Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Nat Rev Mol Cell Biol ; 17(9): 597-604, 2016 09.
Article in English | MEDLINE | ID: mdl-27381243

ABSTRACT

The development of the CRISPR-Cas9 system triggered a revolution in the field of genome engineering. Initially, the use of this system was focused on the study of protein-coding genes but, recently, a number of CRISPR-Cas9-based tools have been developed to study non-coding transcriptional regulatory elements. These technological advances offer unprecedented opportunities for elucidating the functions of enhancers in their endogenous context. Here, we discuss the application, current limitations and future development of CRISPR-Cas9 systems to identify and characterize enhancer elements in a high-throughput manner.


Subject(s)
CRISPR-Cas Systems , Gene Editing/methods , Enhancer Elements, Genetic , Epigenesis, Genetic , Epigenomics/methods
2.
Genome Res ; 34(4): 539-555, 2024 05 15.
Article in English | MEDLINE | ID: mdl-38719469

ABSTRACT

Estrogen Receptor 1 (ESR1; also known as ERα, encoded by ESR1 gene) is the main driver and prime drug target in luminal breast cancer. ESR1 chromatin binding is extensively studied in cell lines and a limited number of human tumors, using consensi of peaks shared among samples. However, little is known about inter-tumor heterogeneity of ESR1 chromatin action, along with its biological implications. Here, we use a large set of ESR1 ChIP-seq data from 70 ESR1+ breast cancers to explore inter-patient heterogeneity in ESR1 DNA binding to reveal a striking inter-tumor heterogeneity of ESR1 action. Of note, commonly shared ESR1 sites show the highest estrogen-driven enhancer activity and are most engaged in long-range chromatin interactions. In addition, the most commonly shared ESR1-occupied enhancers are enriched for breast cancer risk SNP loci. We experimentally confirm SNVs to impact chromatin binding potential for ESR1 and its pioneer factor FOXA1. Finally, in the TCGA breast cancer cohort, we can confirm these variations to associate with differences in expression for the target gene. Cumulatively, we reveal a natural hierarchy of ESR1-chromatin interactions in breast cancers within a highly heterogeneous inter-tumor ESR1 landscape, with the most common shared regions being most active and affected by germline functional risk SNPs for breast cancer development.


Subject(s)
Breast Neoplasms , Chromatin , Enhancer Elements, Genetic , Estrogen Receptor alpha , Hepatocyte Nuclear Factor 3-alpha , Polymorphism, Single Nucleotide , Humans , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , Chromatin/metabolism , Chromatin/genetics , Hepatocyte Nuclear Factor 3-alpha/metabolism , Hepatocyte Nuclear Factor 3-alpha/genetics , Gene Expression Regulation, Neoplastic , Genetic Heterogeneity , Cell Line, Tumor
3.
Nucleic Acids Res ; 47(18): 9557-9572, 2019 10 10.
Article in English | MEDLINE | ID: mdl-31372638

ABSTRACT

Estrogen receptor α (ERα) is an enhancer activating transcription factor, a key driver of breast cancer and a main target for cancer therapy. ERα-mediated gene regulation requires proper chromatin-conformation to facilitate interactions between ERα-bound enhancers and their target promoters. A major determinant of chromatin structure is the CCCTC-binding factor (CTCF), that dimerizes and together with cohesin stabilizes chromatin loops and forms the boundaries of topologically associated domains. However, whether CTCF-binding elements (CBEs) are essential for ERα-driven cell proliferation is unknown. To address this question in a global manner, we implemented a CRISPR-based functional genetic screen targeting CBEs located in the vicinity of ERα-bound enhancers. We identified four functional CBEs and demonstrated the role of one of them in inducing chromatin conformation changes in favor of activation of PREX1, a key ERα target gene in breast cancer. Indeed, high PREX1 expression is a bona-fide marker of ERα-dependency in cell lines, and is associated with good outcome after anti-hormonal treatment. Altogether, our data show that distinct CTCF-mediated chromatin structures are required for ERα- driven breast cancer cell proliferation.


Subject(s)
Breast Neoplasms/genetics , CCCTC-Binding Factor/genetics , Cell Proliferation/genetics , Estrogen Receptor alpha/genetics , Binding Sites/genetics , Breast Neoplasms/pathology , CRISPR-Cas Systems/genetics , Chromatin/genetics , Enhancer Elements, Genetic/genetics , Female , Humans , MCF-7 Cells , Protein Binding/genetics
4.
Nucleic Acids Res ; 46(8): 4213-4227, 2018 05 04.
Article in English | MEDLINE | ID: mdl-29481642

ABSTRACT

Oncogene-induced senescence (OIS), provoked in response to oncogenic activation, is considered an important tumor suppressor mechanism. Long non-coding RNAs (lncRNAs) are transcripts longer than 200 nt without a protein-coding capacity. Functional studies showed that deregulated lncRNA expression promote tumorigenesis and metastasis and that lncRNAs may exhibit tumor-suppressive and oncogenic function. Here, we first identified lncRNAs that were differentially expressed between senescent and non-senescent human fibroblast cells. Using RNA interference, we performed a loss-function screen targeting the differentially expressed lncRNAs, and identified lncRNA-OIS1 (lncRNA#32, AC008063.3 or ENSG00000233397) as a lncRNA required for OIS. Knockdown of lncRNA-OIS1 triggered bypass of senescence, higher proliferation rate, lower abundance of the cell-cycle inhibitor CDKN1A and high expression of cell-cycle-associated genes. Subcellular inspection of lncRNA-OIS1 indicated nuclear and cytosolic localization in both normal culture conditions as well as following oncogene induction. Interestingly, silencing lncRNA-OIS1 diminished the senescent-associated induction of a nearby gene (Dipeptidyl Peptidase 4, DPP4) with established role in tumor suppression. Intriguingly, similar to lncRNA-OIS1, silencing DPP4 caused senescence bypass, and ectopic expression of DPP4 in lncRNA-OIS1 knockdown cells restored the senescent phenotype. Thus, our data indicate that lncRNA-OIS1 links oncogenic induction and senescence with the activation of the tumor suppressor DPP4.


Subject(s)
Cellular Senescence/genetics , Dipeptidyl Peptidase 4/genetics , RNA, Long Noncoding/metabolism , Dipeptidyl Peptidase 4/metabolism , Gene Expression , Genes, ras , Genome , HEK293 Cells , Humans , Neoplasms/genetics , Neoplasms/metabolism
5.
EMBO Rep ; 16(12): 1723-36, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26538417

ABSTRACT

c-Myc is one of the major human proto-oncogenes and is often associated with tumor aggression and poor clinical outcome. Paradoxically, Myc was also reported as a suppressor of cell motility, invasiveness, and metastasis. Among the direct targets of Myc are many components of the protein synthesis machinery whose induction results in an overall increase in protein synthesis that empowers tumor cell growth. At present, it is largely unknown whether beyond the global enhancement of protein synthesis, Myc activation results in translation modulation of specific genes. Here, we measured Myc-induced global changes in gene expression at the transcription, translation, and protein levels and uncovered extensive transcript-specific regulation of protein translation. Particularly, we detected a broad coordination between regulation of transcription and translation upon modulation of Myc activity and showed the connection of these responses to mTOR signaling to enhance oncogenic transformation and to the TGFß pathway to modulate cell migration and invasiveness. Our results elucidate novel facets of Myc-induced cellular responses and provide a more comprehensive view of the consequences of its activation in cancer cells.


Subject(s)
Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic , Neoplasm Invasiveness , Neoplasm Metastasis/genetics , Neoplasms/genetics , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Apoptosis , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Gene Expression Profiling , Genes, myc , Humans , Protein Biosynthesis , Signal Transduction , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
6.
bioRxiv ; 2023 Nov 02.
Article in English | MEDLINE | ID: mdl-37961147

ABSTRACT

Estrogen Receptor alpha (ERα) is the main driver and prime drug target in luminal breast. ERα chromatin binding is extensively studied in cell lines and a limited number of human tumors, using consensi of peaks shared among samples. However, little is known about inter-tumor heterogeneity of ERα chromatin action, along with its biological implications. Here, we use a large set of ERα ChIP-seq data from 70 ERα+ breast cancers to explore inter-patient heterogeneity in ERα DNA binding, to reveal a striking inter-tumor heterogeneity of ERα action. Interestingly, commonly-shared ERα sites showed the highest estrogen-driven enhancer activity and were most-engaged in long-range chromatin interactions. In addition, the most-commonly shared ERα-occupied enhancers were enriched for breast cancer risk SNP loci. We experimentally confirm SNVs to impact chromatin binding potential for ERα and its pioneer factor FOXA1. Finally, in the TCGA breast cancer cohort, we could confirm these variations to associate with differences in expression for the target gene. Cumulatively, we reveal a natural hierarchy of ERα-chromatin interactions in breast cancers within a highly heterogeneous inter-tumor ERα landscape, with the most-common shared regions being most active and affected by germline functional risk SNPs for breast cancer development.

7.
Planta ; 236(4): 1081-92, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22569921

ABSTRACT

An autophagy-related gene Atg8 was cloned for the first time from wild emmer wheat, named as TdAtg8, and its role on autophagy under abiotic stress conditions was investigated. Examination of TdAtg8 expression patterns indicated that Atg8 expression was strongly upregulated under drought stress, especially in the roots when compared to leaves. LysoTracker(®) red marker, utilized to observe autophagosomes, revealed that autophagy is constitutively active in Triticum dicoccoides. Moreover, autophagy was determined to be induced in plants exposed to osmotic stress when compared to plants grown under normal conditions. Functional studies were executed in yeast to confirm that the TdATG8 protein is functional, and showed that the TdAtg8 gene complements the atg8∆::kan MX yeast mutant strain grown under nitrogen deficiency. For further functional analysis, TdATG8 protein was expressed in yeast and analyzed using Western immunoblotting. Atg8-silenced plants were exposed to drought stress and chlorophyll and malondialdehyde (MDA) content measurements demonstrated that Atg8 plays a key role on drought stress tolerance. In addition, Atg8-silenced plants exposed to osmotic stress were found to have decreased Atg8 expression level in comparison to controls. Hence, Atg8 is a positive regulator in osmotic and drought stress response.


Subject(s)
Autophagy/genetics , Gene Expression Regulation, Plant/genetics , Stress, Physiological/genetics , Triticum/genetics , Amino Acid Sequence , Chromosome Mapping , Droughts , Gene Expression Profiling , Gene Silencing , Genes, Plant/genetics , Genetic Complementation Test , Malondialdehyde/analysis , Malondialdehyde/metabolism , Molecular Sequence Data , Mutation , Organ Specificity , Osmosis/physiology , Plant Leaves/cytology , Plant Leaves/genetics , Plant Leaves/metabolism , Plant Leaves/physiology , Plant Roots/cytology , Plant Roots/genetics , Plant Roots/metabolism , Plant Roots/physiology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/physiology , Sequence Alignment , Triticum/cytology , Triticum/metabolism , Triticum/physiology , Up-Regulation/genetics
8.
Cancers (Basel) ; 14(4)2022 Feb 17.
Article in English | MEDLINE | ID: mdl-35205770

ABSTRACT

Intratumor heterogeneity of breast cancer is driven by extrinsic factors from the tumor microenvironment (TME) as well as tumor cell-intrinsic parameters including genetic, epigenetic, and transcriptomic traits. The extracellular matrix (ECM), a major structural component of the TME, impacts every stage of tumorigenesis by providing necessary biochemical and biomechanical cues that are major regulators of cell shape/architecture, stiffness, cell proliferation, survival, invasion, and migration. Moreover, ECM and tissue architecture have a profound impact on chromatin structure, thereby altering gene expression. Considering the significant contribution of ECM to cellular behavior, a large body of work underlined that traditional two-dimensional (2D) cultures depriving cell-cell and cell-ECM interactions as well as spatial cellular distribution and organization of solid tumors fail to recapitulate in vivo properties of tumor cells residing in the complex TME. Thus, three-dimensional (3D) culture models are increasingly employed in cancer research, as these culture systems better mimic the physiological microenvironment and shape the cellular responses according to the microenvironmental cues that will regulate critical cell functions such as cell shape/architecture, survival, proliferation, differentiation, and drug response as well as gene expression. Therefore, 3D cell culture models that better resemble the patient transcriptome are critical in defining physiologically relevant transcriptional changes. This review will present the transcriptional factor (TF) repertoire of breast cancer in 3D culture models in the context of mammary tissue architecture, epithelial-to-mesenchymal transition and metastasis, cell death mechanisms, cancer therapy resistance and differential drug response, and stemness and will discuss the impact of culture dimensionality on breast cancer research.

9.
Chronobiol Int ; 39(4): 590-601, 2022 04.
Article in English | MEDLINE | ID: mdl-34906014

ABSTRACT

This study examines the relationship between high school students' age, gender, personality, computer game addiction, chronotype, and sleep quality using structural equation modelling. For this purpose, the study was planned according to the correlational research design, one of the most common quantitative research methods. The sample of the study consisted of 922 students who accepted to participate and completed the scales. Of the 922 high school students in the sample, 528 were girls, and 394 were boys. In the study, the Computer Game Addiction Scale for Adolescents, Sleep Quality Scale, Composite Scale of Morningness (CSM), and Big Five Inventory were used to measure the variables. Among the variables involved, personality traits, such as consciousness, neuroticism, and openness to experience, were significantly related with morningness-eveningness. Besides, gender and being evening or morning types were found to be significantly related with game addiction. However, sleep quality was predicted by computer game addiction and morningness-eveningness. It was found that the fit indices of the model have an acceptable and good fit in explaining the variables.


Subject(s)
Sleep Quality , Video Games , Adolescent , Circadian Rhythm , Female , Humans , Latent Class Analysis , Male , Personality , Sleep , Students , Surveys and Questionnaires
10.
Genome Biol ; 22(1): 54, 2021 01 29.
Article in English | MEDLINE | ID: mdl-33514403

ABSTRACT

BACKGROUND: Frequent activation of the co-transcriptional factor YAP is observed in a large number of solid tumors. Activated YAP associates with enhancer loci via TEAD4-DNA-binding protein and stimulates cancer aggressiveness. Although thousands of YAP/TEAD4 binding-sites are annotated, their functional importance is unknown. Here, we aim at further identification of enhancer elements that are required for YAP functions. RESULTS: We first apply genome-wide ChIP profiling of YAP to systematically identify enhancers that are bound by YAP/TEAD4. Next, we implement a genetic approach to uncover functions of YAP/TEAD4-associated enhancers, demonstrate its robustness, and use it to reveal a network of enhancers required for YAP-mediated proliferation. We focus on EnhancerTRAM2, as its target gene TRAM2 shows the strongest expression-correlation with YAP activity in nearly all tumor types. Interestingly, TRAM2 phenocopies the YAP-induced cell proliferation, migration, and invasion phenotypes and correlates with poor patient survival. Mechanistically, we identify FSTL-1 as a major direct client of TRAM2 that is involved in these phenotypes. Thus, TRAM2 is a key novel mediator of YAP-induced oncogenic proliferation and cellular invasiveness. CONCLUSIONS: YAP is a transcription co-factor that binds to thousands of enhancer loci and stimulates tumor aggressiveness. Using unbiased functional approaches, we dissect YAP enhancer network and characterize TRAM2 as a novel mediator of cellular proliferation, migration, and invasion. Our findings elucidate how YAP induces cancer aggressiveness and may assist diagnosis of cancer metastasis.


Subject(s)
Carcinogenesis/genetics , Enhancer Elements, Genetic , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Animals , Binding Sites , Cell Line, Tumor , Cell Movement , Cell Proliferation , DNA-Binding Proteins/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Membrane Glycoproteins/chemistry , Mice , Mice, Inbred NOD , Mice, SCID , TEA Domain Transcription Factors/genetics , TEA Domain Transcription Factors/metabolism , Transcription Factors/metabolism , Transcriptome
11.
Cancer Lett ; 436: 87-95, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30145202

ABSTRACT

Breast cancer is the most prevalent type of malignancy in women with ∼1.7 million new cases diagnosed annually, of which the majority express ERα (ESR1), a ligand-dependent transcription factor. Genome-wide chromatin binding maps suggest that ERα may control the expression of thousands of genes, posing a great challenge in identifying functional targets. Recently, we developed a CRISPR-Cas9 functional genetic screening approach to identify enhancers required for ERα-positive breast cancer cell proliferation. We validated several candidates, including CUTE, a putative ERα-responsive enhancer located in the first intron of CUEDC1 (CUE-domain containing protein). Here, we show that CUTE controls CUEDC1 expression, and that this interaction is essential for ERα-mediated cell proliferation. Moreover, ectopic expression of CUEDC1, but not a CUE-domain mutant, rescues the defects in CUTE activity. Finally, CUEDC1 expression correlates positively with ERα in breast cancer. Thus, CUEDC1 is a functional target gene of ERα and is required for breast cancer cell proliferation.


Subject(s)
Breast Neoplasms/genetics , Cell Proliferation/genetics , Estrogen Receptor alpha/genetics , Gene Expression Regulation, Neoplastic , Intracellular Signaling Peptides and Proteins/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , CRISPR-Cas Systems , Cell Line, Tumor , Enhancer Elements, Genetic/genetics , Estrogen Receptor alpha/metabolism , Female , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/metabolism , MCF-7 Cells
12.
Methods Mol Biol ; 1543: 45-55, 2017.
Article in English | MEDLINE | ID: mdl-28349421

ABSTRACT

The advent of next-generation sequencing (NGS) technologies has revolutionized the way we do research on gene expression. High-throughput transcriptomics became possible with the development of microarray technology, but its widespread application only occurred after the emergence of massive parallel sequencing. Especially, RNA sequencing (RNA-seq) has greatly increased our knowledge about the genome and led to the identification and annotation of novel classes of RNAs in different species. However, RNA-seq measures the steady-state level of a given RNA, which is the equilibrium between transcription, processing, and degradation. In recent years, a number of dedicated RNA-seq technologies were developed to measure specifically transcription events. Global run-on sequencing (GRO-seq) is the most widely used method to measure nascent RNA, and in recent years, it has been applied successfully to study the function and mechanism of action of noncoding RNAs. Here, we describe a detailed protocol of GRO-seq that can be readily applied to investigate different aspects of RNA biology in human cells.


Subject(s)
Computational Biology/methods , Gene Expression Profiling/methods , Gene Expression Regulation , Software , Transcription, Genetic , Gene Library , High-Throughput Nucleotide Sequencing , Reverse Transcriptase Polymerase Chain Reaction , Transcriptome
13.
Nat Biotechnol ; 34(2): 192-8, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26751173

ABSTRACT

Systematic identification of noncoding regulatory elements has, to date, mainly relied on large-scale reporter assays that do not reproduce endogenous conditions. We present two distinct CRISPR-Cas9 genetic screens to identify and characterize functional enhancers in their native context. Our strategy is to target Cas9 to transcription factor binding sites in enhancer regions. We identified several functional enhancer elements and characterized the role of two of them in mediating p53 (TP53) and ERα (ESR1) gene regulation. Moreover, we show that a genomic CRISPR-Cas9 tiling screen can precisely map functional domains within enhancer elements. Our approach expands the utility of CRISPR-Cas9 to elucidate the functions of the noncoding genome.


Subject(s)
CRISPR-Cas Systems/genetics , Enhancer Elements, Genetic/genetics , Genetic Engineering/methods , Genome, Human/genetics , Genomics/methods , Animals , Cell Line , Gene Knockout Techniques , Humans , MCF-7 Cells , Mice
14.
Nat Commun ; 6: 6520, 2015 Mar 27.
Article in English | MEDLINE | ID: mdl-25813522

ABSTRACT

p53 binds enhancers to regulate key target genes. Here, we globally mapped p53-regulated enhancers by looking at enhancer RNA (eRNA) production. Intriguingly, while many p53-induced enhancers contained p53-binding sites, most did not. As long non-coding RNAs (lncRNAs) are prominent regulators of chromatin dynamics, we hypothesized that p53-induced lncRNAs contribute to the activation of enhancers by p53. Among p53-induced lncRNAs, we identified LED and demonstrate that its suppression attenuates p53 function. Chromatin-binding and eRNA expression analyses show that LED associates with and activates strong enhancers. One prominent target of LED was located at an enhancer region within CDKN1A gene, a potent p53-responsive cell cycle inhibitor. LED knockdown reduces CDKN1A enhancer induction and activity, and cell cycle arrest following p53 activation. Finally, promoter-associated hypermethylation analysis shows silencing of LED in human tumours. Thus, our study identifies a new layer of complexity in the p53 pathway and suggests its dysregulation in cancer.


Subject(s)
Adenocarcinoma/genetics , Breast Neoplasms/genetics , DNA Methylation/genetics , Gene Expression Regulation, Neoplastic , RNA, Long Noncoding/genetics , Tumor Suppressor Protein p53/genetics , Chromatin Immunoprecipitation , Cyclin-Dependent Kinase Inhibitor p21/genetics , Enhancer Elements, Genetic , Female , Humans , In Situ Hybridization, Fluorescence , MCF-7 Cells , Promoter Regions, Genetic , Real-Time Polymerase Chain Reaction , Sequence Analysis, RNA
15.
Eur J Obstet Gynecol Reprod Biol ; 178: 74-9, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24792541

ABSTRACT

OBJECTIVE: The aim of this study is to evaluate the effects of 2-aminoethoxydiphenyl borate (2-APB) as an antioxidant and analyze biochemical and histopathologic changes in experimental ischemia-reperfusion (I/R) injury in rat ovaries. STUDY DESIGN: Thirty female rats were utilized to create four groups. Group 1: I/R and 2-APB (2mg/kg); Group 2: I/R and 2-APB (4mg/kg); Group 3: I/R; Group 4: sham operation. Ovarian tissue and serum malondialdehyde, nitric oxide (NO) levels; ovarian tissue and serum total antioxidant status (TAS), total oxidant status (TOS), oxidative stress index (OSI) were determined. In ovarian tissue samples histopathologic examination, immunoflourescence staining by TUNEL method was studied. RESULTS: Tissue TOS, serum TOS, and OSI levels were elevated in I/R group. After treatment with 2-APB, tissue and serum TOS levels and OSI levels were markedly decreased. There was a significant difference in terms of tissue and serum NO levels between the sham group and I/R group. Elevation in tissue NO and serum NO levels were decreased after treatment with 2-APB. TUNEL-positive cell number gradually decreased with dose of 2-APB in groups 1 and 2. CONCLUSION: Conservative treatment with 2-APB is beneficial for mitigation of I/R injury, and the ovarian protective effect of 2-APB appears to be mediated through its antiapopitotic and antioxidative effects.


Subject(s)
Antioxidants/therapeutic use , Boron Compounds/therapeutic use , Ovarian Diseases/drug therapy , Reperfusion Injury/drug therapy , Animals , Apoptosis , Female , Nitric Oxide/metabolism , Ovarian Diseases/pathology , Ovary/metabolism , Ovary/pathology , Oxidative Stress/drug effects , Rats, Wistar , Reperfusion Injury/pathology , Torsion, Mechanical
16.
Autophagy ; 9(3): 374-85, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23322078

ABSTRACT

Macroautophagy (autophagy herein) is a cellular catabolic mechanism activated in response to stress conditions including starvation, hypoxia and misfolded protein accumulation. Abnormalities in autophagy were associated with pathologies including cancer and neurodegenerative diseases. Hence, elucidation of the signaling pathways controlling autophagy is of utmost importance. Recently we and others described microRNAs (miRNAs) as novel and potent modulators of the autophagic activity. Here, we describe MIR181A (hsa-miR-181a-1) as a new autophagy-regulating miRNA. We showed that overexpression of MIR181A resulted in the attenuation of starvation- and rapamycin-induced autophagy in MCF-7, Huh-7 and K562 cells. Moreover, antagomir-mediated inactivation of endogenous miRNA activity stimulated autophagy. We identified ATG5 as an MIR181A target. Indeed, ATG5 cellular levels were decreased in cells upon MIR181A overexpression and increased following the introduction of antagomirs. More importantly, overexpression of ATG5 from a miRNA-insensitive cDNA construct rescued autophagic activity in the presence of MIR181A. We also showed that the ATG5 3' UTR contained functional MIR181A responsive sequences sensitive to point mutations. Therefore, MIR181A is a novel and important regulator of autophagy and ATG5 is a rate-limiting miRNA target in this effect.


Subject(s)
Autophagy , Gene Expression Regulation, Neoplastic , MicroRNAs/metabolism , Microtubule-Associated Proteins/metabolism , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/metabolism , Autophagy-Related Protein 5 , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Humans , K562 Cells , Lysosomes/metabolism , MCF-7 Cells , Plasmids/metabolism , Point Mutation , RNA, Messenger/metabolism , Signal Transduction
17.
PLoS One ; 8(12): e82556, 2013.
Article in English | MEDLINE | ID: mdl-24358205

ABSTRACT

BACKGROUND: Autophagy is a vesicular trafficking process responsible for the degradation of long-lived, misfolded or abnormal proteins, as well as damaged or surplus organelles. Abnormalities of the autophagic activity may result in the accumulation of protein aggregates, organelle dysfunction, and autophagy disorders were associated with various diseases. Hence, mechanisms of autophagy regulation are under exploration. METHODS: Over-expression of hsa-miR-376a1 (shortly MIR376A) was performed to evaluate its effects on autophagy. Autophagy-related targets of the miRNA were predicted using Microcosm Targets and MIRanda bioinformatics tools and experimentally validated. Endogenous miRNA was blocked using antagomirs and the effects on target expression and autophagy were analyzed. Luciferase tests were performed to confirm that 3' UTR sequences in target genes were functional. Differential expression of MIR376A and the related MIR376B was compared using TaqMan quantitative PCR. RESULTS: Here, we demonstrated that, a microRNA (miRNA) from the DLK1/GTL2 gene cluster, MIR376A, played an important role in autophagy regulation. We showed that, amino acid and serum starvation-induced autophagy was blocked by MIR376A overexpression in MCF-7 and Huh7 cells. MIR376A shared the same seed sequence and had overlapping targets with MIR376B, and similarly blocked the expression of key autophagy proteins ATG4C and BECN1 (Beclin 1). Indeed, 3' UTR sequences in the mRNA of these autophagy proteins were responsive to MIR376A in luciferase assays. Antagomir tests showed that, endogenous MIR376A was participating to the control of ATG4C and BECN1 transcript and protein levels. Moreover, blockage of endogenous MIR376A accelerated starvation-induced autophagic activity. Interestingly, MIR376A and MIR376B levels were increased with different kinetics in response to starvation stress and tissue-specific level differences were also observed, pointing out to an overlapping but miRNA-specific biological role. CONCLUSIONS: Our findings underline the importance of miRNAs encoded by the DLK1/GTL2 gene cluster in stress-response control mechanisms, and introduce MIR376A as a new regulator of autophagy.


Subject(s)
Autophagy/genetics , MicroRNAs/genetics , Starvation/genetics , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Autophagy-Related Proteins , Beclin-1 , Cell Line, Tumor , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/metabolism , HEK293 Cells , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , MicroRNAs/metabolism
18.
Autophagy ; 8(2): 165-76, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22248718

ABSTRACT

Macroautophagy (autophagy) is the major intracellular degradation pathway for long-lived proteins and organelles. It helps the cell to survive a spectrum of stressful conditions including starvation, growth factor deprivation and misfolded protein accumulation. Moreover, abnormalities of autophagy play a role in major health problems including cancer and neurodegenerative diseases. Yet, mechanisms controlling autophagic activity are not fully understood. Here, we describe hsa-miR-376b (miR-376b) as a new microRNA (miRNA) regulating autophagy. We showed that miR-376b expression attenuated starvation- and rapamycin-induced autophagy in MCF-7 and Huh-7 cells. We discovered autophagy proteins ATG4C and BECN1 (Beclin 1) as cellular targets of miR-376b. Indeed, upon miRNA overexpression, both mRNA and protein levels of ATG4C and BECN1 were decreased. miR-376b target sequences were present in the 3' UTR of ATG4C and BECN1 mRNAs and introduction of mutations abolished their miR-376b responsiveness. Antagomir-mediated inactivation of the endogenous miR-376b led to an increase in ATG4C and BECN1 levels. Therefore, miR-376b controls autophagy by directly regulating intracellular levels of two key autophagy proteins, ATG4C and BECN1.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Autophagy , Cysteine Endopeptidases/metabolism , Membrane Proteins/metabolism , MicroRNAs/metabolism , TOR Serine-Threonine Kinases/antagonists & inhibitors , 3' Untranslated Regions/genetics , Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/genetics , Autophagy/drug effects , Autophagy-Related Proteins , Base Sequence , Beclin-1 , Cell Line, Tumor , Cysteine Endopeptidases/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Membrane Proteins/genetics , MicroRNAs/genetics , Models, Biological , Molecular Sequence Data , Oligoribonucleotides/pharmacology , Proteolysis/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Response Elements/genetics , Sequestosome-1 Protein , Sirolimus/pharmacology , Starvation , TOR Serine-Threonine Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL