Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Bull Math Biol ; 86(3): 30, 2024 02 12.
Article in English | MEDLINE | ID: mdl-38347328

ABSTRACT

One of the most crucial and lethal characteristics of solid tumors is represented by the increased ability of cancer cells to migrate and invade other organs during the so-called metastatic spread. This is allowed thanks to the production of matrix metalloproteinases (MMPs), enzymes capable of degrading a type of collagen abundant in the basal membrane separating the epithelial tissue from the connective one. In this work, we employ a synergistic experimental and mathematical modelling approach to explore the invasion process of tumor cells. A mathematical model composed of reaction-diffusion equations describing the evolution of the tumor cells density on a gelatin substrate, MMPs enzymes concentration and the degradation of the gelatin is proposed. This is completed with a calibration strategy. We perform a sensitivity analysis and explore a parameter estimation technique both on synthetic and experimental data in order to find the optimal parameters that describe the in vitro experiments. A comparison between numerical and experimental solutions ends the work.


Subject(s)
Podosomes , Humans , Podosomes/metabolism , Podosomes/pathology , Gelatin/metabolism , Extracellular Matrix/pathology , Models, Biological , Mathematical Concepts , Matrix Metalloproteinases/metabolism , Neoplasm Invasiveness/pathology
2.
J R Soc Interface ; 21(210): 20230587, 2024 01.
Article in English | MEDLINE | ID: mdl-38196375

ABSTRACT

Glucose is a primary energy source for cancer cells. Several lines of evidence support the idea that monocarboxylate transporters, such as MCT1, elicit metabolic reprogramming of cancer cells in glucose-poor environments, allowing them to re-use lactate, a by-product of glucose metabolism, as an alternative energy source with serious consequences for disease progression. We employ a synergistic experimental and mathematical modelling approach to explore the evolutionary processes at the root of cancer cell adaptation to glucose deprivation, with particular focus on the mechanisms underlying the increase in MCT1 expression observed in glucose-deprived aggressive cancer cells. Data from in vitro experiments on breast cancer cells are used to inform and calibrate a mathematical model that comprises a partial integro-differential equation for the dynamics of a population of cancer cells structured by the level of MCT1 expression. Analytical and numerical results of this model suggest that environment-induced changes in MCT1 expression mediated by lactate-associated signalling pathways enable a prompt adaptive response of glucose-deprived cancer cells, while fluctuations in MCT1 expression due to epigenetic changes create the substrate for environmental selection to act upon, speeding up the selective sweep underlying cancer cell adaptation to glucose deprivation, and may constitute a long-term bet-hedging mechanism.


Subject(s)
Aggression , Neoplasms , Humans , Biological Evolution , Disease Progression , Glucose , Lactic Acid
3.
Food Chem Toxicol ; 185: 114454, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38237855

ABSTRACT

Evidence suggests that meat processing and heat treatment may increase cancer risk through exposure to potentially carcinogenic compounds, polycyclic aromatic hydrocarbons (PAHs), and heterocyclic aromatic amines (HAAs). This study aims to investigate the effect of low concentrations of PAHs and HAAs (from 1 to 100 µmol/L/24h and 48h) in colorectal tumor cells (HT-29, HCT116, and LS174T) and to evaluate the effect of PAHs in the presence of inulin in mice. In vitro, the 4-PAHs have no effect on healthy colon cells but decreased the viability of the colorectal tumor cells and activated the mRNA and protein expressions of CYP1A1 and CYP1B1. In vivo, in mice with colitis induced by 3% DSS, the 4-PAHs (equimolar mix at 50,100, 150 mg/kg.bw, orally 3 times a week for 3 weeks) induced a loss of body weight and tumor formation. Inulin (10 g/L) had no effect on colon length and tumor formation. A significant decrease in the loss of b.w was observed in inulin group as compared to the fiber free group. These results underscore the importance of considering the biological association between low-dose exposure to 4-HAPs and diet-related colon tumors.


Subject(s)
Colorectal Neoplasms , Heterocyclic Compounds , Polycyclic Aromatic Hydrocarbons , Animals , Mice , Inulin/pharmacology , Amines/toxicity , Polycyclic Aromatic Hydrocarbons/toxicity , Dietary Supplements , Heterocyclic Compounds/toxicity
4.
Clin. transl. oncol. (Print) ; 25(5): 1389-1401, mayo 2023.
Article in English | IBECS | ID: ibc-219522

ABSTRACT

Background Adipose tissue is a major component of breast stroma. This study focused on delineating the effects of adipose stem cells (ASCs) derived from breast of healthy women and cancer patients with normal or tumor breast cells. Methods The ASCs were induced to differentiate into adipocytes, and the subsequent adipocyte conditioned media (ACM) were evaluated for their fatty acid profile, adipokine secretion and influence on proliferation, migration and invasion on tumoral (MCF-7 and SUM159) and normal (HMEC) human breast cell lines. Results An enrichment of arachidonic acid was observed in ACM from tumor tissues. Adipose tissues from tumor free secrete twice as much leptin than those from proximal or distal to the tumor. All ACMs display proliferative activity and favor invasiveness of SUM159 cells compared to MCF-7 and HMEC. All ACMs induced lipid droplets accumulation in MCF-7 cells and increased CD36 expression in tumor cells. Conclusion We conclude that among secreted factors analyzed, only arachidonic acid and leptin levels did discriminate ASCs from tumor-bearing and tumor-free breasts emphasizing the importance that other cell types could contribute to the adipose tissue secretome in a tumor context (AU)


Subject(s)
Humans , Female , Breast Neoplasms/pathology , Leptin/metabolism , Adipocytes/metabolism , Adipocytes/pathology , Adipose Tissue/metabolism , Adipose Tissue/pathology , Arachidonic Acid/pharmacology , Cell Line, Tumor , Cell Proliferation , MCF-7 Cells
5.
Clin Transl Oncol ; 25(5): 1389-1401, 2023 May.
Article in English | MEDLINE | ID: mdl-36520383

ABSTRACT

BACKGROUND: Adipose tissue is a major component of breast stroma. This study focused on delineating the effects of adipose stem cells (ASCs) derived from breast of healthy women and cancer patients with normal or tumor breast cells. METHODS: The ASCs were induced to differentiate into adipocytes, and the subsequent adipocyte conditioned media (ACM) were evaluated for their fatty acid profile, adipokine secretion and influence on proliferation, migration and invasion on tumoral (MCF-7 and SUM159) and normal (HMEC) human breast cell lines. RESULTS: An enrichment of arachidonic acid was observed in ACM from tumor tissues. Adipose tissues from tumor free secrete twice as much leptin than those from proximal or distal to the tumor. All ACMs display proliferative activity and favor invasiveness of SUM159 cells compared to MCF-7 and HMEC. All ACMs induced lipid droplets accumulation in MCF-7 cells and increased CD36 expression in tumor cells. CONCLUSION: We conclude that among secreted factors analyzed, only arachidonic acid and leptin levels did discriminate ASCs from tumor-bearing and tumor-free breasts emphasizing the importance that other cell types could contribute to the adipose tissue secretome in a tumor context.


Subject(s)
Breast Neoplasms , Leptin , Female , Humans , Leptin/metabolism , Leptin/pharmacology , Arachidonic Acid/metabolism , Arachidonic Acid/pharmacology , Breast Neoplasms/pathology , Secretome , Adipocytes/metabolism , Adipocytes/pathology , Adipose Tissue/metabolism , Adipose Tissue/pathology , MCF-7 Cells , Cell Proliferation , Culture Media, Conditioned/pharmacology , Cell Line, Tumor
6.
Int J Mol Sci ; 23(24)2022 Dec 18.
Article in English | MEDLINE | ID: mdl-36555812

ABSTRACT

Poly (ADP-ribose) polymerase inhibitors (PARPi) are targeted therapies that inhibit PARP proteins which are involved in a variety of cell functions. PARPi may act as modulators of angiogenesis; however, the relationship between PARPi and the vasculogenic mimicry (VM) in breast cancer remains unclear. To determine whether PARPi regulate the vascular channel formation, we assessed whether the treatment with olaparib, talazoparib and veliparib inhibits the vascular channel formation by breast cancer cell lines. Here, we found that PARPi act as potent inhibitors of the VM formation in triple negative breast cancer cells, independently of the BRCA status. Mechanistically, we find that PARPi trigger and inhibit the NF-κB signaling, leading to the inhibition of the VM. We further show that PARPi decrease the expression of the angiogenic factor PTX3. Moreover, PTX3 rescued the PARPi-inhibited VM inhibition. In conclusion, our results indicate that PARPi, by targeting the VM, may provide a new therapeutic approach for triple negative breast cancer.


Subject(s)
Antineoplastic Agents , Triple Negative Breast Neoplasms , Humans , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , NF-kappa B , Triple Negative Breast Neoplasms/drug therapy , Antineoplastic Agents/therapeutic use , Cell Line, Tumor
7.
Cancers (Basel) ; 14(6)2022 Mar 14.
Article in English | MEDLINE | ID: mdl-35326638

ABSTRACT

Vasculogenic mimicry (VM) formed by aggressive tumor cells to create vascular networks connected with the endothelial cells, plays an important role in breast cancer progression. WISP2 has been considered as a tumor suppressor protein; however, the relationship between WISP2 and VM formation remains unclear. We used the in vitro tube formation assay and in vivo immunohistochemical analysis in a mouse model, and human breast tumors were used to evaluate the effect of WISP2 on VM formation. Here we report that WISP2 acts as a potent inhibitor of VM formation in breast cancer. Enforced expression of WISP2 decreased network formation while knockdown of WISP2 increased VM. Mechanistically, WISP2 increased retention of oncogenic activators YAP/TAZ in cytoplasm, leading to decreased expression of the angiogenic factor CYR61. Studies using an in vivo mouse model and human breast tumors confirmed the in vitro cell lines data. In conclusion, our results indicate that WISP2 may play a critical role in VM and highlight the critical role of WISP2 as a tumor suppressor.

8.
Leukemia ; 36(2): 540-548, 2022 02.
Article in English | MEDLINE | ID: mdl-34556797

ABSTRACT

Myeloma is characterized by bone lesions, which are related to both an increased osteoclast activity and a defect in the differentiation of medullary mesenchymal stem cells (MSCs) into osteoblasts. Outside the medullary environment, adipocyte-derived MSCs (ASCs) could represent a source of functional osteoblasts. However, we recently found a defect in the osteoblastic differentiation of ASCs from myeloma patients (MM-ASCs). We examined the effects of plasma from myeloma patients at diagnosis (MM-plasmas) and in complete remission (CR-plasmas) and from healthy donors on the osteoblastic differentiation of healthy donor-derived ASCs (HD-ASCs). Osteoblastogenesis in HD-ASCs was suppressed by MM-plasmas. Seven cytokines (ANG1, ENA-78, EGF, PDGF-AA/AB/BB, and TARC) were increased in MM-plasmas and separately inhibited the osteoblastic differentiation of HD-ASCs. Comparison of MM-ASCs and HD-ASCs by RNA sequencing showed that two master genes characterizing adipocyte differentiation, CD36 and PPARγ, were upregulated in MM-ASCs as compared to HD-ASCs. Finally, we demonstrated a significant increase in CD36 and PPARγ expression in HD-ASCs in the presence of MM-plasmas or the seven cytokines individually, similarly as in MM-ASCs. We conclude that specific cytokines in MM-plasmas, besides the well-known DKK1, inhibit the osteoblastic differentiation of MM- and HD-ASCs with a skewing towards adipocyte differentiation.


Subject(s)
Adipocytes/cytology , Cell Differentiation , Cytokines/pharmacology , Mesenchymal Stem Cells/cytology , Multiple Myeloma/metabolism , Osteoblasts/cytology , Stem Cells/cytology , Adipocytes/drug effects , Adipocytes/metabolism , Case-Control Studies , Cells, Cultured , Healthy Volunteers , Humans , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Multiple Myeloma/pathology , Osteoblasts/drug effects , Osteoblasts/metabolism , Stem Cells/drug effects , Stem Cells/metabolism
9.
Antioxidants (Basel) ; 10(5)2021 May 04.
Article in English | MEDLINE | ID: mdl-34064498

ABSTRACT

Specific inhibition of NADPH oxidases (NOX) and NO-synthases (NOS), two enzymes associated with redox stress in tumor cells, has aroused great pharmacological interest. Here, we show how these enzymes distinguish between isomeric 2'- and 3'-phosphate derivatives, a difference used to improve the specificity of inhibition by isolated 2'- and 3'-phosphate isomers of our NADPH analogue NS1. Both isomers become fluorescent upon binding to their target proteins as observed by in vitro assay and in vivo imaging. The 2'-phosphate isomer of NS1 exerted more pronounced effects on NOS and NOX-dependent physiological responses than the 3'-phosphate isomer did. Docking and molecular dynamics simulations explain this specificity at the level of the NADPH site of NOX and NOS, where conserved arginine residues distinguished between the 2'-phosphate over the 3'-phosphate group, in favor of the 2'-phosphate.

10.
Mar Drugs ; 18(4)2020 Apr 03.
Article in English | MEDLINE | ID: mdl-32260204

ABSTRACT

High-throughput screening assays have been designed to identify compounds capable of inhibiting phenotypes involved in cancer aggressiveness. However, most studies used commercially available chemical libraries. This prompted us to explore natural products isolated from marine-derived fungi as a new source of molecules. In this study, we established a chemical library from 99 strains corresponding to 45 molecular operational taxonomic units and evaluated their anticancer activity against the MCF7 epithelial cancer cell line and its invasive stem cell-like MCF7-Sh-WISP2 counterpart. We identified the marine fungal Paradendryphiella salina PC 362H strain, isolated from the brown alga Pelvetia caniculata (PC), as one of the most promising fungi which produce active compounds. Further chemical and biological characterizations of the culture of the Paradendryphiella salina PC 362H strain identified (-)-hyalodendrin as the active secondary metabolite responsible for the cytotoxic activity of the crude extract. The antitumor activity of (-)-hyalodendrin was not only limited to the MCF7 cell lines, but also prominent on cancer cells with invasive phenotypes including colorectal cancer cells resistant to chemotherapy. Further investigations showed that treatment of MCF7-Sh-WISP2 cells with (-)-hyalodendrin induced changes in the phosphorylation status of p53 and altered expression of HSP60, HSP70 and PRAS40 proteins. Altogether, our study reveals that this uninvestigated marine fungal crude extract possesses a strong therapeutic potential against tumor cells with aggressive phenotypes and confirms that members of the epidithiodioxopiperazines are interesting fungal toxins with anticancer activities.


Subject(s)
Antineoplastic Agents/pharmacology , Ascomycota/chemistry , Cell Survival/drug effects , Fungi/chemistry , Mycotoxins/pharmacology , Piperazines/pharmacology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Biological Products/pharmacology , Cell Line , Humans , MCF-7 Cells , Mice , Neoplasms/drug therapy , Signal Transduction/drug effects , Thioredoxin-Disulfide Reductase , Thioredoxins , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism
11.
Cancers (Basel) ; 11(12)2019 Dec 13.
Article in English | MEDLINE | ID: mdl-31847105

ABSTRACT

Breast adiposity is correlated with body mass index, menopausal status and mammary density. We here wish to establish how these factors influence the cross-talk between breast adipocytes and normal or malignant breast cells. Adipocyte-derived stem cells (ASCs) were obtained from healthy women and classified into six distinct groups based on body mass index, menopausal status and mammary density. The ASCs were induced to differentiate, and the influence of their conditioned media (ACM) was determined. Unexpectedly, there were no detectable differences in adipogenic differentiation and secretion between the six ASC groups, while their corresponding ACMs had no detectable influence on normal breast cells. In clear contrast, all ACMs profoundly influenced the proliferation, migration and invasiveness of malignant breast cells and increased the number of lipid droplets in their cytoplasm via increased expression of the fatty acid receptor CD36, thereby increasing fatty acid uptake. Importantly, inhibition of CD36 reduced lipid droplet accumulation and attenuated the migration and invasion of the breast cancer cells. These findings suggest that breast-associated adipocytes potentiate the invasiveness of breast cancer cells which, at least in part, is mediated by metabolic reprogramming via CD36-mediated fatty acid uptake.

12.
J Theor Biol ; 479: 73-80, 2019 10 21.
Article in English | MEDLINE | ID: mdl-31283914

ABSTRACT

Three-dimensional cultures of cells are gaining popularity as an in vitro improvement over 2D Petri dishes. In many such experiments, cells have been found to organize in aggregates. We present new results of three-dimensional in vitro cultures of breast cancer cells exhibiting patterns. Understanding their formation is of particular interest in the context of cancer since metastases have been shown to be created by cells moving in clusters. In this paper, we propose that the main mechanism which leads to the emergence of patterns is chemotaxis, i.e., oriented movement of cells towards high concentration zones of a signal emitted by the cells themselves. Studying a Keller-Segel PDE system to model chemotactical auto-organization of cells, we prove that it admits Turing unstable solutions under a time-dependent condition. This result is illustrated by two-dimensional simulations of the model showing spheroidal patterns. They are qualitatively compared to the biological results and their variability is discussed both theoretically and numerically.


Subject(s)
Breast Neoplasms/pathology , Chemotaxis/physiology , Spheroids, Cellular/metabolism , Cell Culture Techniques , Computer Simulation , Humans , Models, Biological , Neoplasm Metastasis/pathology
14.
Clin Cancer Res ; 24(11): 2548-2558, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29490990

ABSTRACT

Purpose: There is extensive cross-talk between VEGF- and EGFR-pathway signaling in colorectal cancer. However, combinations of VEGF- and EGFR-targeted monoclonal antibodies (mAb) show disappointing activity, in particular for patients with mutant RAS Previous results show that tyrosine kinase inhibitors (TKI) can be active in colorectal cancer models resistant to mAbs. This prompted us to examine whether the activity of bevacizumab can be increased by combination with erlotinib.Experimental Design: The antitumor activity of bevacizumab, erlotinib, and their combination was determined in colorectal cancer models with different RAS status and bevacizumab sensitivity. EGFR/VEGF pathway activation was characterized by immunohistochemistry, Western blot, and ELISA assays. The influence of cetuximab and erlotinib on EGF-mediated migration and the EGFR-EGF ligand feedback loop was established in colorectal cancer cell lines with different RAS status.Results: The addition of erlotinib increased bevacizumab activity in all models independent of RAS status. Bevacizumab exposure was accompanied by marked EGFR activation in tumor cells as well as in tumor-associated endothelial cells (TECs) and resulted in strong accumulation of intracellular EGFR, which could be attenuated by erlotinib. In cellular models, erlotinib was able to attenuate EGF-mediated functions in all cell lines independent of RAS status while cetuximab only showed activity in RAS wild-type cells.Conclusions: These results should provide a molecular framework to better understand the increased activity of the bevacizumab-erlotinib combination, compared with bevacizumab alone, in the GERCOR DREAM phase III clinical trial. Differential activity of mAbs and TKIs targeting the same signaling pathway is likely applicable for other tumor types. Clin Cancer Res; 24(11); 2548-58. ©2018 AACR.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , ras Proteins/genetics , Animals , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Bevacizumab/administration & dosage , Cell Line, Tumor , Colorectal Neoplasms/pathology , Disease Models, Animal , Drug Synergism , Erlotinib Hydrochloride/administration & dosage , Humans , Mice , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Prognosis , Proto-Oncogene Proteins B-raf/genetics , Signal Transduction , Treatment Outcome , Xenograft Model Antitumor Assays , ras Proteins/metabolism
15.
Sci Rep ; 7(1): 1749, 2017 05 11.
Article in English | MEDLINE | ID: mdl-28496206

ABSTRACT

WISP1 (Wnt1-inducible signaling pathway protein-1, also known as CCN4) is a member of the CCN family able to mediate cell growth, transformation and survival in a tissue-specific manner. Here, we report that WISP1 expression was highly increased in preadipocytes and decreased during adipocyte differentiation. Moreover, we observed an increase in WISP1 gene expression in adipose tissue from both diet-induced and leptin-deficient ob/ob obese mice, suggesting that WISP1 could be involved in the pathophysiological onset of obesity. Interestingly, overexpression of WISP1 in 3T3-F442A cells prevented adipocyte differentiation via downregulation of peroxisome proliferator-activated receptor (PPARγ) transcriptional activity thereby attenuating the expression of adipogenic markers. Conversely, silencing of WISP1 enhanced adipocyte differentiation. We further show that the inactivation of PPARγ transcriptional activity was mediated, at least in part, by a direct physical association between WISP1 and PPARγ, followed by proteasome-dependent degradation of PPARγ. These results suggest for the first time that WISP1 interacts with PPARγ and that this interaction results in the inhibition of PPARγ activity. Taken together our results suggest that WISP1 functions as a negative regulator of adipogenesis.


Subject(s)
Adipocytes/cytology , Adipocytes/metabolism , CCN Intercellular Signaling Proteins/metabolism , Cell Differentiation , PPAR gamma/metabolism , Proto-Oncogene Proteins/metabolism , 3T3-L1 Cells , Adipogenesis/genetics , Adipose Tissue/metabolism , Animals , CCN Intercellular Signaling Proteins/genetics , Cell Differentiation/genetics , Down-Regulation/genetics , Fibroblasts/metabolism , Gene Knockdown Techniques , Gene Silencing , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Proto-Oncogene Proteins/genetics , Transcription, Genetic , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation/genetics , Wnt Signaling Pathway
16.
J Biol Chem ; 290(34): 21007-21018, 2015 Aug 21.
Article in English | MEDLINE | ID: mdl-26152726

ABSTRACT

Although E3 ubiquitin ligases are deemed to play key roles in normal cell function and homeostasis, whether their alterations contribute to cancer pathogenesis remains unclear. In this study, we sought to investigate potential mechanisms that govern WWP1/Tiul1 (WWP1) ubiquitin ligase activity, focusing on its ability to trigger degradation of TGFß type I receptor (TßRI) in conjunction with Smad7. Our data reveal that the WWP1 protein is very stable at steady states because its autopolyubiquitination activity is silenced due to an intra-interaction between the C2 and/or WW and Hect domains that favors WWP1 monoubiquitination at the expense of its polyubiquitination or polyubiquitination of TßRI. Upon binding of WWP1 to Smad7, this functional interplay is disabled, switching its monoubiquitination activity toward a polyubiquitination activity, thereby driving its own degradation and that of TßRI as well. Intriguingly, a WWP1 point mutation found in human prostate cancer disrupts this regulatory mechanism by relieving the inhibitory effects of C2 and WW on Hect and thereby causing WWP1 hyperactivation. That cancer-driven alteration of WWP1 culminates in excessive TßRI degradation and attenuated TGFß cytostatic signaling, a consequence that could conceivably confer tumorigenic properties to WWP1.


Subject(s)
Carcinogenesis/metabolism , Gene Expression Regulation, Neoplastic , Point Mutation , Protein Serine-Threonine Kinases/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1/metabolism , Ubiquitin-Protein Ligases/metabolism , Carcinogenesis/genetics , Carcinogenesis/pathology , HEK293 Cells , HeLa Cells , Humans , MCF-7 Cells , Protein Binding , Protein Interaction Domains and Motifs , Protein Serine-Threonine Kinases/genetics , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/genetics , Signal Transduction , Smad7 Protein/genetics , Smad7 Protein/metabolism , Transforming Growth Factor beta1/genetics , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/genetics , Ubiquitination
17.
Oncotarget ; 6(21): 18518-33, 2015 Jul 30.
Article in English | MEDLINE | ID: mdl-26041882

ABSTRACT

Most solid tumors contain a subfraction of cells with stem/progenitor cell features. Stem cells are naturally chemoresistant suggesting that chronic chemotherapeutic stress may select for cells with increased "stemness". We carried out a comprehensive molecular and functional analysis of six independently selected colorectal cancer (CRC) cell lines with acquired resistance to three different chemotherapeutic agents derived from two distinct parental cell lines. Chronic drug exposure resulted in complex alterations of stem cell markers that could be classified into three categories: 1) one cell line, HT-29/5-FU, showed increased "stemness" and WNT-signaling, 2) three cell lines showed decreased expression of stem cell markers, decreased aldehyde dehydrogenase activity, attenuated WNT-signaling and lost the capacity to form colonospheres and 3) two cell lines displayed prominent expression of ABC transporters with a heterogeneous response for stem cell markers. While WNT-signaling could be attenuated in the HT-29/5-FU cells by the WNT-signaling inhibitors ICG-001 and PKF-118, this was not accompanied by any selective growth inhibitory effect suggesting that the cytotoxic activity of these compounds is not directly linked to WNT-signaling inhibition. We conclude that classical WNT-signaling inhibitors have toxic off-target activities that need to be addressed for clinical development.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/drug effects , Neoplastic Stem Cells/drug effects , Wnt Signaling Pathway/drug effects , Blotting, Western , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/genetics , Fluorouracil/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , HT29 Cells , Humans , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , Irinotecan , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Organoplatinum Compounds/pharmacology , Oxaliplatin , Protein Isoforms/genetics , Protein Isoforms/metabolism , Pyrimidinones/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Triazines/pharmacology , Wnt Signaling Pathway/genetics
18.
PLoS One ; 9(2): e87878, 2014.
Article in English | MEDLINE | ID: mdl-24498388

ABSTRACT

It has been proposed that the epithelial-mesenchymal transition (EMT) in mammary epithelial cells and breast cancer cells generates stem cell features. WISP2 (Wnt-1-induced signaling protein-2) plays an important role in maintenance of the differentiated phenotype of estrogen receptor-positive breast cancer cells and loss of WISP2 is associated with EMT. We now report that loss of WISP2 in MCF7 breast cancer cells can also promote the emergence of a cancer stem-like cell phenotype characterized by high expression of CD44, increased aldehyde dehydrogenase activity and mammosphere formation. Higher levels of the stem cell markers Nanog and Oct3/4 were observed in those mammospheres. In addition we show that low-cell inoculums are capable of tumor formation in the mammary fat pad of immunodeficient mice. Gene expression analysis show an enrichment of markers linked to stem cell function such as SOX9 and IGFBP7 which is linked to TGF-ß inducible, SMAD3-dependent transcription. Taken together, our data demonstrate that WISP2 loss promotes both EMT and the stem-like cell phenotype.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/pathology , CCN Intercellular Signaling Proteins/metabolism , Epithelial-Mesenchymal Transition , Estrogens/pharmacology , Neoplastic Stem Cells/pathology , Repressor Proteins/metabolism , Animals , Biomarkers, Tumor/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , CCN Intercellular Signaling Proteins/genetics , Cells, Cultured , Female , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , Mice , Mice, Nude , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Phenotype , Repressor Proteins/genetics , Signal Transduction , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
19.
Cell Rep ; 4(3): 530-41, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23911286

ABSTRACT

The homeodomain protein TGIF (TG-interacting factor) restricts TGF-ß/Smad cytostatic signaling by interfering with the nucleocytoplasmic transit of the tumor suppressor cPML. Here, we identify PHRF1 as a ubiquitin ligase that enforces TGIF decay by driving its ubiquitination at lysine 130. In so doing, PHRF1 ensures redistribution of cPML into the cytoplasm, where it associates with SARA and coordinates activation of Smad2 by the TGF-ß receptor. The PHRF1 gene resides within the tumor suppressor locus 11p15.5, which displays frequent loss in a wide variety of malignancies, including breast cancer. Remarkably, we found that the PHRF1 gene is deleted or silenced in a high proportion of human breast cancer samples and cancer cell lines. Reconstitution of PHRF1 into deficient cells impeded their propensity to form tumors in vivo, most likely because of the reemergence of TGF-ß responsiveness. These findings unveil a paradigm behind inactivation of the cPML tumor suppressor network in human malignancies.


Subject(s)
Breast Neoplasms/genetics , Interferon Regulatory Factor-7/genetics , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism , Tumor Suppressor Proteins/genetics , Animals , Breast Neoplasms/metabolism , Dogs , Female , Genes, Tumor Suppressor , Hep G2 Cells , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Interferon Regulatory Factor-7/metabolism , Madin Darby Canine Kidney Cells , Nuclear Proteins/genetics , Phosphorylation , Promyelocytic Leukemia Protein , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction , Transcription Factors/genetics , Transcription, Genetic , Transfection , Transforming Growth Factor beta/genetics , Tumor Suppressor Proteins/metabolism , Ubiquitination
20.
Biochem J ; 447(1): 71-9, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22765757

ABSTRACT

CCN5 (cysteine-rich 61/connective tissue growth factor/nephroblastoma overexpressed 5)/WISP-2 [WNT1 (wingless-type MMTV integration site family, member 1)-inducible signalling pathway protein 2] is an oestrogen-regulated member of the CCN family. CCN5 is a transcriptional repressor of genes associated with the EMT (epithelial-mesenchymal transition) and plays an important role in maintenance of the differentiated phenotype in ER (oestrogen receptor)-positive breast cancer cells. In contrast, CCN5 is undetectable in more aggressive ER-negative breast cancer cells. We now report that CCN5 is induced in ER-negative breast cancer cells such as MDA-MB-231 following glucocorticoid exposure, due to interaction of the endogenous glucocorticoid receptor with a functional glucocorticoid-response element in the CCN5 gene promoter. Glucocorticoid treatment of MDA-MB-231 cells is accompanied by morphological alterations, decreased invasiveness and attenuated expression of mesenchymal markers, including vimentin, cadherin 11 and ZEB1 (zinc finger E-box binding homeobox 1). Interestingly, glucocorticoid exposure did not increase CCN5 expression in ER-positive breast cancer cells, but rather down-regulated ER expression, thereby attenuating oestrogen pathway signalling. Taken together, our results indicate that glucocorticoid treatment of ER-negative breast cancer cells induces high levels of CCN5 expression and is accompanied by the appearance of a more differentiated and less invasive epithelial phenotype. These findings propose a novel therapeutic strategy for high-risk breast cancer patients.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , CCN Intercellular Signaling Proteins/metabolism , Dexamethasone/pharmacology , Estrogen Receptor alpha/metabolism , Glucocorticoids/pharmacology , Repressor Proteins/metabolism , Base Sequence , Breast Neoplasms/genetics , Breast Neoplasms/pathology , CCN Intercellular Signaling Proteins/genetics , Cell Line, Tumor , DNA Primers/genetics , Estradiol/pharmacology , Estrogen Receptor alpha/genetics , Female , Gene Expression/drug effects , Humans , Neoplasm Invasiveness/pathology , Neoplasm Invasiveness/prevention & control , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Repressor Proteins/genetics , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...