Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 507
Filtrar
1.
Cell Chem Biol ; 31(1): 139-149.e14, 2024 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-37967558

RESUMEN

A novel class of benzoxaboroles was reported to induce cancer cell death but the mechanism was unknown. Using a forward genetics platform, we discovered mutations in cleavage and polyadenylation specific factor 3 (CPSF3) that reduce benzoxaborole binding and confer resistance. CPSF3 is the endonuclease responsible for pre-mRNA 3'-end processing, which is also important for RNA polymerase II transcription termination. Benzoxaboroles inhibit this endonuclease activity of CPSF3 in vitro and also curb transcriptional termination in cells, which results in the downregulation of numerous constitutively expressed genes. Furthermore, we used X-ray crystallography to demonstrate that benzoxaboroles bind to the active site of CPSF3 in a manner distinct from the other known inhibitors of CPSF3. The benzoxaborole compound impeded the growth of cancer cell lines derived from different lineages. Our results suggest benzoxaboroles may represent a promising lead as CPSF3 inhibitors for clinical development.


Asunto(s)
Antineoplásicos , Compuestos de Boro , Factor de Especificidad de Desdoblamiento y Poliadenilación , Endonucleasas , Precursores del ARN , Procesamiento Postranscripcional del ARN , Factor de Especificidad de Desdoblamiento y Poliadenilación/antagonistas & inhibidores , Factor de Especificidad de Desdoblamiento y Poliadenilación/química , Endonucleasas/antagonistas & inhibidores , Precursores del ARN/genética , Precursores del ARN/metabolismo , Compuestos de Boro/química , Compuestos de Boro/farmacología , Antineoplásicos/química , Antineoplásicos/farmacología , Procesamiento Postranscripcional del ARN/efectos de los fármacos , Humanos , Línea Celular Tumoral
2.
J Biol Chem ; 299(9): 105093, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37507018

RESUMEN

Epitranscriptomics studies the mechanisms of acquired RNA modifications. The epitranscriptome is dynamically regulated by specific enzymatic reactions, and the proper execution of these enzymatic RNA modifications regulates a variety of physiological RNA functions. However, the lack of experimental tools, such as antibodies for RNA modification, limits the development of epitranscriptomic research. Furthermore, the regulatory enzymes of many RNA modifications have not yet been identified. Herein, we aimed to identify new molecular mechanisms involved in RNA modification by focusing on the AlkB homolog (ALKBH) family molecules, a family of RNA demethylases. We demonstrated that ALKBH4 interacts with small RNA, regulating the formation and metabolism of the (R)-5-carboxyhydroxymethyl uridine methyl ester. We also found that the reaction of ALKBH4 with small RNA enhances protein translation efficiency in an in vitro assay system. These findings indicate that ALKBH4 is involved in the regulation of uridine modification and expand on the role of tRNA-mediated translation control through ALKBH4.


Asunto(s)
Homólogo 4 de AlkB Lisina Desmetilasa , Biosíntesis de Proteínas , Uridina , Procesamiento Postranscripcional del ARN/efectos de los fármacos , Uridina/genética , Uridina/metabolismo , Células HEK293 , Homólogo 4 de AlkB Lisina Desmetilasa/metabolismo , Biosíntesis de Proteínas/genética , Ácidos Cetoglutáricos/farmacología , Hierro/farmacología , Humanos
3.
Cell Cycle ; 22(8): 870-905, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36648057

RESUMEN

Disruption of the complex network that regulates redox homeostasis often underlies resistant phenotypes, which hinder effective and long-lasting cancer eradication. In addition, the RNA methylome-dependent control of gene expression also critically affects traits of cellular resistance to anti-cancer agents. However, few investigations aimed at establishing whether the epitranscriptome-directed adaptations underlying acquired and/or innate resistance traits in cancer could be implemented through the involvement of redox-dependent or -responsive signaling pathways. This is unexpected mainly because: i) the effectiveness of many anti-cancer approaches relies on their capacity to promote oxidative stress (OS); ii) altered redox milieu and reprogramming of mitochondrial function have been acknowledged as critical mediators of the RNA methylome-mediated response to OS. Here we summarize the current state of understanding on this topic, as well as we offer new perspectives that might lead to original approaches and strategies to delay or prevent the problem of refractory cancer and tumor recurrence.


Asunto(s)
Antineoplásicos , Neoplasias , Estrés Oxidativo , Procesamiento Postranscripcional del ARN , ARN , ARN/genética , ARN/metabolismo , Metilación/efectos de los fármacos , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Estrés Oxidativo/efectos de los fármacos , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Neoplasias/terapia , Epigénesis Genética/efectos de los fármacos , Transcriptoma/efectos de los fármacos , Oxidación-Reducción/efectos de los fármacos , Epigenoma/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Recurrencia , Humanos , Animales , Procesamiento Postranscripcional del ARN/efectos de los fármacos , Homeostasis/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo
4.
Proc Natl Acad Sci U S A ; 119(38): e2123529119, 2022 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-36095201

RESUMEN

Cells respond to environmental stress by regulating gene expression at the level of both transcription and translation. The ∼50 modified ribonucleotides of the human epitranscriptome contribute to the latter, with mounting evidence that dynamic regulation of transfer RNA (tRNA) wobble modifications leads to selective translation of stress response proteins from codon-biased genes. Here we show that the response of human hepatocellular carcinoma cells to arsenite exposure is regulated by the availability of queuine, a micronutrient and essential precursor to the wobble modification queuosine (Q) on tRNAs reading GUN codons. Among oxidizing and alkylating agents at equitoxic concentrations, arsenite exposure caused an oxidant-specific increase in Q that correlated with up-regulation of proteins from codon-biased genes involved in energy metabolism. Limiting queuine increased arsenite-induced cell death, altered translation, increased reactive oxygen species levels, and caused mitochondrial dysfunction. In addition to demonstrating an epitranscriptomic facet of arsenite toxicity and response, our results highlight the links between environmental exposures, stress tolerance, RNA modifications, and micronutrients.


Asunto(s)
Arsenitos , Epigénesis Genética , Guanina , ARN de Transferencia , Transcriptoma , Arsenitos/toxicidad , Línea Celular Tumoral , Codón/genética , Guanina/análogos & derivados , Guanina/metabolismo , Humanos , Mitocondrias/efectos de los fármacos , Oxidación-Reducción , Biosíntesis de Proteínas/efectos de los fármacos , Biosíntesis de Proteínas/genética , Procesamiento Postranscripcional del ARN/efectos de los fármacos , ARN de Transferencia/genética
5.
Int J Mol Sci ; 23(3)2022 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-35163183

RESUMEN

Aminoacridines, used for decades as antiseptic and antiparasitic agents, are prospective candidates for therapeutic repurposing and new drug development. Although the mechanisms behind their biological effects are not fully elucidated, they are most often attributed to the acridines' ability to intercalate into DNA. Here, we characterized the effects of 9-aminoacridine (9AA) on pre-rRNA metabolism in cultured mammalian cells. Our results demonstrate that 9AA inhibits both transcription of the ribosomal RNA precursors (pre-rRNA) and processing of the already synthesized pre-rRNAs, thereby rapidly abolishing ribosome biogenesis. Using a fluorescent intercalator displacement assay, we further show that 9AA can bind to RNA in vitro, which likely contributes to its ability to inhibit post-transcriptional steps in pre-rRNA maturation. These findings extend the arsenal of small-molecule compounds that can be used to block ribosome biogenesis in mammalian cells and have implications for the pharmacological development of new ribosome biogenesis inhibitors.


Asunto(s)
Aminacrina/farmacología , Procesamiento Postranscripcional del ARN/efectos de los fármacos , ARN Ribosómico/metabolismo , Animales , Técnicas de Cultivo de Célula , Línea Celular , Nucléolo Celular/metabolismo , Humanos , Ratones , Células 3T3 NIH , Precursores del ARN/genética , Procesamiento Postranscripcional del ARN/fisiología , ARN Ribosómico/biosíntesis , ARN Ribosómico/efectos de los fármacos , Proteínas Ribosómicas/metabolismo , Ribosomas/metabolismo , Transcripción Genética/efectos de los fármacos , Transcripción Genética/fisiología
6.
Cell ; 184(23): 5775-5790.e30, 2021 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-34739832

RESUMEN

RNA, DNA, and protein molecules are highly organized within three-dimensional (3D) structures in the nucleus. Although RNA has been proposed to play a role in nuclear organization, exploring this has been challenging because existing methods cannot measure higher-order RNA and DNA contacts within 3D structures. To address this, we developed RNA & DNA SPRITE (RD-SPRITE) to comprehensively map the spatial organization of RNA and DNA. These maps reveal higher-order RNA-chromatin structures associated with three major classes of nuclear function: RNA processing, heterochromatin assembly, and gene regulation. These data demonstrate that hundreds of ncRNAs form high-concentration territories throughout the nucleus, that specific RNAs are required to recruit various regulators into these territories, and that these RNAs can shape long-range DNA contacts, heterochromatin assembly, and gene expression. These results demonstrate a mechanism where RNAs form high-concentration territories, bind to diffusible regulators, and guide them into compartments to regulate essential nuclear functions.


Asunto(s)
Núcleo Celular/metabolismo , ARN/metabolismo , Animales , Núcleo Celular/efectos de los fármacos , Homólogo de la Proteína Chromobox 5/metabolismo , Cromosomas/metabolismo , ADN/metabolismo , ADN Satélite/metabolismo , Proteínas de Unión al ADN/metabolismo , Dactinomicina/farmacología , Femenino , Genoma , Células HEK293 , Heterocromatina/metabolismo , Humanos , Ratones , Modelos Biológicos , Familia de Multigenes , ARN Polimerasa II/metabolismo , Procesamiento Postranscripcional del ARN/efectos de los fármacos , Procesamiento Postranscripcional del ARN/genética , Empalme del ARN/genética , ARN Largo no Codificante/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Ribosómico/genética , Proteínas de Unión al ARN/metabolismo , Transcripción Genética/efectos de los fármacos
7.
Biomolecules ; 11(9)2021 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-34572607

RESUMEN

In the traditional fermentative model yeast Saccharomyces cerevisiae, ScIxr1 is an HMGB (High Mobility Group box B) protein that has been considered as an important regulator of gene transcription in response to external changes like oxygen, carbon source, or nutrient availability. Kluyveromyces lactis is also a useful eukaryotic model, more similar to many human cells due to its respiratory metabolism. We cloned and functionally characterized by different methodologies KlIXR1, which encodes a protein with only 34.4% amino acid sequence similarity to ScIxr1. Our data indicate that both proteins share common functions, including their involvement in the response to hypoxia or oxidative stress induced by hydrogen peroxide or metal treatments, as well as in the control of key regulators for maintenance of the dNTP (deoxyribonucleotide triphosphate) pool and ribosome synthesis. KlIxr1 is able to bind specific regulatory DNA sequences in the promoter of its target genes, which are well conserved between S. cerevisiae and K. lactis. Oppositely, we found important differences between ScIrx1 and KlIxr1 affecting cellular responses to cisplatin or cycloheximide in these yeasts, which could be dependent on specific and non-conserved domains present in these two proteins.


Asunto(s)
Desoxirribonucleótidos/metabolismo , Proteínas Fúngicas/metabolismo , Regulación Fúngica de la Expresión Génica , Proteínas HMGB/metabolismo , Kluyveromyces/crecimiento & desarrollo , Kluyveromyces/genética , Secuencia de Bases , Cadmio/toxicidad , Carbono/farmacología , Ciclo Celular/efectos de los fármacos , Cisplatino/farmacología , Resistencia a Medicamentos/efectos de los fármacos , Proteínas Fúngicas/química , Eliminación de Gen , Regulación Fúngica de la Expresión Génica/efectos de los fármacos , Proteínas HMGB/química , Hemo/biosíntesis , Peróxido de Hidrógeno/toxicidad , Kluyveromyces/efectos de los fármacos , Mutación/genética , Oxidación-Reducción/efectos de los fármacos , Fenotipo , Regiones Promotoras Genéticas , Unión Proteica/efectos de los fármacos , Procesamiento Postranscripcional del ARN/efectos de los fármacos , ARN Ribosómico/genética , Ribosomas/efectos de los fármacos , Ribosomas/metabolismo , Saccharomyces cerevisiae/metabolismo
8.
Int J Med Sci ; 18(14): 3214-3223, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34400891

RESUMEN

Infantile hemangioma (IH), which threatens the physical and mental health of patients, is the most common benign tumor in infants. Previously, we found that 15,16-dihydrotanshinone I (DHTS) was significantly more effective at inhibiting hemangioma proliferation in vitro and in vivo than the first-line treatment propranolol. To investigate the underlying mechanism of DHTS, we used EOMA cells as a model to study the effect of DHTS. We compared the transcriptomes of control and DHTS-treated EOMA cells. In total, 2462 differentially expressed genes were detected between the groups. Kyoto Encyclopedia of Genes and Genomes pathway analysis revealed downregulated activity of the hypoxia-inducible factor 1 alpha (HIF-1α) signaling pathway in EOMA cells following treatment with DHTS. Thus, we investigated HIF-1α expression at protein and mRNA levels. Our results revealed that DHTS downregulated HIF-1α expression by interfering in its posttranscriptional processing, and the RNA-binding protein HuR participated in this mechanism. Our findings provide a basis for clinical transformation of DHTS and insight into pathogenic mechanisms involved in IH.


Asunto(s)
Furanos/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Hemangioma/tratamiento farmacológico , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Fenantrenos/farmacología , Quinonas/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Regulación hacia Abajo/efectos de los fármacos , Proteína 1 Similar a ELAV/genética , Proteína 1 Similar a ELAV/metabolismo , Furanos/uso terapéutico , Técnicas de Silenciamiento del Gen , Hemangioma/genética , Hemangioma/patología , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Fenantrenos/uso terapéutico , Quinonas/uso terapéutico , Procesamiento Postranscripcional del ARN/efectos de los fármacos , RNA-Seq , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
9.
Cell Rep ; 34(9): 108798, 2021 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-33657363

RESUMEN

Type I interferons (IFNs) induce hundreds of IFN-stimulated genes (ISGs) in response to viral infection. Induction of these ISGs must be regulated for an efficient and controlled antiviral response, but post-transcriptional controls of these genes have not been well defined. Here, we identify a role for the RNA base modification N6-methyladenosine (m6A) in the regulation of ISGs. Using ribosome profiling and quantitative mass spectrometry, coupled with m6A-immunoprecipitation and sequencing, we identify a subset of ISGs, including IFITM1, whose translation is enhanced by m6A and the m6A methyltransferase proteins METTL3 and METTL14. We further determine that the m6A reader YTHDF1 increases the expression of IFITM1 in an m6A-binding-dependent manner. Importantly, we find that the m6A methyltransferase complex promotes the antiviral activity of type I IFN. Thus, these studies identify m6A as having a role in post-transcriptional control of ISG translation during the type I IFN response for antiviral restriction.


Asunto(s)
Adenosina/análogos & derivados , Biosíntesis de Proteínas , Procesamiento Postranscripcional del ARN , Transcripción Genética , Estomatitis Vesicular/genética , Vesiculovirus/patogenicidad , Células A549 , Adenosina/metabolismo , Animales , Antígenos de Diferenciación/biosíntesis , Antígenos de Diferenciación/genética , Antivirales/farmacología , Chlorocebus aethiops , Células HEK293 , Interacciones Huésped-Patógeno , Humanos , Interferón beta/farmacología , Metiltransferasas/biosíntesis , Metiltransferasas/genética , Biosíntesis de Proteínas/efectos de los fármacos , Procesamiento Postranscripcional del ARN/efectos de los fármacos , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Transcripción Genética/efectos de los fármacos , Células Vero , Estomatitis Vesicular/metabolismo , Estomatitis Vesicular/virología , Vesiculovirus/crecimiento & desarrollo , Replicación Viral
10.
Mol Cancer ; 20(1): 29, 2021 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-33557837

RESUMEN

BACKGROUND: The four major RNA adenosine modifications, i.e., m6A, m1A, alternative polyadenylation, and adenosine-to-inosine RNA editing, are mediated mostly by the "writer" enzymes and constitute critical mechanisms of epigenetic regulation in immune response and tumorigenesis. However, the cross-talk and potential roles of these "writers" in the tumor microenvironment (TME), drug sensitivity, and immunotherapy remain unknown. METHODS: We systematically characterized mRNA expression and genetic alterations of 26 RNA modification "writers" in colorectal cancer (CRC), and evaluated their expression pattern in 1697 CRC samples from 8 datasets. We used an unsupervised clustering method to assign the samples into two patterns of expression of RNA modification "writers". Subsequently, we constructed the RNA modification "writer" Score (WM_Score) model based on differentially expressed genes (DEGs) responsible for the RNA modification patterns to quantify the RNA modification-related subtypes of individual tumors. Furthermore, we performed association analysis for WM_Score and characteristics of TME, consensus molecular subtypes (CMSs), clinical features, transcriptional and post-transcriptional regulation, drug response, and the efficacy of immunotherapy. RESULTS: We demonstrated that multi-layer alterations of RNA modification "writer" are associated with patient survival and TME cell-infiltrating characteristics. We identified two distinct RNA modification patterns, characterized by a high and a low WM_Score. The WM_Score-high group was associated with worse patient overall survival and with the infiltration of inhibitory immune cells, such as M2 macrophages, EMT activation, and metastasis, while the WM_Score-low group was associated with a survival advantage, apoptosis, and cell cycle signaling pathways. WM_Score correlated highly with the regulation of transcription and post-transcriptional events contributing to the development of CRC. In response to anti-cancer drugs, WM_Score highly negatively correlated (drug sensitive) with drugs which targeted oncogenic related pathways, such as MAPK, EGFR, and mTOR signaling pathways, positively correlated (drug resistance) with drugs which targeted in apoptosis and cell cycle. Importantly, the WM_Score was associated with the therapeutic efficacy of PD-L1 blockade, suggesting that the development of potential drugs targeting these "writers" to aid the clinical benefits of immunotherapy. CONCLUSIONS: Our study is the first to provide a comprehensive analysis of four RNA modifications in CRC. We revealed the potential function of these writers in TME, transcriptional and post-transcriptional events, and identified their therapeutic liability in targeted therapy and immunotherapy. This work highlights the cross-talk and potential clinical utility of RNA modification "writers" in cancer therapy.


Asunto(s)
Biomarcadores de Tumor , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Regulación Neoplásica de la Expresión Génica , Farmacogenética , Procesamiento Postranscripcional del ARN , Microambiente Tumoral/genética , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/patología , Terapia Combinada , Biología Computacional/métodos , Manejo de la Enfermedad , Susceptibilidad a Enfermedades , Transición Epitelial-Mesenquimal , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Linfocitos Infiltrantes de Tumor/patología , Farmacogenética/métodos , Pronóstico , Modelos de Riesgos Proporcionales , Procesamiento Postranscripcional del ARN/efectos de los fármacos , Transcripción Genética , Transcriptoma , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología
11.
Biochem Pharmacol ; 189: 114402, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33387482

RESUMEN

UDP-glucuronosyltransferases (UGTs) are enzymes catalyzing the glucuronidation of various endogenous and exogenous compounds. In this study, we examined the possibility that N6-methyladenosine (m6A) modification affects hepatic UGT expression. Treatment of HepaRG cells with 3-deazaadenosine, an inhibitor of RNA methylation, significantly increased UGT1A1, UGT1A3, UGT1A4, UGT1A9, UGT2B7, UGT2B10, and UGT2B15 mRNA levels (1.3- to 2.6-fold). Among them, we focused on UGT2B7 because it most highly contributes to glucuronidation of clinically used drugs. Methylated RNA immunoprecipitation assays revealed that UGT2B7 mRNA in HepaRG cells and human livers is subjected to m6A modification mainly at the 5' untranslated region (UTR) and secondarily at the 3'UTR. UGT2B7 mRNA and protein levels in Huh-7 cells were significantly increased by double knockdown of methyltransferase-like 3 (METTL3) and METTL14, whereas those were decreased by knockdown of fat mass and obesity-associated protein (FTO) or alkB homolog 5, RNA demethylase (ALKBH5), suggesting that m6A modification downregulates UGT2B7 expression. By experiments using actinomycin D, an inhibitor of transcription, it was demonstrated that ALKBH5-mediated demethylation would attenuate UGT2B7 mRNA degradation, whereas METTL3/METTL14 or FTO-mediated m6A modification would alter the transactivity of UGT2B7. Luciferase assays revealed that the promoter region at -118 to -106 has a key role in the decrease in transactivity of UGT2B7 by FTO knockdown. We found that hepatocyte nuclear factor 4α (HNF4α) expression was significantly decreased by knockdown of FTO, indicating that this would be the underlying mechanism of the decreased transactivity of UGT2B7 by knockdown of FTO. Interestingly, treatment with entacapone, which is used for the treatment of Parkinson's disease and is an inhibitor of FTO, decreased HNF4α and UGT2B7 expression. In conclusion, this study clarified that RNA methylation posttranscriptionally controls hepatic UGT2B7 expression.


Asunto(s)
Adenosina/análogos & derivados , Glucuronosiltransferasa/biosíntesis , Glucuronosiltransferasa/genética , Hígado/fisiología , Procesamiento Postranscripcional del ARN/fisiología , Adenosina/genética , Adenosina/metabolismo , Adulto , Antiparkinsonianos/farmacología , Catecoles/farmacología , Línea Celular , Femenino , Expresión Génica , Glucuronosiltransferasa/antagonistas & inhibidores , Humanos , Hígado/efectos de los fármacos , Masculino , Persona de Mediana Edad , Nitrilos/farmacología , Procesamiento Postranscripcional del ARN/efectos de los fármacos
12.
Int J Mol Sci ; 22(2)2021 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-33477993

RESUMEN

Information on molecular mechanisms through which sex-steroids regulate oviductal function to support early embryo development is lacking. Here, we hypothesized that the periovulatory endocrine milieu affects the miRNA processing machinery and miRNA expression in bovine oviductal tissues. Growth of the preovulatory follicle was controlled to obtain cows that ovulated a small follicle (SF) and subsequently bore a small corpus luteum (CL; SF-SCL) or a large follicle (LF) and large CL (LF-LCL). These groups differed in the periovulatory plasmatic sex-steroid's concentrations. Ampulla and isthmus samples were collected on day four of the estrous cycle. Abundance of DROSHA, DICER1, and AGO4 transcripts was greater in the ampulla than the isthmus. In the ampulla, transcription of these genes was greater for the SF-SCL group, while the opposite was observed in the isthmus. The expression of the 88 most abundant miRNAs and 14 miRNAs in the ampulla and 34 miRNAs in isthmus were differentially expressed between LF-LCL and SF-SCL groups. Integration of transcriptomic and miRNA data and molecular pathways enrichment showed that important pathways were inhibited in the SF-SCL group due to miRNA control. In conclusion, the endocrine milieu affects the miRNA expression in the bovine oviduct in a region-specific manner.


Asunto(s)
Bovinos , Trompas Uterinas/efectos de los fármacos , Hormonas Esteroides Gonadales/farmacología , MicroARNs , Animales , Bovinos/genética , Bovinos/metabolismo , Ciclo Estral/efectos de los fármacos , Ciclo Estral/genética , Trompas Uterinas/metabolismo , Femenino , Perfilación de la Expresión Génica/veterinaria , Regulación de la Expresión Génica/efectos de los fármacos , MicroARNs/efectos de los fármacos , MicroARNs/genética , MicroARNs/metabolismo , Folículo Ovárico/efectos de los fármacos , Folículo Ovárico/metabolismo , Ovulación/efectos de los fármacos , Ovulación/genética , Procesamiento Postranscripcional del ARN/efectos de los fármacos , Procesamiento Postranscripcional del ARN/genética , Transcriptoma/efectos de los fármacos
13.
Viruses ; 14(1)2021 12 30.
Artículo en Inglés | MEDLINE | ID: mdl-35062264

RESUMEN

Medicinal chemistry optimization of a previously described stilbene inhibitor of HIV-1, 5350150 (2-(2-(5-nitro-2-thienyl)vinyl)quinoline), led to the identification of the thiazole-5-carboxamide derivative (GPS491), which retained potent anti-HIV-1 activity with reduced toxicity. In this report, we demonstrate that the block of HIV-1 replication by GPS491 is accompanied by a drastic inhibition of viral gene expression (IC50 ~ 0.25 µM), and alterations in the production of unspliced, singly spliced, and multiply spliced HIV-1 RNAs. GPS491 also inhibited the replication of adenovirus and multiple coronaviruses. Low µM doses of GPS491 reduced adenovirus infectious yield ~1000 fold, altered virus early gene expression/viral E1A RNA processing, blocked viral DNA amplification, and inhibited late (hexon) gene expression. Loss of replication of multiple coronaviruses (229E, OC43, SARS-CoV2) upon GPS491 addition was associated with the inhibition of viral structural protein expression and the formation of virus particles. Consistent with the observed changes in viral RNA processing, GPS491 treatment induced selective alterations in the accumulation/phosphorylation/function of splicing regulatory SR proteins. Our study establishes that a compound that impacts the activity of cellular factors involved in RNA processing can prevent the replication of several viruses with minimal effect on cell viability.


Asunto(s)
Adenoviridae/efectos de los fármacos , Antivirales/farmacología , Coronavirus/efectos de los fármacos , VIH-1/efectos de los fármacos , Procesamiento Postranscripcional del ARN/efectos de los fármacos , Tiazoles/farmacología , Replicación Viral/efectos de los fármacos , Adenoviridae/fisiología , Antivirales/química , Línea Celular , Coronavirus/clasificación , Coronavirus/fisiología , Expresión Génica/efectos de los fármacos , VIH-1/fisiología , Humanos , Factores de Empalme de ARN/metabolismo , ARN Viral/metabolismo , Tiazoles/química
14.
Epigenetics ; 16(1): 79-91, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32631113

RESUMEN

Fusaric acid (FA) is a food-borne mycotoxin that mediates toxicity with limited information on its epigenetic properties. p53 is a tumour suppressor protein that regulates cell cycle arrest and apoptotic cell death. The expression of p53 is regulated transcriptionally by promoter methylation and post-transcriptionally by N-6-methyladenosine (m6A) RNA methylation. We investigated the effect of FA on p53 expression and its epigenetic regulation via promoter methylation and m6A RNA methylation in human hepatocellular carcinoma (HepG2) cells. HepG2 cells were treated with FA [0, 25, 50, 104, and 150 µg/ml; 24 h] and thereafter, DNA, RNA, and protein was isolated. Promoter methylation and expression of p53 was measured using qPCR and Western blot. RNA immuno-precipitation was used to determine m6A-p53 levels. The expression of m6A methyltransferases (METTL3 and METTL14), demethylases (FTO and ALKBH5), and readers (YTHDF1-3 and YTHDC2) were measured using qPCR. FA induced p53 promoter hypermethylation (p < 0.0001) and decreased p53 expression (p < 0.0001). FA decreased m6A-p53 levels (p < 0.0001) by decreasing METTL3 (p < 0.0001) and METTL14 (p < 0.0001); and suppressed expression of YTHDF1 (p < 0.0001), YTHDF3 (p < 0.0001), and YTHDC2 (p < 0.0001) that ultimately reduced p53 translation (p < 0.0001). Taken together, the data shows that FA epigenetically decreased p53 expression by altering its promoter methylation and m6A RNA methylation in HepG2 cells. This study reveals a mechanism for p53 regulation by FA and provides insight into future therapeutic interventions.


Asunto(s)
Metilación de ADN/efectos de los fármacos , Ácido Fusárico/farmacología , Procesamiento Postranscripcional del ARN/efectos de los fármacos , Proteína p53 Supresora de Tumor/genética , Adenosina/análogos & derivados , Adenosina/metabolismo , Desmetilasa de ARN, Homólogo 5 de AlkB/genética , Desmetilasa de ARN, Homólogo 5 de AlkB/metabolismo , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato/genética , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato/metabolismo , Células Hep G2 , Humanos , Metiltransferasas/genética , Metiltransferasas/metabolismo , Regiones Promotoras Genéticas , ARN Helicasas/genética , ARN Helicasas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
15.
Blood ; 137(7): 994-999, 2021 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-32915956
16.
Mol Neurobiol ; 58(4): 1535-1549, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33210205

RESUMEN

MicroRNAs (miRNA) are small noncoding sequences that control apoptosis, proliferation, and neuroinflammatory pathways in microglia cells. The expression of distinct miRNAs is altered after ischemia in the brain. Only minor information is available about the biogenesis and maturation of miRNAs after ischemia. We aimed at examining the impact of oxygen-glucose deprivation (OGD) and hydrogen peroxide (H2O2)-induced stress on the expression of miRNA regulating proteins such as DROSHA, DGCR8, XPO5, DICER, TARBP2, and AGO2 in the cultured human microglial cell line HMC-3 (human microglial cell line clone 3). OGD duration of 2.5 h or H2O2 stimulation at a concentration of 100 µM for 24 h resulted in a marked increase of the hypoxia sensor hypoxia-inducible factor1-α in HMC-3 cells. These treatments also led to an upregulation of DROSHA, DICER1, and AGO2 detected by semiquantitative real-time PCR (qrtPCR). XPO5 and TARBP2 were only upregulated after stimulation with H2O2, while DGCR8 responded only to OGD. We found elevated DICER1, DROSHA, and AGO2 protein levels by western blot and immunohistochemistry staining. Interestingly, the latter also exposed a colocalization of AGO2 with stress granules (G3BP1) after OGD. Our data indicate that DICER, DROSHA, and AGO2 are induced in microglial cells under hypoxia-like conditions. It might be speculated that their inductions might increase the miRNA synthesis rate. Future studies should investigate this correlation to determine which miRNAs are preferably expressed by microglia cells after ischemia and which functions they could exert.


Asunto(s)
Isquemia Encefálica/genética , Isquemia Encefálica/patología , MicroARNs/biosíntesis , Microglía/metabolismo , Proteínas Argonautas/metabolismo , Línea Celular , ADN Helicasas/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Glucosa/deficiencia , Humanos , Peróxido de Hidrógeno/toxicidad , MicroARNs/genética , Microglía/efectos de los fármacos , Modelos Biológicos , Estrés Oxidativo/efectos de los fármacos , Oxígeno , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , ARN Helicasas/metabolismo , Procesamiento Postranscripcional del ARN/efectos de los fármacos , Proteínas con Motivos de Reconocimiento de ARN/metabolismo , Ribonucleasa III/metabolismo , Estrés Fisiológico/efectos de los fármacos
17.
Methods Mol Biol ; 2192: 59-68, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33230765

RESUMEN

Posttranscriptional RNA modifications have recently emerged as essential posttranscriptional regulators of gene expression. Here we present two methods for single nucleotide resolution detection of 5-formylcytosine (f5C) in RNA. The first relies on chemical protection of f5C against bisulfite treatment, the second method is based on chemical reduction of f5C to hm5C. In combination with regular bisulfite treatment of RNA, the methods allow for precise mapping of f5C. The protocol is used for f5C detection in mtDNA-encoded RNA, however, it can be straightforwardly applied for transcriptome-wide analyses.


Asunto(s)
Citosina/análogos & derivados , Mitocondrias/metabolismo , Nucleótidos/análisis , ARN Mitocondrial/química , Transcriptoma , Citosina/análisis , ADN Mitocondrial/genética , Perfilación de la Expresión Génica , Procesamiento Postranscripcional del ARN/efectos de los fármacos , RNA-Seq/métodos , Sulfitos/farmacología
18.
Signal Transduct Target Ther ; 5(1): 296, 2020 12 26.
Artículo en Inglés | MEDLINE | ID: mdl-33361765

RESUMEN

Hepatocyte nuclear factor 3γ (HNF3γ) is a hepatocyte nuclear factor, but its role and clinical significance in hepatocellular carcinoma (HCC) remain unclear. Herein, we report that HNF3γ expression is downregulated in patient HCC and inversely correlated with HCC malignancy and patient survival. Moreover, our data suggested that the HNF3γ reduction in HCC could be mediated by METTL14-dependent m6A methylation of HNF3γ mRNA. HNF3γ expression was increased during hepatic differentiation and decreased in dedifferentiated HCC cells. Interestingly, HNF3γ delivery promoted differentiation of not only HCC cells but also liver CSCs, which led to suppression of HCC growth. Mechanistic analysis suggested an HNF3γ-centered regulatory network that includes essential liver differentiation-associated transcription factors and functional molecules, which could synergistically facilitate HCC cell differentiation. More importantly, enforced HNF3γ expression sensitized HCC cells to sorafenib-induced growth inhibition and cell apoptosis through transactivation of OATP1B1 and OATP1B3 expression, which are major membrane transporters for sorafenib uptake. Clinical investigation showed that patient-derived HCC xenografts with high HNF3γ expression exhibited a sorafenib response and patients with high HCC HNF3γ levels benefited from sorafenib therapy. Together, these results suggest that HNF3γ plays an essential role in HCC differentiation and may serve as a therapeutic target and predictor of sorafenib benefit in patients.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Desdiferenciación Celular/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Factor Nuclear 3-gamma del Hepatocito/metabolismo , Neoplasias Hepáticas/metabolismo , Proteínas de Neoplasias/metabolismo , Procesamiento Postranscripcional del ARN/efectos de los fármacos , ARN Mensajero/metabolismo , ARN Neoplásico/metabolismo , Sorafenib/farmacología , Animales , Anticuerpos Heterófilos , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Femenino , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Masculino , Ratones , Proteínas de Neoplasias/genética , Trasplante de Neoplasias , ARN Neoplásico/genética
19.
Proc Natl Acad Sci U S A ; 117(47): 29851-29861, 2020 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-33168716

RESUMEN

Kinetoplastids are unicellular eukaryotic parasites responsible for such human pathologies as Chagas disease, sleeping sickness, and leishmaniasis. They have a single large mitochondrion, essential for the parasite survival. In kinetoplastid mitochondria, most of the molecular machineries and gene expression processes have significantly diverged and specialized, with an extreme example being their mitochondrial ribosomes. These large complexes are in charge of translating the few essential mRNAs encoded by mitochondrial genomes. Structural studies performed in Trypanosoma brucei already highlighted the numerous peculiarities of these mitoribosomes and the maturation of their small subunit. However, several important aspects mainly related to the large subunit (LSU) remain elusive, such as the structure and maturation of its ribosomal RNA. Here we present a cryo-electron microscopy study of the protozoans Leishmania tarentolae and Trypanosoma cruzi mitoribosomes. For both species, we obtained the structure of their mature mitoribosomes, complete rRNA of the LSU, as well as previously unidentified ribosomal proteins. In addition, we introduce the structure of an LSU assembly intermediate in the presence of 16 identified maturation factors. These maturation factors act on both the intersubunit and the solvent sides of the LSU, where they refold and chemically modify the rRNA and prevent early translation before full maturation of the LSU.


Asunto(s)
Leishmania/fisiología , Ribosomas Mitocondriales/ultraestructura , Procesamiento Postranscripcional del ARN/fisiología , Subunidades Ribosómicas Grandes de Eucariotas/metabolismo , Trypanosoma cruzi/fisiología , Antiprotozoarios/farmacología , Antiprotozoarios/uso terapéutico , Enfermedad de Chagas/tratamiento farmacológico , Enfermedad de Chagas/parasitología , Microscopía por Crioelectrón , Humanos , Leishmania/citología , Leishmania/efectos de los fármacos , Leishmaniasis/tratamiento farmacológico , Leishmaniasis/parasitología , Ribosomas Mitocondriales/efectos de los fármacos , Ribosomas Mitocondriales/metabolismo , Modelos Moleculares , Procesamiento Postranscripcional del ARN/efectos de los fármacos , ARN Ribosómico/metabolismo , Proteínas Ribosómicas/metabolismo , Subunidades Ribosómicas Grandes de Eucariotas/ultraestructura , Trypanosoma cruzi/citología , Trypanosoma cruzi/efectos de los fármacos
20.
Drug Resist Updat ; 53: 100720, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32892147

RESUMEN

Despite the development of targeted therapy, drug resistance remains a primary hindrance to curative treatment of various cancers. Among several novel approaches to overcome drug resistance, modulating N6-methyladenosine (m6A) RNA modification was found to be an important strategy in various types of cancer cells. Considered as one of the most common epigenetic RNA modifications, m6A regulates multiple biological processes including cellular proliferation, metabolism, and metastasis through modulation of RNA splicing, degradation, and translation, leading to anticancer drug resistance. This regulatory network is orchestrated mainly by several m6A regulators, including "writers", "readers", and "erasers". It is encouraging that several small molecules targeting m6A regulators have shown great potential in overcoming drug resistance in different cancer cell types, two of which entacapone and meclofenamate, are currently undergoing evaluation. However, the m6A modification participates in complex biological processes and its functions are context-dependent, which has challenged the clinical application of targeting the m6A modification in cancer therapy. In this review, we discuss the molecular mechanisms underlying the m6A modification in regulating anticancer drug resistance through modulation of drug-target interaction and drug-mediated cell death signaling. Alteration of the m6A modification interferes with drug efficacy through modulation of the expression of multidrug efflux transporters (e.g., ABCG2, ABCC9, ABCC10), drug metabolizing enzymes (e.g., CYP2C8), and drug targets (e.g., p53 R273 H). Furthermore, alterations of the m6A modification may protect cells from drug-mediated cell death by regulating DNA damage repair (e.g., p53, BRCA1, Pol κ, UBE2B, and ERCC1), downstream adaptive response (e.g., critical regulators of apoptosis, autophagy, pro-survival signaling, and oncogenic bypass signaling), cell stemness, and tumor microenvironment (e.g., ITGA6, ITGB3, and PD-1). We particularly highlight recent advances in therapeutic strategies targeting the m6A modification with the aim to surmount chemoresistance. The comprehensive understanding of the role of the m6A modification integrated with combined therapeutic strategies, should facilitate the development of future therapeutic strategies to circumvent or surmount drug resistance, thus enhancing therapeutic efficacy.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Resistencia a Múltiples Medicamentos/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Procesamiento Postranscripcional del ARN/efectos de los fármacos , Adenosina/análogos & derivados , Adenosina/metabolismo , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Apoptosis/efectos de los fármacos , Apoptosis/genética , Daño del ADN/efectos de los fármacos , Reparación del ADN/efectos de los fármacos , Modelos Animales de Enfermedad , Resistencia a Múltiples Medicamentos/genética , Resistencia a Antineoplásicos/genética , Epigénesis Genética/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Metilación/efectos de los fármacos , Neoplasias/genética , Neoplasias/patología , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Resultado del Tratamiento , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...