1,514 results on '"neat1"'
Search Results
2. Eugenol inhibits NEAT1 as a ceRNA in pre-cancerous breast lesions
- Author
-
Liu, Yusheng, Zhang, Guijuan, Ma, Yi, Ma, Min, and Jiang, Xuefeng
- Published
- 2025
- Full Text
- View/download PDF
3. Upregulation of FAM129B protects against glucocorticoid-induced skeletal muscle atrophy via regulating long non-coding RNA NEAT1
- Author
-
Wang, Yan, Lu, Yushen, Hou, Jinhui, Wang, Yuyang, Luo, Lihuan, Lu, Zuneng, Xie, Yuanlong, Cai, Lin, and Xiao, Zheman
- Published
- 2025
- Full Text
- View/download PDF
4. Potential of long non-coding RNAs as a therapeutic target and molecular markers in glioblastoma pathogenesis
- Author
-
Chaudhary, Rishabh
- Published
- 2021
- Full Text
- View/download PDF
5. In silico analysis of altered expression of long non-coding RNA in SARS-CoV-2 infected cells and their possible regulation by STAT1, STAT3 and interferon regulatory factors
- Author
-
Laha, Sayantan, Saha, Chinmay, Dutta, Susmita, Basu, Madhurima, Chatterjee, Raghunath, Ghosh, Sujoy, and Bhattacharyya, Nitai P.
- Published
- 2021
- Full Text
- View/download PDF
6. Long non-coding RNA NEAT1 promotes ovarian granulosa cell proliferation and cell cycle progression via the miR-29a-3p/IGF1 axis.
- Author
-
He, Lina, Lin, Jie, Qin, Zhengwen, Xu, Qing, Hao, Li, Fu, Yanhong, Ran, Xu, and Chen, Wei
- Subjects
- *
GRANULOSA cells , *CYTOLOGY , *LINCRNA , *CELL physiology , *CELL cycle , *OVARIAN follicle - Abstract
Background: Granulosa cell proliferation and survival are essential for normal ovarian function and follicular development. Long non-coding RNAs (lncRNAs) have emerged as important regulators of cell proliferation and differentiation. Nuclear paraspeckle assembly transcript 1 (NEAT1) has been implicated in various cellular processes, but its role in granulosa cell function remains unclear. Methods: We investigated the function of lncRNA NEAT1 in human ovarian granulosa-like tumor cells (KGN). The effects of NEAT1 overexpression or silencing on cell proliferation and cell cycle were evaluated using CCK-8 assays and flow cytometry. The interaction between NEAT1, miR-29a-3p, and IGF1 was examined using dual-luciferase reporter assays, qRT-PCR, and Western blot analysis. Results: NEAT1 promoted granulosa cell proliferation and cell cycle progression by indirectly upregulated IGF1 expression through acting as a molecular sponge for miR-29a-3p. Cell proliferation and G2/M phase proportions were increased by overexpression of NEAT1, whereas cell proliferation and G2/M phase proportions decreased with NEAT1 silencing. The effects of NEAT1 on cell proliferation and cell cycle-related proteins (CCNB1 and CDK2) were partially reversed by miR-29a-3p mimic, while miR-29a-3p inhibitor rescued the effects of NEAT1 silencing. Conclusion: LncRNA NEAT1 could promote ovarian granulosa cell proliferation and cell cycle progression via the miR-29a-3p/IGF1 axis in polycystic ovary syndrome. Further investigation of this mechanism in clinical samples may have implications for understanding ovarian physiology and pathology. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
7. Role of NEAT1 and HOTAIR long non-coding RNAs in Behcet's Disease pathogenesis and their correlation with target inflammatory cytokines.
- Author
-
Javidi-Aghdam, Kamran, Faghfouri, Amirhossein, Jafarpour, Mehdi, Akbarzadeh-Khiavi, Mostafa, Safary, Azam, Pourbagherian, Omid, and Khabbazi, Alireza
- Abstract
Background: Recent studies have highlighted the potential role of several long non-coding RNAs (lncRNAs) in the pathogenesis of Behçet's disease (BD). This study investigated the expression profiles of lncRNA NEAT1 and lncRNA HOTAIR, and their target cytokine genes, IL-6 and TNF-α, in active and inactive BD patients. Methods: This cross-sectional study was conducted on peripheral blood mononuclear cells (PBMCs) obtained from 25 BD patients and 25 age-sex-matched healthy controls (HCs). BD activity was evaluated using the BDCAF (Behcet's Disease Current Activity Form) score. Correlation analysis assessed the relationship between BD activity and the expression levels of lncRNAs and their target cytokine genes. The diagnostic potential of the genes was evaluated using Receiver Operating Characteristic (ROC) curve analysis. Results: The expression levels of NEAT1, HOTAIR, IL-6, and TNF-α were significantly higher in the BD group compared to the HCs. However, there was no significant correlation between the expression levels of these genes and BD activity or the involvement of various organs. A positive correlation was observed between HOTAIR gene expression and IL-6 (R = 0.594, P-value = 0.009) in the BD group. In contrast, no significant correlation was found between HOTAIR and TNF-α or between NEAT1 and TNF-α or IL-6. The ROC curve analysis indicated strong diagnostic potential for the lncRNAs, with area under the curve (AUC) values of 0.90 for NEAT1 and 0.86 for HOTAIR. Conclusion: The elevated expression levels of NEAT1 and HOTAIR in BD patients suggest their potential involvement in the disease's pathogenesis, indicating promising targets for future diagnostic and therapeutic strategies in BD. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
8. NEAT1 Promotes Valproic Acid-Induced Autism Spectrum Disorder by Recruiting YY1 to Regulate UBE3A Transcription.
- Author
-
He, Chuping, Zhou, Huimei, Chen, Lei, and Liu, Zeying
- Abstract
Evidence suggests that long non-coding RNAs (lncRNAs) play a significant role in autism. Herein, we explored the functional role and possible molecular mechanisms of NEAT1 in valproic acid (VPA)-induced autism spectrum disorder (ASD). A VPA-induced ASD rat model was constructed, and a series of behavioral tests were performed to examine motor coordination and learning-memory abilities. qRT-PCR and western blot assays were used to evaluate target gene expression levels. Loss-and-gain-of-function assays were conducted to explore the functional role of NEAT1 in ASD development. Furthermore, a combination of mechanistic experiments and bioinformatic tools was used to assess the relationship and regulatory role of the NEAT1-YY1-UBE3A axis in ASD cellular processes. Results showed that VPA exposure induced autism-like developmental delays and behavioral abnormalities in the VPA-induced ASD rat model. We found that NEAT1 was elevated in rat hippocampal tissues after VPA exposure. NEAT1 promoted VPA-induced autism-like behaviors and mitigated apoptosis, oxidative stress, and inflammation in VPA-induced ASD rats. Notably, NEAT1 knockdown improved autism-related behaviors and ameliorated hippocampal neuronal damage. Mechanistically, it was observed that NEAT1 recruited the transcription factor YY1 to regulate UBE3A expression. Additionally, in vitro experiments further confirmed that NEAT1 knockdown mitigated hippocampal neuronal damage, oxidative stress, and inflammation through the YY1/UBE3A axis. In conclusion, our study demonstrates that NEAT1 is highly expressed in ASD, and its inhibition prominently suppresses hippocampal neuronal injury and oxidative stress through the YY1/UBE3A axis, thereby alleviating ASD development. This provides a new direction for ASD-targeted therapy. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
9. FUS and METTL3 collaborate to regulate RNA maturation, preventing unfolded protein response and promoting gastric cancer progression.
- Author
-
Liu, Dongtao, Ding, Bo, Liu, Gang, and Yang, Zhijuan
- Subjects
- *
ALTERNATIVE RNA splicing , *UNFOLDED protein response , *LIFE sciences , *MEDICAL sciences , *RNA methylation - Abstract
FUS-mediated alternative splicing and METTL3-regulated RNA methylation play crucial roles in RNA processing. The purpose of this study was to investigate the interactive roles of FUS and METTL3 in gastric cancer (GC) progression. RNA sequencing data were obtained from the TCGA-STAD dataset. Differentially expressed genes (DEGs) were analyzed across groups stratified by the medians of FUS, METTL3, and NEAT1, respectively. Endoplasmic reticulum (ER) stress markers PERK, IRE1, pIRE1, Bip, and CHOP, as well as related apoptosis stress markers PARP, cleaved-PARP, (Cleaved) Caspase 7, and (Cleaved) Caspase 3, were assessed through western blotting. Alternative splicing and N6-methyladenosine (m(6)A) methylation of specific genes were detected with MeRIP-PCR. Finally, in vivo experiments were conducted using nude mice bearing sh-FUS-transfected HGC27 xenograft tumors. FUS and METTL3 expression levels were elevated in GC tissues. A significant overlap of DEGs was observed between the FUS- and METTL3-stratified groups. These overlapping DEGs were predominantly enriched in mRNA processing and protein processing in the ER. ER stress and apoptosis were induced by sh-FUS or sh-METTL3, which was further enhanced by ER stress inducer tunicamycin in both MKN45 and HGC27 cells. Similarly, DEGs for NEAT1 high- and low-expressed groups were enriched in protein processing in the ER and spliceosome. To a lesser extent, ER stress was also induced by sh-NEAT1 and enhanced by tunicamycin in HGC27 cells. Furthermore, sh-FUS or sh-METTL3 influenced alternative splicing and methylation of specific mRNAs, including FUS, NEAT1, PCNA, MCM2, and BIRC5. Tumor progression was inhibited by sh-FUS in mice, and ER stress and apoptosis were induced, which were further enhanced by tunicamycin. FUS and METTL3 collaborate to facilitate RNA maturation. Inhibiting FUS or METTL3 promoted ER stress and apoptosis and inhibited progression in GC. Aberrant levels of FUS and METTL3 can evoke endoplasmic reticulum (ER) stress and apoptosis by generating splicing and methylation variants of mRNAs in gastric cancer, supporting the therapeutic potential of inducing ER stress [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
10. MALAT1 and NEAT1 are Neuroprotective During Hypoxic Preconditioning in the Mouse Hippocampus Possibly by Regulation of NR2B.
- Author
-
Wang, Liping, Fu, Gang, Han, Ruijuan, Fan, Peijia, Yang, Jing, Gong, Kerui, Zhao, Zhijun, Zhang, Chunyang, Sun, Kai, and Shao, Guo
- Subjects
- *
NEURONS , *GENE expression , *RNA , *CEREBRAL anoxia , *POLYMERASE chain reaction - Abstract
Wang, Liping, Gang Fu, Ruijuan Han, Peijia Fan, Jing Yang, Kerui Gong, Zhijun Zhao, Chunyang Zhang, Kai Sun, and Guo Shao. MALAT1 and NEAT1 are neuroprotective during hypoxic preconditioning in the mouse hippocampus possibly by regulation of NR2B. High Alt Med Biol. 25:285–294, 2024. Background: The regulation of noncoding ribonucleic acid (ncRNA) has been shown to be involved in cellular and molecular responses to hypoxic preconditioning (HPC), a situation created by the induction of sublethal hypoxia in the brain. The ncRNAs metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) and nuclear paraspeckle assembly transcript 1 (NEAT1) are abundantly expressed in the brain, where they regulate the expression of various genes in nerve cells. However, the exact roles of MALAT1 and NEAT1 in HPC are not fully understood. Methods: A mouse model of acute repeated hypoxia was used as a model of HPC, and MALAT1 and NEAT1 levels in the hippocampus were measured using real-time polymerase chain reaction (PCR). The mRNA and protein levels of N-methyl-d-aspartate receptor subunit 2 B (NR2B) in the mouse hippocampus were measured using real-time PCR and western blotting, respectively. HT22 cells knocked-down for MALAT1 and NEAT1 were used for in vitro testing. Expression of NR2B, which is involved in nerve cell injury under ischemic and hypoxic conditions, was also evaluated. The levels of spectrin and cleaved caspase-3 in MALAT1 and NEAT1 knockdown HT22 cells under oxygen glucose deprivation/reperfusion (OGD/R) were determined by western blotting. Results: HPC increased the expression of MALAT1 and NEAT1 and decreased the expression of NR2B mRNA in the mouse hippocampus (p < 0.05). Knockdown of MALAT1 and NEAT1 increased both NR2B mRNA and protein levels nearly twofold and caused damage under OGD/R conditions in HT22 cells (p < 0.05). Conclusion: MALAT1 and NEAT1 exert neuroprotective effects by influencing the expression of NR2B. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
11. LIN28A-dependent lncRNA NEAT1 aggravates sepsis-induced acute respiratory distress syndrome through destabilizing ACE2 mRNA by RNA methylation
- Author
-
Jun Liu, Xiang Li, Peng Yang, Yufeng He, Weilong Hong, Yawei Feng, and Zhiqiang Ye
- Subjects
Acute respiratory distress syndrome ,Sepsis ,NEAT1 ,ACE2 ,RNA methylation ,RNA stability ,Medicine - Abstract
Abstract Background Acute respiratory distress syndrome (ARDS) is a life-threatening and heterogeneous disorder leading to lung injury. To date, effective therapies for ARDS remain limited. Sepsis is a frequent inducer of ARDS. However, the precise mechanisms underlying sepsis-induced ARDS remain unclear. Methods Here RNA methylation was detected by methylated RNA immunoprecipitation (MeRIP), RNA stability was determined by RNA decay assay while RNA antisense purification (RAP) was used to identify RNA-protein interaction. Besides, co-immunoprecipitation (Co-IP) was utilized to detect protein-protein interaction. Moreover, mice were injected with lipopolysaccharide (LPS) to establish sepsis-induced ARDS model in vivo. Results This study revealed that long non-coding RNA (lncRNA) nuclear-enriched abundant transcript 1 (NEAT1) aggravated lung injury through suppressing angiotensin-converting enzyme 2 (ACE2) in sepsis-induced ARDS models in vitro and in vivo. Mechanistically, NEAT1 declined ACE2 mRNA stability through heterogeneous nuclear ribonucleoprotein A2/B1 (hnRNPA2B1) in lipopolysaccharide (LPS)-treated alveolar type II epithelial cells (AT-II cells). Besides, NEAT1 destabilized ACE2 mRNA depending on RNA methylation by forming methylated NEAT1/hnRNPA2B1/ACE2 mRNA complex in LPS-treated AT-II cells. Moreover, lin-28 homolog A (LIN28A) improved NEAT1 stability whereas insulin-like growth factor 2 mRNA binding protein 3 (IGF2BP3) augmented NEAT1 destabilization by associating with LIN28A to disrupt the combination of LIN28A and NEAT1 in LPS-treated AT-II cells. Nevertheless, hnRNPA2B1 increased NEAT1 stability by blocking the interaction between LIN28A and IGF2BP3 in LPS-treated AT-II cells. Conclusions These findings uncover mechanisms of sepsis-triggering ARDS and provide promising therapeutic targets for sepsis-induced ARDS.
- Published
- 2025
- Full Text
- View/download PDF
12. Deep Conservation and Unexpected Evolutionary History of Neighboring lncRNAs MALAT1 and NEAT1.
- Author
-
Weghorst, Forrest, Torres Marcén, Martí, Faridi, Garrison, Lee, Yuh, and Cramer, Karina
- Subjects
Birds ,Comparative genomics ,Long non-coding RNA ,MALAT1 ,NEAT1 ,Animals ,RNA ,Long Noncoding ,Phylogeny ,Gene Expression Regulation ,Biological Evolution ,Mammals - Abstract
Long non-coding RNAs (lncRNAs) have begun to receive overdue attention for their regulatory roles in gene expression and other cellular processes. Although most lncRNAs are lowly expressed and tissue-specific, notable exceptions include MALAT1 and its genomic neighbor NEAT1, two highly and ubiquitously expressed oncogenes with roles in transcriptional regulation and RNA splicing. Previous studies have suggested that NEAT1 is found only in mammals, while MALAT1 is present in all gnathostomes (jawed vertebrates) except birds. Here we show that these assertions are incomplete, likely due to the challenges associated with properly identifying these two lncRNAs. Using phylogenetic analysis and structure-aware annotation of publicly available genomic and RNA-seq coverage data, we show that NEAT1 is a common feature of tetrapod genomes except birds and squamates. Conversely, we identify MALAT1 in representative species of all major gnathostome clades, including birds. Our in-depth examination of MALAT1, NEAT1, and their genomic context in a wide range of vertebrate species allows us to reconstruct the series of events that led to the formation of the locus containing these genes in taxa from cartilaginous fish to mammals. This evolutionary history includes the independent loss of NEAT1 in birds and squamates, since NEAT1 is found in the closest living relatives of both clades (crocodilians and tuataras, respectively). These data clarify the origins and relationships of MALAT1 and NEAT1 and highlight an opportunity to study the change and continuity in lncRNA structure and function over deep evolutionary time.
- Published
- 2024
13. LncRNAs SOX2-OT and NEAT1 act as a potential biomarker for esophageal squamous cell carcinoma
- Author
-
Rajiv Ranjan Kumar, Adrija Mohanta, Manjit Kaur Rana, Vivek Uttam, Hardeep Singh Tuli, and Aklank Jain
- Subjects
SOX2-OT ,ESCC ,NEAT1 ,AUC ,ROC ,Clinical settings ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Despite strides in diagnostic and therapeutic approaches for ESCC, patient survival rates remain relatively low. Recent studies highlight the pivotal role of long non-coding RNAs (lncRNAs) in regulating diverse cellular activities in humans. Dysregulated lncRNAs have emerged as potential diagnostic indicators across various cancers, including ESCC. However, further research is necessary to effectively leverage ESCC-associated lncRNAs in clinical settings. Understanding their clinical significance for ESCC diagnosis and their mechanisms can pave the way for more effective therapeutic strategies. Our qRT-PCR analysis revealed significant downregulation of SOX2-OT (~ 2.02-fold) and NEAT1 (~ 1.53-fold) in ESCC blood samples. These lncRNAs show potential as biomarkers for distinguishing ESCC patients from healthy individuals, with ROC curves and AUC values of 0.736 for SOX2-OT and 0.621 for NEAT1. Further analysis examined the correlation between SOX2-OT and NEAT1 expression and various clinicopathological factors, including age, gender, smoking, alcohol use, hot beverage intake, tumor grade, and TNM stages. In-silico studies highlighted their roles in miRNA sponging via mTOR and MAPK pathways, while co-expression network analysis identified associated genes. This research paves the way for future studies on ESCC prognosis using SOX2-OT and NEAT1 as predictive markers. By thoroughly investigating the functions of these lncRNAs, we aim to deepen our understanding of their potential as diagnostic markers and their role in facilitating effective therapeutic interventions for esophageal squamous cell carcinoma (ESCC) within clinical contexts.
- Published
- 2024
- Full Text
- View/download PDF
14. Identifying the ceRNA Regulatory Network in Early-Stage Acute Pancreatitis and Investigating the Therapeutic Potential of NEAT1 in Mouse Models
- Author
-
Lin B and Huang C
- Subjects
acute pancreatitis ,lncrna ,cerna ,neat1 ,pyroptosis ,Pathology ,RB1-214 ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Bi Lin,1 Chaohao Huang2 1Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People’s Republic of China; 2Department of Hepatological Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People’s Republic of ChinaCorrespondence: Chaohao Huang, Department of Hepatological Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People’s Republic of China, Email kirbylin2010@sina.comPurpose: Acute pancreatitis (AP) is a common digestive disorder characterized by high morbidity and mortality. This study aims to uncover differentially expressed long noncoding RNAs (lncRNAs) and mRNAs, as well as related pathways, in the early stage of acute pancreatitis (AP), with a focus on the role of Neat1 in AP and severe acute pancreatitis (SAP).Methods: In this study, we performed high-throughput RNA sequencing on pancreatic tissue samples from three normal mice and three mice with cerulein-induced AP to describe and analyze the expression profiles of long non-coding RNAs (lncRNAs) and mRNAs. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses were conducted on the differentially expressed mRNAs to identify enriched pathways and biological processes. An lncRNA-miRNA-mRNA interaction network was constructed to elucidate potential regulatory mechanisms. Furthermore, we utilized Neat1 knockout mice to investigate the role of Neat1 in the pathogenesis of cerulein-AP and L-arginine-severe acute pancreatitis (SAP).Results: Our results revealed that 261 lncRNAs and 1522 mRNAs were differentially expressed in the cerulein-AP group compared to the control group. GO and KEGG analyses of the differentially expressed mRNAs indicated that the functions of the corresponding genes are enriched in cellular metabolism, intercellular structure, and positive regulation of inflammation, which are closely related to the central events in the pathogenesis of AP. A ceRNA network involving 5 lncRNAs, 226 mRNAs, and 61 miRNAs were constructed. Neat1 was identified to have the potential therapeutic effects in AP. Neat1 knockout in mice inhibited pyroptosis in both the AP/SAP mouse models.Conclusion: We found that lncRNAs, particularly Neat1, play a significant role in the pathogenesis of AP. This finding may provide new insights into further exploring the pathogenesis of SAP and could lead to the identification of new targets for the treatment of AP and SAP.Keywords: acute pancreatitis, lncRNA, ceRNA, NEAT1, pyroptosis
- Published
- 2024
15. Expression Levels of lncRNA NEAT1, miRNA-21, and IL-17 in a Group of Egyptian Patients with Behçet’s Disease: Relation to Disease Manifestations and Activity
- Author
-
Hussein WH, Ramadan H, Labib S, Hegazy GA, Shaker OG, Yusuf SM, Hassanien MA, and Haroon MM
- Subjects
long noncoding ribonucleic acids ,micrornas ,neat1 ,mir-21 ,il-17 ,behçet’s disease. ,Medicine (General) ,R5-920 - Abstract
Wafaa H Hussein,1 Hala Ramadan,2 Safa Labib,2 Gehan A Hegazy,3,4 Olfat G Shaker,5 Sherif M Yusuf,6 Mohammed A Hassanien,7 Maysa M Haroon1 1Rheumatology Department, Faculty of Medicine, Cairo University, Cairo, Egypt; 2Internal Medicine Department, Faculty of Medicine, Cairo University, Cairo, Egypt; 3Clinical Biochemistry Department, Faculty of Medicine, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia; 4Medical Division Department, National Research Centre, Giza, Egypt; 5Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Cairo University, Cairo, Egypt; 6Internal Medicine Department, Medical Research Institute, Alexandria University, Alexandria, Egypt; 7Pharmacy Practice Department, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Kingdom of Saudi ArabiaCorrespondence: Maysa M Haroon, Rheumatology Department, Faculty of Medicine, Cairo University, 71 El Kasr El Aini Street, P.O. Box 11562, Cairo, Egypt, Tel +201025868370, Email maysaharoon@yahoo.comBackground: Long noncoding ribonucleic acids (lncRNAs), small noncoding RNAs known as microRNAs (miRNAs) as well as some cytokines are recently thought to have a role in many inflammatory and autoimmune disorders including Behçet’s disease (BD). This chronic multisystem disease lacks the particular histological or laboratory findings that might aid in its diagnosis. Therefore, any association with such molecules may have an impact on understanding the disease pathogenesis and/or management. The current study compared the levels of NEAT1, miR-21 and IL17 levels in sera of Egyptian BD patients and healthy individuals. The expression levels of these molecules were further investigated for their association with BD manifestations and activity aiming to explore their potential application in disease management.Results: NEAT1 & miR-21 showed down-regulation while IL-17 showed up-regulation among BD patients as compared to controls. IL-17 had significant correlation with major vessels involvement and cyclophosphamide intake. NEAT1 showed a significant negative correlation with colchicine intake. Disease activity did not correlate significantly with any of NEAT1, miR-21 or IL-17.Conclusion: NEAT1, miR-21 and IL17 might have a role in Behçet’s disease pathogenesis, so more research is needed to unveil that role and their potential usage as biomarkers for the diagnosis or therapeutic targets.Keywords: long noncoding ribonucleic acids, microRNAs, NEAT1, miR-21, IL-17, Behçet’s disease
- Published
- 2024
16. Single-cell sequencing analysis revealed that NEAT1 was a potential biomarker and therapeutic target of prostate cancer
- Author
-
Xing Li, Yanjun Li, Lei Zhang, and Huimin Long
- Subjects
Prostate cancer ,Benign prostatic hyperplasia ,scRNA-seq ,NEAT1 ,Cell derived xenograft ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Prostate cancer (PCa) usually manifests atypical symptoms in the early stage, and once symptoms appear, most PCa patients have developed to the advanced stage, failing to undergo radical surgery. In this study, PCa occurrence-related biomarkers were explored based on single-cell RNA sequencing (scRNA-seq) data. Methods scRNA-seq data of prostate normal (Normal), benign prostatic hyperplasia (BPH), and PCa (Tumor) samples were acquired from the Gene Expression Omnibus (GEO). Cellular subsets associated with PCa occurrence were obtained using cell annotation. Additionally, the mRNA expression of nuclear enriched abundant transcript 1 (NEAT1) was detected by quantitative real-time PCR (qRT-PCR). The effects of NEAT1 on cell proliferation and apoptosis were analyzed by 5-ethynyl-2-deoxyuridine (EdU) and flow cytometry. Subsequently, cell-derived xenograft (CDX) models were constructed and divided into the LV-NC and LV-shNEAT1 groups. After the tumor tissues of CDX model mice in each group were extracted, the cell growth and Ki67 expression were observed separately using hematoxylin-eosin (H&E) staining and immunohistochemistry (IHC). Results Ten cellular subsets were obtained via cell annotation, and significantly differential changes were observed between Basal intermediate and Luminal during the course of BPH to PCa. NEAT1−Luminal was highly recruited in the Tumor group with low stemness and high malignancy scores. Matrix metallopeptidase 7 (MMP7)− keratin 17 (KRT17)−Basal intermediate had high ratios in the Tumor group with low stemness and high malignancy scores. The results of pseudotime analysis revealed that NEAT1−Luminal in the Tumor group were consistently distributed with tumor stage cells. In vitro assays showed that NEAT1 expression was elevated in PCa cells, and NEAT1 knockdown could inhibit cell proliferation and induce apoptosis. CDX assays indicated that silencing NEAT1 could reduce the growth rate of PCa tumor volume in CDX model mice. H&E staining results showed that nuclei of tumor cells were reduced and exhibited lighter color in the LV-shNEAT1 group compared with the LV-NC group. IHC results showed that Ki67 positivity was significantly lower in the LV-shNEAT1 group than in the LV-NC group. Conclusion NEAT1 expression is increased in PCa, and NEAT1 can be a potential biomarker and therapeutic target for PCa.
- Published
- 2024
- Full Text
- View/download PDF
17. LncRNAs SOX2-OT and NEAT1 act as a potential biomarker for esophageal squamous cell carcinoma.
- Author
-
Kumar, Rajiv Ranjan, Mohanta, Adrija, Rana, Manjit Kaur, Uttam, Vivek, Tuli, Hardeep Singh, and Jain, Aklank
- Subjects
LINCRNA ,SQUAMOUS cell carcinoma ,OVERALL survival ,RECEIVER operating characteristic curves ,ALCOHOL drinking - Abstract
Despite strides in diagnostic and therapeutic approaches for ESCC, patient survival rates remain relatively low. Recent studies highlight the pivotal role of long non-coding RNAs (lncRNAs) in regulating diverse cellular activities in humans. Dysregulated lncRNAs have emerged as potential diagnostic indicators across various cancers, including ESCC. However, further research is necessary to effectively leverage ESCC-associated lncRNAs in clinical settings. Understanding their clinical significance for ESCC diagnosis and their mechanisms can pave the way for more effective therapeutic strategies. Our qRT-PCR analysis revealed significant downregulation of SOX2-OT (~ 2.02-fold) and NEAT1 (~ 1.53-fold) in ESCC blood samples. These lncRNAs show potential as biomarkers for distinguishing ESCC patients from healthy individuals, with ROC curves and AUC values of 0.736 for SOX2-OT and 0.621 for NEAT1. Further analysis examined the correlation between SOX2-OT and NEAT1 expression and various clinicopathological factors, including age, gender, smoking, alcohol use, hot beverage intake, tumor grade, and TNM stages. In-silico studies highlighted their roles in miRNA sponging via mTOR and MAPK pathways, while co-expression network analysis identified associated genes. This research paves the way for future studies on ESCC prognosis using SOX2-OT and NEAT1 as predictive markers. By thoroughly investigating the functions of these lncRNAs, we aim to deepen our understanding of their potential as diagnostic markers and their role in facilitating effective therapeutic interventions for esophageal squamous cell carcinoma (ESCC) within clinical contexts. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
18. Small Molecule Inhibitor of Protein Kinase C DeltaI (PKCδI) Decreases Inflammatory Pathways and Gene Expression and Improves Metabolic Function in Diet-Induced Obese Mouse Model.
- Author
-
Osborne, Brenna, Patel, Rekha S., Krause-Hauch, Meredith, Lui, Ashley, Vidyarthi, Gitanjali, and Patel, Niketa A.
- Subjects
- *
PROTEIN kinase C , *FAT cells , *SMALL molecules , *TYPE 2 diabetes , *HUMAN stem cells - Abstract
Simple Summary: Obesity results from an excessive level of fat depots and is a global epidemic. Obesity promotes other chronic diseases, such as type 2 diabetes and cardiovascular disease. Adipose (fat cells) is an important endocrine regulator of energy and glucose homeostasis. Protein Kinase Cδ1 (PKCδI) is present in fat cells and regulates their growth and survival. Caspase-3, activated by cell-death signals, cleaves PKCδI and releases a catalytic fragment (PKCδI_C). The PKCδI_C fragment induces inflammation and cell death in adipocytes, advancing the detrimental metabolic effects of obesity. Our prior study had developed a therapeutic, referred to as NP627, to inhibit the release of the PKCδI_C fragment. Previously, the safety, efficacy, and specificity of NP627 have been established using cellular assays with obese adult human adipose stem cells (hASC). The goal of this project was to test the safety and efficacy of NP627 in a diet-induced obese (DIO) mouse model which mimics human obesity. Our results show that NP627 effectively inhibited the release of PKCδI_C, decreased inflammation, and restored glucose metabolism in DIO mice. Unbiased transcriptomics analysis revealed genes that were changed, post-NP627 treatment, in DIO mice. Thus, we believe NP627 has remarkable therapeutic potential for improving the metabolic consequences of obesity. Obesity promotes metabolic diseases such as type 2 diabetes and cardiovascular disease. PKCδI is a serine/threonine kinase which regulates cell growth, differentiation, and survival. Caspase-3 cleavage of PKCδI releases the C-terminal catalytic fragment (PKCδI_C), which promotes inflammation and apoptosis. We previously demonstrated an increase in PKCδI_C in human obese adipose tissue (AT) and adipocytes. Subsequently, we designed a small molecule drug called NP627 and demonstrated that NP627 specifically inhibited the release of PKCδI_C in vitro. Here, we evaluate the in vivo safety and efficacy of NP627 in a diet-induced obese (DIO) mouse model. The results demonstrate that NP627 treatment in DIO mice increased glucose uptake and inhibited the cleavage of PKCδI_C in the AT as well as in the kidney, spleen, and liver. Next, RNAseq analysis was performed on the AT from the NP627-treated DIO mice. The results show increases in ADIPOQ and CIDEC, upregulation of AMPK, PI3K-AKT, and insulin signaling pathways, while inflammatory pathways were decreased post-NP627 administration. Further, levels of lncRNAs associated with metabolic pathways were affected by NP627 treatment. In conclusion, the study demonstrates that NP627, a small-molecule inhibitor of PKCδI activity, is not toxic and that it improves the metabolic function of DIO mice in vivo. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
19. Single-cell sequencing analysis revealed that NEAT1 was a potential biomarker and therapeutic target of prostate cancer.
- Author
-
Li, Xing, Li, Yanjun, Zhang, Lei, and Long, Huimin
- Subjects
BENIGN prostatic hyperplasia ,HEMATOXYLIN & eosin staining ,INHIBITION of cellular proliferation ,GENE expression ,CELL nuclei - Abstract
Background: Prostate cancer (PCa) usually manifests atypical symptoms in the early stage, and once symptoms appear, most PCa patients have developed to the advanced stage, failing to undergo radical surgery. In this study, PCa occurrence-related biomarkers were explored based on single-cell RNA sequencing (scRNA-seq) data. Methods: scRNA-seq data of prostate normal (Normal), benign prostatic hyperplasia (BPH), and PCa (Tumor) samples were acquired from the Gene Expression Omnibus (GEO). Cellular subsets associated with PCa occurrence were obtained using cell annotation. Additionally, the mRNA expression of nuclear enriched abundant transcript 1 (NEAT1) was detected by quantitative real-time PCR (qRT-PCR). The effects of NEAT1 on cell proliferation and apoptosis were analyzed by 5-ethynyl-2-deoxyuridine (EdU) and flow cytometry. Subsequently, cell-derived xenograft (CDX) models were constructed and divided into the LV-NC and LV-shNEAT1 groups. After the tumor tissues of CDX model mice in each group were extracted, the cell growth and Ki67 expression were observed separately using hematoxylin-eosin (H&E) staining and immunohistochemistry (IHC). Results: Ten cellular subsets were obtained via cell annotation, and significantly differential changes were observed between Basal intermediate and Luminal during the course of BPH to PCa. NEAT1
− Luminal was highly recruited in the Tumor group with low stemness and high malignancy scores. Matrix metallopeptidase 7 (MMP7)− keratin 17 (KRT17)− Basal intermediate had high ratios in the Tumor group with low stemness and high malignancy scores. The results of pseudotime analysis revealed that NEAT1− Luminal in the Tumor group were consistently distributed with tumor stage cells. In vitro assays showed that NEAT1 expression was elevated in PCa cells, and NEAT1 knockdown could inhibit cell proliferation and induce apoptosis. CDX assays indicated that silencing NEAT1 could reduce the growth rate of PCa tumor volume in CDX model mice. H&E staining results showed that nuclei of tumor cells were reduced and exhibited lighter color in the LV-shNEAT1 group compared with the LV-NC group. IHC results showed that Ki67 positivity was significantly lower in the LV-shNEAT1 group than in the LV-NC group. Conclusion: NEAT1 expression is increased in PCa, and NEAT1 can be a potential biomarker and therapeutic target for PCa. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
20. Potential role of long non-coding RNA (nuclear paraspeckle assembly transcript 1 and metastasis associated lung adenocarcinoma transcript 1) in periodontitis pathogenesis: A systematic review
- Author
-
Saif M Al-Mufti, Ali A Abdulkareem, Mike Milward, and Paul R Cooper
- Subjects
Periodontitis ,Long non-coding RNA ,NEAT1 ,MALAT1 ,Pathogenesis ,Dentistry ,RK1-715 - Abstract
Background: Dysregulation of Long non-coding (lnc)RNAs has been linked to periodontitis, with potential importance in disease onset and progression. These lncRNAs potentially modulate inflammatory/immune responses during periodontitis. This review aimed to highlight the potential role of Nuclear Paraspeckle Assembly Transcript (NEAT)1 and Metastasis Associated Lung Adenocarcinoma Transcript (MALAT)1 lncRNAs in periodontitis pathogenesis. Methods: A literature search of three electronic databases was performed in SCOPUS, MEDLINE (PubMed) and EMBASE using search terms linking periodontitis/periodontal disease with NEAT1 and MALAT1 lncRNAs. Duplicate publications were removed from the retrieved articles which were then filtered to include the most relevant papers for evidence synthesis. Results: Nine studies (in vitro and in vivo) were included in the final analysis. The total number of studies investigating the role of NEAT1 and MALAT1 in pathogenesis of periodontitis was 4 and 5, respectively. The findings indicated gene expression changes of NEAT1 and MALAT1 in periodontitis compared with periodontal health. Conclusion: No concrete evidence could be withdrawn from this review; however, results suggested that lncRNAs, NEAT1 and MALAT1 could be involved in periodontitis pathogenesis. However, further in vivo studies are required to confirm these findings.
- Published
- 2024
- Full Text
- View/download PDF
21. Involvement of paraspeckle components in viral infections
- Author
-
Romane Milcamps and Thomas Michiels
- Subjects
Interferon ,internal ribosome entry site (IRES) ,long non-coding RNA ,NEAT1 ,paraspeckles ,SFPQ ,Genetics ,QH426-470 ,Cytology ,QH573-671 - Abstract
Paraspeckles are non-membranous subnuclear bodies, formed through the interaction between the architectural long non-coding RNA (lncRNA) nuclear paraspeckle assembly transcript 1 (NEAT1) and specific RNA-binding proteins, including the three Drosophila Behavior/Human Splicing (DBHS) family members (PSPC1 (Paraspeckle Component 1), SFPQ (Splicing Factor Proline and Glutamine Rich) and NONO (Non-POU domain-containing octamer-binding protein)). Paraspeckle components were found to impact viral infections through various mechanisms, such as induction of antiviral gene expression, IRES-mediated translation, or viral mRNA polyadenylation. A complex involving NEAT1 RNA and paraspeckle proteins was also found to modulate interferon gene transcription after nuclear DNA sensing, through the activation of the cGAS-STING axis. This review aims to provide an overview on how these elements actively contribute to the dynamics of viral infections.
- Published
- 2024
- Full Text
- View/download PDF
22. Long non-coding RNA NEAT1 promotes multiple myeloma malignant transformation via targeting miR-485-5p/ABCB8
- Author
-
Yuxiu Xu, Tao Wang, Jiangwei Wan, Dongsheng Ma, Hongyang Zhang, Dongru Cheng, Jing Yang, and Meng Wang
- Subjects
LncRNA ,neat1 ,Multiple myeloma ,Mir-485-5p ,abcb8 ,Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Multiple myeloma (MM) is the second most common hematological cancer all over the world. Long non-coding RNA (lncRNA) nuclear-enriched autosomal transcript-1 (NEAT1) have been reported to play important roles in the development and progression of multiple human malignancies like MM. However, the functional role and molecular mechanism of NEAT1 in MM progression still needs more support to identify potential targets of MM. In the present study, we focused on the clinical and biological significance of NEAT1 in MM. We demonstrated that NEAT1 was up-regulated in MM tissues and cell line. NEAT1 silencing significantly inhibited cell proliferation and promoted cell apoptosis in vitro. And we illustrated that miR-485-5p was a direct target of NEAT1 and the effect of down-regulated NEAT1 on MM cells was partially reversed by the miR-485-5p antisense oligonucleotide (ASO-miR-485-5p). Further investigation revealed that ABCB8 directly interacted with miR-485-5p. Similarly, in vivo experiments confirmed that down-regulated NEAT1 inhibited tumor growth and ABCB8 expression. Taken together, our results demonstrate for the first time that NEAT1/miR-485-5p/ABCB8 axis may be a key pathway for the development and progression of MM, and they may provide a novel avenue for targeted therapy.
- Published
- 2024
- Full Text
- View/download PDF
23. The role of lncRNA NEAT1 in human cancer chemoresistance
- Author
-
Feng Long, Xue Li, Jingyu Pan, Hailin Ye, Cuixia Di, Yong Huang, Jiawei Li, Xuan Zhou, Huiyi Yi, Qiaozhen Huang, and Jing Si
- Subjects
Cancer ,Chemotherapy resistance ,NEAT1 ,Mechanism of action ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 ,Cytology ,QH573-671 - Abstract
Abstract Chemotherapy is currently one of the most effective methods in clinical cancer treatment. However, chemotherapy resistance is an important reason for poor chemotherapy efficacy and prognosis, which has become an urgent problem to be solved in the field of cancer chemotherapy. Therefore, it is very important to deeply study and analyze the mechanism of cancer chemotherapy resistance and its regulatory factors. Long non-coding RNA nuclear paraspeckle assembly transcript 1 (LncRNA NEAT1) has been shown to be closely associated with chemotherapy resistance in cancer. NEAT1 induces cancer cell resistance to chemotherapeutic drugs by regulating cell apoptosis, cell cycle, drug transport and metabolism, DNA damage repair, EMT, autophagy, cancer stem cell characteristics, and metabolic reprogramming. This indicates that NEAT1 may be an important target to overcome chemotherapy resistance and is expected to be a potential biomarker to predict the effect of chemotherapy. This article summarizes the expression characteristics and clinical characteristics of NEAT1 in different cancers, and deeply discusses the regulatory role of NEAT1 in cancer chemotherapy resistance and related molecular mechanisms, aiming to clarify NEAT1 as a new target to overcome cancer chemotherapy resistance and the feasibility of chemotherapy sensitizers, with a view to providing a potential therapeutic direction for overcoming the dilemma of cancer resistance in the future.
- Published
- 2024
- Full Text
- View/download PDF
24. Long noncoding nuclear enriched abundant transcript 1_2 is a promising biomarker for childhood‐onset systemic lupus erythematosus
- Author
-
Shipeng Li, Xia Wang, Xiaozhen Zhao, Jianghong Deng, Weiying Kuang, Junmei Zhang, Xiaohua Tan, Chao Li, and Caifeng Li
- Subjects
Biomarker ,Children ,LncRNA ,SLE ,NEAT1 ,Pediatrics ,RJ1-570 - Abstract
ABSTRACT Importance Systemic lupus erythematosus (SLE) is a diffuse connective tissue disease with complex clinical manifestations and prolonged course. The early diagnosis and condition monitoring of SLE are crucial to disease prognosis. Objective To assess the diagnostic value of long noncoding RNA (lncRNA) nuclear enriched abundant transcript 1 (NEAT1) in childhood‐onset SLE (cSLE). Methods Fifty‐seven children diagnosed with SLE, 40 children diagnosed with juvenile idiopathic arthritis (JIA), and 40 healthy children were included. Peripheral blood samples from each patient were collected. A quantitative polymerase chain reaction was used to confirm the expression of lncNEAT1_1 and lncNEAT1_2 in peripheral blood. Associations among parameters were analyzed using the Mann‐Whitney U test or independent sample t‐test. Results The expression of both lncNEAT1_1 and lncNEAT1_2 in patients with cSLE were significantly higher than that of healthy control and patients with JIA. Receiver operating characteristic curves revealed an area under the curve (AUC) of 0.633 (95% confidence interval [CI], 0.524–0.742; P = 0.024) for lncNEAT1_1. The AUC of lncNEAT1_2 was 0.812 (95% CI, 0.727–0.897; P < 0.0001) to discriminate individuals with cSLE from health control and children with JIA with a sensitivity of 0.622 and a specificity of 0.925. Moreover, lncNEAT1_2 expression was higher in patients with cSLE presenting with fever, lupus nephritis, elevated erythrocyte sedimentation rate, active disease activity, and decreased C3 level, compared with those without these conditions. However, no similar correlation was observed for lncNEAT1_1. Interpretation The expression of lncNEAT1_2 was significantly elevated in children with SLE, especially those with fever, renal involvement, and low C3 levels. These findings suggest that lncNEAT1_2 may represent a potential biomarker for cSLE.
- Published
- 2024
- Full Text
- View/download PDF
25. Glioma-Stem-Cell-Derived Exosomes Remodeled Glioma-Associated Macrophage via NEAT1/miR-125a/STAT3 Pathway.
- Author
-
Pan, Tong, Xie, Dong-Kun, Li, Juan, Qiang, Yu-Jie, Fan, Song-Yuan, Wang, Ting-Ting, Han, Yuan-Yuan, Zang, Jian, Yang, Yang, Zhao, Jun-Long, Li, San-Zhong, and Wu, Shuang
- Subjects
- *
GLIOMA treatment , *GLIOMAS , *MACROPHAGES , *RADIOTHERAPY , *DRUG resistance in cancer cells , *RESEARCH funding , *MICRORNA , *CELLULAR signal transduction , *MICE , *CANCER chemotherapy , *ANIMAL experimentation , *DNA damage , *STEM cells , *STAT proteins , *EXOSOMES , *IMMUNOSUPPRESSION , *DISEASE progression - Abstract
Simple Summary: Glioblastomas (GBMs) are considered the most lethal cancer in the central nervous system (CNS), whose malignant phenotypes are majorly attributed to glioma stem cells (GSCs). Despite combined surgical radiotherapy with temozolomide chemotherapy and tumor-treating fields (TTFs), the tumor almost always recurs near the resection site. Besides the contribution of GSCs, the tumor microenvironment (TME) also plays an important role in glioma recurrence. Our work has demonstrated that GSC-derived exosomes carry lncRNA NEAT1 to promote the M2 polarization of glioma-associated macrophages (GAMs). Further mechanism exploration indicated that NEAT1 represses the expression of miR-125a in GAMs significantly. The decrease in miR-125a induces the elevation of target gene STAT3, which is required for macrophage M2 polarization. The development of M2-like GAMs contributes to the immunosuppressive microenvironment and glioma progression. Our findings elucidate the functions and mechanisms of the crosstalk between GSCs and GAMs via exosomes, providing new therapeutic targets and strategies for glioma. Glioblastoma (GBM), as the most common primary brain tumor, usually results in an extremely poor prognosis, in which glioma stem cells (GSCs) and their immunosuppressive microenvironment prominently intervene in the resistance to radiotherapy and chemotherapy that directly leads to tumor recurrence and shortened survival time. The specific mechanism through which exosomes generated from GSCs support the creation of an immunosuppressive microenvironment remains unknown, while it is acknowledged to be engaged in intercellular communication and the regulation of the glioma immunosuppressive microenvironment. The elevated expression of LncRNA-NEAT1 was found in glioma cells after radiotherapy, chemotherapy, and DNA damage stimulation, and NEAT1 could promote the malignant biological activities of GSCs. Emerging evidence suggests that lncRNAs may reply to external stimuli or DNA damage by playing a role in modulating different aspects of tumor biology. Our study demonstrated a promotive role of the carried NEAT1 by GSC-derived exosomes in the polarization of M2-like macrophages. Further experiments demonstrated the mediative role of miR-125a and its target gene STAT3 in NEAT1-induced polarization of M2-like macrophages that promote glioma progression. Our findings elucidate the mechanism by which GSCs influence the polarization of M2-like macrophages through exosomes, which may contribute to the formation of immunosuppressive microenvironments. Taken together, our study reveals the miR-125a-STAT3 pathway through which exosomal NEAT1 from treatment-resistant GSCs contributes to M2-like macrophage polarization, indicating the potential of exosomal NEAT1 for treating glioma. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
26. LncRNA NEAT1 targets miR‐125/ADAM9 mediated NF‐κB pathway in inflammatory response of rosacea.
- Author
-
Xu, Sijia and Dong, Wenxin
- Subjects
- *
INFLAMMATION , *ROSACEA , *LINCRNA , *WESTERN immunoblotting , *PHENOTYPES , *LUCIFERASES - Abstract
Objective: To investigate the role of NEAT1 targeted regulation of miR‐125/ADAM9 mediated NF‐κB pathway in inflammatory response in rosacea. Method: HaCaT cell rosacea phenotype was induced by LL37. The connection targeted by NEAT1 and miR‐125a‐5p was confirmed by Double‐Luciferase report analysis. qPCR was employed to assess the levels of expression for NEAT1, miR‐125a‐5p, and ADAM9 genes. The levels of expression for ADAM9/TLR2/NF‐κB P65 pathway proteins in each batch of cells were determined by Western blotting. The levels of expression for inflammatory factors, including TNF‐α, IL‐1β, IL‐6, and IL‐18, were measured through ELISA experimentation. Results: LL37 could successfully induce HaCaT cells to exhibit rosacea phenotype. The luciferase report experiment confirmed that NEAT1 could target and bind miR‐125a‐5p and inhibit its expression. ADAM9 exhibited increased expression in LL37‐induced HaCaT cells, showing a positive association with NEAT1 expression and inverse relationship with miR‐125a‐5p activation. LL37 treatment promoted the expression of ADAM9/TLR2/NF‐κB P65 pathway proteins. Silencing ADAM9 can inhibit the inflammatory signaling pathway and reduce the level of TNF‐α, IL‐1β, IL‐6, and IL‐18 in HaCaT cells. Conclusion: NEAT1 can suppress the production of miR‐125a‐5p and activate the TLR2/NF‐κB inflammatory pathway mediated by ADAM9, thereby promoting the inflammatory response in rosacea. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
27. Diagnostic value and clinical significance of lncRNA NEAT1 combined with miR-425-3p in children with viral myocarditis.
- Author
-
Jielin Gao, Lili Qin, Qiaozhi Guo, Dongxia Zhao, Guomei Ma, Kuilong Zhou, Shuang Wang, and Hengrui Hao
- Abstract
Background. Viral myocarditis (VMC) is common in children. Previous studies have reported the clinical value of nuclear paraspeckle assembly transcript 1 (NEAT1) and microRNA-425-3p (miR-425-3p) in certain diseases, but not in VMC. This article was designed to investigate the expression of long noncoding RNA (lncRNA) NEAT1 and miR-425-3p in the serum of patients with VMC and their clinical significance. Methods. We assessed VMC and healthy patients and analyzed differences in the expression levels of NEAT1 and miR-425-3p. The correlation and targeting relationship between the two were reported by Spearman correlation analysis and luciferase reporter assay. ROC curves were plotted to reflect the diagnostic effect of both. In addition, according to the 12-month prognostic effect grouping, patients with VMC were separated into a group with good vs. poor prognosis, and the difference in the expression levels of NEAT1 and miR-425-3p between the two groups were analyzed. The ability of the two markers in the prognosis of VMC was further analyzed by multiple logistic regression. Results. NEAT1 expression was up-regulated in VMC and miR-425-3p expression was down-regulated, and there was a negative correlation and targeting link between the two. The diagnostic efficacy of both NEAT1 and miR-425-3p was higher than that of a single indicator. High expression of NEAT1 and low expression of miR-425-3p were found in VMC patients with poor prognosis. Both were independent influencers of VMC prognosis. Conclusion. NEAT1 and miR-425-3p expressions were affected by VMC and had important clinical implications for VMC, indicating for the first time the clinical function of NEAT1 and miR-425-3p in VMC. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
28. The role of lncRNA NEAT1 in human cancer chemoresistance.
- Author
-
Long, Feng, Li, Xue, Pan, Jingyu, Ye, Hailin, Di, Cuixia, Huang, Yong, Li, Jiawei, Zhou, Xuan, Yi, Huiyi, Huang, Qiaozhen, and Si, Jing
- Subjects
DRUG resistance in cancer cells ,CANCER chemotherapy ,CHEMOSENSITIZERS ,METABOLIC reprogramming ,LINCRNA - Abstract
Chemotherapy is currently one of the most effective methods in clinical cancer treatment. However, chemotherapy resistance is an important reason for poor chemotherapy efficacy and prognosis, which has become an urgent problem to be solved in the field of cancer chemotherapy. Therefore, it is very important to deeply study and analyze the mechanism of cancer chemotherapy resistance and its regulatory factors. Long non-coding RNA nuclear paraspeckle assembly transcript 1 (LncRNA NEAT1) has been shown to be closely associated with chemotherapy resistance in cancer. NEAT1 induces cancer cell resistance to chemotherapeutic drugs by regulating cell apoptosis, cell cycle, drug transport and metabolism, DNA damage repair, EMT, autophagy, cancer stem cell characteristics, and metabolic reprogramming. This indicates that NEAT1 may be an important target to overcome chemotherapy resistance and is expected to be a potential biomarker to predict the effect of chemotherapy. This article summarizes the expression characteristics and clinical characteristics of NEAT1 in different cancers, and deeply discusses the regulatory role of NEAT1 in cancer chemotherapy resistance and related molecular mechanisms, aiming to clarify NEAT1 as a new target to overcome cancer chemotherapy resistance and the feasibility of chemotherapy sensitizers, with a view to providing a potential therapeutic direction for overcoming the dilemma of cancer resistance in the future. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
29. NEAT1 regulates VSMC differentiation and calcification in as long noncoding RNA NEAT1 enhances phenotypic and osteogenic switching of vascular smooth muscle cells in atherosclerosis via scaffolding EZH2.
- Author
-
Yin, Chengye, Ge, Zhuowang, Yuan, Jiali, Chen, Yuhan, Tang, Yong, Xiang, Yin, and Zhang, Yachen
- Subjects
- *
VASCULAR smooth muscle , *LINCRNA , *MUSCLE cells , *PHENOTYPES , *ARTERIAL calcification , *P16 gene , *BONE growth - Abstract
Atherosclerosis (AS) is a significant contributor to cardio-cerebrovascular ischemia diseases, resulting in high mortality rates worldwide. During AS, vascular smooth muscle cells (VSMCs) play a crucial role in plaque formation by undergoing phenotypic and osteogenic switching. Long noncoding RNA nuclear paraspeckle assembly transcript 1 (NEAT1) has previously been identified as a nuclear regulator that promotes tumorigenesis and metastasis, but its role in regulating VSMCs in AS remains unclear. Our study aimed to investigate the biological functions and specific mechanisms of NEAT1 in regulating VSMCs in AS. We found that NEAT1 was upregulated in the aortas of AS mouse models and dedifferentiated primary VSMCs. Silencing NEAT1 in vitro attenuated the proliferation, migration, and osteogenic differentiation of VSMCs, while NEAT1 overexpression had the opposite effect. Furthermore, NEAT1 promoted VSMC osteogenic differentiation and vascular calcification in both in vivo and in vitro vascular calcification models. We also discovered that NEAT1 directly activates enhancer of zeste homolog 2 (EZH2), an epigenetic enzyme that suppresses the expression of senescence- and antimigration-related genes, by translocating it into the nucleus. CUT&Tag assay revealed that NEAT1 guides EZH2 to the promoters of senescence-related genes (P16, P21, and TIMP3), methylating local histones to reduce their transcription. Our findings suggest that NEAT1 functions in AS by modulating the epigenetic function of EZH2, which enhances the proliferation, migration, and osteogenic differentiation of VSMCs. This study provides new insights into the molecular mechanisms underlying the pathogenesis of AS and highlights the potential of NEAT1 as a therapeutic target of AS. NEW & NOTEWORTHY: Our study demonstrates that the upregulation of long noncoding RNA nuclear paraspeckle assembly transcript 1 (NEAT1) promotes proliferation and migration during phenotypic switching of vascular smooth muscle cells in atherosclerosis. We also provide in vivo and in vitro evidence that NEAT1 accelerates vascular calcification. Our findings identified the direct interaction between enhancer of zeste homolog 2 (EZH2) and NEAT1 during atherosclerosis. NEAT1 is necessary for EZH2 to translocate from the cytoplasm to the nucleus, where EZH2 epigenetically inhibits the expression of genes related to senescence and antimigration. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
30. 新疆地区人群非综合征型唇腭裂与外周血 FOXN3-SIN3A复合物表达量相关性研究.
- Author
-
多力昆, 吾甫尔, 地丽拜尔, 依明江, 卡米力江, 买买提明, 李军, 乌丽凡, and 托勒恒
- Abstract
Copyright of West China Journal of Stomatology is the property of Sichuan University, West China College of Stomatology and its content may not be copied or emailed to multiple sites or posted to a listserv without the copyright holder's express written permission. However, users may print, download, or email articles for individual use. This abstract may be abridged. No warranty is given about the accuracy of the copy. Users should refer to the original published version of the material for the full abstract. (Copyright applies to all Abstracts.)
- Published
- 2024
- Full Text
- View/download PDF
31. Long noncoding nuclear enriched abundant transcript 1_2 is a promising biomarker for childhood‐onset systemic lupus erythematosus.
- Author
-
Li, Shipeng, Wang, Xia, Zhao, Xiaozhen, Deng, Jianghong, Kuang, Weiying, Zhang, Junmei, Tan, Xiaohua, Li, Chao, and Li, Caifeng
- Subjects
LUPUS nephritis ,SYSTEMIC lupus erythematosus ,BIOMARKERS ,RECEIVER operating characteristic curves ,JUVENILE idiopathic arthritis ,LINCRNA - Abstract
Importance: Systemic lupus erythematosus (SLE) is a diffuse connective tissue disease with complex clinical manifestations and prolonged course. The early diagnosis and condition monitoring of SLE are crucial to disease prognosis. Objective: To assess the diagnostic value of long noncoding RNA (lncRNA) nuclear enriched abundant transcript 1 (NEAT1) in childhood‐onset SLE (cSLE). Methods: Fifty‐seven children diagnosed with SLE, 40 children diagnosed with juvenile idiopathic arthritis (JIA), and 40 healthy children were included. Peripheral blood samples from each patient were collected. A quantitative polymerase chain reaction was used to confirm the expression of lncNEAT1_1 and lncNEAT1_2 in peripheral blood. Associations among parameters were analyzed using the Mann‐Whitney U test or independent sample t‐test. Results: The expression of both lncNEAT1_1 and lncNEAT1_2 in patients with cSLE were significantly higher than that of healthy control and patients with JIA. Receiver operating characteristic curves revealed an area under the curve (AUC) of 0.633 (95% confidence interval [CI], 0.524–0.742; P = 0.024) for lncNEAT1_1. The AUC of lncNEAT1_2 was 0.812 (95% CI, 0.727–0.897; P < 0.0001) to discriminate individuals with cSLE from health control and children with JIA with a sensitivity of 0.622 and a specificity of 0.925. Moreover, lncNEAT1_2 expression was higher in patients with cSLE presenting with fever, lupus nephritis, elevated erythrocyte sedimentation rate, active disease activity, and decreased C3 level, compared with those without these conditions. However, no similar correlation was observed for lncNEAT1_1. Interpretation: The expression of lncNEAT1_2 was significantly elevated in children with SLE, especially those with fever, renal involvement, and low C3 levels. These findings suggest that lncNEAT1_2 may represent a potential biomarker for cSLE. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
32. Molecular Mechanisms of Tumorgenesis and Metastasis of Long Non-coding RNA (lncRNA) NEAT1 in Human Solid Tumors; An Update.
- Author
-
Alshahrani, Mohammad Y., Saleh, Raed Obaid, Hjazi, Ahmed, Bansal, Pooja, Kaur, Harpreet, Deorari, Mahamedha, Altalbawy, Farag M. A., Kareem, Anaheed Hussein, Hamzah, Hamza Fadhel, and Mohammed, Bahira Abdulrazzaq
- Abstract
The expression of the nuclear paraspeckle assembly transcript 1 (NEAT1), as a well-known long non-coding RNA (lncRNA), is often upregulated in varied types of cancers and associated with poor survival outcomes in patients suffering from tumors. NEAT1 promotes the tumors growth by influencing the various genes' expression profile that regulate various aspects of tumor cell behavior, in particular tumor growth, metastasis and drug resistance. This suggests that NEAT1 are capable of serving as a new diagnostic biomarker and target for therapeutic intervention. Through interrelation with enhancer of zeste homolog 2 (EZH2), NEAT1 acts as a scaffold RNA molecule, and thus regulating the expression EZH2-associated genes. Additionally, by perform as miRNA sponge, it constrains suppressing the interactions between miRNAs-mediated degradation of target mRNAs. In light of this, NEAT1 inhibition by small interfering RNA (siRNA) hampers tumorgenesis. We summarize recent findings about the expression, biological functions, and regulatory process of NEAT1 in human tumors. It specifically emphasizes the clinical significance of NEAT1 as a novel diagnostic biomarker and a promising therapeutic mark for many types of cancers. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
33. AAV mediated repression of Neat1 lncRNA combined with F8 gene augmentation mitigates pathological mediators of joint disease in haemophilia.
- Author
-
Sarangi, Pratiksha, Senthilkumar, Mohankumar B., Amit, Sonal, Kumar, Narendra, and Jayandharan, Giridhara R.
- Subjects
JOINT diseases ,LINCRNA ,JOINT pain ,ADENO-associated virus ,INTRA-articular injections ,BLOOD coagulation factor VIII - Abstract
Haemophilic arthropathy (HA), a common comorbidity in haemophilic patients leads to joint pain, deformity and reduced quality of life. We have recently demonstrated that a long non‐coding RNA, Neat1 as a primary regulator of matrix metalloproteinase (MMP) 3 and MMP13 activity, and its induction in the target joint has a deteriorating effect on articular cartilage. In the present study, we administered an Adeno‐associated virus (AAV) 5 vector carrying an short hairpin (sh)RNA to Neat1 via intra‐articular injection alone or in conjunction with systemic administration of a capsid‐modified AAV8 (K31Q) vector carrying F8 gene (F8‐BDD‐V3) to study its impact on HA. AAV8K31Q‐F8 vector administration at low dose, led to an increase in FVIII activity (16%–28%) in treated mice. We further observed a significant knockdown of Neat1 (~40 fold vs. untreated injured joint, p = 0.005) in joint tissue of treated mice and a downregulation of chondrodegenerative enzymes, MMP3, MMP13 and the inflammatory mediator‐ cPLA2, in mice receiving combination therapy. These data demonstrate that AAV mediated Neat1 knockdown in combination with F8 gene augmentation can potentially impact mediators of haemophilic joint disease. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
34. Inhibiting lncRNA NEAT1 Increases Glioblastoma Response to TMZ by Reducing Connexin 43 Expression
- Author
-
Jinxing Liang, Jia‐xiu Xie, Junhui He, Yi Li, Dongmei Wei, Rongfei Zhou, Guining Wei, Xuehua Liu, Qiudan Chen, and Dongmei Li
- Subjects
chemotherapy sensitivity ,Connexin 43 ,miR‐454‐3p ,NEAT1 ,temozolomide ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
ABSTRACT Objectives Glioblastoma multiforme (GBM) is considered the most assailant subtype of gliomas, presenting a formidable obstacle because of its inherent resistance to temozolomide (TMZ). This study aimed to characterize the function of lncRNA NEAT1 in facilitating the advancement of gliomas. Methods The expression level of NEAT1 in glioma tissues and cells was detected by qRT‐PCR. RNA interference experiment, cell proliferation assay, FITC/PI detection assay, immunoblotting, bioinformatics prediction, a double luciferase reporter gene assay, RNA immunoprecipitation (RIP) assay, SLDT assay and correlation analysis of clinical samples were performed to explore the regulatory effects of NEAT1, miR‐454‐3p and Cx43 and their role in malignant progression of GBM. The role of NEAT1 in vivo was investigated by an intracranial tumor formation experiment in mice. Results The results showed that recurring gliomas displayed elevated levels of NEAT1 compared to primary gliomas. The suppression of NEAT1 led to a restoration of sensitivity in GBM cells to TMZ. NEAT1 functioned as a competitive endogenous RNA against miR‐454‐3p. Connexin 43 was identified as a miR‐454‐3p target. NEAT1 was found to regulate gap junctional intercellular communication by modulating Connexin 43, thereby impacting the response of GBM cells to TMZ chemotherapy. Downregulation of NEAT1 resulted in enhanced chemosensitivity to TMZ and extended the survival of mice. Conclusions Overall, these results indicated that the NEAT1/miR‐454‐3p/Connexin 43 pathway influences GBM cell response to TMZ and could offer a potential new strategy for treating GBM.
- Published
- 2024
- Full Text
- View/download PDF
35. N6‐methyladenosine reader hnRNPA2B1 recognizes and stabilizes NEAT1 to confer chemoresistance in gastric cancer
- Author
-
Jiayao Wang, Jiehao Zhang, Hao Liu, Lingnan Meng, Xianchun Gao, Yihan Zhao, Chen Wang, Xiaoliang Gao, Ahui Fan, Tianyu Cao, Daiming Fan, Xiaodi Zhao, and Yuanyuan Lu
- Subjects
chemoresistance ,gastric cancer ,hnRNPA2B1 ,NEAT1 ,stemness ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Chemoresistance is a major cause of treatment failure in gastric cancer (GC). Heterogeneous nuclear ribonucleoprotein A2B1 (hnRNPA2B1) is an N6‐methyladenosine (m6A)‐binding protein involved in a variety of cancers. However, whether m6A modification and hnRNPA2B1 play a role in GC chemoresistance is largely unknown. In this study, we aimed to investigate the role of hnRNPA2B1 and the downstream mechanism in GC chemoresistance. Methods The expression of hnRNPA2B1 among public datasets were analyzed and validated by quantitative PCR (qPCR), Western blotting, immunofluorescence, and immunohistochemical staining. The biological functions of hnRNPA2B1 in GC chemoresistance were investigated both in vitro and in vivo. RNA sequencing, methylated RNA immunoprecipitation, RNA immunoprecipitation, and RNA stability assay were performed to assess the association between hnRNPA2B1 and the binding RNA. The role of hnRNPA2B1 in maintenance of GC stemness was evaluated by bioinformatic analysis, qPCR, Western blotting, immunofluorescence, and sphere formation assays. The expression patterns of hnRNPA2B1 and downstream regulators in GC specimens from patients who received adjuvant chemotherapy were analyzed by RNAscope and multiplex immunohistochemistry. Results Elevated expression of hnRNPA2B1 was found in GC cells and tissues, especially in multidrug‐resistant (MDR) GC cell lines. The expression of hnRNPA2B1 was associated with poor outcomes of GC patients, especially in those who received 5‐fluorouracil treatment. Silencing hnRNPA2B1 effectively sensitized GC cells to chemotherapy by inhibiting cell proliferation and inducing apoptosis both in vitro and in vivo. Mechanically, hnRNPA2B1 interacted with and stabilized long noncoding RNA NEAT1 in an m6A‐dependent manner. Furthermore, hnRNPA2B1 and NEAT1 worked together to enhance the stemness properties of GC cells via Wnt/β‐catenin signaling pathway. In clinical specimens from GC patients subjected to chemotherapy, the expression levels of hnRNPA2B1, NEAT1, CD133, and CD44 were markedly elevated in non‐responders compared with responders. Conclusion Our findings indicated that hnRNPA2B1 interacts with and stabilizes lncRNA NEAT1, which contribute to the maintenance of stemness property via Wnt/β‐catenin pathway and exacerbate chemoresistance in GC.
- Published
- 2024
- Full Text
- View/download PDF
36. NEAT1 inhibits the angiogenic activity of cerebral arterial endothelial cells by inducing the M1 polarization of microglia through the AMPK signaling pathway
- Author
-
Ting Chen, Xin Huang, Yi-Xuan Zhao, Zhi-wen Zhou, and Wen-sheng Zhou
- Subjects
NEAT1 ,Microglia ,Cerebrovascular endothelial cells ,Angiogenesis ,AMPK ,Cytology ,QH573-671 - Abstract
Abstract Background Enhancing angiogenesis may be an effective strategy to promote functional recovery after ischemic stroke. Inflammation regulates angiogenesis. Microglia are crucial cells that initiate inflammatory responses after various brain injuries. Long noncoding RNA nuclear paraspeckle assembly transcript 1 (NEAT1) plays a role in regulating brain injury. This study aimed to explore the effects of NEAT1-regulated microglial polarization on the neovascularization capacity of cerebrovascular endothelial cells and the underlying molecular regulatory mechanisms. Methods Mouse cerebral arterial endothelial cells (mCAECs) were co-cultured with BV-2 cells in different groups using a Transwell system. NEAT1 expression levels were measured by fluorescence quantitative reverse transcription PCR. Levels of IL-1β, IL-6, TNF-α, Arg-1, IL-4, and IL-10 were determined using ELISA. Expression levels of CD86 and CD163 were detected by immunofluorescence. The neovascularization capacity of mCAECs was assessed using CCK-8, Transwell, Transwell-matrigel, and tube formation assays. Label-free quantification proteomics was carried out to identify differentially expressed proteins. Protein levels were measured by Western blotting. Results NEAT1 overexpression induced M1 polarization in BV-2 cells, whereas NEAT1 knockdown blocked lipopolysaccharide-induced M1 polarization in microglia. NEAT1-overexpressing BV-2 cells suppressed the angiogenic ability of mCAECs, and NEAT1-knocking BV-2 cells promoted the angiogenic ability of mCAECs under lipopolysaccharide treatment. Label-free quantitative proteomic analysis identified 144 upregulated and 131 downregulated proteins that were induced by NEAT1 overexpression. The AMP-activated protein kinase (AMPK) signaling pathway was enriched in the Kyoto Encyclopedia of Genes and Genomes analysis of the differentially expressed proteins. Further verification showed that NEAT1 inactivated the AMPK signaling pathway. Moreover, the AMPK activator 5-aminoimidazole-4-carboxamide ribonucleotide reversed the effect of NEAT1 on BV-2 polarization and the regulatory effect of NEAT1-overexpressing BV-2 cells on the angiogenic ability of mCAECs. Conclusions NEAT1 inhibits the angiogenic activity of mCAECs by inducing M1 polarization of BV-2 cells through the AMPK signaling pathway. This study further clarified the impact and mechanism of NEAT1 on microglia and the angiogenic ability of cerebrovascular endothelial cells.
- Published
- 2024
- Full Text
- View/download PDF
37. Role of ATF3 triggering M2 macrophage polarization to protect against the inflammatory injury of sepsis through ILF3/NEAT1 axis
- Author
-
Wang, Wei, Xu, Rongli, He, Ping, Xiong, Yuqing, Zhao, Haomiao, Fu, Xuewei, Lin, Jie, and Ye, Lijiao
- Published
- 2024
- Full Text
- View/download PDF
38. Knockdown of NEAT1 prevents post-stroke lipid droplet agglomeration in microglia by regulating autophagy
- Author
-
Pan, Yongli, Xin, Wenqiang, Wei, Wei, Tatenhorst, Lars, Graf, Irina, Popa-Wagner, Aurel, Gerner, Stefan T., Huber, Sabine E., Kilic, Ertugrul, Hermann, Dirk M., Bähr, Mathias, Huttner, Hagen B., and Doeppner, Thorsten R.
- Published
- 2024
- Full Text
- View/download PDF
39. PiRNA hsa_piR_019949 promotes chondrocyte anabolic metabolism by inhibiting the expression of lncRNA NEAT1
- Author
-
Zhang, Xinhai, Wang, Xuyi, Yu, Fengbin, Wang, Chenglong, Peng, Jianping, Wang, Chuandong, and Chen, Xiaodong
- Published
- 2024
- Full Text
- View/download PDF
40. NEAT1 repression by MED12 creates chemosensitivity in p53 wild‐type breast cancer cells.
- Author
-
Zhang, Shengjie, Kim, Eui‐Jun, Huang, Junfeng, Liu, Peng, Donahue, Kristine, Wang, Qinchuan, Wang, Yidan, Mcilwain, Sean, Xie, Ling, Chen, Xian, Li, Lingjun, and Xu, Wei
- Subjects
- *
BREAST cancer , *CANCER cells , *P53 antioncogene , *TUMOR antigens , *LINCRNA , *PROTEIN arginine methyltransferases - Abstract
Breast cancer is often treated with chemotherapy. However, the development of chemoresistance results in treatment failure. Long non‐coding RNA nuclear paraspeckle assembly transcript 1 (NEAT1) has been shown to contribute to chemoresistance in breast cancer cells. In studying the transcriptional regulation of NEAT1 using multi‐omics approaches, we showed that NEAT1 is up‐regulated by 5‐fluorouracil in breast cancer cells with wild‐type cellular tumor antigen p53 but not in mutant‐p53‐expressing breast cancer cells. The regulation of NEAT1 involves mediator complex subunit 12 (MED12)‐mediated repression of histone acetylation marks at the promoter region of NEAT1. Knockdown of MED12 but not coactivator‐associated arginine methyltransferase 1 (CARM1) induced histone acetylation at the NEAT1 promoter, leading to elevated NEAT1 mRNAs, resulting in a chemoresistant phenotype. The MED12‐dependent regulation of NEAT1 differs between wild‐type and mutant p53‐expressing cells. MED12 depletion led to increased expression of NEAT1 in a wild‐type p53 cell line, but decreased expression in a mutant p53 cell line. Chemoresistance caused by MED12 depletion can be partially rescued by NEAT1 knockdown in p53 wild‐type cells. Collectively, our study reveals a novel mechanism of chemoresistance dependent on MED12 transcriptional regulation of NEAT1 in p53 wild‐type breast cancer cells. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
41. Long non‐coding RNA NEAT1 promotes aerobic glycolysis and progression of cervical cancer through WNT/β‐catenin/PDK1 axis.
- Author
-
Su, Min, Liang, Ziyan, Shan, Shidong, Gao, Yang, He, Li, Liu, Xuelian, Wang, Anjin, Wang, Hua, and Cai, Hongbing
- Subjects
- *
LINCRNA , *CERVICAL cancer , *PYRUVATE dehydrogenase kinase , *GLYCOLYSIS , *CANCER invasiveness , *LACTATES - Abstract
Background: Cervical cancer is one of the most common gynecological cancers. Accumulated evidence shows that long non‐coding RNAs (lncRNAs) play essential roles in cervical cancer occurrence and progression, but their specific functions and mechanisms remain to be further explored. Methods: The RT‐qPCR assay was used to detect the expression of NEAT1 in cervical cancer tissues and cell lines. CCK‐8, colony formation, flow cytometry, western blotting, and Transwell assays were used to evaluate the impact of NEAT1 on the malignant behavior of cervical cancer cells. Glucose consumption, lactate production, ATP levels, ROS levels, MMP levels, and the mRNA expressions of glycolysis‐related genes and tricarboxylic acid cycle‐related genes were detected to analyze the effect of NEAT1 on metabolism reprograming in cervical cancer cells. The expressions of PDK1, β‐catenin and downstream molecules of the WNT/β‐catenin signaling pathway in cervical cancer cells and tissues were detected by western blotting, RT‐qPCR, immunofluorescence and immunohistochemistry assays. Results: This study investigated the role and possible molecular mechanism of lncRNA nuclear paraspeckle assembly transcript 1 (NEAT1) in cervical cancer. Our results showed that NEAT1 was highly expressed in cervical cancer tissues and cell lines. Downregulation of NEAT1 inhibited the proliferation, migration, invasion and glycolysis of cervical cancer cells, while overexpression of NEAT1 led to the opposite effects. Mechanistically, NEAT1 upregulated pyruvate dehydrogenase kinase (PDK1) through the WNT/β‐catenin signaling pathway, which enhanced glycolysis and then facilitated cervical cancer metastasis. Furthermore, NEAT1 maintained the protein stability of β‐catenin but did not affect its mRNA level. We also excluded the direct binding of NEAT1 to the β‐catenin protein via RNA pull‐down assay. The suppressive impact of NEAT1 knockdown on cell proliferation, invasion, and migration was rescued by β‐catenin overexpression. The WNT inhibitor iCRT3 attenuated the carcinogenic effect induced by NEAT1 overexpression. Conclusion: In summary, these findings indicated that NEAT1 may contribute to the progression of cervical cancer by activating the WNT/β‐catenin/PDK1 signaling axis. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
42. lncRNA Biomarkers of Glioblastoma Multiforme †.
- Author
-
Pokorná, Markéta, Černá, Marie, Boussios, Stergios, Ovsepian, Saak V., and O'Leary, Valerie Bríd
- Subjects
GLIOBLASTOMA multiforme ,LINCRNA ,BIOMARKERS ,NERVE tissue ,NON-coding RNA - Abstract
Long noncoding RNAs (lncRNAs) are RNA molecules of 200 nucleotides or more in length that are not translated into proteins. Their expression is tissue-specific, with the vast majority involved in the regulation of cellular processes and functions. Many human diseases, including cancer, have been shown to be associated with deregulated lncRNAs, rendering them potential therapeutic targets and biomarkers for differential diagnosis. The expression of lncRNAs in the nervous system varies in different cell types, implicated in mechanisms of neurons and glia, with effects on the development and functioning of the brain. Reports have also shown a link between changes in lncRNA molecules and the etiopathogenesis of brain neoplasia, including glioblastoma multiforme (GBM). GBM is an aggressive variant of brain cancer with an unfavourable prognosis and a median survival of 14–16 months. It is considered a brain-specific disease with the highly invasive malignant cells spreading throughout the neural tissue, impeding the complete resection, and leading to post-surgery recurrences, which are the prime cause of mortality. The early diagnosis of GBM could improve the treatment and extend survival, with the lncRNA profiling of biological fluids promising the detection of neoplastic changes at their initial stages and more effective therapeutic interventions. This review presents a systematic overview of GBM-associated deregulation of lncRNAs with a focus on lncRNA fingerprints in patients' blood. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
43. NEAT1 inhibits the angiogenic activity of cerebral arterial endothelial cells by inducing the M1 polarization of microglia through the AMPK signaling pathway.
- Author
-
Chen, Ting, Huang, Xin, Zhao, Yi-Xuan, Zhou, Zhi-wen, and Zhou, Wen-sheng
- Abstract
Background: Enhancing angiogenesis may be an effective strategy to promote functional recovery after ischemic stroke. Inflammation regulates angiogenesis. Microglia are crucial cells that initiate inflammatory responses after various brain injuries. Long noncoding RNA nuclear paraspeckle assembly transcript 1 (NEAT1) plays a role in regulating brain injury. This study aimed to explore the effects of NEAT1-regulated microglial polarization on the neovascularization capacity of cerebrovascular endothelial cells and the underlying molecular regulatory mechanisms. Methods: Mouse cerebral arterial endothelial cells (mCAECs) were co-cultured with BV-2 cells in different groups using a Transwell system. NEAT1 expression levels were measured by fluorescence quantitative reverse transcription PCR. Levels of IL-1β, IL-6, TNF-α, Arg-1, IL-4, and IL-10 were determined using ELISA. Expression levels of CD86 and CD163 were detected by immunofluorescence. The neovascularization capacity of mCAECs was assessed using CCK-8, Transwell, Transwell-matrigel, and tube formation assays. Label-free quantification proteomics was carried out to identify differentially expressed proteins. Protein levels were measured by Western blotting. Results: NEAT1 overexpression induced M1 polarization in BV-2 cells, whereas NEAT1 knockdown blocked lipopolysaccharide-induced M1 polarization in microglia. NEAT1-overexpressing BV-2 cells suppressed the angiogenic ability of mCAECs, and NEAT1-knocking BV-2 cells promoted the angiogenic ability of mCAECs under lipopolysaccharide treatment. Label-free quantitative proteomic analysis identified 144 upregulated and 131 downregulated proteins that were induced by NEAT1 overexpression. The AMP-activated protein kinase (AMPK) signaling pathway was enriched in the Kyoto Encyclopedia of Genes and Genomes analysis of the differentially expressed proteins. Further verification showed that NEAT1 inactivated the AMPK signaling pathway. Moreover, the AMPK activator 5-aminoimidazole-4-carboxamide ribonucleotide reversed the effect of NEAT1 on BV-2 polarization and the regulatory effect of NEAT1-overexpressing BV-2 cells on the angiogenic ability of mCAECs. Conclusions: NEAT1 inhibits the angiogenic activity of mCAECs by inducing M1 polarization of BV-2 cells through the AMPK signaling pathway. This study further clarified the impact and mechanism of NEAT1 on microglia and the angiogenic ability of cerebrovascular endothelial cells. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
44. Elucidating the Role of a Shared lncRNA-miRNA-mRNA Network in Exacerbating Parkinson's Disease Symptoms in the Context of COVID-19 Infection.
- Author
-
Yousefi, Maryam, Hashemi, Motahare-Sadat, Peymani, Maryam, Ghaedi, Kamran, Irani, Shiva, and Etemadifar, Masoud
- Subjects
COMPETITIVE endogenous RNA ,PARKINSON'S disease ,SYMPTOMS ,COVID-19 ,GENE expression ,DEEP brain stimulation ,LINCRNA ,POLYMER networks - Abstract
Background and objectives: Parkinson's disease (PD) is a common neurodegenerative disorder with unclear molecular mechanisms. Noncoding RNAs, such as microRNAs (miRNAs) and long noncoding RNAs (lncRNAs), have been identified as critical regulators of gene expression. This study aimed to investigate the triple network of lncRNA-miRNA-mRNA, known as competing endogenous RNAs (ceRNAs), and to identify essential lncRNAs that regulate PD-related gene expression through their target miRNAs. The study also identified a common triple network between COVID-19 and PD that may contribute to exacerbating PD symptoms. Methods: A bioinformatics approach was employed to construct a PD ceRNA network using common PD genes, miRNAs and lncRNAs with the highest interaction with their targets. Also, a PD-COVID-19 triple network was constructed by integrating PD network nodes into the COVID-19 network. Results: The PD ceRNA network comprised 34 nodes, including 12 lncRNAs, 16 miRNAs with interconnections and six mRNAs, some of which were related to COVID-19. The network showed parallel expression of the SNCA and PARK7 genes as well as the NEAT1 and MALAT1 lncRNAs in both PD and COVID-19. Conclusions: This study provide insights into the molecular mechanisms underlying the worsening of symptoms in PD patients with COVID-19. The PD and COVID-19 ceRNA network indicates that coronavirus could worsen PD symptoms by altering the expression of some genes related to PD. Therefore, COVID-19 could dysregulate the common RNAs involved in PD through lncRNAs, miRNAs. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
45. GOLM1 Promotes Pulmonary Fibrosis through Upregulation of NEAT1.
- Author
-
Wang, Yani, Hu, Danjing, Wan, Linyan, Yang, Shuhui, Liu, Song, Wang, Zixi, Li, Jie, Li, Jia, Zheng, Zhoude, Cheng, Chongsheng, Wang, Yanan, Wang, Hanghang, Tian, Xinlun, Chen, Wenhui, Li, Shanqing, Zhang, Ji, Zha, Xiaojun, Chen, Jingyu, Zhang, Hongbing, and Xu, Kai-Feng
- Subjects
PULMONARY fibrosis ,IDIOPATHIC pulmonary fibrosis ,MEMBRANE proteins ,GENE expression ,EXTRACELLULAR matrix - Abstract
Idiopathic pulmonary fibrosis (IPF) is a lethal progressive disease with elusive molecular mechanisms and limited therapeutic options. Aberrant activation of fibroblasts is a central hallmark of lung fibrosis. Here, we report that Golgi membrane protein 1 (GOLM1, also known as GP73 or GOLPH2) was increased in the lungs of patients with pulmonary fibrosis and mice with bleomycin (BLM)–induced pulmonary fibrosis. Loss of GOLM1 inhibited proliferation, differentiation, and extracellular matrix deposition of fibroblasts, whereas overexpression of GOLM1 exerted the opposite effects. Similarly, worsening pulmonary fibrosis after BLM treatment was observed in GOLM1–knock-in mice, whereas BLM-treated Golm1-knockout mice exhibited alleviated pulmonary fibrosis and collagen deposition. Furthermore, we identified long noncoding RNA NEAT1 downstream of GOLM1 as a potential mediator of pulmonary fibrosis through increased GOLM1 expression. Depletion of NEAT1 inhibited fibroblast proliferation and extracellular matrix production and reversed the profibrotic effects of GOLM1 overexpression. Additionally, we identified KLF4 as a downstream mediator of GOLM1 signaling to NEAT1. Our findings suggest that GOLM1 plays a pivotal role in promoting pulmonary fibrosis through the GOLM1–KLF4–NEAT1 signaling axis. Targeting GOLM1 and its downstream pathways may represent a novel therapeutic strategy for treating pulmonary fibrosis. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
46. Correlation of lncRNA NEAT1 and miRNA-182-5p with the Risk of Liver Fibrosis in Type 2 Diabetes Mellitus Patients with MAFLD
- Author
-
HE Jia, LI Yongping, WEI Feng, LIU Meilan, WU Yaling, SHAO Longge
- Subjects
type 2 diabetes mellitus ,metabolic-related fatty liver disease ,neat1 ,mirna-182-5p ,hepatic fibrosis ,root cause analysis ,Medicine - Abstract
Background With the incidence of chronic metabolic diseases rising by year, which has threatened the national health, the study of non-coding RNA and endocrine metabolism-related diseases has become a research hotspot at home and abroad, while lncRNA NEAT1 and miRNA-182-5p in type 2 diabetes mellitus (T2DM) combined with metabolic-related fatty liver disease (MAFLD) has been rarely reported. Objective To investigate the mechanism and clinical significance of lncRNA NEAT1 and miRNA-182-5p in the development of liver fibrosis in T2DM patients with MAFLD. Methods A total of 236 T2DM patients admitted to the endocrinology department of the First Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology from October 2021 to June 2022 were included as the study subjects, and 49 healthy people were included as the healthy control group. General information and laboratory test results of the subjects were collected. Visceral fat area (VFA) and subcutaneous fat area (SFA) were measured. Peripheral blood was collected and lncRNA NEAT1, miRNA-182-5p were determined. T2DM patients were divided into the T2DM with non-MAFLD group (n=82) and T2DM with MAFLD group (n=154). T2DM with MAFLD group was further divided into the low-risk subgroup (n=55), medium-risk subgroup (n=69) and high-risk subgroup (n=30) according to the liver fibrosis index (FIB-4). In addition, healthy people were selected as the healthy control group (n=49). Spearman rank correlation analysis was used to explore the correlation of lncRNA NEAT1 and miRNA-182-5p expression levels in the high-risk subgroup of liver fibrosis, and multilevel ordinal Logistic regression was used to explore the influencing factors of liver fibrosis risk in T2DM patients with MAFLD. Results Age, neck circumference (NC), fasting blood glucose (FPG) and glycosylated hemoglobin (HbA1c) in the healthy control group were lower than those in the T2DM with non-MAFLD and T2DM with MAFLD groups, the albumin (Alb) in the healthy control group was higher than that in the T2DM with non-MAFLD and T2DM with MAFLD groups (P
- Published
- 2024
- Full Text
- View/download PDF
47. Unraveling NEAT1's complex role in lung cancer biology
- Author
-
Md Sadique Hussain, Obaid Afzal, Gaurav Gupta, Ahsas Goyal, Waleed Hassan Almalki, Imran Kazmi, Sami I. Alzarea, Abdulmalik Saleh Alfawaz Altamimi, Neelam Kukreti, Amlan Chakraborty, Sachin Kumar Singh, and Kamal Dua
- Subjects
long non-coding rna ,neat1 ,lung cancer ,clinical applications ,cancer ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 ,Biology (General) ,QH301-705.5 - Abstract
This review delves into the pivotal role of the long non-coding RNA NEAT1 in cancer biology, particularly in lung cancer (LC). It emphasizes NEAT1's unique subcellular localization and active involvement in gene regulation and chromatin remodeling. The review highlights NEAT1's impact on LC development and progression, including cell processes such as proliferation, migration, invasion, and resistance to therapy, positioning it as a potential diagnostic marker and therapeutic target. The complex web of NEAT1's regulatory interactions with proteins and microRNAs is explored, alongside challenges in targeting it therapeutically. The review concludes optimistically, suggesting future avenues for research and personalized LC therapies, shedding light on NEAT1's crucial role in LC.
- Published
- 2024
- Full Text
- View/download PDF
48. The long non-coding RNA NEAT1 promotes the progression of human ovarian cancer through targeting miR-214-3p and regulating angiogenesis
- Author
-
Yang Liu, Yan Li, Yanzhi Wu, Yiyue Zhao, Xi Hu, and Chunyi Sun
- Subjects
NEAT1 ,miR-214-3p ,Ovarian cancer ,Angiogenesis ,Metastasis ,Gynecology and obstetrics ,RG1-991 - Abstract
Abstract Background Angiogenesis and metastasis contributes substantially to the poor outcome of patients with ovarian cancer. We aimed to explore the role and mechanisms of the long non-coding RNA NEAT1 (nuclear enriched abundant transcript 1) in regulating angiogenesis and metastasis of human ovarian cancer. NEAT1 expression in human ovarian cancer tissues and cell lines including SKOV-3 and A2780 was investigated through in situ hybridization. Gene knockdown and overexpressing were achieved through lentivirus infection, transfection of plasmids or microRNA mimics. Cell viability was measured with the cell counting kit-8 assay, while apoptosis was determined by flow cytometry. Cell migration and invasion were evaluated by transwell experiments, and protein expression was determined by western blot assays or immunohistochemistry. Duo-luciferase reporter assay was employed to confirm the interaction between NEAT1 and target microRNA. In vivo tumor growth was evaluated in nude mice with xenografted SKOV-3/A2780 cells, and blood vessel formation in tumor was examined by histological staining. Results NEAT1 was highly expressed in ovarian cancer tissues of patients and cell lines. MiR-214-3p was identified as a sponging target of NEAT1, and they antagonizedeach other in a reciprocal manner. NEAT1-overexpressing SKOV-3 and A2780 cells had significantly increased proliferation, reduced apoptosis, and augmented abilities of migration and invasion, while cells with NEAT1-knockdown displayed markedly attenuated traits of malignancies. Additionally, the levels of NEAT1 appeared to be positively correlated with the expression levels of angiogenesis-related molecules, including Semaphorin 4D (Sema4D), Sema4D receptor Plexin B1, T-lymphoma invasion and metastasis-inducing protein-1 (Tiam1), and Rho-like GTPases Rac1/2/3. In the xenograft mouse model, more NEAT1 expression resulted in faster in vivo tumor growth, more blood vessel formation in tumor tissues, as well as higher expression levels of angiogenesis-related molecules and CD31. Conclusions NEAT1 promotes angiogenesis and metastasis in human ovarian cancer. NEAT1 and miR-214-3p are promising targets for developing therapeutics to treat human ovarian cancer.
- Published
- 2023
- Full Text
- View/download PDF
49. Genome-wide analyses identify NEAT1 as genetic modifier of age at onset of amyotrophic lateral sclerosis
- Author
-
Chunyu Li, Qianqian Wei, Yanbing Hou, Junyu Lin, Ruwei Ou, Lingyu Zhang, Qirui Jiang, Yi Xiao, Kuncheng Liu, Xueping Chen, TianMi Yang, Wei Song, Bi Zhao, Ying Wu, and Huifang Shang
- Subjects
Amyotrophic Lateral Sclerosis ,Age at onset ,GWAS ,Genetic modifiers ,NEAT1 ,Neurology. Diseases of the nervous system ,RC346-429 ,Geriatrics ,RC952-954.6 - Abstract
Abstract Background Patients with amyotrophic lateral sclerosis (ALS) demonstrate great heterogeneity in the age at onset (AAO), which is closely related to the course of disease. However, most genetic studies focused on the risk of ALS, while the genetic background underlying AAO of ALS is still unknown. Methods To identify genetic determinants influencing AAO of ALS, we performed genome-wide association analysis using a Cox proportional hazards model in 2,841 patients with ALS (Ndiscovery = 2,272, Nreplication = 569) in the Chinese population. We further conducted colocalization analysis using public cis-eQTL dataset, and Mendelian randomization analysis to identify risk factors for AAO of ALS. Finally, functional experiments including dual-luciferase reporter assay and RT-qPCR were performed to explore the regulatory effect of the target variant. Results The total heritability of AAO of ALS was ~ 0.24. One novel locus rs10128627 (FRMD8) was significantly associated with earlier AAO by ~ 3.15 years (P = 1.54E-08, beta = 0.31, SE = 0.05). This locus was cis-eQTL of NEAT1 in multiple brain tissues and blood. Colocalization analysis detected association signals at this locus between AAO of ALS and expression of NEAT1. Furthermore, functional exploration supported the variant rs10128627 was associated with upregulated expression of NEAT1 in cell models and patients with ALS. Causal inference suggested higher total cholesterol, low-density lipoprotein, and eosinophil were nominally associated with earlier AAO of ALS, while monocyte might delay the AAO. Conclusions Collective evidence from genetic, bioinformatic, and functional results suggested NEAT1 as a key player in the disease progression of ALS. These findings improve the current understanding of the genetic role in AAO of ALS, and provide a novel target for further research on the pathogenesis and therapeutic options to delay the disease onset.
- Published
- 2023
- Full Text
- View/download PDF
50. Downregulation of lncRNA NEAT1 interacts with miR-374b-5p/PGAP1 axis to aggravate the development of osteoarthritis
- Author
-
Feiri Huang, Zhongliang Su, Jie Yang, Xizhen Zhao, and Yaozeng Xu
- Subjects
Osteoarthritis ,NEAT1 ,miR374b-5p ,PGAP1 ,Inflammation ,Apoptosis ,Orthopedic surgery ,RD701-811 ,Diseases of the musculoskeletal system ,RC925-935 - Abstract
Abstract Background Osteoarthritis (OA), characterized by inflammation and articular cartilage degradation, is a prevalent arthritis among geriatric population. This paper was to scrutinize the novel mechanism of long noncoding RNA (lncRNA) NEAT1 in OA etiology. Methods A total of 10 OA patients and 10 normal individuals was included in this study. Cell model of OA was built in human normal chondrocytes induced by lipopolysaccharide (LPS). An OA Wistar rat model was established through intra-articular injection of L-cysteine and papain mixtures (proportion at 1:2) into the right knee. Quantitative reverse transcription-polymerase chain reaction was employed to ascertain the expression levels of NEAT1, microRNA (miR)-374b-5p and post-GPI attachment to protein 1 (PGAP1), while dual-luciferase reporter experiments were used for the validation of target relationship among them. Cell cycle and apoptosis were calculated by flow cytometry analysis. CCK-8 assay was done to evaluate the proliferative potentials of chondrocytes. The levels of cell cycle-related proteins (Cyclin A1, Cyclin B1 and Cyclin D2) and pro-apoptotic proteins (Caspase3 and Caspase9) were measured by western blotting. Tumor necrosis factor-alpha (TNF-α), interleukin-1beta (IL-1β) and IL-6 levels were determined via ELISA. Hematoxylin & eosin (HE) Staining was used for pathological examination in OA rats. Results Pronounced downregulation of NEAT1 and PGAP1 and high amounts of miR-374b-5p were identified in OA patients, LPS-induced chondrocytes and OA rats. NEAT1 targeted miR-374b-5p to control PGAP1 expression. Loss of NEAT1 or upregulation of miR-374b-5p dramatically accelerated apoptosis, led to the G1/S arrest and promoted the secretion of inflammatory cytokines in LPS-induced chondrocytes, while ectopic expression of PGAP1 exhibited the opposite influences on chondrocytes. Additionally, we further indicated that upregulation of miR-374b-5p attenuated the effects of PGAP1 overexpression on LPS-induced chondrocytes. Conclusions Reduced NEAT1 induces the development of OA via miR-374b-5p/PGAP1 pathway. This suggests that the regulatory axis NEAT1/miR-374b-5p/PGAP1 is a novel and prospective target for OA treatment.
- Published
- 2023
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.