Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
1.
Mol Cell ; 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-39047724

ABSTRACT

RNA-RNA interactions (RRIs) can dictate RNA molecules to form intricate higher-order structures and bind their RNA substrates in diverse biological processes. To elucidate the function, binding specificity, and regulatory mechanisms of various RNA molecules, especially the vast repertoire of non-coding RNAs, advanced technologies and methods that globally map RRIs are extremely valuable. In the past decades, many state-of-the-art technologies have been developed for this purpose. This review focuses on those high-throughput technologies for the global mapping of RRIs. We summarize the key concepts and the pros and cons of different technologies. In addition, we highlight the novel biological insights uncovered by these RRI mapping methods and discuss the future challenges for appreciating the crucial roles of RRIs in gene regulation across bacteria, viruses, archaea, and mammals.

2.
Mol Cell ; 84(3): 490-505.e9, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38128540

ABSTRACT

SARS-CoV-2 RNA interacts with host factors to suppress interferon responses and simultaneously induces cytokine release to drive the development of severe coronavirus disease 2019 (COVID-19). However, how SARS-CoV-2 hijacks host RNAs to elicit such imbalanced immune responses remains elusive. Here, we analyzed SARS-CoV-2 RNA in situ structures and interactions in infected cells and patient lung samples using RIC-seq. We discovered that SARS-CoV-2 RNA forms 2,095 potential duplexes with the 3' UTRs of 205 host mRNAs to increase their stability by recruiting RNA-binding protein YBX3 in A549 cells. Disrupting the SARS-CoV-2-to-host RNA duplex or knocking down YBX3 decreased host mRNA stability and reduced viral replication. Among SARS-CoV-2-stabilized host targets, NFKBIZ was crucial for promoting cytokine production and reducing interferon responses, probably contributing to cytokine storm induction. Our study uncovers the crucial roles of RNA-RNA interactions in the immunopathogenesis of RNA viruses such as SARS-CoV-2 and provides valuable host targets for drug development.


Subject(s)
COVID-19 , Humans , COVID-19/genetics , SARS-CoV-2/genetics , SARS-CoV-2/metabolism , RNA, Viral/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Interferons/genetics , Cytokines
3.
Mol Cell ; 83(8): 1311-1327.e7, 2023 04 20.
Article in English | MEDLINE | ID: mdl-36958328

ABSTRACT

RNA-binding proteins (RBPs) bind at different positions of the pre-mRNA molecules to promote or reduce the usage of a particular exon. Seeking to understand the working principle of these positional effects, we develop a capture RIC-seq (CRIC-seq) method to enrich specific RBP-associated in situ proximal RNA-RNA fragments for deep sequencing. We determine hnRNPA1-, SRSF1-, and PTBP1-associated proximal RNA-RNA contacts and regulatory mechanisms in HeLa cells. Unexpectedly, the 3D RNA map analysis shows that PTBP1-associated loops in individual introns preferentially promote cassette exon splicing by accelerating asymmetric intron removal, whereas the loops spanning across cassette exon primarily repress splicing. These "positional rules" can faithfully predict PTBP1-regulated splicing outcomes. We further demonstrate that cancer-related splicing quantitative trait loci can disrupt RNA loops by reducing PTBP1 binding on pre-mRNAs to cause aberrant splicing in tumors. Our study presents a powerful method for exploring the functions of RBP-associated RNA-RNA proximal contacts in gene regulation and disease.


Subject(s)
Polypyrimidine Tract-Binding Protein , RNA , Humans , RNA/metabolism , HeLa Cells , Polypyrimidine Tract-Binding Protein/genetics , Polypyrimidine Tract-Binding Protein/metabolism , RNA Splicing/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , RNA Precursors/genetics , RNA Precursors/metabolism , Alternative Splicing , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Serine-Arginine Splicing Factors/genetics
4.
Mol Cell ; 82(21): 4018-4032.e9, 2022 11 03.
Article in English | MEDLINE | ID: mdl-36332605

ABSTRACT

Kinetochore assembly on centromeres is central for chromosome segregation, and defects in this process cause mitotic errors and aneuploidy. Besides the well-established protein network, emerging evidence suggests the involvement of regulatory RNA in kinetochore assembly; however, it has remained elusive about the identity of such RNA, let alone its mechanism of action in this critical process. Here, we report CCTT, a previously uncharacterized long non-coding RNA (lncRNA) transcribed from the arm of human chromosome 17, which plays a vital role in kinetochore assembly. We show that CCTT highly localizes to all centromeres via the formation of RNA-DNA triplex and specifically interacts with CENP-C to help engage this blueprint protein in centromeres, and consequently, CCTT loss triggers extensive mitotic errors and aneuploidy. These findings uncover a non-centromere-derived lncRNA that recruits CENP-C to centromeres and shed critical lights on the function of centromeric DNA sequences as anchor points for kinetochore assembly.


Subject(s)
RNA, Long Noncoding , Humans , Aneuploidy , Centromere Protein A/metabolism , DNA , Kinetochores/metabolism , RNA, Long Noncoding/genetics , Centromere
5.
Cell ; 158(3): 607-19, 2014 Jul 31.
Article in English | MEDLINE | ID: mdl-25083871

ABSTRACT

MicroRNAs are well known to mediate translational repression and mRNA degradation in the cytoplasm. Various microRNAs have also been detected in membrane-compartmentalized organelles, but the functional significance has remained elusive. Here, we report that miR-1, a microRNA specifically induced during myogenesis, efficiently enters the mitochondria where it unexpectedly stimulates, rather than represses, the translation of specific mitochondrial genome-encoded transcripts. We show that this positive effect requires specific miR:mRNA base-pairing and Ago2, but not its functional partner GW182, which is excluded from the mitochondria. We provide evidence for the direct action of Ago2 in mitochondrial translation by crosslinking immunoprecipitation coupled with deep sequencing (CLIP-seq), functional rescue with mitochondria-targeted Ago2, and selective inhibition of the microRNA machinery in the cytoplasm. These findings unveil a positive function of microRNA in mitochondrial translation and suggest a highly coordinated myogenic program via miR-1-mediated translational stimulation in the mitochondria and repression in the cytoplasm.


Subject(s)
Cell Differentiation , MicroRNAs/metabolism , Mitochondria/metabolism , Myoblasts/metabolism , Myocytes, Cardiac/metabolism , Protein Biosynthesis , Animals , Argonaute Proteins/metabolism , Cell Line , Mice , Myoblasts/cytology , Myocytes, Cardiac/cytology
6.
Cell ; 157(3): 651-63, 2014 Apr 24.
Article in English | MEDLINE | ID: mdl-24766810

ABSTRACT

Neurodegenerative diseases can occur so early as to affect neurodevelopment. From a cohort of more than 2,000 consanguineous families with childhood neurological disease, we identified a founder mutation in four independent pedigrees in cleavage and polyadenylation factor I subunit 1 (CLP1). CLP1 is a multifunctional kinase implicated in tRNA, mRNA, and siRNA maturation. Kinase activity of the CLP1 mutant protein was defective, and the tRNA endonuclease complex (TSEN) was destabilized, resulting in impaired pre-tRNA cleavage. Germline clp1 null zebrafish showed cerebellar neurodegeneration that was rescued by wild-type, but not mutant, human CLP1 expression. Patient-derived induced neurons displayed both depletion of mature tRNAs and accumulation of unspliced pre-tRNAs. Transfection of partially processed tRNA fragments into patient cells exacerbated an oxidative stress-induced reduction in cell survival. Our data link tRNA maturation to neuronal development and neurodegeneration through defective CLP1 function in humans.


Subject(s)
Cerebellum/growth & development , Cerebellum/pathology , Cleavage And Polyadenylation Specificity Factor/metabolism , Nuclear Proteins/genetics , Phosphotransferases/genetics , RNA Splicing , RNA, Transfer/genetics , Transcription Factors/genetics , Zebrafish Proteins/metabolism , Animals , Brain/metabolism , Brain/pathology , Cleavage And Polyadenylation Specificity Factor/genetics , Female , Humans , Male , Mice , Models, Molecular , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Nuclear Proteins/metabolism , Pedigree , Phosphotransferases/metabolism , RNA, Transfer/metabolism , Saccharomyces cerevisiae/metabolism , Transcription Factors/metabolism , Zebrafish , Zebrafish Proteins/genetics
7.
Nature ; 619(7971): 868-875, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37438529

ABSTRACT

Enhancers determine spatiotemporal gene expression programs by engaging with long-range promoters1-4. However, it remains unknown how enhancers find their cognate promoters. We recently developed a RNA in situ conformation sequencing technology to identify enhancer-promoter connectivity using pairwise interacting enhancer RNAs and promoter-derived noncoding RNAs5,6. Here we apply this technology to generate high-confidence enhancer-promoter RNA interaction maps in six additional cell lines. Using these maps, we discover that 37.9% of the enhancer-promoter RNA interaction sites are overlapped with Alu sequences. These pairwise interacting Alu and non-Alu RNA sequences tend to be complementary and potentially form duplexes. Knockout of Alu elements compromises enhancer-promoter looping, whereas Alu insertion or CRISPR-dCasRx-mediated Alu tethering to unregulated promoter RNAs can create new loops to homologous enhancers. Mapping 535,404 noncoding risk variants back to the enhancer-promoter RNA interaction maps enabled us to construct variant-to-function maps for interpreting their molecular functions, including 15,318 deletions or insertions in 11,677 Alu elements that affect 6,497 protein-coding genes. We further demonstrate that polymorphic Alu insertion at the PTK2 enhancer can promote tumorigenesis. Our study uncovers a principle for determining enhancer-promoter pairing specificity and provides a framework to link noncoding risk variants to their molecular functions.


Subject(s)
Alu Elements , Enhancer Elements, Genetic , Promoter Regions, Genetic , RNA , Alu Elements/genetics , Cell Line , Enhancer Elements, Genetic/genetics , Focal Adhesion Kinase 1/genetics , Gene Expression Regulation , Nucleic Acid Conformation , Nucleic Acid Heteroduplexes , Promoter Regions, Genetic/genetics , RNA/chemistry , RNA/genetics , RNA/metabolism , Sequence Deletion
8.
Cell ; 152(1-2): 82-96, 2013 Jan 17.
Article in English | MEDLINE | ID: mdl-23313552

ABSTRACT

The induction of pluripotency or trans-differentiation of one cell type to another can be accomplished with cell-lineage-specific transcription factors. Here, we report that repression of a single RNA binding polypyrimidine-tract-binding (PTB) protein, which occurs during normal brain development via the action of miR-124, is sufficient to induce trans-differentiation of fibroblasts into functional neurons. Besides its traditional role in regulated splicing, we show that PTB has a previously undocumented function in the regulation of microRNA functions, suppressing or enhancing microRNA targeting by competitive binding on target mRNA or altering local RNA secondary structure. A key event during neuronal induction is the relief of PTB-mediated blockage of microRNA action on multiple components of the REST complex, thereby derepressing a large array of neuronal genes, including miR-124 and multiple neuronal-specific transcription factors, in nonneuronal cells. This converts a negative feedback loop to a positive one to elicit cellular reprogramming to the neuronal lineage.


Subject(s)
Cell Differentiation , Fibroblasts/cytology , MicroRNAs/genetics , Neurons/cytology , Polypyrimidine Tract-Binding Protein/metabolism , Animals , Cell Line , Cell Lineage , Down-Regulation , Humans , Mice , MicroRNAs/metabolism , Polypyrimidine Tract-Binding Protein/genetics , RNA Splicing , Synapses
9.
Nature ; 591(7849): 259-264, 2021 03.
Article in English | MEDLINE | ID: mdl-33658718

ABSTRACT

Millions of migratory birds occupy seasonally favourable breeding grounds in the Arctic1, but we know little about the formation, maintenance and future of the migration routes of Arctic birds and the genetic determinants of migratory distance. Here we established a continental-scale migration system that used satellite tracking to follow 56 peregrine falcons (Falco peregrinus) from 6 populations that breed in the Eurasian Arctic, and resequenced 35 genomes from 4 of these populations. The breeding populations used five migration routes across Eurasia, which were probably formed by longitudinal and latitudinal shifts in their breeding grounds during the transition from the Last Glacial Maximum to the Holocene epoch. Contemporary environmental divergence between the routes appears to maintain their distinctiveness. We found that the gene ADCY8 is associated with population-level differences in migratory distance. We investigated the regulatory mechanism of this gene, and found that long-term memory was the most likely selective agent for divergence in ADCY8 among the peregrine populations. Global warming is predicted to influence migration strategies and diminish the breeding ranges of peregrine populations of the Eurasian Arctic. Harnessing ecological interactions and evolutionary processes to study climate-driven changes in migration can facilitate the conservation of migratory birds.


Subject(s)
Animal Migration , Falconiformes/physiology , Geographic Mapping , Global Warming/statistics & numerical data , Memory, Long-Term , Animals , Arctic Regions , Falconiformes/genetics , Forecasting
10.
Nature ; 582(7813): 550-556, 2020 06.
Article in English | MEDLINE | ID: mdl-32581380

ABSTRACT

Parkinson's disease is characterized by loss of dopamine neurons in the substantia nigra1. Similar to other major neurodegenerative disorders, there are no disease-modifying treatments for Parkinson's disease. While most treatment strategies aim to prevent neuronal loss or protect vulnerable neuronal circuits, a potential alternative is to replace lost neurons to reconstruct disrupted circuits2. Here we report an efficient one-step conversion of isolated mouse and human astrocytes to functional neurons by depleting the RNA-binding protein PTB (also known as PTBP1). Applying this approach to the mouse brain, we demonstrate progressive conversion of astrocytes to new neurons that innervate into and repopulate endogenous neural circuits. Astrocytes from different brain regions are converted to different neuronal subtypes. Using a chemically induced model of Parkinson's disease in mouse, we show conversion of midbrain astrocytes to dopaminergic neurons, which provide axons to reconstruct the nigrostriatal circuit. Notably, re-innervation of striatum is accompanied by restoration of dopamine levels and rescue of motor deficits. A similar reversal of disease phenotype is also accomplished by converting astrocytes to neurons using antisense oligonucleotides to transiently suppress PTB. These findings identify a potentially powerful and clinically feasible approach to treating neurodegeneration by replacing lost neurons.


Subject(s)
Astrocytes/cytology , Disease Models, Animal , Dopaminergic Neurons/cytology , Parkinson Disease/pathology , Parkinson Disease/therapy , Substantia Nigra/cytology , Substantia Nigra/physiology , Animals , Axons/physiology , Dopamine/biosynthesis , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Female , Heterogeneous-Nuclear Ribonucleoproteins/deficiency , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Humans , In Vitro Techniques , Male , Mice , Neostriatum/cytology , Neostriatum/physiology , Neural Pathways , Neurogenesis , Parkinson Disease/metabolism , Phenotype , Polypyrimidine Tract-Binding Protein/deficiency , Polypyrimidine Tract-Binding Protein/genetics , Polypyrimidine Tract-Binding Protein/metabolism , Substantia Nigra/metabolism
11.
Nature ; 582(7812): 432-437, 2020 06.
Article in English | MEDLINE | ID: mdl-32499643

ABSTRACT

Highly structured RNA molecules usually interact with each other, and associate with various RNA-binding proteins, to regulate critical biological processes. However, RNA structures and interactions in intact cells remain largely unknown. Here, by coupling proximity ligation mediated by RNA-binding proteins with deep sequencing, we report an RNA in situ conformation sequencing (RIC-seq) technology for the global profiling of intra- and intermolecular RNA-RNA interactions. This technique not only recapitulates known RNA secondary structures and tertiary interactions, but also facilitates the generation of three-dimensional (3D) interaction maps of RNA in human cells. Using these maps, we identify noncoding RNA targets globally, and discern RNA topological domains and trans-interacting hubs. We reveal that the functional connectivity of enhancers and promoters can be assigned using their pairwise-interacting RNAs. Furthermore, we show that CCAT1-5L-a super-enhancer hub RNA-interacts with the RNA-binding protein hnRNPK, as well as RNA derived from the MYC promoter and enhancer, to boost MYC transcription by modulating chromatin looping. Our study demonstrates the power and applicability of RIC-seq in discovering the 3D structures, interactions and regulatory roles of RNA.


Subject(s)
Nucleic Acid Conformation , RNA/chemistry , RNA/genetics , Sequence Analysis, RNA/methods , Cell Line , Chromatin/genetics , Chromatin/metabolism , Chromosomes, Human/genetics , Enhancer Elements, Genetic/genetics , Genes, myc/genetics , Genes, rRNA/genetics , Heterogeneous-Nuclear Ribonucleoprotein K/metabolism , Humans , Promoter Regions, Genetic/genetics , RNA, Long Noncoding/chemistry , RNA, Long Noncoding/genetics , Reproducibility of Results , Transcription, Genetic
12.
Nature ; 584(7820): E17, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32724206

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

13.
EMBO Rep ; 24(8): e56416, 2023 08 03.
Article in English | MEDLINE | ID: mdl-37338390

ABSTRACT

Intratumor heterogeneity (ITH) is a barrier to effective therapy. However, it is largely unknown how ITH is established at the onset of tumor progression, such as in colorectal cancer (CRC). Here, we integrate single-cell RNA-seq and functional validation to show that asymmetric division of CRC stem-like cells (CCSC) is critical for early ITH establishment. We find that CCSC-derived xenografts contain seven cell subtypes, including CCSCs, that dynamically change during CRC xenograft progression. Furthermore, three of the subtypes are generated by asymmetric division of CCSCs. They are functionally distinct and appear at the early stage of xenografts. In particular, we identify a chemoresistant and an invasive subtype, and investigate the regulators that control their generation. Finally, we show that targeting the regulators influences cell subtype composition and CRC progression. Our findings demonstrate that asymmetric division of CCSCs contributes to the early establishment of ITH. Targeting asymmetric division may alter ITH and benefit CRC therapy.


Subject(s)
Colorectal Neoplasms , Drug Resistance, Neoplasm , Humans , Drug Resistance, Neoplasm/genetics , Neoplastic Stem Cells/pathology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology
14.
Nucleic Acids Res ; 51(13): 6981-6998, 2023 07 21.
Article in English | MEDLINE | ID: mdl-37246706

ABSTRACT

The molecular mechanism underlying white adipogenesis in humans has not been fully elucidated beyond the transcriptional level. Here, we found that the RNA-binding protein NOVA1 is required for the adipogenic differentiation of human mesenchymal stem cells. By thoroughly exploring the interactions between NOVA1 and its binding RNA, we proved that NOVA1 deficiency resulted in the aberrant splicing of DNAJC10 with an in-frame premature stop codon, reduced DNAJC10 expression at the protein level and hyperactivation of the unfolded protein response (UPR). Moreover, NOVA1 knockdown abrogated the down-regulation of NCOR2 during adipogenesis and up-regulated the 47b+ splicing isoform, which led to decreased chromatin accessibility at the loci of lipid metabolism genes. Interestingly, these effects on human adipogenesis could not be recapitulated in mice. Further analysis of multispecies genomes and transcriptomes indicated that NOVA1-targeted RNA splicing is evolutionarily regulated. Our findings provide evidence for human-specific roles of NOVA1 in coordinating splicing and cell organelle functions during white adipogenesis.


Subject(s)
Chromatin , RNA-Binding Proteins , Unfolded Protein Response , Animals , Humans , Mice , Adipogenesis/genetics , Chromatin/genetics , Neuro-Oncological Ventral Antigen , RNA Splicing , RNA-Binding Proteins/metabolism
15.
Nucleic Acids Res ; 51(10): 5228-5241, 2023 06 09.
Article in English | MEDLINE | ID: mdl-37070178

ABSTRACT

Conversely to canonical splicing, back-splicing connects the upstream 3' splice site (SS) with a downstream 5'SS and generates exonic circular RNAs (circRNAs) that are widely identified and have regulatory functions in eukaryotic gene expression. However, sex-specific back-splicing in Drosophila has not been investigated and its regulation remains unclear. Here, we performed multiple RNA analyses of a variety sex-specific Drosophila samples and identified over ten thousand circular RNAs, in which hundreds are sex-differentially and -specifically back-spliced. Intriguingly, we found that expression of SXL, an RNA-binding protein encoded by Sex-lethal (Sxl), the master Drosophila sex-determination gene that is only spliced into functional proteins in females, promoted back-splicing of many female-differential circRNAs in the male S2 cells, whereas expression of a SXL mutant (SXLRRM) did not promote those events. Using a monoclonal antibody, we further obtained the transcriptome-wide RNA-binding sites of SXL through PAR-CLIP. After splicing assay of mini-genes with mutations in the SXL-binding sites, we revealed that SXL-binding on flanking exons and introns of pre-mRNAs facilitates back-splicing, whereas SXL-binding on the circRNA exons inhibits back-splicing. This study provides strong evidence that SXL has a regulatory role in back-splicing to generate sex-specific and -differential circRNAs, as well as in the initiation of sex-determination cascade through canonical forward-splicing.


Subject(s)
Drosophila Proteins , RNA, Circular , RNA-Binding Proteins , Animals , Female , Male , Drosophila/genetics , Drosophila/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , RNA/genetics , RNA/metabolism , RNA Splicing/genetics , RNA, Circular/metabolism , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
18.
Mol Cell ; 62(6): 875-889, 2016 06 16.
Article in English | MEDLINE | ID: mdl-27211866

ABSTRACT

Increasing evidence suggests that diverse RNA binding proteins (RBPs) interact with regulatory RNAs to regulate transcription. RBFox2 is a well-characterized pre-mRNA splicing regulator, but we now encounter an unexpected paradigm where depletion of this RBP induces widespread increase in nascent RNA production in diverse cell types. Chromatin immunoprecipitation sequencing (ChIP-seq) reveals extensive interaction of RBFox2 with chromatin in a nascent RNA-dependent manner. Bayesian network analysis connects RBFox2 to Polycomb complex 2 (PRC2) and H3K27me3, and biochemical experiments demonstrate the ability of RBFox2 to directly interact with PRC2. Strikingly, RBFox2 inactivation eradicates PRC2 targeting on the majority of bivalent gene promoters and leads to transcriptional de-repression. Together, these findings uncover a mechanism underlying the enigmatic association of PRC2 with numerous active genes, highlight the importance of gene body sequences to gauge transcriptional output, and suggest nascent RNAs as critical signals for transcriptional feedback control to maintain homeostatic gene expression in mammalian genomes.


Subject(s)
Genome , Myocytes, Cardiac/metabolism , Polycomb Repressive Complex 2/metabolism , RNA Splicing Factors/metabolism , RNA/metabolism , Transcription, Genetic , Animals , Bayes Theorem , Chromatin/genetics , Chromatin/metabolism , DNA Methylation , Feedback, Physiological , Gene Expression Regulation , Genotype , HEK293 Cells , Histones/metabolism , Humans , Mice, Knockout , Models, Genetic , Phenotype , Polycomb Repressive Complex 2/genetics , Promoter Regions, Genetic , Protein Binding , RNA/genetics , RNA Interference , RNA Splicing Factors/deficiency , RNA Splicing Factors/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transfection
20.
Proc Natl Acad Sci U S A ; 116(13): 6172-6180, 2019 03 26.
Article in English | MEDLINE | ID: mdl-30867288

ABSTRACT

Heart performance relies on highly coordinated excitation-contraction (EC) coupling, and defects in this critical process may be exacerbated by additional genetic defects and/or environmental insults to cause eventual heart failure. Here we report a regulatory pathway consisting of the RNA binding protein RBFox2, a stress-induced microRNA miR-34a, and the essential EC coupler JPH2. In this pathway, initial cardiac defects diminish RBFox2 expression, which induces transcriptional repression of miR-34a, and elevated miR-34a targets Jph2 to impair EC coupling, which further manifests heart dysfunction, leading to progressive heart failure. The key contribution of miR-34a to this process is further established by administrating its mimic, which is sufficient to induce cardiac defects, and by using its antagomir to alleviate RBFox2 depletion-induced heart dysfunction. These findings elucidate a potential feed-forward mechanism to account for a critical transition to cardiac decompensation and suggest a potential therapeutic avenue against heart failure.


Subject(s)
Heart Failure/metabolism , Heart/physiopathology , Membrane Proteins/metabolism , MicroRNAs/metabolism , Muscle Proteins/metabolism , RNA Splicing Factors/metabolism , Animals , Down-Regulation , Heart Failure/physiopathology , Humans , Mice , Mice, Knockout , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/physiology
SELECTION OF CITATIONS
SEARCH DETAIL