Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
Cell Mol Gastroenterol Hepatol ; : 101390, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39128652

ABSTRACT

BACKGROUND & AIMS: Human sporadic colorectal cancer (CRC) results from a multistep pathway with sequential acquisition of specific genetic mutations in the colorectal epithelium. Modeling CRC in vivo is critical for understanding the tumor microenvironment. To accurately recapitulate human CRC pathogenesis, mouse models must include these multi-step genetic abnormalities. AIMS: Generate a sporadic CRC model that more closely mimics this multi-step process and use this model to study the role of a novel Let7 target PLAGL2 in CRC pathogenesis. METHODS: We generated a CRISPR/Cas9 somatic mutagenesis mouse model that is inducible and multiplexed for simultaneous inactivation of multiple genes involved in CRC pathogenesis. We used both a doxycycline-inducible transcriptional activator and a dox-inactivated transcriptional repressor to achieve tight, non-leaky expression of the Cas9 nickase. This mouse has transgenic expression of multiple guide RNAs to induce sporadic inactivation in the gut epithelium of four tumor suppressor genes commonly mutated in CRC, Apc, Pten, Smad4 and Trp53. These were crossed to Vil-LCL-PLAGL2 mice which have Cre-inducible overexpression of PLAGL2 in the gut epithelium. RESULTS: These mice exhibited random somatic mutations in all four targeted tumor suppressor genes, resulting in multiple adenomas and adenocarcinomas in the small bowel and colon. Crosses with Vil-LCL-PLAGL2 mice demonstrated that gut-specific PLAGL2 overexpression increased colon tumor growth. CONCLUSIONS: This conditional model represents a new CRISPR/Cas9-mediated mouse model of colorectal carcinogenesis. These mice can be used to investigate the role of novel, previously uncharacterized genes in CRC, in the context of multiple commonly mutated tumor suppressor genes and thus more closely mimic human CRC pathogenesis.

2.
Mol Ther ; 31(10): 2816, 2023 Oct 04.
Article in English | MEDLINE | ID: mdl-37582361
3.
Am J Physiol Gastrointest Liver Physiol ; 325(2): G196-G211, 2023 08 01.
Article in English | MEDLINE | ID: mdl-37310750

ABSTRACT

Colorectal cancer (CRC) tumorigenesis and progression are linked to common oncogenic mutations, especially in the tumor suppressor APC, whose loss triggers the deregulation of TCF4/ß-Catenin activity. CRC tumorigenesis is also driven by multiple epimutational modifiers such as transcriptional regulators. We describe the common (and near-universal) activation of the zinc finger transcription factor and Let-7 target PLAGL2 in CRC and find that it is a key driver of intestinal epithelial transformation. PLAGL2 drives proliferation, cell cycle progression, and anchorage-independent growth in CRC cell lines and nontransformed intestinal cells. Investigating effects of PLAGL2 on downstream pathways revealed very modest effects on canonical Wnt signaling. Alternatively, we find pronounced effects on the direct PLAGL2 target genes IGF2, a fetal growth factor, and ASCL2, an intestinal stem cell-specific bHLH transcription factor. Inactivation of PLAGL2 in CRC cell lines has pronounced effects on ASCL2 reporter activity. Furthermore, ASCL2 expression can partially rescue deficits of proliferation and cell cycle progression caused by depletion of PLAGL2 in CRC cell lines. Thus, the oncogenic effects of PLAGL2 appear to be mediated via core stem cell and onco-fetal pathways, with minimal effects on downstream Wnt signaling.NEW & NOTEWORTHY A Let-7 target called PLAGL2 drives oncogenic transformation via Wnt-independent pathways. This work illustrates the robust effects of this zinc finger transcription factor in colorectal cancer (CRC) cell lines and nontransformed intestinal epithelium, with effects mediated, in part, via the direct target genes ASCL2 and IGF2. This has implications for the role of PLAGL2 in activation of onco-fetal and onco-stem cell pathways, contributing to immature and highly proliferative phenotypes in CRC.


Subject(s)
Colorectal Neoplasms , Humans , Colorectal Neoplasms/metabolism , Cell Line, Tumor , Transcription Factors/genetics , Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , beta Catenin/metabolism , Cell Transformation, Neoplastic/genetics , Wnt Signaling Pathway , Carcinogenesis/genetics , Gene Expression Regulation, Neoplastic , Cell Proliferation/genetics , DNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics
4.
Mol Ther Nucleic Acids ; 29: 979-995, 2022 Sep 13.
Article in English | MEDLINE | ID: mdl-36189080

ABSTRACT

The use of T cells from healthy donors for allogeneic chimeric antigen receptor T (CAR-T) cell cancer therapy is attractive because healthy donor T cells can produce versatile off-the-shelf CAR-T treatments. To maximize safety and durability of allogeneic products, the endogenous T cell receptor and major histocompatibility complex class I molecules are often removed via knockout of T cell receptor beta constant (TRBC) (or T cell receptor alpha constant [TRAC]) and B2M, respectively. However, gene editing tools (e.g., CRISPR-Cas9) can display poor fidelity, which may result in dangerous off-target mutations. Additionally, many gene editing technologies require T cell activation, resulting in a low percentage of desirable stem cell memory T cells (TSCM). We characterize an RNA-guided endonuclease, called Cas-CLOVER, consisting of the Clo051 nuclease domain fused with catalytically dead Cas9. In primary T cells from multiple donors, we find that Cas-CLOVER is a high-fidelity site-specific nuclease, with low off-target activity. Notably, Cas-CLOVER yields efficient multiplexed gene editing in resting T cells. In conjunction with the piggyBac transposon for delivery of a CAR transgene against the B cell maturation antigen (BCMA), we produce allogeneic CAR-T cells composed of high percentages of TSCM cells and possessing potent in vivo anti-tumor cytotoxicity.

6.
J Clin Invest ; 131(1)2021 01 04.
Article in English | MEDLINE | ID: mdl-33445170

ABSTRACT

The RNA-binding protein Apobec1 complementation factor (A1CF) regulates posttranscriptional ApoB mRNA editing, but the range of RNA targets and the long-term effect of altered A1CF expression on liver function are unknown. Here we studied hepatocyte-specific A1cf-transgenic (A1cf+/Tg), A1cf+/Tg Apobec1-/-, and A1cf-/- mice fed chow or high-fat/high-fructose diets using RNA-Seq, RNA CLIP-Seq, and tissue microarrays from human hepatocellular cancer (HCC). A1cf+/Tg mice exhibited increased hepatic proliferation and steatosis, with increased lipogenic gene expression (Mogat1, Mogat2, Cidea, Cd36) associated with shifts in polysomal RNA distribution. Aged A1cf+/Tg mice developed spontaneous fibrosis, dysplasia, and HCC, and this development was accelerated on a high-fat/high-fructose diet and was independent of Apobec1. RNA-Seq revealed increased expression of mRNAs involved in oxidative stress (Gstm3, Gpx3, Cbr3), inflammatory response (Il19, Cxcl14, Tnfα, Ly6c), extracellular matrix organization (Mmp2, Col1a1, Col4a1), and proliferation (Kif20a, Mcm2, Mcm4, Mcm6), and a subset of mRNAs (including Sox4, Sox9, Cdh1) were identified in RNA CLIP-Seq. Increased A1CF expression in human HCC correlated with advanced fibrosis and with reduced survival in a subset with nonalcoholic fatty liver disease. In conclusion, we show that hepatic A1CF overexpression selectively alters polysomal distribution and mRNA expression, promoting lipogenic, proliferative, and inflammatory pathways leading to HCC.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Fatty Liver/metabolism , Liver Cirrhosis/metabolism , Liver Neoplasms/metabolism , Neoplasm Proteins/metabolism , RNA-Binding Proteins/metabolism , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Fatty Liver/genetics , Fatty Liver/pathology , Liver Cirrhosis/genetics , Liver Cirrhosis/pathology , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Mice , Mice, Knockout , Neoplasm Proteins/genetics , RNA-Binding Proteins/genetics
7.
J Pathol ; 247(4): 513-523, 2019 04.
Article in English | MEDLINE | ID: mdl-30511397

ABSTRACT

Chronic inflammation of the gastric mucosa, often caused by autoimmune gastritis and/or infection with Helicobacter pylori, can lead to atrophy of acid-secreting parietal cells with metaplasia of remaining cells. The histological pattern marks a critical step in the progression from chronic gastritis to gastric cancer, yet underlying mechanism(s) of inflammation-induced cell death of gastric epithelial cells are poorly understood. We investigated direct effects of a type 1 cytokine associated with autoimmunity and infection, interferon-γ (IFN-γ), on gastric epithelial cells. IFN-γ was applied to three-dimensional organoid cultures of gastric epithelial cells derived from gastric corpus gland (gastroids) of control and IFN-γ receptor-deficient mice. Gastroids were also treated with supernatants from activated immune cells isolated from a mouse model of autoimmune-mediated atrophic gastritis (TxA23) with and without IFN-γ expression. Finally, histopathological analysis of atrophy and metaplasia severity was performed in TxA23 mice and compared to TxA23 × Ifng-/- mice. Gastric epithelial cells in gastroid cultures expressed IFN-γ receptor in the basolateral membrane, and gastroids died when treated with IFN-γ in an IFN-γ receptor-dependent manner. Supernatants from immune cells containing high levels of IFN-γ were highly toxic to gastroids, and toxicity was tempered when IFN-γ was either neutralized using a monoclonal antibody or when supernatants from Ifng-/- mouse immune cells were used. Finally, TxA23 × Ifng-/- mice showed near-complete abrogation of pre-cancerous histopathological atrophy and metaplasia versus IFN-γ-sufficient controls. We identify IFN-γ as a critical promoter of parietal cell atrophy with metaplasia during the progression of gastritis to gastric atrophy and metaplasia. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Gastric Mucosa/pathology , Interferon-gamma/physiology , Stomach Neoplasms/pathology , Animals , Atrophy/pathology , Cell Death/physiology , Cell Transformation, Neoplastic/pathology , Disease Progression , Epithelial Cells/pathology , Gastritis , Interferon-gamma/deficiency , Interferon-gamma/pharmacology , Metaplasia/pathology , Mice, Inbred BALB C , Mice, Inbred C57BL , Parietal Cells, Gastric/pathology , Tumor Cells, Cultured
8.
RNA ; 25(1): 70-81, 2019 01.
Article in English | MEDLINE | ID: mdl-30309881

ABSTRACT

Mammalian C to U RNA is mediated by APOBEC1, the catalytic deaminase, together with RNA binding cofactors (including A1CF and RBM47) whose relative physiological requirements are unresolved. Although A1CF complements APOBEC1 for in vitro RNA editing, A1cf-/- mice exhibited no change in apolipoproteinB (apoB) RNA editing, while Rbm47 mutant mice exhibited impaired intestinal RNA editing of apoB as well as other targets. Here we examined the role of A1CF and RBM47 in adult mouse liver and intestine, following deletion of either one or both gene products and also following forced (liver or intestinal) transgenic A1CF expression. There were minimal changes in hepatic and intestinal apoB RNA editing in A1cf-/- mice and no changes in either liver- or intestine-specific A1CF transgenic mice. Rbm47 liver-specific knockout (Rbm47LKO ) mice demonstrated reduced editing in a subset (11 of 20) of RNA targets, including apoB. By contrast, apoB RNA editing was virtually eliminated (<6% activity) in intestine-specific (Rbm47IKO ) mice with only five of 53 targets exhibiting C-to-U RNA editing. Double knockout of A1cf and Rbm47 in liver (ARLKO ) eliminated apoB RNA editing and reduced editing in the majority of other targets, with no changes following adenoviral APOBEC1 administration. Intestinal double knockout mice (ARIKO ) demonstrated further reduced editing (<10% activity) in four of five of the residual APOBEC1 targets identified in ARIKO mice. These data suggest that A1CF and RBM47 each function independently, yet interact in a tissue-specific manner, to regulate the activity and site selection of APOBEC1 dependent C-to-U RNA editing.


Subject(s)
Heterogeneous-Nuclear Ribonucleoproteins/metabolism , RNA Editing , RNA-Binding Proteins/metabolism , APOBEC-1 Deaminase/genetics , APOBEC-1 Deaminase/metabolism , Animals , Base Sequence , Gene Knockout Techniques , Heterogeneous-Nuclear Ribonucleoproteins/deficiency , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Intestinal Mucosa/metabolism , Liver/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Organ Specificity , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics
9.
Genes Dev ; 32(15-16): 1020-1034, 2018 08 01.
Article in English | MEDLINE | ID: mdl-30068703

ABSTRACT

RNA-binding proteins (RBPs) are expressed broadly during both development and malignant transformation, yet their mechanistic roles in epithelial homeostasis or as drivers of tumor initiation and progression are incompletely understood. Here we describe a novel interplay between RBPs LIN28B and IMP1 in intestinal epithelial cells. Ribosome profiling and RNA sequencing identified IMP1 as a principle node for gene expression regulation downstream from LIN28B In vitro and in vivo data demonstrate that epithelial IMP1 loss increases expression of WNT target genes and enhances LIN28B-mediated intestinal tumorigenesis, which was reversed when we overexpressed IMP1 independently in vivo. Furthermore, IMP1 loss in wild-type or LIN28B-overexpressing mice enhances the regenerative response to irradiation. Together, our data provide new evidence for the opposing effects of the LIN28B-IMP1 axis on post-transcriptional regulation of canonical WNT signaling, with implications in intestinal homeostasis, regeneration and tumorigenesis.


Subject(s)
Carcinogenesis , Gene Expression Regulation , Intestinal Mucosa/metabolism , RNA-Binding Proteins/metabolism , Regulon , Wnt Signaling Pathway , Animals , Cell Line, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Humans , Intestinal Mucosa/physiology , Mice , Mice, Transgenic , Oncogenes , Protein Biosynthesis , RNA-Binding Proteins/physiology , Regeneration , Stem Cells/metabolism
10.
Stem Cell Reports ; 11(2): 410-424, 2018 08 14.
Article in English | MEDLINE | ID: mdl-30017821

ABSTRACT

Intestinal epithelial stem cell (IESC) fate is promoted by two major transcriptional regulators, the TCF4/ß-catenin complex and ASCL2, which drive expression of IESC-specific factors, including Lgr5, Ephb2, and Rnf43. Canonical Wnt signaling via TCF4/ß-catenin directly transactivates Ascl2, which in turn auto-regulates its own expression. Conversely, Let-7 microRNAs antagonize the IESC lineage by repressing specific mRNA targets. Here, we identify the zinc finger transcription factor PLAGL2 as a Let-7 target that regulates IESC fate. PLAGL2 drives an IESC expression signature, activates Wnt gene expression, and enhances a TCF/LEF reporter in intestinal organoids. In parallel, via cell-autonomous mechanisms, PLAGL2 is required for lineage clonal expansion and directly enhances expression of ASCL2. PLAGL2 also supports enteroid growth and survival in the context of Wnt ligand depletion. PLAGL2 expression is strongly associated with an IESC signature in colorectal cancer and may be responsible for contributing to the aberrant activation of an immature phenotype.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , DNA-Binding Proteins/metabolism , Epithelial Cells/metabolism , Intestinal Mucosa/metabolism , RNA-Binding Proteins/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Transcription Factors/metabolism , Transcriptional Activation , Animals , Biomarkers , Cell Culture Techniques , Cell Line, Tumor , DNA-Binding Proteins/genetics , Humans , Mice , RNA, Small Interfering/genetics , RNA-Binding Proteins/genetics , Signal Transduction , Transcription Factors/genetics
11.
Mol Cancer Res ; 16(3): 403-416, 2018 03.
Article in English | MEDLINE | ID: mdl-29330293

ABSTRACT

The RNA-binding protein LIN28B plays an important role in development, stem cell biology, and tumorigenesis. LIN28B has two isoforms: the LIN28B-long and -short isoforms. Although studies have revealed the functions of the LIN28B-long isoform in tumorigenesis, the role of the LIN28B-short isoform remains unclear and represents a major gap in the field. The LIN28B-long and -short isoforms are expressed in a subset of human colorectal cancers and adjacent normal colonic mucosa, respectively. To elucidate the functional and mechanistic aspects of these isoforms, colorectal cancer cells (Caco-2 and LoVo) were generated to either express no LIN28B or the -short or -long isoform. Interestingly, the long isoform suppressed LET-7 expression and activated canonical RAS/ERK signaling, whereas the short isoform did not. The LIN28B-long isoform-expressing cells demonstrated increased drug resistance to 5-fluorouracil and cisplatin through the upregulation of ERCC1, a DNA repair gene, in a LET-7-dependent manner. The LIN28B-short isoform preserved its ability to bind pre-let-7, without inhibiting the maturation of LET-7, and competed with the LIN28B-long isoform for binding to pre-let-7 Coexpression of the short isoform in the LIN28B-long isoform-expressing cells rescued the phenotypes induced by the LIN28B-long isoform.Implications: This study demonstrates the differential antagonistic functions of the LIN28B-short isoform against the LIN28B-long isoform through an inability to degrade LET-7, which leads to the novel premise that the short isoform may serve to counterbalance the long isoform during normal colonic epithelial homeostasis, but its downregulation during colonic carcinogenesis may reveal the protumorigenic effects of the long isoform. Mol Cancer Res; 16(3); 403-16. ©2018 AACR.


Subject(s)
Colorectal Neoplasms/genetics , MicroRNAs/genetics , RNA-Binding Proteins/genetics , Caco-2 Cells , Cell Line, Tumor , Cisplatin/pharmacology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Drug Resistance, Neoplasm , Endonucleases/genetics , Endonucleases/metabolism , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Fluorouracil/pharmacology , HCT116 Cells , Hep G2 Cells , Humans , MicroRNAs/biosynthesis , MicroRNAs/metabolism , Protein Isoforms , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/metabolism , Signal Transduction , ras Proteins/antagonists & inhibitors , ras Proteins/genetics , ras Proteins/metabolism
12.
Dis Model Mech ; 10(3): 197-214, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28250048

ABSTRACT

MicroRNAs (miRNAs) are small single-stranded RNAs that repress mRNA translation and trigger mRNA degradation. Of the ∼1900 miRNA-encoding genes present in the human genome, ∼250 miRNAs are reported to have changes in abundance or altered functions in colorectal cancer. Thousands of studies have documented aberrant miRNA levels in colorectal cancer, with some miRNAs reported to actively regulate tumorigenesis. A recurrent phenomenon with miRNAs is their frequent participation in feedback loops, which probably serve to reinforce or magnify biological outcomes to manifest a particular cellular phenotype. Here, we review the roles of oncogenic miRNAs (oncomiRs), tumor suppressive miRNAs (anti-oncomiRs) and miRNA regulators in colorectal cancer. Given their stability in patient-derived samples and ease of detection with standard and novel techniques, we also discuss the potential use of miRNAs as biomarkers in the diagnosis of colorectal cancer and as prognostic indicators of this disease. MiRNAs also represent attractive candidates for targeted therapies because their function can be manipulated through the use of synthetic antagonists and miRNA mimics.


Subject(s)
Colorectal Neoplasms/genetics , MicroRNAs/metabolism , Signal Transduction/genetics , Animals , Carcinogenesis/genetics , Carcinogenesis/pathology , Colorectal Neoplasms/pathology , Genetic Pleiotropy , Humans , MicroRNAs/genetics , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology
13.
J Biol Chem ; 292(15): 6148-6162, 2017 04 14.
Article in English | MEDLINE | ID: mdl-28228480

ABSTRACT

The discovery and application of CRISPR/Cas9 technology for genome editing has greatly accelerated targeted mutagenesis in a variety of organisms. CRISPR/Cas9-mediated site-specific cleavage is typically exploited for the generation of insertions or deletions (indels) after aberrant dsDNA repair via the endogenous non-homology end-joining (NHEJ) pathway or, alternatively, for enhancing homology-directed repair to facilitate the generation of a specific mutation (or "knock-in"). However, there is a need for efficient cellular assays that can measure Cas9/guide RNA activity. Reliable methods for enriching and identifying desired mutants are also lacking. Here we describe a method using the Piggybac transposon for stable genomic integration of an H2B-GFP reporter or a hygromycin resistance gene for assaying Cas9 target cleavage and homology-directed repair. The H2B-GFP fusion protein provides increased stability and an obvious pattern of nuclear localization. This method, called SRIRACCHA (i.e. a stable, but reversible, integrated reporter for assaying CRISPR/Cas-stimulated HDR activity), enables the enrichment of mutants via selection of GFP-positive or hygromycin-resistant mammalian cells (immortalized or non-immortalized) as a surrogate for the modification of the endogenous target site. Currently available hyperactive Piggybac transposase mutants allow both delivery and removal of the surrogate reporters, with minimal risk of generating undesirable mutations. This assay permits rapid screening for efficient guide RNAs and the accelerated identification of mutant clones and is applicable to many cell types. We foresee the utility of this approach in contexts in which the maintenance of genomic integrity is essential, for example, when engineering cells for therapeutic purposes.


Subject(s)
CRISPR-Cas Systems , Gene Deletion , Gene Targeting/methods , Genetic Vectors/genetics , Animals , Cell Line, Tumor , Mice
14.
Genes Dev ; 31(2): 154-171, 2017 01 15.
Article in English | MEDLINE | ID: mdl-28174210

ABSTRACT

We hypothesized that basic helix-loop-helix (bHLH) MIST1 (BHLHA15) is a "scaling factor" that universally establishes secretory morphology in cells that perform regulated secretion. Here, we show that targeted deletion of MIST1 caused dismantling of the secretory apparatus of diverse exocrine cells. Parietal cells (PCs), whose function is to pump acid into the stomach, normally lack MIST1 and do not perform regulated secretion. Forced expression of MIST1 in PCs caused them to expand their apical cytoplasm, rearrange mitochondrial/lysosome trafficking, and generate large secretory granules. Mist1 induced a cohort of genes regulated by MIST1 in multiple organs but did not affect PC function. MIST1 bound CATATG/CAGCTG E boxes in the first intron of genes that regulate autophagosome/lysosomal degradation, mitochondrial trafficking, and amino acid metabolism. Similar alterations in cell architecture and gene expression were also caused by ectopically inducing MIST1 in vivo in hepatocytes. Thus, MIST1 is a scaling factor necessary and sufficient by itself to induce and maintain secretory cell architecture. Our results indicate that, whereas mature cell types in each organ may have unique developmental origins, cells performing similar physiological functions throughout the body share similar transcription factor-mediated architectural "blueprints."


Subject(s)
Gene Expression Regulation/genetics , Hepatocyte Growth Factor/genetics , Hepatocyte Growth Factor/metabolism , Parietal Cells, Gastric/cytology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Secretory Pathway/genetics , Acinar Cells/cytology , Acinar Cells/drug effects , Acinar Cells/metabolism , Animals , Antineoplastic Agents, Hormonal/pharmacology , Cell Line , Ectopic Gene Expression/drug effects , Gene Deletion , Gene Expression Regulation/drug effects , Mice , Parietal Cells, Gastric/drug effects , Parietal Cells, Gastric/metabolism , Parietal Cells, Gastric/ultrastructure , Tamoxifen/pharmacology
16.
Biomed Res Int ; 2015: 196082, 2015.
Article in English | MEDLINE | ID: mdl-26539468

ABSTRACT

RNA-binding protein (RBP) is a key player in regulating gene expression at the posttranscriptional level. CLIP-Seq, with the ability to provide a genome-wide map of protein-RNA interactions, has been increasingly used to decipher RBP-mediated posttranscriptional regulation. Generating highly reliable binding sites from CLIP-Seq requires not only stringent library preparation but also considerable computational efforts. Here we presented a first systematic evaluation of major computational steps for identifying RBP binding sites from CLIP-Seq data, including preprocessing, the choice of control samples, peak normalization, and motif discovery. We found that avoiding PCR amplification artifacts, normalizing to input RNA or mRNAseq, and defining the background model from control samples can reduce the bias introduced by RNA abundance and improve the quality of detected binding sites. Our findings can serve as a general guideline for CLIP experiments design and the comprehensive analysis of CLIP-Seq data.


Subject(s)
High-Throughput Nucleotide Sequencing/methods , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , Binding Sites/genetics , Caco-2 Cells , Gene Expression Regulation , Humans , MicroRNAs/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Sequence Analysis, RNA
17.
Cancer Cell ; 28(5): 599-609, 2015 Nov 09.
Article in English | MEDLINE | ID: mdl-26481147

ABSTRACT

A more complete understanding of aberrant oncogenic signaling in neuroblastoma, a malignancy of the developing sympathetic nervous system, is paramount to improving patient outcomes. Recently, we identified LIN28B as an oncogenic driver in high-risk neuroblastoma. Here, we identify the oncogene RAN as a LIN28B target and show regional gain of chromosome 12q24 as an additional somatic alteration resulting in increased RAN expression. We show that LIN28B influences RAN expression by promoting RAN Binding Protein 2 expression and by directly binding RAN mRNA. Further, we demonstrate a convergence of LIN28B and RAN signaling on Aurora kinase A activity. Collectively, these findings demonstrate that LIN28B-RAN-AURKA signaling drives neuroblastoma oncogenesis, suggesting that this pathway may be amenable to therapeutic targeting.


Subject(s)
Aurora Kinase A/genetics , Neuroblastoma/genetics , RNA-Binding Proteins/genetics , Signal Transduction/genetics , ran GTP-Binding Protein/genetics , Aurora Kinase A/metabolism , Blotting, Western , Carcinogenesis/genetics , Cell Line, Tumor , Child , Chromosomes, Human, Pair 12/genetics , DNA Copy Number Variations , Gene Amplification , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , MicroRNAs/genetics , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , N-Myc Proto-Oncogene Protein , Neuroblastoma/metabolism , Neuroblastoma/pathology , Nuclear Pore Complex Proteins/genetics , Nuclear Pore Complex Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , RNA-Binding Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , ran GTP-Binding Protein/metabolism
18.
PLoS Genet ; 11(8): e1005408, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26244988

ABSTRACT

Let-7 miRNAs comprise one of the largest and most highly expressed family of miRNAs among vertebrates, and is critical for promoting differentiation, regulating metabolism, inhibiting cellular proliferation, and repressing carcinogenesis in a variety of tissues. The large size of the Let-7 family of miRNAs has complicated the development of mutant animal models. Here we describe the comprehensive repression of all Let-7 miRNAs in the intestinal epithelium via low-level tissue-specific expression of the Lin28b RNA-binding protein and a conditional knockout of the MirLet7c-2/Mirlet7b locus. This ablation of Let-7 triggers the development of intestinal adenocarcinomas concomitant with reduced survival. Analysis of both mouse and human intestinal cancer specimens reveals that stem cell markers were significantly associated with loss of Let-7 miRNA expression, and that a number of Let-7 targets were elevated, including Hmga1 and Hmga2. Functional studies in 3-D enteroids revealed that Hmga2 is necessary and sufficient to mediate many characteristics of Let-7 depletion, namely accelerating cell cycle progression and enhancing a stem cell phenotype. In addition, inactivation of a single Hmga2 allele in the mouse intestine epithelium significantly represses tumorigenesis driven by Lin28b. In aggregate, we conclude that Let-7 depletion drives a stem cell phenotype and the development of intestinal cancer, primarily via Hmga2.


Subject(s)
Adenocarcinoma/genetics , HMGA2 Protein/metabolism , Intestinal Neoplasms/genetics , MicroRNAs/physiology , Neoplastic Stem Cells/metabolism , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cell Proliferation , Gene Expression Regulation, Neoplastic , HMGA2 Protein/genetics , Humans , Intestinal Neoplasms/metabolism , Intestinal Neoplasms/pathology , Mice, Inbred C57BL , Mice, Knockout , Phenotype , RNA Interference , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Tumor Cells, Cultured
19.
Genes Dev ; 27(20): 2233-45, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-24142874

ABSTRACT

The RNA-binding proteins LIN28A and LIN28B have diverse functions in embryonic stem cells, cellular reprogramming, growth, and oncogenesis. Many of these effects occur via direct inhibition of Let-7 microRNAs (miRNAs), although Let-7-independent effects have been surmised. We report that intestine targeted expression of LIN28B causes intestinal hypertrophy, crypt expansion, and Paneth cell loss. Furthermore, LIN28B fosters intestinal polyp and adenocarcinoma formation. To examine potential Let-7-independent functions of LIN28B, we pursued ribonucleoprotein cross-linking, immunoprecipitation, and high-throughput sequencing (CLIP-seq) to identify direct RNA targets. This revealed that LIN28B bound a substantial number of mRNAs and modestly augmented protein levels of these target mRNAs in vivo. Conversely, Let-7 had a profound effect; modulation of Let-7 levels via deletion of the mirLet7c2/mirLet7b genes recapitulated effects of Lin28b overexpression. Furthermore, intestine-specific Let-7 expression could reverse hypertrophy and Paneth cell depletion caused by Lin28b. This was independent of effects on insulin-PI3K-mTOR signaling. Our study reveals that Let-7 miRNAs are critical for repressing intestinal tissue growth and promoting Paneth cell differentiation. Let-7-dependent effects of LIN28B may supersede Let-7-independent effects on intestinal tissue growth. In summary, LIN28B can definitively act as an oncogene in the absence of canonical genetic alterations.


Subject(s)
Carcinogenesis/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation , Intestinal Mucosa/physiopathology , MicroRNAs/genetics , Animals , Cell Differentiation , DNA-Binding Proteins/metabolism , Intestinal Mucosa/cytology , Intestinal Mucosa/growth & development , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mice , Mice, Inbred BALB C , MicroRNAs/metabolism , Paneth Cells/cytology , Protein Binding , Protein Biosynthesis , RNA, Messenger/metabolism , RNA-Binding Proteins , Signal Transduction
20.
Cancer Biol Ther ; 13(14): 1390-5, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23052130

ABSTRACT

The pathogenesis of sporadic colorectal cancer involves distinct pathways, with characteristic genomic alterations. The first pathway, chromosome instability (CIN), is driven by APC mutations and is typified by Kras mutations, p53 mutation/loss of heterozygosity, and deletions at chromosome 18q. The second pathway is referred to as microsatellite instability (MSI), a genetic hallmark of the accumulated mutations that occur as a consequence of derangements in the mismatch repair genes. Finally, proximal colon cancers may involve methylation of a number of genes, which is frequently referred to as the CpG island methylator phenotype (CIMP), and are associated with B-raf mutations. The ability to stratify colorectal cancers by risk would be facilitated by the identification of polymorphisms that might be utilized as biomarkers. LIN28B is an RNA binding protein that is overexpressed in colon cancers. We find that LIN28B rs314277 is associated with significant recurrence of colorectal cancer in Stage II disease, which may have translational therapeutic implications.


Subject(s)
Colonic Neoplasms/genetics , DNA-Binding Proteins/genetics , Neoplasm Recurrence, Local/genetics , Colonic Neoplasms/mortality , Female , Gene Expression Regulation, Neoplastic , Genetic Association Studies , Genotype , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide , RNA-Binding Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL