Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
2.
Nature ; 627(8002): 212-220, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38355801

ABSTRACT

Circular RNAs (circRNAs), which are increasingly being implicated in a variety of functions in normal and cancerous cells1-5, are formed by back-splicing of precursor mRNAs in the nucleus6-10. circRNAs are predominantly localized in the cytoplasm, indicating that they must be exported from the nucleus. Here we identify a pathway that is specific for the nuclear export of circular RNA. This pathway requires Ran-GTP, exportin-2 and IGF2BP1. Enhancing the nuclear Ran-GTP gradient by depletion or chemical inhibition of the major protein exporter CRM1 selectively increases the nuclear export of circRNAs, while reducing the nuclear Ran-GTP gradient selectively blocks circRNA export. Depletion or knockout of exportin-2 specifically inhibits nuclear export of circRNA. Analysis of nuclear circRNA-binding proteins reveals that interaction between IGF2BP1 and circRNA is enhanced by Ran-GTP. The formation of circRNA export complexes in the nucleus is promoted by Ran-GTP through its interactions with exportin-2, circRNA and IGF2BP1. Our findings demonstrate that adaptors such as IGF2BP1 that bind directly to circular RNAs recruit Ran-GTP and exportin-2 to export circRNAs in a mechanism that is analogous to protein export, rather than mRNA export.


Subject(s)
Active Transport, Cell Nucleus , Cell Nucleus , RNA Transport , RNA, Circular , Active Transport, Cell Nucleus/physiology , Cell Nucleus/metabolism , Guanosine Triphosphate/metabolism , Karyopherins/antagonists & inhibitors , Karyopherins/deficiency , Karyopherins/genetics , Karyopherins/metabolism , Nuclear Proteins/metabolism , ran GTP-Binding Protein/metabolism , RNA, Circular/metabolism , RNA Precursors/genetics , RNA Precursors/metabolism , RNA-Binding Proteins/metabolism , Exportin 1 Protein/metabolism , Protein Transport
3.
Nucleic Acids Res ; 52(3): 1387-1403, 2024 Feb 09.
Article in English | MEDLINE | ID: mdl-38015468

ABSTRACT

While the majority of circRNAs are formed from infrequent back-splicing of exons from protein coding genes, some can be produced at quite high level and in a regulated manner. We describe the regulation, biogenesis and function of circDOCK1(2-27), a large, abundant circular RNA that is highly regulated during epithelial-mesenchymal transition (EMT) and whose formation depends on the epithelial splicing regulator ESRP1. CircDOCK1(2-27) synthesis in epithelial cells represses cell motility both by diverting transcripts from DOCK1 mRNA production to circRNA formation and by direct inhibition of migration by the circRNA. HITS-CLIP analysis and CRISPR-mediated deletions indicate ESRP1 controls circDOCK1(2-27) biosynthesis by binding a GGU-containing repeat region in intron 1 and detaining its splicing until Pol II completes its 157 kb journey to exon 27. Proximity-dependent biotinylation (BioID) assay suggests ESRP1 may modify the RNP landscape of intron 1 in a way that disfavours communication of exon 1 with exon 2, rather than physically bridging exon 2 to exon 27. The X-ray crystal structure of RNA-bound ESRP1 qRRM2 domain reveals it binds to GGU motifs, with the guanines embedded in clamp-like aromatic pockets in the protein.


Subject(s)
Alternative Splicing , RNA, Circular , RNA-Binding Proteins , rac GTP-Binding Proteins , RNA/genetics , RNA/metabolism , RNA Splicing , RNA, Circular/genetics , Humans , Cell Line, Tumor , RNA-Binding Proteins/metabolism , rac GTP-Binding Proteins/genetics , rac GTP-Binding Proteins/metabolism
4.
RNA Biol ; 21(1): 1-11, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38112323

ABSTRACT

Epithelial-mesenchymal transition (EMT) plays important roles in tumour progression and is orchestrated by dynamic changes in gene expression. While it is well established that post-transcriptional regulation plays a significant role in EMT, the extent of alternative polyadenylation (APA) during EMT has not yet been explored. Using 3' end anchored RNA sequencing, we mapped the alternative polyadenylation (APA) landscape following Transforming Growth Factor (TGF)-ß-mediated induction of EMT in human mammary epithelial cells and found APA generally causes 3'UTR lengthening during this cell state transition. Investigation of potential mediators of APA indicated the RNA-binding protein Quaking (QKI), a splicing factor induced during EMT, regulates a subset of events including the length of its own transcript. Analysis of QKI crosslinked immunoprecipitation (CLIP)-sequencing data identified the binding of QKI within 3' untranslated regions (UTRs) was enriched near cleavage and polyadenylation sites. Following QKI knockdown, APA of many transcripts is altered to produce predominantly shorter 3'UTRs associated with reduced gene expression. These findings reveal the changes in APA that occur during EMT and identify a potential role for QKI in this process.


Subject(s)
Gene Expression Regulation , Polyadenylation , Humans , Epithelial-Mesenchymal Transition/genetics , Base Sequence , RNA-Binding Proteins/genetics , 3' Untranslated Regions
5.
Blood ; 139(26): 3737-3751, 2022 06 30.
Article in English | MEDLINE | ID: mdl-35443029

ABSTRACT

Inducing cell death by the sphingolipid ceramide is a potential anticancer strategy, but the underlying mechanisms remain poorly defined. In this study, triggering an accumulation of ceramide in acute myeloid leukemia (AML) cells by inhibition of sphingosine kinase induced an apoptotic integrated stress response (ISR) through protein kinase R-mediated activation of the master transcription factor ATF4. This effect led to transcription of the BH3-only protein Noxa and degradation of the prosurvival Mcl-1 protein on which AML cells are highly dependent for survival. Targeting this novel ISR pathway, in combination with the Bcl-2 inhibitor venetoclax, synergistically killed primary AML blasts, including those with venetoclax-resistant mutations, as well as immunophenotypic leukemic stem cells, and reduced leukemic engraftment in patient-derived AML xenografts. Collectively, these findings provide mechanistic insight into the anticancer effects of ceramide and preclinical evidence for new approaches to augment Bcl-2 inhibition in the therapy of AML and other cancers with high Mcl-1 dependency.


Subject(s)
Antineoplastic Agents , Leukemia, Myeloid, Acute , Antineoplastic Agents/therapeutic use , Apoptosis , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Cell Line, Tumor , Ceramides/pharmacology , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism
6.
Cell Rep ; 34(1): 108585, 2021 01 05.
Article in English | MEDLINE | ID: mdl-33406413

ABSTRACT

Potent therapeutic inhibition of the androgen receptor (AR) in prostate adenocarcinoma can lead to the emergence of neuroendocrine prostate cancer (NEPC), a phenomenon associated with enhanced cell plasticity. Here, we show that microRNA-194 (miR-194) is a regulator of epithelial-neuroendocrine transdifferentiation. In clinical prostate cancer samples, miR-194 expression and activity were elevated in NEPC and inversely correlated with AR signaling. miR-194 facilitated the emergence of neuroendocrine features in prostate cancer cells, a process mediated by its ability to directly target a suite of genes involved in cell plasticity. One such target was FOXA1, which encodes a transcription factor with a vital role in maintaining the prostate epithelial lineage. Importantly, a miR-194 inhibitor blocked epithelial-neuroendocrine transdifferentiation and inhibited the growth of cell lines and patient-derived organoids possessing neuroendocrine features. Overall, our study reveals a post-transcriptional mechanism regulating the plasticity of prostate cancer cells and provides a rationale for targeting miR-194 in NEPC.


Subject(s)
Cell Transdifferentiation , Hepatocyte Nuclear Factor 3-alpha/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Animals , Carcinoma, Neuroendocrine/genetics , Carcinoma, Neuroendocrine/metabolism , Cell Culture Techniques/methods , Cell Line, Tumor , Cell Lineage , Gene Expression Regulation, Neoplastic , Humans , MAP Kinase Signaling System , Male , Mice , PC-3 Cells , Signal Transduction
7.
Sci Rep ; 10(1): 14593, 2020 09 03.
Article in English | MEDLINE | ID: mdl-32884024

ABSTRACT

The attachment of unique molecular identifiers (UMIs) to RNA molecules prior to PCR amplification and sequencing, makes it possible to amplify libraries to a level that is sufficient to identify rare molecules, whilst simultaneously eliminating PCR bias through the identification of duplicated reads. Accurate de-duplication is dependent upon a sufficiently complex pool of UMIs to allow unique labelling. In applications dealing with complex libraries, such as total RNA-seq, only a limited variety of UMIs are required as the variation in molecules to be sequenced is enormous. However, when sequencing a less complex library, such as small RNAs for which there is a more limited range of possible sequences, we find increased variation in UMIs are required, even beyond that provided in a commercial kit specifically designed for the preparation of small RNA libraries for sequencing. We show that a pool of UMIs randomly varying across eight nucleotides is not of sufficient depth to uniquely tag the microRNAs to be sequenced. This results in over de-duplication of reads and the marked under-estimation of expression of the more abundant microRNAs. Whilst still arguing for the utility of UMIs, this work demonstrates the importance of their considered design to avoid errors in the estimation of gene expression in libraries derived from select regions of the transcriptome or small genomes.


Subject(s)
Algorithms , Epithelial Cells/metabolism , Mesenchymal Stem Cells/metabolism , MicroRNAs/genetics , RNA/chemistry , Sequence Analysis, DNA/methods , Sequence Analysis, RNA/methods , Epithelial Cells/cytology , Humans , Mesenchymal Stem Cells/cytology , RNA/genetics
8.
Nucleic Acids Res ; 47(16): 8606-8619, 2019 09 19.
Article in English | MEDLINE | ID: mdl-31372646

ABSTRACT

Epithelial-mesenchymal transition (EMT) has been a subject of intense scrutiny as it facilitates metastasis and alters drug sensitivity. Although EMT-regulatory roles for numerous miRNAs and transcription factors are known, their functions can be difficult to disentangle, in part due to the difficulty in identifying direct miRNA targets from complex datasets and in deciding how to incorporate 'indirect' miRNA effects that may, or may not, represent biologically relevant information. To better understand how miRNAs exert effects throughout the transcriptome during EMT, we employed Exon-Intron Split Analysis (EISA), a bioinformatic technique that separates transcriptional and post-transcriptional effects through the separate analysis of RNA-Seq reads mapping to exons and introns. We find that in response to the manipulation of miRNAs, a major effect on gene expression is transcriptional. We also find extensive co-ordination of transcriptional and post-transcriptional regulatory mechanisms during both EMT and mesenchymal to epithelial transition (MET) in response to TGF-ß or miR-200c respectively. The prominent transcriptional influence of miRNAs was also observed in other datasets where miRNA levels were perturbed. This work cautions against a narrow approach that is limited to the analysis of direct targets, and demonstrates the utility of EISA to examine complex regulatory networks involving both transcriptional and post-transcriptional mechanisms.


Subject(s)
Epithelial-Mesenchymal Transition/genetics , Gene Regulatory Networks , MicroRNAs/genetics , RNA Processing, Post-Transcriptional , RNA, Messenger/genetics , Transcription, Genetic , Cell Line , Computational Biology/methods , Datasets as Topic , Epidermal Growth Factor/pharmacology , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition/drug effects , ErbB Receptors/genetics , ErbB Receptors/metabolism , Exons , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Introns , MicroRNAs/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/metabolism , Signal Transduction , Transfection , Transforming Growth Factor beta/pharmacology
9.
EMBO J ; 37(13)2018 07 02.
Article in English | MEDLINE | ID: mdl-29871889

ABSTRACT

Members of the miR-200 family are critical gatekeepers of the epithelial state, restraining expression of pro-mesenchymal genes that drive epithelial-mesenchymal transition (EMT) and contribute to metastatic cancer progression. Here, we show that miR-200c and another epithelial-enriched miRNA, miR-375, exert widespread control of alternative splicing in cancer cells by suppressing the RNA-binding protein Quaking (QKI). During EMT, QKI-5 directly binds to and regulates hundreds of alternative splicing targets and exerts pleiotropic effects, such as increasing cell migration and invasion and restraining tumour growth, without appreciably affecting mRNA levels. QKI-5 is both necessary and sufficient to direct EMT-associated alternative splicing changes, and this splicing signature is broadly conserved across many epithelial-derived cancer types. Importantly, several actin cytoskeleton-associated genes are directly targeted by both QKI and miR-200c, revealing coordinated control of alternative splicing and mRNA abundance during EMT These findings demonstrate the existence of a miR-200/miR-375/QKI axis that impacts cancer-associated epithelial cell plasticity through widespread control of alternative splicing.


Subject(s)
Alternative Splicing/physiology , Cell Plasticity/physiology , Epithelial-Mesenchymal Transition/physiology , MicroRNAs/physiology , RNA-Binding Proteins/physiology , Animals , Cell Line, Tumor , Cell Movement , Dogs , Humans , Madin Darby Canine Kidney Cells , Mice, SCID
10.
Sci Signal ; 8(364): ra18, 2015 Feb 17.
Article in English | MEDLINE | ID: mdl-25690013

ABSTRACT

Factors secreted by tumor cells shape the local microenvironment to promote invasion and metastasis, as well as condition the premetastatic niche to enable secondary-site colonization and growth. In addition to this secretome, tumor cells have increased abundance of growth-promoting receptors at the cell surface. We found that the tyrosine phosphatase PTPN14 (also called Pez, which is mutated in various cancers) suppressed metastasis by reducing intracellular protein trafficking through the secretory pathway. Knocking down PTPN14 in tumor cells or injecting the peritoneum of mice with conditioned medium from PTPN14-deficient cell cultures promoted the growth and metastasis of breast cancer xenografts. Loss of catalytically functional PTPN14 increased the secretion of growth factors and cytokines, such as IL-8 (interleukin-8), and increased the abundance of EGFR (epidermal growth factor receptor) at the cell surface of breast cancer cells and of FLT4 (vascular endothelial growth factor receptor 3) at the cell surface of primary lymphatic endothelial cells. We identified RIN1 (Ras and Rab interactor 1) and PRKCD (protein kinase C-δ) as binding partners and substrates of PTPN14. Similar to cells overexpressing PTPN14, receptor trafficking to the cell surface was inhibited in cells that lacked PRKCD or RIN1 or expressed a nonphosphorylatable RIN1 mutant, and cytokine secretion was decreased in cells treated with PRKCD inhibitors. Invasive breast cancer tissue had decreased expression of PTPN14, and patient survival was worse when tumors had increased expression of the genes encoding RIN1 or PRKCD. Thus, PTPN14 prevents metastasis by restricting the trafficking of both soluble and membrane-bound proteins.


Subject(s)
Breast Neoplasms/metabolism , Neoplasm Metastasis/physiopathology , Protein Transport/physiology , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Tumor Microenvironment/physiology , Animals , Blotting, Western , Cell Line, Tumor , Chromatography, Liquid , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Fluorescent Antibody Technique , Gene Knockdown Techniques , Heterografts/metabolism , Heterografts/physiopathology , Humans , Immunoprecipitation , Intercellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins , Isotope Labeling , Mice , Neoplasm Invasiveness/genetics , Neoplasm Metastasis/prevention & control , Protein Kinase C-delta/antagonists & inhibitors , Protein Kinase C-delta/metabolism , Protein Tyrosine Phosphatases, Non-Receptor/pharmacology , Tandem Mass Spectrometry , rab GTP-Binding Proteins/metabolism
11.
EMBO J ; 33(18): 2040-56, 2014 Sep 17.
Article in English | MEDLINE | ID: mdl-25069772

ABSTRACT

The microRNAs of the miR-200 family maintain the central characteristics of epithelia and inhibit tumor cell motility and invasiveness. Using the Ago-HITS-CLIP technology for transcriptome-wide identification of direct microRNA targets in living cells, along with extensive validation to verify the reliability of the approach, we have identified hundreds of miR-200a and miR-200b targets, providing insights into general features of miRNA target site selection. Gene ontology analysis revealed a predominant effect of miR-200 targets in widespread coordinate control of actin cytoskeleton dynamics. Functional characterization of the miR-200 targets indicates that they constitute subnetworks that underlie the ability of cancer cells to migrate and invade, including coordinate effects on Rho-ROCK signaling, invadopodia formation, MMP activity, and focal adhesions. Thus, the miR-200 family maintains the central characteristics of the epithelial phenotype by acting on numerous targets at multiple levels, encompassing both cytoskeletal effectors that control actin filament organization and dynamics, and upstream signals that locally regulate the cytoskeleton to maintain cell morphology and prevent cell migration.


Subject(s)
Cell Movement , Cell Proliferation , Epithelial Cells/physiology , Gene Expression Regulation , Gene Regulatory Networks , MicroRNAs/genetics , MicroRNAs/metabolism , Cell Line , Cytoskeleton/metabolism , Humans
12.
J Biol Chem ; 289(16): 11194-11205, 2014 Apr 18.
Article in English | MEDLINE | ID: mdl-24627491

ABSTRACT

Epithelial-mesenchymal transition (EMT) is required for the specification of tissues during embryonic development and is recapitulated during the metastatic progression of tumors. The miR-200 family plays a critical role in enforcing the epithelial state with their expression lost in cells undergoing EMT. EMT can be mediated by activation of the ZEB1 and ZEB2 (ZEB) transcription factors, which repress miR-200 expression via a self-reinforcing double negative feedback loop to promote the mesenchymal state. However, it remains unclear what factors drive and maintain epithelial-specific expression of miR-200 in the absence of EMT-inducing factors. Here, we show that the transcription factor Specificity Protein 1 (Sp1) binds to the miR-200b∼200a∼429 proximal promoter and activates miR-200 expression in epithelial cells. In mesenchymal cells, Sp1 expression is maintained, but its ability to activate the miR-200 promoter is perturbed by ZEB-mediated repression. Reduction of Sp1 expression caused changes in EMT-associated markers in epithelial cells. Furthermore, we observed co-expression of Sp1 and miR-200 during mouse embryonic development wherein miR-200 expression was only lost in regions with high ZEB expression. Together, these findings indicate that miR-200 family members require Sp1 to drive basal expression and to maintain an epithelial state.


Subject(s)
Epithelial-Mesenchymal Transition/physiology , MicroRNAs/biosynthesis , Response Elements/physiology , Sp1 Transcription Factor/metabolism , Animals , Cell Line, Tumor , Dogs , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Embryonic Development/physiology , Gene Expression Regulation, Developmental/physiology , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Humans , Kruppel-Like Transcription Factors/biosynthesis , Kruppel-Like Transcription Factors/genetics , Madin Darby Canine Kidney Cells , Mice , MicroRNAs/genetics , Repressor Proteins/biosynthesis , Repressor Proteins/genetics , Sp1 Transcription Factor/genetics , Transcription Factors/biosynthesis , Transcription Factors/genetics , Zinc Finger E-box Binding Homeobox 2 , Zinc Finger E-box-Binding Homeobox 1
13.
PLoS One ; 8(9): e75517, 2013.
Article in English | MEDLINE | ID: mdl-24086551

ABSTRACT

The miR-200b~200a~429 gene cluster is a key regulator of EMT and cancer metastasis, however the transcription-based mechanisms controlling its expression during this process are not well understood. We have analyzed the miR-200b~200a~429 locus for epigenetic modifications in breast epithelial and mesenchymal cell lines using chromatin immunoprecipitation assays and DNA methylation analysis. We discovered a novel enhancer located approximately 5.1kb upstream of the miR-200b~200a~429 transcriptional start site. This region was associated with the active enhancer chromatin signature comprising H3K4me1, H3K27ac, RNA polymerase II and CpG dinucleotide hypomethylation. Luciferase reporter assays revealed the upstream enhancer stimulated the transcription of the miR-200b~200a~429 minimal promoter region approximately 27-fold in breast epithelial cells. Furthermore, we found that a region of the enhancer was transcribed, producing a short, GC-rich, mainly nuclear, non-polyadenylated RNA transcript designated miR-200b eRNA. Over-expression of miR-200b eRNA had little effect on miR-200b~200a~429 promoter activity and its production did not correlate with miR-200b~200a~429 gene expression. While additional investigations of miR-200b eRNA function will be necessary, it is possible that miR-200b eRNA may be involved in the regulation of miR-200b~200a~429 gene expression and silencing. Taken together, these findings reveal the presence of a novel enhancer, which contributes to miR-200b~200a~429 transcriptional regulation in epithelial cells.


Subject(s)
Breast Neoplasms/genetics , Enhancer Elements, Genetic/genetics , Gene Expression Regulation, Neoplastic/genetics , MicroRNAs/genetics , Cell Line, Tumor , Chromatin/genetics , Epigenomics/methods , Epithelial-Mesenchymal Transition/genetics , Female , Humans , Promoter Regions, Genetic/genetics , RNA/genetics , Transcription Initiation Site
14.
J Cell Sci ; 126(Pt 10): 2256-66, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23525011

ABSTRACT

The miR-200 family is a key regulator of the epithelial-mesenchymal transition, however, its role in controlling the transition between cancer stem-cell-like and non-stem-cell-like phenotypes is not well understood. We utilized immortalized human mammary epithelial (HMLE) cells to investigate the regulation of the miR-200 family during their conversion to a stem-like phenotype. HMLE cells were found to be capable of spontaneous conversion from a non-stem to a stem-like phenotype and this conversion was accompanied by the loss of miR-200 expression. Stem-like cell fractions isolated from metastatic breast cancers also displayed loss of miR-200 indicating similar molecular changes may occur during breast cancer progression. The phenotypic change observed in HMLE cells was directly controlled by miR-200 because restoration of its expression decreased stem-like properties while promoting a transition to an epithelial phenotype. Investigation of the mechanisms controlling miR-200 expression revealed both DNA methylation and histone modifications were significantly altered in the stem-like and non-stem phenotypes. In particular, in the stem-like phenotype, the miR-200b-200a-429 cluster was silenced primarily through polycomb group-mediated histone modifications whereas the miR-200c-141 cluster was repressed by DNA methylation. These results indicate that the miR-200 family plays a crucial role in the transition between stem-like and non-stem phenotypes and that distinct epigenetic-based mechanisms regulate each miR-200 gene in this process. Therapy targeted against miR-200 family members and epigenetic modifications might therefore be applicable to breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Mammary Glands, Human/metabolism , MicroRNAs/genetics , Neoplastic Stem Cells/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carcinogenesis/genetics , Cell Line, Transformed , DNA Methylation , Epigenetic Repression , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Histones/metabolism , Humans , Mammary Glands, Human/pathology , Molecular Targeted Therapy , Neoplasm Metastasis , Neoplastic Stem Cells/pathology , Promoter Regions, Genetic/genetics , Transgenes/genetics
15.
Neoplasia ; 15(2): 180-91, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23441132

ABSTRACT

Cancer progression is a complex series of events thought to incorporate the reversible developmental process of epithelial-to-mesenchymal transition (EMT). In vitro, the microRNA-200 family maintains the epithelial phenotype by posttranscriptionally inhibiting the E-cadherin repressors, ZEB1 and ZEB2. Here, we used in situ hybridization and immunohistochemistry to assess expression of miR-200 and EMT biomarkers in formalin-fixed paraffin-embedded human colorectal adenocarcinomas. In addition, laser capture microdissection and quantitative real-time polymerase chain reaction were employed to quantify levels of miR-200 in the normal epithelium, tumor core, invasive front, and stroma. We find that miR-200 is downregulated at the invasive front of colorectal adenocarcinomas that have destroyed and invaded beyond the basement membrane. However, regional lymph node metastases and vascular carcinoma deposits show strong expression of miR-200, suggesting this family of miRNAs is involved in the recapitulation of the primary tumor phenotype at metastatic sites. In contrast, adenomas and adenocarcinomas with intact basement membranes showed uniform miR-200 expression from the tumor core to the tumor-host interface. Taken together, these data support the involvement of EMT and mesenchymal-to-epithelial transition (MET) in the metastasis cascade and show that miR-200 is downregulated in the initial stages of stromal invasion but is restored at metastatic sites.


Subject(s)
Basement Membrane/pathology , Colorectal Neoplasms/genetics , Epithelial-Mesenchymal Transition/genetics , MicroRNAs/genetics , Basement Membrane/metabolism , Cadherins/genetics , Cadherins/metabolism , Cell Line, Tumor , Cell Movement/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Disease Progression , Down-Regulation , Gene Expression Regulation, Neoplastic , Humans , Lymphatic Metastasis/genetics , MicroRNAs/metabolism , Neoplasm Invasiveness/genetics , Stromal Cells/metabolism , Stromal Cells/pathology
16.
J Immunol ; 189(9): 4459-69, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-23024272

ABSTRACT

The closely linked human IL-3 and GM-CSF genes are tightly regulated and are expressed in activated T cells and mast cells. In this study, we used transgenic mice to study the developmental regulation of this locus and to identify DNA elements required for its correct activity in vivo. Because these two genes are separated by a CTCF-dependent insulator, and the GM-CSF gene is regulated primarily by its own upstream enhancer, the main objective in this study was to identify regions of the locus required for correct IL-3 gene expression. We initially found that the previously identified proximal upstream IL-3 enhancers were insufficient to account for the in vivo activity of the IL-3 gene. However, an extended analysis of DNase I-hypersensitive sites (DHSs) spanning the entire upstream IL-3 intergenic region revealed the existence of a complex cluster of both constitutive and inducible DHSs spanning the -34- to -40-kb region. The tissue specificity of these DHSs mirrored the activity of the IL-3 gene, and included a highly inducible cyclosporin A-sensitive enhancer at -37 kb that increased IL-3 promoter activity 40-fold. Significantly, inclusion of this region enabled correct in vivo regulation of IL-3 gene expression in T cells, mast cells, and myeloid progenitor cells.


Subject(s)
Enhancer Elements, Genetic/immunology , Gene Expression Regulation, Developmental/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Interleukin-3/biosynthesis , Interleukin-3/genetics , Animals , Cell Line , Cell Line, Tumor , Cells, Cultured , Deoxyribonuclease I/genetics , Enhancer Elements, Genetic/genetics , Genetic Loci/immunology , Humans , Jurkat Cells , Mice , Mice, Transgenic , Tissue Distribution/genetics , Tissue Distribution/immunology
17.
Mol Biol Cell ; 22(10): 1686-98, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21411626

ABSTRACT

Epithelial-mesenchymal transition (EMT) is a form of cellular plasticity that is critical for embryonic development and tumor metastasis. A double-negative feedback loop involving the miR-200 family and ZEB (zinc finger E-box-binding homeobox) transcription factors has been postulated to control the balance between epithelial and mesenchymal states. Here we demonstrate using the epithelial Madin Darby canine kidney cell line model that, although manipulation of the ZEB/miR-200 balance is able to repeatedly switch cells between epithelial and mesenchymal states, the induction and maintenance of a stable mesenchymal phenotype requires the establishment of autocrine transforming growth factor-ß (TGF-ß) signaling to drive sustained ZEB expression. Furthermore, we show that prolonged autocrine TGF-ß signaling induced reversible DNA methylation of the miR-200 loci with corresponding changes in miR-200 levels. Collectively, these findings demonstrate the existence of an autocrine TGF-ß/ZEB/miR-200 signaling network that regulates plasticity between epithelial and mesenchymal states. We find a strong correlation between ZEBs and TGF-ß and negative correlations between miR-200 and TGF-ß and between miR-200 and ZEBs, in invasive ductal carcinomas, consistent with an autocrine TGF-ß/ZEB/miR-200 signaling network being active in breast cancers.


Subject(s)
Autocrine Communication , Epithelial-Mesenchymal Transition/genetics , Homeodomain Proteins/metabolism , MicroRNAs/genetics , Repressor Proteins/metabolism , Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Carcinoma, Ductal, Breast/genetics , Carcinoma, Ductal, Breast/metabolism , Cell Line , Cofilin 2 , DNA Methylation , Dogs , Feedback, Physiological , Female , Homeodomain Proteins/genetics , Humans , MicroRNAs/metabolism , Repressor Proteins/genetics , Signal Transduction , Transcription Factors/genetics , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/pharmacology , Up-Regulation , Zinc Finger E-box Binding Homeobox 2 , Zinc Finger E-box-Binding Homeobox 1
18.
Leuk Res ; 34(9): 1203-13, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20439113

ABSTRACT

The RUNX1 gene, which is essential for normal hematopoiesis, is frequently rearranged by the t(8;21) chromosomal translocation in acute myeloid leukemia. The resulting RUNX1-ETO fusion protein contributes to leukemic progression by directing aberrant association of transcriptional cofactors and epigenetic modifiers to RUNX1 target genes. For example, the GM-CSF gene is activated by RUNX1, but is repressed by RUNX1-ETO. Here we show that RUNX1 normally cooperates with the histone acetyltransferase, CBP, to regulate GM-CSF expression at two levels. Firstly, it directs the establishment of a competent chromatin environment at the GM-CSF promoter prior to gene activation. It then participates in the transcriptional activation of the promoter in response to immune stimuli. In contrast, RUNX1-ETO, which cannot associate with CBP, is unable to transactivate the GM-CSF promoter and is associated with the generation of a repressive chromatin environment at the promoter.


Subject(s)
Core Binding Factor Alpha 2 Subunit/physiology , Epigenesis, Genetic , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Promoter Regions, Genetic , Transcription, Genetic , Base Sequence , Cell Line , Chromatin Immunoprecipitation , DNA Primers , Humans , RNA, Small Interfering
19.
Epigenetics Chromatin ; 3(1): 3, 2010 Jan 19.
Article in English | MEDLINE | ID: mdl-20180972

ABSTRACT

BACKGROUND: Silencing of transgenes in mice is a common phenomenon typically associated with short multi-copy transgenes. We have investigated the regulation of the highly inducible human granulocyte-macrophage colony-stimulating-factor gene (Csf2) in transgenic mice. RESULTS: In the absence of any previous history of transcriptional activation, this transgene was expressed in T lineage cells at the correct inducible level in all lines of mice tested. In contrast, the transgene was silenced in a specific subset of lines in T cells that had encountered a previous episode of activation. Transgene silencing appeared to be both transcription-dependent and mediated by epigenetic mechanisms. Silencing was accompanied by loss of DNase I hypersensitive sites and inability to recruit RNA polymerase II upon stimulation. This pattern of silencing was reflected by increased methylation and decreased acetylation of histone H3 K9 in the transgene. We found that silenced lines were specifically associated with a single pair of tail-to-tail inverted repeated copies of the transgene embedded within a multi-copy array. CONCLUSIONS: Our study suggests that epigenetic transgene silencing can result from convergent transcription of inverted repeats which can lead to silencing of an entire multi-copy transgene array. This mechanism may account for a significant proportion of the reported cases of transgene inactivation in mice.

20.
Cancer Res ; 68(19): 7846-54, 2008 Oct 01.
Article in English | MEDLINE | ID: mdl-18829540

ABSTRACT

Epithelial to mesenchymal transition occurs during embryologic development to allow tissue remodeling and is proposed to be a key step in the metastasis of epithelial-derived tumors. The miR-200 family of microRNAs plays a major role in specifying the epithelial phenotype by preventing expression of the transcription repressors, ZEB1/deltaEF1 and SIP1/ZEB2. We show here that miR-200a, miR-200b, and the related miR-429 are all encoded on a 7.5-kb polycistronic primary miRNA (pri-miR) transcript. We show that the promoter for the pri-miR is located within a 300-bp segment located 4 kb upstream of miR-200b. This promoter region is sufficient to confer expression in epithelial cells and is repressed in mesenchymal cells by ZEB1 and SIP1 through their binding to a conserved pair of ZEB-type E-box elements located proximal to the transcription start site. These findings establish a double-negative feedback loop controlling ZEB1-SIP1 and miR-200 family expression that regulates cellular phenotype and has direct relevance to the role of these factors in tumor progression.


Subject(s)
Epithelial Cells/physiology , Feedback, Physiological/physiology , Homeodomain Proteins/physiology , Mesoderm/physiology , MicroRNAs/physiology , Nerve Tissue Proteins/physiology , RNA-Binding Proteins/physiology , Transcription Factors/physiology , Animals , Cells, Cultured , Dogs , E-Box Elements/physiology , Feedback, Physiological/genetics , Gene Expression Regulation , Humans , Mice , MicroRNAs/genetics , Models, Biological , Multigene Family/physiology , Promoter Regions, Genetic , RNA, Messenger/metabolism , Zinc Finger E-box-Binding Homeobox 1
SELECTION OF CITATIONS
SEARCH DETAIL
...