Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.015
Filter
Add more filters

Publication year range
1.
Cell ; 185(10): 1728-1744.e16, 2022 05 12.
Article in English | MEDLINE | ID: mdl-35460644

ABSTRACT

As the emerging variants of SARS-CoV-2 continue to drive the worldwide pandemic, there is a constant demand for vaccines that offer more effective and broad-spectrum protection. Here, we report a circular RNA (circRNA) vaccine that elicited potent neutralizing antibodies and T cell responses by expressing the trimeric RBD of the spike protein, providing robust protection against SARS-CoV-2 in both mice and rhesus macaques. Notably, the circRNA vaccine enabled higher and more durable antigen production than the 1mΨ-modified mRNA vaccine and elicited a higher proportion of neutralizing antibodies and distinct Th1-skewed immune responses. Importantly, we found that the circRNARBD-Omicron vaccine induced effective neutralizing antibodies against the Omicron but not the Delta variant. In contrast, the circRNARBD-Delta vaccine protected against both Delta and Omicron or functioned as a booster after two doses of either native- or Delta-specific vaccination, making it a favorable choice against the current variants of concern (VOCs) of SARS-CoV-2.


Subject(s)
COVID-19 , SARS-CoV-2 , Animals , Antibodies, Neutralizing , Antibodies, Viral , COVID-19/prevention & control , COVID-19 Vaccines , Humans , Macaca mulatta , Mice , RNA, Circular/genetics , SARS-CoV-2/genetics , Vaccines, Synthetic/genetics , mRNA Vaccines
2.
Annu Rev Cell Dev Biol ; 38: 263-289, 2022 10 06.
Article in English | MEDLINE | ID: mdl-35609906

ABSTRACT

Covalently closed, single-stranded circular RNAs can be produced from viral RNA genomes as well as from the processing of cellular housekeeping noncoding RNAs and precursor messenger RNAs. Recent transcriptomic studies have surprisingly uncovered that many protein-coding genes can be subjected to backsplicing, leading to widespread expression of a specific type of circular RNAs (circRNAs) in eukaryotic cells. Here, we discuss experimental strategies used to discover and characterize diverse circRNAs at both the genome and individual gene scales. We further highlight the current understanding of how circRNAs are generated and how the mature transcripts function. Some circRNAs act as noncoding RNAs to impact gene regulation by serving as decoys or competitors for microRNAs and proteins. Others form extensive networks of ribonucleoprotein complexes or encode functional peptides that are translated in response to certain cellular stresses. Overall, circRNAs have emerged as an important class of RNAmolecules in gene expression regulation that impact many physiological processes, including early development, immune responses, neurogenesis, and tumorigenesis.


Subject(s)
MicroRNAs , RNA, Circular , Gene Expression Regulation/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , RNA/genetics , RNA/metabolism , RNA, Circular/genetics , RNA, Untranslated , RNA, Viral , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism
3.
Cell ; 183(1): 76-93.e22, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32931733

ABSTRACT

Mitochondria, which play central roles in immunometabolic diseases, have their own genome. However, the functions of mitochondria-located noncoding RNAs are largely unknown due to the absence of a specific delivery system. By circular RNA (circRNA) expression profile analysis of liver fibroblasts from patients with nonalcoholic steatohepatitis (NASH), we observe that mitochondrial circRNAs account for a considerable fraction of downregulated circRNAs in NASH fibroblasts. By constructing mitochondria-targeting nanoparticles, we observe that Steatohepatitis-associated circRNA ATP5B Regulator (SCAR), which is located in mitochondria, inhibits mitochondrial ROS (mROS) output and fibroblast activation. circRNA SCAR, mediated by PGC-1α, binds to ATP5B and shuts down mPTP by blocking CypD-mPTP interaction. Lipid overload inhibits PGC-1α by endoplasmic reticulum (ER) stress-induced CHOP. In vivo, targeting circRNA SCAR alleviates high fat diet-induced cirrhosis and insulin resistance. Clinically, circRNA SCAR is associated with steatosis-to-NASH progression. Collectively, we identify a mitochondrial circRNA that drives metaflammation and serves as a therapeutic target for NASH.


Subject(s)
Mitochondria/genetics , Mitochondrial Proton-Translocating ATPases/genetics , RNA, Circular/genetics , Animals , Cell Line , Diet, High-Fat , Endoplasmic Reticulum Stress/physiology , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression/genetics , Humans , Insulin Resistance , Liver/pathology , Liver Cirrhosis/metabolism , Male , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Mitochondrial Permeability Transition Pore/metabolism , Mitochondrial Proton-Translocating ATPases/metabolism , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , RNA, Circular/metabolism , Reactive Oxygen Species , Transcriptome/genetics
4.
Cell ; 177(4): 865-880.e21, 2019 05 02.
Article in English | MEDLINE | ID: mdl-31031002

ABSTRACT

Circular RNAs (circRNAs) produced from back-splicing of exons of pre-mRNAs are widely expressed, but current understanding of their functions is limited. These RNAs are stable in general and are thought to have unique structural conformations distinct from their linear RNA cognates. Here, we show that endogenous circRNAs tend to form 16-26 bp imperfect RNA duplexes and act as inhibitors of double-stranded RNA (dsRNA)-activated protein kinase (PKR) related to innate immunity. Upon poly(I:C) stimulation or viral infection, circRNAs are globally degraded by RNase L, a process required for PKR activation in early cellular innate immune responses. Augmented PKR phosphorylation and circRNA reduction are found in peripheral blood mononuclear cells (PBMCs) derived from patients with autoimmune disease systemic lupus erythematosus (SLE). Importantly, overexpression of the dsRNA-containing circRNA in PBMCs or T cells derived from SLE can alleviate the aberrant PKR activation cascade, thus providing a connection between circRNAs and SLE.


Subject(s)
RNA, Circular/metabolism , RNA, Circular/physiology , eIF-2 Kinase/metabolism , Adolescent , Adult , Autoimmune Diseases/genetics , Cell Line , Endoribonucleases/metabolism , Female , Humans , Immunity, Innate/genetics , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Lupus Erythematosus, Systemic/genetics , Middle Aged , Phosphorylation , RNA/metabolism , RNA Splicing/genetics , RNA Stability/physiology , RNA, Circular/genetics , RNA, Double-Stranded/metabolism , Virus Diseases/metabolism , eIF-2 Kinase/immunology
5.
Cell ; 178(1): 242-260.e29, 2019 06 27.
Article in English | MEDLINE | ID: mdl-31155234

ABSTRACT

Gene expression in human tissue has primarily been studied on the transcriptional level, largely neglecting translational regulation. Here, we analyze the translatomes of 80 human hearts to identify new translation events and quantify the effect of translational regulation. We show extensive translational control of cardiac gene expression, which is orchestrated in a process-specific manner. Translation downstream of predicted disease-causing protein-truncating variants appears to be frequent, suggesting inefficient translation termination. We identify hundreds of previously undetected microproteins, expressed from lncRNAs and circRNAs, for which we validate the protein products in vivo. The translation of microproteins is not restricted to the heart and prominent in the translatomes of human kidney and liver. We associate these microproteins with diverse cellular processes and compartments and find that many locate to the mitochondria. Importantly, dozens of microproteins are translated from lncRNAs with well-characterized noncoding functions, indicating previously unrecognized biology.


Subject(s)
Myocardium/metabolism , Protein Biosynthesis , Adolescent , Adult , Aged , Animals , Codon/genetics , Female , Gene Expression Regulation , HEK293 Cells , Humans , Infant , Male , Mice , Mice, Inbred C57BL , Middle Aged , Open Reading Frames/genetics , RNA, Circular/genetics , RNA, Circular/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Ribosomes/genetics , Ribosomes/metabolism , Young Adult
6.
Annu Rev Cell Dev Biol ; 35: 407-431, 2019 10 06.
Article in English | MEDLINE | ID: mdl-31403819

ABSTRACT

A large and significant portion of eukaryotic transcriptomes consists of noncoding RNAs (ncRNAs) that have minimal or no protein-coding capacity but are functional. Diverse ncRNAs, including both small RNAs and long ncRNAs (lncRNAs), play essential regulatory roles in almost all biological processes by modulating gene expression at the transcriptional and posttranscriptional levels. In this review, we summarize the current knowledge of plant small RNAs and lncRNAs, with a focus on their biogenesis, modes of action, local and systemic movement, and functions at the nexus of plant development and environmental responses. The complex connections among small RNAs, lncRNAs, and small peptides in plants are also discussed, along with the challenges of identifying and investigating new classes of ncRNAs.


Subject(s)
Plant Development/genetics , Plants/genetics , RNA, Long Noncoding/metabolism , RNA, Plant/metabolism , Stress, Physiological/genetics , Gene Expression Regulation, Plant/genetics , Gene Expression Regulation, Plant/physiology , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Circular/genetics , RNA, Circular/metabolism , RNA, Long Noncoding/genetics , RNA, Plant/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism
7.
Nat Rev Mol Cell Biol ; 21(8): 475-490, 2020 08.
Article in English | MEDLINE | ID: mdl-32366901

ABSTRACT

Many protein-coding genes in higher eukaryotes can produce circular RNAs (circRNAs) through back-splicing of exons. CircRNAs differ from mRNAs in their production, structure and turnover and thereby have unique cellular functions and potential biomedical applications. In this Review, I discuss recent progress in our understanding of the biogenesis of circRNAs and the regulation of their abundance and of their biological functions, including in transcription and splicing, sequestering or scaffolding of macromolecules to interfere with microRNA activities or signalling pathways, and serving as templates for translation. I further discuss the emerging roles of circRNAs in regulating immune responses and cell proliferation, and the possibilities of applying circRNA technologies in biomedical research.


Subject(s)
RNA, Circular/genetics , RNA, Circular/metabolism , RNA, Circular/physiology , Alternative Splicing/genetics , Animals , Exons/genetics , Gene Expression/genetics , Gene Expression Regulation/genetics , Humans , MicroRNAs/metabolism , RNA/genetics , RNA Splicing/genetics , RNA, Messenger/metabolism
8.
Mol Cell ; 84(11): 2007-2008, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38848687

ABSTRACT

We talk to first author Laura Amaya about her paper "Pathways for macrophage uptake of cell-free circular RNAs" (in this issue of Molecular Cell), her path to becoming a scientist, and some of the lessons she's learned along the way.


Subject(s)
RNA, Circular , History, 21st Century , History, 20th Century , Humans , RNA, Circular/genetics , RNA, Circular/metabolism , Macrophages/metabolism , Animals
9.
Mol Cell ; 84(12): 2304-2319.e8, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38838666

ABSTRACT

Circular RNAs (circRNAs) are upregulated during neurogenesis. Where and how circRNAs are localized and what roles they play during this process have remained elusive. Comparing the nuclear and cytoplasmic circRNAs between H9 cells and H9-derived forebrain (FB) neurons, we identify that a subset of adenosine (A)-rich circRNAs are restricted in H9 nuclei but exported to cytosols upon differentiation. Such a subcellular relocation of circRNAs is modulated by the poly(A)-binding protein PABPC1. In the H9 nucleus, newly produced (A)-rich circRNAs are bound by PABPC1 and trapped by the nuclear basket protein TPR to prevent their export. Modulating (A)-rich motifs in circRNAs alters their subcellular localization, and introducing (A)-rich circRNAs in H9 cytosols results in mRNA translation suppression. Moreover, decreased nuclear PABPC1 upon neuronal differentiation enables the export of (A)-rich circRNAs, including circRTN4(2,3), which is required for neurite outgrowth. These findings uncover subcellular localization features of circRNAs, linking their processing and function during neurogenesis.


Subject(s)
Active Transport, Cell Nucleus , Adenosine , Cell Nucleus , Neurogenesis , Neurons , Poly(A)-Binding Protein I , RNA, Circular , RNA , RNA, Circular/metabolism , RNA, Circular/genetics , Neurons/metabolism , Adenosine/metabolism , Cell Nucleus/metabolism , Humans , Poly(A)-Binding Protein I/metabolism , Poly(A)-Binding Protein I/genetics , Animals , RNA/metabolism , RNA/genetics , Cell Line , Cell Differentiation , Cytoplasm/metabolism , Prosencephalon/metabolism
10.
Mol Cell ; 84(11): 2104-2118.e6, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38761795

ABSTRACT

Circular RNAs (circRNAs) are stable RNAs present in cell-free RNA, which may comprise cellular debris and pathogen genomes. Here, we investigate the phenomenon and mechanism of cellular uptake and intracellular fate of exogenous circRNAs. Human myeloid cells and B cells selectively internalize extracellular circRNAs. Macrophage uptake of circRNA is rapid, energy dependent, and saturable. CircRNA uptake can lead to translation of encoded sequences and antigen presentation. The route of internalization influences immune activation after circRNA uptake, with distinct gene expression programs depending on the route of RNA delivery. Genome-scale CRISPR screens and chemical inhibitor studies nominate macrophage scavenger receptor MSR1, Toll-like receptors, and mTOR signaling as key regulators of receptor-mediated phagocytosis of circRNAs, a dominant pathway to internalize circRNAs in parallel to macropinocytosis. These results suggest that cell-free circRNA serves as an "eat me" signal and danger-associated molecular pattern, indicating orderly pathways of recognition and disposal.


Subject(s)
Macrophages , Phagocytosis , RNA, Circular , Signal Transduction , RNA, Circular/genetics , RNA, Circular/metabolism , Humans , Macrophages/metabolism , TOR Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases/genetics , Animals , Toll-Like Receptors/metabolism , Toll-Like Receptors/genetics , B-Lymphocytes/metabolism , B-Lymphocytes/immunology , Scavenger Receptors, Class A/metabolism , Scavenger Receptors, Class A/genetics , Antigen Presentation , Pinocytosis , Mice
11.
Genes Dev ; 38(7-8): 322-335, 2024 05 21.
Article in English | MEDLINE | ID: mdl-38724209

ABSTRACT

Rare, full-length circular intron RNAs distinct from lariats have been reported in several species, but their biogenesis is not understood. We envisioned and tested a hypothesis for their formation using Saccharomyces cerevisiae, documenting full-length and novel processed circular RNAs from multiple introns. Evidence implicates a previously undescribed catalytic activity of the intron lariat spliceosome (ILS) in which the 3'-OH of the lariat tail (with optional trimming and adenylation by the nuclear 3' processing machinery) attacks the branch, joining the intron 3' end to the 5' splice site in a 3'-5' linked circle. Human U2 and U12 spliceosomes produce analogous full-length and processed circles. Postsplicing catalytic activity of the spliceosome may promote intron transposition during eukaryotic genome evolution.


Subject(s)
Introns , RNA Splicing , Saccharomyces cerevisiae , Spliceosomes , Spliceosomes/metabolism , Spliceosomes/genetics , Introns/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Humans , RNA Splicing/genetics , RNA, Circular/genetics , RNA, Circular/metabolism , RNA/metabolism , RNA/genetics
12.
Nature ; 625(7995): 593-602, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38093017

ABSTRACT

Emerging data have shown that previously defined noncoding genomes might encode peptides that bind human leukocyte antigen (HLA) as cryptic antigens to stimulate adaptive immunity1,2. However, the significance and mechanisms of action of cryptic antigens in anti-tumour immunity remain unclear. Here mass spectrometry of the HLA class I (HLA-I) peptidome coupled with ribosome sequencing of human breast cancer samples identified HLA-I-binding cryptic antigenic peptides that were noncanonically translated by a tumour-specific circular RNA (circRNA): circFAM53B. The cryptic peptides efficiently primed naive CD4+ and CD8+ T cells in an antigen-specific manner and induced anti-tumour immunity. Clinically, the expression of circFAM53B and its encoded peptides was associated with substantial infiltration of antigen-specific CD8+ T cells and better survival in patients with breast cancer and patients with melanoma. Mechanistically, circFAM53B-encoded peptides had strong binding affinity to both HLA-I and HLA-II molecules. In vivo, administration of vaccines consisting of tumour-specific circRNA or its encoded peptides in mice bearing breast cancer tumours or melanoma induced enhanced infiltration of tumour-antigen-specific cytotoxic T cells, which led to effective tumour control. Overall, our findings reveal that noncanonical translation of circRNAs can drive efficient anti-tumour immunity, which suggests that vaccination exploiting tumour-specific circRNAs may serve as an immunotherapeutic strategy against malignant tumours.


Subject(s)
Breast Neoplasms , Melanoma , Peptides , Protein Biosynthesis , RNA, Circular , Animals , Female , Humans , Mice , Antigens, Neoplasm/immunology , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Breast Neoplasms/mortality , Breast Neoplasms/pathology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class II/immunology , Mass Spectrometry , Melanoma/genetics , Melanoma/immunology , Melanoma/mortality , Melanoma/pathology , Peptides/genetics , Peptides/immunology , Ribosome Profiling , RNA, Circular/genetics , RNA, Circular/metabolism , Survival Analysis
13.
Mol Cell ; 82(9): 1768-1777.e3, 2022 05 05.
Article in English | MEDLINE | ID: mdl-35358469

ABSTRACT

Circular RNAs are garnering increasing interest as potential regulatory RNAs and a format for gene expression. The characterization of circular RNA using analytical techniques commonly employed in the literature, such as gel electrophoresis, can, under differing conditions, yield different results when attempting to distinguish circular RNA from linear RNA of similar molecular weights. Here, we describe circular RNA migration in different conditions, analyzed by gel electrophoresis and high-performance liquid chromatography (HPLC). We characterize key parameters that affect the migration pattern of circular RNA in gel electrophoresis systems, which include gel type, electrophoresis time, sample buffer composition, and voltage. Finally, we demonstrate the utility of orthogonal analytical tests for circular RNA that take advantage of its covalently closed structure to further distinguish circular RNA from linear RNA following in vitro synthesis.


Subject(s)
RNA, Circular , RNA , Electrophoresis, Agar Gel/methods , Molecular Weight , RNA/genetics , RNA, Circular/genetics
14.
Mol Cell ; 82(2): 420-434.e6, 2022 01 20.
Article in English | MEDLINE | ID: mdl-34951963

ABSTRACT

Exon back-splicing-generated circular RNAs, as a group, can suppress double-stranded RNA (dsRNA)-activated protein kinase R (PKR) in cells. We have sought to synthesize immunogenicity-free, short dsRNA-containing RNA circles as PKR inhibitors. Here, we report that RNA circles synthesized by permuted self-splicing thymidylate synthase (td) introns from T4 bacteriophage or by Anabaena pre-tRNA group I intron could induce an immune response. Autocatalytic splicing introduces ∼74 nt td or ∼186 nt Anabaena extraneous fragments that can distort the folding status of original circular RNAs or form structures themselves to provoke innate immune responses. In contrast, synthesized RNA circles produced by T4 RNA ligase without extraneous fragments exhibit minimized immunogenicity. Importantly, directly ligated circular RNAs that form short dsRNA regions efficiently suppress PKR activation 103- to 106-fold higher than reported chemical compounds C16 and 2-AP, highlighting the future use of circular RNAs as potent inhibitors for diseases related to PKR overreaction.


Subject(s)
Protein Kinase Inhibitors/pharmacology , RNA, Circular/pharmacology , eIF-2 Kinase/antagonists & inhibitors , A549 Cells , Bacteriophage T4/enzymology , Bacteriophage T4/genetics , HEK293 Cells , HeLa Cells , Humans , Immunity, Innate/drug effects , Introns , Nucleic Acid Conformation , Protein Kinase Inhibitors/immunology , RNA Ligase (ATP)/genetics , RNA Ligase (ATP)/metabolism , RNA Precursors/genetics , RNA Precursors/metabolism , RNA, Circular/genetics , RNA, Circular/immunology , Thymidylate Synthase/genetics , Thymidylate Synthase/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism , eIF-2 Kinase/metabolism
15.
Mol Cell ; 82(1): 75-89.e9, 2022 01 06.
Article in English | MEDLINE | ID: mdl-34942120

ABSTRACT

Circular RNAs (circRNAs) are widely expressed in eukaryotes and are regulated in many biological processes. Although several studies indicate their activity as microRNA (miRNA) and protein sponges, little is known about their ability to directly control mRNA homeostasis. We show that the widely expressed circZNF609 directly interacts with several mRNAs and increases their stability and/or translation by favoring the recruitment of the RNA-binding protein ELAVL1. Particularly, the interaction with CKAP5 mRNA, which interestingly overlaps the back-splicing junction, enhances CKAP5 translation, regulating microtubule function in cancer cells and sustaining cell-cycle progression. Finally, we show that circZNF609 downregulation increases the sensitivity of several cancer cell lines to different microtubule-targeting chemotherapeutic drugs and that locked nucleic acid (LNA) protectors against the pairing region on circZNF609 phenocopy such effects. These data set an example of how the small effects tuned by circZNF609/CKAP5 mRNA interaction might have a potent output in tumor growth and drug response.


Subject(s)
Carcinogenesis , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Neoplasms/metabolism , RNA, Circular/metabolism , RNA, Messenger/metabolism , Animals , Antineoplastic Agents/pharmacology , ELAV-Like Protein 1/genetics , ELAV-Like Protein 1/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , K562 Cells , Male , Mice, Nude , Microtubule-Associated Proteins/genetics , Microtubules/drug effects , Microtubules/genetics , Microtubules/pathology , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/pathology , RNA, Circular/genetics , RNA, Messenger/genetics , Signal Transduction , Tumor Burden , Xenograft Model Antitumor Assays
16.
Mol Cell ; 81(20): 4300-4318.e13, 2021 10 21.
Article in English | MEDLINE | ID: mdl-34437836

ABSTRACT

The human genome encodes tens of thousands circular RNAs (circRNAs) with mostly unknown functions. Circular RNAs require internal ribosome entry sites (IRES) if they are to undergo translation without a 5' cap. Here, we develop a high-throughput screen to systematically discover RNA sequences that can direct circRNA translation in human cells. We identify more than 17,000 endogenous and synthetic sequences as candidate circRNA IRES. 18S rRNA complementarity and a structured RNA element positioned on the IRES are important for driving circRNA translation. Ribosome profiling and peptidomic analyses show extensive IRES-ribosome association, hundreds of circRNA-encoded proteins with tissue-specific distribution, and antigen presentation. We find that circFGFR1p, a protein encoded by circFGFR1 that is downregulated in cancer, functions as a negative regulator of FGFR1 oncoprotein to suppress cell growth during stress. Systematic identification of circRNA IRES elements may provide important links among circRNA regulation, biological function, and disease.


Subject(s)
Internal Ribosome Entry Sites , Protein Biosynthesis , RNA, Circular/metabolism , Ribosome Subunits/metabolism , Cell Proliferation , Gene Expression Regulation, Neoplastic , HEK293 Cells , HeLa Cells , Humans , Mutation , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Nucleic Acid Conformation , RNA, Circular/genetics , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Ribosome Subunits/genetics , Structure-Activity Relationship
17.
EMBO J ; 42(10): e112408, 2023 05 15.
Article in English | MEDLINE | ID: mdl-37009655

ABSTRACT

The molecular mechanisms underlying estrogen receptor (ER)-positive breast carcinogenesis and endocrine therapy resistance remain incompletely understood. Here, we report that circPVT1, a circular RNA generated from the lncRNA PVT1, is highly expressed in ERα-positive breast cancer cell lines and tumor samples and is functionally important in promoting ERα-positive breast tumorigenesis and endocrine therapy resistance. CircPVT1 acts as a competing endogenous RNA (ceRNA) to sponge miR-181a-2-3p, promoting the expression of ESR1 and downstream ERα-target genes and breast cancer cell growth. Furthermore, circPVT1 directly interacts with MAVS protein to disrupt the RIGI-MAVS complex formation, inhibiting type I interferon (IFN) signaling pathway and anti-tumor immunity. Anti-sense oligonucleotide (ASO)-targeting circPVT1 inhibits ERα-positive breast cancer cell and tumor growth, re-sensitizing tamoxifen-resistant ERα-positive breast cancer cells to tamoxifen treatment. Taken together, our data demonstrated that circPVT1 can work through both ceRNA and protein scaffolding mechanisms to promote cancer. Thus, circPVT1 may serve as a diagnostic biomarker and therapeutic target for ERα-positive breast cancer in the clinic.


Subject(s)
Breast Neoplasms , RNA, Circular , Female , Humans , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carcinogenesis/genetics , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Drug Resistance, Neoplasm/genetics , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Gene Expression Regulation, Neoplastic , Tamoxifen/pharmacology , Tamoxifen/therapeutic use , RNA, Circular/genetics , RNA, Circular/metabolism
18.
EMBO J ; 42(6): e112039, 2023 03 15.
Article in English | MEDLINE | ID: mdl-36715460

ABSTRACT

Intestinal stem cells (ISCs) at the crypt base are responsible for the regeneration of the intestinal epithelium. However, how ISC self-renewal is regulated still remains unclear. Here we identified a circular RNA, circBtnl1, that is highly expressed in ISCs. Loss of circBtnl1 in mice enhanced ISC self-renewal capacity and epithelial regeneration, without changes in mRNA and protein levels of its parental gene Btnl1. Mechanistically, circBtnl1 and Atf4 mRNA competitively bound the ATP-dependent RNA helicase Ddx3y to impair the stability of Atf4 mRNA in wild-type ISCs. Furthermore, ATF4 activated Sox9 transcription by binding to its promoter via a unique motif, to enhance the self-renewal capacity and epithelial regeneration of ISCs. In contrast, circBtnl1 knockout promoted Atf4 mRNA stability and enhanced ATF4 expression, which caused Sox9 transcription to potentiate ISC stemness. These data indicate that circBtnl1-mediated Atf4 mRNA decay suppresses Sox9 transcription that negatively modulates self-renewal maintenance of ISCs.


Subject(s)
Activating Transcription Factor 4 , Intestinal Mucosa , RNA Stability , RNA, Circular , RNA, Messenger , Regeneration , Stem Cells , Stem Cells/cytology , Stem Cells/physiology , Organoids/cytology , Mice, Inbred C57BL , Animals , Mice , RNA, Circular/genetics , RNA, Circular/metabolism , Intestinal Mucosa/cytology , Intestinal Mucosa/physiology , Regeneration/genetics , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , RNA, Messenger/metabolism , Transcriptional Activation , SOX9 Transcription Factor/genetics , Minor Histocompatibility Antigens/metabolism , DEAD-box RNA Helicases/metabolism
19.
Genome Res ; 34(3): 376-393, 2024 04 25.
Article in English | MEDLINE | ID: mdl-38609186

ABSTRACT

Exon-intron circRNAs (EIciRNAs) are a circRNA subclass with retained introns. Global features of EIciRNAs remain largely unexplored, mainly owing to the lack of bioinformatic tools. The regulation of intron retention (IR) in EIciRNAs and the associated functionality also require further investigation. We developed a framework, FEICP, which efficiently detected EIciRNAs from high-throughput sequencing (HTS) data. EIciRNAs are distinct from exonic circRNAs (EcircRNAs) in aspects such as with larger length, localization in the nucleus, high tissue specificity, and enrichment mostly in the brain. Deep learning analyses revealed that compared with regular introns, the retained introns of circRNAs (CIRs) are shorter in length, have weaker splice site strength, and have higher GC content. Compared with retained introns in linear RNAs (LIRs), CIRs are more likely to form secondary structures and show greater sequence conservation. CIRs are closer to the 5'-end, whereas LIRs are closer to the 3'-end of transcripts. EIciRNA-generating genes are more actively transcribed and associated with epigenetic marks of gene activation. Computational analyses and genome-wide CRISPR screening revealed that SRSF1 binds to CIRs and inhibits the biogenesis of most EIciRNAs. SRSF1 regulates the biogenesis of EIciLIMK1, which enhances the expression of LIMK1 in cis to boost neuronal differentiation, exemplifying EIciRNA physiological function. Overall, our study has developed the FEICP pipeline to identify EIciRNAs from HTS data, and reveals multiple features of CIRs and EIciRNAs. SRSF1 has been identified to regulate EIciRNA biogenesis. EIciRNAs and the regulation of EIciRNA biogenesis play critical roles in neuronal differentiation.


Subject(s)
Exons , Introns , RNA, Circular , RNA, Circular/genetics , RNA, Circular/metabolism , Humans , Serine-Arginine Splicing Factors/genetics , Serine-Arginine Splicing Factors/metabolism , High-Throughput Nucleotide Sequencing , Computational Biology/methods
20.
PLoS Genet ; 20(7): e1011359, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39074152

ABSTRACT

Proper transport of RNAs to synapses is essential for localized translation of proteins in response to synaptic signals and synaptic plasticity. Alzheimer's disease (AD) is a neurodegenerative disease characterized by accumulation of amyloid aggregates and hyperphosphorylated tau neurofibrillary tangles followed by widespread synapse loss. To understand whether RNA synaptic localization is impacted in AD, we performed RNA sequencing on synaptosomes and brain homogenates from AD patients and cognitively healthy controls. This resulted in the discovery of hundreds of mislocalized mRNAs in AD among frontal and temporal brain regions. Similar observations were found in an APPswe/PSEN1dE9 mouse model. Furthermore, major differences were observed among circular RNAs (circRNAs) localized to synapses in AD including two overlapping isoforms of circGSK3ß, one upregulated, and one downregulated. Expression of these distinct isoforms affected tau phosphorylation in neuronal cells substantiating the importance of circRNAs in the brain and pointing to a new class of therapeutic targets.


Subject(s)
Alzheimer Disease , RNA, Circular , RNA, Messenger , Synapses , tau Proteins , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , RNA, Circular/genetics , RNA, Circular/metabolism , Humans , Animals , Synapses/metabolism , Synapses/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Mice , tau Proteins/metabolism , tau Proteins/genetics , Phosphorylation , Disease Models, Animal , Brain/metabolism , Brain/pathology , Male , Neurons/metabolism , Mice, Transgenic , Synaptosomes/metabolism , Female , Aged
SELECTION OF CITATIONS
SEARCH DETAIL