Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Cell Rep Med ; 4(4): 101002, 2023 04 18.
Article in English | MEDLINE | ID: mdl-37044095

ABSTRACT

A genome-wide PiggyBac transposon-mediated screen and a resistance screen in a PIK3CAH1047R-mutated murine tumor model reveal NF1 loss in mammary tumors resistant to the phosphatidylinositol 3-kinase α (PI3Kα)-selective inhibitor alpelisib. Depletion of NF1 in PIK3CAH1047R breast cancer cell lines and a patient-derived organoid model shows that NF1 loss reduces sensitivity to PI3Kα inhibition and correlates with enhanced glycolysis and lower levels of reactive oxygen species (ROS). Unexpectedly, the antioxidant N-acetylcysteine (NAC) sensitizes NF1 knockout cells to PI3Kα inhibition and reverts their glycolytic phenotype. Global phospho-proteomics indicates that combination with NAC enhances the inhibitory effect of alpelisib on mTOR signaling. In public datasets of human breast cancer, we find that NF1 is frequently mutated and that such mutations are enriched in metastases, an indication for which use of PI3Kα inhibitors has been approved. Our results raise the attractive possibility of combining PI3Kα inhibition with NAC supplementation, especially in patients with drug-resistant metastases associated with NF1 loss.


Subject(s)
Breast Neoplasms , Humans , Mice , Animals , Female , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Phosphatidylinositol 3-Kinase , Acetylcysteine/pharmacology , Class I Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/genetics
2.
Commun Biol ; 5(1): 1144, 2022 10 28.
Article in English | MEDLINE | ID: mdl-36307545

ABSTRACT

Biobanking of surplus human healthy and disease-derived tissues is essential for diagnostics and translational research. An enormous amount of formalin-fixed and paraffin-embedded (FFPE), Tissue-Tek OCT embedded or snap-frozen tissues are preserved in many biobanks worldwide and have been the basis of translational studies. However, their usage is limited to assays that do not require viable cells. The access to intact and viable human material is a prerequisite for translational validation of basic research, for novel therapeutic target discovery, and functional testing. Here we show that surplus tissues from multiple solid human cancers directly slow-frozen after resection can subsequently be used for different types of methods including the establishment of 2D, 3D, and ex vivo cultures as well as single-cell RNA sequencing with similar results when compared to freshly analyzed material.


Subject(s)
Formaldehyde , Neoplasms , Humans , Paraffin Embedding , Biological Specimen Banks , Exome Sequencing
3.
Oncogene ; 41(39): 4459-4473, 2022 09.
Article in English | MEDLINE | ID: mdl-36008466

ABSTRACT

Plasticity delineates cancer subtypes with more or less favourable outcomes. In breast cancer, the subtype triple-negative lacks expression of major differentiation markers, e.g., estrogen receptor α (ERα), and its high cellular plasticity results in greater aggressiveness and poorer prognosis than other subtypes. Whether plasticity itself represents a potential vulnerability of cancer cells is not clear. However, we show here that cancer cell plasticity can be exploited to differentiate triple-negative breast cancer (TNBC). Using a high-throughput imaging-based reporter drug screen with 9 501 compounds, we have identified three polo-like kinase 1 (PLK1) inhibitors as major inducers of ERα protein expression and downstream activity in TNBC cells. PLK1 inhibition upregulates a cell differentiation program characterized by increased DNA damage, mitotic arrest, and ultimately cell death. Furthermore, cells surviving PLK1 inhibition have decreased tumorigenic potential, and targeting PLK1 in already established tumours reduces tumour growth both in cell line- and patient-derived xenograft models. In addition, the upregulation of genes upon PLK1 inhibition correlates with their expression in normal breast tissue and with better overall survival in breast cancer patients. Our results indicate that differentiation therapy based on PLK1 inhibition is a potential alternative strategy to treat TNBC.


Subject(s)
Triple Negative Breast Neoplasms , Breast/pathology , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation , Estrogen Receptor alpha , Humans , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism
4.
J Cell Sci ; 134(21)2021 11 01.
Article in English | MEDLINE | ID: mdl-34633031

ABSTRACT

The vast majority of breast cancer-associated deaths are due to metastatic spread of cancer cells, a process aided by epithelial-to-mesenchymal transition (EMT). Mounting evidence has indicated that long non-coding RNAs (lncRNAs) also contribute to tumor progression. We report the identification of 114 novel lncRNAs that change their expression during TGFß-induced EMT in murine breast cancer cells (referred to as EMT-associated transcripts; ETs). Of these, the ET-20 gene localizes in antisense orientation within the tenascin C (Tnc) gene locus. TNC is an extracellular matrix protein that is critical for EMT and metastasis formation. Both ET-20 and Tnc are regulated by the EMT master transcription factor Sox4. Notably, ablation of ET-20 lncRNA effectively blocks Tnc expression and with it EMT. Mechanistically, ET-20 interacts with desmosomal proteins, thereby impairing epithelial desmosomes and promoting EMT. A short transcript variant of ET-20 is shown to be upregulated in invasive human breast cancer cell lines, where it also promotes EMT. Targeting ET-20 appears to be a therapeutically attractive lead to restrain EMT and breast cancer metastasis in addition to its potential utility as a biomarker for invasive breast cancer.


Subject(s)
Breast Neoplasms , RNA, Long Noncoding , Animals , Breast Neoplasms/genetics , Cell Line, Tumor , Desmosomes/genetics , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , Neoplasm Invasiveness/genetics , RNA, Long Noncoding/genetics , SOXC Transcription Factors
5.
Oncogene ; 39(24): 4728-4740, 2020 06.
Article in English | MEDLINE | ID: mdl-32404986

ABSTRACT

An epithelial to mesenchymal transition (EMT) is an embryonic dedifferentiation program which is aberrantly activated in cancer cells to acquire cellular plasticity. This plasticity increases the ability of breast cancer cells to invade into surrounding tissue, to seed metastasis at distant sites and to resist to chemotherapy. In this study, we have observed a higher expression of interferon-related factors in basal-like and claudin-low subtypes of breast cancer in patients, known to be associated with EMT. Notably, Irf1 exerts essential functions during the EMT process, yet it is also required for the maintenance of an epithelial differentiation status of mammary gland epithelial cells: RNAi-mediated ablation of Irf1 in mammary epithelial cells results in the expression of mesenchymal factors and Smad transcriptional activity. Conversely, ablation of Irf1 during TGFß-induced EMT prevents a mesenchymal transition and stabilizes the expression of E-cadherin. In the basal-like murine breast cancer cell line 4T1, RNAi-mediated ablation of Irf1 reduces colony formation and cell migration in vitro and shedding of circulating tumor cells and metastasis formation in vivo. This context-dependent dual role of Irf1 in the regulation of epithelial-mesenchymal plasticity provides important new insights into the functional contribution and therapeutic potential of interferon-regulated factors in breast cancer.


Subject(s)
Epithelial-Mesenchymal Transition , Interferon Regulatory Factor-1/biosynthesis , Mammary Glands, Animal/metabolism , Mammary Neoplasms, Experimental/metabolism , Neoplasm Proteins/biosynthesis , Animals , Cell Line, Tumor , Female , Interferon Regulatory Factor-1/genetics , Mammary Glands, Animal/pathology , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Neoplasm Metastasis , Neoplasm Proteins/genetics
6.
BMC Cancer ; 19(1): 312, 2019 Apr 04.
Article in English | MEDLINE | ID: mdl-30947697

ABSTRACT

BACKGROUND: The tumour microenvironment is a critical regulator of malignant cancer progression. While endothelial cells have been widely studied in the context of tumour angiogenesis, their role as modulators of cancer cell invasion and migration is poorly understood. METHODS: We have investigated the influence of endothelial cells on the invasive and migratory behaviour of human cancer cells in vitro. RESULTS: Upon exposure to culture supernatants of endothelial cells, distinct cancer cells, such as SK-BR-3 cells, showed significantly increased invasion and cell migration concomitant with changes in cell morphology and gene expression reminiscent of an epithelial-mesenchymal transition (EMT). Interestingly, the pro-migratory effect on SK-BR-3 cells was significantly enhanced by supernatants obtained from subconfluent, proliferative endothelial cells rather than from confluent, quiescent endothelial cells. Systematically comparing the supernatants of subconfluent and confluent endothelial cells by quantitative MS proteomics revealed eight candidate proteins that were secreted at significantly higher levels by confluent endothelial cells representing potential inhibitors of cancer cell migration. Among these proteins, nidogen-1 was exclusively expressed in confluent endothelial cells and was found to be necessary and sufficient for the inhibition of SK-BR-3 cell migration. Indeed, SK-BR-3 cells exposed to nidogen-1-depleted endothelial supernatants showed increased promigratory STAT3 phosphorylation along with increased cell migration. This reflects the situation of enhanced SK-BR-3 migration upon stimulation with conditioned medium from subconfluent endothelial cells with inherent absence of nidogen-1 expression. CONCLUSION: The identification of nidogen-1 as an endothelial-derived inhibitor of migration of distinct cancer cell types reveals a novel mechanism of endothelial control over cancer progression.


Subject(s)
Breast Neoplasms/pathology , Endothelial Cells/metabolism , Membrane Glycoproteins/metabolism , Tumor Microenvironment , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , Epithelial-Mesenchymal Transition/physiology , Female , Gene Expression Regulation, Neoplastic , Human Umbilical Vein Endothelial Cells , Humans , Membrane Glycoproteins/genetics , Neoplasm Invasiveness/pathology , Phosphorylation , Primary Cell Culture , RNA, Small Interfering/metabolism , STAT3 Transcription Factor/metabolism
7.
Dev Cell ; 48(4): 539-553.e6, 2019 02 25.
Article in English | MEDLINE | ID: mdl-30713070

ABSTRACT

Epithelial-mesenchymal transition (EMT) enables cells to gain migratory and invasive features underlined by major transcriptional and epigenetic reprogramming. However, most studies have focused on the endpoints of the EMT process, and the epistatic hierarchy of the transcriptional networks underlying EMT has remained elusive. We have used a siRNA-based, functional high-content microscopy screen to identify 46 (co)transcription factors ((co)TFs) and 13 miRNAs critically required for EMT in normal murine mammary gland (NMuMG) cells. We compared dynamic gene expression during EMT kinetics and used functional perturbation of critical (co)TFs and miRNAs to identify groups and networks of EMT genes. Computational analysis as well as functional validation experiments revealed interaction networks between TFs and miRNAs and delineated the hierarchical and functional interactions of multiple EMT regulatory networks in NMuMG cells.


Subject(s)
Cell Movement/genetics , Epithelial-Mesenchymal Transition/genetics , MicroRNAs/genetics , Transcription Factors/metabolism , Animals , Cells, Cultured , Epithelial-Mesenchymal Transition/physiology , Gene Regulatory Networks/genetics , Humans , Mice
8.
Cancer Cell ; 35(1): 17-32.e6, 2019 01 14.
Article in English | MEDLINE | ID: mdl-30645973

ABSTRACT

Cancer cell plasticity facilitates the development of therapy resistance and malignant progression. De-differentiation processes, such as an epithelial-mesenchymal transition (EMT), are known to enhance cellular plasticity. Here, we demonstrate that cancer cell plasticity can be exploited therapeutically by forcing the trans-differentiation of EMT-derived breast cancer cells into post-mitotic and functional adipocytes. Delineation of the molecular pathways underlying such trans-differentiation has motivated a combination therapy with MEK inhibitors and the anti-diabetic drug Rosiglitazone in various mouse models of murine and human breast cancer in vivo. This combination therapy provokes the conversion of invasive and disseminating cancer cells into post-mitotic adipocytes leading to the repression of primary tumor invasion and metastasis formation.


Subject(s)
Adipocytes/cytology , Breast Neoplasms/drug therapy , Cell Transdifferentiation/drug effects , Flavonoids/administration & dosage , Neoplasm Metastasis/drug therapy , Rosiglitazone/administration & dosage , 3T3-L1 Cells , Adipogenesis , Animals , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Movement , Epithelial-Mesenchymal Transition/drug effects , Female , Flavonoids/pharmacology , Humans , Mice , Neoplasm Transplantation , Proto-Oncogene Proteins c-met/metabolism , Rosiglitazone/therapeutic use , Signal Transduction/drug effects , Transforming Growth Factor beta/metabolism
9.
Oncogene ; 37(31): 4197-4213, 2018 08.
Article in English | MEDLINE | ID: mdl-29713055

ABSTRACT

An epithelial to mesenchymal transition (EMT) has been correlated to malignant tumor progression and metastasis by promoting cancer cell migration and invasion and chemoresistance. Hence, finding druggable EMT effectors is critical to efficiently interfere with metastasis formation and to overcome therapy resistance. We have employed a high-content microscopy screen in combination with a kinome and phosphatome-wide siRNA library to identify signaling pathways underlying an EMT of murine mammary epithelial cells and breast cancer cells. This screen identified the MEK5-ERK5 axis as a critical player in TGFß-mediated EMT. Suppression of MEK5-ERK5 signaling completely prevented the morphological and molecular changes occurring during a TGFß-induced EMT and, conversely, forced highly metastatic breast cancer cells into a differentiated epithelial state. Inhibition of MEK5-ERK5 signaling also repressed breast cancer cell migration and invasion and substantially reduced lung metastasis without affecting primary tumor growth. The results suggest that the MEK5-ERK5 signaling axis via activation of MEF2B and other transcription factors plays an important role in the induction and maintenance of breast cancer cell migration and invasion and thus represents an exploitable target for the pharmacological inhibition of cancer cell metastasis.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Epithelial-Mesenchymal Transition/physiology , MAP Kinase Kinase 5/metabolism , Mitogen-Activated Protein Kinase 7/metabolism , Neoplasm Metastasis/pathology , RNA, Small Interfering/metabolism , Animals , Cell Line , Cell Line, Tumor , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Gene Expression Regulation, Neoplastic/physiology , Humans , Mice , Signal Transduction/physiology , Transforming Growth Factor beta/metabolism
10.
Nat Commun ; 8(1): 1168, 2017 10 27.
Article in English | MEDLINE | ID: mdl-29079737

ABSTRACT

Epithelial tumour cells can gain invasive and metastatic capabilities by undergoing an epithelial-mesenchymal transition. Transcriptional regulators and post-transcriptional effectors like microRNAs orchestrate this process of high cellular plasticity and its malignant consequences. Here, using microRNA sequencing in a time-resolved manner and functional validation, we have identified microRNAs that are critical for the regulation of an epithelial-mesenchymal transition and of mesenchymal tumour cell migration. We report that miR-1199-5p is downregulated in its expression during an epithelial-mesenchymal transition, while its forced expression prevents an epithelial-mesenchymal transition, tumour cell migration and invasion in vitro, and lung metastasis in vivo. Mechanistically, miR-1199-5p acts in a reciprocal double-negative feedback loop with the epithelial-mesenchymal transition transcription factor Zeb1. This function resembles the activities of miR-200 family members, guardians of an epithelial cell phenotype. However, miR-1199-5p and miR-200 family members share only six target genes, indicating that, besides regulating Zeb1 expression, they exert distinct functions during an epithelial-mesenchymal transition.


Subject(s)
Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Mammary Neoplasms, Animal/metabolism , MicroRNAs/metabolism , Neoplasm Metastasis , Zinc Finger E-box-Binding Homeobox 1/metabolism , Animals , Cell Line, Tumor , Cell Movement , Down-Regulation , Female , Gene Expression Profiling , Humans , Mammary Neoplasms, Animal/genetics , Mice , MicroRNAs/genetics , Phenotype , Transforming Growth Factor beta/metabolism , Zinc Finger E-box-Binding Homeobox 1/genetics
11.
Cell Stress ; 1(3): 139-140, 2017 Dec 01.
Article in English | MEDLINE | ID: mdl-31225444

ABSTRACT

Epithelial to mesenchymal transition (EMT) as well as its reversal process, mesenchymal to epithelial transition (MET), are essential and well-controlled cellular processes during embryonic development. Tightly controlled regulatory mechanisms guide an EMT/MET plasticity and enable cells to switch forth and back between different cell morphologies and functional capabilities to endow the necessity of tissue plasticity. However, aberrant and uncontrolled activation of these processes during malignant tumor progression promotes primary tumor cell invasion, cancer cell dissemination and metastatic outgrowth. In a recent study (Nat Commun; doi: 10.1038/s41467-017-01197-w), we have reported on the post-transcriptional control of normal and cancer-associated EMT by miRNAs and identified a novel, critical double-negative feedback regulation of the thus far unknown miRNA miR1199 and the key EMT transcription factor Zeb1.

12.
Curr Opin Cell Biol ; 43: 7-13, 2016 12.
Article in English | MEDLINE | ID: mdl-27371787

ABSTRACT

An epithelial to mesenchymal transition (EMT) is a process of cell remodeling critical during embryonic development and organogenesis. During an EMT, epithelial cells lose their polarized organization and acquire migratory and invasive capabilities. While a plethora of experimental results have indicated that manipulating an EMT also affects cancer metastasis, its reverse process, a mesenchymal to epithelial transition (MET), seems to support metastatic outgrowth in distant organs. Moreover, recent reports investigating cancer cells circulating in the blood stream or employing genetic lineage-tracing have questioned a critical role of an EMT in metastasis formation. Hence, we need to better understand the molecular networks underlying the cell plasticity conferred by an EMT or a MET and its functional contribution to malignant tumor progression.


Subject(s)
Epithelial-Mesenchymal Transition , Neoplasm Metastasis/pathology , Animals , Disease Progression , Epithelial-Mesenchymal Transition/genetics , Gene Regulatory Networks , Humans , Models, Biological , Neoplasm Metastasis/genetics , Neoplasms/genetics , Neoplasms/pathology
13.
J Cell Sci ; 127(Pt 7): 1523-36, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24554433

ABSTRACT

The cellular changes during an epithelial-mesenchymal transition (EMT) largely rely on global changes in gene expression orchestrated by transcription factors. Tead transcription factors and their transcriptional co-activators Yap and Taz have been previously implicated in promoting an EMT; however, their direct transcriptional target genes and their functional role during EMT have remained elusive. We have uncovered a previously unanticipated role of the transcription factor Tead2 during EMT. During EMT in mammary gland epithelial cells and breast cancer cells, levels of Tead2 increase in the nucleus of cells, thereby directing a predominant nuclear localization of its co-factors Yap and Taz via the formation of Tead2-Yap-Taz complexes. Genome-wide chromatin immunoprecipitation and next generation sequencing in combination with gene expression profiling revealed the transcriptional targets of Tead2 during EMT. Among these, zyxin contributes to the migratory and invasive phenotype evoked by Tead2. The results demonstrate that Tead transcription factors are crucial regulators of the cellular distribution of Yap and Taz, and together they control the expression of genes critical for EMT and metastasis.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , DNA-Binding Proteins/biosynthesis , Epithelial-Mesenchymal Transition/physiology , Phosphoproteins/metabolism , Transcription Factors/biosynthesis , Transcription Factors/metabolism , Zyxin/biosynthesis , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Cycle Proteins , Cell Growth Processes/physiology , Cell Line, Tumor , DNA-Binding Proteins/genetics , Female , Mammary Glands, Animal/cytology , Mammary Glands, Animal/metabolism , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Mice, Transgenic , Phosphoproteins/genetics , Signal Transduction , TEA Domain Transcription Factors , Trans-Activators , Transcription Factors/genetics , Transcriptional Activation , YAP-Signaling Proteins , Zyxin/genetics
14.
PLoS One ; 7(11): e48651, 2012.
Article in English | MEDLINE | ID: mdl-23144919

ABSTRACT

INTRODUCTION: Increasing evidence supports a role of an epithelial to mesenchymal transition (EMT) process in endowing subsets of tumor cells with properties driving malignant tumor progression and resistance to cancer therapy. To advance our understanding of the underlying mechanisms, we sought to generate a transplantable cellular model system that allows defined experimental manipulation and analysis of EMT in vitro and at the same time recapitulates oncogenic EMT in vivo. METHODOLOGY/RESULTS: We have established a stable murine breast cancer cell line (Py2T) from a breast tumor of an MMTV-PyMT transgenic mouse. Py2T cells display a metastable epithelial phenotype characterized by concomitant expression of luminal and basal cytokeratins and sheet migration. Exposure of Py2T cells to transforming growth factor ß (TGFß) in vitro induces reversible EMT accompanied by downregulation of E-cadherin and upregulation of mesenchymal markers, including EMT transcription factors, and a gain in single cell motility and invasiveness. Py2T cells give rise to tumors after orthotopic injection into syngeneic FVB/N mice. Notably, transplantation of epithelial Py2T cells results in the formation of invasive primary tumors with low to absent E-cadherin expression, indicating that the cells undergo EMT-like changes in vivo. This process appears to at least in part depend on TGFß signaling, since tumors formed by Py2T cells expressing a dominant-negative version of TGFß receptor widely maintain their epithelial differentiation status. CONCLUSIONS/SIGNIFICANCE: Together, the data demonstrate that the Py2T cell line represents a versatile model system to study the EMT process in vitro and in vivo. The observation that Py2T cells give rise to tumors and collectively undergo EMT-like changes in vivo highlights the suitability of the Py2T model system as a tool to study tumor-related EMT. In particular, Py2T cells may serve to corroborate recent findings relating EMT to cancer cell stemness, to therapy resistance and to tumor recurrence.


Subject(s)
Epithelial-Mesenchymal Transition/drug effects , Mammary Neoplasms, Animal/pathology , Transforming Growth Factor beta/pharmacology , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Shape/drug effects , Disease Models, Animal , Epithelial Cells/drug effects , Epithelial Cells/pathology , Female , Humans , Kinetics , Mice , Mice, Transgenic , Neoplasm Invasiveness , Neoplasm Transplantation , Phenotype , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...