Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Cancer Res Commun ; 3(2): 235-244, 2023 02.
Article in English | MEDLINE | ID: mdl-36968142

ABSTRACT

Disease recurrence and drug resistance are major challenges in the clinical management of patients with colorectal cancer liver metastases (CLM), and because tumors are generally microsatellite stable (MSS), responses to immune therapies are poor. The mesenchymal phenotype is overrepresented in treatment-resistant cancers and is associated with an immunosuppressed microenvironment. The aim of this work was to molecularly identify and characterize a mesenchymal subgroup of MSS CLM to identify novel therapeutic approaches. We here generated a mesenchymal gene expression signature by analysis of resection specimens from 38 patients with CLM using ranked expression level of the epithelial-to-mesenchymal transition-related transcription factor PRRX1. Downstream pathway analysis based on the resulting gene signature was performed and independent, publicly available datasets were used to validate the findings. A subgroup comprising 16% of the analyzed CLM samples were classified as mesenchymal, or belonging to the PRRX1 high group. Analysis of the PRRX1 signature genes revealed a distinct immunosuppressive phenotype with high expression of immune checkpoints HAVCR2/TIM-3 and VISTA, in addition to the M2 macrophage marker CD163. The findings were convincingly validated in datasets from three external CLM cohorts. Upregulation of immune checkpoints HAVCR2/TIM-3 and VISTA in the PRRX1 high subgroup is a novel finding, and suggests immune evasion beyond the PD-1/PD-L1 axis, which may contribute to poor response to PD-1/PD-L1-directed immune therapy in MSS colorectal cancer. Importantly, these checkpoints represent potential novel opportunities for immune-based therapy approaches in a subset of MSS CLM. Significance: CLM is an important cause of colorectal cancer mortality where the majority of patients have yet to benefit from immunotherapies. In this study of gene expression profiling analyses, we uncovered novel immune checkpoint targets in a subgroup of patients with MSS CLMs harboring a mesenchymal phenotype.


Subject(s)
Colorectal Neoplasms , Liver Neoplasms , Humans , B7-H1 Antigen/genetics , Hepatitis A Virus Cellular Receptor 2/genetics , Programmed Cell Death 1 Receptor/genetics , Neoplasm Recurrence, Local/genetics , Colorectal Neoplasms/genetics , Liver Neoplasms/genetics , Microsatellite Repeats , Tumor Microenvironment/genetics , Homeodomain Proteins/genetics
2.
Article in English | MEDLINE | ID: mdl-34036235

ABSTRACT

PURPOSE: Antiangiogenic therapy using bevacizumab has proven effective for a number of cancers; however, in breast cancer (BC), there is an unmet need to identify patients who benefit from such treatment. PATIENTS AND METHODS: In the NeoAva phase II clinical trial, patients (N = 132) with large (≥ 25 mm) human epidermal growth factor receptor 2 (HER2)-negative primary tumors were randomly assigned 1:1 to treatment with neoadjuvant chemotherapy (CTx) alone or in combination with bevacizumab (Bev plus CTx). The ratio of the tumor size after relative to before treatment was calculated into a continuous response scale. Tumor biopsies taken prior to neoadjuvant treatment were analyzed by reverse-phase protein arrays (RPPA) for expression levels of 210 BC-relevant (phospho-) proteins. Lasso regression was used to derive a predictor of tumor shrinkage from the expression of selected proteins prior to treatment. RESULTS: We identified a nine-protein signature score named vascular endothelial growth factor inhibition response predictor (ViRP) for use in the Bev plus CTx treatment arm able to predict with accuracy pathologic complete response (pCR) (area under the curve [AUC] = 0.85; 95% CI, 0.74 to 0.97) and low residual cancer burden (RCB 0/I) (AUC = 0.80; 95% CI, 0.68 to 0.93). The ViRP score was significantly lower in patients with pCR (P < .001) and in patients with low RCB (P < .001). The ViRP score was internally validated on mRNA data and the resultant surrogate mRNA ViRP score significantly separated the pCR patients (P = .016). Similarly, the mRNA ViRP score was validated (P < .001) in an independent phase II clinical trial (PROMIX). CONCLUSION: Our ViRP score, integrating the expression of nine proteins and validated on mRNA data both internally and in an independent clinical trial, may be used to increase the likelihood of benefit from treatment with bevacizumab combined with chemotherapy in patients with HER2-negative BC.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bevacizumab/therapeutic use , Breast Neoplasms/drug therapy , Neoadjuvant Therapy , Breast Neoplasms/chemistry , Female , Humans , Neoplasm Proteins , Predictive Value of Tests , Receptor, ErbB-2/analysis , Treatment Outcome
3.
Mol Cancer Ther ; 19(3): 895-905, 2020 03.
Article in English | MEDLINE | ID: mdl-31871265

ABSTRACT

Receptor tyrosine kinase AXL is found upregulated in various types of cancer, including melanoma, and correlates with an aggressive cancer phenotype, inducing cell proliferation and epithelial-to-mesenchymal transition. In addition, AXL has recently been linked to chemotherapy resistance, and inhibition of AXL is found to increase DNA damage and reduce expression of DNA repair proteins. In light of this, we aimed to investigate whether targeting AXL together with DNA damage response proteins would be therapeutically beneficial. Using melanoma cell lines, we observed that combined reduction of AXL and CHK1/CHK2 signaling decreased proliferation, deregulated cell-cycle progression, increased apoptosis, and reduced expression of DNA damage response proteins. Enhanced therapeutic effect of combined treatment, as compared with mono-treatment, was further observed in a patient-derived xenograft model and, of particular interest, when applying a three-dimensional ex vivo spheroid drug sensitivity assay on tumor cells harvested directly from 27 patients with melanoma lymph node metastases. Together, these results indicate that targeting AXL together with the DNA damage response pathway could be a promising treatment strategy in melanoma, and that further investigations in patient groups lacking treatment alternatives should be pursued.


Subject(s)
DNA Damage , DNA Repair , Drug Resistance, Neoplasm , Melanoma/pathology , Proto-Oncogene Proteins/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Thiophenes/pharmacology , Urea/analogs & derivatives , Animals , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Cycle Checkpoints , Cell Proliferation , Checkpoint Kinase 1/antagonists & inhibitors , Checkpoint Kinase 2/antagonists & inhibitors , Drug Therapy, Combination , Female , Gene Expression Regulation, Neoplastic , Humans , Lymphatic Metastasis , Melanoma/drug therapy , Melanoma/genetics , Mice , Mice, Nude , Neoplasm Invasiveness , Proto-Oncogene Proteins/genetics , RNA, Small Interfering/genetics , Receptor Protein-Tyrosine Kinases/genetics , Tumor Cells, Cultured , Urea/pharmacology , Xenograft Model Antitumor Assays , Axl Receptor Tyrosine Kinase
4.
Breast Cancer Res ; 21(1): 61, 2019 05 14.
Article in English | MEDLINE | ID: mdl-31088535

ABSTRACT

INTRODUCTION: Glutaminase inhibitors target cancer cells by blocking the conversion of glutamine to glutamate, thereby potentially interfering with anaplerosis and synthesis of amino acids and glutathione. The drug CB-839 has shown promising effects in preclinical experiments and is currently undergoing clinical trials in several human malignancies, including triple-negative breast cancer (TNBC). However, response to glutaminase inhibitors is variable and there is a need for identification of predictive response biomarkers. The aim of this study was to determine how glutamine is utilized in two patient-derived xenograft (PDX) models of breast cancer representing luminal-like/ER+ (MAS98.06) and basal-like/triple-negative (MAS98.12) breast cancer and to explore the metabolic effects of CB-839 treatment. EXPERIMENTAL: MAS98.06 and MAS98.12 PDX mice received CB-839 (200 mg/kg) or drug vehicle two times daily p.o. for up to 28 days (n = 5 per group), and the effect on tumor growth was evaluated. Expression of 60 genes and seven glutaminolysis key enzymes were determined using gene expression microarray analysis and immunohistochemistry (IHC), respectively, in untreated tumors. Uptake and conversion of glutamine were determined in the PDX models using HR MAS MRS after i.v. infusion of [5-13C] glutamine when the models had received CB-839 (200 mg/kg) or vehicle for 2 days (n = 5 per group). RESULTS: Tumor growth measurements showed that CB-839 significantly inhibited tumor growth in MAS98.06 tumors, but not in MAS98.12 tumors. Gene expression and IHC analysis indicated a higher proline synthesis from glutamine in untreated MAS98.06 tumors. This was confirmed by HR MAS MRS of untreated tumors demonstrating that MAS98.06 used glutamine to produce proline, glutamate, and alanine, and MAS98.12 to produce glutamate and lactate. In both models, treatment with CB-839 resulted in accumulation of glutamine. In addition, CB-839 caused depletion of alanine, proline, and glutamate ([1-13C] glutamate) in the MAS98.06 model. CONCLUSION: Our findings indicate that TNBCs may not be universally sensitive to glutaminase inhibitors. The major difference in the metabolic fate of glutamine between responding MAS98.06 xenografts and non-responding MAS98.12 xenografts is the utilization of glutamine for production of proline. We therefore suggest that addiction to proline synthesis from glutamine is associated with response to CB-839 in breast cancer. The effect of glutaminase inhibition in two breast cancer patient-derived xenograft (PDX) models. 13C HR MAS MRS analysis of tumor tissue from CB-839-treated and untreated models receiving 13C-labeled glutamine ([5-13C] Gln) shows that the glutaminase inhibitor CB-839 is causing an accumulation of glutamine (arrow up) in two PDX models representing luminal-like breast cancer (MAS98.06) and basal-like breast cancer (MAS98.12). In MAS98.06 tumors, CB-839 is in addition causing depletion of proline ([5-13C] Pro), alanine ([1-13C] Ala), and glutamate ([1-13C] Glu), which could explain why CB-839 causes tumor growth inhibition in MAS98.06 tumors, but not in MAS98.12 tumors.


Subject(s)
Breast Neoplasms/metabolism , Glutaminase/metabolism , Glutamine/metabolism , Proline/metabolism , Animals , Biomarkers , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Computational Biology , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Female , Gene Expression Profiling , Glutaminase/antagonists & inhibitors , Humans , Immunohistochemistry , Magnetic Resonance Spectroscopy , Metabolomics/methods , Mice , Models, Biological , Xenograft Model Antitumor Assays
5.
Nat Commun ; 10(1): 1600, 2019 04 08.
Article in English | MEDLINE | ID: mdl-30962452

ABSTRACT

In the preceding decades, molecular characterization has revolutionized breast cancer (BC) research and therapeutic approaches. Presented herein, an unbiased analysis of breast tumor proteomes, inclusive of 9995 proteins quantified across all tumors, for the first time recapitulates BC subtypes. Additionally, poor-prognosis basal-like and luminal B tumors are further subdivided by immune component infiltration, suggesting the current classification is incomplete. Proteome-based networks distinguish functional protein modules for breast tumor groups, with co-expression of EGFR and MET marking ductal carcinoma in situ regions of normal-like tumors and lending to a more accurate classification of this poorly defined subtype. Genes included within prognostic mRNA panels have significantly higher than average mRNA-protein correlations, and gene copy number alterations are dampened at the protein-level; underscoring the value of proteome quantification for prognostication and phenotypic classification. Furthermore, protein products mapping to non-coding genomic regions are identified; highlighting a potential new class of tumor-specific immunotherapeutic targets.


Subject(s)
Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/pathology , Protein Interaction Maps , Proteome/metabolism , Breast/pathology , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Carcinoma, Ductal, Breast/genetics , Carcinoma, Ductal, Breast/immunology , DNA Copy Number Variations , Datasets as Topic , Female , Gene Expression Profiling , Humans , Oligonucleotide Array Sequence Analysis , Proteogenomics/methods , Proteome/genetics , Proteome/immunology , RNA, Messenger/metabolism
6.
Mol Oncol ; 12(9): 1540-1558, 2018 09.
Article in English | MEDLINE | ID: mdl-29741811

ABSTRACT

The tumor microenvironment (TME) may influence both cancer progression and therapeutic response. In breast cancer, particularly in the aggressive triple-negative/basal-like subgroup, patient outcome is strongly associated with the tumor's inflammatory profile. Tumor-associated macrophages (TAMs) are among the most abundant immune cells in the TME, shown to be linked to poor prognosis and therapeutic resistance. In this study, we investigated the effect of the metastasis- and inflammation-associated microenvironmental factor S100A4 on breast cancer cells (BCCs) of different subtypes and explored their further interactions with myeloid cells. We demonstrated that extracellular S100A4 activates BCCs, particularly the basal-like subtype, to elevate secretion of pro-inflammatory cytokines. The secreted factors promoted conversion of monocytes to TAM-like cells that exhibited protumorigenic activities: stimulated epithelial-mesenchymal transition, proliferation, chemoresistance, and motility in cancer cells. In conclusion, we have shown that extracellular S100A4 instigates a tumor-supportive microenvironment, involving a network of cytokines and TAM-like cells, which was particularly characteristic for basal-like BCCs and potentiated their aggressive properties. The S100A4-BCC-TAM interaction cascade could be an important contributor to the aggressive behavior of this subtype and should be further explored for therapeutic targeting.


Subject(s)
Cytokines/metabolism , Macrophages/metabolism , Macrophages/pathology , S100 Calcium-Binding Protein A4/metabolism , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Tumor Microenvironment , Biopsy , Cell Line, Tumor , Cell Movement , Cell Proliferation , Drug Resistance, Neoplasm , Epithelial-Mesenchymal Transition , Female , Heterografts , Humans , Inflammation/metabolism , MCF-7 Cells , Monocytes/pathology , Spheroids, Cellular
7.
PLoS One ; 11(12): e0167754, 2016.
Article in English | MEDLINE | ID: mdl-28002446

ABSTRACT

BACKGROUND: The novel A/J Min/+ mouse, which is a model for human Familial Adenomatous Polyposis (FAP), develops spontaneously multiple adenocarcinomas in the colon as well as in the small intestine. Agaricus blazei Murill (AbM) is an edible Basidiomycetes mushroom that has been used in traditional medicine against cancer and other diseases. The mushroom contains immunomodulating ß-glucans and is shown to have antitumor effects in murine cancer models. Andosan™ is a water extract based on AbM (82%), but it also contains the medicinal Basidiomycetes mushrooms Hericeum erinaceus and Grifola frondosa. METHODS AND FINDINGS: Tap water with 10% Andosan™ was provided as the only drinking water for 15 or 22 weeks to A/J Min/+ mice and A/J wild-type mice (one single-nucleotide polymorphism (SNP) difference), which then were exsanguinated and their intestines preserved in formaldehyde and the serum frozen. The intestines were examined blindly by microscopy and also stained for the tumor-associated protease, legumain. Serum cytokines (pro- and anti-inflammatory, Th1-, Th2 -and Th17 type) were measured by Luminex multiplex analysis. Andosan™ treated A/J Min/+ mice had a significantly lower number of adenocarcinomas in the intestines, as well as a 60% significantly reduced intestinal tumor load (number of tumors x size) compared to control. There was also reduced legumain expression in intestines from Andosan™ treated animals. Moreover, Andosan™ had a significant cytotoxic effect correlating with apoptosis on the human cancer colon cell line, Caco-2, in vitro. When examining serum from both A/J Min/+ and wild type mice, there was a significant increase in anti-tumor Th1 type and pro-inflammatory cytokines in the Andosan™ treated mice. CONCLUSIONS: The results from this mouse model for colorectal cancer shows significant protection of orally administered Andosan™ against development of intestinal cancer. This is supported by the finding of less legumain in intestines of Andosan™ treated mice and increased systemic Th1 cytokine response. The mechanism is probably both immuno-modulatory and growth inhibition of tumor cells by induction of apoptosis.


Subject(s)
Agaricus/chemistry , Intestinal Mucosa/metabolism , Plant Extracts/chemistry , Protective Agents/chemistry , Agaricus/metabolism , Animals , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/isolation & purification , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Caco-2 Cells , Cysteine Endopeptidases/metabolism , Cytokines/blood , Enzyme-Linked Immunosorbent Assay , Female , Humans , Intestines/pathology , Male , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Plant Extracts/pharmacology , Protective Agents/pharmacology
8.
Oncotarget ; 7(15): 19997-20015, 2016 Apr 12.
Article in English | MEDLINE | ID: mdl-26918352

ABSTRACT

The knowledge on how tumor-associated stroma influences efficacy of anti-cancer therapy just started to emerge. Here we show that lung fibroblasts reduce melanoma sensitivity to the BRAF inhibitor (BRAFi) vemurafenib only if the two cell types are in close proximity. In the presence of fibroblasts, the adjacent melanoma cells acquire de-differentiated mesenchymal-like phenotype. Upon treatment with BRAFi, such melanoma cells maintain high levels of phospho ribosomal protein S6 (pS6), i.e. active mTOR signaling, which is suppressed in the BRAFi sensitive cells without stromal contacts. Inhibitors of PI3K/mTOR in combination with BRAFi eradicate pS6high cell subpopulations and potentiate anti-cancer effects in melanoma protected by the fibroblasts. mTOR and BRAF co-inhibition also delayed the development of early-stage lung metastases in vivo. In conclusion, we demonstrate that upon influence from fibroblasts, melanoma cells undergo a phenotype switch to the mesenchymal state, which can support PI3K/mTOR signaling. The lost sensitivity to BRAFi in such cells can be overcome by co-targeting PI3K/mTOR. This knowledge could be explored for designing BRAFi combination therapies aiming to eliminate both stroma-protected and non-protected counterparts of metastases.


Subject(s)
Fibroblasts/pathology , Melanoma/pathology , Mesoderm/pathology , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Animals , Apoptosis , Cell Proliferation , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Melanoma/drug therapy , Melanoma/metabolism , Mesoderm/drug effects , Mesoderm/metabolism , Mice, Nude , Signal Transduction/drug effects , Stromal Cells/drug effects , Stromal Cells/metabolism , Stromal Cells/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
9.
Biochimie ; 122: 208-18, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26343556

ABSTRACT

Prominent tasks of cysteine cathepsins involve endo-lysosomal proteolysis and turnover of extracellular matrix constituents or plasma membrane proteins for maintenance of intestinal homeostasis. Here we report on enhanced levels and altered subcellular localization of distinct cysteine cathepsins in adenocarcinoma tissue in comparison to adjacent normal colon. Immunofluorescence and immunoblotting investigations revealed the presence of cathepsin L in the nuclear compartment in addition to its expected endo-lysosomal localization in colorectal carcinoma cells. Cathepsin L was represented as the full-length protein in the nuclei of HCT116 cells from which stefin B, a potent cathepsin L inhibitor, was absent. Fluorescence activated cell sorting analyses with synchronized cell cultures revealed deceleration of cell cycle progression of HCT116 cells upon inhibition of cathepsin L activity, while expression of cathepsin L-enhanced green fluorescent protein chimeras accelerated S-phase entry. We conclude that the activity of cathepsin L is high in the nucleus of colorectal carcinoma cells because of lacking stefin B inhibitory activity. Furthermore, we hypothesize that nuclear cathepsin L accelerates cell cycle progression of HCT116 cells thereby supporting the notion that cysteine cathepsins may play significant roles in carcinogenesis due to deregulated trafficking.


Subject(s)
Cathepsin L/metabolism , Cell Cycle , Cell Nucleus/enzymology , Colorectal Neoplasms/enzymology , Endosomes/enzymology , Lysosomes/enzymology , Animals , Caco-2 Cells , Cell Line , Cell Line, Tumor , Colorectal Neoplasms/pathology , Epithelial Cells/enzymology , Fluorescent Antibody Technique , HCT116 Cells , Humans , Immunoblotting , Intestines/cytology , Intestines/enzymology , Microscopy, Confocal , S Phase
10.
Data Brief ; 5: 468-75, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26594658

ABSTRACT

The endo-lysosomal cysteine cathepsin L has recently been shown to have moonlighting activities in that its unexpected nuclear localization in colorectal carcinoma cells is involved in cell cycle progression (Tamhane et al., 2015) [1]. Here, we show data on the construction and sequence of a plasmid coding for human cathepsin L tagged with an enhanced green fluorescent protein (phCL-EGFP) in which the fluorescent protein is covalently attached to the C-terminus of the protease. The plasmid was used for transfection of HCT116 colorectal carcinoma cells, while data from non-transfected and pEGFP-N1-transfected cells is also shown. Immunoblotting data of lysates from non-transfected controls and HCT116 cells transfected with pEGFP-N1 and phCL-EGFP, showed stable expression of cathepsin L-enhanced green fluorescent protein chimeras, while endogenous cathepsin L protein amounts exceed those of hCL-EGFP chimeras. An effect of phCL-EGFP expression on proliferation and metabolic states of HCT116 cells at 24 h post-transfection was observed.

11.
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
12.
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
13.
Protoplasma ; 252(3): 755-74, 2015 May.
Article in English | MEDLINE | ID: mdl-25398648

ABSTRACT

Proteases play essential roles in protein degradation, protein processing, and extracellular matrix remodeling in all cell types and tissues. They are also involved in protein turnover for maintenance of homeostasis and protein activation or inactivation for cell signaling. Proteases range in function and specificity, with some performing distinct substrate cleavages, while others accomplish proteolysis of a wide range of substrates. As such, different cell types use specialized molecular mechanisms to regulate the localization of proteases and their function within the compartments to which they are destined. Here, we focus on the cysteine family of cathepsin proteases and legumain, which act predominately within the endo-lysosomal pathway. In particular, recent knowledge on cysteine cathepsins and their primary regulator legumain is scrutinized in terms of their trafficking to endo-lysosomal compartments and other less recognized cellular locations. We further explore the mechanisms that regulate these processes and point to pathological cases which arise from detours taken by these proteases. Moreover, the emerging biological roles of specific forms and variants of cysteine cathepsins and legumain are discussed. These may be decisive, pathogenic, or even deadly when localizing to unusual cellular compartments in their enzymatically active form, because they may exert unexpected effects by alternative substrate cleavage. Hence, we propose future perspectives for addressing the actions of cysteine cathepsins and legumain as well as their specific forms and variants. The increasing knowledge in non-canonical aspects of cysteine cathepsin- and legumain-mediated proteolysis may prove valuable for developing new strategies to utilize these versatile proteases in therapeutic approaches.


Subject(s)
Cathepsins/metabolism , Cell Biology , Cysteine Endopeptidases/metabolism , Proteolysis , Animals , Humans , Lysosomes/metabolism , Models, Biological
14.
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
15.
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
17.
BMC Cancer ; 10: 17, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-20074384

ABSTRACT

BACKGROUND: High activity of cysteine proteases such as legumain and the cathepsins have been shown to facilitate growth and invasion of a variety of tumor types. In breast cancer, several recent studies have indicated that loss of the cysteine protease inhibitor cystatin E/M leads to increased growth and metastasis. Although cystatin E/M is normally expressed in the skin, its role in cysteine protease regulation and progression of malignant melanoma has not been studied. METHODS: A panel of various non-melanoma and melanoma cell lines was used. Cystatin E/M and C were analyzed in cell media by immunoblotting and ELISA. Legumain, cathepsin B and L were analyzed in cell lysates by immunoblotting and their enzymatic activities were analyzed by peptide substrates. Two melanoma cell lines lacking detectable secretion of cystatin E/M were transfected with a cystatin E/M expression plasmid (pCST6), and migration and invasiveness were studied by a Matrigel invasion assay. RESULTS: Cystatin E/M was undetectable in media from all established melanoma cell lines examined, whereas strong immunobands were detected in two of five primary melanoma lines and in two of six lines derived from patients with metastatic disease. Among the four melanoma lines secreting cystatin E/M, the glycosylated form (17 kD) was predominant compared to the non-glycosylated form (14 kD). Legumain, cathepsin B and L were expressed and active in most of the cell lines, although at low levels in the melanomas expressing cystatin E/M. In the melanoma lines where cystatin E/M was secreted, cystatin C was generally absent or expressed at a very low level. When melanoma cells lacking secretion of cystatin E/M were transfected with pCST6, their intracellular legumain activity was significantly inhibited. In contrast, cathepsin B activity was not affected. Furthermore, invasion was suppressed in cystatin E/M over-expressing melanoma cell lines as measured by the transwell Matrigel assay. CONCLUSIONS: These results suggest that the level of cystatin E/M regulates legumain activity and hence the invasive potential of human melanoma cells.


Subject(s)
Cystatin M/biosynthesis , Cysteine Endopeptidases/biosynthesis , Gene Expression Regulation, Neoplastic , Melanoma/metabolism , Skin Neoplasms/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Collagen/chemistry , Culture Media, Serum-Free/metabolism , Cysteine Proteases/metabolism , Drug Combinations , Humans , Laminin/chemistry , Neoplasm Invasiveness , Neoplasm Metastasis , Proteoglycans/chemistry
18.
BMC Cancer ; 8: 172, 2008 Jun 13.
Article in English | MEDLINE | ID: mdl-18554396

ABSTRACT

BACKGROUND: S100A4 is a metastasis-associated protein which has been linked to multiple cellular events, and has been identified extracellularly, in the cytoplasm and in the nucleus of tumor cells; however, the biological implications of subcellular location are unknown. Associations between a variety of posttranslational protein modifications and altered biological functions of proteins are becoming increasingly evident. Identification and characterization of posttranslationally modified S100A4 variants could thus contribute to elucidating the mechanisms for the many cellular functions that have been reported for this protein, and might eventually lead to the identification of novel drugable targets. METHODS: S100A4 was immuoprecipitated from a panel of in vitro and in vivo sources using a monoclonal antibody and the samples were separated by 2D-PAGE. Gels were analyzed by western blot and silver staining, and subsequently, several of the observed spots were identified as S100A4 by the use of MALDI-TOF and MALDI-TOF/TOF. RESULTS: A characteristic pattern of spots was observed when S100A4 was separated by 2D-PAGE suggesting the presence of at least three charge variants. These charge variants were verified as S100A4 both by western immunoblotting and mass spectrometry, and almost identical patterns were observed in samples from different tissues and subcellular compartments. Interestingly, recombinant S100A4 displayed a similar pattern on 2D-PAGE, but with different quantitative distribution between the observed spots. CONCLUSION: Endogenously expressed S100A4 were shown to exist in several charge variants, which indicates the presence of posttranslational modifications altering the net charge of the protein. The different variants were present in all subcellular compartments and tissues/cell lines examined, suggesting that the described charge variants is a universal phenomenon, and cannot explain the localization of S100A4 in different subcellular compartments. However, the identity of the specific posttranslational modification and its potential contribution to the many reported biological events induced by S100A4, are subject to further studies.


Subject(s)
Colorectal Neoplasms/chemistry , Colorectal Neoplasms/metabolism , Protein Processing, Post-Translational , S100 Proteins/isolation & purification , S100 Proteins/metabolism , Amino Acid Sequence , Blotting, Western , Colorectal Neoplasms/pathology , Electrophoresis, Gel, Two-Dimensional , HCT116 Cells , Humans , Immunoprecipitation , Molecular Sequence Data , Neoplasm Metastasis , Protein Isoforms , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , S100 Calcium-Binding Protein A4 , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
SELECTION OF CITATIONS
SEARCH DETAIL
...