Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 5.057
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 187(3): 733-749.e16, 2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-38306984

RESUMEN

Autoimmune diseases disproportionately affect females more than males. The XX sex chromosome complement is strongly associated with susceptibility to autoimmunity. Xist long non-coding RNA (lncRNA) is expressed only in females to randomly inactivate one of the two X chromosomes to achieve gene dosage compensation. Here, we show that the Xist ribonucleoprotein (RNP) complex comprising numerous autoantigenic components is an important driver of sex-biased autoimmunity. Inducible transgenic expression of a non-silencing form of Xist in male mice introduced Xist RNP complexes and sufficed to produce autoantibodies. Male SJL/J mice expressing transgenic Xist developed more severe multi-organ pathology in a pristane-induced lupus model than wild-type males. Xist expression in males reprogrammed T and B cell populations and chromatin states to more resemble wild-type females. Human patients with autoimmune diseases displayed significant autoantibodies to multiple components of XIST RNP. Thus, a sex-specific lncRNA scaffolds ubiquitous RNP components to drive sex-biased immunity.


Asunto(s)
Autoanticuerpos , Enfermedades Autoinmunes , ARN Largo no Codificante , Animales , Femenino , Humanos , Masculino , Ratones , Autoanticuerpos/genética , Enfermedades Autoinmunes/genética , Autoinmunidad/genética , Ribonucleoproteínas/genética , Ribonucleoproteínas/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Cromosoma X/genética , Cromosoma X/metabolismo , Inactivación del Cromosoma X , Caracteres Sexuales
2.
Cell ; 184(10): 2633-2648.e19, 2021 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-33864768

RESUMEN

Long non-coding RNA (lncRNA) genes have well-established and important impacts on molecular and cellular functions. However, among the thousands of lncRNA genes, it is still a major challenge to identify the subset with disease or trait relevance. To systematically characterize these lncRNA genes, we used Genotype Tissue Expression (GTEx) project v8 genetic and multi-tissue transcriptomic data to profile the expression, genetic regulation, cellular contexts, and trait associations of 14,100 lncRNA genes across 49 tissues for 101 distinct complex genetic traits. Using these approaches, we identified 1,432 lncRNA gene-trait associations, 800 of which were not explained by stronger effects of neighboring protein-coding genes. This included associations between lncRNA quantitative trait loci and inflammatory bowel disease, type 1 and type 2 diabetes, and coronary artery disease, as well as rare variant associations to body mass index.


Asunto(s)
Enfermedad/genética , Herencia Multifactorial/genética , Población/genética , ARN Largo no Codificante/genética , Transcriptoma , Enfermedad de la Arteria Coronaria/genética , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 2/genética , Perfilación de la Expresión Génica , Variación Genética , Humanos , Enfermedades Inflamatorias del Intestino/genética , Especificidad de Órganos/genética , Sitios de Carácter Cuantitativo
3.
Cell ; 184(7): 1790-1803.e17, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33735607

RESUMEN

The long non-coding RNA (lncRNA) XIST establishes X chromosome inactivation (XCI) in female cells in early development and thereafter is thought to be largely dispensable. Here, we show XIST is continually required in adult human B cells to silence a subset of X-linked immune genes such as TLR7. XIST-dependent genes lack promoter DNA methylation and require continual XIST-dependent histone deacetylation. XIST RNA-directed proteomics and CRISPRi screen reveal distinctive somatic cell-type-specific XIST complexes and identify TRIM28 that mediates Pol II pausing at promoters of X-linked genes in B cells. Single-cell transcriptome data of female patients with either systemic lupus erythematosus or COVID-19 infection revealed XIST dysregulation, reflected by escape of XIST-dependent genes, in CD11c+ atypical memory B cells (ABCs). XIST inactivation with TLR7 agonism suffices to promote isotype-switched ABCs. These results indicate cell-type-specific diversification and function for lncRNA-protein complexes and suggest expanded roles for XIST in sex-differences in biology and medicine.


Asunto(s)
Linfocitos B/inmunología , COVID-19 , Lupus Eritematoso Sistémico , ARN Largo no Codificante/fisiología , Receptor Toll-Like 7/inmunología , Inactivación del Cromosoma X , COVID-19/genética , COVID-19/inmunología , Línea Celular , Metilación de ADN , Femenino , Silenciador del Gen , Humanos , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/inmunología
4.
Mol Cell ; 2024 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-39383879

RESUMEN

The role of long non-coding RNAs (lncRNAs) in malignant cell transformation remains elusive. We previously identified an enhancer-associated lncRNA, LINC01116 (named HOXDeRNA), as a transformative factor converting human astrocytes into glioma-like cells. Employing a combination of CRISPR editing, chromatin isolation by RNA purification coupled with sequencing (ChIRP-seq), in situ mapping RNA-genome interactions (iMARGI), chromatin immunoprecipitation sequencing (ChIP-seq), HiC, and RNA/DNA FISH, we found that HOXDeRNA directly binds to CpG islands within the promoters of 35 glioma-specific transcription factors (TFs) distributed throughout the genome, including key stem cell TFs SOX2, OLIG2, POU3F2, and ASCL1, liberating them from PRC2 repression. This process requires a distinct RNA quadruplex structure and other segments of HOXDeRNA, interacting with EZH2 and CpGs, respectively. Subsequent transformation activates multiple oncogenes (e.g., EGFR, miR-21, and WEE1), driven by the SOX2- and OLIG2-dependent glioma-specific super enhancers. These results help reconstruct the sequence of events underlying the process of astrocyte transformation, highlighting HOXDeRNA's central genome-wide activity and suggesting a shared RNA-dependent mechanism in otherwise heterogeneous and multifactorial gliomagenesis.

5.
Mol Cell ; 83(23): 4318-4333.e10, 2023 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-37989319

RESUMEN

RNA unwinding by DExH-type helicases underlies most RNA metabolism and function. It remains unresolved if and how the basic unwinding reaction of helicases is regulated by auxiliary domains. We explored the interplay between the RecA and auxiliary domains of the RNA helicase maleless (MLE) from Drosophila using structural and functional studies. We discovered that MLE exists in a dsRNA-bound open conformation and that the auxiliary dsRBD2 domain aligns the substrate RNA with the accessible helicase tunnel. In an ATP-dependent manner, dsRBD2 associates with the helicase module, leading to tunnel closure around ssRNA. Furthermore, our structures provide a rationale for blunt-ended dsRNA unwinding and 3'-5' translocation by MLE. Structure-based MLE mutations confirm the functional relevance of our model for RNA unwinding. Our findings contribute to our understanding of the fundamental mechanics of auxiliary domains in DExH helicase MLE, which serves as a model for its human ortholog and potential therapeutic target, DHX9/RHA.


Asunto(s)
Proteínas de Drosophila , ARN Helicasas , Animales , Humanos , Proteínas Cromosómicas no Histona/genética , ADN Helicasas/genética , Drosophila/genética , Drosophila/metabolismo , Proteínas de Drosophila/metabolismo , Homeostasis , ARN/metabolismo , ARN Helicasas/metabolismo , ARN Bicatenario/genética , Factores de Transcripción/metabolismo
6.
Trends Biochem Sci ; 49(6): 532-544, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38582689

RESUMEN

Interactions of RNA with DNA are principles of gene expression control that have recently gained considerable attention. Among RNA-DNA interactions are R-loops and RNA-DNA hybrid G-quadruplexes, as well as RNA-DNA triplexes. It is proposed that RNA-DNA triplexes guide RNA-associated regulatory proteins to specific genomic locations, influencing transcription and epigenetic decision making. Although triplex formation initially was considered solely an in vitro event, recent progress in computational, biochemical, and biophysical methods support in vivo functionality with relevance for gene expression control. Here, we review the central methodology and biology of triplexes, outline paradigms required for triplex function, and provide examples of physiologically important triplex-forming long non-coding RNAs.


Asunto(s)
ADN , ARN , ADN/metabolismo , ADN/química , ARN/metabolismo , ARN/química , ARN/genética , Humanos , Animales , Conformación de Ácido Nucleico
7.
Immunity ; 50(3): 600-615.e15, 2019 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-30824325

RESUMEN

CCR7 chemokine receptor stimulation induces rapid but transient dendritic cell (DC) migration toward draining lymph nodes, which is critical for the initiation of protective immunity and maintenance of immune homeostasis. The mechanisms for terminating CCR7-mediated DC migration remain incompletely understood. Here we have identified a long non-coding RNA lnc-Dpf3 whose feedback restrained CCR7-mediated DC migration. CCR7 stimulation upregulated lnc-Dpf3 via removing N6-methyladenosine (m6A) modification to prevent RNA degradation. DC-specific lnc-Dpf3 deficiency increased CCR7-mediated DC migration, leading to exaggerated adaptive immune responses and inflammatory injuries. Mechanistically, CCR7 stimulation activated the HIF-1α transcription factor pathway in DCs, leading to metabolic reprogramming toward glycolysis for DC migration. lnc-Dpf3 directly bound to HIF-1α and suppressed HIF-1α-dependent transcription of the glycolytic gene Ldha, thus inhibiting DC glycolytic metabolism and migratory capacity. We demonstrate a critical role for CCR7-inducible lnc-Dpf3 in coupling epigenetic and metabolic pathways to feedback-control DC migration and inflammatory responses.


Asunto(s)
Movimiento Celular/genética , Proteínas de Unión al ADN/genética , Glucólisis/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Receptores CCR7/genética , Factores de Transcripción/genética , Inmunidad Adaptativa/genética , Animales , Línea Celular , Células Dendríticas/patología , Epigénesis Genética/genética , Regulación de la Expresión Génica/genética , Células HEK293 , Humanos , Inflamación/genética , Inflamación/patología , Ganglios Linfáticos/patología , Redes y Vías Metabólicas/genética , Ratones , Ratones Endogámicos C57BL , Transcripción Genética/genética , Regulación hacia Arriba/genética
8.
Hum Mol Genet ; 2024 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-39239979

RESUMEN

Common variants in the MicroRNA 137 host gene MIR137HG and its adjacent gene DPYD have been associated with schizophrenia risk and the latest Psychiatric Genomics Consortium (PGC). Genome-Wide Association Study on schizophrenia has confirmed and extended these findings. To elucidate the association of schizophrenia risk-associated SNPs in this genomic region, we examined the expression of both mature and immature transcripts of the miR-137 host gene (MIR137HG) in the dorsolateral prefrontal cortex (DLPFC) and subgenual anterior cingulate cortex (sgACC) of postmortem brain samples of donors with schizophrenia and psychiatrically-unaffected controls using qPCR and RNA-Seq approaches. No differential expression of miR-137, MIR137HG, or its transcripts was observed. Two schizophrenia risk-associated SNPs identified in the PGC study, rs11165917 (DLPFC: P = 2.0e-16; sgACC: P = 6.4e-10) and rs4274102 (DLPFC: P = 0.036; sgACC: P = 0.002), were associated with expression of the MIR137HG long non-coding RNA transcript MIR137HG-203 (ENST00000602672.2) in individuals of European ancestry. Carriers of the minor (risk) allele of rs11165917 had significantly lower expression of MIR137HG-203 compared with those carrying the major allele. However, we were unable to validate this result by short-read sequencing of RNA extracted from DLPFC or sgACC tissue. This finding suggests that immature transcripts of MIR137HG may contribute to genetic risk for schizophrenia.

9.
Mol Cell ; 72(6): 970-984.e7, 2018 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-30449723

RESUMEN

Extensive tracts of the mammalian genome that lack protein-coding function are still transcribed into long noncoding RNA. While these lncRNAs are generally short lived, length restricted, and non-polyadenylated, how their expression is distinguished from protein-coding genes remains enigmatic. Surprisingly, depletion of the ubiquitous Pol-II-associated transcription elongation factor SPT6 promotes a redistribution of H3K36me3 histone marks from active protein coding to lncRNA genes, which correlates with increased lncRNA transcription. SPT6 knockdown also impairs the recruitment of the Integrator complex to chromatin, which results in a transcriptional termination defect for lncRNA genes. This leads to the formation of extended, polyadenylated lncRNAs that are both chromatin restricted and form increased levels of RNA:DNA hybrid (R-loops) that are associated with DNA damage. Additionally, these deregulated lncRNAs overlap with DNA replication origins leading to localized DNA replication stress and a cellular senescence phenotype. Overall, our results underline the importance of restricting lncRNA expression.


Asunto(s)
Proliferación Celular , Senescencia Celular , Daño del ADN , Replicación del ADN , ADN de Neoplasias/biosíntesis , ARN Largo no Codificante/metabolismo , ARN Neoplásico/metabolismo , Factores de Transcripción/metabolismo , Neoplasias Uterinas/metabolismo , Animales , Ensamble y Desensamble de Cromatina , ADN Polimerasa II/genética , ADN Polimerasa II/metabolismo , ADN de Neoplasias/genética , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Células HeLa , Histonas/metabolismo , Humanos , Metilación , Conformación de Ácido Nucleico , Ácidos Nucleicos Heterodúplex/genética , Ácidos Nucleicos Heterodúplex/metabolismo , Estabilidad del ARN , ARN Largo no Codificante/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Neoplásico/genética , Factores de Transcripción/genética , Transcripción Genética , Neoplasias Uterinas/genética
10.
Mol Cell ; 71(6): 956-972.e9, 2018 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-30146317

RESUMEN

Gene regulation requires selective targeting of DNA regulatory enhancers over megabase distances. Here we show that Evf2, a cloud-forming Dlx5/6 ultraconserved enhancer (UCE) lncRNA, simultaneously localizes to activated (Umad1, 1.6 Mb distant) and repressed (Akr1b8, 27 Mb distant) chr6 target genes, precisely regulating UCE-gene distances and cohesin binding in mouse embryonic forebrain GABAergic interneurons (INs). Transgene expression of Evf2 activates Lsm8 (12 Mb distant) but fails to repress Akr1b8, supporting trans activation and long-range cis repression. Through both short-range (Dlx6 antisense) and long-range (Akr1b8) repression, the Evf2-5'UCE links homeodomain and mevalonate pathway-regulated enhancers to IN diversity. The Evf2-3' end is required for long-range activation but dispensable for RNA cloud localization, functionally dividing the RNA into 3'-activator and 5'UCE repressor and targeting regions. Together, these results support that Evf2 selectively regulates UCE interactions with multi-megabase distant genes through complex effects on chromosome topology, linking lncRNA-dependent topological and transcriptional control with interneuron diversity and seizure susceptibility.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/genética , Proteínas de Homeodominio/genética , Prosencéfalo/embriología , Oxidorreductasas de Alcohol/genética , Animales , Proteínas de Ciclo Celular/genética , Proteínas Cromosómicas no Histona/genética , Secuencia Conservada , Elementos de Facilitación Genéticos/genética , Proteínas de Homeodominio/fisiología , Interneuronas/fisiología , Ratones , Neurogénesis/genética , Neurogénesis/fisiología , ARN Largo no Codificante/genética , Factores de Transcripción , Cohesinas
11.
Plant J ; 2024 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-39226395

RESUMEN

Long non-coding RNAs (lncRNAs) play crucial roles in various biological processes in plants. However, the functional mechanism of lncRNAs in fruit ripening, particularly the transition from unripe to ripe stages, remains elusive. One such lncRNA1840, reported by our group, was found to have important role in tomato fruit ripening. In the present study, we gain insight into its functional role in fruit ripening. CRISPR-Cas9 mediated lncRNA1840 mutants caused the delayed tomato fruit ripening. Notably, loss function of lncRNA1840 did not directly impact ethylene signaling but rather delay ethylene synthesis. Transcriptomic analysis revealed differences in the expression of ripening related genes in lncRNA1840 mutants, suggesting that it is involved in gene regulation of fruit ripening. We used Chromatin Isolation by RNA Purification (ChIRP)-Seq to identify lncRNA1840 binding sites on chromatin. ChIRP-seq suggested that lncRNA1840 had occupancy on 40 genes, but none of them is differentially expressed genes in transcriptomic analysis, which indicated lncRNA1840 might indirectly modulate the gene expression. ChIRP-mass spectrometry analysis identified potential protein interactors of lncRNA1840, Pre-mRNA processing splicing factor 8, highlighting its involvement in post-transcriptional regulatory pathways. In summary, lncRNA1840 is key player in tomato plant growth and fruit ripening, with multifaceted roles in gene expression and regulatory networks.

12.
Cancer Metastasis Rev ; 43(1): 5-27, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37552389

RESUMEN

The hypoxic environment is prominently witnessed in most solid tumors and is associated with the promotion of cell proliferation, epithelial-mesenchymal transition (EMT), angiogenesis, metabolic reprogramming, therapeutic resistance, and metastasis of tumor cells. All the effects are mediated by the expression of a transcription factor hypoxia-inducible factor-1α (HIF-1α). HIF-1α transcriptionally modulates the expression of genes responsible for all the aforementioned functions. The stability of HIF-1α is regulated by many proteins and non-coding RNAs (ncRNAs). In this article, we have critically discussed the crucial role of ncRNAs [such as microRNAs (miRNAs), long non-coding RNAs (lncRNAs), circular RNAs (circRNAs), Piwi-interacting RNAs (piRNAs), and transfer RNA (tRNA)-derived small RNAs (tsRNAs)] in the regulation of stability and expression of HIF-1α. We have comprehensively discussed the molecular mechanisms and relationship of HIF-1α with each type of ncRNA in either promotion or repression of human cancers and therapeutic resistance. We have also elaborated on ncRNAs that are in clinical examination for the treatment of cancers. Overall, the majority of aspects concerning the relationship between HIF-1α and ncRNAs have been discussed in this article.


Asunto(s)
MicroARNs , Neoplasias , Humanos , Proliferación Celular/genética , Resistencia a Antineoplásicos/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , MicroARNs/genética , MicroARNs/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/genética , ARN no Traducido/genética
13.
EMBO J ; 40(15): e107134, 2021 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-34180064

RESUMEN

Long non-coding RNAs (lncRNAs) are emerging as key regulators of endothelial cell function. Here, we investigated the role of a novel vascular endothelial-associated lncRNA (VEAL2) in regulating endothelial permeability. Precise editing of veal2 loci in zebrafish (veal2gib005Δ8/+ ) induced cranial hemorrhage. In vitro and in vivo studies revealed that veal2 competes with diacylglycerol for interaction with protein kinase C beta-b (Prkcbb) and regulates its kinase activity. Using PRKCB2 as bait, we identified functional ortholog of veal2 in humans from HUVECs and named it as VEAL2. Overexpression and knockdown of VEAL2 affected tubulogenesis and permeability in HUVECs. VEAL2 was differentially expressed in choroid tissue in eye and blood from patients with diabetic retinopathy, a disease where PRKCB2 is known to be hyperactivated. Further, VEAL2 could rescue the effects of PRKCB2-mediated turnover of endothelial junctional proteins thus reducing hyperpermeability in hyperglycemic HUVEC model of diabetic retinopathy. Based on evidence from zebrafish and hyperglycemic HUVEC models and diabetic retinopathy patients, we report a hitherto unknown VEAL2 lncRNA-mediated regulation of PRKCB2, for modulating junctional dynamics and maintenance of endothelial permeability.


Asunto(s)
Retinopatía Diabética/genética , Proteína Quinasa C beta/genética , ARN Largo no Codificante/genética , Pez Cebra/genética , Anciano , Anciano de 80 o más Años , Animales , Animales Modificados Genéticamente , Estudios de Casos y Controles , Retinopatía Diabética/fisiopatología , Embrión no Mamífero , Endotelio Vascular , Regulación de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana , Humanos , Persona de Mediana Edad , Permeabilidad , Proteína Quinasa C beta/metabolismo , ARN Largo no Codificante/sangre , Pez Cebra/embriología , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
14.
J Virol ; 98(6): e0170523, 2024 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-38742902

RESUMEN

Long non-coding RNAs (lncRNAs) represent a new group of host factors involved in viral infection. Current study identified an intergenic lncRNA, LINC08148, as a proviral factor of Zika virus (ZIKV) and Dengue virus 2 (DENV2). Knockout (KO) or silencing of LINC08148 decreases the replication of ZIKV and DENV2. LINC08148 mainly acts at the endocytosis step of ZIKV but at a later stage of DENV2. RNA-seq analysis reveals that LINC08148 knockout downregulates the transcription levels of five endocytosis-related genes including AP2B1, CHMP4C, DNM1, FCHO1, and Src. Among them, loss of Src significantly decreases the uptake of ZIKV. Trans-complementation of Src in the LINC08148KO cells largely restores the caveola-mediated endocytosis of ZIKV, indicating that the proviral effect of LINC08148 is exerted through Src. Finally, LINC08148 upregulates the Src transcription through associating with its transcription factor SP1. This work establishes an essential role of LINC08148 in the ZIKV entry, underscoring a significance of lncRNAs in the viral infection. IMPORTANCE: Long non-coding RNAs (lncRNAs), like proteins, participate in viral infection. However, functions of most lncRNAs remain unknown. In this study, we performed a functional screen based on microarray data and identified a new proviral lncRNA, LINC08148. Then, we uncovered that LINC08148 is involved in the caveola-mediated endocytosis of ZIKV, rather than the classical clathrin-mediated endocytosis. Mechanistically, LINC08148 upregulates the transcription of Src, an initiator of caveola-mediated endocytosis, through binding to its transcription factor SP1. This study identifies a new lncRNA involved in the ZIKV infection, suggesting lncRNAs and cellular proteins are closely linked and cooperate to regulate viral infection.


Asunto(s)
Endocitosis , ARN Largo no Codificante , Internalización del Virus , Infección por el Virus Zika , Virus Zika , ARN Largo no Codificante/metabolismo , ARN Largo no Codificante/genética , Virus Zika/genética , Virus Zika/fisiología , Humanos , Infección por el Virus Zika/virología , Infección por el Virus Zika/metabolismo , Infección por el Virus Zika/genética , Factor de Transcripción Sp1/metabolismo , Factor de Transcripción Sp1/genética , Caveolas/metabolismo , Animales , Replicación Viral , Regulación hacia Arriba , Virus del Dengue/fisiología , Virus del Dengue/genética , Chlorocebus aethiops , Células HEK293 , Células Vero , Familia-src Quinasas/metabolismo , Familia-src Quinasas/genética
15.
Brief Bioinform ; 24(1)2023 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-36545797

RESUMEN

The subcellular localization of long non-coding RNAs (lncRNAs) is crucial for understanding lncRNA functions. Most of existing lncRNA subcellular localization prediction methods use k-mer frequency features to encode lncRNA sequences. However, k-mer frequency features lose sequence order information and fail to capture sequence patterns and motifs of different lengths. In this paper, we proposed GraphLncLoc, a graph convolutional network-based deep learning model, for predicting lncRNA subcellular localization. Unlike previous studies encoding lncRNA sequences by using k-mer frequency features, GraphLncLoc transforms lncRNA sequences into de Bruijn graphs, which transforms the sequence classification problem into a graph classification problem. To extract the high-level features from the de Bruijn graph, GraphLncLoc employs graph convolutional networks to learn latent representations. Then, the high-level feature vectors derived from de Bruijn graph are fed into a fully connected layer to perform the prediction task. Extensive experiments show that GraphLncLoc achieves better performance than traditional machine learning models and existing predictors. In addition, our analyses show that transforming sequences into graphs has more distinguishable features and is more robust than k-mer frequency features. The case study shows that GraphLncLoc can uncover important motifs for nucleus subcellular localization. GraphLncLoc web server is available at http://csuligroup.com:8000/GraphLncLoc/.


Asunto(s)
ARN Largo no Codificante , ARN Largo no Codificante/genética , Aprendizaje Automático
16.
Mol Cell ; 67(2): 163-164, 2017 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-28732203

RESUMEN

In this issue of Molecular Cell, two papers by Chen et al. (2017) and Li et al. (2017) describe new insights into circRNA biogenesis and function, connecting circRNAs to innate immune pathways.


Asunto(s)
ARN Helicasas DEAD-box/inmunología , ARN , Antivirales , Humanos , Inmunidad Innata
17.
BMC Biol ; 22(1): 159, 2024 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-39075446

RESUMEN

BACKGROUND: Recent studies have shown that several long non-coding RNAs (lncRNAs) in the placenta are associated with preeclampsia (PE). However, the extent to which lncRNAs may contribute to the pathological progression of PE is unclear. RESULTS: Here, we report a hierarchical regulatory network involved in early-onset severe PE (EOSPE). We have carried out transcriptome sequencing on the placentae from patients and normal subjects to identify the differentially expressed genes (DEGs), including some lncRNAs (DElncRNAs). We then constructed a high-quality hierarchical regulatory network of lncRNAs, transcription factors (TFs), and target DEGs, containing 1851 lncRNA-TF interactions and 6901 TF-promoter interactions. The lncRNA-to-target regulatory interactions were further validated by the triplex structures between the DElncRNAs and the promoters of the target DEGs. The DElncRNAs in the regulatory network were clustered into 3 clusters, one containing DElncRNAs correlated with the blood pressure, including FLNB-AS1 with targeting 27.89% (869/3116) DEGs in EOSPE. We further demonstrated that FLNB-AS1 could bind the transcription factor JUNB to regulate a series members of the HIF-1 signaling pathway in trophoblast cells. CONCLUSIONS: Our results suggest that the differential expression of lncRNAs may perturb the lncRNA-TF-DEG hierarchical regulatory network, leading to the dysregulation of many genes involved in EOSPE. Our study provides a new strategy and a valuable resource for studying the mechanism underlying gene dysregulation in EOSPE patients.


Asunto(s)
Redes Reguladoras de Genes , Preeclampsia , ARN Largo no Codificante , Preeclampsia/genética , Humanos , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Femenino , Embarazo , Placenta/metabolismo
18.
Genomics ; 116(2): 110803, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38290592

RESUMEN

N6-methyladenosine (m6A) methylation is the most prevalent internal epigenetic posttranscriptional mechanism for regulating mammalian RNA. Despite recent advances in determining the biological functions of m6A methylation, its association with the pathology of ovarian endometriosis remains uncertain. Herein, we performed m6A transcriptome-wide profiling to identify key lncRNAs with m6A modification involved in ovarian endometriosis development by bioinformatics analysis. We found the total m6A level was lower in ovarian endometriosis than in normal endometrium samples, with 9663 m6A peaks associated with 8989 lncRNAs detected in ovarian endometriosis and 9902 m6A peaks associated with 9210 lncRNAs detected in normal endometrium samples. These m6A peaks were primarily enriched within AAACU motifs. Functional enrichment analysis indicated that pathways involving the regulation of adhesion and development were significantly enriched in these differentially methylated lncRNAs. The regulatory relationships among lncRNAs, microRNAs (miRNAs), and mRNAs were identified by competing endogenous RNA (ceRNA) analysis and determination of the network regulating lncRNA-mRNA expression. Several specific lncRNA, including LINC00665, LINC00937, FZD10-AS1, DIO3OS and GATA2-AS1 which were differently expressed and modified by m6A, were validated using qRT-PCR and its interaction with infiltrating immune cells was explored. Furthermore, we found LncRNA DIO3OS promotes the invasion and migration of Human endometrial stromal cells (THESCs) and ALKBH5 regulates the expression of the lncRNA DIO3OS through m6A modification in vitro. Our study firstly revealed the transcriptome-wide map of m6A modification in lncRNAs of ovarian endometriosis. These findings may enable the determination of the underlying mechanism governing the pathogenesis of ovarian endometriosis and provide theoretical basis for further deeper research on the role of m6A in the development of ovarian endometriosis.


Asunto(s)
Endometriosis , ARN Largo no Codificante , Femenino , Humanos , Animales , ARN Largo no Codificante/genética , Transcriptoma , Endometriosis/genética , Adenosina , Metilación , Mamíferos
19.
BMC Bioinformatics ; 25(1): 187, 2024 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-38741200

RESUMEN

MOTIVATION: Long non-coding RNAs (lncRNAs) are a class of molecules involved in important biological processes. Extensive efforts have been provided to get deeper understanding of disease mechanisms at the lncRNA level, guiding towards the detection of biomarkers for disease diagnosis, treatment, prognosis and prevention. Unfortunately, due to costs and time complexity, the number of possible disease-related lncRNAs verified by traditional biological experiments is very limited. Computational approaches for the prediction of disease-lncRNA associations allow to identify the most promising candidates to be verified in laboratory, reducing costs and time consuming. RESULTS: We propose novel approaches for the prediction of lncRNA-disease associations, all sharing the idea of exploring associations among lncRNAs, other intermediate molecules (e.g., miRNAs) and diseases, suitably represented by tripartite graphs. Indeed, while only a few lncRNA-disease associations are still known, plenty of interactions between lncRNAs and other molecules, as well as associations of the latters with diseases, are available. A first approach presented here, NGH, relies on neighborhood analysis performed on a tripartite graph, built upon lncRNAs, miRNAs and diseases. A second approach (CF) relies on collaborative filtering; a third approach (NGH-CF) is obtained boosting NGH by collaborative filtering. The proposed approaches have been validated on both synthetic and real data, and compared against other methods from the literature. It results that neighborhood analysis allows to outperform competitors, and when it is combined with collaborative filtering the prediction accuracy further improves, scoring a value of AUC equal to 0966. AVAILABILITY: Source code and sample datasets are available at: https://github.com/marybonomo/LDAsPredictionApproaches.git.


Asunto(s)
Biología Computacional , ARN Largo no Codificante , ARN Largo no Codificante/genética , Humanos , Biología Computacional/métodos , Algoritmos , MicroARNs/genética , MicroARNs/metabolismo , Predisposición Genética a la Enfermedad/genética
20.
Artículo en Inglés | MEDLINE | ID: mdl-39265177

RESUMEN

Pulmonary hypertension (PH) is a life-threatening condition characterized by pulmonary vascular remodeling and endothelial dysfunction. Current therapies primarily target vasoactive imbalances but often fail to address adverse vascular remodeling. Long non-coding RNA (lncRNA), which are key regulators of various cellular processes, remain underexplored in the context of PH. To investigate the role of lncRNA in PH, we performed a comprehensive analysis using Weighted Gene Co-expression Network Analysis (WGCNA) on the GSE113439 dataset, comprising human lung tissue samples from different PH subtypes. Our analysis identified the lncRNA SNHG11 as consistently downregulated in PH. Functional assays in human pulmonary artery endothelial cells (HPAECs) demonstrated that SNHG11 plays a critical role in modulating inflammation, cell proliferation, apoptosis, and the JAK/STAT and MAPK signaling pathways. Mechanistically, SNHG11 influences the stability of PRPF8, a crucial mRNA spliceosome component, thereby affecting multiple cellular functions beyond splicing. In vivo experiments using a hypoxic rat model showed that knockdown of SNHG11 alleviates PH development and improves right ventricular function. These findings highlight SNHG11 as a key regulator in PH pathogenesis and suggest it as a potential therapeutic target.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA