exhibited an intrinsically high level of PAI-1 and a low level of uPA. This change in the ratio of activator and inhibitor activities was attributed to altered fibrin degradation by keloid fibroblasts. The PAI-1 increase was also demonstrated at the RNA level by Northern analysis. In terms of the pivotal role of the plasmin/plasminogen activator system in matrix remodeling, the elevated PAI-1 level exhibited by keloid fibroblasts may have significant consequences not only in altered fibrin degradation, but also in subsequent repair steps that lead to keloids and fibrosis.


Subject(s)
Fibrinolysis , Keloid/physiopathology , Plasminogen Activator Inhibitor 1/analysis , Fibrin/metabolism , Fibroblasts/physiology , Humans , Plasminogen Activator Inhibitor 1/genetics , RNA, Messenger/analysis , Urokinase-Type Plasminogen Activator/analysis , Urokinase-Type Plasminogen Activator/genetics
15.
Enzyme Protein ; 49(1-3): 72-84, 1996.
Article in English | MEDLINE | ID: mdl-8796998

ABSTRACT

Several classes of extracellular matrix (ECM)-degrading proteases have been shown to play an important role in tumor invasion and metastasis. Among them, the matrix metalloproteinases (MMPs) and the plasminogen activator (PA)-plasmin system have been the focus of numerous studies. However, few of those have examined the interaction of these two classes of proteases during tumor progression and their specific roles in this complex process have remained unclear. In this article, comparative information on the structure, function, and regulation of these two classes of proteases is reviewed and their interaction on various levels is discussed. This review shows that MMPs and the PA-plasmin system closely cooperate to achieve optimal degradation of the ECM during the invasive and metastatic process.


Subject(s)
Extracellular Matrix/enzymology , Fibrinolysin/metabolism , Metalloendopeptidases/metabolism , Neoplasm Invasiveness , Plasminogen Activators/metabolism , Amino Acid Sequence , Animals , Humans , Models, Biological , Molecular Sequence Data , Proteins/metabolism , Tissue Inhibitor of Metalloproteinase-2
16.
Proc Natl Acad Sci U S A ; 92(1): 205-9, 1995 Jan 03.
Article in English | MEDLINE | ID: mdl-7816818

ABSTRACT

A metastatic human melanoma cell line that produces urokinase-type plasminogen activator was stably transfected with cDNA encoding human plasminogen activator inhibitor 2 (PAI-2). Transfected clones expressed PAI-2 at levels two to nine times higher than both the parental cell line and mock transfectants, as detected by ELISA of cell lysates and conditioned medium. The clone with the highest PAI-2 expression exhibited complete inhibition of soluble and cell-surface-bound plasminogen activator activity. The level of PAI-2 overexpression in these clonal cell lines correlated positively with the inhibition of their ability to degrade extracellular matrix in vitro. Parental, mock-transfected, and PAI-2-transfected cell lines produced rapidly growing tumors when injected s.c. into the skin of mice with severe combined immunodeficiency. The tumors producing the highest levels of PAI-2 were surrounded by a dense tumor capsule. Both parental cells and mock-transfected cells invariably metastasized from s.c. tumors to lymph nodes and lungs of mice. PAI-2-transfected cell lines produced significantly less or no metastases. Taken together, these data indicate a critical role for plasminogen activator activity in melanoma invasion and metastasis.


Subject(s)
Gene Expression , Lung Neoplasms/secondary , Melanoma/pathology , Neoplasm Metastasis/prevention & control , Plasminogen Activator Inhibitor 2/biosynthesis , Skin Neoplasms/pathology , Animals , Cell Line , Extracellular Matrix , Humans , Lung Neoplasms/pathology , Lung Neoplasms/prevention & control , Melanoma/metabolism , Melanoma/secondary , Mice , Mice, SCID , Skin Neoplasms/metabolism , Transfection , Transplantation, Heterologous
17.
Cancer Res ; 53(24): 6051-7, 1993 Dec 15.
Article in English | MEDLINE | ID: mdl-8261421

ABSTRACT

Plasminogen activators (PA) elaborated by tumor cells play an important role in the complex process of tissue invasion and metastasis. In the present study the effect of the PA inhibitor type 2 (PAI-2) on tissue invasion in vitro and in vivo was investigated. Clones either expressing (B-) or not expressing the endogenous PAI-2 gene (C+) were isolated from the human HT1080 fibrosarcoma cell line and transfected with full-length PAI-2 cDNA. Recombinant PAI-2 (rPAI-2) expressed by these cells completely inhibited receptor-bound urokinase activity and partially neutralized secreted PA activity. Degradation of extracellular matrix proteins by these transfected cells was markedly decreased when compared to mock or untransfected control cells. The rPAI-2-expressing cells did not penetrate a multilayer of rat smooth muscle cells in vitro, which was readily invaded and destroyed by control cells. The PAI-2 transfectants remained tumorigenic in athymic/nude mice, but tumors originating from these cells showed the presence of a thick, collagenous capsule absent in tumors formed by control cells. Thus, expression of rPAI-2 in HT1080 cells resulted in neutralization of receptor-bound urokinase with subsequent inhibition of matrix protein degradation and invasion in vitro and induction of a thick, peritumoral capsule in vivo.


Subject(s)
Fibrosarcoma/prevention & control , Plasminogen Activator Inhibitor 2/biosynthesis , Recombinant Proteins/biosynthesis , Animals , Extracellular Matrix/metabolism , Fibrosarcoma/metabolism , Fibrosarcoma/pathology , Mice , Neoplasm Invasiveness , Neoplasm Metastasis , Plasminogen Activator Inhibitor 2/genetics , Rats , Transfection , Tumor Cells, Cultured , Urokinase-Type Plasminogen Activator/genetics
18.
Int J Cancer ; 52(2): 298-304, 1992 Sep 09.
Article in English | MEDLINE | ID: mdl-1325952

ABSTRACT

The human sarcoma cell line HT1080 was found, by in situ hybridization, to consist of cells expressing various levels of urokinase (uPA) and tissue type (tPA) plasminogen activator (PA) suggesting clonal variation of expression of these genes. Colonies originating from single HT1080 cells were, therefore, established and screened for PA activity using a fibrin agarose overlay. Colonies inducing lysis (clone C+ and H+) or no lysis (clones B- and M-) were isolated and tested for mRNA levels of uPA, tPA, uPA receptor (uPAR) and the 3 PA inhibitors (PAI), PAI-1, PAI-2 and protease-nexin I. The different clones revealed considerable variation of expression of the different PA and PAI genes, with lysis-inducing clones expressing mainly the PA genes, whereas non-lysing clones demonstrated higher expression of the PAI genes. Amplification or loss of specific genes was excluded by Southern blotting. The protein levels of cellular and secreted PA and PAI determined by ELISA and Western blots demonstrated a pattern similar to that observed for PA and PAI mRNA concentrations, suggesting clonal differences either on the level of transcription or in RNA processing and/or stability. Due to complex interactions between PA and PAI, neither mRNA nor protein levels of the different genes were predictive for the amount of functional PA activity present in the supernatant or on the cell surface of the different clones. Receptor-bound uPA activity was found to be considerably higher in lysis-inducing than in non-lysing clones and the activity was dependent on neutralization by PAI-1 rather than on the level of uPAR mRNA.


Subject(s)
Gene Expression Regulation, Neoplastic , RNA, Messenger/analysis , Receptors, Cell Surface/genetics , Sarcoma/genetics , Tissue Plasminogen Activator/genetics , Urokinase-Type Plasminogen Activator/genetics , Fibrinolysin/analysis , Gene Expression Regulation, Enzymologic , Humans , RNA, Neoplasm/analysis , Receptors, Urokinase Plasminogen Activator , Tissue Plasminogen Activator/antagonists & inhibitors , Tumor Cells, Cultured , Urokinase-Type Plasminogen Activator/antagonists & inhibitors
19.
Cell Growth Differ ; 3(3): 191-7, 1992 Mar.
Article in English | MEDLINE | ID: mdl-1633110

ABSTRACT

As cells progress through the multistep process of neoplastic transformation, they eventually acquire the property of invasive behavior. Although both plasminogen activators (PA) and their inhibitors (PAI) contribute to this process, their regulation in normal and transformed cells remains poorly defined. Because somatic cell hybrids provide useful tools for examining the transformation pathway, tumorigenic and invasive HeLa cells were fused with human normal vascular smooth muscle cells and tested for invasion-related parameters, including the expression of PA and PAI genes, and matrix degradation. Both parental cell lines produced large amounts of PAI activities with no detectable PA in either cellular or secreted form. Opposite findings were obtained with the hybrid cell lines, which demonstrated the presence of receptor-bound and secreted PA but absence of enzymatically measurable PAI activities. Both urokinase-type and tissue-type PA were found in cell-associated and secreted form in the hybrid cells. In addition, expression of the urokinase-type PA receptor gene was found in the three hybrid cells and the vascular smooth muscle cells but not in the HeLa cells. Expression of active, receptor-bound and secreted PA provided the nontumorigenic hybrid cells with the enzymatic tools to degrade extracellular proteins in a plasminogen-dependent manner. Thus, the hybrid cells lost tumorigenicity while retaining the tissue-degrading capability of HeLa cells. These hybrid cell lines should prove to be important reagents for investigating the complex regulatory control of PA and PAI gene expression.


Subject(s)
Hybrid Cells/physiology , Plasminogen Activators/physiology , Plasminogen Inactivators/metabolism , Extracellular Matrix/enzymology , Gene Expression Regulation , HeLa Cells , Humans , Hybrid Cells/enzymology , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/enzymology , Plasminogen Activators/analysis , Plasminogen Activators/genetics , RNA, Messenger/physiology
20.
Am J Respir Cell Mol Biol ; 6(2): 133-9, 1992 Feb.
Article in English | MEDLINE | ID: mdl-1540377

ABSTRACT

Co-secretion of plasminogen activator inhibitor type 1 (PAI-1) and urokinase-type plasminogen activator was identified in short-term cultures of primary type II pneumocytes isolated from adult rats. After separation by sodium dodecyl sulfate (SDS)-PAGE and reverse fibrin autography (reverse FA) of serum-free conditioned medium (SFCM), cellular lysate, and extracellular matrix (ECM), the inhibitor was seen as a zone of spared lysis at an apparent molecular mass of 46 to 48 kD. The plasminogen activator (PA) activity could only be visualized when human instead of bovine fibrin was used in the indicator gel. It presented as a single band of lysis at an apparent molecular mass of 45 kD when tested by regular FA and was found adjacent to PAI-1 when examined by reverse FA. Immunoblot analysis of type II pneumocyte SFCM, cellular lysate, and ECM revealed two bands at 46 and 48 kD, consistent with the apparent molecular masses (Mr) reported for rat PAI-1 from HTC hepatoma cells. Type II pneumocyte PAI-1 formed SDS-resistant complexes with tissue-type and urokinase-type plasminogen activator and was found to be stable to acid, to short-term exposure to heat, and to the denaturants guanidine HCl and SDS, while being sensitive to treatment with alkali and urea. When levels of type II pneumocyte PAI-1 activity were monitored over time during short-term culture conditions, the level of PAI-1 in SFCM remained stable, whereas activity in the lysate accumulated and activity in the ECM declined.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Lung/metabolism , Plasminogen Inactivators/metabolism , Animals , Blotting, Western , Cells, Cultured , Electrophoresis, Polyacrylamide Gel , Fibrin/metabolism , Lung/cytology , Male , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...