Open Access

Long non‑coding RNA Dlx6os1 serves as a potential treatment target for diabetic nephropathy via regulation of apoptosis and inflammation

  • Authors:
    • Li Cheng
    • Jie Cheng
    • Wenfang Peng
    • Xiaohong Jiang
    • Shan Huang
  • View Affiliations

  • Published online on: August 7, 2020     https://doi.org/10.3892/etm.2020.9112
  • Pages: 3791-3797
  • Copyright: © Cheng 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

The present study investigated the effect of long non‑coding RNA (lncRNA) Dlx6os1 silencing on cell proliferation, apoptosis and fibrosis, and further explored its influence on the mRNA expression profile in mouse mesangial cells (MMCs) of a diabetic nephropathy (DN) cellular model. A DN cellular model was constructed in SV40 MES13 MMCs under high glucose conditions (30 mmol/l glucose culture). lncRNA Dlx6os1 short hairpin (sh)RNA plasmids and negative control (NC) shRNA plasmids were transfected into the MMCs of the DN cellular model as the sh‑lncRNA group and sh‑NC group respectively. The mRNA expression profile was determined in the sh‑lncRNA and sh‑NC groups. Compared with the sh‑NC group, the cell proliferation, mRNA and protein expression levels of proliferative markers (cyclin D1 and proliferating cell nuclear antigen) as well as fibrosis markers (fibronectin and collagen I) were suppressed, whereas cell apoptosis was promoted in the sh‑lncRNA group. The mRNA expression profile identified 423 upregulated mRNAs and 438 downregulated mRNAs in the sh‑lncRNA group compared with the sh‑NC group. Additionally, Gene Ontology/Kyoto Encyclopedia of Genes and Genomes enrichment analyses revealed that the differentially expressed mRNAs were enriched in apoptosis and inflammation‑related pathways. Further gene‑set enrichment analysis of apoptosis and inflammation revealed that lncRNA Dlx6os1 inhibition promoted apoptosis and suppressed inflammation in MMCs of the DN cellular model. In conclusion, lncRNA Dlx6os1 may serve as a potential treatment target for DN via regulation of multiple apoptosis‑ and inflammation‑related pathways.

Introduction

Diabetic nephropathy (DN), which occurs in 20-40% of all diabetes cases, is one of the systemic microvascular complications of diabetes as well as the leading cause of end-stage renal disease (1). DN is a chronic process whose early clinical symptoms are often not evident (2). In addition, there is a large risk of DN patients developing uremia, which is a key contributor to diabetes-related disability or death (2). Hyperglycemia, in the long term, causes hypertrophy, infiltration of inflammatory cells, thickening of the basement membrane, glomerular sclerosis and tubular atrophy, eventually leading to kidney failure (3). Once kidney failure is developed, DN is even more difficult to treat than other kidney diseases (3). Thus, the early prevention of DN is important for diabetes management. However, the current methodology is still insufficient for timely identification of disease onset and prognosis in DN.

Long non-coding RNAs (lncRNAs) are transcripts that measure more than 200 nucleotides with limited protein-coding ability (4). The effects of lncRNAs have been revealed in the pathogenesis of various human diseases including diabetes (5). Several lncRNAs, including lnc-Rpph1, lnc-MEG3 and lnc-CYP4B1-PS1-001, are known to regulate fibrosis and inflammation in DN (3,6,7). Our previous study selected five candidate lncRNAs from lncRNA microarray data and found that lncRNA Dlx6os1 was highly expressed in high glucose-treated mouse mesangial cells (MMCs) (8). The next stage of the investigation observed that inhibition of lncRNA Dlx6os1 suppressed proliferation and fibrosis but promoted apoptosis in MMCs of the DN cellular model (8). lncRNA Dlx6os1 is a novel gene that has not been studied in DN, despite the aforementioned evidence from our previous study (8). The present study further analyzed the effect of lncRNA Dlx6os1 suppression on the mRNA expression profile in MMCs of a DN cellular model to explore the mechanism of lncRNA Dlx6os1 inhibition in the pathogenesis of DN.

Materials and methods

Construction of DN cellular model

lncRNA Dlx6os1 exhibits good homology between mice and humans. A DN cellular model was constructed according to the method described in our previous study (8). In brief, SV40 MES13 cells (MMCs) were purchased from Shanghai Institutes for Biological Sciences and were cultured in Dulbecco modified Eagle's medium (Gibco; Thermo Fisher Scientific, Inc.) with 10% fetal bovine serum (Gibco; Thermo Fisher Scientific, Inc.) in a humidified incubator with 5% CO2 at 37˚C. Then 30 mmol/l glucose was added to treat SV40 MES13 cells for 96 h to construct the DN cellular model.

Construction and transfection of short hairpin (sh)RNA plasmids

Negative control (NC) shRNA and lncRNA Dlx6os1 shRNA plasmids were constructed by Genewiz, Inc. and transfected into a DN cellular model (SV40 MES13 cells under 30 mmol/l glucose culture), then termed the sh-NC group and sh-lncRNA group. Subsequently, i) lncRNA Dlx6os1 expression in the two groups was detected by reverse transcription-quantitative (RT-q) PCR at 48 h; ii) proliferation ability was detected by Cell Counting Kit-8 (Sigma-Aldrich; Merck KGaA) at 0, 24, 48 and 72 h according to the manufacturers' instructions; iii) the mRNA and protein expression levels of proliferative markers (cyclin D1 and proliferating cell nuclear antigen) as well as fibrosis markers (fibronectin and collagen I) were detected by RT-qPCR and western blotting at 48 h; iv) the cell apoptosis rate was detected by FITC Annexin V Apoptosis Detection kit II with propidium iodide (AV/PI; BD Biosciences) at 48 h according to the manufacturer's protocol (Fig. S1). Data were analyzed using a flow cytometer (BD FACSCalibur) with FlowJo 7.6 software (FlowJo, LLC). A wavelength of 488/535 nm was used.

mRNA sequencing

mRNA sequencing was performed by Genergy Bio Company (http://www.genenergy.cn/). In brief, cells were harvested at 48 h after transfection, and total RNA was extracted using TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.), then RNA concentration, purity and integrity were assessed. Subsequently, mRNA was captured by Dynabeads Oligo (dT)25 (Thermo Fisher Scientific, Inc.), then first and second strands of the cDNA were synthesized and library fragments were purified using AMPure XP system (Beckman Coulter, Inc.). PCR assay and assessment of library quality were conducted using Bioanalyzer 2100 system (Agilent Technologies, Inc.), then clustering of index-coded samples was performed using HiSeq PE Cluster kit v4 cBot (Illumina, Inc.), and the libraries were sequenced on Illumina Hiseq X10 platform (Illumina, Inc.). A total of 150 bp paired-end reads was produced after cluster generation for mRNA. Automated quality control and adapter trimming were conducted using Trim Galore, Cutadapt and FastQC (9). Then the trimmed reads were mapped to the mouse genome mm10 by Hisat2 (Version 2.1.0; http://ccb.jhu.edu/software/hisat2/manual.shtml), and mapping quality control was conducted using RSeQC (Version 3.0.0; http://rseqc.sourceforge.net/). Finally, the read counts of mRNA were then calculated using FeatureCounts (Rsubread; http://www.bioconductor.org). The top 20 dysregulated mRNAs are presented in Table I as a reference for future studies that investigate specific mRNA targeted by lncRNA D1x6os1 in DN.

Table I

Top 20 dysregulated mRNAs (10 upregulated and 10 downregulated) in sh-lncRNA cells vs. sh-NC cells.

Table I

Top 20 dysregulated mRNAs (10 upregulated and 10 downregulated) in sh-lncRNA cells vs. sh-NC cells.

Gene symbolIDChromosomeLog2 FCP-valueTrend
Ramp2 ENSMUSG00000001240112.078303 4.52x10-6Up
Tagln ENSMUSG0000003208592.016348 6.7x10-6Up
Krt7 ENSMUSG00000023039151.749297 2.09x10-5Up
Aifm3 ENSMUSG00000022763161.757504 3.1x10-5Up
Sult1c2 ENSMUSG00000023122171.781638 3.4x10-5Up
1700001P01Rik ENSMUSG00000018543115.796657 3.42x10-5Up
Foxp2 ENSMUSG0000002956362.688633 4.09x10-5Up
Sirt2 ENSMUSG0000001514972.458172 4.93x10-5Up
Sep15 ENSMUSG0000003707232.153584 7.67x10-5Up
Slc39a4 ENSMUSG00000063354153.04629 8.55x10-5Up
Ccdc160 ENSMUSG00000073207X-6.79158 4.98x10-7Down
Myom1 ENSMUSG0000002404917-2.09334 6.97x10-6Down
Lmbr1l ENSMUSG0000002299915-2.19777 8.64x10-6Down
Ddx43 ENSMUSG000000702919-1.72587 1.94x10-5Down
Vwa5b2 ENSMUSG0000004661316-2.63226 2.39x10-5Down
Slc11a1 ENSMUSG000000261771-5.77347 2.84x10-5Down
Ccdc7b ENSMUSG000000560188-4.83542 4.45x10-5Down
Fmn1 ENSMUSG000000440422-1.85445 4.85x10-5Down
Fer1l4 ENSMUSG000000133382-5.69906 5.37x10-5Down
As3mt ENSMUSG0000000355919-1.94909 5.43x10-5Down

[i] Ten upregulated and ten downregulated mRNAs were selected according to the rank of the P-value. sh, short hairpin; lnc, long non-coding; NC, negative control; FC, fold change.

Bioinformatics

The mRNAs which were identified in ≥50% samples were included in the analysis and raw read counts were normalized and logarithmically transformed for bioinformatics analysis. The bioinformatics analysis was conducted using R software (Version 3.3.3; http://www.R-project.org/) (10). In brief, i) differentially expressed mRNAs were detected using DeSeq2 package (Version 1.12.3; http://www.bioconductor.org/), and statistical significance was defined as adjusted P-value <0.05; the biological significance was defined as a difference of at least abs [log2 (fold change)]>2, which was presented as a Volcano plot; ii) Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis were performed using Database for Annotation, Visualization and Integrated Discovery web servers; iii) gene-set enrichment analysis (GSEA) on apoptosis and inflammation pathways was performed using the phenoTest package (11). The datasets of the present study were uploaded on Gene Expression Omnibus (GEO) (https://www.ncbi.nlm.nih.gov/geo/) for public availability. The GSE number is GSE145301.

Statistical analysis

Statistical analysis and figures were performed using GraphPad Prism 7.00 (GraphPad Software, Inc.). Comparison between two groups was determined by paired t-test. P<0.05 was considered to indicate a statistically significant difference.

Results

lncRNA D1x6os1 expression following transfection

In order to detect the relative expression of lncRNA D1x6os1 after transfection, qPCR was conducted at 48 h. lncRNA D1x6os1 expression was decreased in the sh-lncRNA group compared with the sh-NC group following transfection (P<0.001), which indicated successful transfection under high glucose exposure (Fig. 1).

Effect of lncRNA D1x6os1 silencing on cell proliferation, fibrosis and apoptosis in MMCs of a DN cellular model

Cell proliferation, fibrosis and apoptosis were determined to assess the influence of lncRNA D1x6os1 silencing. Cell proliferation was reduced in the sh-lncRNA group compared with the sh-NC group at 24 (P<0.05), 48 (P<0.05), and 72 h (P<0.01; Fig. 2A). The relative expression levels of cyclin D1 (P<0.05), PCNA (P<0.01), FN (P<0.05) and collagen I (P<0.05) were suppressed in the sh-lncRNA group compared with the sh-NC group (Fig. 2B and C). In addition, the cell apoptosis rate was increased in the sh-lncRNA group compared with the sh-NC group (P<0.001; Fig. 2D).

Volcano plot of differentially expressed mRNAs

A Volcano plot is an intuitive plot that screens out the differentially expressed genes between the two group of samples. In the present study, it revealed 423 upregulated mRNAs and 438 downregulated mRNAs in the sh-lncRNA group compared with the sh-NC group in MMCs of a DN cellular model (absolute value of FC>2, Padj<0.05; Fig. 3). The top 20 dysregulated mRNAs are presented in Table I.

GO and KEGG enrichment analysis of differentially expressed mRNAs

To evaluate the possible mechanisms of dysregulated mRNAs in response to lncRNA D1x6os1 silencing, GO and KEGG enrichment analyses were conducted. GO enrichment analysis revealed that the differentially expressed mRNAs were mainly enriched in biological processes including ‘apoptotic process’, ‘inflammatory response’ and ‘cytoskeleton organization’; and were mainly present in cellular components such as ‘integral component of membrane’, ‘cell surface’ and ‘extracellular space’; in addition, they were involved in molecular functions including ‘cytokine receptor activity’, ‘phospholipase binding’ and ‘cysteine-type endopeptidase activity’ (Fig. 4A). According to KEGG enrichment analysis, the dysregulated mRNAs were mainly enriched in inflammation and apoptosis-related pathways such as ‘NF-κB signaling pathway’, ‘cytokine-cytokine receptor interaction’ and apoptosis pathways (Fig. 4B).

GSEA

Since apoptosis and inflammation are the two main pathological mechanisms of DN, GSEA of apoptosis and inflammation was performed. In GSEA analysis of apoptosis, the leading-edge subset of mRNAs were overexpressed in sh-lncRNA cells compared with sh-NC cells according to the location of the maximum enrichment score (Fig. 5A). While in GSEA analysis of inflammation, the leading-edge subset of mRNAs were downregulated in sh-lncRNA cells compared with sh-NC cells (Fig. 5B). These results indicated that lncRNA D1x6os1 inhibition promoted cell apoptosis and suppressed inflammation in MMCs of a DN cellular model.

Discussion

As a progressive microvascular complication, DN is heralded by a subsequent reduction in glomerular filtration rate and marked by microalbuminuria (12). The renal inflammation underlies the pathogenic mechanism of DN (13). Principle inflammatory cells such as macrophages are responsible for inflammation response via the release of cytokines, chemokines and reactive oxygen species, which eventually increase the production of extracellular matrix in glomeruli as well as progressive tubulointerstitial fibrosis (13). In addition, T-lymphocyte activation induced by hyperglycemia induces kidney damage in the early stages by disturbing albumin glomerular excretion as well as renal infiltration (14). Apoptosis-induced dysfunction of podocytes and damage to mesangial cells and renal tubular epithelial cells are indicators of glomerular filtration dysfunction (15-18). Although the pathology and pathophysiology of DN have been investigated for some time and there has been progress, the complex molecular network is still not fully understood and identification of genes or molecules by conventional tools remains deficient. Therefore, screening of genes and molecules by novel strategies, such as high-throughput platforms are necessary for determining valuable information on the numerous molecules involved in DN pathology.

From the published microarray data, lncRNA expression profile is critical in the pathogenesis and progression of DN (3,19). Previous studies report that several lncRNAs regulate inflammation and cell proliferation in DN (3,19). For instance, lncRNA Rpph1 was demonstrated to facilitate inflammation and promote proliferation of MMCs by interacting with DN-related factor galectin-3 in a DN cellular model (3). Knockdown of lncRNA Gm4419 was revealed to inhibit pro-inflammatory cytokine expression and renal fibrosis biomarkers and reduce cell proliferation of MMCs under high glucose conditions (19). lncRNA Dlx6os1 was upregulated in the kidneys of diabetic mice and its expression was induced under high glucose conditions (20). Therefore, in order to mimic the DN environment, SV40 MES13 cells (MMCs) cultured under high glucose conditions were used for the transfection of lncRNA Dlx6os1 inhibitor plasmids in our previous study (8). In the present study, the same DN cellular model derived from MMCs was constructed, and it was confirmed that lncRNA Dlx6os1 silencing suppressed cell proliferation and fibrosis and promoted cell apoptosis in MMCs of the DN cellular model. The following theories may account for these effects: i) lncRNA Dlx6os1 silencing may influence the expression of target mRNAs and attenuate the cell cycle to suppress cell proliferation and facilitate apoptosis. It was demonstrated in the following GSEA analysis of apoptosis that lncRNA Dlx6os1 silencing promoted cell apoptosis in MMCs of the DN cellular model. The reduced cell apoptosis may attenuate the mesangial expansion as well as formation of glomerulosclerosis in DN (21). ii) Inhibition of lncRNA Dlx6os1 may suppress inflammation by blocking signaling pathways such as the NF-κB pathway, interfering with cytokine-cytokine receptor interaction (as demonstrated in the KEGG enrichment analysis) and reducing the inflammatory cytokines, as well as reactive oxygen species, leading to reduced secretion of the extracellular matrix and subsequent fibrosis.

Since the pathogenic mechanism of DN is complex and involves multiple molecular pathways, a more refined screening of the genetic background of lncRNA Dlx6os1 will certainly provide a greater insight into the disease. Therefore, additional RNA sequencing for the mRNA expression profile in lncRNA Dlx6os1-silenced MMCs of the DN cellular model was performed. The mRNA expression profile revealed that lncRNA Dlx6os1 silencing led to 423 upregulated mRNAs and 438 downregulated mRNAs in MMCs of the DN cellular model. The KEGG enrichment analyses revealed that the differentially expressed mRNAs were enriched in apoptotic and inflammatory responses/pathways. GSEA analysis of apoptosis and inflammation pathways was also performed and suggested that lncRNA Dlx6os1 silencing promoted cell apoptosis and inhibited inflammation in MMCs of the DN cellular model. These observations supplemented the potential molecular mechanism of lncRNA Dlx6os1 in the pathogenesis of DN to some extent. However, deeper exploration of downstream mRNAs of lncRNA Dlx6os1 in regulating cell proliferation, fibrosis and cell apoptosis in DN is still required.

There were several limitations to the present study: i) The effect of lncRNA Dlx6os1 silencing on the mRNA expression profile was evaluated by RNA sequencing, which is high-throughput and lacks accuracy (22). Therefore, detecting individual mRNA expression using qPCR and corresponding protein expression using western blotting may be helpful to validate the influence of lncRNA Dlx6os1 on mRNA expression in MMCs of the DN cellular model. ii) Although commonly used in DN research, knocking down an lncRNA in a short-term as well as an acute kidney damage cellular model does not substantially represent the real DN condition as a chronic disease. Therefore, further animal study is required to validate the results of the present study. iii) lncRNA Dlx6os1 was revealed to regulate apoptosis and inflammation in MMCs of the DN cellular model, whereas the clinical implications, including its correlation with disease risk, disease progression or prognosis of DN patients, remains to be elucidated, which may be a valuable research direction for diabetic diseases in the future. iv) The present study only reported general information concerning lncRNA Dlx6os1 silencing on biological processes and molecular functions such as apoptosis and inflammation in DN, whereas the detailed pathways or the protein markers were not investigated.

In conclusion, lncRNA Dlx6os1 silencing attenuated disease progression by regulating cell apoptosis and inflammation-related pathways in MMCs of a DN cellular model, suggesting that it could assist with developing a novel treatment target for DN.

Supplementary Material

Annexin V/propidium iodide analysis for cell apoptosis. A total of three repetitions were performed for sh-NC cells and sh-lncRNA cells. sh, short hairpin; NC, negative control; lnc, long non-coding.

Acknowledgements

Not applicable.

Funding

The present study was supported by the Foundation of Guizhou Science and Technology Plan Project [Guizhou Science and Technology platform talents; grant no. (2017) 5735-03] and the Dominant Discipline of the Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Changning District Committee of Science and Technology (grant no. CNKW2017Y06).

Availability of data and materials

The datasets used and/or analyzed during the present study are available from the corresponding author on reasonable request.

Authors' contributions

SH conceived and designed the study. LC and JC collected the data. WP and XJ performed the experiments and drafted the manuscript. All authors reviewed and edited the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Sun HJ, Wu ZY, Cao L, Zhu MY, Liu TT, Guo L, Lin Y, Nie XW and Bian JS: Hydrogen Sulfide: Recent progression and perspectives for the treatment of diabetic nephropathy. Molecules. 24(2857)2019.PubMed/NCBI View Article : Google Scholar

2 

Fried LF, Emanuele N, Zhang JH, Brophy M, Conner TA, Duckworth W, Leehey DJ, McCullough PA, O'Connor T, Palevsky PM, et al: Combined angiotensin inhibition for the treatment of diabetic nephropathy. N Engl J Med. 369:1892–1903. 2013.PubMed/NCBI View Article : Google Scholar

3 

Zhang P, Sun Y, Peng R, Chen W, Fu X, Zhang L, Peng H and Zhang Z: Long non-coding RNA Rpph1 promotes inflammation and proliferation of mesangial cells in diabetic nephropathy via an interaction with Gal-3. Cell Death Dis. 10(526)2019.PubMed/NCBI View Article : Google Scholar

4 

Reichelt-Wurm S, Wirtz T, Chittka D, Lindenmeyer M, Reichelt RM, Beck S, Politis P, Charonis A, Kretz M, Huber TB, et al: Glomerular expression pattern of long non-coding RNAs in the type 2 diabetes mellitus BTBR mouse model. Sci Rep. 9(9765)2019.PubMed/NCBI View Article : Google Scholar

5 

Moran I, Akerman I, van de Bunt M, Xie R, Benazra M, Nammo T, Arnes L, Nakić N, García-Hurtado J, Rodríguez-Seguí S, et al: Human β cell transcriptome analysis uncovers lncRNAs that are tissue-specific, dynamically regulated, and abnormally expressed in type 2 diabetes. Cell Metab. 16:435–448. 2012.PubMed/NCBI View Article : Google Scholar

6 

Zha F, Qu X, Tang B, Li J, Wang Y, Zheng P, Ji T, Zhu C and Bai S: Long non-coding RNA MEG3 promotes fibrosis and inflammatory response in diabetic nephropathy via miR-181a/Egr-1/TLR4 axis. Aging (Albany NY). 11:3716–3730. 2019.PubMed/NCBI View Article : Google Scholar

7 

Wang S, Chen X, Wang M, Yao D, Chen T, Yan Q and Lu W: Long non-coding RNA CYP4B1-PS1-001 inhibits proliferation and fibrosis in diabetic nephropathy by interacting with nucleolin. Cell Physiol Biochem. 49:2174–2187. 2018.PubMed/NCBI View Article : Google Scholar

8 

Cheng J, Cheng L, Tang Y, Li H, Peng W and Huang S: Inhibition of lncRNA Dlx6os1 decreases cell proliferation and fibrosis and increases cell apoptosis in diabetic nephropathy. Int J Clin Exp Pathol. 11:3302–3309. 2018.PubMed/NCBI

9 

Kim D, Paggi JM, Park C, Bennett C and Salzberg SL: Graph-based genome alignment and genotyping with HISAT2 and HISAT-genotype. Nat Biotechnol. 37:907–915. 2019.PubMed/NCBI View Article : Google Scholar

10 

The R Foundation for Statistical Computing, Vienna, Austria. URL http://www.R-project.org.

11 

Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, Paulovich A, Pomeroy SL, Golub TR, Lander ES and Mesirov JP: Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci USA. 102:15545–15550. 2005.PubMed/NCBI View Article : Google Scholar

12 

Zheng M, Lv LL, Cao YH, Liu H, Ni J, Dai HY, Liu D, Lei XD and Liu BC: A pilot trial assessing urinary gene expression profiling with an mRNA array for diabetic nephropathy. PLoS One. 7(e34824)2012.PubMed/NCBI View Article : Google Scholar

13 

Barutta F, Bruno G, Grimaldi S and Gruden G: Inflammation in diabetic nephropathy: Moving toward clinical biomarkers and targets for treatment. Endocrine. 48:730–742. 2015.PubMed/NCBI View Article : Google Scholar

14 

Duran-Salgado MB and Rubio-Guerra AF: Diabetic nephropathy and inflammation. World J Diabetes. 5:393–398. 2014.PubMed/NCBI View Article : Google Scholar

15 

Gao L, Sun N, Xu Q, Jiang Z and Li C: Comparative analysis of mRNA expression profiles in type 1 and type 2 diabetes mellitus. Epigenomics. 11:685–699. 2019.PubMed/NCBI View Article : Google Scholar

16 

Yi W and OuYang Q: Adiponectin improves diabetic nephropathy by inhibiting necrotic apoptosis. Arch Med Sci. 15:1321–1328. 2019.PubMed/NCBI View Article : Google Scholar

17 

Wang T, Gao Y, Wang X, Shi Y, Xu J, Wu B, He J and Li Y: Calpain-10 drives podocyte apoptosis and renal injury in diabetic nephropathy. Diabetes Metab Syndr Obes. 12:1811–1820. 2019.PubMed/NCBI View Article : Google Scholar

18 

Tsai YC, Kuo PL, Hung WW, Wu LY, Wu PH, Chang WA, Kuo MC and Hsu YL: Angpt2 induces mesangial cell apoptosis through the MicroRNA-33-5p-SOCS5 loop in diabetic nephropathy. Mol Ther Nucleic Acids. 13:543–555. 2018.PubMed/NCBI View Article : Google Scholar

19 

Yi H, Peng R, Zhang LY, Sun Y, Peng HM, Liu HD, Yu LJ, Li AL, Zhang YJ, Jiang WH and Zhang Z: LincRNA-Gm4419 knockdown ameliorates NF-κB/NLRP3 inflammasome-mediated inflammation in diabetic nephropathy. Cell Death Dis. 8(e2583)2017.PubMed/NCBI View Article : Google Scholar

20 

Wang M, Wang S, Yao D, Yan Q and Lu W: A novel long non-coding RNA CYP4B1-PS1-001 regulates proliferation and fibrosis in diabetic nephropathy. Mol Cell Endocrinol. 426:136–145. 2016.PubMed/NCBI View Article : Google Scholar

21 

Han F, Xue M, Chang Y, Li X, Yang Y, Sun B and Chen L: Triptolide suppresses glomerular mesangial cell proliferation in diabetic nephropathy is associated with inhibition of PDK1/Akt/mTOR pathway. Int J Biol Sci. 13:1266–1275. 2017.PubMed/NCBI View Article : Google Scholar

22 

Li W, Wang P, Li Y, Zhang K, Ding F, Nie T, Yang X, Lv Q and Zhao L: Identification of MicroRNAs in response to different day lengths in soybean using high-throughput sequencing and qRT-PCR. PLoS One. 10(e0132621)2015.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

October-2020
Volume 20 Issue 4

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Cheng L, Cheng J, Peng W, Jiang X and Huang S: Long non‑coding RNA Dlx6os1 serves as a potential treatment target for diabetic nephropathy via regulation of apoptosis and inflammation . Exp Ther Med 20: 3791-3797, 2020
APA
Cheng, L., Cheng, J., Peng, W., Jiang, X., & Huang, S. (2020). Long non‑coding RNA Dlx6os1 serves as a potential treatment target for diabetic nephropathy via regulation of apoptosis and inflammation . Experimental and Therapeutic Medicine, 20, 3791-3797. https://doi.org/10.3892/etm.2020.9112
MLA
Cheng, L., Cheng, J., Peng, W., Jiang, X., Huang, S."Long non‑coding RNA Dlx6os1 serves as a potential treatment target for diabetic nephropathy via regulation of apoptosis and inflammation ". Experimental and Therapeutic Medicine 20.4 (2020): 3791-3797.
Chicago
Cheng, L., Cheng, J., Peng, W., Jiang, X., Huang, S."Long non‑coding RNA Dlx6os1 serves as a potential treatment target for diabetic nephropathy via regulation of apoptosis and inflammation ". Experimental and Therapeutic Medicine 20, no. 4 (2020): 3791-3797. https://doi.org/10.3892/etm.2020.9112