Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Cell ; 174(3): 716-729.e27, 2018 07 26.
Article in English | MEDLINE | ID: mdl-29961576

ABSTRACT

Single-cell RNA sequencing technologies suffer from many sources of technical noise, including under-sampling of mRNA molecules, often termed "dropout," which can severely obscure important gene-gene relationships. To address this, we developed MAGIC (Markov affinity-based graph imputation of cells), a method that shares information across similar cells, via data diffusion, to denoise the cell count matrix and fill in missing transcripts. We validate MAGIC on several biological systems and find it effective at recovering gene-gene relationships and additional structures. Applied to the epithilial to mesenchymal transition, MAGIC reveals a phenotypic continuum, with the majority of cells residing in intermediate states that display stem-like signatures, and infers known and previously uncharacterized regulatory interactions, demonstrating that our approach can successfully uncover regulatory relations without perturbations.


Subject(s)
Gene Expression Profiling/methods , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , Algorithms , Cell Line , Epistasis, Genetic/genetics , Gene Regulatory Networks/genetics , Humans , Markov Chains , MicroRNAs/genetics , RNA, Messenger/genetics , Software
2.
Cell ; 154(1): 61-74, 2013 Jul 03.
Article in English | MEDLINE | ID: mdl-23827675

ABSTRACT

The recent discovery that normal and neoplastic epithelial cells re-enter the stem cell state raised the intriguing possibility that the aggressiveness of carcinomas derives not from their existing content of cancer stem cells (CSCs) but from their proclivity to generate new CSCs from non-CSC populations. Here, we demonstrate that non-CSCs of human basal breast cancers are plastic cell populations that readily switch from a non-CSC to CSC state. The observed cell plasticity is dependent on ZEB1, a key regulator of the epithelial-mesenchymal transition. We find that plastic non-CSCs maintain the ZEB1 promoter in a bivalent chromatin configuration, enabling them to respond readily to microenvironmental signals, such as TGFß. In response, the ZEB1 promoter converts from a bivalent to active chromatin configuration, ZEB1 transcription increases, and non-CSCs subsequently enter the CSC state. Our findings support a dynamic model in which interconversions between low and high tumorigenic states occur frequently, thereby increasing tumorigenic and malignant potential.


Subject(s)
Breast Neoplasms/pathology , Chromatin/metabolism , Homeodomain Proteins/metabolism , Neoplastic Stem Cells/pathology , Promoter Regions, Genetic , Transcription Factors/metabolism , Animals , Breast Neoplasms/genetics , Epithelial Cells/pathology , Gene Knockdown Techniques , Homeodomain Proteins/genetics , Humans , Hyaluronan Receptors/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , MicroRNAs/metabolism , Neoplastic Stem Cells/metabolism , Transcription Factors/genetics , Transforming Growth Factor beta/metabolism , Zinc Finger E-box-Binding Homeobox 1
3.
Cell ; 145(6): 926-40, 2011 Jun 10.
Article in English | MEDLINE | ID: mdl-21663795

ABSTRACT

The epithelial-mesenchymal transition (EMT) has been associated with the acquisition of motility, invasiveness, and self-renewal traits. During both normal development and tumor pathogenesis, this change in cell phenotype is induced by contextual signals that epithelial cells receive from their microenvironment. The signals that are responsible for inducing an EMT and maintaining the resulting cellular state have been unclear. We describe three signaling pathways, involving transforming growth factor (TGF)-ß and canonical and noncanonical Wnt signaling, that collaborate to induce activation of the EMT program and thereafter function in an autocrine fashion to maintain the resulting mesenchymal state. Downregulation of endogenously synthesized inhibitors of autocrine signals in epithelial cells enables the induction of the EMT program. Conversely, disruption of autocrine signaling by added inhibitors of these pathways inhibits migration and self-renewal in primary mammary epithelial cells and reduces tumorigenicity and metastasis by their transformed derivatives.


Subject(s)
Autocrine Communication , Breast Neoplasms/metabolism , Breast/cytology , Neoplastic Stem Cells/metabolism , Paracrine Communication , Stem Cells/metabolism , Animals , Bone Morphogenetic Proteins/metabolism , Breast/metabolism , Breast/pathology , Breast Neoplasms/pathology , Cell Movement , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition , Female , Humans , Mammary Glands, Animal/cytology , Mammary Glands, Animal/drug effects , Mesoderm/metabolism , Mice , Signal Transduction , Transforming Growth Factor beta/metabolism , Wnt Proteins/metabolism
4.
Proc Natl Acad Sci U S A ; 114(12): E2337-E2346, 2017 03 21.
Article in English | MEDLINE | ID: mdl-28270621

ABSTRACT

Neoplastic cells within individual carcinomas often exhibit considerable phenotypic heterogeneity in their epithelial versus mesenchymal-like cell states. Because carcinoma cells with mesenchymal features are often more resistant to therapy and may serve as a source of relapse, we sought to determine whether such cells could be further stratified into functionally distinct subtypes. Indeed, we find that a basal epithelial marker, integrin-ß4 (ITGB4), can be used to enable stratification of mesenchymal-like triple-negative breast cancer (TNBC) cells that differ from one another in their relative tumorigenic abilities. Notably, we demonstrate that ITGB4+ cancer stem cell (CSC)-enriched mesenchymal cells reside in an intermediate epithelial/mesenchymal phenotypic state. Among patients with TNBC who received chemotherapy, elevated ITGB4 expression was associated with a worse 5-year probability of relapse-free survival. Mechanistically, we find that the ZEB1 (zinc finger E-box binding homeobox 1) transcription factor activity in highly mesenchymal SUM159 TNBC cells can repress expression of the epithelial transcription factor TAp63α (tumor protein 63 isoform 1), a protein that promotes ITGB4 expression. In addition, we demonstrate that ZEB1 and ITGB4 are important in modulating the histopathological phenotypes of tumors derived from mesenchymal TNBC cells. Hence, mesenchymal carcinoma cell populations are internally heterogeneous, and ITGB4 is a mechanistically driven prognostic biomarker that can be used to identify the more aggressive subtypes of mesenchymal carcinoma cells in TNBC. The ability to rapidly isolate and mechanistically interrogate the CSC-enriched, partially mesenchymal carcinoma cells should further enable identification of novel therapeutic opportunities to improve the prognosis for high-risk patients with TNBC.


Subject(s)
Carcinoma/metabolism , Integrin beta4/metabolism , Mesoderm/metabolism , Neoplastic Stem Cells/metabolism , Triple Negative Breast Neoplasms/metabolism , Carcinoma/genetics , Carcinoma/mortality , Cell Line, Tumor , Epithelial-Mesenchymal Transition , Female , Humans , Integrin beta4/genetics , Mesoderm/cytology , Prognosis , Transcription Factors/genetics , Transcription Factors/metabolism , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/mortality , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Zinc Finger E-box-Binding Homeobox 1/genetics , Zinc Finger E-box-Binding Homeobox 1/metabolism
5.
Cancer Metastasis Rev ; 35(4): 645-654, 2016 12.
Article in English | MEDLINE | ID: mdl-27878502

ABSTRACT

Carcinoma cells that are induced to suppress their epithelial features and upregulate mesenchymal gene expression programs acquire traits that promote an invasive and metastatic phenotype. This is achieved through the expression of a program termed the epithelial-to-mesenchymal transition (EMT)-a fundamental cell-biological process that plays key roles in embryogenesis and wound healing. Re-activation of the EMT during cancer promotes disease progression and enhances the metastatic phenotype by bestowing upon previously benign carcinoma cell traits such as migration, invasion, resistance to anoikis, chemoresistance and tumour-initiating potential. Herein, we discuss recent insights into the function of the EMT and cancer cell plasticity during cancer progression, with a focus on their role in promoting successful completion of the later stages of the metastatic cascade.


Subject(s)
Breast Neoplasms/pathology , Cell Plasticity/physiology , Animals , Breast Neoplasms/drug therapy , Cell Plasticity/drug effects , Epithelial-Mesenchymal Transition/drug effects , Female , Humans , Neoplasm Metastasis
6.
Proc Natl Acad Sci U S A ; 108(19): 7950-5, 2011 May 10.
Article in English | MEDLINE | ID: mdl-21498687

ABSTRACT

Current models of stem cell biology assume that normal and neoplastic stem cells reside at the apices of hierarchies and differentiate into nonstem progeny in a unidirectional manner. Here we identify a subpopulation of basal-like human mammary epithelial cells that departs from that assumption, spontaneously dedifferentiating into stem-like cells. Moreover, oncogenic transformation enhances the spontaneous conversion, so that nonstem cancer cells give rise to cancer stem cell (CSC)-like cells in vitro and in vivo. We further show that the differentiation state of normal cells-of-origin is a strong determinant of posttransformation behavior. These findings demonstrate that normal and CSC-like cells can arise de novo from more differentiated cell types and that hierarchical models of mammary stem cell biology should encompass bidirectional interconversions between stem and nonstem compartments. The observed plasticity may allow derivation of patient-specific adult stem cells without genetic manipulation and holds important implications for therapeutic strategies to eradicate cancer.


Subject(s)
Breast Neoplasms/pathology , Breast/cytology , Cell Dedifferentiation , Adult Stem Cells/cytology , Adult Stem Cells/physiology , Animals , Breast/physiology , Breast Neoplasms/physiopathology , CD24 Antigen/metabolism , Cell Dedifferentiation/physiology , Cell Transformation, Neoplastic/pathology , Cells, Cultured , Epithelial Cells/cytology , Epithelial Cells/physiology , Female , Humans , Hyaluronan Receptors/metabolism , Mammary Glands, Animal/cytology , Membrane Proteins/metabolism , Mice , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/physiology , Stem Cell Transplantation , Transplantation, Heterologous
7.
Cancer Discov ; 14(4): 643-647, 2024 Apr 04.
Article in English | MEDLINE | ID: mdl-38571433

ABSTRACT

SUMMARY: Understandably, conventional therapeutic strategies have focused on controlling primary tumors. We ask whether the cost of such strategies is actually an increased likelihood of metastatic relapse.


Subject(s)
Neoplasms , Humans , Neoplasms/therapy , Tumor Microenvironment
8.
Clin Chem ; 59(1): 168-79, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23220226

ABSTRACT

BACKGROUND: Heterogeneity within a given cancer arises from diverse cell types recruited to the tumor and from genetic and/or epigenetic differences amongst the cancer cells themselves. These factors conspire to create a disease with various phenotypes. There are 2 established models of cancer development and progression to metastatic disease. These are the clonal evolution and cancer stem cell models. CONTENT: The clonal evolution theory suggests that successive mutations accumulating in a given cell generate clonal outgrowths that thrive in response to microenvironmental selection pressures, dictating the phenotype of the tumor. The alternative cancer stem cell (CSC) model suggests that cancer cells with similar genetic backgrounds can be hierarchically organized according to their tumorigenic potential. Accordingly, CSCs reside at the apex of the hierarchy and are thought to possess the majority of a cancer's tumor-initiating and metastatic ability. A defining feature of this model is its apparent unidirectional nature, whereby CSCs undergo symmetric division to replenish the CSC pool and irreversible asymmetric division to generate daughter cells (non-CSCs) with low tumorigenic potential. However, evolving evidence supports a new model of tumorigenicity, in which considerable plasticity exists between the non-CSC and CSC compartments, such that non-CSCs can reacquire a CSC phenotype. These findings suggest that some tumors may adhere to a plastic CSC model, in which bidirectional conversions are common and essential components of tumorigenicity. SUMMARY: Accumulating evidence surrounding the plasticity of cancer cells, in particular, suggests that aggressive CSCs can be created de novo within a tumor. Given the current focus on therapeutic targeting of CSCs, we discuss the implications of non-CSC-to-CSC conversions on the development of future therapies.


Subject(s)
Neoplasms/pathology , Humans , Models, Biological
9.
Adv Sci (Weinh) ; 10(22): e2301802, 2023 08.
Article in English | MEDLINE | ID: mdl-37217832

ABSTRACT

Epithelial-mesenchymal transition (EMT) is a reversible transcriptional program invoked by cancer cells to drive cancer progression. Transcription factor ZEB1 is a master regulator of EMT, driving disease recurrence in poor-outcome triple negative breast cancers (TNBCs). Here, this work silences ZEB1 in TNBC models by CRISPR/dCas9-mediated epigenetic editing, resulting in highly-specific and nearly complete suppression of ZEB1 in vivo, accompanied by long-lasting tumor inhibition. Integrated "omic" changes promoted by dCas9 linked to the KRAB domain (dCas9-KRAB) enabled the discovery of a ZEB1-dependent-signature of 26 genes differentially-expressed and -methylated, including the reactivation and enhanced chromatin accessibility in cell adhesion loci, outlining epigenetic reprogramming toward a more epithelial state. In the ZEB1 locus transcriptional silencing is associated with induction of locally-spread heterochromatin, significant changes in DNA methylation at specific CpGs, gain of H3K9me3, and a near complete erasure of H3K4me3 in the ZEB1 promoter. Epigenetic shifts induced by ZEB1-silencing are enriched in a subset of human breast tumors, illuminating a clinically-relevant hybrid-like state. Thus, the synthetic epi-silencing of ZEB1 induces stable "lock-in" epigenetic reprogramming of mesenchymal tumors associated with a distinct and stable epigenetic landscape. This work outlines epigenome-engineering approaches for reversing EMT and customizable precision molecular oncology approaches for targeting poor outcome breast cancers.


Subject(s)
Triple Negative Breast Neoplasms , Humans , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology , Clustered Regularly Interspaced Short Palindromic Repeats , Neoplasm Recurrence, Local/genetics , Transcription Factors/genetics , Epigenesis, Genetic/genetics
10.
bioRxiv ; 2023 Oct 02.
Article in English | MEDLINE | ID: mdl-37873432

ABSTRACT

Intra-tumoral phenotypic heterogeneity promotes tumor relapse and therapeutic resistance and remains an unsolved clinical challenge. It manifests along multiple phenotypic axes and decoding the interconnections among these different axes is crucial to understand its molecular origins and to develop novel therapeutic strategies to control it. Here, we use multi-modal transcriptomic data analysis - bulk, single-cell and spatial transcriptomics - from breast cancer cell lines and primary tumor samples, to identify associations between epithelial-mesenchymal transition (EMT) and luminal-basal plasticity - two key processes that enable heterogeneity. We show that luminal breast cancer strongly associates with an epithelial cell state, but basal breast cancer is associated with hybrid epithelial/mesenchymal phenotype(s) and higher phenotypic heterogeneity. These patterns were inherent in methylation profiles, suggesting an epigenetic crosstalk between EMT and lineage plasticity in breast cancer. Mathematical modelling of core underlying gene regulatory networks representative of the crosstalk between the luminal-basal and epithelial-mesenchymal axes recapitulate and thus elucidate mechanistic underpinnings of the observed associations from transcriptomic data. Our systems-based approach integrating multi-modal data analysis with mechanism-based modeling offers a predictive framework to characterize intra-tumor heterogeneity and to identify possible interventions to restrict it.

11.
Cancer Discov ; 12(8): 1847-1859, 2022 08 05.
Article in English | MEDLINE | ID: mdl-35736000

ABSTRACT

ABSTRACT: Phenotypic plasticity describes the ability of cancer cells to undergo dynamic, nongenetic cell state changes that amplify cancer heterogeneity to promote metastasis and therapy evasion. Thus, cancer cells occupy a continuous spectrum of phenotypic states connected by trajectories defining dynamic transitions upon a cancer cell state landscape. With technologies proliferating to systematically record molecular mechanisms at single-cell resolution, we illuminate manifold learning techniques as emerging computational tools to effectively model cell state dynamics in a way that mimics our understanding of the cell state landscape. We anticipate that "state-gating" therapies targeting phenotypic plasticity will limit cancer heterogeneity, metastasis, and therapy resistance. SIGNIFICANCE: Nongenetic mechanisms underlying phenotypic plasticity have emerged as significant drivers of tumor heterogeneity, metastasis, and therapy resistance. Herein, we discuss new experimental and computational techniques to define phenotypic plasticity as a scaffold to guide accelerated progress in uncovering new vulnerabilities for therapeutic exploitation.


Subject(s)
Epithelial-Mesenchymal Transition , Neoplasms , Adaptation, Physiological , Humans , Neoplasms/drug therapy
12.
Patterns (N Y) ; 3(9): 100577, 2022 Sep 09.
Article in English | MEDLINE | ID: mdl-36124302

ABSTRACT

Exciting advances in technologies to measure biological systems are currently at the forefront of research. The ability to gather data along an increasing number of omic dimensions has created a need for tools to analyze all of this information together, rather than siloing each technology into separate analysis pipelines. To advance this goal, we introduce a framework called the single-cell multi-modal generative adversarial network (scMMGAN) that integrates data from multiple modalities into a unified representation in the ambient data space for downstream analysis using a combination of adversarial learning and data geometry techniques. The framework's key improvement is an additional diffusion geometry loss with a new kernel that constrains the otherwise over-parameterized GAN. We demonstrate scMMGAN's ability to produce more meaningful alignments than alternative methods on a wide variety of data modalities and that its output can be used to draw conclusions from real-world biological experimental data.

13.
Cells Tissues Organs ; 192(3): 141-57, 2010.
Article in English | MEDLINE | ID: mdl-20357428

ABSTRACT

Recreating an environment that supports and promotes fundamental homeostatic mechanisms is a significant challenge in tissue engineering. Optimizing cell survival, proliferation, differentiation, apoptosis and angiogenesis, and providing suitable stromal support and signalling cues are keys to successfully generating clinically useful tissues. Interestingly, those components are often subverted in the cancer setting, where aberrant angiogenesis, cellular proliferation, cell signalling and resistance to apoptosis drive malignant growth. In contrast to tissue engineering, identifying and inhibiting those pathways is a major challenge in cancer research. The recent discovery of adult tissue-specific stem cells has had a major impact on both tissue engineering and cancer research. The unique properties of these cells and their role in tissue and organ repair and regeneration hold great potential for engineering tissue-specific constructs. The emerging body of evidence implicating stem cells and progenitor cells as the source of oncogenic transformation prompts caution when using these cells for tissue-engineering purposes. While tissue engineering and cancer research may be considered as opposed fields of research with regard to their proclaimed goals, the compelling overlap in fundamental pathways underlying these processes suggests that cross-disciplinary research will benefit both fields. In this review article, tissue engineering and cancer research are brought together and explored with regard to discoveries that may be of mutual benefit.


Subject(s)
Neoplasms/etiology , Neoplasms/pathology , Tissue Engineering/trends , Animals , Biomedical Research , Humans , Neoplasms/metabolism , Regeneration/physiology , Stem Cells/cytology , Stem Cells/pathology , Stem Cells/physiology , Tissue Engineering/adverse effects
14.
Cancers (Basel) ; 11(10)2019 Oct 16.
Article in English | MEDLINE | ID: mdl-31623163

ABSTRACT

Therapies that prevent metastatic dissemination and tumor growth in secondary organs are severely lacking. A better understanding of the mechanisms that drive metastasis will lead to improved therapies that increase patient survival. Within a tumor, cancer cells are equipped with different phenotypic and functional capacities that can impact their ability to complete the metastatic cascade. That phenotypic heterogeneity can be derived from a combination of factors, in which the genetic make-up, interaction with the environment, and ability of cells to adapt to evolving microenvironments and mechanical forces play a major role. In this review, we discuss the specific properties of those cancer cell subgroups and the mechanisms that confer or restrict their capacity to metastasize.

15.
Cancer Res ; 66(23): 11271-8, 2006 Dec 01.
Article in English | MEDLINE | ID: mdl-17145872

ABSTRACT

Epithelial-to-mesenchymal transition (EMT) increases cell migration and invasion, and facilitates metastasis in multiple carcinoma types, but belies epithelial similarities between primary and secondary tumors. This study addresses the importance of mesenchymal-to-epithelial transition (MET) in the formation of clinically significant metastasis. The previously described bladder carcinoma TSU-Pr1 (T24) progression series of cell lines selected in vivo for increasing metastatic ability following systemic seeding was used in this study. It was found that the more metastatic sublines had acquired epithelial characteristics. Epithelial and mesenchymal phenotypes were confirmed in the TSU-Pr1 series by cytoskeletal and morphologic analysis, and by performance in a panel of in vitro assays. Metastatic ability was examined following inoculation at various sites. Epithelial characteristics associated with dramatically increased bone and soft tissue colonization after intracardiac or intratibial injection. In contrast, the more epithelial sublines showed decreased lung metastases following orthotopic inoculation, supporting the concept that EMT is important for the escape of tumor cells from the primary tumor. We confirmed the overexpression of the IIIc subtype of multiple fibroblast growth factor receptors (FGFR) through the TSU-Pr1 series, and targeted abrogation of FGFR2IIIc reversed the MET and associated functionality in this system and increased survival following in vivo inoculation in severe combined immunodeficient mice. This model is the first to specifically model steps of the latter part of the metastatic cascade in isogenic cell lines, and confirms the suspected role of MET in secondary tumor growth.


Subject(s)
Carcinoma, Transitional Cell/pathology , Epithelial Cells/pathology , Mesoderm/pathology , Urinary Bladder Neoplasms/pathology , Animals , Blotting, Western , Carcinoma, Transitional Cell/genetics , Carcinoma, Transitional Cell/metabolism , Cell Differentiation , Cell Line, Tumor , Epithelial Cells/metabolism , Gene Expression Regulation, Neoplastic , Humans , Keratins/metabolism , Mesoderm/metabolism , Mice , Neoplasm Metastasis , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptor, Fibroblast Growth Factor, Type 2/genetics , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Receptor, Fibroblast Growth Factor, Type 2/physiology , Reverse Transcriptase Polymerase Chain Reaction , Survival Analysis , Time Factors , Transplantation, Heterologous , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/metabolism , Vimentin/metabolism
16.
Dev Cell ; 47(6): 691-693, 2018 12 17.
Article in English | MEDLINE | ID: mdl-30562512

ABSTRACT

Inhibition of metastatic cancer cell colonization and outgrowth is arguably one of the greatest therapeutic challenges. Reporting in Cancer Discovery, Liu et al. (2018) describe how homophilic interactions of CD44, a classical breast cancer stem cell marker, drive tumor cell aggregation outside the primary tumor to augment their metastatic potential.


Subject(s)
Breast Neoplasms , Cell Aggregation , Humans , Hyaluronan Receptors , Neoplastic Stem Cells
17.
Nat Cell Biol ; 20(9): 1084-1097, 2018 09.
Article in English | MEDLINE | ID: mdl-30154549

ABSTRACT

Lack of insight into mechanisms governing breast cancer metastasis has precluded the development of curative therapies. Metastasis-initiating cancer cells (MICs) are uniquely equipped to establish metastases, causing recurrence and therapeutic resistance. Using various metastasis models, we discovered that certain primary tumours elicit a systemic inflammatory response involving interleukin-1ß (IL-1ß)-expressing innate immune cells that infiltrate distant MIC microenvironments. At the metastatic site, IL-1ß maintains MICs in a ZEB1-positive differentiation state, preventing MICs from generating highly proliferative E-cadherin-positive progeny. Thus, when the inherent plasticity of MICs is impeded, overt metastases cannot be established. Ablation of the pro-inflammatory response or inhibition of the IL-1 receptor relieves the differentiation block and results in metastatic colonization. Among patients with lymph node-positive breast cancer, high primary tumour IL-1ß expression is associated with better overall survival and distant metastasis-free survival. Our data reveal complex interactions that occur between primary tumours and disseminated MICs that could be exploited to improve patient survival.


Subject(s)
Breast Neoplasms/metabolism , Inflammation/metabolism , Interleukin-1beta/metabolism , Lung Neoplasms/metabolism , Myeloid Cells/metabolism , Tumor Microenvironment , Animals , Anti-Inflammatory Agents/pharmacology , Antigens, CD/genetics , Antigens, CD/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cadherins/genetics , Cadherins/metabolism , Cell Communication , Cell Differentiation , Cell Line, Tumor , Cell Plasticity , Cell Proliferation , Female , Humans , Inflammation/immunology , Inflammation/pathology , Inflammation/prevention & control , Interleukin-1beta/genetics , Interleukin-1beta/pharmacology , Lung Neoplasms/immunology , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Lymphatic Metastasis , Mice, Nude , Myeloid Cells/drug effects , Myeloid Cells/immunology , Myeloid Cells/pathology , Signal Transduction , Time Factors , Xenograft Model Antitumor Assays , Zinc Finger E-box-Binding Homeobox 1/genetics , Zinc Finger E-box-Binding Homeobox 1/metabolism
18.
Elife ; 72018 07 30.
Article in English | MEDLINE | ID: mdl-30059005

ABSTRACT

Alternative splicing of mRNA precursors represents a key gene expression regulatory step and permits the generation of distinct protein products with diverse functions. In a genome-scale expression screen for inducers of the epithelial-to-mesenchymal transition (EMT), we found a striking enrichment of RNA-binding proteins. We validated that QKI and RBFOX1 were necessary and sufficient to induce an intermediate mesenchymal cell state and increased tumorigenicity. Using RNA-seq and eCLIP analysis, we found that QKI and RBFOX1 coordinately regulated the splicing and function of the actin-binding protein FLNB, which plays a causal role in the regulation of EMT. Specifically, the skipping of FLNB exon 30 induced EMT by releasing the FOXC1 transcription factor. Moreover, skipping of FLNB exon 30 is strongly associated with EMT gene signatures in basal-like breast cancer patient samples. These observations identify a specific dysregulation of splicing, which regulates tumor cell plasticity and is frequently observed in human cancer.


Subject(s)
Alternative Splicing/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Filamins/genetics , Mesenchymal Stem Cells/metabolism , Animals , Base Sequence , Cell Line, Tumor , Epithelial-Mesenchymal Transition/genetics , Exons/genetics , Female , Filamins/metabolism , Gene Expression Regulation, Neoplastic , Genome, Human , Humans , Hyaluronan Receptors/metabolism , Mice, Nude , Neoplasm Proteins/metabolism , Open Reading Frames/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Reproducibility of Results
19.
Cells Tissues Organs ; 185(1-3): 7-19, 2007.
Article in English | MEDLINE | ID: mdl-17587803

ABSTRACT

Cellular plasticity is fundamental to embryonic development. The importance of cellular transitions in development is first apparent during gastrulation when the process of epithelial to mesenchymal transition transforms polarized epithelial cells into migratory mesenchymal cells that constitute the embryonic and extraembryonic mesoderm. It is now widely accepted that this developmental pathway is exploited in various disease states, including cancer progression. The loss of epithelial characteristics and the acquisition of a mesenchymal-like migratory phenotype are crucial to the development of invasive carcinoma and metastasis. However, given the morphological similarities between primary tumour and metastatic lesions, it is likely that tumour cells re-activate certain epithelial properties through a mesenchymal to epithelial transition (MET) at the secondary site, although this is yet to be proven. MET is also an essential developmental process and has been extensively studied in kidney organogenesis and somitogenesis. In this review we describe the process of MET, highlight important mediators, and discuss their implication in the context of cancer progression.


Subject(s)
Carcinoma/pathology , Cell Transformation, Neoplastic/pathology , Epithelial Cells/pathology , Mesoderm/cytology , Urinary Bladder Neoplasms/pathology , Animals , Carcinoma/genetics , Carcinoma/metabolism , Cell Transformation, Neoplastic/genetics , Disease Progression , Epithelial Cells/metabolism , Humans , Models, Biological , Neoplasm Metastasis , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/metabolism
20.
BMC Cancer ; 6: 53, 2006 Mar 06.
Article in English | MEDLINE | ID: mdl-16519808

ABSTRACT

BACKGROUND: Although PPARgamma antagonists have shown considerable pre-clinical efficacy, recent studies suggest PPARgamma ligands induce PPARgamma-independent effects. There is a need to better define such effects to permit rational utilization of these agents. METHODS: We have studied the effects of a range of endogenous and synthetic PPARgamma ligands on proliferation, growth arrest (FACS analysis) and apoptosis (caspase-3/7 activation and DNA fragmentation) in multiple prostate carcinoma cell lines (DU145, PC-3 and LNCaP) and in a series of cell lines modelling metastatic transitional cell carcinoma of the bladder (TSU-Pr1, TSU-Pr1-B1 and TSU-Pr1-B2). RESULTS: 15-deoxy-prostaglandin J2 (15dPGJ2), troglitazone (TGZ) and to a lesser extent ciglitazone exhibited inhibitory effects on cell number; the selective PPARgamma antagonist GW9662 did not reverse these effects. Rosiglitazone and pioglitazone had no effect on proliferation. In addition, TGZ induced G0/G1 growth arrest whilst 15dPGJ2 induced apoptosis. CONCLUSION: Troglitazone and 15dPGJ2 inhibit growth of prostate and bladder carcinoma cell lines through different mechanisms and the effects of both agents are PPARgamma-independent.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma/drug therapy , Chromans/pharmacology , PPAR gamma/agonists , Prostaglandin D2/analogs & derivatives , Prostatic Neoplasms/drug therapy , Thiazolidinediones/pharmacology , Urinary Bladder Neoplasms/drug therapy , Antineoplastic Agents/therapeutic use , Apoptosis , Carcinoma/metabolism , Carcinoma/pathology , Carcinoma, Transitional Cell/drug therapy , Carcinoma, Transitional Cell/metabolism , Carcinoma, Transitional Cell/pathology , Caspase 3 , Caspase 7 , Caspases/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Chromans/therapeutic use , DNA Fragmentation , Humans , Ligands , Male , PPAR gamma/antagonists & inhibitors , PPAR gamma/metabolism , Prostaglandin D2/pharmacology , Prostaglandin D2/therapeutic use , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , RNA, Messenger/metabolism , Thiazolidinediones/therapeutic use , Troglitazone , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL