Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Mol Life Sci ; 78(2): 733-755, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32385587

ABSTRACT

Previous clinical and experimental evidence strongly supports a breast cancer-promoting function of the lysosomal protease cathepsin B. However, the cathepsin B-dependent molecular pathways are not completely understood. Here, we studied the cathepsin-mediated secretome changes in the context of the MMTV-PyMT breast cancer mouse model. Employing the cell-conditioned media from tumor-macrophage co-cultures, as well as tumor interstitial fluid obtained by a novel strategy from PyMT mice with differential cathepsin B expression, we identified an important proteolytic and lysosomal signature, highlighting the importance of this organelle and these enzymes in the tumor micro-environment. The Cellular Repressor of E1A Stimulated Genes 1 (CREG1), a secreted endolysosomal glycoprotein, displayed reduced abundance upon over-expression of cathepsin B as well as increased abundance upon cathepsin B deletion or inhibition. Moreover, it was cleaved by cathepsin B in vitro. CREG1 reportedly could act as tumor suppressor. We show that treatment of PyMT tumor cells with recombinant CREG1 reduced proliferation, migration, and invasion; whereas, the opposite was observed with reduced CREG1 expression. This was further validated in vivo by orthotopic transplantation. Our study highlights CREG1 as a key player in tumor-stroma interaction and suggests that cathepsin B sustains malignant cell behavior by reducing the levels of the growth suppressor CREG1 in the tumor microenvironment.


Subject(s)
Breast Neoplasms/pathology , Cathepsin B/metabolism , Neoplasm Invasiveness/pathology , Repressor Proteins/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cathepsin B/genetics , Cell Proliferation , Female , Gene Deletion , Gene Expression Regulation, Neoplastic , Mice , Neoplasm Invasiveness/genetics , Repressor Proteins/genetics , Tumor Cells, Cultured , Tumor Microenvironment , Up-Regulation
2.
Clin Proteomics ; 15: 11, 2018.
Article in English | MEDLINE | ID: mdl-29527141

ABSTRACT

BACKGROUND: Proteomic analyses of clinical specimens often rely on human tissues preserved through formalin-fixation and paraffin embedding (FFPE). Minimal sample consumption is the key to preserve the integrity of pathological archives but also to deal with minimal invasive core biopsies. This has been achieved by using the acid-labile surfactant RapiGest in combination with a direct trypsinization (DTR) strategy. A critical comparison of the DTR protocol with the most commonly used filter aided sample preparation (FASP) protocol is lacking. Furthermore, it is unknown how common histological stainings influence the outcome of the DTR protocol. METHODS: Four single consecutive murine kidney tissue specimens were prepared with the DTR approach or with the FASP protocol using both 10 and 30 k filter devices and analyzed by label-free, quantitative liquid chromatography-tandem mass spectrometry (LC-MS/MS). We compared the different protocols in terms of proteome coverage, relative label-free quantitation, missed cleavages, physicochemical properties and gene ontology term annotations of the proteins. Additionally, we probed compatibility of the DTR protocol for the analysis of common used histological stainings, namely hematoxylin & eosin (H&E), hematoxylin and hemalaun. These were proteomically compared to an unstained control by analyzing four human tonsil FFPE tissue specimens per condition. RESULTS: On average, the DTR protocol identified 1841 ± 22 proteins in a single, non-fractionated LC-MS/MS analysis, whereas these numbers were 1857 ± 120 and 1970 ± 28 proteins for the FASP 10 and 30 k protocol. The DTR protocol showed 15% more missed cleavages, which did not adversely affect quantitation and intersample comparability. Hematoxylin or hemalaun staining did not adversely impact the performance of the DTR protocol. A minor perturbation was observed for H&E staining, decreasing overall protein identification by 13%. CONCLUSIONS: In essence, the DTR protocol can keep up with the FASP protocol in terms of qualitative and quantitative reproducibility and performed almost as well in terms of proteome coverage and missed cleavages. We highlight the suitability of the DTR protocol as a viable and straightforward alternative to the FASP protocol for proteomics-based clinical research.

3.
J Cancer ; 8(19): 4065-4074, 2017.
Article in English | MEDLINE | ID: mdl-29187882

ABSTRACT

Studies in the MMTV-PyMT (PyMT) breast cancer mouse model have shown a strong influence of the lysosomal cysteine cathepsins B or L on lung metastasis formation. Transgenic expression of human CTSB (tgCTSB) or CTSL (tgCTSL) both led to similar metastatic phenotypes with increased metastatic burden in the PyMT mice. However, recent studies in other tumor models proved marked differences in effects of either cathepsin on the proteome composition. We sought to analyze and compare proteome changes in the metastatic proteome of PyMT mice expressing either tgCTSB or tgCTSL to evaluate similarities and differences in those models. Performing an explorative, quantitative proteome comparison based on LC-MS/MS, we identified up to 3,000 proteins from murine lung metastases in three independent biological replicates per genotype. In both cases, when compared to wild-type (WT) mice, we noticed a pronounced impact of transgene cathepsin expression on the metastasis proteome. Highlights include increased moesin, integrin beta 1 and vinexin levels in the tgCTSB dataset and increased saposin and granulin levels in the tgCTSL dataset. Importantly, non-supervised hierarchical clustering clearly separated tgCTSB vs. tgCTSL induced proteome changes. In summary, tgCTSB and tgCTSL both display a strong and distinct impact on proteome composition of lung macrometastases in the PyMT model. Our observations suggest that they impact malignant behavior in distinct ways, thus further emphasizing interest into their tumor-contextual functionality.

4.
Cell Oncol (Dordr) ; 40(6): 639-650, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28808957

ABSTRACT

BACKGROUND: Solid tumors contain various components that together form the tumor microenvironment. Cancer associated fibroblasts (CAFs) are capable of secreting and responding to signaling molecules and growth factors. Due to their role in tumor development, CAFs are considered as potential therapeutic targets. A prominent tumor-associated signaling molecule is transforming growth factor ß (TGFß), an inducer of epithelial-to-mesenchymal transition (EMT). The differential action of TGFß on CAFs and ETCs (epithelial tumor cells) has recently gained interest. Here, we aimed to investigate the effects of TGFß on CAFs and ETCs at the proteomic level. METHODS: We established a 2D co-culture system of differentially fluorescently labeled CAFs and ETCs and stimulated this co-culture system with TGFß. The respective cell types were separated using FACS and subjected to quantitative analyses of individual proteomes using mass spectrometry. RESULTS: We found that TGFß treatment had a strong impact on the proteome composition of CAFs, whereas ETCs responded only marginally to TGFß. Quantitative proteomic analyses of the different cell types revealed up-regulation of extracellular matrix (ECM) proteins in TGFß treated CAFs. In addition, we found that the TGFß treated CAFs exhibited increased N-cadherin levels. CONCLUSIONS: From our data we conclude that CAFs respond to TGFß treatment by changing their proteome composition, while ETCs appear to be rather resilient.


Subject(s)
Cancer-Associated Fibroblasts/cytology , Cancer-Associated Fibroblasts/drug effects , Coculture Techniques/methods , Transforming Growth Factor beta/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Epithelial-Mesenchymal Transition/drug effects , HCT116 Cells , Humans , Proteomics , Tumor Microenvironment
5.
Biochim Biophys Acta ; 1863(11): 2700-2709, 2016 11.
Article in English | MEDLINE | ID: mdl-27526672

ABSTRACT

Cathepsin B (CTSB) is a lysosomal endo- and exopeptidase that is also secreted in high amounts by malignant and non-malignant cells. We determined the effect of CTSB on the tumor cell secretome by shRNA-mediated silencing of CTSB mRNA expression and subsequent proteomic LC-MS/MS analysis of the cell supernatants. We identified significant protein changes of 17 secreted or shed proteins. Notably, we found a general reduction in protein abundance of ADAM10 substrates and lysosomal proteins. We corroborated reduced amounts of soluble ADAM10 (sADAM10) and soluble APP (sAPP) in the two cancer cell lines MDA-MB-231 and U2OS by immunoblotting. Interestingly, reductions in sADAM10 and sAPP could be reversed by re-introducing a catalytically inactive variant of CTSB, suggesting a formerly unknown non-catalytic function of the protease.


Subject(s)
Cathepsin B/metabolism , Neoplasms/enzymology , RNA Interference , ADAM10 Protein/genetics , ADAM10 Protein/metabolism , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Blotting, Western , Cathepsin B/genetics , Cell Line, Tumor , Chromatography, Liquid , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mutation , Neoplasms/genetics , Neoplasms/pathology , Proteomics/methods , Signal Transduction , Tandem Mass Spectrometry , Time Factors , Transfection
6.
Biochimie ; 122: 88-98, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26455267

ABSTRACT

Carcinomas establish a molecular cross talk between malignant tumor cells and the activated non-malignant cells of the tumor stroma. This cell-cell communication in tumor-stroma interaction includes soluble, secreted proteins that act in a paracrine or autocrine manner. Proteases are crucial factors in tumor-stroma interaction by degrading or truncating secreted bioactive proteins. The cysteine protease cathepsin B is frequently overexpressed in several cancer types, including breast cancer. Its abundance often correlates with poor prognosis. In the murine polyoma virus middle T oncogene (PyMT) breast cancer model, cathepsin B is equally pro-tumorigenic. In this study, we investigate how cathepsin B shapes the secreted proteome of PyMT breast cancers. We employed a novel strategy to harvest tumor interstitial fluid (IF) in combination with chemical stable isotope tagging for quantitative proteomic comparison of IF stemming from PyMT tumors from wild-type mice, mice lacking cathepsin B, and mice over-expressing human cathepsin B. In three biological replicates, we achieve good proteome coverage (∼1700 proteins), with a large content (>70%) of secreted proteins. This characterizes IF as a robust source for the investigation of cancer secretomes. We also identified a large number of shed ectodomains, thus highlighting the importance of tumor-contextual cell surface proteolysis. Furthermore, IF contained >190 proteases and protease inhibitors, which span the entire range of absolute protein abundances; an observation testifying for an important role of proteolysis in tumor-stroma interaction. The cathepsin B genotype consistently affected proteins including alpha-1B-glycoprotein and major urinary proteins 11 and 8 (MUP8). Our study establishes tumor IF as a rich source for the investigation of secreted proteins in tumor biology and sheds light on complex proteolytic networks in the breast cancer secretome.


Subject(s)
Cathepsin B/metabolism , Extracellular Fluid/metabolism , Mammary Neoplasms, Experimental/metabolism , Proteome/metabolism , Proteomics/methods , Animals , Antigens, Polyomavirus Transforming/genetics , Blotting, Western , Chromatography, Liquid , Mammary Neoplasms, Experimental/genetics , Mice, Knockout , Mice, Transgenic , Peptide Hydrolases/metabolism , Polyomavirus/genetics , Polyomavirus/immunology , Protease Inhibitors/metabolism , Proteins/metabolism , Tandem Mass Spectrometry
7.
Oncotarget ; 6(27): 23874-89, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26203557

ABSTRACT

Expression of miR-200c is a molecular switch to determine cellular fate towards a mesenchymal or epithelial phenotype. miR-200c suppresses the early steps of tumor progression by preventing epithelial-mesenchymal transition (EMT) and intravasation of tumor cells. Unraveling the underlying molecular mechanisms might pinpoint to novel therapeutic options. To better understand these mechanisms it is crucial to identify targets of miR-200c. Here, we employ a combination of quantitative proteomic and bioinformatic strategies to identify novel miR-200c targets. We identify and confirm two subunits of the central cellular kinase protein kinase A (PKA), namely PRKAR1A and PRKACB, to be directly regulated by miR-200c. Notably, siRNA-mediated downregulation of both proteins phenocopies the migratory behavior of breast cancer cells after miR-200c overexpression. Patient data from publicly accessible databases supports a miR-200c-PKA axis. Thus, our study identifies the PKA heteroprotein as an important mediator of miR-200c induced repression of migration in breast cancer cells. By bioinformatics, we define a miRNA target cluster consisting of PRKAR1A, PRKAR2B, PRKACB, and COF2, which is targeted by a group of 14 miRNAs.


Subject(s)
Breast Neoplasms/pathology , Cell Movement/genetics , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/genetics , Cyclic AMP-Dependent Protein Kinase RIalpha Subunit/genetics , Epithelial-Mesenchymal Transition/genetics , MicroRNAs/genetics , Breast Neoplasms/genetics , Cell Line, Tumor , Chromatography, Liquid , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/biosynthesis , Cyclic AMP-Dependent Protein Kinase RIalpha Subunit/biosynthesis , Female , Gene Expression Regulation, Neoplastic , Humans , Lim Kinases/genetics , RNA Interference , RNA, Small Interfering/genetics , Tandem Mass Spectrometry
SELECTION OF CITATIONS
SEARCH DETAIL
...