Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 247
Filter
Add more filters

Publication year range
1.
Cell ; 185(10): 1777-1792.e21, 2022 05 12.
Article in English | MEDLINE | ID: mdl-35512705

ABSTRACT

Spatially resolved transcriptomic technologies are promising tools to study complex biological processes such as mammalian embryogenesis. However, the imbalance between resolution, gene capture, and field of view of current methodologies precludes their systematic application to analyze relatively large and three-dimensional mid- and late-gestation embryos. Here, we combined DNA nanoball (DNB)-patterned arrays and in situ RNA capture to create spatial enhanced resolution omics-sequencing (Stereo-seq). We applied Stereo-seq to generate the mouse organogenesis spatiotemporal transcriptomic atlas (MOSTA), which maps with single-cell resolution and high sensitivity the kinetics and directionality of transcriptional variation during mouse organogenesis. We used this information to gain insight into the molecular basis of spatial cell heterogeneity and cell fate specification in developing tissues such as the dorsal midbrain. Our panoramic atlas will facilitate in-depth investigation of longstanding questions concerning normal and abnormal mammalian development.


Subject(s)
Organogenesis , Transcriptome , Animals , DNA/genetics , Embryo, Mammalian , Female , Gene Expression Profiling/methods , Mammals/genetics , Mice , Organogenesis/genetics , Pregnancy , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , Transcriptome/genetics
2.
Nat Rev Mol Cell Biol ; 21(6): 341-352, 2020 06.
Article in English | MEDLINE | ID: mdl-32300252

ABSTRACT

Epithelial-mesenchymal transition (EMT) encompasses dynamic changes in cellular organization from epithelial to mesenchymal phenotypes, which leads to functional changes in cell migration and invasion. EMT occurs in a diverse range of physiological and pathological conditions and is driven by a conserved set of inducing signals, transcriptional regulators and downstream effectors. With over 5,700 publications indexed by Web of Science in 2019 alone, research on EMT is expanding rapidly. This growing interest warrants the need for a consensus among researchers when referring to and undertaking research on EMT. This Consensus Statement, mediated by 'the EMT International Association' (TEMTIA), is the outcome of a 2-year-long discussion among EMT researchers and aims to both clarify the nomenclature and provide definitions and guidelines for EMT research in future publications. We trust that these guidelines will help to reduce misunderstanding and misinterpretation of research data generated in various experimental models and to promote cross-disciplinary collaboration to identify and address key open questions in this research field. While recognizing the importance of maintaining diversity in experimental approaches and conceptual frameworks, we emphasize that lasting contributions of EMT research to increasing our understanding of developmental processes and combatting cancer and other diseases depend on the adoption of a unified terminology to describe EMT.


Subject(s)
Biomedical Research/standards , Epithelial-Mesenchymal Transition , Animals , Cell Movement , Cell Plasticity , Consensus , Developmental Biology/standards , Humans , Neoplasms/pathology , Terminology as Topic
3.
Cell ; 166(1): 21-45, 2016 Jun 30.
Article in English | MEDLINE | ID: mdl-27368099

ABSTRACT

The significant parallels between cell plasticity during embryonic development and carcinoma progression have helped us understand the importance of the epithelial-mesenchymal transition (EMT) in human disease. Our expanding knowledge of EMT has led to a clarification of the EMT program as a set of multiple and dynamic transitional states between the epithelial and mesenchymal phenotypes, as opposed to a process involving a single binary decision. EMT and its intermediate states have recently been identified as crucial drivers of organ fibrosis and tumor progression, although there is some need for caution when interpreting its contribution to metastatic colonization. Here, we discuss the current state-of-the-art and latest findings regarding the concept of cellular plasticity and heterogeneity in EMT. We raise some of the questions pending and identify the challenges faced in this fast-moving field.


Subject(s)
Epithelial-Mesenchymal Transition , Fibrosis/pathology , Neoplasms/pathology , Animals , Embryonic Development , Epigenesis, Genetic , Humans , Transcription, Genetic
5.
EMBO J ; 42(21): e113448, 2023 11 02.
Article in English | MEDLINE | ID: mdl-37737560

ABSTRACT

The nucleosome remodeling and histone deacetylase (NuRD) complex physically associates with BCL11B to regulate murine T-cell development. However, the function of NuRD complex in mature T cells remains unclear. Here, we characterize the fate and metabolism of human T cells in which key subunits of the NuRD complex or BCL11B are ablated. BCL11B and the NuRD complex bind to each other and repress natural killer (NK)-cell fate in T cells. In addition, T cells upregulate the NK cell-associated receptors and transcription factors, lyse NK-cell targets, and are reprogrammed into NK-like cells (ITNKs) upon deletion of MTA2, MBD2, CHD4, or BCL11B. ITNKs increase OPA1 expression and exhibit characteristically elongated mitochondria with augmented oxidative phosphorylation (OXPHOS) activity. OPA1-mediated elevated OXPHOS enhances cellular acetyl-CoA levels, thereby promoting the reprogramming efficiency and antitumor effects of ITNKs via regulating H3K27 acetylation at specific targets. In conclusion, our findings demonstrate that the NuRD complex and BCL11B cooperatively maintain T-cell fate directly by repressing NK cell-associated transcription and indirectly through a metabolic-epigenetic axis, providing strategies to improve the reprogramming efficiency and antitumor effects of ITNKs.


Subject(s)
Histones , Mi-2 Nucleosome Remodeling and Deacetylase Complex , Animals , Humans , Mice , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics , Mitochondrial Dynamics , Repressor Proteins/genetics , Repressor Proteins/metabolism , T-Lymphocytes/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism
6.
Development ; 151(3)2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38300897

ABSTRACT

Epithelial-mesenchymal transition (EMT) and its reverse mechanism, mesenchymal-epithelial transition (MET), are evolutionarily conserved mechanisms initially identified in studies of early metazoan development. EMT may even have been established in choanoflagellates, the closest unicellular relative of Metazoa. These crucial morphological transitions operate during body plan formation and subsequently in organogenesis. These findings have prompted an increasing number of investigators in biomedicine to assess the importance of such mechanisms that drive epithelial cell plasticity in multiple diseases associated with congenital disabilities and fibrosis, and, most importantly, in the progression of carcinoma. EMT and MET also play crucial roles in regenerative medicine, notably by contributing epigenetic changes in somatic cells to initiate reprogramming into stem cells and their subsequent differentiation into distinct lineages.


Subject(s)
Epithelial Cells , Epithelial-Mesenchymal Transition , Animals , Humans , Cell Differentiation , Fibrosis , Organogenesis
7.
Cell Mol Life Sci ; 81(1): 233, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38780775

ABSTRACT

Patients with head and neck squamous cell carcinoma (HNSCC) are at a high risk of developing recurrence and secondary cancers. This study evaluates the prognostic and surveillance utilities of circulating tumour cells (CTCs) in HNSCC. A total of 154 HNSCC patients were recruited and followed up for 4.5 years. Blood samples were collected at baseline and follow-up. CTCs were isolated using a spiral microfluid device. Recurrence and death due to cancer were assessed during the follow-up period. In patients with HNSCC, the presence of CTCs at baseline was a predictor of recurrence (OR = 8.40, p < 0.0001) and death (OR= ∞, p < 0.0001). Patients with CTCs at baseline had poor survival outcomes (p < 0.0001). Additionally, our study found that patients with CTCs in a follow-up appointment were 2.5 times more likely to experience recurrence or death from HNSCC (p < 0.05) prior to their next clinical visit. Our study highlights the prognostic and monitoring utilities of CTCs' in HNSCC patients. Early identification of CTCs facilitates precise risk assessment, guiding treatment choices and ultimately enhancing patient outcomes.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Neoplasm Recurrence, Local , Neoplastic Cells, Circulating , Squamous Cell Carcinoma of Head and Neck , Humans , Neoplastic Cells, Circulating/pathology , Neoplastic Cells, Circulating/metabolism , Male , Head and Neck Neoplasms/pathology , Head and Neck Neoplasms/mortality , Head and Neck Neoplasms/blood , Head and Neck Neoplasms/diagnosis , Female , Middle Aged , Neoplasm Recurrence, Local/pathology , Squamous Cell Carcinoma of Head and Neck/pathology , Squamous Cell Carcinoma of Head and Neck/mortality , Squamous Cell Carcinoma of Head and Neck/blood , Squamous Cell Carcinoma of Head and Neck/diagnosis , Aged , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/blood , Carcinoma, Squamous Cell/diagnosis , Prognosis , Adult , Follow-Up Studies
8.
Semin Cancer Biol ; 97: 1-11, 2023 12.
Article in English | MEDLINE | ID: mdl-37944215

ABSTRACT

Transforming growth factor-ß (TGF-ß) signaling regulates cell-specific programs involved in embryonic development, wound-healing, and immune homeostasis. Yet, during tumor progression, these TGF-ß-mediated programs are altered, leading to epithelial cell plasticity and a reprogramming of epithelial cells into mesenchymal lineages through epithelial-to-mesenchymal transition (EMT), a critical developmental program in morphogenesis and organogenesis. These changes, in turn, lead to enhanced carcinoma cell invasion, metastasis, immune cell differentiation, immune evasion, and chemotherapy resistance. Here, we discuss EMT as one of the critical programs associated with carcinoma cell plasticity and the influence exerted by TGF-ß on carcinoma status and function. We further explore the composition of carcinoma and other cell populations within the tumor microenvironment, and consider the relevant outcomes related to the programs associated with cancer treatment resistance.


Subject(s)
Carcinoma , Transforming Growth Factor beta , Humans , Epithelial-Mesenchymal Transition/genetics , Epithelial Cells , Signal Transduction , Cell Line, Tumor , Tumor Microenvironment
9.
BMC Cancer ; 24(1): 611, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38773399

ABSTRACT

RNA interactomes and their diversified functionalities have recently benefited from critical methodological advances leading to a paradigm shift from a conventional conception on the regulatory roles of RNA in pathogenesis. However, the dynamic RNA interactomes in adenoma-carcinoma sequence of human CRC remain unexplored. The coexistence of adenoma, cancer, and normal tissues in colorectal cancer (CRC) patients provides an appropriate model to address this issue. Here, we adopted an RNA in situ conformation sequencing technology for mapping RNA-RNA interactions in CRC patients. We observed large-scale paired RNA counts and identified some unique RNA complexes including multiple partners RNAs, single partner RNAs, non-overlapping single partner RNAs. We focused on the antisense RNA OIP5-AS1 and found that OIP5-AS1 could sponge different miRNA to regulate the production of metabolites including pyruvate, alanine and lactic acid. Our findings provide novel perspectives in CRC pathogenesis and suggest metabolic reprogramming of pyruvate for the early diagnosis and treatment of CRC.


Subject(s)
Adenoma , Colorectal Neoplasms , MicroRNAs , Pyruvic Acid , RNA, Long Noncoding , Humans , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , MicroRNAs/genetics , MicroRNAs/metabolism , Adenoma/genetics , Adenoma/metabolism , Adenoma/pathology , Pyruvic Acid/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Gene Expression Regulation, Neoplastic , Metabolic Reprogramming
10.
Cell ; 139(5): 871-90, 2009 Nov 25.
Article in English | MEDLINE | ID: mdl-19945376

ABSTRACT

The epithelial to mesenchymal transition (EMT) plays crucial roles in the formation of the body plan and in the differentiation of multiple tissues and organs. EMT also contributes to tissue repair, but it can adversely cause organ fibrosis and promote carcinoma progression through a variety of mechanisms. EMT endows cells with migratory and invasive properties, induces stem cell properties, prevents apoptosis and senescence, and contributes to immunosuppression. Thus, the mesenchymal state is associated with the capacity of cells to migrate to distant organs and maintain stemness, allowing their subsequent differentiation into multiple cell types during development and the initiation of metastasis.


Subject(s)
Cell Transformation, Neoplastic , Neoplasm Metastasis , Neoplasms/pathology , Neoplastic Stem Cells/cytology , Animals , Epithelial Cells/cytology , Gastrulation , Humans
11.
Breast Cancer Res ; 25(1): 59, 2023 05 30.
Article in English | MEDLINE | ID: mdl-37254190

ABSTRACT

Metastasis accounts for most cancer-associated deaths; yet, this complex process remains poorly understood, particularly the relationship between distant metastasis and primary site-derived cells. Here, we modified the classical MMTV-PyMT breast carcinoma model to trace the fate of mammary-derived carcinoma cells. We show that within the lung, when the metastatic breast carcinoma cells are conditionally depleted, transformed lung epithelial cells generate new metastases. Metastatic breast carcinoma cells transmit H19 long noncoding (lnc) RNA to lung epithelial cells through exosomes. SF3B1 bearing mutations at arginine-625 alternatively splices H19 lncRNA in lung epithelial cells, which selectively acts like a molecular sponge to sequester let-7a and induces Myc upregulation. Under the conditional elimination of primary site-derived breast carcinoma cells, lung malignant cells expressing the mutated SF3B1 splice variant dominate the newly created tumors. Our study suggests that these new carcinoma cells originating from within the colonized organ can replace the primary site-derived malignant cells whenever their expansion is abrogated using an inducible diphtheria toxin receptor in our designed system. These findings should call for a better understanding of metastatic tumors with the specific origin during cancer metastasis.


Subject(s)
Breast Neoplasms , Lung Neoplasms , Mammary Neoplasms, Animal , RNA, Long Noncoding , Animals , Humans , Female , Breast Neoplasms/genetics , Breast Neoplasms/pathology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Alternative Splicing , Breast/pathology , Lung/pathology , Mammary Neoplasms, Animal/pathology , Lung Neoplasms/secondary , Transcription Factors/genetics , Cell Line, Tumor
12.
Liver Int ; 43(12): 2776-2793, 2023 12.
Article in English | MEDLINE | ID: mdl-37804055

ABSTRACT

BACKGROUND & AIMS: The class I- phosphatidylinositol-3 kinases (PI3Ks) signalling is dysregulated in almost all human cancers whereas the isoform-specific roles remain poorly investigated. We reported that the isoform δ (PI3Kδ) regulated epithelial cell polarity and plasticity and recent developments have heightened its role in hepatocellular carcinoma (HCC) and solid tumour progression. However, its role in cholangiocarcinoma (CCA) still lacks investigation. APPROACH & RESULTS: Immunohistochemical analyses of CCA samples reveal a high expression of PI3Kδ in the less differentiated CCA. The RT-qPCR and immunoblot analyses performed on CCA cells stably overexpressing PI3Kδ using lentiviral construction reveal an increase of mesenchymal and stem cell markers and the pluripotency transcription factors. CCA cells stably overexpressing PI3Kδ cultured in 3D culture display a thick layer of ECM at the basement membrane and a wide single lumen compared to control cells. Similar data are observed in vivo, in xenografted tumours established with PI3Kδ-overexpressing CCA cells in immunodeficient mice. The expression of mesenchymal and stemness genes also increases and tumour tissue displays necrosis and fibrosis, along with a prominent angiogenesis and lymphangiogenesis, as in mice liver of AAV8-based-PI3Kδ overexpression. These PI3Kδ-mediated cell morphogenesis and stroma remodelling were dependent on TGFß/Src/Notch signalling. Whole transcriptome analysis of PI3Kδ using the cancer cell line encyclopedia allows the classification of CCA cells according to cancer progression. CONCLUSIONS: Overall, our results support the critical role of PI3Kδ in the progression and aggressiveness of CCA via TGFß/src/Notch-dependent mechanisms and open new directions for the classification and treatment of CCA patients.


Subject(s)
Bile Duct Neoplasms , Carcinoma, Hepatocellular , Cholangiocarcinoma , Liver Neoplasms , Humans , Animals , Mice , Carcinoma, Hepatocellular/pathology , Phosphatidylinositol 3-Kinase , Phosphatidylinositol 3-Kinases/metabolism , Liver Neoplasms/pathology , Cholangiocarcinoma/pathology , Bile Ducts, Intrahepatic/pathology , Bile Duct Neoplasms/genetics , Bile Duct Neoplasms/metabolism , Fibrosis , Transforming Growth Factor beta , Protein Isoforms , Cell Line, Tumor
13.
PLoS Comput Biol ; 17(7): e1009193, 2021 07.
Article in English | MEDLINE | ID: mdl-34297718

ABSTRACT

Epithelial-mesenchymal transition (EMT) and its reverse process, mesenchymal-epithelial transition (MET), are believed to play key roles in facilitating the metastatic cascade. Metastatic lesions often exhibit a similar epithelial-like state to that of the primary tumour, in particular, by forming carcinoma cell clusters via E-cadherin-mediated junctional complexes. However, the factors enabling mesenchymal-like micrometastatic cells to resume growth and reacquire an epithelial phenotype in the target organ microenvironment remain elusive. In this study, we developed a workflow using image-based cell profiling and machine learning to examine morphological, contextual and molecular states of individual breast carcinoma cells (MDA-MB-231). MDA-MB-231 heterogeneous response to the host organ microenvironment was modelled by substrates with controllable stiffness varying from 0.2kPa (soft tissues) to 64kPa (bone tissues). We identified 3 distinct morphological cell types (morphs) varying from compact round-shaped to flattened irregular-shaped cells with lamellipodia, predominantly populating 2-kPa and >16kPa substrates, respectively. These observations were accompanied by significant changes in E-cadherin and vimentin expression. Furthermore, we demonstrate that the bone-mimicking substrate (64kPa) induced multicellular cluster formation accompanied by E-cadherin cell surface localisation. MDA-MB-231 cells responded to different substrate stiffness by morphological adaptation, changes in proliferation rate and cytoskeleton markers, and cluster formation on bone-mimicking substrate. Our results suggest that the stiffest microenvironment can induce MET.


Subject(s)
Epithelial-Mesenchymal Transition/physiology , Machine Learning , Models, Biological , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/physiopathology , Adaptation, Physiological , Antigens, CD/metabolism , Biomarkers, Tumor/metabolism , Biophysical Phenomena , Cadherins/metabolism , Cell Adhesion/physiology , Cell Count , Cell Line, Tumor , Cell Proliferation/physiology , Cell Shape/physiology , Computational Biology , Extracellular Matrix/pathology , Extracellular Matrix/physiology , Female , Humans , Neoplasm Metastasis/pathology , Neoplasm Metastasis/physiopathology , Tumor Microenvironment/physiology , Vimentin/metabolism
14.
Brief Bioinform ; 20(2): 717-731, 2019 03 25.
Article in English | MEDLINE | ID: mdl-29726962

ABSTRACT

With the advent of high-throughput technologies leading to big data generation, increasing number of gene signatures are being published to predict various features of diseases such as prognosis and patient survival. However, to use these signatures for identifying therapeutic targets, use of additional bioinformatic tools is indispensible part of research. Here, we have generated a pipeline comprised of nearly 15 bioinformatic tools and enrichment statistical methods to propose and validate a drug combination strategy from already approved drugs and present our approach using published pan-cancer epithelial-mesenchymal transition (EMT) signatures as a case study. We observed that histone deacetylases were critical targets to tune expression of multiple epithelial versus mesenchymal genes. Moreover, SRC and IKBK were the principal intracellular kinases regulating multiple signaling pathways. To confirm the anti-EMT efficacy of the proposed target combination in silico, we validated expression of targets in mesenchymal versus epithelial subtypes of ovarian cancer. Additionally, we inhibited the pinpointed proteins in vitro using an invasive lung cancer cell line. We found that whereas low-dose mono-therapy failed to limit cell dispersion from collagen spheroids in a microfluidic device as a metric of EMT, the combination fully inhibited dissociation and invasion of cancer cells toward cocultured endothelial cells. Given the approval status and safety profiles of the suggested drugs, the proposed combination set can be considered in clinical trials.


Subject(s)
Computational Biology , Histone Deacetylases/metabolism , I-kappa B Kinase/metabolism , Neoplasms/pathology , src-Family Kinases/metabolism , Cell Adhesion/drug effects , Cell Adhesion/genetics , Cell Line, Tumor , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Humans , Neoplasms/genetics , Neoplasms/metabolism
16.
Cell Mol Life Sci ; 77(14): 2701-2722, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32008085

ABSTRACT

Epithelial to mesenchymal transition (EMT) is a complex plastic and reversible cellular process that has critical roles in diverse physiological and pathological phenomena. EMT is involved in embryonic development, organogenesis and tissue repair, as well as in fibrosis, cancer metastasis and drug resistance. In recent years, the ability to edit the genome using the clustered regularly interspaced palindromic repeats (CRISPR) and associated protein (Cas) system has greatly contributed to identify or validate critical genes in pathway signaling. This review delineates the complex EMT networks and discusses recent studies that have used CRISPR/Cas technology to further advance our understanding of the EMT process.


Subject(s)
CRISPR-Cas Systems/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Editing/methods , Embryonic Development/genetics , Humans , Organogenesis/genetics , Signal Transduction/genetics
17.
EMBO Rep ; 19(8)2018 08.
Article in English | MEDLINE | ID: mdl-29907679

ABSTRACT

In ovarian cancer, the prometastatic RTK AXL promotes motility, invasion and poor prognosis. Here, we show that reduced survival caused by AXL overexpression can be mitigated by the expression of the GPI-anchored tumour suppressor OPCML Further, we demonstrate that AXL directly interacts with OPCML, preferentially so when AXL is activated by its ligand Gas6. As a consequence, AXL accumulates in cholesterol-rich lipid domains, where OPCML resides. Here, phospho-AXL is brought in proximity to the lipid domain-restricted phosphatase PTPRG, which de-phosphorylates the RTK/ligand complex. This prevents AXL-mediated transactivation of other RTKs (cMET and EGFR), thereby inhibiting sustained phospho-ERK signalling, induction of the EMT transcription factor Slug, cell migration and invasion. From a translational perspective, we show that OPCML enhances the effect of the phase II AXL inhibitor R428 in vitro and in vivo We therefore identify a novel mechanism by which two spatially restricted tumour suppressors, OPCML and PTPRG, coordinate to repress AXL-dependent oncogenic signalling.


Subject(s)
Cell Adhesion Molecules/metabolism , Ovarian Neoplasms/metabolism , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 5/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Benzocycloheptenes/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Chickens , Cholesterol/metabolism , Enzyme Activation/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Fallopian Tubes/pathology , Female , GPI-Linked Proteins/metabolism , Gene Silencing/drug effects , Humans , Intercellular Signaling Peptides and Proteins/metabolism , MAP Kinase Signaling System/drug effects , Membrane Microdomains/metabolism , Neoplasm Invasiveness , Ovarian Neoplasms/pathology , Phosphorylation/drug effects , Protein Binding/drug effects , Treatment Outcome , Triazoles/pharmacology , Axl Receptor Tyrosine Kinase
18.
Proc Natl Acad Sci U S A ; 114(11): E2215-E2224, 2017 03 14.
Article in English | MEDLINE | ID: mdl-28251929

ABSTRACT

Robust prognostic gene signatures and therapeutic targets are difficult to derive from expression profiling because of the significant heterogeneity within breast cancer (BC) subtypes. Here, we performed forward genetic screening in mice using Sleeping Beauty transposon mutagenesis to identify candidate BC driver genes in an unbiased manner, using a stabilized N-terminal truncated ß-catenin gene as a sensitizer. We identified 134 mouse susceptibility genes from 129 common insertion sites within 34 mammary tumors. Of these, 126 genes were orthologous to protein-coding genes in the human genome (hereafter, human BC susceptibility genes, hBCSGs), 70% of which are previously reported cancer-associated genes, and ∼16% are known BC suppressor genes. Network analysis revealed a gene hub consisting of E1A binding protein P300 (EP300), CD44 molecule (CD44), neurofibromin (NF1) and phosphatase and tensin homolog (PTEN), which are linked to a significant number of mutated hBCSGs. From our survival prediction analysis of the expression of human BC genes in 2,333 BC cases, we isolated a six-gene-pair classifier that stratifies BC patients with high confidence into prognostically distinct low-, moderate-, and high-risk subgroups. Furthermore, we proposed prognostic classifiers identifying three basal and three claudin-low tumor subgroups. Intriguingly, our hBCSGs are mostly unrelated to cell cycle/mitosis genes and are distinct from the prognostic signatures currently used for stratifying BC patients. Our findings illustrate the strength and validity of integrating functional mutagenesis screens in mice with human cancer transcriptomic data to identify highly prognostic BC subtyping biomarkers.


Subject(s)
Breast Neoplasms/genetics , Cell Transformation, Neoplastic/genetics , DNA Transposable Elements , Genetic Association Studies , Genetic Predisposition to Disease , Mutagenesis, Insertional , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Transformation, Neoplastic/metabolism , Computational Biology/methods , Disease Models, Animal , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , High-Throughput Nucleotide Sequencing , Humans , Mice , Mice, Knockout , Mutation , Prognosis , Reproducibility of Results , Risk , Signal Transduction , Survival Analysis , Transcriptome
19.
Phys Biol ; 16(4): 041004, 2019 05 07.
Article in English | MEDLINE | ID: mdl-30939460

ABSTRACT

Epithelial-to-mesenchymal transition (EMT) is a fundamental developmental process wherein polarized epithelial cells lose their junctional architecture and apical-basal polarity to become motile mesenchymal cells, and there is emerging evidence for its role in propagating tumor dissemination. While many multifaceted nodules converge onto the EMT program, in this review we will highlight the fundamental biology of the signaling schemas that enable EMT. In many cancers, the property of tumor dissemination and metastasis is closely associated with re-enabling developmental properties such as EMT. We discuss the molecular complexity of the tumor heterogeneity in terms of EMT-based gene expression molecular subtypes, and the rewiring of critical signaling nodules in the subtypes displaying higher degrees of EMT can be therapeutically exploited. Specifically in the context of a deadly malignancy such as ovarian cancer where there are no defined mutations or limited biomarkers for developing targeted therapy or personalized medicine, we highlight the importance of identifying EMT-based subtypes that will improve therapeutic intervention. In ovarian cancer, the poor prognosis mesenchymal 'Mes' subtype presents with amplified signaling of the receptor tyrosine kinase (RTK) AXL, extensive crosstalk with other RTKs such as cMET, EGFR and HER2, and sustained temporal activation of extracellular-signal regulated kinase (ERK) leading to induction of EMT transcription factor Slug, underscoring a pathway addiction in Mes that can be therapeutically targeted. We will further examine the emergence of therapeutic modalities in these EMT subtypes and finally conclude with potential interdisciplinary biophysical methodologies to provide additional insights in deciphering the mechanistic and biochemical aspects of EMT. This review intends to provide an overview of the cellular and molecular changes accompanying epithelial-to-mesenchymal transition (EMT) in development and the requisition of this evolutionarily conserved pathway in cancer progression and metastatic disease. Specifically, in a heterogeneous disease such as ovarian cancer lacking defined targetable mutations, the identification of EMT-based subtypes has opened avenues to tailor precision personalized medicine. In particular, using the oncogenic RTK AXL as an example, we will highlight how this classification enables EMT-subtype specific identification of targets that could improve treatment options for patients and how there is a growing need for biophysical approaches to model dynamic processes such as EMT.


Subject(s)
Epithelial-Mesenchymal Transition , Cell Line, Tumor , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/physiology , Female , Humans , Neoplasm Invasiveness , Neoplasm Metastasis , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Precision Medicine/methods , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Axl Receptor Tyrosine Kinase
20.
Breast Cancer Res ; 20(1): 83, 2018 08 02.
Article in English | MEDLINE | ID: mdl-30071870

ABSTRACT

BACKGROUND: Although numerous studies have reported that tricho-rhino-phalangeal syndrome type I (TRPS1) protein, the only reported atypical GATA transcription factor, is overexpressed in various carcinomas, the underlying mechanism(s) by which it contributes to cancer remain unknown. METHODS: Both overexpression and knockdown of TRPS1 assays were performed to examine the effect of TRPS1 on histone deacetylase 2 (HDAC2) protein level and luminal breast cancer cell proliferation. Also, RT-qRCR, luciferase reporter assay and RNA-sequencing were used for transcription detection. Chromatin immunoprecipitation (ChIP) using H4K16ac antibody in conjunction with qPCR was used for determining H4K16ac levels in targeted genes. Furthermore, in vitro cell proliferation assay and in vivo tumor xenografts were used to detect the effect of TRPS1 on tumor growth. RESULTS: We found that TRPS1 scaffolding recruits and enhances interaction between USP4 and HDAC2 leading to HDAC2 de-ubiquitination and H4K16 deacetylation. We detected repression of a set of cellular growth-related genes by the TRPS1-USP4-HDAC2 axis indicating it is essential in tumor growth. In vitro and in vivo experiments confirmed that silencing TRPS1 reduced tumor growth, whereas overexpression of HDAC2 restored tumor growth. CONCLUSION: Our study deciphered the TRPS1-USP4-HDAC2 axis as a novel mechanism that contributes to tumor growth. Significantly, our results revealed the scaffolding function of TPRS1 in USP4-directed HDAC2 de-ubiquitination and provided new mechanistic insights into the crosstalk between TRPS1, ubiquitin, and histone modification systems leading to tumor growth.


Subject(s)
Breast Neoplasms/pathology , Carcinogenesis/pathology , DNA-Binding Proteins/metabolism , Histone Deacetylase 2/metabolism , Transcription Factors/metabolism , Ubiquitin-Specific Proteases/metabolism , Animals , Breast Neoplasms/genetics , Carcinogenesis/genetics , Cell Line, Tumor , Cell Proliferation , DNA-Binding Proteins/genetics , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , HEK293 Cells , Histone Deacetylase 2/genetics , Histones/metabolism , Humans , Mice , Mice, Nude , Protein Binding , Protein Interaction Domains and Motifs/genetics , Repressor Proteins , Transcription Factors/genetics , Ubiquitination , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL