Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 5.201
Filtrar
Más filtros











Intervalo de año de publicación
1.
Microb Cell Fact ; 23(1): 162, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38824548

RESUMEN

BACKGROUND: Syringic acid (SA) is a high-value natural compound with diverse biological activities and wide applications, commonly found in fruits, vegetables, and herbs. SA is primarily produced through chemical synthesis, nonetheless, these chemical methods have many drawbacks, such as considerable equipment requirements, harsh reaction conditions, expensive catalysts, and numerous by-products. Therefore, in this study, a novel biotransformation route for SA production was designed and developed by using engineered whole cells. RESULTS: An O-methyltransferase from Desulfuromonas acetoxidans (DesAOMT), which preferentially catalyzes a methyl transfer reaction on the meta-hydroxyl group of catechol analogues, was identified. The whole cells expressing DesAOMT can transform gallic acid (GA) into SA when S-adenosyl methionine (SAM) is used as a methyl donor. We constructed a multi-enzyme cascade reaction in Escherichia coli, containing an endogenous shikimate kinase (AroL) and a chorismate lyase (UbiC), along with a p-hydroxybenzoate hydroxylase mutant (PobA**) from Pseudomonas fluorescens, and DesAOMT; SA was biosynthesized from shikimic acid (SHA) by using whole cells catalysis. The metabolic system of chassis cells also affected the efficiency of SA biosynthesis, blocking the chorismate metabolism pathway improved SA production. When the supply of the cofactor NADPH was optimized, the titer of SA reached 133 µM (26.2 mg/L). CONCLUSION: Overall, we designed a multi-enzyme cascade in E. coli for SA biosynthesis by using resting or growing whole cells. This work identified an O-methyltransferase (DesAOMT), which can catalyze the methylation of GA to produce SA. The multi-enzyme cascade containing four enzymes expressed in an engineered E. coli for synthesizing of SA from SHA. The metabolic system of the strain and biotransformation conditions influenced catalytic efficiency. This study provides a new green route for SA biosynthesis.


Asunto(s)
Biocatálisis , Escherichia coli , Ácido Gálico , Ingeniería Metabólica , Ácido Gálico/metabolismo , Ácido Gálico/análogos & derivados , Escherichia coli/metabolismo , Escherichia coli/genética , Ingeniería Metabólica/métodos , Metiltransferasas/metabolismo , Metiltransferasas/genética , Ácido Shikímico/metabolismo , Pseudomonas fluorescens/metabolismo , Pseudomonas fluorescens/enzimología , Pseudomonas fluorescens/genética , Biotransformación
2.
Oncol Res ; 32(6): 1047-1061, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38827317

RESUMEN

Background: Colorectal cancer (CRC) is one of the most frequently diagnosed cancers. In many cases, the poor prognosis of advanced CRC is associated with resistance to treatment with chemotherapeutic drugs such as 5-Fluorouracil (5-FU). The epithelial-to-mesenchymal transition (EMT) and dysregulation in protein methylation are two mechanisms associated with chemoresistance in many cancers. This study looked into the effect of 5-FU dose escalation on EMT and protein methylation in CRC. Materials and Methods: HCT-116, Caco-2, and DLD-1 CRC cell lines were exposed to dose escalation treatment of 5-FU. The motility and invasive potentials of the cells before and after treatment with 5-FU were investigated through wound healing and invasion assays. This was followed by a Western blot which analyzed the protein expressions of the epithelial marker E-cadherin, mesenchymal marker vimentin, and the EMT transcription factor (EMT-TF), the snail family transcriptional repressor 1 (Snail) in the parental and desensitized cells. Western blotting was also conducted to study the protein expressions of the protein methyltransferases (PMTs), Euchromatic histone lysine methyltransferase 2 (EHMT2/G9A), protein arginine methyltransferase (PRMT5), and SET domain containing 7/9 (SETD7/9) along with the global lysine and arginine methylation profiles. Results: The dose escalation method generated 5-FU desensitized CRC cells with distinct morphological features and increased tolerance to high doses of 5-FU. The 5-FU desensitized cells experienced a decrease in migration and invasion when compared to the parental cells. This was reflected in the observed reduction in E-cadherin, vimentin, and Snail in the desensitized cell lines. Additionally, the protein expressions of EHMT2/G9A, PRMT5, and SETD7/9 also decreased in the desensitized cells and global protein lysine and arginine methylation became dysregulated with 5-FU treatment. Conclusion: This study showed that continuous, dose-escalation treatment of 5-FU in CRC cells generated 5-FU desensitized cancer cells that seemed to be less aggressive than parental cells.


Asunto(s)
Movimiento Celular , Neoplasias Colorrectales , Transición Epitelial-Mesenquimal , Fluorouracilo , Humanos , Fluorouracilo/farmacología , Fluorouracilo/administración & dosificación , Transición Epitelial-Mesenquimal/efectos de los fármacos , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/metabolismo , Movimiento Celular/efectos de los fármacos , Línea Celular Tumoral , Antimetabolitos Antineoplásicos/farmacología , Resistencia a Antineoplásicos , Relación Dosis-Respuesta a Droga , Metiltransferasas/metabolismo , Metiltransferasas/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Metilación , Proteína-Arginina N-Metiltransferasas/metabolismo , Proteína-Arginina N-Metiltransferasas/genética
3.
J Exp Clin Cancer Res ; 43(1): 154, 2024 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-38822363

RESUMEN

BACKGROUND: RNA modifications of transfer RNAs (tRNAs) are critical for tRNA function. Growing evidence has revealed that tRNA modifications are related to various disease processes, including malignant tumors. However, the biological functions of methyltransferase-like 1 (METTL1)-regulated m7G tRNA modifications in breast cancer (BC) remain largely obscure. METHODS: The biological role of METTL1 in BC progression were examined by cellular loss- and gain-of-function tests and xenograft models both in vitro and in vivo. To investigate the change of m7G tRNA modification and mRNA translation efficiency in BC, m7G-methylated tRNA immunoprecipitation sequencing (m7G tRNA MeRIP-seq), Ribosome profiling sequencing (Ribo-seq), and polysome-associated mRNA sequencing were performed. Rescue assays were conducted to decipher the underlying molecular mechanisms. RESULTS: The tRNA m7G methyltransferase complex components METTL1 and WD repeat domain 4 (WDR4) were down-regulated in BC tissues at both the mRNA and protein levels. Functionally, METTL1 inhibited BC cell proliferation, and cell cycle progression, relying on its enzymatic activity. Mechanistically, METTL1 increased m7G levels of 19 tRNAs to modulate the translation of growth arrest and DNA damage 45 alpha (GADD45A) and retinoblastoma protein 1 (RB1) in a codon-dependent manner associated with m7G. Furthermore, in vivo experiments showed that overexpression of METTL1 enhanced the anti-tumor effectiveness of abemaciclib, a cyclin-dependent kinases 4 and 6 (CDK4/6) inhibitor. CONCLUSION: Our study uncovered the crucial tumor-suppressive role of METTL1-mediated tRNA m7G modification in BC by promoting the translation of GADD45A and RB1 mRNAs, selectively blocking the G2/M phase of the cell cycle. These findings also provided a promising strategy for improving the therapeutic benefits of CDK4/6 inhibitors in the treatment of BC patients.


Asunto(s)
Neoplasias de la Mama , Metiltransferasas , ARN de Transferencia , Humanos , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Femenino , Ratones , Animales , Metiltransferasas/metabolismo , Metiltransferasas/genética , ARN de Transferencia/genética , ARN de Transferencia/metabolismo , Metilación , Línea Celular Tumoral , Proliferación Celular , Carcinogénesis/genética , Puntos de Control del Ciclo Celular , Biosíntesis de Proteínas , Ensayos Antitumor por Modelo de Xenoinjerto , Ratones Desnudos
4.
FASEB J ; 38(11): e23729, 2024 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-38847786

RESUMEN

Diabetic nephropathy (DN) is one of the common microvascular complications in diabetic patients. Marrow mesenchymal stem cells (MSCs) have attracted attention in DN therapy but the underlying mechanism remains unclear. Here, we show that MSC administration alleviates high glucose (HG)-induced human kidney tubular epithelial cell (HK-2 cell) injury and ameliorates renal injury in DN mice. We identify that Smad2/3 is responsible for MSCs-regulated DN progression. The activity of Smad2/3 was predominantly upregulated in HG-induced HK-2 cell and DN mice and suppressed with MSC administration. Activation of Smad2/3 via transforming growth factor-ß1 (TGF-ß1) administration abrogates the protective effect of MSCs on HG-induced HK-2 cell injury and renal injury of DN mice. Smad2/3 has been reported to interact with methyltransferase of N6-methyladenosine (m6A) complex and we found a methyltransferase, Wilms' tumor 1-associating protein (WTAP), is involved in MSCs-Smad2/3-regulated DN development. Moreover, WTAP overexpression abrogates the improvement of MSCs on HG-induced HK-2 cell injury and renal injury of DN mice. Subsequently, α-enolase (ENO1) is the downstream target of WTAP-mediated m6A modification and contributes to the MSCs-mediated regulation. Collectively, these findings reveal a molecular mechanism in DN progression and indicate that Smad2/3/WTAP/ENO1 may present a target for MSCs-mediated DN therapy.


Asunto(s)
Nefropatías Diabéticas , Células Madre Mesenquimatosas , Proteína Smad2 , Proteína smad3 , Nefropatías Diabéticas/metabolismo , Nefropatías Diabéticas/patología , Animales , Células Madre Mesenquimatosas/metabolismo , Proteína Smad2/metabolismo , Ratones , Humanos , Proteína smad3/metabolismo , Masculino , Ratones Endogámicos C57BL , Adenosina/metabolismo , Adenosina/análogos & derivados , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Transducción de Señal , Metiltransferasas/metabolismo , Metiltransferasas/genética , Trasplante de Células Madre Mesenquimatosas/métodos , Factor de Crecimiento Transformador beta1/metabolismo , Línea Celular
5.
Biol Direct ; 19(1): 44, 2024 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-38849910

RESUMEN

BACKGROUND: The molecular mechanisms of osteosarcoma (OS) are complex. In this study, we focused on the functions of melanoma cell adhesion molecule (MCAM), methyltransferase 3 (METTL3) and insulin like growth factor 2 mRNA binding protein 1 (IGF2BP1) in OS development. METHODS: qRT-PCR assay and western blot assay were performed to determine mRNA and protein expression of MCAM, METTL3, IGF2BP1 and YY1. MTT assay and colony formation assay were conducted to assess cell proliferation. Cell apoptosis, invasion and migration were evaluated by flow cytometry analysis, transwell assay and wound-healing assay, respectively. Methylated RNA Immunoprecipitation (MeRIP), dual-luciferase reporter, Co-IP, RIP and ChIP assays were performed to analyze the relationships of MCAM, METTL3, IGF2BP1 and YY1. The functions of METTL3 and MCAM in tumor growth were explored through in vivo experiments. RESULTS: MCAM was upregulated in OS, and MCAM overexpression promoted OS cell growth, invasion and migration and inhibited apoptosis. METTL3 and IGF2BP1 were demonstrated to mediate the m6A methylation of MCAM. Functionally, METTL3 or IGF2BP1 silencing inhibited OS cell progression, while MCAM overexpression ameliorated the effects. Transcription factor YY1 promoted the transcription level of METTL3 and regulated METTL3 expression in OS cells. Additionally, METTL3 deficiency suppressed tumor growth in vivo, while MCAM overexpression abated the effect. CONCLUSION: YY1/METTL3/IGF2BP1/MCAM axis aggravated OS development, which might provide novel therapy targets for OS.


Asunto(s)
Adenosina , Metiltransferasas , Osteosarcoma , Proteínas de Unión al ARN , Osteosarcoma/genética , Osteosarcoma/metabolismo , Metiltransferasas/metabolismo , Metiltransferasas/genética , Humanos , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Adenosina/análogos & derivados , Adenosina/metabolismo , Adenosina/genética , Línea Celular Tumoral , Animales , Ratones , Proliferación Celular , Neoplasias Óseas/genética , Neoplasias Óseas/metabolismo , Progresión de la Enfermedad , Ratones Desnudos , Apoptosis , Movimiento Celular , Regulación Neoplásica de la Expresión Génica
6.
Crit Rev Immunol ; 44(6): 49-61, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38848293

RESUMEN

BACKGROUND: Sustained expression of the long noncoding RNA (lncRNA) LINC01106 in tumors is crucial for the malignant phenotype of tumor cells. Nevertheless, the mechanisms and clinical effects of LINC01106 in lung adenocarcinoma (LUAD) are limited. This study shows the effect of vir-like m6A methyltransferase-associated (KIAA1429)-mediated N6-methyladenosine (m6A) modification on steady LINC01106 expression on LUAD progression. METHODS: Quantitative real-time polymerase chain reaction (qRT-PCR) was performed to determine LINC01106 and KIAA1429 levels in LUAD tissues. Transwell, 5-ethynyl-2'-deoxyuridine (EdU), and cell counting kit-8 (CCK-8) assays were used to analyze the functional roles of LINC01106. A xenograft was constructed to verify the function of silencing LINC01106 in tumor growth. The regulatory role of LINC01106 was investigated using methylated RNA immunoprecipitation (MeRIP), qRT-PCR, and the actinomycin D assay. Western blotting was used to identify key proteins in the JAK/STAT3 (JAK2, STAT3) pathway. RESULTS: LINC01106 and KIAA1429 were highly expressed in LUAD, and LINC01106 was interconnected with high tumor grade, stage, and poor prognosis. Data revealed that LINC01106 inhibition reduced LUAD cell proliferation, invasion, and migration and restrained LUAD cell tumorigenicity. In addition, LINC01106 silencing reduced phosphorylated JAK2 and STAT3 levels. KIAA1429-mediated LINC01106 enhances its m6A modification and expression in LUAD cells. Moreover, KIAA1429 promotion eliminated the malignant phenotypic suppression induced by low expression in LUAD cells. CONCLUSION: This study showed that KIAA1429 enhanced LINC01106 m6A modification to promote LUAD development. These results may lead to a better understanding of the mechanism of KIAA1429-m6A-LINC01106 in LUAD and offer a valuable therapeutic target for LUAD.


Asunto(s)
Adenocarcinoma del Pulmón , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares , ARN Largo no Codificante , Factor de Transcripción STAT3 , Transducción de Señal , Humanos , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción STAT3/genética , Adenocarcinoma del Pulmón/genética , Adenocarcinoma del Pulmón/metabolismo , Adenocarcinoma del Pulmón/patología , Animales , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Ratones , Proliferación Celular/genética , Línea Celular Tumoral , Adenosina/análogos & derivados , Adenosina/metabolismo , Metiltransferasas/metabolismo , Metiltransferasas/genética , Ratones Desnudos , Janus Quinasa 2/metabolismo , Janus Quinasa 2/genética , Movimiento Celular/genética , Femenino , Quinasas Janus/metabolismo , Masculino , Proteínas de Unión al ARN
7.
Breast Cancer Res ; 26(1): 94, 2024 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-38844963

RESUMEN

BACKGROUND: RNA m5C methylation has been extensively implicated in the occurrence and development of tumors. As the main methyltransferase, NSUN2 plays a crucial regulatory role across diverse tumor types. However, the precise impact of NSUN2-mediated m5C modification on breast cancer (BC) remains unclear. Our study aims to elucidate the molecular mechanism underlying how NSUN2 regulates the target gene HGH1 (also known as FAM203) through m5C modification, thereby promoting BC progression. Additionally, this study targets at preliminarily clarifying the biological roles of NSUN2 and HGH1 in BC. METHODS: Tumor and adjacent tissues from 5 BC patients were collected, and the m5C modification target HGH1 in BC was screened through RNA sequencing (RNA-seq) and single-base resolution m5C methylation sequencing (RNA-BisSeq). Methylation RNA immunoprecipitation-qPCR (MeRIP-qPCR) and RNA-binding protein immunoprecipitation-qPCR (RIP-qPCR) confirmed that the methylation molecules NSUN2 and YBX1 specifically recognized and bound to HGH1 through m5C modification. In addition, proteomics, co-immunoprecipitation (co-IP), and Ribosome sequencing (Ribo-Seq) were used to explore the biological role of HGH1 in BC. RESULTS: As the main m5C methylation molecule, NSUN2 is abnormally overexpressed in BC and increases the overall level of RNA m5C. Knocking down NSUN2 can inhibit BC progression in vitro or in vivo. Combined RNA-seq and RNA-BisSeq analysis identified HGH1 as a potential target of abnormal m5C modifications. We clarified the mechanism by which NSUN2 regulates HGH1 expression through m5C modification, a process that involves interactions with the YBX1 protein, which collectively impacts mRNA stability and protein synthesis. Furthermore, this study is the first to reveal the binding interaction between HGH1 and the translation elongation factor EEF2, providing a comprehensive understanding of its ability to regulate transcript translation efficiency and protein synthesis in BC cells. CONCLUSIONS: This study preliminarily clarifies the regulatory role of the NSUN2-YBX1-m5C-HGH1 axis from post-transcriptional modification to protein translation, revealing the key role of abnormal RNA m5C modification in BC and suggesting that HGH1 may be a new epigenetic biomarker and potential therapeutic target for BC.


Asunto(s)
Neoplasias de la Mama , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Metiltransferasas , Estabilidad del ARN , Proteína 1 de Unión a la Caja Y , Humanos , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Neoplasias de la Mama/metabolismo , Femenino , Metilación , Metiltransferasas/metabolismo , Metiltransferasas/genética , Proteína 1 de Unión a la Caja Y/metabolismo , Proteína 1 de Unión a la Caja Y/genética , Ratones , Animales , Línea Celular Tumoral , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proliferación Celular
8.
Nat Commun ; 15(1): 5033, 2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38866783

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease due to gradual motoneurons (MN) degeneration. Among the processes associated to ALS pathogenesis, there is the formation of cytoplasmic inclusions produced by aggregation of mutant proteins, among which the RNA binding protein FUS. Here we show that, in neuronal cells and in iPSC-derived MN expressing mutant FUS, such inclusions are significantly reduced in number and dissolve faster when the RNA m6A content is diminished. Interestingly, stress granules formed in ALS conditions showed a distinctive transcriptome with respect to control cells, which reverted to similar to control after m6A downregulation. Notably, cells expressing mutant FUS were characterized by higher m6A levels suggesting a possible link between m6A homeostasis and pathological aggregates. Finally, we show that FUS inclusions are reduced also in patient-derived fibroblasts treated with STM-2457, an inhibitor of METTL3 activity, paving the way for its possible use for counteracting aggregate formation in ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral , Células Madre Pluripotentes Inducidas , Neuronas Motoras , Proteína FUS de Unión a ARN , Proteína FUS de Unión a ARN/metabolismo , Proteína FUS de Unión a ARN/genética , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/patología , Humanos , Neuronas Motoras/metabolismo , Neuronas Motoras/patología , Células Madre Pluripotentes Inducidas/metabolismo , Gránulos Citoplasmáticos/metabolismo , Fibroblastos/metabolismo , Adenosina/metabolismo , Adenosina/análogos & derivados , Metiltransferasas/metabolismo , Metiltransferasas/genética , Mutación , Cuerpos de Inclusión/metabolismo , Gránulos de Estrés/metabolismo , Transcriptoma
9.
Cell Mol Life Sci ; 81(1): 256, 2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38866991

RESUMEN

Pulmonary hypertension (PH) is characterized by vascular remodeling predominantly driven by a phenotypic switching in pulmonary artery smooth muscle cells (PASMCs). However, the underlying mechanisms for this phenotypic alteration remain incompletely understood. Here, we identified that RNA methyltransferase METTL3 is significantly elevated in the lungs of hypoxic PH (HPH) mice and rats, as well as in the pulmonary arteries (PAs) of HPH rats. Targeted deletion of Mettl3 in smooth muscle cells exacerbated hemodynamic consequences of hypoxia-induced PH and accelerated pulmonary vascular remodeling in vivo. Additionally, the absence of METTL3 markedly induced phenotypic switching in PASMCs in vitro. Mechanistically, METTL3 depletion attenuated m6A modification and hindered the processing of pri-miR-143/145, leading to a downregulation of miR-143-3p and miR-145-5p. Inhibition of hnRNPA2B1, an m6A mediator involved in miRNA maturation, similarly resulted in a significant reduction of miR-143-3p and miR-145-5p. We demonstrated that miR-145-5p targets Krüppel-like factor 4 (KLF4) and miR-143-3p targets fascin actin-bundling protein 1 (FSCN1) in PASMCs. The decrease of miR-145-5p subsequently induced an upregulation of KLF4, which in turn suppressed miR-143/145 transcription, establishing a positive feedback circuit between KLF4 and miR-143/145. This regulatory circuit facilitates the persistent suppression of contractile marker genes, thereby sustaining PASMC phenotypic switch. Collectively, hypoxia-induced upregulation of METTL3, along with m6A mediated regulation of miR-143/145, might serve as a protective mechanism against phenotypic switch of PASMCs. Our results highlight a potential therapeutic strategy targeting m6A modified miR-143/145-KLF4 loop in the treatment of PH.


Asunto(s)
Adenosina , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel , Metiltransferasas , MicroARNs , Miocitos del Músculo Liso , Arteria Pulmonar , Factor 4 Similar a Kruppel/metabolismo , Animales , MicroARNs/genética , MicroARNs/metabolismo , Arteria Pulmonar/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Factores de Transcripción de Tipo Kruppel/genética , Miocitos del Músculo Liso/metabolismo , Ratones , Adenosina/análogos & derivados , Adenosina/metabolismo , Metiltransferasas/metabolismo , Metiltransferasas/genética , Ratas , Fenotipo , Masculino , Hipertensión Pulmonar/metabolismo , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/patología , Músculo Liso Vascular/metabolismo , Ratones Endogámicos C57BL , Remodelación Vascular/genética , Ratas Sprague-Dawley , Humanos
10.
Planta ; 259(6): 152, 2024 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-38735012

RESUMEN

MAIN CONCLUSION: Overexpression of Artemisia annua jasmonic acid carboxyl methyltransferase (AaJMT) leads to enhanced artemisinin content in Artemisia annua. Artemisinin-based combination therapies remain the sole deterrent against deadly disease malaria and Artemisia annua remains the only natural producer of artemisinin. In this study, the 1101 bp gene S-adenosyl-L-methionine (SAM): Artemisia annua jasmonic acid carboxyl methyltransferase (AaJMT), was characterised from A. annua, which converts jasmonic acid (JA) to methyl jasmonate (MeJA). From phylogenetic analysis, we confirmed that AaJMT shares a common ancestor with Arabidopsis thaliana, Eutrema japonica and has a close homology with JMT of Camellia sinensis. Further, the Clustal Omega depicted that the conserved motif I, motif III and motif SSSS (serine) required to bind SAM and JA, respectively, are present in AaJMT. The relative expression of AaJMT was induced by wounding, MeJA and salicylic acid (SA) treatments. Additionally, we found that the recombinant AaJMT protein catalyses the synthesis of MeJA from JA with a Km value of 37.16 µM. Moreover, site-directed mutagenesis of serine-151 in motif SSSS to tyrosine, asparagine-10 to threonine and glutamine-25 to histidine abolished the enzyme activity of AaJMT, thus indicating their determining role in JA substrate binding. The GC-MS analysis validated that mutant proteins of AaJMT were unable to convert JA into MeJA. Finally, the artemisinin biosynthetic and trichome developmental genes were upregulated in AaJMT overexpression transgenic lines, which in turn increased the artemisinin content.


Asunto(s)
Acetatos , Artemisia annua , Artemisininas , Ciclopentanos , Metiltransferasas , Oxilipinas , Filogenia , Artemisia annua/genética , Artemisia annua/enzimología , Artemisia annua/metabolismo , Ciclopentanos/metabolismo , Ciclopentanos/farmacología , Artemisininas/metabolismo , Oxilipinas/metabolismo , Oxilipinas/farmacología , Metiltransferasas/metabolismo , Metiltransferasas/genética , Acetatos/farmacología , Acetatos/metabolismo , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo , Plantas Modificadas Genéticamente , Regulación de la Expresión Génica de las Plantas , Ácido Salicílico/metabolismo
11.
FASEB J ; 38(11): e23693, 2024 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-38809685

RESUMEN

N6-methylated adenosine (m6A) is a crucial RNA modification in eukaryotes, particularly in cancer. However, its role in cervical cancer (CC) is unclear. We aimed to elucidate the part of m6A in CC by analyzing methyltransferase-like 3 (METTL3) expression, identifying downstream targets, and exploring the underlying mechanism. We assessed METTL3 expression in CC using western blotting, quantitative polymerase chain reaction (qPCR), and immunohistochemistry. In vitro and in vivo experiments examined METTL3's role in CC. We employed RNA sequencing, methylated RNA immunoprecipitation sequencing, qPCR, and RNA immunoprecipitation qPCR to explore METTL3's mechanism in CC. METTL3 expression was upregulated in CC, promoting cell proliferation and metastasis. METTL3 knockdown inhibited human cervical cancer by inactivating AKT/mTOR signaling pathway. METTL3-mediated m6A modification was observed in CC cells, targeting phosphodiesterase 3A (PDE3A). METTL3 catalyzed m6A modification on PDE3A mRNA through YTH domain family protein 3 (YTHDF3). Our study indicated the mechanism of m6A modification in CC and suggested the METTL3/YTHDF3/PDE3A axis as a potential clinical target for CC treatment.


Asunto(s)
Adenosina , Proliferación Celular , Metiltransferasas , Neoplasias del Cuello Uterino , Metiltransferasas/metabolismo , Metiltransferasas/genética , Neoplasias del Cuello Uterino/metabolismo , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/patología , Humanos , Femenino , Adenosina/análogos & derivados , Adenosina/metabolismo , Animales , Ratones , Regulación Neoplásica de la Expresión Génica , Línea Celular Tumoral , Ratones Desnudos , Transducción de Señal , Ratones Endogámicos BALB C
12.
Zhongguo Zhong Yao Za Zhi ; 49(10): 2680-2688, 2024 May.
Artículo en Chino | MEDLINE | ID: mdl-38812168

RESUMEN

Methyleugenol is one of the main active constituents in the volatile oil of the traditional Chinese medicine Asari Radix et Rhizoma. It possesses various pharmacological effects such as analgesic, anesthetic, and anti-inflammatory properties. In biosynthesis, the initial precursor phenylalanine is finally converted into methyleugenol through a series of intermediate compounds including coniferyl acid, courmaryl acid, caffeic acid, ferulic acid/ferulic-CoA, coniferyl aldehyde, conferyl alcohol, cnfiferyl acetate, and eugenol/isoeugenol, which are produced through catalysis of a large number of enzymes. Eugenol O-methyltransferase(EOMT) is one of the key enzymes in the biosynthesis pathway, capable of methylating eugenol on the para-site hydroxyl group of the benzene ring, thereby generating methyleugenol. Here, an(iso)eugenol O-methyltransferase(IEMT) gene was cloned for the first time from Asarum siebo-ldii, holding an open reading frame that consisted of 1 113 bp and encoded a protein containing 370 amino acid residues. Bioinformatics analysis results showed that this protein was equipped with the characteristic structural domains of methyltransferases such as S-adenosylmethionine(SAM) binding sites and dimerization domains. The prokaryotic expression recombinant plasmid pET28a(+)-AsIEMT was constructed, and the candidate protein was induced and purified. In vitro enzyme assays confirmed that AsIEMT had dual functions. The enzyme could catalyze the production either of methyleugenol from eugenol or of methylisoeugenol from isoeugenol, although the latter was more prevalent. When isoeugenol was used as the substrate, the kinetics parameters K_m and V_(max) of catalytic reaction were(0.90±0.06) mmol·L~(-1) and(1.32±0.04)nmol·s~(-1)·mg~(-1), respectively. This study expanded our understandings of critical enzyme genes involved in phenylpropanoid metabolic pathways, and would facilitate the elucidation of quality formation mechanisms of the TCM Asari Radix et Rhizoma.


Asunto(s)
Asarum , Eugenol , Metiltransferasas , Metiltransferasas/genética , Metiltransferasas/química , Metiltransferasas/metabolismo , Eugenol/análogos & derivados , Eugenol/metabolismo , Eugenol/química , Asarum/genética , Asarum/química , Asarum/enzimología , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo , Proteínas de Plantas/química , Filogenia , Secuencia de Aminoácidos , Clonación Molecular
13.
Int J Nanomedicine ; 19: 4181-4197, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38766656

RESUMEN

Purpose: The committed differentiation fate regulation has been a difficult problem in the fields of stem cell research, evidence showed that nanomaterials could promote the differentiation of stem cells into specific cell types. Layered double hydroxide (LDH) nanoparticles possess the regulation function of stem cell fate, while the underlying mechanism needs to be investigated. In this study, the process of embryonic stem cells (ESCs) differentiate to neural progenitor cells (NPCs) by magnesium aluminum LDH (MgAl-LDH) was investigated. Methods: MgAl-LDH with diameters of 30, 50, and 100 nm were synthesized and characterized, and their effects on the cytotoxicity and differentiation of NPCs were detected in vitro. Dot blot and MeRIP-qPCR were performed to detect the level of m6A RNA methylation in nanoparticles-treated cells. Results: Our work displayed that LDH nanoparticles of three different sizes were biocompatible with NPCs, and the addition of MgAl-LDH could significantly promote the process of ESCs differentiate to NPCs. 100 nm LDH has a stronger effect on promoting NPCs differentiation compared to 30 nm and 50 nm LDH. In addition, dot blot results indicated that the enhanced NPCs differentiation by MgAl-LDH was closely related to m6A RNA methylation process, and the major modification enzyme in LDH controlled NPCs differentiation may be the m6A RNA methyltransferase METTL3. The upregulated METTL3 by LDH increased the m6A level of Sox1 mRNA, enhancing its stability. Conclusion: This work reveals that MgAl-LDH nanoparticles can regulate the differentiation of ESCs into NPCs by increasing m6A RNA methylation modification of Sox1.


Asunto(s)
Diferenciación Celular , Nanopartículas , Células-Madre Neurales , Diferenciación Celular/efectos de los fármacos , Animales , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Ratones , Nanopartículas/química , Metilación/efectos de los fármacos , Hidróxidos/química , Hidróxidos/farmacología , Metiltransferasas/metabolismo , Metiltransferasas/genética , Tamaño de la Partícula , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/citología , Adenosina/farmacología , Adenosina/química , Adenosina/análogos & derivados , Hidróxido de Aluminio/química , Hidróxido de Aluminio/farmacología , Hidróxido de Magnesio/química , Hidróxido de Magnesio/farmacología
14.
J Biochem Mol Toxicol ; 38(6): e23749, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38800929

RESUMEN

Colon adenocarcinoma (COAD) is a common and fatal malignant tumor of digestive system with complex etiology. 5-Methylcytosine (m5C) modification of RNA by the NSUN gene family (NSUN1-NSUN7) and DNMT2 reshape cell biology and regulate tumor development. However, the expression profile, prognostic significance and function of these m5C modifiers in COAD remain largely unclear. By mining multiple integrated tumor databases, we found that NSUN1, NSUN2, NSUN5, and NSUN6 were overexpressed in COAD tumor samples relative to normal samples. Clinically, high expression of NSUN6 was significantly associated with shorter survival (including both disease-free survival and overall survival) in COAD patients. NSUN6 was further confirmed to be upregulated at both tissue and cellular levels of COAD, suggesting that NSUN6 plays a critical role in disease progression. Through comprehensive gene enrichment analysis and cell-based functional validation, it was revealed that NSUN6 promoted the cell cycle progression and cell proliferation of COAD. Mechanistically, NSUN6 upregulates the expression of oncogenic METTL3 and catalyzes its m5C modification in COAD cells. Overexpression of METTL3 significantly relieved the cell cycle inhibition of COAD caused by NSUN6 deficiency. Furthermore, NSUN6 was negatively associated with the abundance of infiltrating immune cells in COAD tumors, such as activated B cells, natural killer cells, effector memory CD8 T cells, and regulatory T cells. Importantly, pan-cancer analysis further uncovered that NSUN6 was dysregulated and heterogeneous in various tumors. Thus our findings extend the role of m5C transferase in COAD and suggest that NSUN6 is a potential biomarker and target for this malignancy.


Asunto(s)
5-Metilcitosina , Adenocarcinoma , Neoplasias del Colon , Progresión de la Enfermedad , Metiltransferasas , Humanos , Metiltransferasas/metabolismo , Metiltransferasas/genética , Neoplasias del Colon/patología , Neoplasias del Colon/metabolismo , Neoplasias del Colon/genética , 5-Metilcitosina/metabolismo , 5-Metilcitosina/análogos & derivados , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Adenocarcinoma/genética , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica
15.
Sci Rep ; 14(1): 12090, 2024 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-38802444

RESUMEN

Micropeptides hidden in long non-coding RNAs (lncRNAs) have been uncovered to program various cell-biological changes associated with malignant transformation-glioblastoma (GBM) cascade. Here, we identified and characterized a novel hidden micropeptide implicated in GBM. We screened potential candidate lncRNAs by establishing a workflow involving ribosome-bound lncRNAs, publicly available MS/MS data, and prognosis-related lncRNAs. Micropeptide expression was detected by western blot (WB), immunofluorescence (IF), and immunohistochemistry (IHC). Cell proliferation rate was assessed by calcein/PI staining and EdU assay. Proteins interacted with the micropeptide were analyzed by proteomics after co-immunoprecipitation (Co-IP). We discovered that lncRNA AF127577.4 indeed encoded an endogenous micropeptide, named AF127577.4-ORF. AF127577.4-ORF was associated with GBM clinical grade. In vitro, AF127577.4-ORF could suppress GBM cell proliferation. Moreover, AF127577.4-ORF reduced m6A methylation level of GBM cells. Mechanistically, AF127577.4-ORF diminished ERK2 interaction with m6A reader methyltransferase like 3 (METTL3) and downregulated phosphorylated ERK (p-ERK) level. The ERK inhibitor reduced p-ERK level and downregulated METTL3 protein expression. AF127577.4-ORF weakened the stability of METTL3 protein by ERK. Also, AF127577.4-ORF suppressed GBM cell proliferation via METTL3. Our study identifies a novel micropeptide AF127577.4-ORF hidden in a lncRNA, with a potent anti-proliferating function in GBM by diminishing METTL3 protein stability by reducing the ERK2/METTL3 interaction. This micropeptide may be beneficial for development of therapeutic strategies against GBM.


Asunto(s)
Proliferación Celular , Glioblastoma , Metiltransferasas , Proteína Quinasa 1 Activada por Mitógenos , ARN Largo no Codificante , Humanos , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Glioblastoma/genética , Glioblastoma/metabolismo , Glioblastoma/patología , Línea Celular Tumoral , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/genética , Metiltransferasas/metabolismo , Metiltransferasas/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Péptidos/metabolismo
16.
Int J Biol Sci ; 20(7): 2491-2506, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38725850

RESUMEN

Colon inflammation is characterized by disturbances in the intestinal microbiota and inflammation. Melatonin (Mel) can improve colon inflammation. However, the underlying mechanism remains unclear. Recent studies suggest that m6A methylation modification may play an important role in inflammatory responses. This study aimed to explore the effects of melatonin and LPS-mediated m6A methylation on colon inflammation. Our study found that melatonin inhibits M1 macrophages, activates M2 macrophages, inhibit the secretion of pro-inflammatory factors, maintain colon homeostasis and improves colon inflammation through MTNR1B. In addition, the increased methylation level of m6A is associated with the occurrence of colon inflammation, and melatonin can also reduce the level of colon methylation to improve colon inflammation. Among them, the main methylated protein METTL3 can be inhibited by melatonin through MTNR1B. In a word, melatonin regulates m6A methylation by improving abnormal METTL3 protein level to reshape the microflora and activate macrophages to improve colon inflammation, mainly through MTNR1B.


Asunto(s)
Adenosina , Lipopolisacáridos , Macrófagos , Melatonina , Melatonina/farmacología , Melatonina/metabolismo , Animales , Ratones , Adenosina/metabolismo , Adenosina/análogos & derivados , Adenosina/farmacología , Metilación/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos/efectos de los fármacos , Metiltransferasas/metabolismo , Metiltransferasas/genética , Inflamación/metabolismo , Colon/metabolismo , Colon/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , Colitis/inducido químicamente , Colitis/metabolismo , Receptor de Melatonina MT2/metabolismo , Receptor de Melatonina MT2/genética , Células RAW 264.7
17.
Cell Death Dis ; 15(5): 338, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38744809

RESUMEN

Epitranscriptomic RNA modifications are crucial for the maintenance of glioma stem cells (GSCs), the most malignant cells in glioblastoma (GBM). 3-methylcytosine (m3C) is a new epitranscriptomic mark on RNAs and METTL8 represents an m3C writer that is dysregulated in cancer. Although METTL8 has an established function in mitochondrial tRNA (mt-tRNA) m3C modification, alternative splicing of METTL8 can also generate isoforms that localize to the nucleolus where they may regulate R-loop formation. The molecular basis for METTL8 dysregulation in GBM, and which METTL8 isoform(s) may influence GBM cell fate and malignancy remain elusive. Here, we investigated the role of METTL8 in regulating GBM stemness and tumorigenicity. In GSC, METTL8 is exclusively localized to the mitochondrial matrix where it installs m3C on mt-tRNAThr/Ser(UCN) for mitochondrial translation and respiration. High expression of METTL8 in GBM is attributed to histone variant H2AZ-mediated chromatin accessibility of HIF1α and portends inferior glioma patient outcome. METTL8 depletion impairs the ability of GSC to self-renew and differentiate, thus retarding tumor growth in an intracranial GBM xenograft model. Interestingly, METTL8 depletion decreases protein levels of HIF1α, which serves as a transcription factor for several receptor tyrosine kinase (RTK) genes, in GSC. Accordingly, METTL8 loss inactivates the RTK/Akt axis leading to heightened sensitivity to Akt inhibitor treatment. These mechanistic findings, along with the intimate link between METTL8 levels and the HIF1α/RTK/Akt axis in glioma patients, guided us to propose a HIF1α/Akt inhibitor combination which potently compromises GSC proliferation/self-renewal in vitro. Thus, METTL8 represents a new GBM dependency that is therapeutically targetable.


Asunto(s)
Glioblastoma , Subunidad alfa del Factor 1 Inducible por Hipoxia , Metiltransferasas , Células Madre Neoplásicas , Proteínas Proto-Oncogénicas c-akt , Humanos , Glioblastoma/metabolismo , Glioblastoma/patología , Glioblastoma/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Animales , Metiltransferasas/metabolismo , Metiltransferasas/genética , Ratones , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/genética , Línea Celular Tumoral , Carcinogénesis/genética , Carcinogénesis/patología , Carcinogénesis/metabolismo , Transducción de Señal , ARN de Transferencia/metabolismo , ARN de Transferencia/genética , Mitocondrias/metabolismo , Regulación Neoplásica de la Expresión Génica , Ratones Desnudos , Proliferación Celular
18.
Biochem Biophys Res Commun ; 716: 150039, 2024 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-38701556

RESUMEN

The objective of this study was to better characterize the role of the glutamine transporter SLC38A1 in cervical cancer and explore the underlying mechanisms. Data from public databases and clinical cervical cancer tissue samples were used to assess the expression of SLC38A1 and its prognostic significance. Immunohistochemical staining, qRT-PCR, and Western blotting were used to evaluate the expression of relevant genes and proteins. Cell viability, cell cycle, apoptosis, and intracellular glutamine content were measured using CCK-8, flow cytometry, and biochemical assays. Additionally, the RNA immunoprecipitation (RIP) assay was used to examine the impact of METTL3/IGF2BP3 on the m6A modification of the SLC38A1 3'UTR. Both cervical cancer specimens and cells showed significantly increased expression of SLC38A1 and its expression correlated with an unfavorable prognosis. Knockdown of SLC38A1 inhibited cell viability and cell cycle progression, induced apoptosis, and suppressed tumor growth in vivo. Glutaminase-1 inhibitor CB-839 reversed the effects of SLC38A1 overexpression. METTL3 promoted m6A modification of SLC38A1 and enhanced its mRNA stability through IGF2BP3 recruitment. Moreover, METTL3 silencing inhibited cell viability, cell cycle progression, intracellular glutamine content, and induced apoptosis, but these effects were reversed by SLC38A1 overexpression. In conclusion, METTL3-mediated m6A methylation of SLC38A1 stimulates cervical cancer progression. SLC38A1 inhibition is a potential therapeutic strategy for cervical cancer.


Asunto(s)
Adenosina , Metiltransferasas , Neoplasias del Cuello Uterino , Neoplasias del Cuello Uterino/metabolismo , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/patología , Humanos , Femenino , Metiltransferasas/metabolismo , Metiltransferasas/genética , Adenosina/análogos & derivados , Adenosina/metabolismo , Adenosina/genética , Metilación , Línea Celular Tumoral , Proliferación Celular/genética , Animales , Sistema de Transporte de Aminoácidos A/metabolismo , Sistema de Transporte de Aminoácidos A/genética , Apoptosis/genética , Regulación Neoplásica de la Expresión Génica , Ratones Desnudos , Ratones , Pronóstico , Supervivencia Celular/genética
19.
Biochem Biophys Res Commun ; 716: 150011, 2024 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-38704890

RESUMEN

Methionine adenosyltransferase 2 A (MAT2A) mediates the synthesis of methyl donor S-Adenosylmethionine (SAM), providing raw materials for methylation reactions in cells. MAT2A inhibitors are currently used for the treatment of tumors with methylthioadenosine phosphorylase (MTAP) deficiency in clinical research. Methyltransferase like 3 (METTL3) catalyzes N6-methyladenosine (m6A) modification of mRNA in mammalian cells using SAM as the substrate which has been shown to affect the tumorigenesis of non-small cell lung cancer (NSCLC) from multiple perspectives. MAT2A-induced SAM depletion may have the potential to inhibit the methyl transfer function of METTL3. Therefore, in order to expand the applicability of inhibitors, improve anti-tumor effects and reduce toxicity, the combinational effect of MAT2A inhibitor AG-270 and METTL3 inhibitor STM2457 was evaluated in NSCLC. The results showed that this combination induced cell apoptosis rather than cell cycle arrest, which was non-tissue-specific and was independent of MTAP expression status, resulting in a significant synergistic anti-tumor effect. We further elucidated that the combination-induced enhanced apoptosis was associated with the decreased m6A level, leading to downregulation of PI3K/AKT protein, ultimately activating the apoptosis-related proteins. Unexpectedly, although combination therapy resulted in metabolic recombination, no significant change in methionine metabolic metabolites was found. More importantly, the combination also exerted synergistic effects in vivo. In summary, the combination of MAT2A inhibitor and METTL3 inhibitor showed synergistic effects both in vivo and in vitro, which laid a theoretical foundation for expanding the clinical application research of the two types of drugs.


Asunto(s)
Apoptosis , Carcinoma de Pulmón de Células no Pequeñas , Sinergismo Farmacológico , Neoplasias Pulmonares , Metionina Adenosiltransferasa , Metiltransferasas , Metionina Adenosiltransferasa/metabolismo , Metionina Adenosiltransferasa/antagonistas & inhibidores , Metionina Adenosiltransferasa/genética , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Humanos , Apoptosis/efectos de los fármacos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/metabolismo , Animales , Metiltransferasas/metabolismo , Metiltransferasas/antagonistas & inhibidores , Línea Celular Tumoral , Antineoplásicos/farmacología , Inhibidores Enzimáticos/farmacología , Ratones , Ratones Desnudos , Ratones Endogámicos BALB C , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Cell Mol Biol Lett ; 29(1): 69, 2024 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-38741032

RESUMEN

BACKGROUND: Pulmonary hypertension (PH) is a progressive disease characterized by pulmonary vascular remodeling. Increasing evidence indicates that endothelial-to-mesenchymal transition (EndMT) in pulmonary artery endothelial cells (PAECs) is a pivotal trigger initiating this remodeling. However, the regulatory mechanisms underlying EndMT in PH are still not fully understood. METHODS: Cytokine-induced hPAECs were assessed using RNA methylation quantification, qRT-PCR, and western blotting to determine the involvement of N6-methyladenosine (m6A) methylation in EndMT. Lentivirus-mediated silencing, overexpression, tube formation, and wound healing assays were utilized to investigate the function of METTL3 in EndMT. Endothelial-specific gene knockout, hemodynamic measurement, and immunostaining were performed to explore the roles of METTL3 in pulmonary vascular remodeling and PH. RNA-seq, RNA Immunoprecipitation-based qPCR, mRNA stability assay, m6A mutation, and dual-luciferase assays were employed to elucidate the mechanisms of RNA methylation in EndMT. RESULTS: The global levels of m6A and METTL3 expression were found to decrease in TNF-α- and TGF-ß1-induced EndMT in human PAECs (hPAECs). METTL3 inhibition led to reduced endothelial markers (CD31 and VE-cadherin) and increased mesenchymal markers (SM22 and N-cadherin) as well as EndMT-related transcription factors (Snail, Zeb1, Zeb2, and Slug). The endothelial-specific knockout of Mettl3 promoted EndMT and exacerbated pulmonary vascular remodeling and hypoxia-induced PH (HPH) in mice. Mechanistically, METTL3-mediated m6A modification of kruppel-like factor 2 (KLF2) plays a crucial role in the EndMT process. KLF2 overexpression increased CD31 and VE-cadherin levels while decreasing SM22, N-cadherin, and EndMT-related transcription factors, thereby mitigating EndMT in PH. Mutations in the m6A site of KLF2 mRNA compromise KLF2 expression, subsequently diminishing its protective effect against EndMT. Furthermore, KLF2 modulates SM22 expression through direct binding to its promoter. CONCLUSIONS: Our findings unveil a novel METTL3/KLF2 pathway critical for protecting hPAECs against EndMT, highlighting a promising avenue for therapeutic investigation in PH.


Asunto(s)
Adenosina , Células Endoteliales , Transición Epitelial-Mesenquimal , Hipertensión Pulmonar , Factores de Transcripción de Tipo Kruppel , Metiltransferasas , Adenosina/análogos & derivados , Adenosina/metabolismo , Animales , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/metabolismo , Humanos , Metiltransferasas/metabolismo , Metiltransferasas/genética , Ratones , Células Endoteliales/metabolismo , Transición Epitelial-Mesenquimal/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Factores de Transcripción de Tipo Kruppel/genética , Arteria Pulmonar/metabolismo , Arteria Pulmonar/patología , Metilación , Ratones Endogámicos C57BL , Cadherinas/metabolismo , Cadherinas/genética , Masculino , Remodelación Vascular/genética , Células Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA