Journal articles on the topic 'Bioinformatics, RNA-Seq, lncRNA'

To see the other types of publications on this topic, follow the link: Bioinformatics, RNA-Seq, lncRNA.

Create a spot-on reference in APA, MLA, Chicago, Harvard, and other styles

Select a source type:

Consult the top 50 journal articles for your research on the topic 'Bioinformatics, RNA-Seq, lncRNA.'

Next to every source in the list of references, there is an 'Add to bibliography' button. Press on it, and we will generate automatically the bibliographic reference to the chosen work in the citation style you need: APA, MLA, Harvard, Chicago, Vancouver, etc.

You can also download the full text of the academic publication as pdf and read online its abstract whenever available in the metadata.

Browse journal articles on a wide variety of disciplines and organise your bibliography correctly.

1

Han, Zhijie, Weiwei Xue, Lin Tao, Yan Lou, Yunqing Qiu, and Feng Zhu. "Genome-wide identification and analysis of the eQTL lncRNAs in multiple sclerosis based on RNA-seq data." Briefings in Bioinformatics 21, no. 3 (April 24, 2019): 1023–37. http://dx.doi.org/10.1093/bib/bbz036.

Full text
Abstract:
Abstract The pathogenesis of multiple sclerosis (MS) is significantly regulated by long noncoding RNAs (lncRNAs), the expression of which is substantially influenced by a number of MS-associated risk single nucleotide polymorphisms (SNPs). It is thus hypothesized that the dysregulation of lncRNA induced by genomic variants may be one of the key molecular mechanisms for the pathology of MS. However, due to the lack of sufficient data on lncRNA expression and SNP genotypes of the same MS patients, such molecular mechanisms underlying the pathology of MS remain elusive. In this study, a bioinformatics strategy was applied to obtain lncRNA expression and SNP genotype data simultaneously from 142 samples (51 MS patients and 91 controls) based on RNA-seq data, and an expression quantitative trait loci (eQTL) analysis was conducted. In total, 2383 differentially expressed lncRNAs were identified as specifically expressing in brain-related tissues, and 517 of them were affected by SNPs. Then, the functional characterization, secondary structure changes and tissue and disease specificity of the cis-eQTL SNPs and lncRNA were assessed. The cis-eQTL SNPs were substantially and specifically enriched in neurological disease and intergenic region, and the secondary structure was altered in 17.6% of all lncRNAs in MS. Finally, the weighted gene coexpression network and gene set enrichment analyses were used to investigate how the influence of SNPs on lncRNAs contributed to the pathogenesis of MS. As a result, the regulation of lncRNAs by SNPs was found to mainly influence the antigen processing/presentation and mitogen-activated protein kinases (MAPK) signaling pathway in MS. These results revealed the effectiveness of the strategy proposed in this study and give insight into the mechanism (SNP-mediated modulation of lncRNAs) underlying the pathology of MS.
APA, Harvard, Vancouver, ISO, and other styles
2

Hao, Qing, Lei Yang, Dingyu Fan, Bin Zeng, and Juan Jin. "The transcriptomic response to heat stress of a jujube (Ziziphus jujuba Mill.) cultivar is featured with changed expression of long noncoding RNAs." PLOS ONE 16, no. 5 (May 27, 2021): e0249663. http://dx.doi.org/10.1371/journal.pone.0249663.

Full text
Abstract:
Long non-coding RNA (lncRNA) of plant species undergoes dynamic regulation and acts in developmental and stress regulation. Presently, there is little information regarding the identification of lncRNAs in jujube (Ziziphus jujuba Mill.), and it is uncertain whether the lncRNAs could respond to heat stress (HS) or not. In our previous study, a cultivar (Hqing1-HR) of Z. jujuba were treated by HS (45°C) for 0, 1, 3, 5 and 7 days, and it was found that HS globally changed the gene expression by RNA sequencing (RNA-seq) experiments and informatics analyses. In the current study, 8260 lncRNAs were identified successfully from the previous RNA-seq data, and it indicated that lncRNAs expression was also altered globally, suggesting that the lncRNAs might play vital roles in response to HS. Furthermore, bioinformatics analyses of potential target mRNAs of lncRNAs with cis-acting mechanism were performed, and it showed that multiple differentially expressed (DE) mRNAs co-located with DElncRNAs were highly enriched in pathways associated with response to stress and regulation of metabolic process. Taken together, these findings not only provide a comprehensive identification of lncRNAs but also useful clues for molecular mechanism response to HS in jujube.
APA, Harvard, Vancouver, ISO, and other styles
3

Zheng, Yuan Y., Sheng D. Sheng, Tai Y. Hui, Chang Yue, Jia M. Sun, Dan Guo, Su L. Guo, et al. "An Integrated Analysis of Cashmere Fineness lncRNAs in Cashmere Goats." Genes 10, no. 4 (April 2, 2019): 266. http://dx.doi.org/10.3390/genes10040266.

Full text
Abstract:
Animal growth and development are regulated by long non-coding RNAs (lncRNAs). However, the functions of lncRNAs in regulating cashmere fineness are poorly understood. To identify the key lncRNAs that are related to cashmere fineness in skin, we have collected skin samples of Liaoning cashmere goats (LCG) and Inner Mongolia cashmere goats (MCG) in the anagen phase, and have performed RNA sequencing (RNA-seq) approach on these samples. The high-throughput sequencing and bioinformatics analyses identified 437 novel lncRNAs, including 93 differentially expressed lncRNAs. We also identified 3,084 differentially expressed messenger RNAs (mRNAs) out of 27,947 mRNAs. Gene ontology (GO) analyses of lncRNAs and target genes in cis show a predominant enrichment of targets that are related to intermediate filament and intermediate filament cytoskeleton. According to the Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis, sphingolipid metabolism is a significant pathway for lncRNA targets. In addition, this is the first report to reveal the possible lncRNA–mRNA regulatory network for cashmere fineness in cashmere goats. We also found that lncRNA XLOC_008679 and its target gene, KRT35, may be related to cashmere fineness in the anagen phase. The characterization and expression analyses of lncRNAs will facilitate future studies on the potential value of fiber development in LCG.
APA, Harvard, Vancouver, ISO, and other styles
4

Jin, Qiao, Qian Gong, Xuan Le, Jin He, and Lenan Zhuang. "Bioinformatics and Experimental Analyses Reveal Immune-Related LncRNA–mRNA Pair AC011483.1-CCR7 as a Biomarker and Therapeutic Target for Ischemic Cardiomyopathy." International Journal of Molecular Sciences 23, no. 19 (October 9, 2022): 11994. http://dx.doi.org/10.3390/ijms231911994.

Full text
Abstract:
Ischemic cardiomyopathy (ICM), which increases along with aging, is the leading cause of heart failure. Currently, immune response is believed to be critical in ICM whereas the roles of immune-related lncRNAs remain vague. In this study, we aimed to systematically analyze immune-related lncRNAs in the aging-related disease ICM. Here, we downloaded publicly available RNA-seq data from ischemic cardiomyopathy patients and non-failing controls (GSE116250). Weighted gene co-expression network analysis (WGCNA) was performed to identify key ICM-related modules. The immune-related lncRNAs of key modules were screened by co-expression analysis of immune-related mRNAs. Then, a competing endogenous RNA (ceRNA) network, including 5 lncRNAs and 13 mRNAs, was constructed using lncRNA–mRNA pairs which share regulatory miRNAs and have significant correlation. Among the lncRNA–mRNA pairs, one pair (AC011483.1-CCR7) was verified in another publicly available ICM dataset (GSE46224) and ischemic cell model. Further, the immune cell infiltration analysis of the GSE116250 dataset revealed that the proportions of monocytes and CD8+ T cells were negatively correlated with the expression of AC011483.1-CCR7, while plasma cells were positively correlated, indicating that AC011483.1-CCR7 may participate in the occurrence and development of ICM through immune cell infiltration. Together, our findings revealed that lncRNA–mRNA pair AC011483.1-CCR7 may be a novel biomarker and therapeutic target for ICM.
APA, Harvard, Vancouver, ISO, and other styles
5

Liu, Fen, Yang Yang, Tong Liu, Jun Deng, Heng Zhang, Dan Luo, and Yuan-Lei Lou. "Analysis of Differentially Expressed Long Noncoding RNA in Renal Ischemia-Reperfusion Injury." Kidney and Blood Pressure Research 45, no. 5 (2020): 686–701. http://dx.doi.org/10.1159/000508217.

Full text
Abstract:
Background: Renal ischemia-reperfusion (IR) injury is one of the major causes of acute renal failure which seriously endangers the health and life of patients. Currently, there is still lack of comprehensive knowledge of the molecular mechanism of renal IR injury, and the regulatory role of long noncoding RNA (lncRNA) in renal IR damage remains poorly understood. Aim: The aim of this study was to analyze the expression spectrum of lncRNA in renal IR damage in mice and to explore specific lncRNA that may be involved in regulating the development of human renal IR injury. Methods: RNA-Seq was used to investigate the lncRNA profile of renal IR injury in a mouse model, and conservation analysis was performed on mouse lncRNAs with differential expression (fragments per kilobase of transcript per million mapped reads ≥2) by BLASTN. The potential functions and associated pathways of the differentially expressed lncRNA were explored by bioinformatics analysis. The cell hypoxia model was used to detect the expression of the candidate lncRNA. Results: Of the 45,923 lncRNA transcripts detected in the samples, and 5,868 lncRNAs were found to be significantly differentially expressed (p < 0.05 and fold change ≥ 2) in 24-h IR kidney tissue compared to the expression in the control group. It was found that 56 differently expressed mouse lncRNA transcripts have human homology by analyzing the conserved sequences. We also found that lncRNA-NONHSAT183385.1 expression significantly increased in HK2 cells after 24 h of hypoxia and increased further 6 h after reoxygenation, and after 24 h of reoxygenation it was dramatically downregulated, indicating that NONHSAT183385.1 may be involved in the pathophysiological process of renal tubular epithelial cells in response to ischemia in human renal IR. Conclusion: Our study revealed differentially expressed lncRNAs in renal IR damage in mice and identified a set of conserved lncRNAs, which would help to explore lncRNAs that may play important regulatory roles in human renal IR injury.
APA, Harvard, Vancouver, ISO, and other styles
6

Joachims, Michelle L., Bhuwan Khatri, Chuang Li, Kandice L. Tessneer, John A. Ice, Anna M. Stolarczyk, Nicolas Means, et al. "Dysregulated long non-coding RNA in Sjögren’s disease impacts both interferon and adaptive immune responses." RMD Open 8, no. 2 (November 2022): e002672. http://dx.doi.org/10.1136/rmdopen-2022-002672.

Full text
Abstract:
ObjectiveSjögren’s disease (SjD) is an autoimmune disease characterised by inflammatory destruction of exocrine glands. Patients with autoantibodies to Ro/SSA (SjDRo+) exhibit more severe disease. Long non-coding RNAs (lncRNAs) are a functionally diverse class of non-protein-coding RNAs whose role in autoimmune disease pathology has not been well characterised.MethodsWhole blood RNA-sequencing (RNA-seq) was performed on SjD cases (n=23 Ro/SSA negative (SjDRo−); n=27 Ro/SSA positive (SjDRo+) and healthy controls (HCs; n=27). Bioinformatics and pathway analyses of differentially expressed (DE) transcripts (log2fold change ≥2 or ≤0.5; padj<0.05) were used to predict lncRNA function.LINC01871was characterised by RNA-seq analyses of HSB-2 cells with CRISPR-targetedLINC01871deletion (LINC01871−/−) and in vitro stimulation assays.ResultsWhole blood RNA-seq revealed autoantibody-specific transcription profiles and disproportionate downregulation of DE transcripts in SjD cases relative to HCs. Sixteen DE lncRNAs exhibited correlated expression with the interferon (IFN)-regulated gene,RSAD2, in SjDRo+(r≥0.65 or ≤−0.6); four antisense lncRNAs exhibited IFN-regulated expression in immune cell lines.LINC01871was upregulated in all SjD cases. RNA-seq and pathway analyses ofLINC01871−/−cells implicated roles in cytotoxic function, differentiation and IFNγ induction.LINC01871was induced by IFNγ in a myeloid cell line and regulated by calcineurin/NFAT pathway and T cell receptor (TCR) signalling in primary human T cells.ConclusionLINC01871influences expression of many immune cell genes and growth factors, is IFNγ inducible, and regulated by calcineurin signalling and TCR ligand engagement. AlteredLINC01871expression may influence the dysregulated T cell inflammatory pathways implicated in SjD.
APA, Harvard, Vancouver, ISO, and other styles
7

Judy, Jen, Xunde Wang, Fayaz Seifuddin, Laxminath Tumburu, Mehdi Pirooznia, and Swee Lay Thein. "RNA Seq Profiles and Bioinformatics Validation in a Large Sample of Sickle Cell Disease Patients." Blood 136, Supplement 1 (November 5, 2020): 13–14. http://dx.doi.org/10.1182/blood-2020-139382.

Full text
Abstract:
Background Sickle Cell Disease (SCD) is a genetic disorder caused by a single amino acid substitution in the ß-hemoglobin chain. Clinical manifestations of SCD are multisystemic and heterogeneous, with a wide range of organ damage between patients despite identical genetic mutations. The sub-phenotypes of SCD reflect damage to different organs arising from the pathophysiology of the disease. Biomarkers, such as N-terminal-pro-Brain natriuretic peptide (NT-proBNP), tricuspid regurgitant velocity (TRV), and hemolytic parameters provide readouts of the degree of different organ damage and are known risk markers. Dissecting its pathophysiology along known biological pathways has led to development of therapeutic targets, but we still do not fully understand the pathogenesis for much of the organ damage. Given the complexity of the pathophysiology of SCD that is likely to involve multiple overlapping and interacting biological pathways, an agnostic, network-based approach using gene expression data with annotated risk markers provides an attractive alternative. We assessed the plausibility of using such an approach with bilirubin levels as a phenotype. Bilirubin is a breakdown product of red blood cells (RBCs) that is measured in routine labs and a validated hemolytic and disease severity marker of SCD. Methods Using RNA-Seq data from 224 patients with SCD (80% HbSS and HbSBeta0 thalassemia combined, 15% HbSC, 5% HbSBeta+ thalassemia, and 0.4% HbSD), we performed a differential gene expression (DGE) analysis and weighted gene co-expression network analysis (WGCNA) of 12,450 genes. For the DGE analysis we used total bilirubin levels as a continuous variable for the main phenotype of interest, adjusting for age, sex, and hemoglobin genotype. We then conducted WGCNA to identify modules of genes with similar coexpression patterns, which we functionally annotated (via the ClusterProfiler R package) for interesting biological themes. Results Of the 12,450 genes, 10,604 were protein coding, and 1,560 were lncRNAs, and 286 were other non-coding transcripts. 3,509 (2,975 coding, 447 lncRNA, and 87 other non-coding) were considered differentially expressed (unadjusted p-value &lt; 0.05) according to bilirubin level. Through the WCGNA, the genes clustered into 11 co-expression modules, representing a range of biological pathways. Four of these modules had representative eigengenes that were significantly correlated with bilirubin (p &lt; 0.05), and we focused on 2 of these that had biologically interesting functions. The first module of interest (turquoise) consisted of 3,636 genes (3,371 protein-coding and 210 lncRNA) (25% differentially expressed, and 62% upregulated). Genes in this module have been associated with cellular functioning of younger RBCs, validating the presence of the significant Gene Ontology (GO) biological pathways listed in the bar graph (mitochondria gene expression, ribonucleotide processes, etc.), and aligning with expected processes as the young RBCs replace the degraded RBCs that had caused the high bilirubin. Notably, this module contained the biliverdin reductase A (BLVRA) gene (differentially expressed at p &lt; 0.008), which reduces biliverdin to bilirubin. The other highlighted module (red) consisted of 914 genes (846 protein-coding and 57 lncRNA) (42% differentially expressed, and 97% upregulated). Multiple pathways in the red module were related to catabolic processes. As previously stated, bilirubin is the byproduct of hemoprotein catabolism, suggesting an interesting correlation. Additionally, this module contained 2 other pathways associated with porphyrin, which is a component of RBCs that contains the heme group and is involved in heme biosynthesis. Conclusion Taken together, these results provide interesting insights into the biological pathways driving one facet of sickle cell disease's many clinical manifestations. Figure 1 Disclosures No relevant conflicts of interest to declare.
APA, Harvard, Vancouver, ISO, and other styles
8

Rizalhanafi, Fatin Nabihah, Norlisah Ramli, Vairavan Narayanan, Khairunnisa Rashid, Jesminder Kaur Singh, and Kamariah Ibrahim. "OTEH-5. Characterization of long-non coding RNA associated ceRNA network hub gene involved in glioblastoma multiforme lipid metabolism." Neuro-Oncology Advances 3, Supplement_2 (July 1, 2021): ii11. http://dx.doi.org/10.1093/noajnl/vdab070.044.

Full text
Abstract:
Abstract Background Glioblastoma multiforme (GBM) are the major death contributor in primary brain tumour. Despite having an improved diagnostic criterion by integrating both histological and molecular features such as Isocitrate Dehydrogenase (IDH) detection, the prognosis of GBM patients still remain poor. Lipid metabolism is an essential pathway that fuel GBM aggressiveness. IDH1 one of the key enzyme that regulates it. Long non-coding RNAs (lncRNAs) act as competing endogenous RNAs (ceRNAs) in tumour initiation and progression. In parallel, miRNA-mediate ceRNA crosstalk between lncRNAs and mRNAs. In this study, we aim to investigate the IDH1 subgroup lncRNA associated ceRNA network hub gene responsible in the coordination of glioblastoma multiforme lipid metabolism using bioinformatics approach. Methods TCGA-GBM dataset consist of 168 GBM RNA-seq (159 IDH1 wt and 9 IDH1 mutation) were downloaded. Differentially expressed genes (DEG) were then obtained using Limma. Gene sets related with lipid metabolism from GSEA-MSigDB were overlapped with DEG using Venn diagram to identify the DEmRNA that are related with lipid metabolism. Construction of mRNA-miRNA and lncRNA-miRNA interaction networks were performed using miRNet. The ceRNA interaction network were later combined in the Cytoscape software. Potential lncRNA hub genes were identified by CytoHubba analysis. Results From 1389 DEG, 67 genes were identified to be significant in the regulation of lipid metabolism. By analysing the lncRNA-miRNA-mRNA interaction network, candidate hub lncRNAs consists of three genes with highest connective nodes; CYTOR, LOXL1-AS1 and HOTAIR. These genes are significantly upregulated in glioma. LOXL1-AS1 serve as an excellent prognostic biomarker for both glioma and glioblastoma as the effect of high and low LOXL1-AS1 expression on patients’ survival is significant (p&lt;0.05). Conclusions Data mining and bioinformatics approach guided the identification of the potential hub lncRNAs associated ceRNA network in GBM lipid metabolism. This allows us to uncover the novel role of lncRNA in GBM tumorigenesis.
APA, Harvard, Vancouver, ISO, and other styles
9

Li, Rongyang, Bojiang Li, Aiwen Jiang, Yan Cao, Liming Hou, Zengkai Zhang, Xiying Zhang, Honglin Liu, Kee-Hong Kim, and Wangjun Wu. "Exploring the lncRNAs Related to Skeletal Muscle Fiber Types and Meat Quality Traits in Pigs." Genes 11, no. 8 (August 4, 2020): 883. http://dx.doi.org/10.3390/genes11080883.

Full text
Abstract:
The alteration in skeletal muscle fiber is a critical factor affecting livestock meat quality traits and human metabolic diseases. Long non-coding RNAs (lncRNAs) are a diverse class of non-coding RNAs with a length of more than 200 nucleotides. However, the mechanisms underlying the regulation of lncRNAs in skeletal muscle fibers remain elusive. To understand the genetic basis of lncRNA-regulated skeletal muscle fiber development, we performed a transcriptome analysis to identify the key lncRNAs affecting skeletal muscle fiber and meat quality traits on a pig model. We generated the lncRNA expression profiles of fast-twitch Biceps femoris (Bf) and slow-twitch Soleus (Sol) muscles and identified the differentially expressed (DE) lncRNAs using RNA-seq and performed bioinformatics analyses. This allowed us to identify 4581 lncRNA genes among six RNA libraries and 92 DE lncRNAs between Bf and Sol which are the key candidates for the conversion of skeletal muscle fiber types. Moreover, we detected the expression patterns of lncRNA MSTRG.42019 in different tissues and skeletal muscles of various development stages. In addition, we performed a correlation analyses between the expression of DE lncRNA MSTRG.42019 and meat quality traits. Notably, we found that DE lncRNA MSTRG.42019 was highly expressed in skeletal muscle and its expression was significantly higher in Sol than in Bf, with a positive correlation with the expression of Myosin heavy chain 7 (MYH7) (r = 0.6597, p = 0.0016) and a negative correlation with meat quality traits glycolytic potential (r = −0.5447, p = 0.0130), as well as drip loss (r = −0.5085, p = 0.0221). Moreover, we constructed the lncRNA MSTRG.42019–mRNAs regulatory network for a better understanding of a possible mechanism regulating skeletal muscle fiber formation. Our data provide the groundwork for studying the lncRNA regulatory mechanisms of skeletal muscle fiber conversion, and given the importance of skeletal muscle fiber types in muscle-related diseases, our data may provide insight into the treatment of muscular diseases in humans.
APA, Harvard, Vancouver, ISO, and other styles
10

Li, Ying, Chao Zhang, Luwei Qin, Dong Li, Guangyuan Zhou, Dejian Dang, Shuaiyin Chen, et al. "Characterization of Critical Functions of Long Non-Coding RNAs and mRNAs in Rhabdomyosarcoma Cells and Mouse Skeletal Muscle Infected by Enterovirus 71 Using RNA-Seq." Viruses 10, no. 10 (October 11, 2018): 556. http://dx.doi.org/10.3390/v10100556.

Full text
Abstract:
Enterovirus 71 (EV71) is the main pathogen of severe hand-foot-mouth disease (HFMD). Long non-coding RNAs (lncRNAs) are recognized as pivotal factors during the pathogenesis of viral infection. However, the critical functions of lncRNAs in EV71–host interactions have not been characterized. Here, for the first time, we performed global transcriptome analysis of lncRNA and mRNA expression profiles in EV71-infected human rhabdomyosarcoma (RD) cells and skeletal muscle of mice using second-generation sequencing. In our study, a total of 3801 novel lncRNAs were identified. In addition, 23 lncRNAs and 372 mRNAs exhibited remarkable differences in expression levels between infected and uninfected RD cells, while 104 lncRNAs and 2647 mRNAs were differentially expressed in infected skeletal muscle from neonatal mice. Comprehensive bioinformatics analysis included target gene prediction, lncRNA‑mRNA co-expression network construction, as well as gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis mainly focused on differentially-expressed genes (DEGs). Our results suggest that lncRNAs may participate in EV71 infection-induced pathogenesis through regulating immune responses, protein binding, cellular component biogenesis and metabolism. The present study provides novel insights into the functions of lncRNAs and the possible pathogenic mechanism following EV71 infection.
APA, Harvard, Vancouver, ISO, and other styles
11

Zhang, Liwen, Ying Zhou, Fangfang Zhou, Xialian Yu, Jian Liu, Yunzi Liu, Yufei Zhu, Weiming Wang, and Nan Chen. "Altered Expression of Long Noncoding and Messenger RNAs in Diabetic Nephropathy following Treatment with Rosiglitazone." BioMed Research International 2020 (January 14, 2020): 1–17. http://dx.doi.org/10.1155/2020/1360843.

Full text
Abstract:
Diabetic nephropathy (DN) is characterized by metabolic disorder and inflammation. However, the regulatory effects that long noncoding RNAs (lncRNAs) have on the pathogenesis of DN and on the efficacy of rosiglitazone treatment have yet to be clearly defined. Herein, we performed unbiased RNA sequencing to characterize the transcriptomic profiles in db/db diabetic mouse model with or without rosiglitazone treatment that served to improve the phenotypes of DN. Moreover, RNA-seq profiling revealed that the development of DN caused an upregulation in the expression of 1176 mRNAs and a downregulation in the expression of 1010 mRNAs compared to controls, with the expression of 251 mRNAs being returned to normal following treatment with rosiglitazone. Further, 88 upregulated and 68 downregulated lncRNAs were identified in db/db mice compared to controls, 10 of which had their normal expression restored following treatment with rosiglitazone. Bioinformatic analysis revealed that the primary pathways involved in the pathogenesis of DN, and subsequently in the therapeutic effects of PPARγ, are related to inflammatory and metabolic processes. From bioinformatics analysis, lncRNA-AI838599 emerged as a novel molecular mechanism for rosiglitazone treatment in DN through TNFα-NFκb pathway. These findings may indicate a new molecular regulatory approach for the development of DN therapeutic agents.
APA, Harvard, Vancouver, ISO, and other styles
12

Zhang, Li-Hai, Jiao Wang, Bai-Hong Tan, Yan-Bin Yin, and Yu-Ming Kang. "The Association of lncRNA and mRNA Changes in Adipose Tissue with Improved Insulin Resistance in Type 2 Obese Diabetes Mellitus Rats after Roux-en-Y Gastric Bypass." Disease Markers 2022 (July 18, 2022): 1–11. http://dx.doi.org/10.1155/2022/8902916.

Full text
Abstract:
Objective. Roux-en-Y gastric bypass (RYGB) has shown good effects in improving obesity and type II diabetes mellitus (T2DM), but the underlying mechanisms remain unclear. This study explored the changes of related lncRNAs, mRNAs, and signaling pathways in white adipose tissue of T2DM rats after RYGB based on RNA-Seq sequencing, with the aim to provide a theoretical basis for RYGB treatment. Methods. T2DM rat models were established by continuous feeding with a high-fat diet and injection of streptozotocin (STZ), after which they underwent RYGB or sham surgery. After the surgery, their body weight was measured weekly. Their fasting blood glucose (FBG) and fasting serum insulin (FSI) were also measured. A homeostasis model assessment of insulin resistance (HOMA-IR) was calculated at weeks 0, 8, and 12. Besides, white adipose tissue of T2DM rats was collected for RNA-Seq sequencing and validated by qRT-PCR. A series of bioinformatics analyses, such as differential expression genes (DEGs) screening, was performed. GO and KEGG functional enrichment analysis and protein-protein interaction (PPI) network construction were conducted based on the sequencing data. Results. RYGB surgery could significantly inhibit the weight growth rate and decrease the FBG, FSI, and HOMA-IR of T2DM rats. Bioinformatics analysis of RNA sequencing (RNA-Seq) results revealed that 87 DE- lncRNAs (49 upregulated and 38 downregulated) and 1,824 DEGs (896 upregulated and 928 downregulated) were present in between the RYGB group and Sham group. GO and KEGG analysis showed that the target genes of DEGs and differentially expressed lncRNAs (DE-lncRNAs) were mainly associated with amino acid metabolism, fatty acid metabolism, channel activity, and other processes. In addition, the PPI network diagram also displayed that genes such as Fasn, Grin3a, and Nog could be key genes playing a role after RYGB. qRT-PCR showed that the expression level of Grin3a in the RYGB group was significantly increased compared with the Sham group, while the expression of Fasn and Nog was significantly decreased, which was consistent with the sequencing results. Conclusion. Using RNA-Seq sequencing, this study revealed the changes of related lncRNAs, mRNAs, and signaling pathways in the white adipose tissue of T2DM rats after RYGB and identified Fasn, Grin3a, and Nog as potential key genes to function after RYGB.
APA, Harvard, Vancouver, ISO, and other styles
13

Zhu, Xiaojian, Jinfeng Zhu, Ting Tan, Fanqin Bu, Jiefeng Zhao, Chen Luo, and Hongliang Luo. "RP11-51O6.1 sponges miR-206 to accelerate colorectal cancer carcinogenesis and metastasis through upregulating YAP1." Carcinogenesis 42, no. 7 (May 26, 2021): 984–94. http://dx.doi.org/10.1093/carcin/bgab044.

Full text
Abstract:
Abstract Long non-coding RNAs (lncRNAs) have been characterized by playing a crucial role in tumorigenesis. However, the detail biological function and clinical importance of lncRNAs in colorectal cancer (CRC) are unclear and have attracted different levels of in-depth research. In this context, we explored the differentially expressed profiles of lncRNAs in six CRC tissues and three adjacent non-tumor tissues from RNA-sequencing (RNA-seq) study and noted a lncRNA, RP11-51O6.1, which is markedly overexpressed in CRC tissues, particularly in aggressive cases. Impressively, an elevated RP11-51O6.1 level was highly correlated with poor prognosis in clinical patients. Functional analyses revealed that RP11-51O6.1 could promote cell proliferation in vitro and in vivo. Furthermore, we reported that RP11-51O6.1 enhances cell migration and invasion in vitro. Mechanistic studies (Bioinformatics binding site analyses, the Luciferase reporter, Ago2 immunoprecipitation, the RNA pull-down, immunofluorescence colocalization, rescued assays and western blotting) implicated that RP11-51O6.1 could regulate YAP1 expression by competitively sponging miR-206 and blocking its activity in promoting CRC progression. Conclusively, our findings identify a novel RP11-51O6.1/miR-206/YAP1 regulatory axis that participates in CRC progression and development, suggesting RP11-51O6.1 is an exploitable biomarker and appealing therapeutic target in treating CRC.
APA, Harvard, Vancouver, ISO, and other styles
14

He, Haibo, Hanwen Huang, Panyong Hu, and Zhong Chen. "Microarray and Bioinformatics Analysis of Differential Gene and lncRNA Expression during Erythropoietin Treatment of Acute Spinal Cord Injury in Rats." Computational and Mathematical Methods in Medicine 2022 (September 2, 2022): 1–13. http://dx.doi.org/10.1155/2022/4121910.

Full text
Abstract:
Purpose. We performed a genome-wide analysis of long noncoding RNA (lncRNA) expression to identify novel targets for the further study of recombinant human erythropoietin (rhEPO) treatment of acute spinal cord injury (SCI) in rats. Methods. Nine rats were randomly divided into 3 groups. No operation was performed in group 1. In groups 2 and 3, a laminectomy was performed at the 10th thoracic vertebra, and a contusion injury was induced by extradural application of an aneurysm clip. Group 1 rats did not receive any treatment, group 2 rats received a single intraperitoneal injection of normal saline, and group 3 rats received rhEPO. Three days after injury, spinal cord tissues were collected for RNA-Seq, microarray, differentially expressed genes (DEGs), Gene Ontology (GO) function enrichment, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment, and protein-protein interaction (PPI) analyses. Results. Compared with group 1, 4,446 genes were found to be differentially expressed in group 2. Furthermore, 99 lncRNAs were found to be changed in the injury group. The data indicate that 2,471 mRNAs were upregulated, and 1,975 mRNAs were downregulated in group 2 as compared with group 1. In addition, 45 of the lncRNAs were upregulated, and the other 44 lncRNAs were downregulated. The top 5 upregulated and top 5 downregulated lncRNAs that were different between group 2 and group 1 are shown. The top 5 downregulated and the top 5 upregulated lncRNAs that were different between group 3 and group 2 are shown. Conclusion. RhEPO treatment alters the expression profiles of the differentially expressed lncRNAs and genes beneficial to the development of new treatments.
APA, Harvard, Vancouver, ISO, and other styles
15

Li, Yang, Ling Deng, Xiaofeng Pan, Chunyan Liu, and Rong Fu. "The Role of lncRNA AF117829.1 in the Immunological Pathogenesis of Severe Aplastic Anaemia." Oxidative Medicine and Cellular Longevity 2021 (March 15, 2021): 1–19. http://dx.doi.org/10.1155/2021/5587921.

Full text
Abstract:
Objective. Severe aplastic anaemia (SAA) is an autoimmune disease with immune tolerance dysfunction mediated by hyperactivated T lymphocytes that target the haematopoietic system. Numerous studies suggest that long noncoding RNAs (lncRNAs) play a significant role in almost every level of gene function/regulation. However, their specific mechanisms in SAA remain undetermined. This study is aimed at determining the role of key lncRNAs in CD8+ T lymphocytes in the mechanisms of SAA. Methods. RNA-seq was performed to detect all lncRNAs and mRNAs in peripheral CD8+ T lymphocytes from SAA patients and healthy controls. The lncRNA targets were predicted by bioinformatics, Gene Ontology (GO) analysis, and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis. RT-qPCR was used to verify the expression of key lncRNAs and their predicted targets. We screened lncRNA AF117829.1, which was correlated with autoimmune diseases and downregulated in CD8+ T lymphocytes, and further validated its effects on CD8+ T lymphocytes from SAA patients. Results. We systematically described the lncRNA/mRNA expression changes in CD8+ T lymphocytes in SAA patients and assessed their possible biological functions and signalling pathways. A total of 194 lncRNAs and 2099 mRNAs were changed in SAA patients versus healthy controls. These differentially expressed lncRNAs/mRNAs were associated with organelle components, catalytic activity, the response to stimulation, signal transduction, the immune system and metabolic processes. The downregulated expression of one altered factor, lncRNA AF117829.1, in CD8+ T lymphocytes from SAA patients increased CD8+ T lymphocyte immune function by promoting RIP2 expression. lncRNA AF117829.1 overexpression in CD8+ T lymphocytes reduced perforin and granzyme B expression. The same effect was achieved with GSK583, a RIP2 kinase inhibitor. Conclusions. The proliferation and overactivation of CD8+ T lymphocytes, also known as cytotoxic T cells (CTLs), directly induce bone marrow (BM) failure in SAA patients, but the specific mechanism remains unclear. We found that lncRNA AF117829.1 and its target genes were associated with T cell proliferation, differentiation, and immune dysregulation and that lncRNA AF117829.1 regulated CD8+ T lymphocyte function in SAA patients by promoting RIP2 expression. These findings improve our understanding of the molecular mechanism of immune pathogenesis and provide potential targets for SAA diagnosis and treatment.
APA, Harvard, Vancouver, ISO, and other styles
16

Wang, Hongxian, Lirong Shu, Nan Niu, Chenyang Zhao, Shuqi Lu, Yanhua Li, Huanyu Wang, et al. "Novel lncRNAs with diagnostic or prognostic value screened out from breast cancer via bioinformatics analyses." PeerJ 10 (July 14, 2022): e13641. http://dx.doi.org/10.7717/peerj.13641.

Full text
Abstract:
Background Recent studies have shown that long non-coding RNAs (lncRNAs) may play key regulatory roles in many malignant tumors. This study investigated the use of novel lncRNA biomarkers in the diagnosis and prognosis of breast cancer. Materials and Methods The database subsets of The Cancer Genome Atlas (TCGA) by RNA-seq for comparing analysis of tissue samples between breast cancer and normal control groups were downloaded. Additionally, anticoagulant peripheral blood samples were collected and used in this cohort study. The extracellular vesicles (EVs) from the plasma were extracted and sequenced, then analyzed to determine the expressive profiles of the lncRNAs, and the cancer-related differentially expressed lncRNAs were screened out. The expressive profiles and associated downstream-mRNAs were assessed using bioinformatics (such as weighted correlation network analysis (WGCNA), Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genome (KEGG) enrichments, Receiver-Operating Characteristic (ROC) curve and survival analysis, etc.) to investigate the diagnostic and prognostic values of these EV lncRNAs and their effectors. Results In this study, 41 breast cancer-related lncRNAs were screen out from two datasets of tissue and fresh collected plasma samples of breast cancer via the transcriptomic and bioinformatics techniques. A total of 19 gene modules were identified with WGCNA analysis, of which five modules were significantly correlated with the clinical stage of breast cancer, including 28 lncRNA candidates. The ROC curves of these lncRNAs revealed that the area under the curve (AUC) of all candidates were great than 70%. However, eight lncRNAs had an AUC >70%, indicating that the combined one has a good diagnostic value. In addition, the results of survival analysis suggested that two lncRNAs with low expressive levels may indicate the poor prognosis of breast cancer. By tissue sample verification, C15orf54, AL157935.1, LINC01117, and SNHG3 were determined to have good diagnostic ability in breast cancer lesions, however, there was no significant difference in the plasma EVs of patients. Moreover, survival analysis data also showed that AL355974.2 may serve as an independent prognostic factor and as a protective factor. Conclusion A total of five lncRNAs found in this study could be developed as biomarkers for breast cancer patients, including four diagnostic markers (C15orf54, AL157935.1, LINC01117, and SNHG3) and a potential prognostic marker (AL355974.2).
APA, Harvard, Vancouver, ISO, and other styles
17

Bjeije, Hassan, Bahram Mohammad Soltani, Mehrdad Behmanesh, and Mohammad Reza Zali. "YWHAE long non-coding RNA competes with miR-323a-3p and miR-532-5p through activating K-Ras/Erk1/2 and PI3K/Akt signaling pathways in HCT116 cells." Human Molecular Genetics 28, no. 19 (May 27, 2019): 3219–31. http://dx.doi.org/10.1093/hmg/ddz146.

Full text
Abstract:
AbstractYWHAE gene product belongs to the 14-3-3 protein family that mediates signal transduction in plants and mammals. Protein-coding and non-coding RNA (lncRNA) transcripts have been reported for this gene in human. Here, we aimed to functionally characterize YWHAE-encoded lncRNA in colorectal cancer-originated cells. RNA-seq analysis showed that YWHAE gene is upregulated in colorectal cancer specimens. Additionally, bioinformatics analysis suggested that YWHAE lncRNA sponges miR-323a-3p and miR-532-5p that were predicted to target K-Ras 3′UTR sequence. Overexpression of YWHAE lncRNA resulted in upregulation of K-Ras gene expression, while overexpression of both miR-323a-3p and miR-532-5p had an inverse effect, detected by RT-qPCR. Consistently, western blot analysis confirmed that YWHAE lncRNA overexpression upregulated K-Ras/Erk1/2 and PI3K/Akt signaling pathways, while miR-323a-3p and miR-532-5p overexpression suppressed both pathways in HCT116 cells. Furthermore, dual luciferase assay validated the direct interaction of miR-323a-3p and miR-532-5p with K-Ras 3′UTR sequence and supported the sponging effect of YWHAE lncRNA over both miRNAs. These results suggested YWHAE lncRNA as an oncogene that exerts its effect through sponging miR-323a-3p and miR-532-5p and in turn, upregulates K-Ras/Erk1/2 and PI3K/Akt signaling pathways. Consistently, flow cytometry analysis, MTT assay and measuring cyclin D1 gene expression, confirmed the cell cycle stimulatory effect of YWHAE lncRNA, while miR-323a-3p and miR-532-5p showed an inhibitory effect on cell cycle progression. Finally, wound-healing assay supported the cell migratory effect of YWHAE lncRNA in HCT116 cells. This study identified a novel mechanism involving YWHAE-encoded lncRNA, miR-323a-3p and miR-532-5p in regulating HCT116 cell survival and suggested a potential therapeutic avenue for colorectal cancer.
APA, Harvard, Vancouver, ISO, and other styles
18

Wu, Baojin, Xinjie Tang, Zhaoping Zhou, Honglin Ke, Shao Tang, and Ronghu Ke. "RNA sequencing analysis of FGF2-responsive transcriptome in skin fibroblasts." PeerJ 9 (January 15, 2021): e10671. http://dx.doi.org/10.7717/peerj.10671.

Full text
Abstract:
Background Fibroblast growth factor 2 (FGF2) is a highly pleiotropic cytokine with antifibrotic activity in wound healing. During the process of wound healing and fibrosis, fibroblasts are the key players. Although accumulating evidence has suggested the antagonistic effects of FGF2 in the activation process of fibroblasts, the mechanisms by which FGF2 hinders the fibroblast activation remains incompletely understood. This study aimed to identify the key genes and their regulatory networks in skin fibroblasts treated with FGF2. Methods RNA-seq was performed to identify the differentially expressed mRNA (DEGs) and lncRNA between FGF2-treated fibroblasts and control. DEGs were analyzed by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG). Furthermore, the networks between mRNAs and lncRNAs were constructed by Pearson correlation analysis and the networkanalyst website. Finally, hub genes were validated by real time-PCR. Results Between FGF2-treated fibroblasts and control fibroblasts, a total of 1475 DEGs was obtained. These DEGs were mainly enriched in functions such as the ECM organization, cell adhesion, and cell migration. They were mainly involved in ECM-receptor interaction, PI3K-Akt signaling, and the Hippo pathway. The hub DEGs included COL3A1, COL4A1, LOX, PDGFA, TGFBI, and ITGA10. Subsequent real-time PCR, as well as bioinformatics analysis, consistently demonstrated that the expression of ITGA10 was significantly upregulated while the other five DEGs (COL3A1, COL4A1, LOX, PDGFA, TGFBI) were downregulated in FGF2-treated fibroblasts. Meanwhile, 213 differentially expressed lncRNAs were identified and three key lncRNAs (HOXA-AS2, H19, and SNHG8) were highlighted in FGF2-treated fibroblasts. Conclusion The current study comprehensively analyzed the FGF2-responsive transcriptional profile and provided candidate mechanisms that may account for FGF2-mediated wound healing.
APA, Harvard, Vancouver, ISO, and other styles
19

Li, Yuchen, Jingjing Zhao, Shulin Yu, Zhen Wang, Xigan He, Yonghui Su, Tianan Guo, et al. "Extracellular Vesicles Long RNA Sequencing Reveals Abundant mRNA, circRNA, and lncRNA in Human Blood as Potential Biomarkers for Cancer Diagnosis." Clinical Chemistry 65, no. 6 (June 1, 2019): 798–808. http://dx.doi.org/10.1373/clinchem.2018.301291.

Full text
Abstract:
Abstract BACKGROUND Extracellular vesicles (EVs) contain a rich cargo of different RNA species with specialized functions and clinical applications. However, the landscape and characteristics of extracellular vesicle long RNA (exLR) in human blood remain largely unknown. METHODS We presented an optimized strategy for exLR sequencing (exLR-seq) of human plasma. The sample cohort included 159 healthy individuals, 150 patients with cancer (5 cancer types), and 43 patients with other diseases. Bioinformatics approaches were used to analyze the distribution and features of exLRs. Support vector machine algorithm was performed to construct the diagnosis classifier, and diagnostic efficiency was evaluated by ROC analysis. RESULTS More than 10000 exLRs, including mRNA, circRNA, and lncRNA, were reliably detected in each exLR-seq sample from 1–2 mL of plasma. We observed that blood EVs contain a substantial fraction of intact mRNAs and a large number of assembling spliced junctions; circRNA was also enriched in blood EVs. Interestingly, blood exLRs reflected their tissue origins and the relative fractions of different immune cell types. Additionally, the exLR profile could distinguish patients with cancer from healthy individuals. We further showed that 8 exLRs can serve as biomarkers for hepatocellular carcinoma (HCC) diagnosis with high diagnostic efficiency in training [area under the curve (AUC) = 0.9527; 95% CI, 0.9170–0.9883], validation cohort (AUC = 0.9825; 95% CI, 0.9606–1), and testing cohort (AUC = 0.9627; 95% CI, 0.9263–0.9991). CONCLUSIONS In summary, this study revealed abundant exLRs in human plasma and identified diverse specific markers potentially useful for cancer diagnosis.
APA, Harvard, Vancouver, ISO, and other styles
20

Huang, Wanlong, Xiuxiu Zhang, Ai Li, Lingli Xie, and Xiangyang Miao. "Genome-Wide Analysis of mRNAs and lncRNAs of Intramuscular Fat Related to Lipid Metabolism in Two Pig Breeds." Cellular Physiology and Biochemistry 50, no. 6 (2018): 2406–22. http://dx.doi.org/10.1159/000495101.

Full text
Abstract:
Background/Aims: Long non-coding RNAs (lncRNAs) can regulate adipogenesis and lipid accumulation. Intramuscular fat deposition appears to vary in different pig breeds, and the regulation mechanism has not yet been fully elucidated at molecular level. Moreover, little is known about the function and profile of lncRNAs in intramuscular fat deposition and metabolism in pig. The aim of this study was thus to explore the regulatory functions of lncRNAs in intramuscular fat deposition. Methods: In this study, Laiwu (LW) pig and Large White (LY) pig with significant difference in fat deposition were selected for use. RNA-seq technology and bioinformatics methods were used to comparatively analyze the gene expression profiles of intramuscular fat between LW and LY pigs to identify key mRNAs and lncRNAs associated with lipid metabolism and adipogenesis. Real-time fluorescence-based quantitative PCR was applied to verify the expression level of the differentially expressed mRNAs and lncRNAs. Results: A total of 513 mRNAs and 55 lncRNAs were differentially expressed between two pig breeds. By co-expression network construction as well as cis- and trans-regulated target gene analysis, 31 key lncRNAs were identified. Gene Ontology and KEGG pathway analyses revealed that differentially expressed genes and lncRNAs were mainly involved in the biological processes and pathways related to adipogenesis and lipid metabolism. Conclusion: XLOC_046142, XLOC_004398 and XLOC_015408 may target MAPKAPK2, NR1D2 and AKR1C4, respectively, and play critical regulatory roles in intramuscular adipogenesis and lipid accumulation in pig. XLOC_064871 and XLOC_011001 may play a role in lipid metabolism-related disease via regulating TRIB3 and BRCA1. This study provides a valuable resource for lncRNA study and improves our understanding of the biological roles of lipid metabolism- related genes and molecular mechanism of intramuscular fat metabolism and deposition.
APA, Harvard, Vancouver, ISO, and other styles
21

Luo, Yuhao, Rui Zhou, Na Huang, Li Sun, and Wangjun Liao. "Effect of long non-coding RNA EIF3J-AS1 on multi-drug resistance and autophagy in gastric cancer." Journal of Clinical Oncology 35, no. 15_suppl (May 20, 2017): e15581-e15581. http://dx.doi.org/10.1200/jco.2017.35.15_suppl.e15581.

Full text
Abstract:
e15581 Background: Gastric cancer (GC) is a leading cause of cancer mortality worldwide, oxaliplatin and epirubicin based chemotherapy are one of the most important treatment options for GC patients. However, drug resistance, especially multi-drug resistance remains a major obstacle for successful chemotherapy. Recently, long non-coding RNAs (lncRNAs) have been widely identified to play emerging roles in diverse physiological and pathophysiological processes including drug resistance. Our previous bioinformatics analysis showed long non-coding RNA EIF3J-AS1 was a potential multi-drug resistance gene, but the underlying mechanism is still unknown. Methods: We generated oxaliplatin resistance cells (MGC803/OXA) and epirubicin resistance cells(MGC803/EPI) based on parental gastric cancer cells MGC803. Relative expression levels of EIF3J-AS1 were measured by qRT-PCR. Transmission electron microscopy was used to measure autophagosomes. Rapamycin was applied to inducing autophagy while chloroquine and 3-methyladenine were used to block autophagy. Protein level of autophagy related genes were examined by Western Blot. Coexpression genes of EIF3J-AS1 from TCGA RNA-seq datas were analyzed by cBiportal. RNA immunoprecipitation was used to analyze endogenous microRNAs and mRNAs. Results: EIF3J-AS1 was significantly upregulated in MGC803/OXA and MGC803/EPI cells compared with parental cells MGC803. EIF3J-AS1 inhibition increased chemosensitivity to both oxaliplatin and epirubicin. Moreover, EIF3J-AS1 silence lead to the decrease of autophagy. Autophagy related gene ATG14 was identified as a downstream target gene. EIF3J-AS1 promoted ATG14 expression by directly interacting with and increasing stability of ATG14 mRNA, On the other hand, EIF3J-AS1 competitively sponged miR-188-3p and promoted ATG14 expression in a ceRNA-dependent way. Conclusions: LncRNA EIF3J-AS1 is a crucial regulator of multi-drug resistance by inducing autophagy in gastric cancer. Targeting EIF3J-AS1/ATG14 axis might be a new paradigm for cancer therapeutics.
APA, Harvard, Vancouver, ISO, and other styles
22

Flower, Cameron T., Lihe Chen, Hyun Jun Jung, Viswanathan Raghuram, Mark A. Knepper, and Chin-Rang Yang. "An integrative proteogenomics approach reveals peptides encoded by annotated lincRNA in the mouse kidney inner medulla." Physiological Genomics 52, no. 10 (October 1, 2020): 485–91. http://dx.doi.org/10.1152/physiolgenomics.00048.2020.

Full text
Abstract:
Long noncoding RNAs (lncRNAs) are intracellular transcripts longer than 200 nucleotides and lack protein-coding information. A subclass of lncRNA known as long intergenic noncoding RNAs (lincRNAs) are transcribed from genomic regions that share no overlap with annotated protein-coding genes. Increasing evidence has shown that some annotated lincRNA transcripts do in fact contain open reading frames (ORFs) encoding functional short peptides in the cell. Few robust methods for lincRNA-encoded peptide identification have been reported, and the tissue-specific expression of these peptides has been largely unexplored. Here we propose an integrative workflow for lincRNA-encoded peptide discovery and test it on the mouse kidney inner medulla (IM). In brief, low molecular weight protein fractions were enriched from homogenate of IMs and trypsinized into shorter peptides, which were sequenced by high resolution liquid chromatography-tandem mass spectrometry (LC-MS/MS). To curate a hypothetical lincRNA-encoded peptide database for peptide-spectrum matching following LC-MS/MS, we performed RNA-Seq on IMs, computationally removed reads overlapping with annotated protein-coding genes, and remapped the remaining reads to a database of mouse noncoding transcripts to infer lincRNA expression. Expressed lincRNAs were searched for ORFs by an existing rule-based algorithm, and translated ORFs were used for peptide-spectrum matching. Peptides identified by LC-MS/MS were further evaluated by using several quality control criteria and bioinformatics methods. We discovered three novel lincRNA-encoded peptides, which are conserved in mouse, rat, and human. The workflow can be adapted for discovery of small protein-coding genes in any species or tissue where noncoding transcriptome information is available.
APA, Harvard, Vancouver, ISO, and other styles
23

Medina, José María, Muhammad Nadeem Abbas, Chaima Bensaoud, Michael Hackenberg, and Michail Kotsyfakis. "Bioinformatic Analysis of Ixodes ricinus Long Non-Coding RNAs Predicts Their Binding Ability of Host miRNAs." International Journal of Molecular Sciences 23, no. 17 (August 28, 2022): 9761. http://dx.doi.org/10.3390/ijms23179761.

Full text
Abstract:
Ixodes ricinus ticks are distributed across Europe and are a vector of tick-borne diseases. Although I. ricinus transcriptome studies have focused exclusively on protein coding genes, the last decade witnessed a strong increase in long non-coding RNA (lncRNA) research and characterization. Here, we report for the first time an exhaustive analysis of these non-coding molecules in I. ricinus based on 131 RNA-seq datasets from three different BioProjects. Using this data, we obtained a consensus set of lncRNAs and showed that lncRNA expression is stable among different studies. While the length distribution of lncRNAs from the individual data sets is biased toward short length values, implying the existence of technical artefacts, the consensus lncRNAs show a more homogeneous distribution emphasizing the importance to incorporate data from different sources to generate a solid reference set of lncRNAs. KEGG enrichment analysis of host miRNAs putatively targeting lncRNAs upregulated upon feeding showed that these miRNAs are involved in several relevant functions for the tick-host interaction. The possibility that at least some tick lncRNAs act as host miRNA sponges was further explored by identifying lncRNAs with many target regions for a given host miRNA or sets of host miRNAs that consistently target lncRNAs together. Overall, our findings suggest that lncRNAs that may act as sponges have diverse biological roles related to the tick–host interaction in different tissues.
APA, Harvard, Vancouver, ISO, and other styles
24

Paralkar, Vikram R., Tejaswini Mishra, Jing Luan, Yu Yao, Neeraja Konuthula, David M. Bodine, Ross C. Hardison, and Mitchell J. Weiss. "Long Noncoding RNAs in Erythropoiesis and Megakaryopoiesis." Blood 120, no. 21 (November 16, 2012): 2331. http://dx.doi.org/10.1182/blood.v120.21.2331.2331.

Full text
Abstract:
Abstract Abstract 2331 Lnc (long noncoding) RNAs are RNA transcripts greater than 200nt that regulate gene expression independent of protein coding potential. It is estimated that thousands of lncRNAs play vital roles in diverse cellular processes and are involved in numerous diseases, including cancer. We hypothesize that multiple lncRNAs regulate erythrocyte and megakaryocyte formation by modulating gene expression. To identify lncRNAs in erythro-megakaryopoiesis, we purified two biological replicates each of murine Ter119+ erythroblasts, CD41+ megakaryocytes and bipotential megakaryocyte-erythroid progenitors (MEPs) [Lin− Kit+, Sca1−, CD16/32−, CD34−]. We performed strand-specific, paired-end, 200nt-read-length deep sequencing (RNA-Seq) to a depth of ∼200 million reads per sample using the Illumina GAII platform. We used the Tophat and Cufflinks suite of bioinformatic tools to assemble and compare de-novo transcriptomes from these three cell types, producing a high-confidence set of 69,488 transcripts. We confirmed that the RNA-seq assemblies accurately reflect gene expression predicted from prior studies. For example, Ter119+ cells were highly enriched for key erythroid transcripts encoding globins, heme synthetic enzymes and specialized membrane proteins. Megakaryocytes expressed high levels of gene encoding lineage-specific integrins and platelet markers. MEPs expressed numerous progenitor genes including Gata2, Kit and Myc. Thus, the RNA-seq data are of high-quality and sufficient complexity to accurately represent erythroid, megakaryocytic and MEP transcriptomes. We used a series of Unix-based bioinformatic filtering tools to identify lncRNAs that are expressed in these transcriptomes. We identified 605 “stringent” lncRNAs, and 813 “potential noncoding” transcripts. 47% of the lncRNAs are novel unannotated transcripts, validating the use of de-novo RNA-Seq in unique cell populations for lncRNA discovery. Among the 605 “stringent” lncRNAs, 103 are erythroid-restricted, 133 are meg-restricted and 280 are MEP-restricted, consistent with reports that lncRNAs exhibit exquisitely cell-type specific expression. Current efforts are aimed at generating a more comprehensive map of lncRNA expression at specific stages of erythroid and megakaryocyte/platelet development, and performing high throughput functional screens to analyze currently identified lncRNAs. Our studies are beginning to define new layers of gene regulation in normal erythro-megakaryopoiesis and are relevant to the pathophysiology of related disorders including various anemias, myeloproliferative and myelodysplastic syndromes and leukemias. Disclosures: No relevant conflicts of interest to declare.
APA, Harvard, Vancouver, ISO, and other styles
25

Kunz, Meik, Beat Wolf, Maximilian Fuchs, Jan Christoph, Ke Xiao, Thomas Thum, David Atlan, Hans-Ulrich Prokosch, and Thomas Dandekar. "A comprehensive method protocol for annotation and integrated functional understanding of lncRNAs." Briefings in Bioinformatics 21, no. 4 (October 3, 2019): 1391–96. http://dx.doi.org/10.1093/bib/bbz066.

Full text
Abstract:
Abstract Long non-coding RNAs (lncRNAs) are of fundamental biological importance; however, their functional role is often unclear or loosely defined as experimental characterization is challenging and bioinformatic methods are limited. We developed a novel integrated method protocol for the annotation and detailed functional characterization of lncRNAs within the genome. It combines annotation, normalization and gene expression with sequence-structure conservation, functional interactome and promoter analysis. Our protocol allows an analysis based on the tissue and biological context, and is powerful in functional characterization of experimental and clinical RNA-Seq datasets including existing lncRNAs. This is demonstrated on the uncharacterized lncRNA GATA6-AS1 in dilated cardiomyopathy.
APA, Harvard, Vancouver, ISO, and other styles
26

Affinito, Ornella, Katia Pane, Giovanni Smaldone, Francesca Maria Orlandella, Peppino Mirabelli, Giuliana Beneduce, Rosanna Parasole, Mimmo Ripaldi, Marco Salvatore, and Monica Franzese. "lncRNAs–mRNAs Co–Expression Network Underlying Childhood B–Cell Acute Lymphoblastic Leukaemia: A Pilot Study." Cancers 12, no. 9 (September 2, 2020): 2489. http://dx.doi.org/10.3390/cancers12092489.

Full text
Abstract:
Long non–coding RNAs (lncRNAs) are emerging as key gene regulators in the pathogenesis and development of various cancers including B lymphoblastic leukaemia (B–ALL). In this pilot study, we used RNA–Seq transcriptomic data for identifying novel lncRNA–mRNA cooperative pairs involved in childhood B–ALL pathogenesis. We conceived a bioinformatic pipeline based on unsupervised PCA feature extraction approach and stringent statistical criteria to extract potential childhood B–ALL lncRNA signatures. We then constructed a co–expression network of the aberrantly expressed lncRNAs (30) and protein–coding genes (754). We cross–validated our in–silico findings on an independent dataset and assessed the expression levels of the most differentially expressed lncRNAs and their co–expressed mRNAs through ex vivo experiments. Using the guilt–by–association approach, we predicted lncRNA functions based on their perfectly co–expressed mRNAs (Spearman’s correlation) that resulted closely disease–associated. We shed light on 24 key lncRNAs and their co–expressed mRNAs which may play an important role in B–ALL pathogenesis. Our results may be of clinical utility for diagnostic and/or prognostic purposes in paediatric B–ALL management.
APA, Harvard, Vancouver, ISO, and other styles
27

Zhao, Yixuan, Xin Huang, Zewei Zhang, Haizhou Li, and Tao Zan. "The Long Noncoding Transcript HNSCAT1 Activates KRT80 and Triggers Therapeutic Efficacy in Head and Neck Squamous Cell Carcinoma." Oxidative Medicine and Cellular Longevity 2022 (August 4, 2022): 1–19. http://dx.doi.org/10.1155/2022/4156966.

Full text
Abstract:
Head and neck squamous carcinoma (HNSC) is the most prevalent malignancy of the head and neck regions. Long noncoding RNAs (lncRNAs) are vital in tumorigenesis regulation. However, the role of lncRNAs in HNSC requires further exploration. Herein, through bioinformatic assays using The Cancer Genome Atlas (TCGA) datasets, rapid amplification of cDNA ends (RACE) assays, and RNA-FISH, we revealed that a novel cytoplasmic transcript, HNSC-associated transcript 1 (HNSCAT1, previously recognized as linc01269), was downregulated in tumor samples and advanced tumor stages and was also associated with favorable outcomes in HNSC. Overexpression of HNSCAT1 triggered treatment efficacy in HNSCs both in vivo and in vitro. More importantly, through high-throughput transcriptome analysis (RNA-seq, in NODE database, OEZ007550), we identified KRT80, a tumor suppressor in HNSC, as the target of HNSCAT1. KRT80 expression was modulated by lncRNA HNSCAT1 and presented a positive correlation in tumor samples ( R = 0.52 , p < 0.001 ). Intriguingly, we identified that miR-1245 simultaneously interacts with KRT80 and HNSCAT1, which bridges the regulatory function between KRT80 and HNSCAT1. Conclusively, our study demonstrated that lncRNA HNSCAT1 functions as a necessary tumor inhibitor in HNSC, which provides a novel mechanism of lncRNA function and provides alternative targets for the diagnosis and treatment of HNSC.
APA, Harvard, Vancouver, ISO, and other styles
28

Wu, Hao, Tiantian Liu, Jianni Qi, Chengyong Qin, and Qiang Zhu. "Four Autophagy-Related lncRNAs Predict the Prognosis of HCC through Coexpression and ceRNA Mechanism." BioMed Research International 2020 (October 9, 2020): 1–19. http://dx.doi.org/10.1155/2020/3801748.

Full text
Abstract:
Abnormally expressed long noncoding RNAs (lncRNAs) have been reported to affect the occurrence and progression of hepatocellular carcinoma (HCC) by modulating the autophagy axis. However, none of studies has explored the clinical significance of these autophagy-related lncRNAs in HCC comprehensively. In this study, the RNA-seq, miRNA-seq, and clinical data of normal and HCC patients from the TCGA database and autophagy genes from the Human Autophagy Database were extracted. Subsequently, we screened out 78 differentially expressed autophagy-related lncRNAs, and four prognostic-related lncRNAs (LUCAT1, AC099850.3, ZFPM2-AS1, and AC009005.1) were eventually used to develop the prognostic model. This signature could be regarded as an independent prognostic signature for HCC patients and has the highest prediction efficiency than other clinicopathological factors for the 1-, 3-, and 5-year survival (AUC=0.764,0.738,and 0.717, respectively). Additionally, regardless of whether the clinical information is complete for HCC patients, the autophagy-related lncRNA model shows a good predictive power for the overall survival. Importantly, the coexpression network of 4 lncRNAs and 11 autophagy-related genes was constructed. Moreover, based on the bioinformatic analyses, our results found that LUCAT1 and ZFPM2-AS1 may affect the autophagic activity in HCC through the hsa-miR-495-3p/DLC1 and hsa-miR-515-5p/DAPK2 axis, respectively. In conclusion, we establish an effective prognostic model for HCC patients and shed new light on the autophagy-related regulatory mechanisms of the identified lncRNAs.
APA, Harvard, Vancouver, ISO, and other styles
29

Liu, Hongxia, Qianping Chen, Wang Zheng, Yuchuan Zhou, Yang Bai, Yan Pan, Jianghong Zhang, and Chunlin Shao. "LncRNA CASC19 Enhances the Radioresistance of Nasopharyngeal Carcinoma by Regulating the miR-340-3p/FKBP5 Axis." International Journal of Molecular Sciences 24, no. 3 (February 3, 2023): 3047. http://dx.doi.org/10.3390/ijms24033047.

Full text
Abstract:
Radioresistance remains a serious obstacle encountered in the radiotherapy of nasopharyngeal carcinoma (NPC). Both mRNAs and non-coding RNAs (ncRNAs), including long ncRNA (lncRNA) and microRNA (miRNA), play essential roles in radiosensitivity. However, the comprehensive expression profiles and competing endogenous RNA (ceRNA) regulatory networks among lncRNAs, miRNAs, and mRNAs in NPC radioresistance are still bewildering. In this study, we performed an RNA-sequencing (RNA-seq) assay in the radioresistant NPC cells CNE2R and its parental cells CNE2 to identify the differentially expressed lncRNAs, miRNAs, and mRNAs. The ceRNA networks containing lncRNAs, miRNAs, and mRNAs were predicted on the basis of the Pearson correlation coefficients and authoritative miRanda databases. In accordance with bioinformatic analysis of the data of the tandem mass tag (TMT) assay of CNE2R and CNE2 cells and the gene chip assay of radioresistant NPC samples in pre- and post-radiotherapy, the radioresistance-related signaling network of lncRNA CASC19, miR-340-3p, and FKBP5 was screened and further verified using an RT-qPCR assay. CASC19 was positively associated with FKBP5 expression while negatively correlated with miR-340-3p, and the target binding sites of CASC19/miR-340-3p and miR-340-3p/FKBP5 were confirmed using a dual-luciferase reporter assay. Moreover, using an mRFP–GFP–LC3 maker, it was found that autophagy contributed to the radioresistance of NPC. MiR-340-3p inhibition or FKBP5 overexpression could rescue the suppression of autophagy and radioresistance induced by CASC19 knockdown in CNE2R cells. In conclusion, the CASC19/miR-340-3p/FKBP5 network may be instrumental in regulating NPC radioresistance by enhancing autophagy, which provides potential new therapeutic targets for NPC.
APA, Harvard, Vancouver, ISO, and other styles
30

Lipka, Aleksandra, Jan Pawel Jastrzebski, Lukasz Paukszto, Karol Gustaw Makowczenko, Elzbieta Lopienska-Biernat, Marek Gowkielewicz, Ewa Lepiarczyk, Marta Wiszpolska, Mariusz Krzysztof Majewski, and Marta Majewska. "Sex-Biased lncRNA Signature in Fetal Growth Restriction (FGR)." Cells 10, no. 4 (April 16, 2021): 921. http://dx.doi.org/10.3390/cells10040921.

Full text
Abstract:
Impaired fetal growth is one of the most important causes of prematurity, stillbirth and infant mortality. The pathogenesis of idiopathic fetal growth restriction (FGR) is poorly understood but is thought to be multifactorial and comprise a range of genetic causes. This research aimed to investigate non-coding RNAs (lncRNAs) in the placentas of male and female fetuses affected by FGR. RNA-Seq data were analyzed to detect lncRNAs, their potential target genes and circular RNAs (circRNAs); a differential analysis was also performed. The multilevel bioinformatic analysis enabled the detection of 23,137 placental lncRNAs and 4263 of them were classified as novel. In FGR-affected female fetuses’ placentas (ff-FGR), among 19 transcriptionally active regions (TARs), five differentially expressed lncRNAs (DELs) and 12 differentially expressed protein-coding genes (DEGs) were identified. Within 232 differentially expressed TARs identified in male fetuses (mf-FGR), 33 encompassed novel and 176 known lncRNAs, and 52 DEGs were upregulated, while 180 revealed decreased expression. In ff-FGR ACTA2-AS1, lncRNA expression was significantly correlated with five DEGs, and in mf-FGR, 25 TARs were associated with DELs correlated with 157 unique DEGs. Backsplicing circRNA processes were detected in the range of H19 lncRNA, in both ff- and mf-FGR placentas. The performed global lncRNAs characteristics in terms of fetal sex showed dysregulation of DELs, DEGs and circRNAs that may affect fetus growth and pregnancy outcomes. In female placentas, DELs and DEGs were associated mainly with the vasculature, while in male placentas, disturbed expression predominantly affected immune processes.
APA, Harvard, Vancouver, ISO, and other styles
31

Joachims, M. L., B. Khatri, K. L. Tessneer, A. M. Stolarczyk, G. B. Wiley, A. Rasmussen, J. Guthridge, et al. "OP0140 DYSREGULATED EXPRESSION OF THE LONG NON-CODING RNA, LINC01871, IMPLICATED IN SJÖGREN’S SYNDROME PATHOGENESIS." Annals of the Rheumatic Diseases 79, Suppl 1 (June 2020): 90.1–90. http://dx.doi.org/10.1136/annrheumdis-2020-eular.3950.

Full text
Abstract:
Background:Sjögren’s syndrome (SS) is a chronic, heterogenous autoimmune disease characterized by inflammatory destruction of the exocrine glands. Long non-coding RNAs (lncRNAs) have emerged as a functionally diverse class of non-protein coding RNA (ncRNA) with increasing implications in interferon signaling, immune cell regulation, and autoimmune disease pathology. The potential role of lncRNAs in SS pathogenesis is unknown.Objectives:To identify and characterize candidate lncRNAs with potential relevance to SS pathology.Methods:RNA-seq was used on whole blood from SS patients (n=30 antibody negative (Ro-); n=27 antibody positive (Ro+)) and healthy controls (HC, n=27) to identify differentially expressed (DE) lncRNAs (log2 fold change (FC) ≥ 2 or ≤ 0.5; padj<0.05). Bioinformatic and pathway analyses were used to predict lncRNA function.In vitrotime course experiments in HSB2 T cell lymphoblasts stimulated with PMA/Ionomycin (PMA/I) or type I interferon (IFN) were used to assess biological relevance.LINC01871function was further investigated by RNA-seq on a single cell clone of HSB2 with confirmed CRISPR-targetedLINC01871deletion (LINC01871-/-).Results:We identified a total of 1054 unique DE ncRNAs between Ro+, Ro-and/or a combined analysis relative to HC; of these, 45 (1 long intergenic ncRNA (lincRNA), 1 antisense, 43 pseudogenes) were overexpressed in all 3 SS subsets. To begin investigating the function of the previously undescribed lincRNA,LINC01871(SSRo-: FC=2.85; padj=1.1x10-4), we performed a correlation analysis of the SSRo-transcriptome, which found several co-expressed protein coding RNAs involved in immune regulation (THEMIS,TBX21,IL10RA,IL2RB,among many others). Similarly, Ingenuity Pathway Analysis of the SS transcriptome compared to HC, as well as several gene ontology enrichment analyses of publicly available RNA expression correlation databases, identified shared immune-related pathways including cytotoxic T cell, natural killer cell, and T cell regulation. To further study the role ofLINC01871in cytotoxic T cells, we used qRT-PCR to resolve the effects of PMA/I or type I IFN stimulation onLINC01871expression in the T lymphoblastoid HSB2 cells.LINC01871expression was downregulated after PMA/I stimulation, but unchanged with type I IFN stimulation. To explore the regulatory function ofLINC01871in T cells, we targetedLINC01871in HSB2 cells using CRISPR. To this end, we generated a single cellLINC01871-/-clone with no RNA expression by qPCR and confirmed homozygous deletion using DNA sequencing. RNA-seq analysis ofLINC01871-/-compared to unmodified HSB2 cells identified 1166 DE transcripts. Pathway analyses clustered the DE transcripts into similar immune regulatory, cytotoxic and T cell pathways identified in SSRo-whole blood RNA-seq and publicly available RNA-seq databases. Further, several prominent T cell regulatory transcripts that exhibited correlated upregulation withLINC01871in SSRo-whole blood RNA-seq also demonstrated downregulation afterLINC01871deletion:CD109(FC=-9.7; padj=5.3x10-16),IL22(FC=-8.1; padj=7.6x10-11),PDCD1(FC=-6.2; padj=1.1x10-6),THEMIS(FC=-3.8; padj=2.7x10-165) andTBX21(FC=-2.1; padj=3.3x10-25).Conclusion:LncRNAs are emerging as important regulators of immune function with increasing evidence of autoimmune disease relevance. Here, we leveraged RNA-seq, extensive bioinformatic data, and CRISPR technology to identify and functionally characterizeLINC01871as a potential mediator of the dysregulated T cell inflammatory response pathways implicated in SS pathogenesis.Disclosure of Interests:Michelle L Joachims: None declared, Bhuwan Khatri: None declared, Kandice L Tessneer: None declared, Anna M Stolarczyk: None declared, Graham B Wiley: None declared, Astrid Rasmussen Speakers bureau: Novartis, ThermoFischer, Joel Guthridge Grant/research support from: Xencor, Bristol Myers Squibb, DXterity, Judith A. James Grant/research support from: Progentec Diagnostics, Inc, Consultant of: Abbvie, Novartis, Jannsen, R Hal Scofield Grant/research support from: Pfizer, Kathy L Sivils: None declared, Indra Adrianto: None declared, Christopher Lessard: None declared
APA, Harvard, Vancouver, ISO, and other styles
32

Wang, Jian, Hui-Zeng Sun, Eóin O’Hara, Hong Chen, and Leluo Guan. "20 Profiling circRNA and lncRNA Expression in Key Bovine Metabolic Tissues." Journal of Animal Science 99, Supplement_3 (October 8, 2021): 9. http://dx.doi.org/10.1093/jas/skab235.016.

Full text
Abstract:
Abstract CircRNAs and lncRNAs are non-coding RNAs that regulate many biological processes at the cellular level. However, knowledge of their function and expression patterns in cattle remains scarce. We investigated circRNA and lncRNA expression profiles in four key metabolic tissues of beef cattle (rumen, liver, muscle, and subcutaneous adipose) using RNA-Seq. Bioinformatic analysis of 189 samples collected from 48 cattle identified 19,766 circRNAs and 10,615 lncRNAs across all four tissues. PCA revealed the expression profiles of both circ- and lncRNA were clearly separated by tissue, suggesting their expression patterns are tissue dependent. Moreover, both datasets contained large numbers of transcripts that were unique to each tissue, underlining the divergence in function across metabolic sites. Further functional analysis of tissue specific circRNAs using the Kyoto Encyclopedia of Genes and Genomes (KEGG) showed that lysine degradation, peroxisome, insulin resistance, and ubiquitin-mediated proteolysis unique to the rumen, liver, muscle, and adipose tissues, respectively. The same analysis of the lncRNAs revealed that purine metabolism, fatty acid degradation, cAMP signaling pathway, and AGE-RAGE signaling pathway in diabetic complications were uniquely enriched in the rumen, liver, muscle, and subcutaneous adipose tissues, respectively. Together, these results revealed tissue specific metabolic pathways that may be regulated by the multiple non-coding RNAs. More specifically, circDLG1 and lncMSTRG.12092.8 were predicted to regulate the expression of Lymphocyte Antigen 6 Family Member G5B (LY6G5B) and pyruvate kinase M1/2 (PKM), Adenylate Kinase 1 (AK1), and Mitofusin 2 (MFN2), respectively, indicating regulatory roles in host immune response, glycolysis, energy balance control, and mitochondrial fusion, all of which may contribute to regulation of energy metabolism. Our findings show that expression profiles of circRNAs and lncRNAs differ among metabolic tissues, and these transcripts may play crucial roles in regulating metabolism in cattle.
APA, Harvard, Vancouver, ISO, and other styles
33

Gao, Yubang, Feihu Xi, Hangxiao zhang, Xuqing Liu, Huiyuan Wang, Liangzhen zhao, Anireddy S. N. Reddy, and Lianfeng Gu. "Single-molecule Real-time (SMRT) Isoform Sequencing (Iso-Seq) in Plants: The Status of the Bioinformatics Tools to Unravel the Transcriptome Complexity." Current Bioinformatics 14, no. 7 (September 17, 2019): 566–73. http://dx.doi.org/10.2174/1574893614666190204151746.

Full text
Abstract:
Background: The advent of the Single-Molecule Real-time (SMRT) Isoform Sequencing (Iso-Seq) has paved the way to obtain longer full-length transcripts. This method was found to be much superior in identifying full-length splice variants and other post-transcriptional events as compared to the Next Generation Sequencing (NGS)-based short read sequencing (RNA-Seq). Several different bioinformatics tools to analyze the Iso-Seq data have been developed and some of them are still being refined to address different aspects of transcriptome complexity. However, a comprehensive summary of the available tools and their utility is still lacking. Objective: Here, we summarized the existing Iso-Seq analysis tools and presented an integrated bioinformatics pipeline for Iso-Seq analysis, which overcomes the limitations of NGS and generates long contiguous Full-Length Non-Chimeric (FLNC) reads for the analysis of posttranscriptional events. Results: In this review, we summarized recent applications of Iso-Seq in plants, which include improved genome annotations, identification of novel genes and lncRNAs, identification of fulllength splice isoforms, detection of novel Alternative Splicing (AS) and Alternative Polyadenylation (APA) events. In addition, we also discussed the bioinformatics pipeline for comprehensive Iso-Seq data analysis, including how to reduce the error rate in the reads and how to identify and quantify post-transcriptional events. Furthermore, the visualization approach of Iso-Seq was discussed as well. Finally, we discussed methods to combine Iso-Seq data with RNA-Seq for transcriptome quantification. Conclusion: Overall, this review demonstrates that the Iso-Seq is pivotal for analyzing transcriptome complexity and this new method offers unprecedented opportunities to comprehensively understand transcripts diversity.
APA, Harvard, Vancouver, ISO, and other styles
34

Zafar, Junaid, Junlin Huang, Xiaoxia Xu, and Fengliang Jin. "Recent Advances and Future Potential of Long Non-Coding RNAs in Insects." International Journal of Molecular Sciences 24, no. 3 (January 30, 2023): 2605. http://dx.doi.org/10.3390/ijms24032605.

Full text
Abstract:
Over the last decade, long non-coding RNAs (lncRNAs) have witnessed a steep rise in interest amongst the scientific community. Because of their functional significance in several biological processes, i.e., alternative splicing, epigenetics, cell cycle, dosage compensation, and gene expression regulation, lncRNAs have transformed our understanding of RNA’s regulatory potential. However, most knowledge concerning lncRNAs comes from mammals, and our understanding of the potential role of lncRNAs amongst insects remains unclear. Technological advances such as RNA-seq have enabled entomologists to profile several hundred lncRNAs in insect species, although few are functionally studied. This article will review experimentally validated lncRNAs from different insects and the lncRNAs identified via bioinformatic tools. Lastly, we will discuss the existing research challenges and the future of lncRNAs in insects.
APA, Harvard, Vancouver, ISO, and other styles
35

Weiss, Mitchell J. "Role of Long Coding RNAs in Epigenetic Modulation of Hematopoiesis." Blood 122, no. 21 (November 15, 2013): SCI—28—SCI—28. http://dx.doi.org/10.1182/blood.v122.21.sci-28.sci-28.

Full text
Abstract:
Abstract Long noncoding (Lnc) RNAs are RNA transcripts greater than 200 nucleotides (nt) that regulate gene expression independent of protein coding potential (1-3). It is estimated that thousands of lncRNAs play vital roles in diverse cellular processes. LncRNAs modulate many stages of gene expression by regulating transcription, epigenetics, splicing, translation, and protein localization. We hypothesize that multiple lncRNAs are expressed specifically during erythrocyte and megakaryocyte differentiation, and are likely to have important roles. To identify lncRNAs in erythro-megakaryopoiesis, we performed strand-specific, paired-end deep sequencing (RNA-Seq) to a depth of 200 million reads per sample on two replicates each of murine Ter119+erythroblasts, CD41+ megakaryocytes and bipotential megakaryocyte-erythroid progenitors (MEPs) [lin- Kit+ Sca1- CD16/32- CD34-], and used bioinformatic filtering tools to identify approximately 1,100 candidate lncRNAs. Over 60 percent of these lncRNAs are novel unannotated transcripts with exquisite lineage-specific expression. Using erythroid and megakaryocytic primary cell ChIP-Seq for key transcription factors (TFs) GATA1, TAL1, GATA2,and FLI1, we found that the loci of lncRNAs show similar degree of TF binding as coding genes. We used the erythroid line G1E-ER4 (which expresses estrogen-activated GATA1) to confirm that lncRNAs bound by GATA1 are also directly regulated by it. Furthermore, we used histone methylation ChIP-Seq to show that most lncRNAs arise from classical “promoters” with high H3K4me3 levels and low H3K4me1 levels. Thus, we find that lncRNAs show epigenetic features similar to the promoters of coding genes and are directly regulated by similar TF networks. Comparison of the transcriptomes of mouse fetal liver and human cord blood erythroblasts demonstrated that lncRNAs are expressed in a highly species-specific fashion, i.e., most lncRNAs identifiable in one species are not transcribed in the other, even though the corresponding genomic region is present in both species. Numerous non-conserved but functional lncRNAs are reported in the literature, and the significance of conservation in lncRNA biology is greatly debated. In order to identify functional lncRNAs, we are currently performing RNAi knockdown on numerous candidates to assess how loss of function affects erythroid maturation. We are also performing HITS-CLIP of key chromatin modifying complexes and erythroid transcription factors to identify lncRNAs bound to them. Our studies are beginning to define new layers of gene regulation in normal erythro-megakaryopoiesis, which may be relevant to the pathophysiology of related disorders including various anemias, myeloproliferative and myelodysplastic syndromes and leukemias. 1. Wang K.C., Chang H.Y. Molecular mechanisms of long noncoding RNAs. Molecular Cell. 2011;43(6):904-914. Prepublished on 2011/09/20 as DOI 10.1016/j.molcel.2011.08.018. 2. Hu W., Alvarez-Dominguez J.R., Lodish H.F. Regulation of mammalian cell differentiation by long non-coding RNAs. EMBO reports. 2012;13(11):971-983. Prepublished on 2012/10/17 as DOI 10.1038/embor.2012.145. 3. Paralkar V.R., Weiss M.J. Long noncoding RNAs in biology and hematopoiesis. Blood. 2013;121(24):4842-4846. Prepublished on 2013/05/07 as DOI 10.1182/blood-2013-03-456111. Disclosures: No relevant conflicts of interest to declare.
APA, Harvard, Vancouver, ISO, and other styles
36

Tao, Yifan, Siqi Lu, Tao Zheng, Mingxiao Li, Jun Qiang, and Pao Xu. "RNA-Seq Analysis of the Key Long Noncoding RNAs and mRNAs Related to the Regulation of Hepatic Lipid Metabolism in Oreochromis niloticus." Fishes 7, no. 6 (November 11, 2022): 332. http://dx.doi.org/10.3390/fishes7060332.

Full text
Abstract:
Genetically improved farmed tilapia (Oreochromis niloticus, GIFT) is prone to hepatic metabolic imbalances and fatty liver disease during intensive farming. Long non-coding RNAs (lncRNAs) perform essential roles in various biological processes, including lipid metabolism. However, the lncRNAs involved in hepatic lipid metabolism in tilapia have not yet been identified. In this study, Illumina sequencing and bioinformatic analyses were performed on the liver of juvenile male GIFT fed a high-fat diet (HFD, 18.5% lipid) or a normal-fat diet (NFD, 8% lipid) for 56 days. RNA-seq analyses revealed 299 differentially expressed (DE)-mRNAs and 284 DE-lncRNAs between these two groups. The transcript profiles of 14 candidates (seven DE-mRNA and seven DE-lncRNAs) were verified by qRT-PCR, and the results were consistent with the RNA-seq results. Furthermore, 65 cis target genes and 3610 trans target genes of DE-lncRNAs were predicted. Functional analyses suggested that multiple metabolic pathways are affected by a high fat intake, including the PPAR signaling, fatty acid degradation, and fatty acid metabolism pathways. A co-expression network analysis indicated that many lncRNAs interact with numerous genes involved in lipid metabolism, and that some genes are regulated by multiple lncRNAs. The expression patterns of three lncRNAs (MSTRG.14598.1, MSTRG.6725.3, and MSTRG.13364.2) and their potential target genes (faldh, slc25a48, and fabp7a) in the PPAR signaling pathway were investigated. Our study provides new information about lncRNAs associated with lipid metabolism in tilapia.
APA, Harvard, Vancouver, ISO, and other styles
37

Deng, Keyong, Xiaotong Ning, Xiaoxiao Ren, Bin Yang, Jianxin Li, Jie Cao, Jichun Chen, Xiangfeng Lu, Shufeng Chen, and Laiyuan Wang. "Transcriptome-wide N6-methyladenosine methylation landscape of coronary artery disease." Epigenomics 13, no. 10 (May 2021): 793–808. http://dx.doi.org/10.2217/epi-2020-0372.

Full text
Abstract:
Aim: To reveal transcriptome-wide N6-methyladenosine (m6A) methylome of coronary artery disease (CAD). Materials & methods: The m6A levels of RNA from peripheral blood mononuclear cells measured by colorimetry were significantly decreased in CAD cases. Transcriptome-wide m6A methylome profiled by methylated RNA immunoprecipitation sequencing (MeRIP-seq) identified differentially methylated m6A sites within both mRNAs and lncRNAs between CAD and control group. Results: Bioinformatic analysis indicated that differentially methylated genes were involved in the pathogenesis of atherosclerosis. MeRIP-quantitative real-time PCR assay confirmed the reliability of MeRIP-seq data. Finally, the rat carotid artery balloon injury model was performed to confirm the role of m6A demethylase FTO in neointima formation. Conclusion: Our study provided a resource of differentially methylated m6A profile for uncovering m6A biological functions in the pathogenesis of CAD.
APA, Harvard, Vancouver, ISO, and other styles
38

Pashler, Amy L., Benjamin P. Towler, Christopher I. Jones, Hope J. Haime, Tom Burgess, and Sarah F. Newbury. "Genome-wide analyses of XRN1-sensitive targets in osteosarcoma cells identify disease-relevant transcripts containing G-rich motifs." RNA 27, no. 10 (July 15, 2021): 1265–80. http://dx.doi.org/10.1261/rna.078872.121.

Full text
Abstract:
XRN1 is a highly conserved exoribonuclease which degrades uncapped RNAs in a 5′–3′ direction. Degradation of RNAs by XRN1 is important in many cellular and developmental processes and is relevant to human disease. Studies in D. melanogaster demonstrate that XRN1 can target specific RNAs, which have important consequences for developmental pathways. Osteosarcoma is a malignancy of the bone and accounts for 2% of all pediatric cancers worldwide. Five-year survival of patients has remained static since the 1970s and therefore furthering our molecular understanding of this disease is crucial. Previous work has shown a down-regulation of XRN1 in osteosarcoma cells; however, the transcripts regulated by XRN1 which might promote osteosarcoma remain elusive. Here, we confirm reduced levels of XRN1 in osteosarcoma cell lines and patient samples and identify XRN1-sensitive transcripts in human osteosarcoma cells. Using RNA-seq in XRN1-knockdown SAOS-2 cells, we show that 1178 genes are differentially regulated. Using a novel bioinformatic approach, we demonstrate that 134 transcripts show characteristics of direct post-transcriptional regulation by XRN1. Long noncoding RNAs (lncRNAs) are enriched in this group, suggesting that XRN1 normally plays an important role in controlling lncRNA expression in these cells. Among potential lncRNAs targeted by XRN1 is HOTAIR, which is known to be up-regulated in osteosarcoma and contributes to disease progression. We have also identified G-rich and GU motifs in post-transcriptionally regulated transcripts which appear to sensitize them to XRN1 degradation. Our results therefore provide significant insights into the specificity of XRN1 in human cells which are relevant to disease.
APA, Harvard, Vancouver, ISO, and other styles
39

Sun, Yi, Yeying Wen, Qishuang Ruan, Le Yang, Shuna Huang, Xingyan Xu, Yingying Cai, Huangyuan Li, and Siying Wu. "Exploring the association of long noncoding RNA expression profiles with intracranial aneurysms, based on sequencing and related bioinformatics analysis." BMC Medical Genomics 13, no. 1 (October 6, 2020). http://dx.doi.org/10.1186/s12920-020-00805-x.

Full text
Abstract:
Abstract Background The present study aims to investigate the complete long non-coding RNA (lncRNA) and messenger RNA (mRNA) expression profiles in Intracranial aneurysm (IA) patients and controls by RNA sequencing, which reveals the lncRNA with predictive value for IA risk. Methods The comprehensive lncRNA and mRNA expression profiles were detected by RNA-Seq in human IA walls and superficial temporal arteries (STAs), followed by bioinformatics analyses, such as GO analysis, KEGG pathway analysis, and CNC network construction. Subsequently, qRT-PCR was used to profile the expression levels of selected lncRNA (lncRNA ENST000000576153, lncRNA ENST00000607042, lncRNA ENST00000471220, lncRNA ENST00000478738, lncRNA MALAT1, lncRNA ENST00000508090 and lncRNA ENST00000579688) in 30 (small) or 130 (large) peripheral blood leukocytes, respectively. Multivariate logistic regression was utilized to analyze the effects of lncRNA on IA. Receiver operating characteristic (ROC) curve was further drawn to explore the value of lncRNA in predicting IA. Results Totally 900 up-regulated and 293 down-regulated lncRNAs, as well as 1297 up-regulated and 831 down-regulated mRNAs were discovered in sequencing. Enrichment analyses revealed that they were actively involved in immune/inflammatory response and cell adhesion/extracellular matrix. Co-expression analysis and further enrichment analyses showed that five candidate lncRNAs might participate in IA’s inflammatory response. Besides, after controlling other conventional risk factors, multivariate logistic regression analysis disclosed that low expression of lncRNA ENST00000607042, lncRNA ENST00000471220, lncRNA ENST00000478738, lncRNA MALAT1 in peripheral blood leukocytes were independent risk factors for IA. LncRNA ENST00000607042 has superior diagnostic value for IA. Conclusions This study reveals the complete lncRNAs expression profiles in IA. The inflammatory response was closely related to IA. Besides, lncRNA ENST00000607042 might be a novel biomarker for IA risk.
APA, Harvard, Vancouver, ISO, and other styles
40

Li, Hui-hui, Lin-tao Sai, Yuan Liu, Colman I. Freel, Kai Wang, Chi Zhou, Jing Zheng, Qiang Shu, and Ying-jie Zhao. "Systemic lupus erythematosus dysregulates the expression of long noncoding RNAs in placentas." Arthritis Research & Therapy 24, no. 1 (June 14, 2022). http://dx.doi.org/10.1186/s13075-022-02825-7.

Full text
Abstract:
Abstract Background Systemic lupus erythematosus (SLE) can cause placental dysfunctions, which may result in pregnancy complications. Long noncoding RNAs (lncRNAs) are actively involved in the regulation of immune responses during pregnancy. The present study aimed to determine the lncRNA expression profiles in placentas from women with SLE to gain new insights into the underlying molecular mechanisms in SLE pregnancies. Methods RNA sequencing (RNA-seq) analysis was performed to identify SLE-dysregulated lncRNAs and mRNAs in placentas from women with SLE and normal full-term (NT) pregnancies. Bioinformatics analysis was conducted to predict the biological functions of these SLE-dysregulated lncRNAs and mRNAs. Results RNA-seq analysis identified 52 dysregulated lncRNAs in SLE placentas, including 37 that were upregulated and 15 downregulated. Additional 130 SLE-dysregulated mRNAs were discovered, including 122 upregulated and 8 downregulated. Bioinformatics analysis revealed that SLE-dysregulated genes were associated with biological functions and gene networks, such as regulation of type I interferon-mediated signaling pathway, response to hypoxia, regulation of MAPK (mitogen-activated protein kinase) cascade, response to steroid hormone, complement and coagulation cascades, and Th1 and Th2 cell differentiation. Conclusions This is the first report of the lncRNA profiles in placentas from SLE pregnancies. These results suggest that the aberrant expression and the potential regulatory function of lncRNAs in placentas may play comprehensive roles in the pathogenesis of SLE pregnancies. SLE-dysregulated lncRNAs may potentially serve as biomarkers for SLE.
APA, Harvard, Vancouver, ISO, and other styles
41

Huang, Rong, and Li-Jing Sun. "Identification of circulating lncRNA in chronic kidney disease based on bioinformatics analysis." Experimental Biology and Medicine, June 25, 2022, 153537022211040. http://dx.doi.org/10.1177/15353702221104035.

Full text
Abstract:
Chronic kidney disease (CKD) is a high mortality disease and generally remains asymptomatic in the early stages. Long non-coding RNA (lncRNA) is defined as a non-protein-coding transcript more than 200 nucleotides which participate in numerous biological processes and have been identified as novel diagnostic markers for many diseases. Detection of circulating lncRNAs is a rapidly evolving, new area of molecular diagnosis. The purpose of our research was to identify circulating lncRNA expression profiles and possible molecular mechanisms involved in CKD. Blood samples were obtained from patients with CKD and healthy volunteers, and high-throughput sequencing was performed to identify differentially expressed (DE) lncRNAs and mRNAs. DE lncRNAs and mRNAs in peripheral blood mononuclear cells (PBMCs) were confirmed by quantitative reverse transcription polymerase chain reaction (qRT-PCR) to ensure the reliability and validity of RNA-seq data. Bioinformatics analysis was used to obtain biological functions and key pathways related to the pathogenesis of CKD. The interaction and co-expression functional networks for DE lncRNAs and mRNAs were also constructed. Our data showed that of the 425 DE lncRNAs detected, 196 lncRNAs were upregulated, while that of 229 lncRNAs were downregulated. A total of 433 DE mRNAs were identified in patients with CKD compared to healthy individuals. GO analysis revealed that DE lncRNAs were highly correlated with binding and pathway regulation. KEGG analysis suggested that DE lncRNAs were obviously enriched in regulatory pathways, such as antigen processing and presentation. We successfully constructed a potential DE lncRNA-mRNA co-expression network and analyzed the target genes of DE lncRNAs to predict cis- and trans-regulation in CKD. 100 lncRNAs that corresponded to 14 transcription factors (TFs) were identified in the TF-lncRNA binary network. Our findings on the lncRNA expression profiles and functional networks may help to interpret the possible molecular mechanisms implied in the pathogenesis of CKD; the results demonstrated that lncRNAs could potentially to be used as diagnostic biomarkers in CKD.
APA, Harvard, Vancouver, ISO, and other styles
42

Shi, Xin, Panpan Li, Xiang Wu, Zhihua Wang, Gang Zhao, and Jun Shu. "RNA-Seq Comprehensive Analysis Reveals the Long Noncoding RNA Expression Profile and Coexpressed mRNA in Adult Degenerative Scoliosis." Frontiers in Genetics 13 (August 11, 2022). http://dx.doi.org/10.3389/fgene.2022.902943.

Full text
Abstract:
Objective: Owing to the intensification of the aging process worldwide, the prevalence of adult degenerative scoliosis (ADS) is increasing at an alarming rate. However, genomic research related to the etiology of ADS is rarely reported worldwide. Since long noncoding RNAs (lncRNAs) play a pivotal role in the progression of human diseases, this study aimed to investigate ADS-associated messenger RNAs (mRNAs) and lncRNAs by RNA sequencing (RNA-seq), as well as performed comprehensive bioinformatics analysis based on the lncRNA–mRNA coexpression network and protein–protein interaction (PPI) network.Methods: Initially, six whole blood (WB) samples were obtained from three ADS and three nondegenerative lumbar trauma patients who underwent surgical operation for RNA-seq exploration to construct differential mRNA and lncRNA expression profiles. Subsequently, quantitative RT-PCR (qRT-PCR) was performed to validate three randomly selected differentially expressed mRNAs and lncRNAs derived from the nucleus pulposus (NP) tissue of 14 other subjects (seven ADS patients and seven nondegenerative lumbar trauma patients), respectively.Results: A total of 1,651 upregulated and 1,524 downregulated mRNAs and 147 upregulated and 83 downregulated lncRNAs were screened out from the RNA-Seq data, which constructed coexpression networks to investigate their regulatory interactions further. GO gene function prediction revealed that lncRNA-targeted genes might play a vital role in ADS via participation in multiple biological processes such as the AMPK signaling pathway, lysosomes, and ubiquitin-mediated proteolysis, as well as cellular metabolic processes. Moreover, the expression levels of three selected lncRNAs and mRNAs were validated by qRT-PCR, respectively, demonstrating that the relative expression levels were consistent with the RNA-seq data. Notably, the dysregulated RNAs, AKT1, UBA52, PTPN12, and CLEC16A, were significantly differentially expressed in ADS WB samples and might serve as potentially regulated genes for research in the future.Conclusions: This study provides the first insight into the altered transcriptome profile of long-stranded noncoding RNAs associated with ADS, which paves the way for further exploration of the clinical biomarkers and molecular regulatory mechanisms for this poorly understood degenerative disease. However, the detailed biological mechanisms underlying these candidate lncRNAs in ADS necessitate further elucidation in future studies.
APA, Harvard, Vancouver, ISO, and other styles
43

Sun, Yuxiu, Chen Li, Qingyi Lu, Haixu Jiang, Mengmeng Zhu, Guangrui Huang, and Ting Wang. "Integrative Analysis of lncRNA-mRNA Profile Reveals Potential Predictors for SAPHO Syndrome." Frontiers in Genetics 12 (June 21, 2021). http://dx.doi.org/10.3389/fgene.2021.684520.

Full text
Abstract:
Synovitis, acne, pustulosis, hyperostosis, and osteitis (SAPHO) syndrome is known as a rare disease characterized by inflammatory lesions on bones and skin. Polymorphism of clinical manifestation and lack of molecular biomarkers have both limited its diagnosis. Our study performed RNA sequencing (RNA-seq) and integrative bioinformatics analysis of long noncoding RNA (lncRNA)-messenger RNA (mRNA) profile in patients with SAPHO syndrome and healthy controls. A total of 4,419 differentially expressed (DE) mRNAs and 2,713 lncRNAs were identified (p &lt; 0.05, fold change &gt; 2) and a coexpression network was constructed to further investigate their regulatory interactions. The DE lncRNAs were predicted to interact with mRNAs in both cis and trans manners. Functional prediction found that the lncRNA-targeted genes may function in SAPHO syndrome by participating in biological process such as adipocytokine signaling pathway, ErbB signaling pathway, FoxO signaling pathway, as well as production and function of miRNAs. The expression levels of three pairs of coexpressed lncRNA-mRNAs were validated by qRT-PCR, and their relative expression levels were consistent with the RNA-seq data. The deregulated RNAs GAS7 and lnc-CLLU1.1-1:2 may serve as potential diagnostic biomarkers, and the combined receiver operating characteristic (ROC) curve of the two showed more reliable diagnostic ability with an AUC value of 0.871 in distinguishing SAPHO patients from healthy controls. In conclusion, this study provides a first insight into long noncoding RNA transcriptome profile changes associated with SAPHO syndrome and inspiration for further investigation on clinical biomarkers and molecular regulators of this inadequately understood clinical entity.
APA, Harvard, Vancouver, ISO, and other styles
44

Hu, Huiyan, Qing Jia, Jianzhong Xi, Bo Zhou, and Zhiqiang Li. "Integrated analysis of lncRNA, miRNA and mRNA reveals novel insights into the fertility regulation of large white sows." BMC Genomics 21, no. 1 (September 14, 2020). http://dx.doi.org/10.1186/s12864-020-07055-2.

Full text
Abstract:
Abstract Background Improving sow fertility is extremely important as it can lead to increased reproductive efficiency and thus profitability for swine producers. There are considerable differences in fertility rates among individual animals, but the underlying molecular mechanisms remain unclear. In this study, by using different types of RNA libraries, we investigated the complete transcriptome of ovarian tissue during the luteal (L) and follicular (F) phases of the estrous cycle in Large White pigs with high (H) and low (L) fecundity, and performed a comprehensive analysis of long noncoding RNAs (lncRNAs), mRNAs and micro RNAs (miRNAs) from 16 samples by combining RNA sequencing (RNA-seq) with bioinformatics. Results In total, 24,447 lncRNAs, 27,370 mRNAs, and 216 known miRNAs were identified in ovarian tissues. The genomic features of lncRNAs, such as length distribution and number of exons, were further analyzed. We selected a threshold of P < 0.05 and |log2 (fold change)| ≥ 1 to obtain the differentially expressed lncRNAs, miRNAs and mRNAs by pairwise comparison (LH vs. LL, FH vs. FL). Bioinformatics analysis of these differentially expressed RNAs revealed multiple significantly enriched pathways (P < 0.05) that were closely involved in the reproductive process, such as ovarian steroidogenesis, lysosome, steroid biosynthesis, and the estrogen and GnRH signaling pathways. Moreover, bioinformatics screening of differentially expressed miRNAs that share common miRNA response elements (MREs) with lncRNAs and their downstream mRNA targets were performed. Finally, we constructed lncRNA–miRNA–mRNA regulation networks. The key genes in these networks were verified by Reverse Transcription Real-time Quantitative PCR (RT-qRCR), which were consistent with the results from RNA-Seq data. Conclusions These results provide further insights into the fertility of pigs andcan contribute to further experimental investigation of the functions of these genes.
APA, Harvard, Vancouver, ISO, and other styles
45

Mei, Bujun, and Rong Liu. "Identification of lncRNAs Differentially Expressed during Natural and Induced Estrus in Sheep." Indian Journal of Animal Research, Of (August 21, 2021). http://dx.doi.org/10.18805/ijar.b-1347.

Full text
Abstract:
Background: The manipulation of the estrous cycle or induction of estrus is a commonly used technique in sheep industry. The goal of this study was to identify and characterize differences of non-coding RNAs (lincRNAs) expression between induced estrus and natural estrus using the BGISEQ-500 plat form in 7 Mongolian sheep, which will provide insights into the regulation mechanisms of lncRNAs in different reproduction mode of sheep. Methods: During the late spring, ovarian, pituitary, hypothalamic, pineal and uterine tissue samples were collected from four artificially induced estrus and three naturally estrus Mongolian sheep. Total RNA was extracted from the five tissues using TRIzol reagent (Invitrogen) and treated with DNase I following the manufacturer’s instructions. A total of 35 sheep samples were sequenced using the BGISEQ-500 plat form. Bioinformatics methods were used to analysis expression difference analysis between groups, SNP and InDel, alternative splicing, lncRNA’s miRNA precursor prediction, lncRNA target gene and family prediction. Result: 211 novel lncRNAs were systematically identified using RNA-Seq technology. Meanwhile, we found that there are diversifications of lncRNAs in induced estrus vs. nature estrus of ewes. Therefore, we predict that, under the action of exogenous hormones, many physiological processes of ewes may be affected to varying degrees through the change of LncRNA to a variety of pathways.
APA, Harvard, Vancouver, ISO, and other styles
46

Liu, Tianyi, Hui Feng, Salsabeel Yousuf, Lingli Xie, and Xiangyang Miao. "Differential regulation of mRNAs and lncRNAs related to lipid metabolism in Duolang and Small Tail Han sheep." Scientific Reports 12, no. 1 (July 1, 2022). http://dx.doi.org/10.1038/s41598-022-15318-z.

Full text
Abstract:
AbstractThe function of long non-coding RNA (lncRNA) can be achieved through the regulation of target genes, and the deposition of fat is regulated by lncRNA. Fat has an important effect on meat quality. However, there are relatively few studies on lncRNAs in the subcutaneous adipose tissue of Duolang sheep and Small Tail Han sheep. In this study, RNA-Seq technology and bioinformatics methods were used to identify and analyze the lncRNA and mRNA in the subcutaneous adipose tissue of the two breeds of sheep. The results showed that 107 lnRNAs and 1329 mRNAs were differentially expressed. The differentially expressed genes and lncRNA target genes were significantly enriched in the biosynthesis of unsaturated fatty acids signaling pathway, fatty acid metabolism, adipocyte differentiation and other processes related to fat deposition. Among them, LOC105616076, LOC114118103, LOC105607837, LOC101116622, and LOC105603235 target FADS1, SCD, ELOVL6, HSD17B12 and HACD2, respectively. They play a key regulatory role in the biosynthesis of unsaturated fatty acids. This study lays a foundation for the study of the molecular mechanism of lncRNA on fat development, and has reference value for studying the differences in fat deposition between Duolang sheep and Small Tail Han sheep.
APA, Harvard, Vancouver, ISO, and other styles
47

Tang, Wen, Qing Liu, Wei Tan, Tianshi Sun, and Youwen Deng. "LncRNA expression profile analysis of Mg2+-induced osteogenesis by RNA-seq and bioinformatics." Genes & Genomics, August 24, 2021. http://dx.doi.org/10.1007/s13258-021-01140-w.

Full text
APA, Harvard, Vancouver, ISO, and other styles
48

Zeng, Junkai, Ming Chen, Yeqing Yang, and Buling Wu. "A novel hypoxic lncRNA, HRL-SC, promotes the proliferation and migration of human dental pulp stem cells through the PI3K/AKT signaling pathway." Stem Cell Research & Therapy 13, no. 1 (June 28, 2022). http://dx.doi.org/10.1186/s13287-022-02970-5.

Full text
Abstract:
Abstract Background Human dental pulp stem cells (hDPSCs) are critical for pulp generation. hDPSCs proliferate faster under hypoxia, but the mechanism by which long noncoding RNA (lncRNA) regulates this process is not fully understood. Methods Novel lncRNAs were obtained by reanalysis of transcriptome datasets from RNA-Seq under hypoxia compared with normoxia, and a differential expression analysis of target genes was performed. Bioinformatics analyses, including gene ontology analysis, Kyoto Encyclopedia of Genes and Genomes pathway analysis and gene set enrichment analysis, were used to understand the function of key novel lncRNAs. hDPSCs were isolated from dental pulp tissue. EdU and scratch wound healing assays were used to detect the proliferation and migration of hDPSCs. qRT-PCR was used to detect changes in the RNA expression of selected genes. RNA fluorescence in situ hybridization, small interfering RNA, qRT-PCR and Western blot analysis were used to explore the function of key novel lncRNAs. Results We identified 496 novel lncRNAs in hDPSCs under hypoxia, including 45 differentially expressed novel lncRNAs. Of these, we focused on a key novel lncRNA, which we designated HRL-SC (hypoxia-responsive lncRNA in stem cells). Functional annotation revealed that HRL-SC was associated with hypoxic conditions and the PI3K/AKT signaling pathway. HRL-SC was mainly located in the cytoplasm of hDPSCs and had stable high expression under hypoxia. Knockdown of HRL-SC inhibited the proliferation and migration of hDPSCs and the expression levels of PI3K/AKT-related marker proteins. Furthermore, the AKT activator SC79 partially offset the inhibitory effect caused by the knockdown, indicating that HRL-SC promoted hDPSCs through the PI3K/AKT signaling pathway. Conclusions Hypoxia-responsive lncRNA HRL-SC promotes the proliferation and migration of hDPSCs through the PI3K/AKT signaling pathway, and this understanding may facilitate the regenerative application of hDPSCs.
APA, Harvard, Vancouver, ISO, and other styles
49

Azadeh, Mansoureh, Ali Salehzadeh, Kamran Ghaedi, and Soheila Talesh Sasani. "NEAT1 can be a diagnostic biomarker in the breast cancer and gastric cancer patients by targeting XIST, hsa-miR-612, and MTRNR2L8: integrated RNA targetome interaction and experimental expression analysis." Genes and Environment 44, no. 1 (May 17, 2022). http://dx.doi.org/10.1186/s41021-022-00244-3.

Full text
Abstract:
Abstract Background The most frequent malignancy in women is breast cancer (BC). Gastric cancer (GC) is also the leading cause of cancer-related mortality. Long non-coding RNAs (lncRNAs) are thought to be important neurotic regulators in malignant tumors. In this study, we aimed to evaluate the expression level of NEAT1 and the interaction of this non-coding RNA with correlated microRNAs, lncRNAs, and mRNAs or protein coding genes, experimentally and bioinformatically. Methods For the bioinformatics analyses, we performed RNA-RNA and protein–protein interaction analyses, using ENCORI and STRING. The expression analyses were performed by five tools: Microarray data analysis, TCGA data analysis (RNA-seq, R Studio), GEPIA2, ENCORI, and real-time PCR experiment. qRT-PCR experiment was performed on 50 GC samples and 50 BC samples, compared to adjacent control tissue. Results Based on bioinformatics and experimental analyses, lncRNA NEAT1 have a significant down-regulation in the breast cancer samples with tumor size lower than 2 cm. Also, it has a significant high expression in the gastric cancer patients. Furthermore, NEAT1 have a significant interaction with XIST, hsa-miR-612 and MTRNR2L8. High expression of NEAT1 have a correlation with the lower survival rate of breast cancer samples and higher survival rate of gastric cancer patients. Conclusion This integrated computational and experimental investigation revealed some new aspects of the lncRNA NEAT1 as a potential prognostic biomarker for the breast cancer and gastric cancer samples. Further investigations about NEA1 and correlated mRNAs, lncRNAs, and microRNAs – specially the mentioned RNAs in this study – can lead the researchers to more clear information about the role of NEAT1 in the breast cancer and gastric cancer.
APA, Harvard, Vancouver, ISO, and other styles
50

Liu, Jiandong, Xiangna Guo, Lu Yang, Tao Tao, Jun Cao, Zexuan Hong, Fanning Zeng, Yitian Lu, Chunshui Lin, and Zaisheng Qin. "Effect of Celastrol on LncRNAs and mRNAs Profiles of Cerebral Ischemia-Reperfusion Injury in Transient Middle Cerebral Artery Occlusion Mice Model." Frontiers in Neuroscience 16 (May 23, 2022). http://dx.doi.org/10.3389/fnins.2022.889292.

Full text
Abstract:
Celastrol plays a significant role in cerebral ischemia-reperfusion injury. Although previous studies have confirmed that celastrol post-treatment has a protective effect on ischemic stroke, the therapeutic effect of celastrol on ischemic stroke and the underlying molecular mechanism remain unclear. In the present study, focal transient cerebral ischemia was induced by transient middle cerebral artery occlusion (tMCAO) in mice and celastrol was administered immediately after reperfusion. We performed lncRNA and mRNA analysis in the ischemic hemisphere of adult mice with celastrol post-treatment through RNA-Sequencing (RNA-Seq). A total of 50 differentially expressed lncRNAs (DE lncRNAs) and 696 differentially expressed mRNAs (DE mRNAs) were identified between the sham and tMCAO group, and a total of 544 DE lncRNAs and 324 DE mRNAs were identified between the tMCAO and tMCAO + celastrol group. Bioinformatic analysis was done on the identified deregulated genes through gene ontology (GO) analysis, KEGG pathway analysis and network analysis. Pathway analysis indicated that inflammation-related signaling pathways played vital roles in the treatment of ischemic stroke by celastrol. Four DE lncRNAs and 5 DE mRNAs were selected for further validation by qRT-PCR in brain tissue, primary neurons, primary astrocytes, and BV2 cells. The results of qRT-PCR suggested that most of selected differentially expressed genes showed the same fold change patterns as those in RNA-Seq results. Our study suggests celastrol treatment can effectively reduce cerebral ischemia-reperfusion injury. The bioinformatics analysis of lnRNAs and mRNAs profiles in the ischemic hemisphere of adult mice provides a new perspective in the neuroprotective effects of celastrol, particularly with regards to ischemic stroke.
APA, Harvard, Vancouver, ISO, and other styles
We offer discounts on all premium plans for authors whose works are included in thematic literature selections. Contact us to get a unique promo code!

To the bibliography