Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
Biochimie ; 139: 27-37, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28528272

ABSTRACT

The asparaginyl endopeptidase legumain and its inhibitor cystatin E/M are endogenously glycosylated. However, little is known about the nature of the carbohydrate groups and whether they affect the functions of these proteins. In this study both glycosylated and unglycosylated forms of legumain and cystatin E/M were studied. HEK293 cell lines stably over-expressing legumain or cystatin E/M, and HCT116 cells were used as cell models, and mature legumain was purified from bovine kidneys. To obtain unglycosylated proteins, cells were treated with tunicamycin, an inhibitor of N-linked glycosylation, whereas PNGase F and Endo H were used to characterize the glycosylation types. Cells were incubated with glycosylated, unglycosylated proteins and/or legumain selective activity-based probe, and legumain and/or cystatin E/M was studied by activity measurement, ELISA or immunoblotting in cell lysates or conditioned media. Legumain and probe in whole cells were studied by immunofluorescence. The carbohydrates on legumain were shown to be of the hybrid or high mannose type, whereas cystatin E/M was characterized as complex mannose-linked. While glycosylated prolegumain was able to autoactivate, the unglycosylated form was not, and addition of glycosaminoglycans did not facilitate autoactivation of unglycosylated prolegumain. Glycosylated prolegumain was internalized and processed to the mature active form, but no internalization of unglycosylated prolegumain was observed. A Cy5-labelled legumain specific activity-based probe (MP-L09) was synthesized and shown to be a novel tool to study intracellular legumain. Also, internalization of mature legumain (36 kDa) was visualized both alone and complexed with probe. Contrary to the importance of legumain glycosylation, both glycosylated and unglycosylated cystatin E/M showed similar capacity to inhibit legumain. In conclusion, glycosylation of prolegumain is necessary for correct processing to active forms and internalization, whereas the inhibitory property of cystatin E/M is independent of the glycosylation status.


Subject(s)
Cystatin M/pharmacology , Cysteine Endopeptidases/metabolism , Glycosaminoglycans/metabolism , Mannose/chemistry , Protein Processing, Post-Translational , Cysteine Endopeptidases/chemistry , Glycosylation , HCT116 Cells , HEK293 Cells , Humans
2.
Cancer Res Treat ; 49(2): 374-386, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27488871

ABSTRACT

PURPOSE: When integrating molecularly targeted compounds in radiotherapy, synergistic effects of the systemic agent and radiation may extend the limits of patient tolerance, increasing the demand for understanding the pathophysiological mechanisms of treatment toxicity. In this Pelvic Radiation and Vorinostat (PRAVO) study, we investigated mechanisms of adverse effects in response to the histone deacetylase (HDAC) inhibitor vorinostat (suberoylanilide hydroxamic acid, SAHA) when administered as a potential radiosensitiser. MATERIALS AND METHODS: This phase I study for advanced gastrointestinal carcinoma was conducted in sequential patient cohorts exposed to escalating doses of vorinostat combined with standard-fractionated palliative radiotherapy to pelvic target volumes. Gene expression microarray analysis of the study patient peripheral blood mononuclear cells (PBMC) was followed by functional validation in cultured cell lines and mice treated with SAHA. RESULTS: PBMC transcriptional responses to vorinostat, including induction of apoptosis, were confined to the patient cohort reporting dose-limiting intestinal toxicities. At relevant SAHA concentrations, apoptotic features (annexin V staining and caspase 3/7 activation, but not poly-(ADP-ribose)-polymerase cleavage) were observed in cultured intestinal epithelial cells. Moreover, SAHA-treated mice displayed significant weight loss. CONCLUSION: The PRAVO study design implemented a strategy to explore treatment toxicity caused by an HDAC inhibitor when combined with radiotherapy and enabled the identification of apoptosis as a potential mechanism responsible for the dose-limiting effects of vorinostat. To the best of our knowledge, this is the first report deciphering mechanisms of normal tissue adverse effects in response to an HDAC inhibitor within a combined-modality treatment regimen.


Subject(s)
Antineoplastic Agents/adverse effects , Apoptosis/drug effects , Histone Deacetylase Inhibitors/adverse effects , Intestinal Mucosa/metabolism , Intestines/drug effects , Aged , Aged, 80 and over , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/genetics , Biomarkers , Cell Line , Clinical Trials, Phase I as Topic , Combined Modality Therapy , Female , Gastrointestinal Neoplasms/genetics , Gastrointestinal Neoplasms/metabolism , Gastrointestinal Neoplasms/pathology , Gastrointestinal Neoplasms/therapy , Gene Expression Profiling , Histone Deacetylase Inhibitors/therapeutic use , Humans , Hydroxamic Acids/adverse effects , Hydroxamic Acids/therapeutic use , Intestines/pathology , Male , Mice , Middle Aged , Pelvis/radiation effects , Radiotherapy/methods , Rats , Transcriptome , Vorinostat
3.
Clin Exp Metastasis ; 32(8): 755-67, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26349943

ABSTRACT

S100A4 promotes metastasis in several types of cancer, but the involved molecular mechanisms are still incompletely described. The protein is associated with a wide variety of biological functions and it locates to different subcellular compartments, including nuclei, cytoplasm and extracellular space. Nuclear expression of S100A4 has been associated with more advanced disease stage as well as poor outcome in colorectal cancer (CRC). The present study was initiated to investigate the nuclear function of S100A4 and thereby unravel potential biological mechanisms linking nuclear expression to a more aggressive phenotype. CRC cell lines show heterogeneity in nuclear S100A4 expression and preliminary experiments revealed cells in G2/M to have increased nuclear accumulation compared to G1 and S cells, respectively. Synchronization experiments validated nuclear S100A4 expression to be most prominent in the G2/M phase, but manipulating nuclear levels of S100A4 using lentiviral modified cells failed to induce changes in cell cycle distribution and proliferation. Proximity ligation assay did, however, demonstrate proximity between S100A4 and cyclin B1 in vitro, while confocal microscopy showed S100A4 to localize to areas corresponding to centrosomes in mitotic cells prior to chromosome segregation. This might indicate a novel and uncharacterized function of the metastasis-associated protein in CRC cells.


Subject(s)
Cell Nucleus/chemistry , Centrosome/physiology , Colorectal Neoplasms/pathology , Cyclin B1/physiology , S100 Proteins/physiology , Animals , Cell Division , Cell Line, Tumor , G2 Phase , Humans , Mice , S100 Calcium-Binding Protein A4 , Tumor Suppressor Protein p53/physiology
4.
Eur J Cancer ; 51(1): 9-17, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25466510

ABSTRACT

BACKGROUND: The cysteine proteinase legumain is highly expressed in cancer. Legumain is a potential biomarker and has been suggested to be utilised for prodrug activation in cancer therapy. However, to define the suitability of legumain for such purposes, detailed knowledge of cell type-specific and subcellular expression together with proteolytic activity patterns in tumour tissue is necessary. METHODS: Expression of legumain was examined in a panel of 277 primary tumours from colorectal cancer (CRC) patients using immunohistochemistry. Tumour (cytoplasmic diffuse, cytoplasmic granulated, and nuclear) and stromal cell expression of legumain was quantified, and associations with clinicopathological parameters and outcome were analysed. Additionally, normal colon tissue and spontaneous mouse tumours were stained for legumain. RESULTS: Legumain was highly expressed in tumour and stromal cells. Nuclear legumain was detected in 30% of the tumours. In colon cancer patients, high legumain expression was associated with overall and metastasis-free survival (OS; MFS) in uni- and multivariate analysis. Nuclear legumain was associated with poor OS, but not MFS in the colon cancer subgroup. Cytoplasmic granulated or diffuse expression was not associated with OS or MFS. Normal epithelial cells exhibited granulated legumain mainly at the apical pole, and legumain was highly expressed in CD68 positive macrophages. CONCLUSIONS: Legumain is a highly expressed proteinase in CRC and associated with poor outcome in colon cancer. Diversified localisation of legumain expression in tumour and stromal cells suggests multiple functions in CRC, representing both a challenge and an opportunity for use in therapeutic targeting.


Subject(s)
Colorectal Neoplasms/therapy , Cysteine Endopeptidases/metabolism , Cysteine Proteases/metabolism , Cohort Studies , Colorectal Neoplasms/pathology , Cysteine Endopeptidases/adverse effects , Cysteine Proteases/adverse effects , Female , Humans , Male , Prodrugs , Prognosis
5.
Cancer Lett ; 344(1): 28-39, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24215866

ABSTRACT

Tumor cells have the ability to exploit stromal cells to facilitate metastasis. By using malignant melanoma as a model, we show that the stroma adjacent to metastatic lesions is enriched in the known metastasis-promoting protein S100A4. S100A4 stimulates cancer cells to secrete paracrine factors, such as inflammatory cytokines IL8, CCL2 and SAA, which activate stromal cells (endothelial cells and monocytes) so that they acquire tumor-supportive properties. Our data establishes S100A4 as an inducer of a cytokine network enabling tumor cells to engage angiogenic and inflammatory stromal cells, which might contribute to pro-metastatic activity of S100A4.


Subject(s)
Melanoma/metabolism , S100 Proteins/metabolism , Stromal Cells/metabolism , Tumor Microenvironment/physiology , Animals , Cell Line, Tumor , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Heterografts , Humans , Immunohistochemistry , Inflammation/metabolism , Melanoma/pathology , Mice , Mice, Nude , Neoplasm Invasiveness , Real-Time Polymerase Chain Reaction , Receptor Cross-Talk/physiology , S100 Calcium-Binding Protein A4 , Stromal Cells/pathology
6.
PLoS One ; 8(1): e52980, 2013.
Article in English | MEDLINE | ID: mdl-23326369

ABSTRACT

The cysteine protease legumain is involved in several biological and pathological processes, and the protease has been found over-expressed and associated with an invasive and metastatic phenotype in a number of solid tumors. Consequently, legumain has been proposed as a prognostic marker for certain cancers, and a potential therapeutic target. Nevertheless, details on how legumain advances malignant progression along with regulation of its proteolytic activity are unclear. In the present work, legumain expression was examined in colorectal cancer cell lines. Substantial differences in amounts of pro- and active legumain forms, along with distinct intracellular distribution patterns, were observed in HCT116 and SW620 cells and corresponding subcutaneous xenografts. Legumain is thought to be located and processed towards its active form primarily in the endo-lysosomes; however, the subcellular distribution remains largely unexplored. By analyzing subcellular fractions, a proteolytically active form of legumain was found in the nucleus of both cell lines, in addition to the canonical endo-lysosomal residency. In situ analyses of legumain expression and activity confirmed the endo-lysosomal and nuclear localizations in cultured cells and, importantly, also in sections from xenografts and biopsies from colorectal cancer patients. In the HCT116 and SW620 cell lines nuclear legumain was found to make up approximately 13% and 17% of the total legumain, respectively. In similarity with previous studies on nuclear variants of related cysteine proteases, legumain was shown to process histone H3.1. The discovery of nuclear localized legumain launches an entirely novel arena of legumain biology and functions in cancer.


Subject(s)
Cell Nucleus/enzymology , Colorectal Neoplasms/enzymology , Cysteine Endopeptidases/metabolism , Lysosomes/enzymology , Amino Acid Sequence , Animals , Cell Line, Tumor , Colorectal Neoplasms/pathology , Cysteine Endopeptidases/genetics , Female , HCT116 Cells , HT29 Cells , Humans , Immunoblotting , Immunohistochemistry , Mice , Mice, Inbred BALB C , Mice, Nude , Microscopy, Confocal , Molecular Sequence Data , Mutation , Nuclear Localization Signals/genetics , Proteolysis , RNA Interference , Transplantation, Heterologous
7.
Biochimie ; 94(12): 2590-9, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22902879

ABSTRACT

Legumain, an asparaginyl endopeptidase, is up-regulated in tumour and tumour-associated cells, and is linked to the processing of cathepsin B, L, and proMMP-2. Although legumain is mainly localized to the endosomal/lysosomal compartments, legumain has been reported to be localized extracellularly in the tumour microenvironment and associated with extracellular matrix and cell surfaces. The most potent endogenous inhibitor of legumain is cystatin E/M, which is a secreted protein synthesised with an export signal. Therefore, we investigated the cellular interplay between legumain and cystatin E/M. As a cell model, HEK293 cells were transfected with legumain cDNA, cystatin E/M cDNA, or both, and over-expressing monoclonal cell lines were selected (termed M38L, M4C, and M3CL, respectively). Secretion of prolegumain from M38L cells was inhibited by treatment with brefeldin A, whereas bafilomycin A1 enhanced the secretion. Cellular processing of prolegumain to the 46 and 36 kDa enzymatically active forms was reduced by treatment with either substance alone. M38L cells showed increased, but M4C cells decreased, cathepsin L processing suggesting a crucial involvement of legumain activity. Furthermore, we observed internalization of cystatin E/M and subsequently decreased intracellular legumain activity. Also, prolegumain was shown to internalize followed by increased intracellular legumain processing and activation. In addition, in M4C cells incomplete processing of the internalized prolegumain was observed, as well as nuclear localized cystatin E/M. Furthermore, auto-activation of secreted prolegumain was inhibited by cystatin E/M, which for the first time shows a regulatory role of cystatin E/M in controlling both intra- and extracellular legumain activity.


Subject(s)
Cystatin M/metabolism , Cysteine Endopeptidases/metabolism , Extracellular Space/metabolism , Intracellular Space/metabolism , Brefeldin A/pharmacology , Cathepsin L/metabolism , Cell Nucleus/metabolism , Cystatin M/genetics , Cysteine Endopeptidases/genetics , DNA, Complementary/genetics , Enzyme Inhibitors/pharmacology , Enzyme Precursors/genetics , Enzyme Precursors/metabolism , Extracellular Space/drug effects , HEK293 Cells , Humans , Immunoblotting , Intracellular Space/drug effects , Macrolides/pharmacology , Microscopy, Confocal , Protein Synthesis Inhibitors/pharmacology , Transfection
8.
Shock ; 24(3): 214-8, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16135959

ABSTRACT

Enhanced plasma levels of matrix metalloproteinase 9 (MMP-9) detected in patients with severe sepsis are thought to contribute to the development of organ dysfunction in endotoxemia. We have recently reported that peptidoglycan, the major wall component of gram-positive bacteria, increases MMP-9 levels in lung and liver and organ injury in the rat. Thus far, it is unclear whether MMP-9 is part of the septic response to peptidoglycan in human blood. The aim of the present study was to examine the regulation of MMP-9 by peptidoglycan in human leukocytes. The addition of peptidoglycan to whole human blood caused enhanced levels of MMP-9 after 1 h of incubation (306 vs. 75 ng/mL, P < or = 0.05) and onward, as measured by enzyme-linked immunoabsorbant assay. In neutrophil cultures, MMP-9 values increased significantly after 30 min of incubation with peptidoglycan (242 vs. 121 ng/mL, P < or = 0.05), whereas muramyl dipeptide had no effect. In contrast, adherent monocytes released insignificant amounts of MMP-9. To examine whether the released MMP-9 resulted from de novo synthesis, intracellular and secreted MMP-9 was measured during stimulation of neutrophils. The total MMP-9 values (the sum of intracellular and secreted MMP-9) before and after stimulation were mainly unaltered. The enhanced MMP-9 levels induced by peptidoglycan was attenuated by inhibitors of p38 mitogen-activated protein kinases (MAPK), (SB202190, 25 microM) and ERK1/2 (PD98059, 25 microM) and inhibitors of Src Tyrosine kinase (PP2, 5 microM) and PI3-K (LY294002, 25 microM).


Subject(s)
Gene Expression Regulation, Enzymologic/drug effects , Matrix Metalloproteinase 9/blood , Neutrophils/enzymology , Peptidoglycan/metabolism , Staphylococcus aureus/metabolism , Chromones/pharmacology , Dose-Response Relationship, Drug , Endotoxemia , Enzyme Inhibitors/pharmacology , Enzyme-Linked Immunosorbent Assay , Flavonoids/pharmacology , Humans , Imidazoles/pharmacology , Inflammation , Kinetics , Leukocytes/enzymology , Lipopolysaccharides/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Monocytes/metabolism , Morpholines/pharmacology , Neutrophils/metabolism , Peptidoglycan/chemistry , Pyridines/pharmacology , Pyrimidines/pharmacology , Time Factors , p38 Mitogen-Activated Protein Kinases/metabolism , src-Family Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...