Open Access

Construction of chronic glomerulonephritis‑related lncRNA‑mRNA regulatory network and lncRNA‑­miRNA‑mRNA ceRNA network by bioinformatics analysis

  • Authors:
    • Xing-Xing Zhuang
    • Tao Liu
    • Liang-Bing Wei
    • Jia-Rong Gao
  • View Affiliations

  • Published online on: July 7, 2023     https://doi.org/10.3892/etm.2023.12102
  • Article Number: 403
  • Copyright: © Zhuang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Long non‑coding RNAs (lncRNAs) are ncRNA transcripts >200 nucleotides that are important genetic regulators. LncRNAs can directly regulate mRNA through a lncRNA‑mRNA regulatory mode and can also regulate mRNA through competitive binding to micro (mi)RNA, which is generally known as the competitive endogenous RNA (ceRNA) network. The present study evaluated the functional roles and regulatory networks of lncRNAs in chronic glomerulonephritis (CGN). The proliferative ability of mouse glomerular mesangial cells (GMCs) induced by different concentrations of lipopolysaccharide (LPS) was assessed using the Cell Counting Kit‑8 assay, and RNA sequencing (RNA‑seq) was performed to identify differentially expressed lncRNAs in LPS‑induced GMCs. Based on the sequencing results, six lncRNAs were selected for validation using reverse transcription‑quantitative PCR (RT‑qPCR). Furthermore, the lncRNA‑mRNA regulatory network and the lncRNA‑miRNA‑mRNA ceRNA network were constructed to assess the role and mechanism of CGN‑related lncRNAs. To elucidate the biological functions of lncRNAs, Gene Ontology (GO) biological process term enrichment and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed on all mRNAs involved in the lncRNA‑mRNA regulatory network and in the ceRNA network. A total of 1,532 differentially expressed lncRNAs, including 594 upregulated lncRNAs and 938 downregulated lncRNAs, were identified using RNA‑seq. The results of RT‑qPCR validation were consistent with RNA‑seq results. An lncRNA‑mRNA regulatory network, including 236 lncRNAs and 556 mRNAs, and a ceRNA network, including 6 lncRNAs, 18 miRNAs and 419 mRNAs, were successfully constructed. The GO biological process term enrichment and KEGG pathway enrichment analyses demonstrated that those lncRNAs were often related to inflammatory response and substance metabolism. The present study identified key CGN‑related lncRNAs in LPS‑induced GMCs, and further demonstrated a global view of the lncRNA‑mRNA regulatory network and ceRNA network involved in CGN. These results offered novel insights into the roles of lncRNAs in the pathogenesis of CGN and identified potential diagnostic biomarkers.

Introduction

Chronic glomerulonephritis (CGN) is an autoimmune glomerulopathy, characterized by the excessive proliferation of mesangial cells, accumulation of extracellular matrix and infiltration of circulating inflammatory cells (1). Glomerular mesangial cells (GMCs), distributed in the mesangial matrix of the glomerulus, constitute the mesangial region of the glomerulus together with the mesangial matrix. GMCs have multiple physiological functions, including stabilizing the structure of glomerular capillaries, maintaining mesangial matrix homeostasis, regulating filtration surface area, phagocytosis of apoptotic cells and immune complexes (2). Under the influence of certain pathological factors such as hyperglycemia and inflammation, GMCs can proliferate abnormally, increase intracellular protein synthesis and, increase the secretion of the extracellular matrix (3-4). Therefore, excessive proliferation of GMCs is an important pathological feature of certain human kidney diseases, including chronic glomerulonephritis and diabetic nephropathies. Knowledge of the responses of GMCs to pathological stimuli is crucial to the understanding of the pathogenesis of chronic glomerulonephritis. Thus, a deeper understanding of the excessive proliferation of GMCs is required to devise more effective prevention and therapies for CGN.

Long non-coding RNAs (lncRNAs) are generally considered as non-coding transcripts >200 nucleotides that are important genetic regulators. LncRNAs have been previously reported to be crucial determinants of epigenetic regulation and serve a key role in regulation of chromatin structure, scaffolding or the decoy function of mRNAs, and post-transcriptional regulation of mRNAs (5). The regulatory role of lncRNA can be summarized as cis-action on neighboring genes or as trans-action through effects on mRNA stability, mRNA translation or the regulation of microRNA-mRNA interactions and RNA binding proteins (6-7). For example, Gao et al (8) reported that lncRNA NONRATG001910.2 promoted CTNNB1 expression by targeting miR-339-3p in lipopolysaccharide (LPS)-stimulated rat mesangial cells, which suggested that NONRATG001910.2 may be a potential biomarker for CGN. Moreover, Zhou et al (9) suggested that lncRNA NORAD directly regulated RUNX2 transcription and promoted the proliferation, migration and invasion of breast cancer cells. Competitive endogenous (ce)RNA is the most frequently reported mechanism of lncRNAs.

Materials and methods

GMC culture conditions

Mouse SV40-MES-13 GMCs were purchased from BNCC Biological Technology and cultured in DMEM (Beijing Solarbio Science & Technology Co., Ltd.) supplemented with 10% FBS (Biological Industries Sartorius AG) and penicillin/streptomycin at 37˚C in a humidified, 5% CO2 atmosphere. GMCs stimulated with 3.0 µg/ml LPS (MilliporeSigma) (10-11) were defined as the model group (n=3) and unstimulated GMCs were defined as the control group (n=3).

Cell proliferation assay

Cell proliferation rates were assessed using the Cell Counting Kit-8 (CCK-8) assay (cat. no. BB19071X; Shanghai Besto Biological Technology Co., Ltd.). GMCs were seeded (5.0x104 cells/well) into 96-well tissue culture plates with DMEM medium and treated with CCK-8 solution (10 µl/well) for 1 h. The optical density (absorbance at 450 nm) was assessed using an RT-6100 ELISA reader (Rayto Life and Analytical Sciences Co., Ltd.).

LncRNA sequence analysis

To ensure the quality of paired-end sequencing reads, FastQC software (version 0.10.1) was used to evaluate the quality of the original sequencing data. The input reads were considered good data quality when the Q20 base percentage in ‘Reads’ was ≥90% and the Q30 base percentage in ‘Reads’ was ≥80%.

High-throughput RNA-seq was performed by Genesky Bio-tech Co., Ltd. RNA fragmentation was performed using Bioruptor Pico (cat. no. B01060001; Diagenode SA) sonication in RNase-free water. RNA integrity was detected by denaturing gel electrophoresis and quantified using a NanoDrop 2000 spectrophotometer (Thermo Fisher Scientific, Inc.). The purified RNA fragments were then used to construct libraries using the TruSeq RNA Sample Prep Kit (cat. no. RS-122-2002; Illumina, Inc.). Libraries underwent quality control and were quantified using an Agilent 2100 bioanalyzer system (Agilent Technologies, Inc.). Paired-end 150 bp sequencing was performed using an Illumina HiSeq 2500 (Illumina, Inc.).

Isolation of RNA and reverse transcription-quantitative (RT-qPCR)

The expression levels of NONMMUG036949.2, NONMMUG089165.1, NONMMUG030447.2, NONMMUG028702.2, NONMMUG039651.2 and NONMMUG032587.2 were assessed using RT-qPCR. Total RNA was extracted from the cells using TRIzol® (Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions. Primers (Table I) were designed based on the cDNA sequences, which were accessed from the National Center for Biotechnology Information (NCBI) and tested using NCBI Primer BLAST, using Primer Premier 5 (Premier Biosoft International), and synthesized by Sangon Biotech Co., Ltd. The concentration and purity of the isolated RNA were determined using an OD1000+ Ultra Micro Spectrophotometer (WuYi Technologies), and reverse transcription was performed using the PrimeScript™ RT Reagent Kit with gDNA Eraser (cat. no. RR047A; Takara Biotechnology Co., Ltd.). qPCR was performed using a StepOne Plus fluorescence quantitative PCR instrument (Applied Biosystems; Thermo Fisher Scientific, Inc.) with SYBR Green qPCR Master Mix (cat. no. G3322-05; Wuhan Servicebio Technology Co., Ltd.). β-actin was used as the internal control; for quantitative results, the expression of lncRNA was expressed as fold change using the 2-ΔΔCq method (12).

Table I

Primer sequences used for reverse transcription-quantitative PCR.

Table I

Primer sequences used for reverse transcription-quantitative PCR.

GeneAmplicon size, bpSequence (5'-3')
NONMMUG036949.2185F: TTCTCCAGGACACTCACCAC
  R: AGAGAGCCAGGCATAGTGTG
NONMMUG089165.1140F: AGACCACGACGCCTTAAGTA
  R: GAATGGGAGTCCGAATGCAG
NONMMUG030447.273F: GCGTCGCCTAACGGTCC
  R: GGGACAAGAAGGTCATCGGT
NONMMUG028702.2181F: ACTGATACCATGCACCTCTCA
  R: GCATGTCACTTCAGCCTCTG
NONMMUG039651.2160F: TCCCTGCTGCAGTGTCATAA
  R: AGCAAAGCTCCCTTGTCTCT
NONMMUG032587.2175F: CAAGCCTGGATGTTCCATCG
  R: AGGGCACACCCTTCAAAGAT

[i] F, forward; R, reverse

Construction of the lncRNA-mRNA regulatory network

LncTar (http://www.cuilab.cn/lnctar) was used to predict lncRNA-mRNA interactions utilizing free energy minimization (13-14). LncTar utilizes a variation on the standard ‘sliding’ algorithm approach to calculate the normalized binding free energy (ndG) and predicts the minimum free energy joint structure. ndG ≤0.1 was regarded as the cutoff to determine the paired RNAs as interacting, ndG >0.1 was considered to indicate that paired RNAs did not interact. Subsequently, Cytoscape 3.8.1 was used to visualize the lncRNA-mRNA network after screening the target mRNAs of differentially expressed lncRNAs.

Construction of the lncRNA-associated ceRNA network

Interactions between miRNAs and lncRNAs were predicted using the miRNA target prediction software, miRanda (http://www.miranda.org) (15), and the three miRNAs of the highest confidence that can bind to lncRNAs were chosen. The target mRNAs of miRNAs were predicted using TargetScan 8.0 (https://www.targetscan.org/vert_80/) (16), and mRNAs with high confidence (cumulative weighted context score cutoff level <-0.8) bound to the miRNAs were screened out (17). Cytoscape 3.8.1 was then used to delineate the lncRNA-miRNA-mRNA ceRNA network.

Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis

To evaluate the role of differentially expressed lncRNAs in LPS-induced GMCs, GO biological process (BP) term enrichment and KEGG pathway enrichment were assessed (18). GO terms with P<0.05 were considered statistically significant. KEGG pathway enrichment analysis was used to identify significantly enriched signal transduction pathways or metabolic pathways (P<0.05). GO and KEGG enrichment analysis were performed using the free online data analysis platform OmicShare tools (https://www.omicshare.com/tools).

Protein-protein interaction (PPI) network construction and core gene screening

The PPI network of the mRNA involved in the ceRNA network was generated using the STRING online database tool (Version 11.5, https://string-db.org) (19); interaction pairs with a confidence value >0.4 were deemed significant and were retained. The top 10 genes in the PPI network were evaluated using the MCC, MNC and Degree algorithms through the CytoHubba plug-in for Cytoscape (20). The final hub genes were identified as those that were identified by all three of the algorithms.

GEO public dataset analysis

The public datasets (GSE104066) utilized in this study were obtained from the GEO database (https://www.ncbi.nlm.nih.gov/geo) and were initially screened based on disease (CGN), organism (Homo sapiens) and experiment type (expression profiling by array). Ultimately, the GSE104066 dataset was selected for use in CGN analysis.

Statistical analysis

Statistical analysis was performed using SPSS 22.0 software (IBM Corp.), and data are reported as the mean + SD. The data were subjected to one-way ANOVA with Tukey's post hoc multiple comparison tests. P<0.05 was considered to indicate a statistically significant difference.

Results

Optimal concentration and treatment time of LPS were determined using the CCK-8 assay

The present study evaluated the proliferative ability of GMCs induced by different concentrations of LPS (0.5, 1.0, 3.0, 5.0 and 10.0 µg/ml) using the CCK-8 assay at 24 and 48 h following treatment. The relationship between cell proliferation, LPS concentration and intervention time were assessed (Fig. 1). The results of the CCK-8 proliferation assay demonstrated that LPS could effectively induce cell proliferation in GMCs, with the highest absorbance (optical density 450 nm) observed at a concentration of 3.0 µg/ml (Table SI), which indicated that 3.0 µg/ml was the optimal concentration of LPS for the induction of cell proliferation in GMCs. Furthermore, there was no significant difference demonstrated between the treatment times of 24 and 48 h (P=0.62) at a concentration of 3.0 µg/ml LPS. Consequently, the concentration of 3.0 µg/ml and the treatment time of 24 h were selected for use in subsequent experiments.

Characteristics of differentially expressed lncRNAs

In the present study, six groups of cells were used in subsequent experiments, including three groups of LPS-induced GMCs as the model group (LPS1-3) and three groups of normal GMCs as the control group (CON1-3).

After deduplication, quality trimming and quality filtering, the sequencing data at both ends of the R1 and R2 paired reads were assessed as being of good quality (Q20 base and Q30 base were both >95%). The proportion of ‘clean reads’ retained after cleaning was >95% for each of the six samples which met the pre-determined quality requirements for sequencing. The quality control results of the sequencing data were presented (Table II; Table SII).

Table II

The quality control results for the sequencing data.

Table II

The quality control results for the sequencing data.

 R1R2 
SampleQ20, %Q30, %Q20, %Q30, %Clean reads, % of total
CON-198.195.398.595.997.7
CON-298.195.298.597.597.5
CON-398.195.398.597.597.5
LPS-198.195.298.297.797.7
LPS-298.295.498.396.996.9
LPS-398.195.398.497.397.3

[i] CON, control; LPS, lipopolysaccharide.

The violin plot demonstrated the relative abundance of lncRNAs in each sample (Fig. 2A). The violin plot demonstrated that there were significant differences in the expression of lncRNAs in LPS-induced GMCs compared with control GMCs. The threshold for differentially expressed lncRNAs was set at absolute fold change |(FC)|≥1.5 and P<0.05 (21-22). A total of 1,532 differentially expressed lncRNAs, including 594 upregulated lncRNAs and 938 downregulated lncRNAs, were identified from a total of 46,879 lncRNAs (Fig. 2B; Table SIII). A heat map of the top 50 differentially expressed lncRNAs was presented (Fig. 2C). The top 10 upregulated and downregulated lncRNAs are presented in Table III, and the radar map in Fig. 2D presents the top 10 upregulated and downregulated lncRNA expression levels in LPS-induced GMCs compared with control GMCs.

Table III

Top 10 upregulated and downregulated expressed log non-coding RNAs.

Table III

Top 10 upregulated and downregulated expressed log non-coding RNAs.

Gene ID Log2(FC)P-valueRegulation
NONMMUG011832.26.80680.0013Up
NONMMUG017581.26.82260.0052Up
NONMMUG090379.16.84730.0049Up
NONMMUG054724.16.88940.001Up
NONMMUG023960.26.8950.0001Up
NONMMUG025222.26.93140.0001Up
NONMMUG085342.17.0671<0.0001Up
NONMMUG007118.28.61840.0274Up
NONMMUG076324.18.74890.0252Up
NONMMUG079107.18.81340.0241Up
NONMMUG006074.2-10.4113<0.0001Down
NONMMUG041623.2-8.8981<0.0001Down
NONMMUG020465.2-8.56090.0285Down
NONMMUG019560.2-8.32420.0332Down
NONMMUG095800.1-8.17960.0364Down
NONMMUG025486.2-7.9163<0.0001Down
NONMMUG020727.2-7.89280.0435Down
NONMMUG056215.1-7.85810.0444Down
NONMMUG036634.2-7.5271<0.0001Down
NONMMUG019631.2-7.2817<0.0001Down

[i] FC, fold change.

Construction of the lncRNA-mRNA regulatory network

To evaluate the functions of differentially expressed lncRNAs, 556 target mRNAs of the 236 lncRNAs were predicted using LncTar (Table SIV). The highly coordinated expression between lncRNAs and target mRNAs may be due to complementary base pairing between lncRNA and mRNA (23). The lncRNA-mRNA regulatory network presented in Fig. 3 shows the interaction relationship between lncRNA and mRNA. For example, lncRNA NONMMUG029023.2 may regulate Stoml2 mRNA expression. Ptdss2 may be affected by both lncRNA NONMMUG039651.2 and lncRNA NONMMUG095401.1.

To elucidate the biological functions of differentially expressed lncRNAs in LPS-induced GMCs, GO BP term enrichment and KEGG pathway analyses were performed on all mRNAs in the lncRNA-mRNA regulatory network (Fig. 4). Within the GO BP classification, ‘cellular macromolecule metabolic process’, ‘negative regulation of biological process’ and ‘nitrogen compound metabolic process’ were the top three over-represented terms (Fig. 4A). Within the GO cellular components (CC) classification, ‘intracellular’, ‘intracellular part’ and ‘membrane-bounded organelle’ were the top three over-represented terms (Fig. 4B). Within the GO molecular function (MF) classification, ‘galactoside 2-α-L-fucosyltransferase activity’, ‘α-(1,2)-fucosyltransferase activity’ and ‘chromatin insulator sequence’ binding were the top three over-represented terms (Fig. 4C).

The KEGG metabolic pathway enrichment analysis demonstrated that ‘GnRH secretion’, ‘Melanoma’, ‘Ras signaling pathway’, ‘Glycosphingolipid biosynthesis-globo and isoglobo series’ and ‘β-Alanine metabolism’ were the top five most significantly enriched KEGG pathways (Fig. 4D). In addition, inflammation-related signaling pathways, such as the ‘Rap1 signaling pathway’ and ‘Ras signaling pathway’, and substance metabolism pathways, such as ‘Ribofiavin metabolism’, ‘β-Alanine metabolism’ and ‘Histidine metabolism’ were also significantly enriched. These results suggested that inflammation and substance metabolism disorder may be the underlying CGN pathogenesis.

RT-qPCR validation

To verify the reliability of the sequencing data and to support more clinically meaningful follow-up research, RT-qPCR validation of certain highly conserved lncRNAs between humans and mice was performed. The conservation information of lncRNAs was obtained from the NONCODE database (http://www.noncode.org). The conservation analysis of lncRNAs was assessed using the E-value, where a threshold of E-value <1x10-5 was used (24). After conservation analysis among species (Table IV), NONMMUG036949.2, NONMMUG089165.1, NONMMUG030447.2, NONMMUG028702.2, NONMMUG039651.2 and NONMMUG032587.2 were selected for RT-qPCR validation (25). The RT-qPCR results demonstrated the same expression trend as the RNA-Seq results, with the six selected lncRNAs all significantly upregulated in LPS-induced GMCs compared with the control group (Fig. 5; Table SV).

Table IV

Conservative information for the six selected long non-coding RNAs.

Table IV

Conservative information for the six selected long non-coding RNAs.

GeneChromosome log2(FC)P-valueAlignmentScore, bitsE-value
NONMMUG036949.261.28990.0476 NONHSAG058656.260 6.00x10-7
NONMMUG089165.1121.96080.0054 NONHSAG048570.2283 3.00x10-74
NONMMUG030447.241.41350.0250 NONHSAG056378.1232 7.00x10-59
NONMMUG028702.241.21520.0287 NONHSAG038858.256 2.00x10-5
NONMMUG039651.271.05440.0353 NONHSAG063259.1168 3.00x10-39
NONMMUG032587.251.39380.0013 NONHSAG088273.166 2.00x10-8

[i] FC, fold change.

Construction of the lncRNA-associated ceRNA network

In addition to the lncRNA-mRNA regulatory network, lncRNAs can also serve a role in other gene networks that regulate diverse biological processes, such as the ceRNA network (26). To construct an lncRNA-associated ceRNA network, the aforementioned six highly conserved and RT-qPCR-validated lncRNAs were used to construct ceRNA networks. The ceRNA network consisted of miRNAs of the highest confidence with lncRNAs, and mRNAs with a high confidence (cumulative weighted context score cutoff level <-0.8) (27) bound to the miRNAs, which included 6 lncRNAs, 18 miRNAs and 419 mRNAs (Fig. 6; Tables SVI and SVII). lncRNAs can be competing targets of shared miRNAs with other mRNAs and form a complex regulatory ceRNA network. For example, the lncRNA NONMMUG089165.1 may act as a sponge for mmu-miR-3960 to affect EVX1 expression. PARD3B could be affected by both the lncRNA NONMMUG039651.2/mmu-miR-7081-5P axis and the lncRNA NONMMUG028702.2/mmu-miR-7044-5P axis at the same time.

To evaluate the biological functions of the lncRNAs in the ceRNA network, GO BP and KEGG pathway analysis was performed on all mRNAs involved in the ceRNA network (Fig. 7). Within the GO BP classification, ‘system development’, ‘animal organ development’ and ‘multicellular organism development’ were the top three over-represented terms (Fig. 7A). Within the GO CC classification, ‘neuronal cell body membrane’, ‘cell body membrane’ and ‘α-β T cell receptor complex’ were the top three over-represented terms (Fig. 7B). Within the GO MF classification, ‘DNA-binding transcription activator activity, RNA polymerase II-specific’, ‘α-1,6-mannosylglycoprotein 6-β-N-acetylglucosaminyltransferase activity’ and ‘DNA-binding transcription factor activity’ were the top three over-represented terms (Fig. 7C). The KEGG metabolic pathway enrichment analysis demonstrated that ‘Collecting duct acid secretion’, ‘Circadian rhythm’, ‘Proteoglycans in cancer’, ‘Pathways in cancer’ and ‘Oxytocin signaling pathway’ were the top five most significantly enriched KEGG pathways (Fig. 7D). Furthermore, inflammation-related signaling pathways, such as ‘MAPK signaling pathway’, ‘mTOR signaling pathway’ and ‘Rap1 signaling pathway’ were significantly enriched KEGG pathways. These results suggested that inflammation may be the underlying CGN pathogenesis.

Screening of PPI core genes and validation on GEO dataset

The PPI network of all the mRNAs involved in ceRNA was successfully constructed, including 396 nodes and 260 edges (Fig. 8A). The top 10 genes were evaluated in the PPI network using the MCC, MNC and Degree algorithms; the results of the three algorithms were cross-compared to identify five core genes, including IKAROS family zinc finger 1 (IKZF1), CD3 ε subunit of T-cell receptor complex (CD3E), synapsin 1 (SYN1), glutamate ionotropic receptor NMDA subtype 2B (GRIN2B) and SPI1) (Fig. 8B). According to the ceRNA hypothesis, lncRNAs compete with miRNAs, to bind to the target genes, which increases their mRNA expression. Public dataset GSE104066 from the GEO database was used to validate the expression of IKZF1, CD3E, SYN1, GRIN2B and SPI1 in CGN. The data demonstrated that IKZF1, CD3E, SYN1 and GRIN2B were significantly upregulated; however, no significant difference was demonstrated for SPI1 (Fig. 8C; Table SVIII).

Discussion

Chronic inflammation is an essential factor in the occurrence and development of CGN, which may activate cell proliferation and induce the deregulation of cells (28-29). Excessive proliferation of GMCs, commonly observed in glomerulonephritis, is an important pathological basis of kidney disease (30-31). LPS is a major constituent of the outer membrane of gram-negative bacteria that can stimulate the activation of inflammatory factors in cells, leading to systemic inflammatory response and activation of the immune system. LPS has frequently been used as an inducer of cell proliferation in numerous previous studies. For example, it has been previously reported that LPS can induce THP-1 cell (32) and naive B cell (33) proliferation. Our research group has previously used LPS-induced GMCs as an in vitro model of CGN (34); therefore, in the present study, RNA-seq was used to evaluate CGN-related lncRNAs in LPS-induced GMCs, and an lncRNA-mRNA regulatory network and an lncRNA-miRNA-mRNA ceRNA network were constructed to elucidate the possible molecular mechanism of CGN.

The ceRNA theory was first proposed by Salmena (35) in 2011; it provided a new perspective for studying the role of RNA biological behavior in the occurrence and development of disease. According to the ceRNA theory, RNA transcripts, including non-coding RNAs, circular RNAs and pseudogene transcripts, could function as miRNA sponges and so regulate miRNA expression. Previous studies have reported that lncRNAs could affect mRNA stability through a ceRNA theory. For example, it has been previously reported that lncRNA linc00673 acts as a ceRNA by sponging miR-150-5p and thus regulating ZEB1 expression; as such, it was suggested that linc00673 served an essential role in the regulation of non-small cell lung cancer proliferation, migration and invasion (36). Previous studies have reported on the ceRNA theory and the link between lncRNAs and miRNAs (37). Therefore, the construction of a lncRNA-associated ceRNA network in CGN could have important research significance.

The enrichment analysis of the GO terms and KEGG pathways of lncRNA-targeted genes presented a preliminary depiction of the lncRNA function under investigation. The present study performed a comprehensive analysis of the GO terms and KEGG pathways for all mRNAs involved in the lncRNA-mRNA regulatory network and lncRNA-associated ceRNA network. The results of the two KEGG pathway analyses indicated significant enrichment of classical inflammatory signaling pathways, including MAPK, Rap1, Ras and mTOR. In instances of renal dysfunction, the timely clearance of pro-inflammatory cytokines and small to medium-sized molecular toxins is hindered, which leads to an elevation in inflammatory cytokines. Furthermore, the accumulation of these toxins within the body can stimulate the production of additional inflammatory cytokines (38), as evidenced by the enrichment of numerous inflammatory signaling pathways in the present study.

Furthermore, the metabolism of certain substances appeared to be affected, with pathways such as ‘N-Glycan biosynthesis’, ‘Glycosphingolipid biosynthesis’, ‘Galactose metabolism’, ‘Glycosaminoglycan degradation’, ‘β-Alanine metabolism’, ‘Riboflavin metabolism’ and ‘Histidine metabolism’ being significantly enriched in the KEGG enrichment analysis of the lncRNA-mRNA regulatory network. The kidneys are vital organs responsible for the elimination of metabolic waste products from the bloodstream and the regulation of the homeostatic levels of electrolytes and metabolites, while simultaneously eliminating harmful toxins from the body. In instances where kidney function is compromised, the metabolic equilibrium of specific substances within the body is disrupted (39). Previous studies have reported notable variances in the plasma amino acid profile of individuals with chronic kidney disease in comparison with those of healthy individuals, which is typically characterized by alterations in the levels of endogenous and essential amino acids (40,41).

There were certain limitations in the present study. Firstly, the number of samples tested in RNA-seq was small and should be increased in future studies to reduce possible bias in the sequencing results. Secondly, both the lncRNA-miRNA and miRNA-mRNA regulatory relationships were only predicted using bioinformatics analysis software, and all hypotheses and relevant mechanisms need to be verified by further experimental molecular studies.

In conclusion, 1,532 differentially expressed lncRNAs, including 594 upregulated lncRNAs and 938 downregulated lncRNAs, were identified using RNA-seq in LPS-induced GMCs. Furthermore, the lncRNA-mRNA regulatory network including 236 lncRNAs and 556 mRNAs, and the lncRNA-miRNA-mRNA ceRNA network including 6 lncRNAs, 18 miRNAs and 419 mRNAs were constructed. KEGG pathway analysis demonstrated that certain classical inflammatory signaling pathways and substances metabolism were significantly enriched. The present study demonstrated a global view of the lncRNA-associated ceRNA network, and may offer novel insights into the roles of lncRNAs in the pathogenesis of CGN as well as in identifying promising diagnostic biomarkers.

Supplementary Material

Raw data of CCK-8 proliferation assay.
The quality control result of sequencing data
All long non-coding RNAs detected by RNA sequencing
The target mRNAs of 236 differentially expressed lncRNAs
Raw reverse transcription-quantitative PCR data
The target miRNAs of long non-coding RNAs
The target mRNAs of miRNAs
The expression of IKZF1, CD3E, SYN1, GRIN2B, and SPI1 in dataset GSE104066

Acknowledgements

Not applicable.

Funding

Funding: This study was financially supported by The National Natural Science Foundation of China (grant no. 81973546) and The Key Scientific Research Projects of Natural Science in Colleges and Universities in Anhui Province (grant no. 2022AH050747).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request. The raw RNA sequencing data were deposited in the National Genomics Data Center (accession number PRJCA017179; https://ngdc.cncb.ac.cn/bioproject/browse/PRJCA017179.

Authors' contributions

XXZ, TL, LBW and JRG wrote the manuscript. JRG conceived and designed the experiment. XXZ performed the cell experiment and acquisition of data. TL and LBW interpreted and analyzed the sequencing data. LBW and JRG revised the manuscript critically for important intellectual content. XXZ, TL, LBW and JRG confirm the authenticity of all the raw data. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

All authors declare that they have no competing interests.

References

1 

Sethi S and Fervenza FC: Standardized classification and reporting of glomerulonephritis. Nephrol Dial Transplant. 34:193–199. 2019.PubMed/NCBI View Article : Google Scholar

2 

Avraham S, Korin B, Chung JJ, Oxburgh L and Shaw AS: The Mesangial cell-the glomerular stromal cell. Nat Rev Nephrol. 17:855–864. 2021.PubMed/NCBI View Article : Google Scholar

3 

Chao S, Xu Q, Dong S, Guo M, Liu X and Cheng X: Polygala fallax Hemsl combined with compound Sanqi granules relieves glomerulonephritis by regulating proliferation and apoptosis of glomerular mesangial cells. J Int Med Res. 48(300060519894124)2020.PubMed/NCBI View Article : Google Scholar

4 

Shen J, Wu Q, Liang T, Zhang J, Bai J, Yuan M and Shen P: TRIM40 inhibits IgA1-induced proliferation of glomerular mesangial cells by inactivating NLRP3 inflammasome through ubiquitination. Mol Immunol. 140:225–232. 2021.PubMed/NCBI View Article : Google Scholar

5 

Bridges MC, Daulagala AC and Kourtidis A: LNCcation: lncRNA localization and function. J Cell Biol. 220(e202009045)2021.PubMed/NCBI View Article : Google Scholar

6 

Zhu P, He F, Hou Y, Tu G, Li Q, Jin T, Zeng H, Qin Y, Wan X, Qiao Y, et al: A novel hypoxic long noncoding RNA KB-1980E6.3 maintains breast cancer stem cell stemness via interacting with IGF2BP1 to facilitate c-Myc mRNA stability. Oncogene. 40:1609–1627. 2021.PubMed/NCBI View Article : Google Scholar

7 

Tan YT, Lin JF, Li T, Li JJ, Xu RH and Ju HQ: LncRNA-mediated posttranslational modifications and reprogramming of energy metabolism in cancer. Cancer Commun (Lond). 41:109–120. 2021.PubMed/NCBI View Article : Google Scholar

8 

Gao J, Zhu X, Chen H, Jiang H, Shi M, Wei L and Qin X: Long non-coding NONRATG001910.2 promotes the proliferation of rat mesangial cell line HBZY-1 through the miR-339-3p/CTNNB1 axis. Front Genet. 28(834144)2022.PubMed/NCBI View Article : Google Scholar

9 

Zhou K, Ou Q, Wang G, Zhang W, Hao Y and Li W: High long non-coding RNA NORAD expression predicts poor prognosis and promotes breast cancer progression by regulating TGF-β pathway. Cancer Cell Int. 19(63)2019.PubMed/NCBI View Article : Google Scholar

10 

Gao JR, Shi MM, Jiang H, Zhu XL, Wei LB and Qin XJ: MicroRNA-339-5p inhibits lipopolysaccharide-induced rat mesangial cells by regulating the Syk/Ras/c-Fos pathway. Naunyn Schmiedebergs Arch Pharmacol. 395:1075–1085. 2022.PubMed/NCBI View Article : Google Scholar

11 

Liu T, Zhuang XX, Qin XJ, Wei LB and Gao JR: The potential role of N6-methyladenosine modification of LncRNAs in contributing to the pathogenesis of chronic glomerulonephritis. Inflamm Res. 72:623–638. 2023.PubMed/NCBI View Article : Google Scholar

12 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2-(Delta Delta C(T)) method. Methods. 25:402–408. 2001.PubMed/NCBI View Article : Google Scholar

13 

Khoei MA, Karimi M, Karamian R, Amini S and Soorni A: Identification of the complex interplay between nematode-related lncRNAs and their target genes in glycine max L. Front Plant Sci. 12(779597)2021.PubMed/NCBI View Article : Google Scholar

14 

Li J, Ma W, Zeng P, Wang J, Geng B, Yang J and Cui Q: LncTar: A tool for predicting the RNA targets of long noncoding RNAs. Brief Bioinform. 16:806–812. 2015.PubMed/NCBI View Article : Google Scholar

15 

John B, Enright AJ, Aravin A, Tuschl T, Sander C and Marks DS: Human MicroRNA targets. PLoS Biol. 2(e363)2004.PubMed/NCBI View Article : Google Scholar

16 

McGeary SE, Lin KS, Shi CY, Pham TM, Bisaria N, Kelley GM and Bartel DP: The biochemical basis of microRNA targeting efficacy. Science. 366(eaav1741)2019.PubMed/NCBI View Article : Google Scholar

17 

Su H, Tao T, Yang Z, Kang X, Zhang X, Kang D, Wu S and Li C: Circular RNA cTFRC acts as the sponge of MicroRNA-107 to promote bladder carcinoma progression. Mol Cancer. 18(27)2019.PubMed/NCBI View Article : Google Scholar

18 

Chen L, Zhang YH, Lu G, Huang T and Cai YD: Analysis of cancer-related lncRNAs using gene ontology and KEGG pathways. Artif Intell Med. 76:27–36. 2017.PubMed/NCBI View Article : Google Scholar

19 

Szklarczyk D, Kirsch R, Koutrouli M, Nastou K, Mehryary F, Hachilif R, Gable AL, Fang T, Doncheva NT, Pyysalo S, et al: The STRING database in 2023: Protein-protein association networks and functional enrichment analyses for any sequenced genome of interest. Nucleic Acids Res. 51:D638–D646. 2023.PubMed/NCBI View Article : Google Scholar

20 

Doncheva NT, Morris JH, Gorodkin J and Jensen LJ: Cytoscape StringApp: Network analysis and visualization of proteomics data. J Proteome Res. 18:623–632. 2019.PubMed/NCBI View Article : Google Scholar

21 

Gao R, Liang X, Cheedipudi S, Cordero J, Jiang X, Zhang Q, Caputo L, Günther S, Kuenne C, Ren Y, et al: Pioneering function of Isl1 in the epigenetic control of cardiomyocyte cell fate. Cell Res. 29:486–501. 2019.PubMed/NCBI View Article : Google Scholar

22 

Weber MD, McKim DB, Niraula A, Witcher KG, Yin W, Sobol CG, Wang Y, Sawicki CM, Sheridan JF and Godbout JP: The influence of microglial elimination and repopulation on stress sensitization induced by repeated social defeat. Biol Psychiatry. 85:667–678. 2019.PubMed/NCBI View Article : Google Scholar

23 

Yang KC, Yamada KA, Patel AY, Topkara VK, George I, Cheema FH, Ewald GA, Mann DL and Nerbonne JM: Deep RNA sequencing reveals dynamic regulation of myocardial noncoding RNAs in failing human heart and remodeling with mechanical circulatory support. Circulation. 129:1009–1021. 2014.PubMed/NCBI View Article : Google Scholar

24 

Liang F, Zhang Y, Wang X, Yang S, Fang T, Zheng S and Zeng L: Integrative mRNA and long noncoding RNA analysis reveals the regulatory network of floral bud induction in longan (Dimocarpus longan Lour.). Front Plant Sci. 13(923183)2022.PubMed/NCBI View Article : Google Scholar

25 

Wang Y, Zhu P, Luo J, Wang J, Liu Z, Wu W, Du Y, Ye B, Wang D, He L, et al: LncRNA HAND2-AS1 promotes liver cancer stem cell self-renewal via BMP signaling. EMBO J. 38(e101110)2019.PubMed/NCBI View Article : Google Scholar

26 

Li S, Cao Y, Zhang H, Lu X, Wang T, Xu S, Kong T, Bo C, Li L, Ning S, et al: Construction of lncRNA-Mediated ceRNA network for investigating immune pathogenesis of ischemic stroke. Mol Neurobiol. 58:4758–4769. 2021.PubMed/NCBI View Article : Google Scholar

27 

Mulari S, Eskin A, Lampinen M, Nummi A, Nieminen T, Teittinen K, Ojala T, Kankainen M, Vento A, Laurikka J, et al: Ischemic heart disease selectively modifies the right atrial appendage transcriptome. Front Cardiovasc Med. 8(728198)2021.PubMed/NCBI View Article : Google Scholar

28 

Chung H, Komada T, Lau A, Chappellaz M, Platnich JM, de Koning HD, Petri B, Luque Y, Walker S, Benediktsson H, et al: AIM2 Suppresses inflammation and epithelial cell proliferation during glomerulonephritis. J Immunol. 207:2799–2812. 2021.PubMed/NCBI View Article : Google Scholar

29 

Eaton JE, Fritcher EG, Gores GJ, Atkinson EJ, Tabibian JH, Topazian MD, Gossard AA, Halling KC, Kipp BR and Lazaridis KN: Biliary multifocal chromosomal polysomy and cholangiocarcinoma in primary sclerosing cholangitis. Am J Gastroenterol. 110:299–309. 2015.PubMed/NCBI View Article : Google Scholar

30 

Harendza S, Schneider A, Helmchen U and Stahl RA: Extracellular matrix deposition and cell proliferation in a model of chronic glomerulonephritis in the rat. Nephrol Dial Transplant. 14:2873–2879. 1999.PubMed/NCBI View Article : Google Scholar

31 

Harendza S, Behrens U, Zahner G, Schneider A and Stahl RA: In vitro characterization of the mesangial phenotype in a proliferative glomerulonephritis of the rat. Nephrol Dial Transplant. 12:2537–2541. 1997.PubMed/NCBI View Article : Google Scholar

32 

Sun J, Shigemi H, Cao M, Qin E, Tang J, Shen J and Iwasaki H: Minocycline induces autophagy and inhibits cell proliferation in LPS-Stimulated THP-1 cells. Biomed Res Int. 2020(5459209)2020.PubMed/NCBI View Article : Google Scholar

33 

Ribeiro de Almeida C, Dhir S, Dhir A, Moghaddam AE, Sattentau Q, Meinhart A and Proudfoot NJ: RNA helicase DDX1 converts RNA G-quadruplex structures into R-loops to promote IgH class switch recombination. Mol Cell. 70:650–662. 2018.PubMed/NCBI View Article : Google Scholar

34 

Liu T, Zhuang XX, Qin XJ, Wei LB and Gao JR: Alteration of N6-methyladenosine epitranscriptome profile in lipopolysaccharide-induced mouse mesangial cells. Naunyn Schmiedebergs Arch Pharmacol. 395:445–458. 2022.PubMed/NCBI View Article : Google Scholar

35 

Salmena L, Poliseno L, Tay Y, Kats L and Pandolfi PP: A ceRNA hypothesis: The Rosetta Stone of a hidden RNA language? Cell. 146:353–358. 2011.PubMed/NCBI View Article : Google Scholar

36 

Lu W, Zhang H, Niu Y, Wu Y, Sun W, Li H, Kong J, Ding K, Shen HM, Wu H, et al: Long non-coding RNA linc00673 regulated non-small cell lung cancer proliferation, migration, invasion and epithelial mesenchymal transition by sponging miR-150-5p. Mol Cancer. 16(118)2017.PubMed/NCBI View Article : Google Scholar

37 

Wang L, Cho KB, Li Y, Tao G, Xie Z and Guo B: Long noncoding RNA (lncRNA)-mediated competing endogenous RNA networks provide novel potential biomarkers and therapeutic targets for colorectal cancer. Int J Mol Sci. 20(5758)2019.PubMed/NCBI View Article : Google Scholar

38 

Suliman ME, Qureshi AR, Stenvinkel P, Pecoits-Filho R, Bárány P, Heimbürger O, Anderstam B, Ayala ER, Filho JCD, Alvestrand A and Lindholm B: Inflammation contributes to low plasma amino acid concentrations in patients with chronic kidney disease. Am J Clin Nutr. 82:342–349. 2015.PubMed/NCBI View Article : Google Scholar

39 

Yan LJ: Redox imbalance and mitochondrial abnormalities in kidney disease. Biomolecules. 12(476)2022.PubMed/NCBI View Article : Google Scholar

40 

Taherkhani A, Kalantari S, Oskouie AA, Nafar M, Taghizadeh M and Tabar K: Network analysis of membranous glomerulonephritis based on metabolomics data. Mol Med Rep. 18:4197–4212. 2018.PubMed/NCBI View Article : Google Scholar

41 

Carter JL, Parker CT, Stevens PE, Eaglestone G, Knight S, Farmer CK and Lamb EJ: Biological variation of plasma and urinary markers of acute kidney injury in patients with chronic kidney disease. Clin Chem. 62:876–883. 2016.PubMed/NCBI View Article : Google Scholar

Related Articles

Copy and paste a formatted citation
x
Spandidos Publications style
Zhuang X, Liu T, Wei L and Gao J: Construction of chronic glomerulonephritis‑related lncRNA‑mRNA regulatory network and lncRNA‑­miRNA‑mRNA ceRNA network by bioinformatics analysis. Exp Ther Med 26: 403, 2023
APA
Zhuang, X., Liu, T., Wei, L., & Gao, J. (2023). Construction of chronic glomerulonephritis‑related lncRNA‑mRNA regulatory network and lncRNA‑­miRNA‑mRNA ceRNA network by bioinformatics analysis. Experimental and Therapeutic Medicine, 26, 403. https://doi.org/10.3892/etm.2023.12102
MLA
Zhuang, X., Liu, T., Wei, L., Gao, J."Construction of chronic glomerulonephritis‑related lncRNA‑mRNA regulatory network and lncRNA‑­miRNA‑mRNA ceRNA network by bioinformatics analysis". Experimental and Therapeutic Medicine 26.2 (2023): 403.
Chicago
Zhuang, X., Liu, T., Wei, L., Gao, J."Construction of chronic glomerulonephritis‑related lncRNA‑mRNA regulatory network and lncRNA‑­miRNA‑mRNA ceRNA network by bioinformatics analysis". Experimental and Therapeutic Medicine 26, no. 2 (2023): 403. https://doi.org/10.3892/etm.2023.12102