Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Cancer Res ; 54(24): 6517-25, 1994 Dec 15.
Article in English | MEDLINE | ID: mdl-7987851

ABSTRACT

Redistribution of lysosomes to the cell surface and secretion of lysosomal proteases appear to be general phenomena in cells that participate in local proteolysis. In the present study, we have determined whether malignant progression affects the intracellular distribution and secretion of the lysosomal protease cathepsin B in three model systems, each of which consists of cell pairs that differ in their degree of malignancy. The intracellular distribution of vesicles staining for cathepsin B was evaluated by immunofluorescent microscopy and the secretion of cathepsin B was evaluated by two complementary techniques: stopped assays of activity secreted into culture media; and continuous assays of activity secreted from viable (> or = 95%) cells growing on coverslips. We observed that the intracellular distribution of cathepsin B+ vesicles was more peripheral in the cells of higher malignancy in all three model systems and that active cathepsin B was secreted constitutively from these cells. Because an acidic pericellular pH has been shown to induce translocation of lysosomes in macrophages and fibroblasts, we evaluated the intracellular distribution of cathepsin B+ vesicles and secretion of cathepsin B in cell pairs incubated at slightly acidic pH. Acidic pericellular pH induced a redistribution of cathepsin B+ vesicles toward the cell periphery. In the more malignant cells, this resulted with time in reduced intracellular staining for cathepsin B and enhanced secretion of active cathepsin B. Translocation and secretion of cathepsin B were dependent on a functional microtubular system. Both the redistribution of cathepsin B+ vesicles toward the cell surface induced by acidic pH and the constitutive and acidic pH-induced secretion of active cathepsin B could be inhibited by microtubule poisons and stabilizers. We suggest that the redistribution of active cathepsin B to the surface of malignant cells and its secretion may facilitate invasion of these cells.


Subject(s)
Cathepsin B/metabolism , Colorectal Neoplasms/metabolism , Fibrocystic Breast Disease/metabolism , Hydrogen-Ion Concentration , Melanoma, Experimental/metabolism , Animals , Cathepsin B/analysis , Cell Membrane/metabolism , Colorectal Neoplasms/chemistry , Colorectal Neoplasms/pathology , Fibrocystic Breast Disease/chemistry , Fibrocystic Breast Disease/pathology , Humans , Male , Melanoma, Experimental/chemistry , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Microtubules/drug effects , Nocodazole/pharmacology , Paclitaxel/pharmacology , Tumor Cells, Cultured
2.
Cancer Res ; 54(23): 6027-31, 1994 Dec 01.
Article in English | MEDLINE | ID: mdl-7954439

ABSTRACT

The poor prognosis of human malignant gliomas is due to their invasion and recurrence, the molecular mechanisms of which remain poorly characterized. We have accumulated substantial evidence implicating the cysteine protease cathepsin B in human glioma malignancy. Increases in cathepsin B expression were observed throughout progression. In primary brain tumor tissue, transcript abundance (Northern blot analysis) increased in low-grade astrocytoma to high-grade glioblastoma from 3- to 6-fold, respectively, above normal brain levels. This increase correlated with increases in protein abundance (from + to ) as measured by immunohistochemistry. Furthermore, in glioblastoma cell lines increases in transcript abundance (ranging from 3- to 12-fold) were accompanied by increases in enzyme activity (44-133 nmol/min x mg protein). Altered subcellular localization was observed both immunohistochemically and by indirect immunofluorescence confocal microscopy and was found to correlate with increased grade. In addition, this increase in cathepsin B expression and altered subcellular localization correlated with histomorphological invasion and clinical evidence of invasion as detected by magnetic resonance imaging. These data support the hypothesis that cathepsin B plays a role in human glioma progression and invasion.


Subject(s)
Cathepsin B/analysis , Glioma/enzymology , Animals , Blotting, Northern , Cathepsin B/genetics , Glioma/diagnosis , Glioma/pathology , Humans , Immunohistochemistry , Magnetic Resonance Imaging , Neoplasm Invasiveness , Rabbits
3.
Burns ; 42(1): 131-140, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26603913

ABSTRACT

The wound healing process is frequently associated with a number of major clinical challenges, due to the failure of commonly used antibiotics as a remedy for wounds. There have always been fascinating questions about the novel applications of bioactive glasses (BGs) and it is expected that in the next few years these types of materials may play an important role in many aspects of soft tissue regeneration. This research focuses on the feasibility of using silver- and fluoride-containing BGs against multidrug-resistant bacterial strains isolated from patients with burns. According to the results obtained, fluoride did not exhibit antibacterial activity against the tested bacteria, while both 1% and 2% silver-containing BGs inhibited the bacterial growth. It is an important finding that 1% silver-containing BGs showed a potential antibacterial activity without any toxicity against fibroblasts, suggesting that this class of BGs could play a key role in the prevention of infection, reduction of pain, and removal of excessive exudates.


Subject(s)
Anti-Bacterial Agents/pharmacology , Burns/microbiology , Drug Resistance, Multiple, Bacterial , Fluorides/pharmacology , Glass , Pseudomonas aeruginosa/drug effects , Silver/pharmacology , Wound Infection/microbiology , Amikacin/pharmacology , Animals , Aztreonam/pharmacology , Carbenicillin/pharmacology , Cefepime , Ceftazidime/pharmacology , Ceftriaxone/pharmacology , Cell Survival/drug effects , Cephalosporins/pharmacology , Ciprofloxacin/pharmacology , Gentamicins/pharmacology , Humans , Imipenem/pharmacology , Meropenem , Mice , Microbial Sensitivity Tests , NIH 3T3 Cells , Penicillanic Acid/analogs & derivatives , Penicillanic Acid/pharmacology , Piperacillin/pharmacology , Piperacillin, Tazobactam Drug Combination , Thienamycins/pharmacology , Tobramycin/pharmacology
4.
Cell Prolif ; 49(1): 115-21, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26840647

ABSTRACT

OBJECTIVES: Human amniotic membrane (HAM) has been widely used in soft tissue engineering both in its fresh form and decellularized; its efficiency to aid treatment of burn injuries is well known. On the other hand, it has been reported clinically by several studies that human adipose-derived stem cells (hADSC) are a promising cell source for cell therapy for burns. Recently, we have reported a new technique for decellularization of HAM. In this study, potential of prepared decellularized HAM (dHAM) as a viable support for proliferation and delivery of hADSC was investigated. MATERIALS AND METHODS: Amniotic membranes were collected, decellularized and preserved according to the protocol described in our previously published study. hADSC were obtained from the patients undergoing elective liposuction surgery and cells were then seeded on the decellularized membrane for various times. Efficiency of the decellularized membrane as a delivery system for hADSC was investigated by MTT, LDH specific activity, DAPI staining and SEM. RESULTS: The results showed that dHAM provided a supporting microenvironment for cell growth without producing any cytotoxic effects. In addition, the cells were spread out and actively attached to the dHAM scaffold. CONCLUSION: These results strongly suggest that dHAMs have considerable potential as 3D cell-carrier scaffolds for delivery of hADSC, in tissue engineering and regenerative medicine applications.


Subject(s)
Adipose Tissue/cytology , Amnion/cytology , Tissue Engineering/methods , Tissue Survival , Cell Death , Cell Survival , Cells, Cultured , Humans , Indoles/metabolism , Staining and Labeling , Stromal Cells/cytology , Stromal Cells/ultrastructure , Tissue Scaffolds
5.
Burns ; 41(7): 1488-97, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26048133

ABSTRACT

Human amniotic membranes (HAMs) have attracted the attention of burn surgeons for decades due to favorable properties such as their antibacterial activity and promising support of cell proliferation. On the other hand, as a major implication in the health of burn patients, the prevalence of bacteria resistant to multiple antibiotics is increasing due to overuse of antibiotics. The aim of this study was to investigate whether HAMs (both fresh and acellular) are an effective antibacterial agent against antibiotic-resistant bacteria isolated from burn patients. Therefore, a HAM was decellularized and tested for its antibacterial activity. Decellularization of the tissue was confirmed by hematoxylin and eosin (H&E) and 4,6-diamidino-2-phenylindole (DAPI) staining. In addition, the cyto-biocompatibility of the acellular HAM was proven by the cell viability test (3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide, MTT) and scanning electron microscopy (SEM). The resistant bacteria were isolated from burns, identified, and tested for their susceptibility to antibiotics using both the antibiogram and polymerase chain reaction (PCR) techniques. Among the isolated bacteria, three blaIMP gene-positive Pseudomonas aeruginosa strains were chosen for their high resistance to the tested antibiotics. The antibacterial activity of the HAM was also tested for Klebsiella pneumoniae (American Type Culture Collection (ATCC) 700603) as a resistant ATCC bacterium; Staphylococcus aureus (mecA positive); and three standard strains of ATCC bacteria including Escherichia coli (ATCC 25922), Pseudomonas aeruginosa (ATCC 27833), and S. aureus (ATCC 25923). Antibacterial assay revealed that only the latter three bacteria were susceptible to the HAM. All the data obtained from this study suggest that an alternative strategy is required to complement HAM grafting in order to fully protect burns from nosocomial infections.


Subject(s)
Amnion , Bacterial Infections/prevention & control , Biological Dressings , Burns/therapy , Drug Resistance, Bacterial , Amnion/cytology , Analysis of Variance , Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Bacteria/isolation & purification , Bacterial Infections/microbiology , Burns/microbiology , Humans , Microbial Sensitivity Tests , Polymerase Chain Reaction , Wound Healing
6.
Neoplasia ; 2(6): 496-504, 2000.
Article in English | MEDLINE | ID: mdl-11228542

ABSTRACT

Malignant progression is accompanied by degradation of extracellular matrix proteins. Here we describe a novel confocal assay in which we can observe proteolysis by living human breast cancer cells (BT20 and BT549) through the use of quenched-fluorescent protein substrates. Degradation thus was imaged, by confocal optical sectioning, as an accumulation of fluorescent products. With the BT20 cells, fluorescence was localized to pericellular focal areas that coincide with pits in the underlying matrix. In contrast, fluorescence was localized to intracellular vesicles in the BT549 cells, vesicles that also label for lysosomal markers. Neither intracellular nor pericellular fluorescence was observed in the BT549 cells in the presence of cytochalasin B, suggesting that degradation occurred intracellularly and was dependent on endocytic uptake of substrate. In the presence of a cathepsin B-selective cysteine protease inhibitor, intracellular fluorescence was decreased approximately 90% and pericellular fluorescence decreased 67% to 96%, depending on the protein substrate. Matrix metallo protease inhibitors reduced pericellular fluorescence approximately 50%, i.e., comparably to a serine and a broad spectrum cysteine protease inhibitor. Our results suggest that: 1) a proteolytic cascade participates in pericellular digestion of matrix proteins by living human breast cancer cells, and 2) the cysteine protease cathepsin B participates in both pericellular and intracellular digestion of matrix proteins by living human breast cancer cells.


Subject(s)
Breast Neoplasms/enzymology , Peptide Hydrolases/metabolism , Tumor Cells, Cultured/cytology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cathepsin B/antagonists & inhibitors , Cathepsin B/metabolism , Cerebrospinal Fluid Proteins/pharmacology , Cystatin C , Cystatins/pharmacology , Cysteine Proteinase Inhibitors/pharmacology , Dipeptides/pharmacology , Extracellular Matrix/metabolism , Fluorescent Dyes , Humans , Image Processing, Computer-Assisted , Immunoenzyme Techniques , Liver/enzymology , Lysosomes/enzymology , Lysosomes/metabolism , Microscopy, Confocal , Serum Albumin, Bovine/metabolism , Tumor Cells, Cultured/metabolism
7.
J Histochem Cytochem ; 46(6): 745-51, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9603786

ABSTRACT

The cystatin superfamily of cysteine protease inhibitors and target cysteine proteases such as cathepsin B have been implicated in malignant progression. The respective cellular/extracellular localization of cystatins and cysteine proteases in tumors may be critical in regulating activity of the enzymes. Confocal microscopy has enabled us to demonstrate the differential localization of cystatins and cathepsin B in an embryonic liver cell line and an invasive hepatoma cell line. In both, stefins A and B were distributed diffusely throughout the cytoplasm, whereas cystatin C was distributed in juxtanuclear vesicles. Stefin A and cystatin C, but not stefin B, were present on the cell surface. Cystatin C was found on the top surfaces of both cell lines, whereas stefin A was found only on the top surface of the embryonic liver cells. Cathepsin B staining was concentrated in perinuclear vesicles in the embryonic liver cells. In the hepatoma cells, staining for cathepsin B was also present in vesicles adjacent to the cell membrane and on localized regions of the bottom surface. Such a disparate distribution of cathepsin B and its endogenous inhibitors may facilitate proteolysis by the hepatoma cells and thereby contribute to their invasive phenotype.


Subject(s)
Cathepsin B/metabolism , Cysteine Proteinase Inhibitors/metabolism , Cytoplasm/metabolism , Membrane Proteins/metabolism , Animals , Carcinoma, Hepatocellular/metabolism , Cell Line , Cell Nucleus/metabolism , Cystatin A , Cystatin B , Cystatin C , Cystatins/metabolism , Liver/metabolism , Mice , Microscopy, Confocal , Microscopy, Fluorescence , Tumor Cells, Cultured
8.
Int J Dev Neurosci ; 17(5-6): 483-94, 1999.
Article in English | MEDLINE | ID: mdl-10571410

ABSTRACT

Increased expression of cathepsin B has been reported in a number of human and animal tumors. This has also been observed in human gliomas where increases in cathepsin B mRNA, protein, activity and secretion parallel malignant progression. In the present study, we showed that cathepsin B was directly involved in glioma cell invasion. Activity of cathepsin B was an order of magnitude higher in glioma tissue than in matched normal brain. Inhibitors of cysteine proteases reduced invasion of glioma cells in two in vitro models: invasion through Matrigel and infiltration of a glioma spheroid into a normal brain aggregate. Glioma spheroids expressed higher levels of cathepsin B than did monolayers and the ability of subclones differing in cathepsin B activity to infiltrate normal brain aggregates paralleled their cathepsin B activity. We confirmed that intracellular staining for cathepsin B occurs at the cell periphery and in cell processes and observed extracellular staining on the cell surface. In addition, we demonstrated that intracellular cathepsin B located at the cell periphery and in processes was active. The cell surface cathepsin B colocalized with areas of degradation of an extracellular matrix component. We hypothesize that the increased expression of active cathepsin B in gliomas leads to increases in invasion in vitro and in vivo and have developed a xenotransplant model in which this hypothesis can be tested.


Subject(s)
Brain Neoplasms/metabolism , Cathepsin B/genetics , Gene Expression Regulation, Neoplastic/physiology , Glioma/metabolism , Animals , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Cell Line , Cysteine Proteinase Inhibitors/pharmacology , Disease Progression , Glioma/drug therapy , Glioma/pathology , Humans , Neoplasm Invasiveness , Rats , Reference Values , Transplantation, Heterologous
9.
J Neurosurg ; 83(2): 285-90, 1995 Aug.
Article in English | MEDLINE | ID: mdl-7542317

ABSTRACT

The poor prognosis of patients with malignant gliomas is at least partially due to the invasive nature of these tumors. In this study, the authors investigated the possibility that the cysteine protease cathepsin B (CB) is a participant in the process of glial tumor cell invasion. To accomplish this, an immunohistochemical analysis was made of the localization of antibodies to CB in biopsies of five specimens of normal brain, 16 astrocytomas, 33 anaplastic astrocytomas, and 33 glioblastomas multiforme. Staining was scored according to the percentage of positive cells and the intensity of the stain, graded from 0 to 3+. Staining for CB was not seen in any of five samples of normal brain except for occasional neuronal cell bodies and microglia. Only five (31%) of 16 astrocytomas showed a small percentage of positive cells (0.01%-3%) that were stained in a light, diffuse cytoplasmic pattern (1+). Twenty-nine (87.8%) of 33 anaplastic astrocytomas showed positive light, granular staining in 2% to 40% of cells. In anaplastic astrocytoma, the staining within a tumor was heterogeneous with intensities of 1+ (17%), 1+ to 2+ (29%), or 2+ (55%). In contrast, all 33 (100%) glioblastomas were positive in 10% to 90% of cells. The staining was present in a coarse, granular pattern with an intensity of 2+ (12%) or 3+ (88%). Tumor cells infiltrating into brain adjacent to malignant gliomas stained positively in 26 cases that could be evaluated for glioblastoma multiforme; these invading cells frequently followed penetrating blood vessels as typical "secondary structures of Scherer." Moderate to intense CB staining associated with endothelial proliferation in high-grade tumors was also observed, especially in regions of tumor infiltration into adjacent normal brain. These results provide evidence consistent with the hypothesis that CB is functionally significant in the process of tumor invasion and angiogenesis in the clinical progression of the malignant phenotype in astrocytes.


Subject(s)
Brain Neoplasms/pathology , Cathepsin B/analysis , Glioma/pathology , Antibodies, Monoclonal , Astrocytoma/pathology , Brain/cytology , Brain Neoplasms/blood supply , Cell Division , Cytoplasm/pathology , Disease Progression , Endothelium, Vascular/pathology , Glioblastoma/pathology , Glioma/blood supply , Humans , Immunoenzyme Techniques , Immunohistochemistry , Microglia/ultrastructure , Neoplasm Invasiveness , Neovascularization, Pathologic/pathology , Neurons/ultrastructure , Staining and Labeling
10.
Pathol Oncol Res ; 4(4): 283-96, 1998.
Article in English | MEDLINE | ID: mdl-9887359

ABSTRACT

Increased expression and alteration of intracellular trafficking of lysosomal cathepsins have been reported in malignant tumors, or in cells transformed by the transfection with the ras oncogene. In the present study, immortal MCF-10A human breast epithelial cells were transformed with the mutated ras oncogene. Both cell lines were investigated for changes in the intracellular localization of lysosomal cathepsin D and lamp-1 (lysosome-associated membrane protein) employing specific antibodies and confocal immunofluorescence microscopy. The results revealed that staining for cathepsin D along with for lamp-1 was mostly localized in the perinuclear region of MCF-10A cells. In contrast, the staining for these proteins was found to be widely distributed throughout the cytoplasm and at the cell periphery in MCF-10AneoT cells. The organization of microtubules, but not actin, appeared to differ between MCF-10A cells and their oncogenic ras transfectants. When the microtubules were depolymerized by treatment of MCF-10A cells with nocodazole, vesicles containing the lysosomal cathepsin D were dispersed in the cytoplasm and translocation of these vesicles to the cell periphery was observed. The intracellular localization of cathepsin D in the nocodazole-treated MCF-10A cells seemed to be similar to that observed in the oncogenic ras transfectants of these cells. When taxol, which inhibits microtubule depolymerization, was added to the culture medium of neoT cells, a polymerized microtubule network was observed, and the reclustering of cathepsin D and lamp-1 occurred in an unidirectional manner towards the perinuclear region. These findings support a model in which cytoskeletal microtubule organization is closely related to the trafficking of lysosomes/endosomes, and in which oncogenic ras interferes with such organization in human breast epithelial cells.


Subject(s)
Breast/cytology , Cathepsin D/metabolism , Cell Transformation, Neoplastic/ultrastructure , Epithelial Cells/enzymology , Lysosomes/enzymology , Animals , Antigens, CD/metabolism , Biological Transport , Breast/enzymology , Cell Line, Transformed/drug effects , Cell Line, Transformed/enzymology , Cell Line, Transformed/ultrastructure , Cytochalasin D/pharmacology , Epithelial Cells/ultrastructure , Female , Fibrocystic Breast Disease/pathology , Humans , Immunoglobulin G/immunology , Lysosomal Membrane Proteins , Male , Membrane Glycoproteins/metabolism , Microscopy, Confocal , Microscopy, Fluorescence , Microtubules/drug effects , Microtubules/ultrastructure , Nocodazole/pharmacology , Oncogene Protein p21(ras)/genetics , Oncogene Protein p21(ras)/physiology , Paclitaxel/pharmacology , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/physiology , Rabbits , Rats , Recombinant Fusion Proteins/physiology , Transfection
11.
Oncogene ; 33(36): 4474-84, 2014 Sep 04.
Article in English | MEDLINE | ID: mdl-24077280

ABSTRACT

The cysteine protease cathepsin B (CTSB) is frequently overexpressed in human breast cancer and correlated with a poor prognosis. Genetic deficiency or pharmacological inhibition of CTSB attenuates tumor growth, invasion and metastasis in mouse models of human cancers. CTSB is expressed in both cancer cells and cells of the tumor stroma, in particular in tumor-associated macrophages (TAM). In order to evaluate the impact of tumor- or stromal cell-derived CTSB on Polyoma Middle T (PyMT)-induced breast cancer progression, we used in vivo and in vitro approaches to induce human CTSB overexpression in PyMT cancer cells or stromal cells alone or in combination. Orthotopic transplantation experiments revealed that CTSB overexpression in cancer cells rather than in the stroma affects PyMT tumor progression. In 3D cultures, primary PyMT tumor cells showed higher extracellular matrix proteolysis and enhanced collective cell invasion when CTSB was overexpressed and proteolytically active. Coculture of PyMT cells with bone marrow-derived macrophages induced a TAM-like macrophage phenotype in vitro, and the presence of such M2-polarized macrophages in 3D cultures enhanced sprouting of tumor spheroids. We employed a doxycycline (DOX)-inducible CTSB expression system to selectively overexpress human CTSB either in cancer cells or in macrophages in 3D cocultures. Tumor spheroid invasiveness was only enhanced when CTSB was overexpressed in cancer cells, whereas CTSB expression in macrophages alone did not further promote invasiveness of tumor spheroids. We conclude that CTSB overexpression in the PyMT mouse model promotes tumor progression not by a stromal effect, but by a direct, cancer cell-inherent mode of action: CTSB overexpression renders the PyMT cancers more invasive by increasing proteolytic extracellular matrix protein degradation fostering collective cell invasion into adjacent tissue.


Subject(s)
Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/pathology , Cathepsin B/metabolism , Extracellular Matrix Proteins/metabolism , Macrophages/metabolism , Stromal Cells/transplantation , Animals , Antigens, Polyomavirus Transforming/genetics , Breast Neoplasms/metabolism , Carcinoma, Ductal, Breast/metabolism , Cathepsin B/genetics , Disease Progression , Doxycycline/pharmacology , Female , Gene Expression Regulation, Neoplastic , Humans , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Transgenic
12.
Biol Chem ; 382(5): 785-8, 2001 May.
Article in English | MEDLINE | ID: mdl-11517931

ABSTRACT

Degradation of basement membrane is an essential step for tumor invasion. In order to study degradation in real time as well as localize the site of proteolysis, we have established an assay with living human cancer cells in which we image cleavage of quenched-fluorescent basement membrane type IV collagen (DQ-collagen IV). Accumulation of fluorescent products is imaged with a confocal microscope and localized by optically sectioning both the cells and the matrix on which they are growing. For the studies described here, we seeded U87 human glioma cells as either monolayers or spheroids on a 3-dimensional gelatin matrix in which DQ-collagen IV had been embedded. As early as 24 hours after plating as monolayers, U87 cells were present throughout the 3-dimensional matrix. Cells at all levels had accumulated fluorescent degradation products of DQ-collagen IV intracellularly within vesicles. Similar observations were made for U87 spheroids and the individual cells migrating from the spheroids into the gelatin matrix. Both the migrating cells and those within the spheroid contained fluorescent degradation products of DQ-collagen IV intracellularly within vesicles. Thus, glioma cells like breast cancer cells are able to degrade type IV collagen intracellularly, suggesting that this is an important pathway for matrix degradation.


Subject(s)
Glioma/enzymology , Image Processing, Computer-Assisted , Peptide Hydrolases/metabolism , Cathepsin B/metabolism , Collagen Type IV/metabolism , Extracellular Matrix/metabolism , Fluorescent Dyes , Glioma/metabolism , Glioma/pathology , Humans , Lysosomes/enzymology , Lysosomes/metabolism , Microscopy, Confocal , Neoplasm Invasiveness , Tumor Cells, Cultured
13.
Biol Chem ; 379(2): 113-23, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9524062

ABSTRACT

Cathepsin B has been implicated in progression of various human tumors. Overexpression of cathepsin B mRNA, increased cathepsin B staining and elevated cathepsin B activity have been found in different human cancers. These occur especially at the invasive edges of cancers, suggesting a role for cathepsin B in tumor invasion. In some tumors, mRNA expression and protein staining for cathepsin B correlate with clinical progression. Cathepsin B can facilitate tumor progression directly through degradation of components of the basement membrane and extracellular matrix. In vitro studies show that cathepsin B may exert its degradative effects intracellularly or extracellularly, depending on the cell type and location of cathepsin B. Cathepsin B can also facilitate tumor progression indirectly through activation of other latent proteases and/or degradation of protein inhibitors of other proteases. Thus cathepsin B may be an integral component of the proteolytic cascade linked to malignant progression of human tumors.


Subject(s)
Cathepsin B/metabolism , Neoplasms/physiopathology , Animals , Disease Progression , Humans
14.
Biomed Biochim Acta ; 50(4-6): 549-54, 1991.
Article in English | MEDLINE | ID: mdl-1801721

ABSTRACT

Expression, membrane association and secretion of both cathepsin B and cathepsin L have been associated with the malignant phenotype of murine B16 melanomas. Only native forms of the two enzymes were found in the lysosomal fractions, whereas both native and latent forms were found in the membrane fractions. Brief exposure to acid pH induced secretion of only native forms of both cathepsin B and cathepsin L and a concomitant reduction in membrane-associated activities. Thus, the pericellular acidification associated with malignant tumors may provide optimal conditions for proteolysis by the cysteine proteinases in terms of their enhanced stability and their induced release in native forms not requiring proteolytic activation.


Subject(s)
Cathepsin B/metabolism , Cathepsins/metabolism , Endopeptidases , Tumor Cells, Cultured/enzymology , Animals , Cathepsin L , Cell Membrane/enzymology , Cysteine Endopeptidases , Hydrogen-Ion Concentration , Lysosomes/enzymology , Melanoma, Experimental/enzymology , Phenotype
15.
Biochem J ; 285 ( Pt 2): 427-34, 1992 Jul 15.
Article in English | MEDLINE | ID: mdl-1637335

ABSTRACT

Cathepsin B was purified from normal human liver and several human tumour tissues and partially characterized. Three forms of cathepsin B, with molecular masses of 25 kDa, 26 kDa (the two appearing as a doublet) and 30 kDa, were detected in SDS/polyacrylamide gels. The 25-26 kDa doublet was associated with the fractions from tumours and normal liver containing the highest cathepsin B activity. Cathepsin B from both sources showed similar pH optima. Both normal liver and tumour cathepsin B exhibited similar kinetics against selected synthetic substrates. At neutral pH and 24 degrees C, cathepsin B from both normal liver and tumour exhibited a lower Km and a higher kcat./Km than at pH 6.0. Their inhibitory profiles against synthetic inhibitors were also similar. Immunological studies with a monospecific antibody against the mature double-chain form of human liver cathepsin B and an antibody against a cathepsin B-derived synthetic peptide established the immunological similarity of liver and tumour enzymes. The N-terminal sequences of the 25 kDa and 26 kDa forms were identical with that of the heavy chain of the mature double-chain form of human cathepsin B, whereas the N-terminal sequence of the 30 kDa species was identical with that of the single-chain form of human cathepsin B. Treatment of the double-chain form of cathepsin B from normal liver and tumours with the endoglycosidase peptide-N4-(N-acetyl-beta-glucosaminyl)asparagine amidase converted the 26 kDa form into 25 kDa in SDS/polyacrylamide gels, suggesting that cathepsin B may exist as both glycosylated and unglycosylated forms. Our results, in contrast with those reported earlier for mouse cathepsin B, indicate that human liver and tumour cathepsin B are similar.


Subject(s)
Cathepsin B/metabolism , Liver/enzymology , Neoplasms/enzymology , Amino Acid Sequence , Binding Sites , Blotting, Western , Carbohydrate Metabolism , Cathepsin B/genetics , Cathepsin B/isolation & purification , Electrophoresis, Polyacrylamide Gel , Humans , Hydrogen-Ion Concentration , Kinetics , Molecular Sequence Data , Molecular Weight
16.
Exp Cell Res ; 260(2): 292-303, 2000 Nov 01.
Article in English | MEDLINE | ID: mdl-11035924

ABSTRACT

Transfection of Rat1 fibroblasts with an activated form of rac1 (V12rac1) stimulated cell migration in vitro compared to transfection of Rat1 fibroblasts with vector only or with dominant negative rac1 (N17rac1). To investigate the involvement of proteases in this migration, we used a novel confocal assay to evaluate the ability of the Rat1 transfectants to degrade a quenched fluorescent protein substrate (DQ-green bovine serum albumin) embedded in a three-dimensional gelatin matrix. Cleavage of the substrate results in fluorescence, thus enabling one to image extracellular and intracellular proteolysis by living cells. The Rat1 transfectants accumulated degraded substrate intracellularly. V12rac1 increased accumulation of the fluorescent product in vesicles that also labeled with the lysosomal marker LysoTracker. Treatment of the V12rac1-transfected cells with membrane-permeable inhibitors of lysosomal cysteine proteases and a membrane-permeable selective inhibitor of the cysteine protease cathepsin B significantly reduced intracellular accumulation of degraded substrate, indicating that degradation occurred intracellularly. V12rac1 stimulated uptake of dextran 70 (a marker of macropinocytosis) and polystyrene beads (markers of phagocytosis) into vesicles that also labeled for cathepsin B. Thus, stimulation of the endocytic pathways of macropinocytosis and phagocytosis by activated Rac1 may be responsible for the increased internalization and subsequent degradation of extracellular proteins.


Subject(s)
Cathepsin B/metabolism , Cell Movement/physiology , Endocytosis/physiology , rac1 GTP-Binding Protein/metabolism , Animals , Biomarkers , Cathepsin B/antagonists & inhibitors , Cell Line , Cysteine Proteinase Inhibitors/pharmacology , Dextrans/metabolism , Dipeptides/pharmacology , Enzyme Activation , Gelatin , Humans , Intracellular Membranes/enzymology , Leucine/analogs & derivatives , Leucine/pharmacology , Lysosomes/metabolism , Phagocytosis/physiology , Pinocytosis/physiology , Rats , Serum Albumin, Bovine/metabolism , rac1 GTP-Binding Protein/genetics
17.
Biochem Cell Biol ; 74(6): 799-810, 1996.
Article in English | MEDLINE | ID: mdl-9164649

ABSTRACT

Experimental and clinical evidence reveals that the growth of solid tumors is dependent on angiogenesis. Proteolytic enzymes such as plasminogen activators and matrix metalloproteinases have been implicated in this neovascularization. The role of lysosomal proteases in this process has yet to be explored. Increased expression of the lysosomal cysteine protease cathepsin B has been observed in many etiologically different tumors, including human brain, prostate, breast, and gastrointestinal cancers. Immunohistochemical and in situ histochemical studies have demonstrated expression of cathepsin B in neovessels induced during malignant progression of human glioblastoma and prostate carcinomas. In these two tumor types, neovessels stain strongly for cathepsin B compared with the normal microvasculature. As an initial point to elucidate whether cathepsin B is an important component of the angiogenic response in tumours, we analyzed expression of cathepsin B in endothelial cells during neovessel formation. We present evidence for strong immunostaining of cathepsin B in rat brain microvascular endothelial cells as they form capillary tubes in vitro. This finding is discussed within the general framework of the role of proteolytic enzymes in tumor invasion and angiogenesis.


Subject(s)
Cathepsin B/physiology , Neoplasms/blood supply , Neovascularization, Pathologic , Animals , Cathepsin B/analysis , Endothelium, Vascular/chemistry , Humans , Neoplasm Invasiveness
18.
J Cell Sci ; 107 ( Pt 2): 373-84, 1994 Feb.
Article in English | MEDLINE | ID: mdl-8207069

ABSTRACT

Alterations in trafficking and increases in expression of the lysosomal proteases cathepsins B, D and L have been observed in transformed cells and malignant tumors, including human breast carcinoma. ras and the related rab proteins participate in the vesicular transport processes required for normal trafficking of lysosomal enzymes. In addition, transfection of murine fibroblasts with the ras oncogene has been shown to increase the expression of cathepsins L and B. As human cancers are primarily epithelial in origin, we have investigated whether there are alterations in the trafficking and expression of cathepsin B in MCF-10 human breast epithelial cells transfected with wild-type and mutated ras. In all cells examined, i.e. mortal MCF-10M cells, immortal MCF-10A or MCF-10F cells, and transfected MCF-10A cells (transfected with the neomycin resistance gene (MCF-10Aneo) or cotransfected with wild-type proto-oncogenic ras (MCF-10AneoN) or mutated oncogenic ras (MCF-10AneoT)), levels of mRNA transcripts for cathepsin B were similar. However, alterations in trafficking of cathepsin B were observed in the cells transfected with oncogenic ras. In these cells there was an increased association of cathepsin B activity and cathepsin B protein with plasma membrane/endosomal fractions and a more peripheral distribution of immunofluorescent staining for cathepsin B. At the electron microscopic level, immunogold labeling for cathepsin B was localized to the cell membrane as well as to vesicles in the microvilli and adjacent to the cell membrane. In the parental MCF-10A cells, in contrast, cathepsin B was localized to vesicles in the perinuclear region. The cathepsin B associated with plasma membrane/endosomal fractions in the cells transfected with oncogenic ras was mature cathepsin B as demonstrated by immunoblot analysis. This was confirmed further by showing an absence of peripheral immunofluorescent staining in these cells using an antibody specific for the propeptide of cathepsin B. Thus, we have demonstrated by multiple techniques that transfection of human breast epithelial cells with oncogenic ras results in alterations in the trafficking of cathepsin B similar to those observed previously in human and animal tumors of both epithelial and mesenchymal origin.


Subject(s)
Cathepsin B/metabolism , Genes, ras , Breast/cytology , Breast/metabolism , Cell Line , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/ultrastructure , Epithelial Cells , Epithelium/metabolism , Female , Humans , Immunohistochemistry , Microscopy, Immunoelectron , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transfection
19.
J Neurochem ; 65(3): 1035-45, 1995 Sep.
Article in English | MEDLINE | ID: mdl-7643083

ABSTRACT

The cysteine proteinase cathepsin B (CB) was isolated from immortalized murine BV-2 microglial cells and examined via sodium dodecyl sulfate-polyacrylamide gel electrophoresis and immunoblotting to establish physicochemical properties of CB from what is generally considered the resident CNS macrophage. Microglial proteases have been implicated in several pathological processes occurring in the CNS, including neurodegeneration. Murine microglial CB was observed to consist of two major single-chain species of 32 and 34 kDa, with pls of 5.5-5.2 and 5.1-4.5, respectively. In addition, a minor 24-kDa CB species was also observed in some microglial preparations. The major CB isozymes in microglia differed from those observed in murine liver and brain, which consisted of both single- and double-chain CB variants of 31 and 24-25 kDa/5 kDa, respectively, with pl values of 5.5-4.5. A microglial pro-CB of 37 kDa was also isolated, which could be processed to the 34-kDa single-chain CB species. Cystatin was observed to prevent pro-CB processing, whereas E-64 and leupeptin were only partially inhibitory. The 37-kDa pro-CB species was observed to undergo processing into the 34-kDa CB species when incubated at pH 5.5 but remained stable with respect to molecular mass when incubated at pH 7.0. In contrast, the 34-kDa single-chain CB species was observed to autodegrade when incubated at pH 7.0, whereas incubation at pH 5.5 did not affect the integrity of the species as monitored by immunoblotting. Both pro-CB and 32-kDa single-chain CB species were observed extracellularly following lipopolysaccharide activation of BV-2 microglial cells.


Subject(s)
Cathepsin B/metabolism , Neuroglia/enzymology , Animals , Brain/enzymology , Cathepsin B/chemistry , Cathepsin B/isolation & purification , Cell Line , Culture Media , Electrophoresis, Polyacrylamide Gel , Enzyme Precursors/metabolism , Enzyme Stability , Hydrogen-Ion Concentration , Immunoblotting , Isoelectric Point , Isoenzymes/chemistry , Isoenzymes/isolation & purification , Isoenzymes/metabolism , Liver/enzymology , Male , Mice , Protease Inhibitors/pharmacology
20.
Eur J Biochem ; 264(1): 100-9, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10447678

ABSTRACT

Lysosomal cathepsin B has been implicated in parasitic, inflammatory and neoplastic diseases. Most of these pathologies suggest a role for cathepsin B outside the cells, although the origin of extracellular active enzyme is not well defined. The activity of extracellular cathepsin B is difficult to assess because of the presence of inhibitors and inactivation of the enzyme by oxidizing agents. Therefore, we have developed a continuous assay for measurement of cathepsin B activity produced pericellularly by living cells. The kinetic rate of Z-Arg-Arg-NHMec conversion was monitored and the assay optimized for enzyme stability, cell viability and sensitivity. To validate the assay, we determined that human liver cathepsin B was stable and active under the conditions of the assay and its activity could be inhibited by the selective epoxide derivative CA-074. Via this assay, we were able to demonstrate that active cathepsin B was secreted pericellularly by viable cells. Both preneoplastic and malignant cells secreted active cathepsin B. Pretreatment of cells with the membrane-permeant proinhibitor CA-074Me completely abolished pericellular and total cathepsin B activity whereas pretreatment with the active drug CA-074 had no effect. Immunoprecipitation and immunoblotting experiments suggested that the active enzyme species was 31-kDa single-chain cathepsin B. Exocytosis of cathepsin B was not related to secretion of proenzyme or secretion from mature lysosomes. Our results suggest an alternative pathway for exocytosis of active cathepsin B.


Subject(s)
Cathepsin B/metabolism , Exocytosis , Cathepsin B/antagonists & inhibitors , Cathepsin B/chemistry , Cell Line, Transformed , Fluorescent Antibody Technique , Humans , Hydrogen-Ion Concentration , Kinetics , Molecular Weight , Reproducibility of Results , Sensitivity and Specificity , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL