Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
1.
Mol Cell ; 84(3): 490-505.e9, 2024 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-38128540

RESUMO

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.


Assuntos
COVID-19 , Humanos , COVID-19/genética , SARS-CoV-2/genética , SARS-CoV-2/metabolismo , RNA Viral/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Interferons/genética , Citocinas
2.
Mol Cell ; 83(8): 1311-1327.e7, 2023 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-36958328

RESUMO

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.


Assuntos
Proteína de Ligação a Regiões Ricas em Polipirimidinas , RNA , Humanos , RNA/metabolismo , Células HeLa , Proteína de Ligação a Regiões Ricas em Polipirimidinas/genética , Proteína de Ligação a Regiões Ricas em Polipirimidinas/metabolismo , Splicing de RNA/genética , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Precursores de RNA/genética , Precursores de RNA/metabolismo , Processamento Alternativo , Ribonucleoproteínas Nucleares Heterogêneas/genética , Ribonucleoproteínas Nucleares Heterogêneas/metabolismo , Fatores de Processamento de Serina-Arginina/genética
3.
Mol Cell ; 82(21): 4018-4032.e9, 2022 11 03.
Artigo em Inglês | MEDLINE | ID: mdl-36332605

RESUMO

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.


Assuntos
RNA Longo não Codificante , Humanos , Aneuploidia , Proteína Centromérica A/metabolismo , DNA , Cinetocoros/metabolismo , RNA Longo não Codificante/genética , Centrômero
4.
Cell ; 158(3): 607-19, 2014 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-25083871

RESUMO

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.


Assuntos
Diferenciação Celular , MicroRNAs/metabolismo , Mitocôndrias/metabolismo , Mioblastos/metabolismo , Miócitos Cardíacos/metabolismo , Biossíntese de Proteínas , Animais , Proteínas Argonautas/metabolismo , Linhagem Celular , Camundongos , Mioblastos/citologia , Miócitos Cardíacos/citologia
5.
Cell ; 157(3): 651-63, 2014 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-24766810

RESUMO

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.


Assuntos
Cerebelo/crescimento & desenvolvimento , Cerebelo/patologia , Fator de Especificidade de Clivagem e Poliadenilação/metabolismo , Proteínas Nucleares/genética , Fosfotransferases/genética , Splicing de RNA , RNA de Transferência/genética , Fatores de Transcrição/genética , Proteínas de Peixe-Zebra/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Fator de Especificidade de Clivagem e Poliadenilação/genética , Feminino , Humanos , Masculino , Camundongos , Modelos Moleculares , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/patologia , Proteínas Nucleares/metabolismo , Linhagem , Fosfotransferases/metabolismo , RNA de Transferência/metabolismo , Saccharomyces cerevisiae/metabolismo , Fatores de Transcrição/metabolismo , Peixe-Zebra , Proteínas de Peixe-Zebra/genética
6.
Nature ; 619(7971): 868-875, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37438529

RESUMO

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.


Assuntos
Elementos Alu , Elementos Facilitadores Genéticos , Regiões Promotoras Genéticas , RNA , Elementos Alu/genética , Linhagem Celular , Elementos Facilitadores Genéticos/genética , Quinase 1 de Adesão Focal/genética , Regulação da Expressão Gênica , Conformação de Ácido Nucleico , Ácidos Nucleicos Heteroduplexes , Regiões Promotoras Genéticas/genética , RNA/química , RNA/genética , RNA/metabolismo , Deleção de Sequência
7.
Cell ; 152(1-2): 82-96, 2013 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-23313552

RESUMO

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.


Assuntos
Diferenciação Celular , Fibroblastos/citologia , MicroRNAs/genética , Neurônios/citologia , Proteína de Ligação a Regiões Ricas em Polipirimidinas/metabolismo , Animais , Linhagem Celular , Linhagem da Célula , Regulação para Baixo , Humanos , Camundongos , MicroRNAs/metabolismo , Proteína de Ligação a Regiões Ricas em Polipirimidinas/genética , Splicing de RNA , Sinapses
8.
Nature ; 591(7849): 259-264, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33658718

RESUMO

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.


Assuntos
Migração Animal , Falconiformes/fisiologia , Mapeamento Geográfico , Aquecimento Global/estatística & dados numéricos , Memória de Longo Prazo , Animais , Regiões Árticas , Falconiformes/genética , Previsões
9.
Nature ; 584(7820): E17, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32724206

RESUMO

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

10.
Nature ; 582(7812): 432-437, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32499643

RESUMO

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.


Assuntos
Conformação de Ácido Nucleico , RNA/química , RNA/genética , Análise de Sequência de RNA/métodos , Linhagem Celular , Cromatina/genética , Cromatina/metabolismo , Cromossomos Humanos/genética , Elementos Facilitadores Genéticos/genética , Genes myc/genética , Genes de RNAr/genética , Ribonucleoproteínas Nucleares Heterogêneas Grupo K/metabolismo , Humanos , Regiões Promotoras Genéticas/genética , RNA Longo não Codificante/química , RNA Longo não Codificante/genética , Reprodutibilidade dos Testes , Transcrição Gênica
11.
Nature ; 582(7813): 550-556, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32581380

RESUMO

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.


Assuntos
Astrócitos/citologia , Modelos Animais de Doenças , Neurônios Dopaminérgicos/citologia , Doença de Parkinson/patologia , Doença de Parkinson/terapia , Substância Negra/citologia , Substância Negra/fisiologia , Animais , Axônios/fisiologia , Dopamina/biossíntese , Dopamina/metabolismo , Neurônios Dopaminérgicos/metabolismo , Feminino , Ribonucleoproteínas Nucleares Heterogêneas/deficiência , Ribonucleoproteínas Nucleares Heterogêneas/genética , Ribonucleoproteínas Nucleares Heterogêneas/metabolismo , Humanos , Técnicas In Vitro , Masculino , Camundongos , Neostriado/citologia , Neostriado/fisiologia , Vias Neurais , Neurogênese , Doença de Parkinson/metabolismo , Fenótipo , Proteína de Ligação a Regiões Ricas em Polipirimidinas/deficiência , Proteína de Ligação a Regiões Ricas em Polipirimidinas/genética , Proteína de Ligação a Regiões Ricas em Polipirimidinas/metabolismo , Substância Negra/metabolismo
12.
EMBO Rep ; 24(8): e56416, 2023 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-37338390

RESUMO

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.


Assuntos
Neoplasias Colorretais , Resistencia a Medicamentos Antineoplásicos , Humanos , Resistencia a Medicamentos Antineoplásicos/genética , Células-Tronco Neoplásicas/patologia , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia
13.
Nucleic Acids Res ; 51(13): 6981-6998, 2023 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-37246706

RESUMO

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.


Assuntos
Cromatina , Proteínas de Ligação a RNA , Resposta a Proteínas não Dobradas , Animais , Humanos , Camundongos , Adipogenia/genética , Cromatina/genética , Antígeno Neuro-Oncológico Ventral , Splicing de RNA , Proteínas de Ligação a RNA/metabolismo
14.
Nucleic Acids Res ; 51(10): 5228-5241, 2023 06 09.
Artigo em Inglês | MEDLINE | ID: mdl-37070178

RESUMO

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.


Assuntos
Proteínas de Drosophila , RNA Circular , Proteínas de Ligação a RNA , Animais , Feminino , Masculino , Drosophila/genética , Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , RNA/genética , RNA/metabolismo , Splicing de RNA/genética , RNA Circular/metabolismo , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo
17.
Mol Cell ; 62(6): 875-889, 2016 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-27211866

RESUMO

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.


Assuntos
Genoma , Miócitos Cardíacos/metabolismo , Complexo Repressor Polycomb 2/metabolismo , Fatores de Processamento de RNA/metabolismo , RNA/metabolismo , Transcrição Gênica , Animais , Teorema de Bayes , Cromatina/genética , Cromatina/metabolismo , Metilação de DNA , Retroalimentação Fisiológica , Regulação da Expressão Gênica , Genótipo , Células HEK293 , Histonas/metabolismo , Humanos , Camundongos Knockout , Modelos Genéticos , Fenótipo , Complexo Repressor Polycomb 2/genética , Regiões Promotoras Genéticas , Ligação Proteica , RNA/genética , Interferência de RNA , Fatores de Processamento de RNA/deficiência , Fatores de Processamento de RNA/genética , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Transfecção
19.
Proc Natl Acad Sci U S A ; 116(13): 6172-6180, 2019 03 26.
Artigo em Inglês | MEDLINE | ID: mdl-30867288

RESUMO

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.


Assuntos
Insuficiência Cardíaca/metabolismo , Coração/fisiopatologia , Proteínas de Membrana/metabolismo , MicroRNAs/metabolismo , Proteínas Musculares/metabolismo , Fatores de Processamento de RNA/metabolismo , Animais , Regulação para Baixo , Insuficiência Cardíaca/fisiopatologia , Humanos , Camundongos , Camundongos Knockout , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/fisiologia
20.
Nature ; 511(7509): 358-61, 2014 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-25030175

RESUMO

The surface of the cornea consists of a unique type of non-keratinized epithelial cells arranged in an orderly fashion, and this is essential for vision by maintaining transparency for light transmission. Cornea epithelial cells (CECs) undergo continuous renewal from limbal stem or progenitor cells (LSCs), and deficiency in LSCs or corneal epithelium--which turns cornea into a non-transparent, keratinized skin-like epithelium--causes corneal surface disease that leads to blindness in millions of people worldwide. How LSCs are maintained and differentiated into corneal epithelium in healthy individuals and which key molecular events are defective in patients have been largely unknown. Here we report establishment of an in vitro feeder-cell-free LSC expansion and three-dimensional corneal differentiation protocol in which we found that the transcription factors p63 (tumour protein 63) and PAX6 (paired box protein PAX6) act together to specify LSCs, and WNT7A controls corneal epithelium differentiation through PAX6. Loss of WNT7A or PAX6 induces LSCs into skin-like epithelium, a critical defect tightly linked to common human corneal diseases. Notably, transduction of PAX6 in skin epithelial stem cells is sufficient to convert them to LSC-like cells, and upon transplantation onto eyes in a rabbit corneal injury model, these reprogrammed cells are able to replenish CECs and repair damaged corneal surface. These findings suggest a central role of the WNT7A-PAX6 axis in corneal epithelial cell fate determination, and point to a new strategy for treating corneal surface diseases.


Assuntos
Doenças da Córnea/metabolismo , Doenças da Córnea/patologia , Epitélio Corneano/citologia , Epitélio Corneano/metabolismo , Proteínas do Olho/metabolismo , Proteínas de Homeodomínio/metabolismo , Homeostase , Fatores de Transcrição Box Pareados/metabolismo , Proteínas Repressoras/metabolismo , Proteínas Wnt/metabolismo , Animais , Diferenciação Celular , Linhagem da Célula , Modelos Animais de Doenças , Epitélio Corneano/patologia , Proteínas do Olho/genética , Proteínas de Homeodomínio/genética , Humanos , Limbo da Córnea/citologia , Limbo da Córnea/metabolismo , Masculino , Fator de Transcrição PAX6 , Fatores de Transcrição Box Pareados/genética , Coelhos , Proteínas Repressoras/genética , Transdução de Sinais , Pele/citologia , Pele/metabolismo , Pele/patologia , Transplante de Células-Tronco , Células-Tronco/citologia , Células-Tronco/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Proteínas Wnt/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA