Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 36.606
Filtrer
2.
Med Oncol ; 41(9): 218, 2024 Aug 05.
Article de Anglais | MEDLINE | ID: mdl-39103705

RÉSUMÉ

Gastric cancers (GCs) are among the most common and fatal malignancies in the world. Despite our increasing understanding of the molecular mechanisms underlying GC, further biomarkers are still needed for more in-depth examination, focused prognosis, and treatment. GC is one among the long non-coding RNAs, or lncRNAs, that have emerged as key regulators of the pathophysiology of cancer. This comprehensive review focuses on the diverse functions of long noncoding RNAs (lncRNAs) in the development of GC and their interactions with important intracellular signaling pathways. LncRNAs affect GC-related carcinogenic signaling cascades including pathways for EGFR, PI3K/AKT/mTOR, p53, Wnt/ß-catenin, JAK/STAT, Hedgehog, NF-κB, and hypoxia-inducible factor. Dysregulated long non-coding RNA (lncRNA) expression has been associated with multiple characteristics of cancer, such as extended growth, apoptosis resistance, enhanced invasion and metastasis, angiogenesis, and therapy resistance. For instance, lncRNAs such as HOTAIR, MALAT1, and H19 promote the development of GC via altering these pathways. Beyond their main roles, GC lncRNAs exhibit potential as diagnostic and prognostic biomarkers. The overview discusses CRISPR/Cas9 genome-modifying methods, antisense oligonucleotides, small molecules, and RNA interference as potential therapeutic approaches to regulate the expression of long noncoding RNAs (lncRNAs). An in-depth discussion of the intricate functions that lncRNAs play in the development of the majority of stomach malignancies is provided in this review. It provides the groundwork for future translational research in lncRNA-based whole processes toward GC by highlighting their carcinogenic effects, regulatory roles in significant signaling cascades, and practical scientific uses as biomarkers and therapeutic targets.


Sujet(s)
ARN long non codant , Transduction du signal , Tumeurs de l'estomac , Humains , ARN long non codant/génétique , Tumeurs de l'estomac/génétique , Tumeurs de l'estomac/anatomopathologie , Tumeurs de l'estomac/métabolisme , Transduction du signal/génétique , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/métabolisme , Régulation de l'expression des gènes tumoraux
3.
Oral Health Prev Dent ; 22: 381-388, 2024 Aug 06.
Article de Anglais | MEDLINE | ID: mdl-39105315

RÉSUMÉ

PURPOSE: To analyse the relative expression and diagnostic potential of lncRNA XIST (XIST) in peri-implantitis, and explore the related mechanism of XIST in peri-implantitis. MATERIALS AND METHODS: XIST expression in saliva of patients with peri-implantitis was detected by qRT-PCR. The diagnostic significance of XIST in peri-implantitis was assessed by ROC curve. Clinical indicators of the included patients were collected and the correlation between XIST levels and peri-implant indicators was determined by Pearson correlation analysis. Bioinformatic prediction and luciferase reporter assay confirmed the targeting relationship of XIST with downstream factors. RESULTS: Salivary XIST levels were obviously higher in patients with peri-implantitis than in the healthy control group, and the AUC value for identifying patients was 0.8742 with a sensitivity and specificity of 83.5% and 81.4%. Patients in the peri-implantitis group had higher levels of plaque index (PLI), sulcus bleeding index (SBI) and probing depth (PD) than those in the healthy control group, and the expression of XIST was positively correlated with PLI, SBI, and PD levels. In addition, miR-150-5p was confirmed to be a potential downstream target of XIST. CONCLUSION: XIST was overexpressed in the saliva of patients with peri-implantitis and correlated with the severity of the disease. XIST has high diagnostic significance for detecting peri-implantitis.


Sujet(s)
Péri-implantite , ARN long non codant , Salive , Humains , Salive/composition chimique , Salive/métabolisme , Péri-implantite/diagnostic , Péri-implantite/métabolisme , Péri-implantite/génétique , Femelle , Mâle , Adulte d'âge moyen , Études cas-témoins , Indice parodontal , Adulte
4.
Urolithiasis ; 52(1): 113, 2024 Aug 06.
Article de Anglais | MEDLINE | ID: mdl-39105900

RÉSUMÉ

Long non-coding ribose nucleic acids (lncRNAs) have been implicated in the development of nephrolithiasis. The study aims to investigate the interplay of lncRNA SBF2-AS1 (SETbinding factor 2 antisense RNA 1) and NLR family pyrin domain containing 3 (NLRP3) in regulating the calcium oxalate monohydrate (COM)-induced human kidney HK-2 cell injury. HK-2 cells were treated with COM (100 µg/mL) to create a cellular model of kidney injury. Gene and protein expression was assessed by quantitative reverse-transcriptase polymerase chain reaction (qRT-PCR) and Western blot. Proliferation and apoptosis rates, as well as levels of malondialdehyde (MDA), lactate dehydrogenase (LDH), superoxide dismutase (SOD), tumor necrosis factor (TNF)-α, interleukin (IL)-1ß, and IL-6 were measured. Additionally, potential miRNAs interacting with SBF2-AS1 and NLRP3 were predicted utilizing the starBase and TargetScan databases. The interference of SBF2-AS1 resulted in increased cell proliferation and SOD levels in HK-2 cells after COM induction. SBF2-AS1 silencing also reduced COM-induced cell death and inflammatory cytokine production by down-regulating NLRP3 protein expression. Conversely, forced upregulation of NLRP3 abrogated the effect of SBF2-AS1 interference. Notably, SBF2-AS1 interference on COM-induced oxidative stress and COM-induced cellular damage was rescued by antioxidant, indicating the involvement of oxidative burden in COM-induced damage. miR-302e acted as a mediator miRNA linking the functional association of SBF2-AS1 and NLRP3. Silencing SBF2-AS1 promoted miR-302e level and miR-302e reduced NLRP3 expression in HK-2 cells to protect against COM-induced damage. In summary, these findings suggest that downregulation of lncRNA SBF2-AS1 can potentially protect HK-2 cells from COM-induced injury by modulating the miR-302e/NLRP3 pathway.


Sujet(s)
Oxalate de calcium , microARN , Protéine-3 de la famille des NLR contenant un domaine pyrine , ARN long non codant , Humains , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/génétique , microARN/métabolisme , microARN/génétique , ARN long non codant/génétique , ARN long non codant/métabolisme , Oxalate de calcium/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Lignée cellulaire , Prolifération cellulaire/génétique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques , Apoptose/génétique , Techniques de knock-down de gènes , Stress oxydatif/effets des médicaments et des substances chimiques
5.
Clinics (Sao Paulo) ; 79: 100400, 2024.
Article de Anglais | MEDLINE | ID: mdl-39089097

RÉSUMÉ

BACKGROUND: Aortic Dissection (AD) is a vascular disease with a high mortality rate and limited treatment strategies. The current research analyzed the function and regulatory mechanism of lncRNA HCG18 in AD. METHODS: HCG18, miR-103a-3p, and HMGA2 levels in the aortic tissue of AD patients were examined by RT-qPCR. After transfection with relevant plasmids, the proliferation of rat aortic Vascular Smoothing Muscle Cells (VSMCs) was detected by CCK-8 and colony formation assay, Bcl-2 and Bax was measured by Western blot, and apoptosis was checked by flow cytometry. Then, the targeting relationship between miR-103a-3p and HCG18 or HMGA2 was verified by bioinformation website analysis and dual luciferase reporter assay. Finally, the effect of HCG18 was verified in an AD rat model induced by ß-aminopropionitrile. RESULTS: HCG18 and HMGA2 were upregulated and miR-103a-3p was downregulated in the aortic tissues of AD patients. Downregulating HCG18 or upregulating miR-103a-3p enhanced the proliferation of VSMCs and limited cell apoptosis. HCG18 promoted HMGA2 expression by competing with miR-103a-3p and restoring HMGA2 could impair the effect of HCG18 downregulation or miR-103a-3p upregulation in mediating the proliferation and apoptosis of VSMCs. In addition, down-regulation of HCG18 could improve the pathological injury of the aorta in AD rats. CONCLUSION: HCG18 reduces proliferation and induces apoptosis of VSMCs through the miR-103a-3p/HMGA2 axis, thus aggravating AD.


Sujet(s)
, Apoptose , Prolifération cellulaire , microARN , ARN long non codant , microARN/génétique , microARN/métabolisme , Apoptose/génétique , Apoptose/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/génétique , Animaux , ARN long non codant/génétique , ARN long non codant/métabolisme , /génétique , /métabolisme , Humains , Protéine HMGA2/génétique , Protéine HMGA2/métabolisme , Mâle , Rats , Muscles lisses vasculaires/métabolisme , Régulation négative , Rat Sprague-Dawley , Régulation positive , Adulte d'âge moyen , Myocytes du muscle lisse/métabolisme , Modèles animaux de maladie humaine
6.
BMC Genomics ; 25(1): 756, 2024 Aug 02.
Article de Anglais | MEDLINE | ID: mdl-39095710

RÉSUMÉ

BACKGROUND: Long non-coding RNAs (lncRNAs) are RNA transcripts of more than 200 nucleotides that do not encode canonical proteins. Their biological structure is similar to messenger RNAs (mRNAs). To distinguish between lncRNA and mRNA transcripts quickly and accurately, we upgraded the PLEK alignment-free tool to its next version, PLEKv2, and constructed models tailored for both animals and plants. RESULTS: PLEKv2 can achieve 98.7% prediction accuracy for human datasets. Compared with classical tools and deep learning-based models, this is 8.1%, 3.7%, 16.6%, 1.4%, 4.9%, and 48.9% higher than CPC2, CNCI, Wen et al.'s CNN, LncADeep, PLEK, and NcResNet, respectively. The accuracy of PLEKv2 was > 90% for cross-species prediction. PLEKv2 is more effective and robust than CPC2, CNCI, LncADeep, PLEK, and NcResNet for primate datasets (including chimpanzees, macaques, and gorillas). Moreover, PLEKv2 is not only suitable for non-human primates that are closely related to humans, but can also predict the coding ability of RNA sequences in plants such as Arabidopsis. CONCLUSIONS: The experimental results illustrate that the model constructed by PLEKv2 can distinguish lncRNAs and mRNAs better than PLEK. The PLEKv2 software is freely available at https://sourceforge.net/projects/plek2/ .


Sujet(s)
ARN long non codant , ARN messager , ARN long non codant/génétique , ARN messager/génétique , Humains , Animaux , Logiciel , Biologie informatique/méthodes
7.
J Exp Clin Cancer Res ; 43(1): 217, 2024 Aug 05.
Article de Anglais | MEDLINE | ID: mdl-39098911

RÉSUMÉ

Aberrant alternative splicing events play a critical role in cancer biology, contributing to tumor invasion, metastasis, epithelial-mesenchymal transition, and drug resistance. Recent studies have shown that alternative splicing is a key feature for transcriptomic variations in colorectal cancer, which ranks third among malignant tumors worldwide in both incidence and mortality. Long non-coding RNAs can modulate this process by acting as trans-regulatory agents, recruiting splicing factors, or driving them to specific targeted genes. LncH19 is a lncRNA dis-regulated in several tumor types and, in colorectal cancer, it plays a critical role in tumor onset, progression, and metastasis. In this paper, we found, that in colorectal cancer cells, the long non-coding RNA H19 can bind immature RNAs and splicing factors as hnRNPM and RBFOX2. Through bioinformatic analysis, we identified 57 transcripts associated with lncH19 and containing binding sites for both splicing factors, hnRNPM, and RBFOX2. Among these transcripts, we identified the mRNA of the GTPase-RAC1, whose alternatively spliced isoform, RAC1B, has been ascribed several roles in the malignant transformation. We confirmed, in vitro, the binding of the splicing factors to both the transcripts RAC1 and lncH19. Loss and gain of expression experiments in two colorectal cancer cell lines (SW620 and HCT116) demonstrated that lncH19 is required for RAC1B expression and, through RAC1B, it induces c-Myc and Cyclin-D increase. In vivo, investigation from biopsies of colorectal cancer patients showed higher levels of all the explored genes (lncH19, RAC1B, c-Myc and Cyclin-D) concerning the healthy counterpart, thus supporting our in vitro model. In addition, we identified a positive correlation between lncH19 and RAC1B in colorectal cancer patients. Finally, we demonstrated that lncH19, as a shuttle, drives the splicing factors RBFOX2 and hnRNPM to RAC1 allowing exon retention and RAC1B expression. The data shown in this paper represent the first evidence of a new mechanism of action by which lncH19 carries out its functions as an oncogene by prompting colorectal cancer through the modulation of alternative splicing.


Sujet(s)
Épissage alternatif , Tumeurs colorectales , Régulation de l'expression des gènes tumoraux , Facteurs d'épissage des ARN , ARN long non codant , Protéine G rac1 , Humains , Tumeurs colorectales/génétique , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/métabolisme , ARN long non codant/génétique , Protéine G rac1/génétique , Protéine G rac1/métabolisme , Facteurs d'épissage des ARN/génétique , Facteurs d'épissage des ARN/métabolisme , Lignée cellulaire tumorale , Ribonucléoprotéine nucléaire hétérogène du groupe M/métabolisme , Ribonucléoprotéine nucléaire hétérogène du groupe M/génétique , Protéines de répression/génétique , Protéines de répression/métabolisme
8.
Wiley Interdiscip Rev RNA ; 15(4): e1864, 2024.
Article de Anglais | MEDLINE | ID: mdl-39087253

RÉSUMÉ

A considerable proportion of the eukaryotic genome undergoes transcription, leading to the generation of noncoding RNA molecules that lack protein-coding information and are not subjected to translation. These noncoding RNAs (ncRNAs) are well recognized to have essential roles in several biological processes. Long noncoding RNAs (lncRNAs) represent the most extensive category of ncRNAs found in the human genome. Much research has focused on investigating the roles of cis-acting lncRNAs in the regulation of specific target gene expression. In the majority of instances, the regulation of sense gene expression by its corresponding antisense pair occurs in a negative (discordant) manner, resulting in the suppression of the target genes. The notion that a negative correlation exists between sense and antisense pairings is, however, not universally valid. In fact, several recent studies have reported a positive relationship between corresponding cis antisense pairs within plants, budding yeast, and mammalian cancer cells. The positive (concordant) correlation between anti-sense and sense transcripts leads to an increase in the level of the sense transcript within the same genomic loci. In addition, mechanisms such as altering chromatin structure, the formation of R loops, and the recruitment of transcription factors can either enhance transcription or stabilize sense transcripts through their antisense pairs. The primary objective of this work is to provide a comprehensive understanding of both aspects of antisense regulation, specifically focusing on the positive correlation between sense and antisense transcripts in the context of eukaryotic gene expression, including its implications towards cancer progression. This article is categorized under: RNA Processing > 3' End Processing Regulatory RNAs/RNAi/Riboswitches > Regulatory RNAs.


Sujet(s)
Régulation de l'expression des gènes , Humains , Animaux , ARN antisens/génétique , ARN antisens/métabolisme , ARN long non codant/génétique , ARN long non codant/métabolisme
9.
Sci Rep ; 14(1): 17804, 2024 08 01.
Article de Anglais | MEDLINE | ID: mdl-39090162

RÉSUMÉ

Limited treatment options and poor prognosis present significant challenges in the treatment of lung squamous cell carcinoma (LUSC). Disulfidptosis impacts cancer progression and prognosis. We developed a prognostic signature using disulfidptosis-related long non-coding RNAs (lncRNAs) to predict the prognosis of LUSC patients. Gene expression matrices and clinical information for LUSC were downloaded from the TCGA database. Co-expression analysis identified 209 disulfidptosis-related lncRNAs. LASSO-Cox regression analysis identified nine key lncRNAs, forming the basis for establishing a prognostic model. The model's validity was confirmed by Kaplan-Meier and ROC curves. Cox regression analysis identified the risk score (RS) as an independent prognostic factor inversely correlated with overall survival. A nomogram based on the RS demonstrated good predictive performance for LUSC patient prognosis. The relationship between RS and immune function was explored using ESTIMATE, CIBERSORT, and ssGSEA algorithms. According to the TIDE database, a negative correlation was found between RS and immune therapy responsiveness. The GDSC database revealed that 49 drugs were beneficial for the low-risk group and 25 drugs for the high-risk group. Silencing C10orf55 expression in SW900 cells reduced invasiveness and migration potential. In summary, this lncRNA model based on TCGA-LUSC data effectively predicts prognosis and assists clinical decision-making.


Sujet(s)
Carcinome épidermoïde , Régulation de l'expression des gènes tumoraux , Tumeurs du poumon , ARN long non codant , Humains , ARN long non codant/génétique , Tumeurs du poumon/génétique , Tumeurs du poumon/mortalité , Tumeurs du poumon/anatomopathologie , Pronostic , Carcinome épidermoïde/génétique , Carcinome épidermoïde/anatomopathologie , Carcinome épidermoïde/thérapie , Marqueurs biologiques tumoraux/génétique , Mâle , Nomogrammes , Femelle , Estimation de Kaplan-Meier , Lignée cellulaire tumorale , Analyse de profil d'expression de gènes
10.
J Transl Med ; 22(1): 731, 2024 Aug 05.
Article de Anglais | MEDLINE | ID: mdl-39103911

RÉSUMÉ

Targeting non-coding RNAs (ncRNAs), including microRNAs (miRNAs) and long non-coding RNAs (lncRNAs), has recently emerged as a promising strategy for treating malignancies and other diseases. In recent years, the development of ncRNA-based therapeutics for targeting protein-coding and non-coding genes has also gained momentum. This review systematically examines ongoing and completed clinical trials to provide a comprehensive overview of the emerging landscape of ncRNA-based therapeutics. Significant efforts have been made to advance ncRNA therapeutics to early clinical studies. The most advanced trials have been conducted with small interfering RNAs (siRNAs), miRNA replacement using nanovector-entrapped miRNA mimics, or miRNA silencing by antisense oligonucleotides. While siRNA-based therapeutics have already received FDA approval, miRNA mimics, inhibitors, and lncRNA-based therapeutics are still under evaluation in preclinical and early clinical studies. We critically discuss the rationale and methodologies of ncRNA targeting strategies to illustrate this rapidly evolving field.


Sujet(s)
Essais cliniques comme sujet , Tumeurs , ARN non traduit , Humains , Tumeurs/génétique , Tumeurs/thérapie , ARN non traduit/génétique , ARN non traduit/usage thérapeutique , microARN/génétique , microARN/usage thérapeutique , microARN/métabolisme , ARN long non codant/génétique , ARN long non codant/métabolisme , Animaux , Petit ARN interférent/usage thérapeutique
11.
Front Immunol ; 15: 1419576, 2024.
Article de Anglais | MEDLINE | ID: mdl-39108268

RÉSUMÉ

Long non-coding RNAs (lncRNAs), defined as RNA molecules exceeding 200 nucleotides in length, have been implicated in the regulation of various biological processes and the progression of tumors. Among them, LINC00518, a recently identified lncRNA encoded by a gene located on chromosome 6p24.3, consists of three exons and is predicted to positively regulate the expression of specific genes. LINC00518 has emerged as a key oncogenic lncRNA in multiple cancer types. It exerts its tumor-promoting effects by modulating the expression of several target genes, primarily through acting as a sponge for microRNAs (miRNAs). Additionally, LINC00518 influences critical signaling pathways, including the Wnt/ß-catenin, JAK/STAT, and integrin ß3/FAK pathways. Elevated levels of LINC00518 in tumor tissues are associated with increased tumor size, advanced clinical stage, metastasis, and poor survival prognosis. This review provides a comprehensive summary of the genetic characteristics, expression patterns, biological functions, and underlying mechanisms of LINC00518 in human diseases.


Sujet(s)
Évolution de la maladie , Régulation de l'expression des gènes tumoraux , Tumeurs , ARN long non codant , Humains , ARN long non codant/génétique , Tumeurs/génétique , Tumeurs/anatomopathologie , Animaux , Marqueurs biologiques tumoraux/génétique , Transduction du signal , Pronostic , microARN/génétique
12.
Gen Physiol Biophys ; 43(5): 399-409, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39140680

RÉSUMÉ

Acute lung injury (ALI) is a significant health condition with notable rates of morbidity and mortality globally. Long non-coding ribose nucleic acids (lncRNAs) play vital roles in mitigating various inflammation-related diseases, including ALI. The study aimed to investigate the functional role and molecular mechanisms of lncRNA SNHG1 on ALI in lipopolysaccharide (LPS)-treated A549 cells and in LPS-induced ALI mice. The expression of SNHG1 was initially examined in LPS-treated A549 cells. We further demonstrated the critical function of SNHG1 through various cellular assessments following SNHG1 knockdown, including cell counting kit (CCK)-8 assay, flow cytometry analysis, as well as enzyme-linked immunosorbent assay (ELISA). Reducing SNHG1 levels hindered the negative effects of LPS on cell viability, apoptosis, and inflammation. Moreover, SNHG1 acted as a negative regulator for miR-199a-3p, which targeted downstream ROCK2. Depletion of miR-199a-3p or enhanced expression of ROCK2 abolished the protective effects of SNHG1 knockdown on LPS-induced apoptosis and inflammation. Consistently, silencing SNHG1 alleviated LPS-induced lung injury in mice, demonstrating its potential therapeutic benefits in managing ALI. Overall, this study sheds light on the role of SNHG1 in modulating inflammation and apoptosis in ALI through the miR-199a-3p/ROCK2 pathway, offering new insights for the treatment of this condition.


Sujet(s)
Lésion pulmonaire aigüe , Techniques de knock-down de gènes , Lipopolysaccharides , microARN , ARN long non codant , rho-Associated Kinases , ARN long non codant/génétique , ARN long non codant/métabolisme , Lésion pulmonaire aigüe/induit chimiquement , Lésion pulmonaire aigüe/métabolisme , Lésion pulmonaire aigüe/génétique , microARN/métabolisme , microARN/génétique , Animaux , Souris , Humains , rho-Associated Kinases/métabolisme , rho-Associated Kinases/génétique , Mâle , Cellules A549 , Transduction du signal , Souris de lignée C57BL , Apoptose/génétique
13.
Mol Biol Rep ; 51(1): 895, 2024 Aug 08.
Article de Anglais | MEDLINE | ID: mdl-39115693

RÉSUMÉ

BACKGROUND: Insufficient trophoblast invasion, culminating in suboptimal uterine spiral artery remodeling, is pinpointed as a pivotal contributor to preeclampsia (PE) development. LINC01410 has been documented to be increased in various neoplasms, and is significantly associated with the invasive capabilities of tumor cells. Nonetheless, its function and the mechanisms in the pathogenesis of PE require further investigation. METHODS AND RESULTS: LINC01410 and methyltransferase-like 3 (METTL3) were ectopically expressed in HTR-8/Svneo cells via lentiviral transduction. Subsequently, the cells' invasive capabilities and apoptosis rates were evaluated employing Transwell assays and flow cytometry, respectively. The interplay between LINC01410 and METTL3, alongside the m6A methylation of FAS, was probed through RNA immunoprecipitation (RIP). Additionally, the association between FAS and METTL3 was elucidated via Coimmunoprecipitation (Co-IP) assays. The protein level of NF-κB, BAX, and BCL-2 in LINC01410-overexpressing cells was detected by Western blot. Our findings revealed that LINC01410 elevation increased the invasive ability of HTR-8/Svneo cells, directly impacting METTL3 then leading to its reduced expression. Conversely, heightened METTL3 expression mitigated invasiveness while enhancing apoptosis in these cells. Moreover, METTL3's interaction with FAS led to increased FAS expression, subject to m6A methylation. A surge in LINC01410 markedly decreased both mRNA and protein levels of FAS. Furthermore, LINC01410 overexpression significantly reduced NF-κB and BAX protein levels while augmenting BCL-2. CONCLUSIONS: Upregulation of LINC01410 expression promotes trophoblast cell invasion by inhibiting FAS levels through modified m6A alteration and suppressing the NF-κB pathway. These findings underscore the pivotal role of LINC01410 in regulating trophoblast cell invasion and propose it as a promising therapeutic strategy for preventing or alleviating PE. This offers valuable insights for the clinical treatment of PE, for which definitive targeted therapy methods are currently lacking.


Sujet(s)
Apoptose , Methyltransferases , Pré-éclampsie , ARN long non codant , Trophoblastes , Antigènes CD95 , Humains , Trophoblastes/métabolisme , Methyltransferases/métabolisme , Methyltransferases/génétique , Antigènes CD95/métabolisme , Antigènes CD95/génétique , Femelle , Apoptose/génétique , Grossesse , ARN long non codant/génétique , ARN long non codant/métabolisme , Pré-éclampsie/génétique , Pré-éclampsie/métabolisme , Pré-éclampsie/anatomopathologie , Lignée cellulaire , Mouvement cellulaire/génétique , Facteur de transcription NF-kappa B/métabolisme , Transduction du signal/génétique
14.
Sci Rep ; 14(1): 18146, 2024 08 05.
Article de Anglais | MEDLINE | ID: mdl-39103417

RÉSUMÉ

Urate nephropathy, a common complication of hyperuricemia, has garnered increasing attention worldwide. However, the exact pathogenesis of this condition remains unclear. Currently, inflammation is widely accepted as the key factor in urate nephropathy. Therefore, the aim of this study was to elucidate the interaction of lincRNA-p21/AIF-1/CMPK2/NLRP3 via exosomes in urate nephropathy. This study evaluated the effect of lincRNA-p21/AIF-1/CMPK2/NLRP3 using clinical data collected from patients with urate nephropathy and human renal tubular epithelial cells (HK2) cultured with different concentrations of urate. In clinical research section, the level of lincRNA-p21/AIF-1 in exosomes of urine in patients with hyperuricemia or urate nephropathy was found to be increased, particularly in patients with urate nephropathy. In vitro study section, the level of exosomes, inflammation, autophagy, and apoptosis was increased in HK2 cells induced by urate. Additionally, the expression of lincRNA-p21, AIF-1, CMPK2, and NLRP3 was upregulated in exosomes and HK2 cells. Furthermore, manipulating the activity of lincRNA-p21, AIF-1, CMPK2, and NLRP3 through overexpression or interference vectors regulated the level of inflammation, autophagy, and apoptosis in HK2 cells. In conclusion, the pathway of lincRNA-p21/AIF-1/CMPK2/NLRP3 contributed to inflammation, autophagy, and apoptosis of human renal tubular epithelial cell induced by urate via exosomes. Additionally, the specific exosomes in urine might serve as novel biomarkers for urate nephropathy.


Sujet(s)
Apoptose , Autophagie , Cellules épithéliales , Exosomes , Protéine-3 de la famille des NLR contenant un domaine pyrine , ARN long non codant , Acide urique , Humains , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Acide urique/métabolisme , Exosomes/métabolisme , Cellules épithéliales/métabolisme , Cellules épithéliales/anatomopathologie , ARN long non codant/métabolisme , ARN long non codant/génétique , Transduction du signal , Inflammation/métabolisme , Inflammation/anatomopathologie , Tubules rénaux/métabolisme , Tubules rénaux/anatomopathologie , Lignée cellulaire , Mâle , Facteur inducteur d'apoptose/métabolisme , Femelle , Adulte d'âge moyen , Hyperuricémie/métabolisme , Hyperuricémie/urine , Protéines de liaison au calcium , Protéines des microfilaments
15.
Int J Mol Sci ; 25(15)2024 Jul 24.
Article de Anglais | MEDLINE | ID: mdl-39125649

RÉSUMÉ

lncRNAs are noncoding transcripts with tissue and cancer specificity. Particularly, in breast cancer, lncRNAs exhibit subtype-specific expression; they are particularly upregulated in luminal tumors. However, no gene signature-based laboratory tests have been developed for luminal breast cancer identification or the differential diagnosis of luminal tumors, since no luminal A- or B-specific genes have been identified. Particularly, luminal B patients are of clinical interest, since they have the most variable response to neoadjuvant treatment; thus, it is necessary to develop diagnostic and predictive biomarkers for these patients to optimize treatment decision-making and improve treatment quality. In this study, we analyzed the lncRNA expression profiles of breast cancer cell lines and patient tumor samples from RNA-Seq data to identify an lncRNA signature specific for luminal phenotypes. We identified an lncRNA signature consisting of LINC01016, GATA3-AS1, MAPT-IT1, and DSCAM-AS1 that exhibits luminal subtype-specific expression; among these lncRNAs, GATA3-AS1 is associated with the presence of residual disease (Wilcoxon test, p < 0.05), which is related to neoadjuvant chemotherapy resistance in luminal B breast cancer patients. Furthermore, analysis of GATA3-AS1 expression using RNA in situ hybridization (RNA ISH) demonstrated that this lncRNA is detectable in histological slides. Similar to estrogen receptors and Ki67, both commonly detected biomarkers, GATA3-AS1 proves to be a suitable predictive biomarker for clinical application in breast cancer laboratory tests.


Sujet(s)
Marqueurs biologiques tumoraux , Tumeurs du sein , Résistance aux médicaments antinéoplasiques , Régulation de l'expression des gènes tumoraux , Traitement néoadjuvant , ARN long non codant , Humains , ARN long non codant/génétique , Tumeurs du sein/génétique , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/anatomopathologie , Tumeurs du sein/métabolisme , Femelle , Résistance aux médicaments antinéoplasiques/génétique , Marqueurs biologiques tumoraux/génétique , Lignée cellulaire tumorale , Analyse de profil d'expression de gènes , Facteur de transcription GATA-3/génétique , Facteur de transcription GATA-3/métabolisme , Transcriptome
16.
BMC Cancer ; 24(1): 961, 2024 Aug 06.
Article de Anglais | MEDLINE | ID: mdl-39107726

RÉSUMÉ

BACKGROUND: Pancreatic cancer (PaCa) is one of the most intractable and fatal malignancies and is associated with the dysregulation of long noncoding RNAs (lncRNAs), which are a large class of noncoding RNAs larger than 200 nt that act as competing endogenous RNAs or sponges for miRNAs to induce tumour biological behaviours. However, their clinical value in treating pancreatic cancer has been poorly explained, but they are essential for improving the prognosis of PaCa patients. METHODS: We analysed the plasma-derived exosomal lncRNA profiles of PaCa patients by using whole-transcriptome sequencing analysis and identified significantly differentially expressed lncRNAs, including LINC01268, LINC02802, AC124854.1, and AL132657.1. In the current study, the expression levels of four plasma-derived exosomal lncRNAs in PaCa plasma were validated via quantitative real-time polymerase chain reaction (qRT‒PCR). The relationship between the expression of the four lncRNAs and the clinicopathological features of patients with PaCa was also evaluated. RESULTS: We demonstrated that exosomal LINC01268, LINC02802, AC124854.1 and AL132657.1 were highly expressed in PaCa plasma compared with those in normal controls; moreover, they were positively correlated with the serum expression of carbohydrate antigen 19-9 (CA19-9). The receiver operating characteristic curves (AUCs) of the four lncRNAs were 0.8421, 0.6544, 0.7190, and 0.6321, and the AUC value of the combination of the four exosomal lncRNAs increased to 0.8476, with a sensitivity of 0.72 and specificity of 0.89. These results suggested that the plasma-derived exosomal genes LINC01268, LINC02802, AC124854.1, and AL132657.1 may be novel diagnostic markers for PaCa. CONCLUSIONS: Our research demonstrated that the plasma-derived exosomal lncRNAs of PaCa patients are novel blood-based biomarkers of disease.


Sujet(s)
Marqueurs biologiques tumoraux , Exosomes , Tumeurs du pancréas , ARN long non codant , Humains , ARN long non codant/sang , ARN long non codant/génétique , Tumeurs du pancréas/génétique , Tumeurs du pancréas/sang , Tumeurs du pancréas/anatomopathologie , Exosomes/génétique , Exosomes/métabolisme , Marqueurs biologiques tumoraux/sang , Marqueurs biologiques tumoraux/génétique , Mâle , Femelle , Adulte d'âge moyen , Pronostic , Régulation de l'expression des gènes tumoraux , Sujet âgé , Analyse de profil d'expression de gènes/méthodes , Courbe ROC , Antigène CA 19-9/sang
17.
Mol Biol Rep ; 51(1): 909, 2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-39145884

RÉSUMÉ

BACKGROUND: Inflammatory cytokines such as Interleukin 1ß(IL1ß), IL6,Tumor Necrosis Factor-α (TNF-α) can inhibit osteoblast differentiation and induce osteoblast apoptosis. PANoptosis, a newly identified type of programmed cell death (PCD), may be influenced by long noncoding RNA (lncRNAs) which play important roles in regulating inflammation. However, the potential role of lncRNAs in inflammation and PANoptosis during osteogenic differentiation remains unclear. This study aimed to investigate the regulatory functions of lncRNAs in inflammation and apoptosis during osteogenic differentiation. METHODS AND RESULTS: High-throughput sequencing was used to identify differentially expressed genes involved in osteoblast differentiation under inflammatory conditions. Two lncRNAs associated with inflammation and PANoptosis during osteogenic differentiation were identified from sequencing data and Gene Expression Omnibus (GEO) databases. Their functionalities were analyzed using diverse bioinformatics methodologies, resulting in the construction of the lncRNA-miRNA-mRNA network. Among these, lncRNA (MIR17HG) showed a high correlation with PANoptosis. Bibliometric methods were employed to collect literature data on PANoptosis, and its components were inferred. PCR and Western Blotting experiments confirmed that lncRNA MIR17HG is related to PANoptosis in osteoblasts during inflammation. CONCLUSIONS: Our data suggest that TNF-α-induced inhibition of osteogenic differentiation and PANoptosis in MC3T3-E1 osteoblasts is associated with MIR17HG. These findings highlight the critical role of MIR17HG in the interplay between inflammation, PANoptosis, and osteogenic differentiation, suggesting potential therapeutic targets for conditions involving impaired bone formation and inflammatory responses.


Sujet(s)
Différenciation cellulaire , Réseaux de régulation génique , microARN , Ostéoblastes , Ostéogenèse , ARN long non codant , Facteur de nécrose tumorale alpha , ARN long non codant/génétique , ARN long non codant/métabolisme , Ostéogenèse/génétique , Différenciation cellulaire/génétique , Facteur de nécrose tumorale alpha/génétique , Facteur de nécrose tumorale alpha/métabolisme , Ostéoblastes/métabolisme , Ostéoblastes/effets des médicaments et des substances chimiques , microARN/génétique , microARN/métabolisme , Souris , Animaux , Apoptose/génétique , Inflammation/génétique , Humains , Biologie informatique/méthodes , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , ARN messager/génétique , ARN messager/métabolisme , Analyse de profil d'expression de gènes/méthodes ,
18.
Sci Rep ; 14(1): 18928, 2024 08 15.
Article de Anglais | MEDLINE | ID: mdl-39147766

RÉSUMÉ

This study aimed to develop a prognostic risk model based on immune-related long non-coding RNAs (lncRNAs). By analyzing the expression profiles of specific long non-coding RNAs, the objective was to construct a predictive model to accurately assess the survival prognosis of breast cancer (BC) patients. This effort seeks to provide personalized treatment strategies for patients and improve clinical outcomes. Based on the median risk value, 300 samples of triple-negative BC (TNBC) patients were rolled into a high-risk group (HR group, n = 140) and a low-risk group (LR group, n = 160). Multivariate Cox (MVC) analysis was performed by combining the patient risk score and clinical information to evaluate the prognostic value of the prognostic risk (PR) model. A total of 371 immune-related lncRNAs associated with the prognosis of TNBC were obtained from 300 TNBC samples. Nine associated with prognosis were obtained by univariate Cox (UVC) analysis, and 3 (AC090181.2, LINC01235, and LINC01943) were selected by MVC analysis for the construction of TNBC PR model. Survival analysis showed a great difference in TNBC patients in different groups (P < 0.001). The receiver operator characteristic (ROC) curve showed the model possessed a good area under ROC curve (AUC), which was 0.928. The patient RS jointing with clinical information as well as the MVC analysis revealed that RS was an independent risk factor (IRF) for prognosis of TNBC (P < 0.05, HR = 1.033286). Therefore, the lncRNAs associated with TNBC immunity can be screened by bioinformatics analysis, and the established PR model of TNBC could better predict the prognosis of patients with TNBC, exhibiting a high application value in clinic.


Sujet(s)
ARN long non codant , Humains , ARN long non codant/génétique , Femelle , Pronostic , Adulte d'âge moyen , Marqueurs biologiques tumoraux/génétique , Tumeurs du sein triple-négatives/génétique , Tumeurs du sein triple-négatives/mortalité , Tumeurs du sein triple-négatives/immunologie , Régulation de l'expression des gènes tumoraux , Tumeurs du sein/génétique , Tumeurs du sein/mortalité , Tumeurs du sein/immunologie , Appréciation des risques/méthodes , Courbe ROC , Analyse de profil d'expression de gènes , Analyse de survie , Facteurs de risque
19.
Sci Rep ; 14(1): 18500, 2024 08 09.
Article de Anglais | MEDLINE | ID: mdl-39122807

RÉSUMÉ

Programmed cell death (PCD) is a process that eliminates infected, damaged, or possibly neoplastic cells to sustain homeostatic multicellular organisms. Although long noncoding RNAs (lncRNAs) are involved in various types of PCD and regulate tumor growth, invasion, and migration, the role of PCD-related lncRNAs in bladder cancer still lacks systematic exploration. In this research, we integrated multiple types of PCD as pan-PCD and identified eight pan-PCD-related lncRNAs (LINC00174, HCP5, HCG27, UCA1, SNHG15, GHRLOS, CYB561D2, and AGAP11). Then, we generated a pan-PCD-related lncRNA prognostic signature (PPlncPS) with excellent predictive power and reliability, which performed equally well in the E-MTAB-4321 cohort. In comparison with the low-PPlncPS score group, the high-PPlncPS score group had remarkably higher levels of angiogenesis, matrix, cancer-associated fibroblasts, myeloid cell traffic, and protumor cytokine signatures. In addition, the low-PPlncPS score group was positively correlated with relatively abundant immune cell infiltration, upregulated expression levels of immune checkpoints, and high tumor mutation burden (TMB). Immunogenomic profiles revealed that patients with both low PPlncPS scores and high TMB had the best prognosis and may benefit from immune checkpoint inhibitors. Furthermore, for patients with high PPlncPS scores, docetaxel, staurosporine, and luminespib were screened as potential therapeutic candidates. In conclusion, we generated a pan-PCD-related lncRNA signature, providing precise and individualized prediction for clinical prognosis and some new insights into chemotherapy and immune checkpoint inhibitor therapy for bladder cancer.


Sujet(s)
Régulation de l'expression des gènes tumoraux , ARN long non codant , Tumeurs de la vessie urinaire , Tumeurs de la vessie urinaire/génétique , Tumeurs de la vessie urinaire/immunologie , Tumeurs de la vessie urinaire/traitement médicamenteux , Tumeurs de la vessie urinaire/anatomopathologie , Humains , ARN long non codant/génétique , Pronostic , Marqueurs biologiques tumoraux/génétique , Apoptose/génétique , Analyse de profil d'expression de gènes , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique
20.
Cell Transplant ; 33: 9636897241266725, 2024.
Article de Anglais | MEDLINE | ID: mdl-39126321

RÉSUMÉ

Many studies support the idea that long noncoding RNAs (lncRNAs) are significantly involved in the process of cardiomyocyte (CM) regeneration following a myocardial infarction (MI). This study aimed to systematically review the emerging role of lncRNAs in cardiac regeneration by promoting CM proliferation after MI. Furthermore, the review summarized potential targets and the underlying mechanisms of lncRNAs to induce heart regeneration, suggesting utilizing lncRNAs as innovative therapeutic targets for mitigating MI injuries. We searched the PubMed, Scopus, and Web of Science databases for studies on lncRNAs that play a role in heart regeneration after MI. We used search terms that included MI, lncRNAs, CM, and proliferation. Relevant English articles published until June 11, 2023, were systematically reviewed based on inclusion and exclusion criteria. A total of 361 publications were initially identified, and after applying the inclusion and exclusion criteria, nine articles were included in this systematic review. These studies investigated the role of critical lncRNAs in cardiac regeneration after MI, including five upregulated and four downregulated lncRNAs. Acting as a competitive endogenous RNA is one of the main roles of lncRNAs in regulating genes involved in CM proliferation through binding to target microRNAs. The main molecular processes that greatly increase CM proliferation are those that turn on the Hippo/YAP1, PI3K/Akt, JAK2-STAT3, and E2F1-ECRAR-ERK1/2 signaling pathways. This systematic review highlights the significant role of lncRNAs in heart regeneration after MI and their impact on CM proliferation. The findings suggest that lncRNAs could serve as potential targets for therapeutic interventions aiming to enhance cardiac function.


Sujet(s)
Prolifération cellulaire , Infarctus du myocarde , Myocytes cardiaques , ARN long non codant , Régénération , ARN long non codant/génétique , ARN long non codant/métabolisme , Myocytes cardiaques/métabolisme , Prolifération cellulaire/génétique , Infarctus du myocarde/thérapie , Infarctus du myocarde/génétique , Infarctus du myocarde/métabolisme , Infarctus du myocarde/physiopathologie , Humains , Animaux
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE