Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 509
Filtrar
1.
Environ Health Perspect ; 132(8): 87003, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39133094

RESUMO

BACKGROUND: Currently, many emerging polycyclic aromatic hydrocarbons (PAHs) have been found to be widely present in the environment. However, little has been reported about their toxicity, particularly in relation to CYP1A1. OBJECTIVES: This study aimed to explore the toxicity of naphtho[2,1-a]pyrene (N21aP) and elucidate the mechanism underlying N21aP-induced expression of CYP1A1. METHODS: The concentration and sources of N21aP were detected and analyzed by gas chromatography-triple quadrupole mass spectrometry (GC-MS/MS) and diagnostic ratio analysis. Then the effects of CYP1A1 on the toxicity of N21aP were conducted in male wild-type (WT) and Cyp1a1 knockout mice exposed to N21aP (0.02, 0.2, and 2mg/kg) through intratracheal instillation. Further, the aryl hydrocarbon receptor (AhR) pathway was examined through luciferase and chromatin immunoprecipitation (ChIP) assays. N6-methyladenosine (m6A) modification levels were measured on global RNA and specifically on CYP1A1 mRNA using dot blotting and methylated RNA immunoprecipitation-quantitative real-time polymerase chain reaction (MeRIP qRT-PCR), with validation by m6A inhibitors, DAA and SAH. m6A sites on CYP1A1 were identified by bioinformatics and luciferase assays, and CYP1A1 mRNA's interaction with IGF2BP3 was confirmed by RNA pull-down, luciferase, and RNA binding protein immunoprecipitation (RIP) assays. RESULTS: N21aP was of the same environmental origin as benzo[a]pyrene (BaP) but was more stably present in the environment. N21aP could be metabolically activated by CYP1A1 to produce epoxides, causing DNA damage and further leading to lung inflammation. Importantly, in addition to the classical AhR pathway (i.e., BaP), N21aP also induced CYP1A1 expression with a posttranscriptional modification of m6A in CYP1A1 mRNA via the METTL14-IGF2BP3-CYP1A1 axis. Specifically, in the two recognition sites of METTL14 on the CYP1A1 mRNA transcript (position at 2700 and 5218), a methylation site (position at 5218) in the 3'-untranslated region (UTR) was recognized by IGF2BP3, enhanced the stability of CYP1A1 mRNA, and finally resulted in an increase in CYP1A1 expression. DISCUSSION: This study systematically demonstrated that in addition to AhR-mediated transcriptional regulation, N21aP, had a new additional mechanism of m6A-mediated posttranscriptional modification, jointly contributing to CYP1A1 expression. Given that PAHs are the metabolic substrates of CYP1A1, this study not only helps to understand the significance of environment-genetic interactions for the toxicity of PAHs but also helps to better understand the health risks of the emerging PAHs at environmental exposure levels. https://doi.org/10.1289/EHP14055.


Assuntos
Citocromo P-450 CYP1A1 , Receptores de Hidrocarboneto Arílico , Animais , Citocromo P-450 CYP1A1/metabolismo , Citocromo P-450 CYP1A1/genética , Camundongos , Masculino , Receptores de Hidrocarboneto Arílico/metabolismo , Receptores de Hidrocarboneto Arílico/genética , Camundongos Knockout , Adenosina/análogos & derivados , Adenosina/metabolismo , Poluentes Ambientais/toxicidade , Processamento Pós-Transcricional do RNA/efeitos dos fármacos
2.
Eur J Med Chem ; 274: 116526, 2024 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-38805939

RESUMO

Epitranscriptomics, the field of post-translational RNA modifications, is a burgeoning domain of research that has recently received significant attention for its role in multiple diseases, including cancer. N6-methyladenosine (m6A) is the most prominent post-translational RNA modification and plays a critical role in RNA transcription, processing, translation, and metabolism. The m6A modification is controlled by three protein classes known as writers (methyltransferases), erasers (demethylases), and readers (m6A-binding proteins). Each class of m6A regulatory proteins has been implicated in cancer initiation and progression. As such, many of these proteins have been identified as potential targets for anti-cancer chemotherapeutics. In this work, we provide an overview of the role m6A-regulating proteins play in cancer and discuss the current state of small molecule therapeutics targeting these proteins.


Assuntos
Adenosina , Antineoplásicos , Neoplasias , Bibliotecas de Moléculas Pequenas , Humanos , Adenosina/análogos & derivados , Adenosina/química , Adenosina/farmacologia , Adenosina/metabolismo , Antineoplásicos/farmacologia , Antineoplásicos/química , Antineoplásicos/síntese química , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Neoplasias/metabolismo , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia , RNA/metabolismo , Animais , Estrutura Molecular , Processamento Pós-Transcricional do RNA/efeitos dos fármacos
3.
Cell Chem Biol ; 31(1): 139-149.e14, 2024 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-37967558

RESUMO

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.


Assuntos
Antineoplásicos , Compostos de Boro , Fator de Especificidade de Clivagem e Poliadenilação , Endonucleases , Precursores de RNA , Processamento Pós-Transcricional do RNA , Fator de Especificidade de Clivagem e Poliadenilação/antagonistas & inibidores , Fator de Especificidade de Clivagem e Poliadenilação/química , Endonucleases/antagonistas & inibidores , Precursores de RNA/genética , Precursores de RNA/metabolismo , Compostos de Boro/química , Compostos de Boro/farmacologia , Antineoplásicos/química , Antineoplásicos/farmacologia , Processamento Pós-Transcricional do RNA/efeitos dos fármacos , Humanos , Linhagem Celular Tumoral
4.
J Biol Chem ; 299(9): 105093, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37507018

RESUMO

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.


Assuntos
Homólogo AlkB 4 da Lisina Desmetilase , Biossíntese de Proteínas , Uridina , Processamento Pós-Transcricional do RNA/efeitos dos fármacos , Uridina/genética , Uridina/metabolismo , Células HEK293 , Homólogo AlkB 4 da Lisina Desmetilase/metabolismo , Biossíntese de Proteínas/genética , Ácidos Cetoglutáricos/farmacologia , Ferro/farmacologia , Humanos
5.
Cell Cycle ; 22(8): 870-905, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36648057

RESUMO

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.


Assuntos
Antineoplásicos , Neoplasias , Estresse Oxidativo , Processamento Pós-Transcricional do RNA , RNA , RNA/genética , RNA/metabolismo , Metilação/efeitos dos fármacos , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Estresse Oxidativo/efeitos dos fármacos , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , Neoplasias/terapia , Epigênese Genética/efeitos dos fármacos , Transcriptoma/efeitos dos fármacos , Oxirredução/efeitos dos fármacos , Epigenoma/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Recidiva , Humanos , Animais , Processamento Pós-Transcricional do RNA/efeitos dos fármacos , Homeostase/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo
6.
Proc Natl Acad Sci U S A ; 119(38): e2123529119, 2022 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-36095201

RESUMO

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.


Assuntos
Arsenitos , Epigênese Genética , Guanina , RNA de Transferência , Transcriptoma , Arsenitos/toxicidade , Linhagem Celular Tumoral , Códon/genética , Guanina/análogos & derivados , Guanina/metabolismo , Humanos , Mitocôndrias/efeitos dos fármacos , Oxirredução , Biossíntese de Proteínas/efeitos dos fármacos , Biossíntese de Proteínas/genética , Processamento Pós-Transcricional do RNA/efeitos dos fármacos , RNA de Transferência/genética
7.
Int J Mol Sci ; 23(3)2022 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-35163183

RESUMO

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.


Assuntos
Aminacrina/farmacologia , Processamento Pós-Transcricional do RNA/efeitos dos fármacos , RNA Ribossômico/metabolismo , Animais , Técnicas de Cultura de Células , Linhagem Celular , Nucléolo Celular/metabolismo , Humanos , Camundongos , Células NIH 3T3 , Precursores de RNA/genética , Processamento Pós-Transcricional do RNA/fisiologia , RNA Ribossômico/biossíntese , RNA Ribossômico/efeitos dos fármacos , Proteínas Ribossômicas/metabolismo , Ribossomos/metabolismo , Transcrição Gênica/efeitos dos fármacos , Transcrição Gênica/fisiologia
8.
Cell ; 184(23): 5775-5790.e30, 2021 11 11.
Artigo em Inglês | MEDLINE | ID: mdl-34739832

RESUMO

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.


Assuntos
Núcleo Celular/metabolismo , RNA/metabolismo , Animais , Núcleo Celular/efeitos dos fármacos , Homólogo 5 da Proteína Cromobox/metabolismo , Cromossomos/metabolismo , DNA/metabolismo , DNA Satélite/metabolismo , Proteínas de Ligação a DNA/metabolismo , Dactinomicina/farmacologia , Feminino , Genoma , Células HEK293 , Heterocromatina/metabolismo , Humanos , Camundongos , Modelos Biológicos , Família Multigênica , RNA Polimerase II/metabolismo , Processamento Pós-Transcricional do RNA/efeitos dos fármacos , Processamento Pós-Transcricional do RNA/genética , Splicing de RNA/genética , RNA Longo não Codificante/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Ribossômico/genética , Proteínas de Ligação a RNA/metabolismo , Transcrição Gênica/efeitos dos fármacos
9.
Biomolecules ; 11(9)2021 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-34572607

RESUMO

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.


Assuntos
Desoxirribonucleotídeos/metabolismo , Proteínas Fúngicas/metabolismo , Regulação Fúngica da Expressão Gênica , Proteínas HMGB/metabolismo , Kluyveromyces/crescimento & desenvolvimento , Kluyveromyces/genética , Sequência de Bases , Cádmio/toxicidade , Carbono/farmacologia , Ciclo Celular/efeitos dos fármacos , Cisplatino/farmacologia , Resistência a Medicamentos/efeitos dos fármacos , Proteínas Fúngicas/química , Deleção de Genes , Regulação Fúngica da Expressão Gênica/efeitos dos fármacos , Proteínas HMGB/química , Heme/biossíntese , Peróxido de Hidrogênio/toxicidade , Kluyveromyces/efeitos dos fármacos , Mutação/genética , Oxirredução/efeitos dos fármacos , Fenótipo , Regiões Promotoras Genéticas , Ligação Proteica/efeitos dos fármacos , Processamento Pós-Transcricional do RNA/efeitos dos fármacos , RNA Ribossômico/genética , Ribossomos/efeitos dos fármacos , Ribossomos/metabolismo , Saccharomyces cerevisiae/metabolismo
10.
Int J Med Sci ; 18(14): 3214-3223, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34400891

RESUMO

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.


Assuntos
Furanos/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Hemangioma/tratamento farmacológico , Subunidade alfa do Fator 1 Induzível por Hipóxia/antagonistas & inibidores , Fenantrenos/farmacologia , Quinonas/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Regulação para Baixo/efeitos dos fármacos , Proteína Semelhante a ELAV 1/genética , Proteína Semelhante a ELAV 1/metabolismo , Furanos/uso terapêutico , Técnicas de Silenciamento de Genes , Hemangioma/genética , Hemangioma/patologia , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Fenantrenos/uso terapêutico , Quinonas/uso terapêutico , Processamento Pós-Transcricional do RNA/efeitos dos fármacos , RNA-Seq , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
11.
Cell Rep ; 34(9): 108798, 2021 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-33657363

RESUMO

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.


Assuntos
Adenosina/análogos & derivados , Biossíntese de Proteínas , Processamento Pós-Transcricional do RNA , Transcrição Gênica , Estomatite Vesicular/genética , Vesiculovirus/patogenicidade , Células A549 , Adenosina/metabolismo , Animais , Antígenos de Diferenciação/biossíntese , Antígenos de Diferenciação/genética , Antivirais/farmacologia , Chlorocebus aethiops , Células HEK293 , Interações Hospedeiro-Patógeno , Humanos , Interferon beta/farmacologia , Metiltransferases/biossíntese , Metiltransferases/genética , Biossíntese de Proteínas/efeitos dos fármacos , Processamento Pós-Transcricional do RNA/efeitos dos fármacos , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Transcrição Gênica/efeitos dos fármacos , Células Vero , Estomatite Vesicular/metabolismo , Estomatite Vesicular/virologia , Vesiculovirus/crescimento & desenvolvimento , Replicação Viral
12.
Mol Cancer ; 20(1): 29, 2021 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-33557837

RESUMO

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.


Assuntos
Biomarcadores Tumorais , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Regulação Neoplásica da Expressão Gênica , Farmacogenética , Processamento Pós-Transcricional do RNA , Microambiente Tumoral/genética , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/patologia , Terapia Combinada , Biologia Computacional/métodos , Gerenciamento Clínico , Suscetibilidade a Doenças , Transição Epitelial-Mesenquimal , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/metabolismo , Linfócitos do Interstício Tumoral/patologia , Farmacogenética/métodos , Prognóstico , Modelos de Riscos Proporcionais , Processamento Pós-Transcricional do RNA/efeitos dos fármacos , Transcrição Gênica , Transcriptoma , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/imunologia
13.
Biochem Pharmacol ; 189: 114402, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33387482

RESUMO

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.


Assuntos
Adenosina/análogos & derivados , Glucuronosiltransferase/biossíntese , Glucuronosiltransferase/genética , Fígado/fisiologia , Processamento Pós-Transcricional do RNA/fisiologia , Adenosina/genética , Adenosina/metabolismo , Adulto , Antiparkinsonianos/farmacologia , Catecóis/farmacologia , Linhagem Celular , Feminino , Expressão Gênica , Glucuronosiltransferase/antagonistas & inibidores , Humanos , Fígado/efeitos dos fármacos , Masculino , Pessoa de Meia-Idade , Nitrilas/farmacologia , Processamento Pós-Transcricional do RNA/efeitos dos fármacos
14.
Int J Mol Sci ; 22(2)2021 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-33477993

RESUMO

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.


Assuntos
Bovinos , Tubas Uterinas/efeitos dos fármacos , Hormônios Esteroides Gonadais/farmacologia , MicroRNAs , Animais , Bovinos/genética , Bovinos/metabolismo , Ciclo Estral/efeitos dos fármacos , Ciclo Estral/genética , Tubas Uterinas/metabolismo , Feminino , Perfilação da Expressão Gênica/veterinária , Regulação da Expressão Gênica/efeitos dos fármacos , MicroRNAs/efeitos dos fármacos , MicroRNAs/genética , MicroRNAs/metabolismo , Folículo Ovariano/efeitos dos fármacos , Folículo Ovariano/metabolismo , Ovulação/efeitos dos fármacos , Ovulação/genética , Processamento Pós-Transcricional do RNA/efeitos dos fármacos , Processamento Pós-Transcricional do RNA/genética , Transcriptoma/efeitos dos fármacos
15.
Viruses ; 14(1)2021 12 30.
Artigo em Inglês | MEDLINE | ID: mdl-35062264

RESUMO

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.


Assuntos
Adenoviridae/efeitos dos fármacos , Antivirais/farmacologia , Coronavirus/efeitos dos fármacos , HIV-1/efeitos dos fármacos , Processamento Pós-Transcricional do RNA/efeitos dos fármacos , Tiazóis/farmacologia , Replicação Viral/efeitos dos fármacos , Adenoviridae/fisiologia , Antivirais/química , Linhagem Celular , Coronavirus/classificação , Coronavirus/fisiologia , Expressão Gênica/efeitos dos fármacos , HIV-1/fisiologia , Humanos , Fatores de Processamento de RNA/metabolismo , RNA Viral/metabolismo , Tiazóis/química
16.
Epigenetics ; 16(1): 79-91, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32631113

RESUMO

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.


Assuntos
Metilação de DNA/efeitos dos fármacos , Ácido Fusárico/farmacologia , Processamento Pós-Transcricional do RNA/efeitos dos fármacos , Proteína Supressora de Tumor p53/genética , Adenosina/análogos & derivados , Adenosina/metabolismo , Homólogo AlkB 5 da RNA Desmetilase/genética , Homólogo AlkB 5 da RNA Desmetilase/metabolismo , Dioxigenase FTO Dependente de alfa-Cetoglutarato/genética , Dioxigenase FTO Dependente de alfa-Cetoglutarato/metabolismo , Células Hep G2 , Humanos , Metiltransferases/genética , Metiltransferases/metabolismo , Regiões Promotoras Genéticas , RNA Helicases/genética , RNA Helicases/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteína Supressora de Tumor p53/metabolismo
17.
Methods Mol Biol ; 2192: 59-68, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33230765

RESUMO

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.


Assuntos
Citosina/análogos & derivados , Mitocôndrias/metabolismo , Nucleotídeos/análise , RNA Mitocondrial/química , Transcriptoma , Citosina/análise , DNA Mitocondrial/genética , Perfilação da Expressão Gênica , Processamento Pós-Transcricional do RNA/efeitos dos fármacos , RNA-Seq/métodos , Sulfitos/farmacologia
18.
Mol Neurobiol ; 58(4): 1535-1549, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33210205

RESUMO

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.


Assuntos
Isquemia Encefálica/genética , Isquemia Encefálica/patologia , MicroRNAs/biossíntese , Microglia/metabolismo , Proteínas Argonautas/metabolismo , Linhagem Celular , DNA Helicases/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Glucose/deficiência , Humanos , Peróxido de Hidrogênio/toxicidade , MicroRNAs/genética , Microglia/efeitos dos fármacos , Modelos Biológicos , Estresse Oxidativo/efeitos dos fármacos , Oxigênio , Proteínas de Ligação a Poli-ADP-Ribose/metabolismo , RNA Helicases/metabolismo , Processamento Pós-Transcricional do RNA/efeitos dos fármacos , Proteínas com Motivo de Reconhecimento de RNA/metabolismo , Ribonuclease III/metabolismo , Estresse Fisiológico/efeitos dos fármacos
19.
Blood ; 137(7): 994-999, 2021 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-32915956
20.
Signal Transduct Target Ther ; 5(1): 296, 2020 12 26.
Artigo em Inglês | MEDLINE | ID: mdl-33361765

RESUMO

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.


Assuntos
Carcinoma Hepatocelular/metabolismo , Desdiferenciação Celular/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Fator 3-gama Nuclear de Hepatócito/metabolismo , Neoplasias Hepáticas/metabolismo , Proteínas de Neoplasias/metabolismo , Processamento Pós-Transcricional do RNA/efeitos dos fármacos , RNA Mensageiro/metabolismo , RNA Neoplásico/metabolismo , Sorafenibe/farmacologia , Animais , Anticorpos Heterófilos , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Feminino , Humanos , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , Masculino , Camundongos , Proteínas de Neoplasias/genética , Transplante de Neoplasias , RNA Neoplásico/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA