Kidney Res Clin Pract > Epub ahead of print
Rong, Xue, Hao, Liu, and Xu: Long non-coding RNA MEG3 silencing weakens high glucose-induced mesangial cell injury by decreasing LIN28B expression by sponging and sequestering miR-23c

Abstract

Background

Diabetic nephropathy (DN) is a common kidney disease in diabetic patients. Long non-coding RNA maternally expressed gene 3 (MEG3) and microRNA (miR)-23c are reported to be implicated in DN development. Nevertheless, it is unclear that the molecular mechanism between MEG3 and miR-23c in DN remains unclear.

Methods

Human mesangial cells (HMCs) were treated with high glucose (HG) to simulate the DN status in vitro. Expression of MEG3 and miR-23c was measured. Effects of MEG3 silencing on HG-stimulated HMC injury were determined. The relationship between MEG3 and miR-23c was verified by the dual-luciferase reporter and RNA immunoprecipitation assays.

Results

MEG3 was overexpressed in serums from DN patients and HG-stimulated HMCs. MEG3 knockdown weakened HG-stimulated HMC proliferation, extracellular matrix (ECM) accumulation, and inflammation. MEG3 regulated lin-28 homolog B (LIN28B) expression through adsorbing miR-23c. MiR-23c inhibitor reversed MEG3 knockdown-mediated effects on HG-stimulated HMC proliferation, ECM accumulation, and inflammation. LIN28B overexpression overturned miR-23c mimic-mediated effects on HG-stimulated HMC proliferation, ECM accumulation, and inflammation.

Conclusion

MEG3 regulated HMC injury via regulation of the miR-23c/LIN28B axis in DN, which can help us better understand the mechanism of DN mediated by MEG3.

Introduction

Diabetic nephropathy (DN), a common diabetic complication, is the main cause of end-stage renal disease [1]. DN is related to the structural changes of the kidney, which is manifested by the thickening of the glomerular basement membrane and the accumulation of extracellular matrix (ECM) [2]. Currently, hyperglycemia is recognized as the main driving force for the development of DN. Studies have uncovered that hyperglycemia can cause inflammation, which promotes the development of DN [3,4]. Also, high glucose (HG) induces proliferation and ECM accumulation in mesangial cells [5,6]. As reported, epithelial-to-mesenchymal transition (EMT) accelerates the accumulation of ECM components [7,8].
Experiments and clinical studies on mesangial cells have confirmed that the phenotypic transformation of mesangial cells exists in DN, characterized by mesangial hypertrophy and massive secretion of ECM [9,10]. Phenotypic transformation of the mesangium may be the cytological basis for the gradual accumulation of ECM in the glomerulus, eventually resulting in glomerulosclerosis [11,12]. Therefore, exploring the pathogenesis of DN using human mesangial cells (HMCs) is of great significance for understanding the pathogenesis of the disease.
Long non-coding RNAs (lncRNAs) are a type of non-coding transcripts that exert vital roles in the development of many diseases, including DN [13,14]. For instance, lncRNA NR_033515 facilitated DN progression via accelerating proliferation, EMT, and fibrogenesis of mesangial cells [15]. Also, lncRNA plasmacytoma variant translocation 1 could contribute to podocyte damage and apoptosis in DN [16]. LncRNA maternally expressed gene 3 (MEG3) is located at chromosome 14q32.3 [17]. It has been reported that MEG3 plays a vital role in some diseases. Mounting studies revealed the tumor-inhibiting role of MEG3 in diverse cancers [18]. MEG3 could protect endothelial function by modulating the DNA damage response [19]. Also, MEG3 played an inhibitory role by inactivating the AKT/mTOR pathway in rheumatoid arthritis [20]. In DN, MEG3 could alleviate proliferation, fibrosis, and inflammation of mesangial cells [21,22]. However, the regulatory mechanism of MEG3 on HG-induced mesangial cell damage has not been fully elucidated.
According to the competing endogenous RNA (ceRNA) hypothesis, lncRNAs act as microRNA (miR) sponges and can bind miRs competitively to indirectly regulate messenger RNAs (mRNAs) using shared miR response elements. MiRs can modulate gene expression by inducing translational repression or degradation [23]. MiRs are implicated in almost all pathological and physiological processes, including cell differentiation, immune inflammation, apoptosis, and proliferation [24]. Recent studies have revealed that miRs have great potential as therapeutic targets and diagnostic markers in many diseases, including DN [24]. The starBase predicted that many miRNAs may interact with MEG3. By reviewing previously reported literature, we searched for five miRNAs (miR-145-5p, miR-143-3p, miR-181a-5p, miR-485-5p, and miR-23c) that are low expressed and inhibit mesangial cell proliferation in DN mesangial cells. The miR-23c with the largest decrease in MEG3 overexpression in cells had been selected for further analysis (Supplementary Fig. 1, available online). MiR-23c plays a tumor-repressive role in endometrial cancer [25] and hepatocellular cancer [26]. In DN, high miR-23c expression inhibits pyroptosis of renal tubular epithelial [27] and decreases proliferation, fibrosis, and EMT of mouse mesangial cells [28]. However, the mechanism of miR-23c dysregulation in mesangial cells under HG stimulation is unclear.
Herein, the relationship between MEG3 and miR-23c in HG-induced mesangial cell damage was explored. Our results uncovered that MEG3 inhibition reduced HG-induced proliferation, ECM accumulation, and inflammation in HMCs via upregulating lin-28 homolog B (LIN28B) through adsorbing miR-23c.

Methods

Serum samples

Peripheral blood (5 mL) was obtained from 25 healthy controls (normal) and 25 type 2 diabetes with DN. Twenty-five kidney tissues were collected from type 2 diabetes with DN through ultrasound-guided kidney biopsy. Twenty-five normal kidney samples were obtained from patients who underwent renal tumor resection. All recruited subjects were diagnosed or underwent basic examination at the People’s Hospital of Zhengzhou University. The permission for this research was approved by the Ethics Committee of People’s Hospital of Zhengzhou University (No. 20210628), and all participants signed informed consent. The clinical and biochemical characteristics are exhibited in Table 1.

Cell culture and treatment

HMCs (BNCC341276; BNCC) were cultured in Dulbecco’s modified eagle medium (DMEM; Sigma-Aldrich) supplemented with glucose (5 mM, #G8270; Sigma-Aldrich), fetal bovine serum (FBS; #10100147, 10%; Life Technologies [LT]), and strep/penicillin (#P0781, 1%; Sigma-Aldrich). For HG stimulation, 30 mM of glucose was used, with 5 mM of glucose serving as a control.

Isolation of human primary mesangial cells

The separated cortical tissue on the superficial surface of the kidney is finely cut into extremely fine pieces and lightly ground with a tissue grinder. The cell suspension was filtered on a 200 mesh stainless steel screen and washed three times with D-Hank’s solution containing penicillin-streptomycin double antibody to remove blood cells as much as possible. The tissue on the sieve was digested using a DMEM medium containing 0.15% type IV collagenase (Sigma-Aldrich). After removing the supernatant, add an appropriate amount of DMEM medium and centrifuge again to obtain the cell precipitation. The cells were cultured in DMEM supplemented with 20% FBS.

Cell transfection

Oligonucleotides or vectors were transfected into HMCs using Lipofectamine 3000 reagent (#L3000001; Invitrogen). Small interfering (si) RNA against MEG3 (si-MEG3: GGUUGUUGUGAGAAUUAAAUG) and its negative control (NC) (si-NC: 5’-UUCUCCGAACGUGUCACGUTT-3’), miR-23c inhibitor and mimic (anti-miR-23c: 5’-GGGUAAUCACUGGCAAUGUGAU-3’ and miR-23c: 5’-AUCACAUUGCCAGUGAUUACCC-3’), anti-miR-NC (5’-CGGUUCGCTAGCGCCGACUCG-3’), and miR-NC (5’-CGAUCGCAUCAGCAUCGAUUGC-3’) were purchased from RiboBio. The full-length complementary DNA (cDNA) of MEG3 (NR_002766) or LIN28B (NM_001004317) was digested with KpnI-XhoI or BamHI-NotI and then ligated into the pcDNA3.1(+) vector (Life Technologies) to construct the MEG3 and LIN28B overexpression plasmid.

Quantitative real-time polymerase chain reaction

Plasma/Serum RNA Purification Mini Kit (#RNB700; Sigma-Aldrich) and RNeasy Mini Kit (#74104; QIAGEN) were used to isolate total RNA from serum samples or HMCs. The M-MLV First Strand Kit (#28025013; Life Technologies) was for cDNA generation. Quantitative PCR was conducted with the SYBR Green PCR Master Mix (#1725270; Bio-Rad). The 2–ΔΔCt method was applied to figure the expression of MEG3, LIN28B, and miR-23c in serum samples or HMCs with β-actin or U6 as a housekeeping gene, and the primers were added to Table 2.

Cell proliferation analysis

The proliferation of HMCs was analyzed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and 5-ethynyl-2’-deoxyuridine (EdU) assays. For MTT assays, HMCs (5 × 103 cells/well) were cultured for 24 hours, 48 hours, or 72 hours. Then, the MTT solution (#CT01-5, 100 μL, 0.5 mg/mL; Sigma-Aldrich) was supplemented into each well. Dimethylsulfoxide (#D8418, 150 μL; Sigma-Aldrich) was added to each well to dissolve the crystals after 4 hours of incubation. Next, the absorbance at 570 nm was analyzed by Microplate Reader (Bio-Rad). The EdU assay was performed using the Cell-Light EdU Kit (#C10310-1; RiboBio) based on the manufacturer’s instructions. Nuclear staining was performed with DAPI (#MBD0015; Sigma-Aldrich), and the cells were observed under a fluorescence microscope (Olympus).

Cell cycle progression analysis

In brief, HMCs were harvested and fixed with ethanol (70%) at 4 °C overnight, followed by incubation with RNase A containing propidium iodide (40%, #81845; Sigma-Aldrich). Cell distribution was assessed with FACScan flow cytometry (BD Biosciences) and FACS Diva Software (BD Biosciences).

Western blotting

HMCs were lysed in radioimmunoprecipitation assay buffer containing a protease inhibitor cocktail (Roche). Total protein of 40 μg was isolated by 10% sodium dodecyl sulfate-polyacrylamide gel electrophoresis and then transferred to polyvinylidene difluoride (PVDF) membranes (Roche). After blocking with tris buffered saline tween buffer with 5% skim milk, the PVDF membranes were incubated with primary antibodies. Thereafter, the PVDF membranes were incubated with second antibodies. The primary antibodies were exhibited as follows: anti-glyceraldehyde-3-phosphate dehydrogenase (GAPDH) (ab9484), anti-LIN28B (ab115698), anti-N-cadherin (sc-8424), anti-E-cadherin (sc-8426), anti-collagen IV (sc-59814), and anti-fibronectin (sc-18825). Protein bands were visualized with an ImmunoStar LD (Wako Pure Chemical). The primary antibodies for LIN28B and GAPDH were bought from Abcam, and other antibodies were purchased from Santa Cruz Biotechnology. All original protein bands of western blotting were exhibited in Supplementary Fig. 2 (available online).

Enzyme-linked immunosorbent assay

The levels of interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-α) were detected with the enzyme-linked immunosorbent assay (ELISA) kits (R&D Systems). Briefly, the supernatants of HMCs were collected after culture for 48 hours, followed by the analysis of the absorbance with the Microplate Reader (Bio-Rad).

Dual-luciferase reporter assay

The binding sites of MEG3 or LIN28B in miR-23c were predicted with the starBase database. The luciferase reporters containing wild type (WT)-MEG3, mutant (MUT)-MEG3, LIN28B 3’ untranslated regions (UTR)-WT, or LIN28B 3’ UTR-MUT were synthesized by GenePharma. The luciferase reporter vectors were cotransfected into HMCs together with miR-23c or miR-NC when the cell reached 80% confluence. After 48 hours of transfection, the luciferase activities of Firefly and Renilla were measured with the luciferase reporter assay kit (Promega). The ratio of Firefly luciferase activity to Renilla luciferase activity was the relative luciferase activity.

RNA immunoprecipitation assay

RNA immunoprecipitation (RIP) analysis was executed with the Magna RIP Kit (Millipore). Briefly, about 2 × 107 HMCs were lysed in complete RIP lysis buffer containing protease/RNase inhibitors (Roche) and then centrifuged at 16,000 ×g at 4 °C for 5 minutes. Subsequently, cell extracts were incubated with magnetic beads conjugated with anti-immunoglobulin G (ab172730; Abcam) or anti-Ago2 (ab186733; Abcam). Immunoprecipitated RNAs were isolated by TRIzol (Life Technologies) after the sample was treated with protease K at 55 °C for 30 minutes. The coprecipitated RNAs were purified and then analyzed by quantitative real-time polymerase chain reaction (qRT-PCR).

Statistical analysis

All experiments were repeated at least three times. Statistical analysis was implemented by IBM SPSS version 20.0 (IBM Corp.) with the unpaired Student t test and one-way analysis of variance. Data were exhibited as mean ± standard deviation. A p-value of <0.05 was considered statistically significant.

Results

The negative correlation between MEG3 and miR-23c in diabetic nephropathy

Previous studies have revealed that MEG3 and miR-23c exert opposite roles in DN [22,27]. We detected the expression of MEG3 and miR-23c in serums of DN patients (n = 25) and healthy controls (n = 25) to investigate the relationship between MEG3 and miR-23c. qRT-PCR exhibited higher MEG3 levels in serums derived from DN patients than healthy controls, while miR-23c expression had an adverse result (Fig. 1A, B). Pearson correlation analysis indicated a negative correlation between MEG3 and miR-23c expression in serums derived from DN patients (Fig. 1C). Additionally, we detected MEG3 and miR-23c levels in 25 DN kidney tissues and 25 normal kidney tissues. The results exhibited that MEG3 was observably upregulated in DN kidney tissues than in normal kidney tissues, while miR-23c had an opposing result (Supplementary Fig. 3, available online). MEG3 expression in patients with normoalbuminuria, diabetic microalbuminuria, and macroalbuminuria was elevated successively. However, miR-23c expression had an opposite result (Supplementary Fig. 4A, B; available online). In addition, DN patients with high MEG3 expression had higher fasting blood glucose levels, while DN patients with high miR-23c expression had lower fasting blood glucose levels (Supplementary Fig. 4C, D; available online). Subsequently, we examined MEG3 and miR-23c expression in HG-stimulated HMCs. Likewise, MEG3 expression was elevated while miR-23c expression was decreased in HG-stimulated HMCs relative to the control group (Fig. 1D, E). Taken together, the negative correlation between MEG3 and miR-23c in DN might be related to the progression of DN.

MEG3 inhibition reduced high glucose-stimulated human mesangial cell proliferation, extracellular matrix accumulation, and inflammation

Next, we explored the biological role of MEG3 in DN. qRT-PCR exhibited that transfection of si-MEG3 reversed the elevation of MEG3 in HMCs induced by HG treatment (Fig. 2A). MTT and EdU assays showed that MEG3 inhibition weakened HG stimulation-induced HMC proliferation (Fig. 2B, C). Moreover, MEG3 silencing caused HG-induced HMCs to be arrested in G0/G1 phase, suggesting that MEG3 silencing weakened HG-induced cell cycle progression in HMCs (Fig. 2D). Western blotting presented that MEG3 silencing weakened upregulation of fibronectin and collagen IV protein levels in HG-stimulated HMCs (Fig. 2E). Moreover, MEG3 inhibition reversed the downregulation of E-cadherin and the upregulation of N-cadherin in HMCs induced by HG treatment (Fig. 2F). Subsequently, ELISA exhibited that MEG3 downregulation could decrease the release of IL-6 and TNF-α in HMCs mediated by HG (Fig. 2G). Additionally, we surveyed the biological function of MEG3 in mesangial cells isolated from DN kidney tissues. The results exhibited that MEG3 knockdown reduced proliferation and ECM accumulation of mesangial cells in vitro (Supplementary Fig. 5, available online). Together, these findings indicated that MEG3 silencing decreased HMC injury induced by HG treatment.

MEG3 was verified as a miR-23c sponge

Based on the opposite expression levels of MEG3 and miR-23c in DN, we speculated that MEG3 might be a sponge of miR-23c. Online bioinformatics prediction (starBase) exhibited that MEG3 and miR-23c had complementary binding sites (Fig. 3A). And miR-23c overexpression could reduce the luciferase activity of the luciferase reporter with WT-MEG3 (Fig. 3B). RIP assay manifested that miR-23c and MEG3 were enriched in the anti-Ago2 group (Fig. 3C). After MEG3 transfection, MEG3 expression was intensified in HG-stimulated HMCs (Fig. 3D). Furthermore, MEG3 inhibition weakened the decrease in miR-23c expression in HG-stimulated HMCs, but MEG3 overexpression had an opposing effect (Fig. 3E). Collectively, MEG3 acted as a miR-23c sponge.

MiR-23c inhibitor abolished MEG3 downregulation-mediated effects on high glucose-induced human mesangial cell injury

Subsequently, we further surveyed whether miR-23c was implicated in DN progression regulated by MEG3. qRT-PCR showed that introduction of anti-miR-23c alleviated MEG3 knockdown-mediated downregulation of miR-23c in HMCs induced by HG (Fig. 4A). Moreover, MEG3 silencing resulted in the impaired ability of cell proliferation and the arrest of cell cycle progression in the G0/G1 phase of HG-induced HMCs, but these effects mediated by MEG3 inhibition were lightened after miR-23c silencing (Fig. 4BE). Also, miR-23c downregulation overturned MEG3 silencing-mediated inhibition on ECM accumulation, EMT, and inflammation of HMCs under HG treatment (Fig. 4FH). Therefore, these findings revealed that MEG3 could modulate HG-induced HMC injury via sponging miR-23c.

MiR-23c directly targeted LIN28B in human mesangial cells

We predicted the downstream target of miR-23c through the starBase database. As exhibited in Fig. 5A, LIN28B possessed complementary binding sites with miR-23c. We also observed that the luciferase activity of luciferase reporters with LIN28B 3’UTR-WT was reduced in HMCs after co-transfection with miR-23c (Fig. 5B). Moreover, miR-23c and LIN28B enrichment were higher in the anti-Ago2 group (Fig. 5C). Also, LIN28B mRNA levels were also increased in serums from DN patients (Fig. 5D). LIN28B mRNA and miR-23c expression had a negative correlation in serums of DN patients (Fig. 5E). IHC staining also showed the elevated expression of LIN28B in human kidney samples (Supplementary Fig. 6, available online). Analogously, LIN28B protein levels were elevated in HG-induced HMCs (Fig. 5F). These results suggested that LIN28B was a downstream target of miR-23c in HMCs.

LIN28B overexpression overturned miR-23c mimic-mediated impacts on high glucose-induced human mesangial cell injury

Considering the above results, we further checked whether miR-23c regulated HMCs injury via targeting LIN28B. Western blotting displayed that LIN28B introduction impaired downregulation of LIN28B in HG-stimulated HMCs mediated by miR-23c mimic (Fig. 6A). Also, miR-23c mimic-mediated inhibition of cell proliferation and arrest of cell cycle progression in the G0/G1 phase were reversed by LIN28B elevation in HG-stimulated HMCs (Fig. 6BE). Furthermore, forcing LIN28B expression overturned the decrease of ECM accumulation, EMT, and inflammation in HG-stimulated HMCs mediated by miR-23c overexpression (Fig. 6FH). These data indicated that miR-23c modulated HG-induced proliferation, ECM accumulation, and inflammation via targeting LIN28B in HMCs.

LIN28B was regulated by the MEG3/miR-23c axis in human mesangial cells

Given that lncRNA can function as a ceRNA in the advancement of some diseases, we further analyzed whether MEG3 regulated LIN28B expression via binding to miR-23c. The results exhibited that MEG3 knockdown decreased LIN28B mRNA and protein levels in HG-stimulated HMCs, while this reduction was relieved by miR-23c downregulation (Fig. 7). These data manifested that MEG3 regulated LIN28B expression via binding to miR-23c.

Discussion

Increasing evidence has manifested that lncRNAs can be used as promising therapeutic targets and diagnostic biomarkers for DN [29]. HG is characteristic of DN, which can induce proliferation, EMT, ECM accumulation, and inflammation of HMCs [30,31]. EMT, a phenotypic change process, is accompanied by the loss of E-cadherin and the rise of N-cadherin and is a factor that directly leads to ECM accumulation, especially in DN [32]. Recent studies revealed that MEG3 silencing could induce apoptosis, curb proliferation, and decrease fibrosis of HMCs by sponging miR-145 under HG treatment [21]. Another report uncovered that MEG3 contributed to inflammation and fibrosis of HMCs via regulation of the miR-181a/Egr-1/TLR4 axis [22]. In this study, MEG3 was highly expressed in DN. MEG3 downregulation decreased HMC proliferation, ECM accumulation, and inflammation under HG treatment, which was consistent with the previous studies [21,22], indicating that HG-induced HMC injury relied on MEG3-mediated mechanisms.
More and more evidence showed that lncRNAs regulate gene expression by competitively binding to miRs, thereby participating in the development of various diseases [33]. In DN, lncRNA MALAT1 accelerated pyroptosis of renal tubular epithelial through elevating ELAVL1 expression via sponging miR-23c [27]. Also, lncRNA NEAT1 adsorbed miR-23c in mouse mesangial cells, resulting in facilitating cell proliferation, fibrosis, and EMT [28]. These data indicated that miR-23c played a protective role in the pathogenesis of DN. Our results manifested the sponge action of MEG3 on miR-23c in HMCs. Moreover, miR-23c inhibitor abolished the suppressive impact of MEG3 knockdown on HG-stimulated HMC proliferation, ECM accumulation, and inflammation. Therefore, we inferred that HG-induced MEG3 modulated HMC injury via sponging miR-23c.
LIN28B, an RNA-binding protein, is an important regulator of stem cell maintenance [34] and tumorigenesis [35]. A study revealed that miR-152 could repress HG-induced angiogenesis by targeting LIN28B and inhibiting vascular endothelial growth factor signaling [36]. In DN, transforming growth factor-β1 induced collagen accumulation via the LIN28B/let-7 pathway [37]. Furthermore, miR-379-5p inhibited renal fibrosis through the regulation of the LIN28B/let-7 pathway [38]. Herein, we verified that LIN28B acted as a miR-23c target. LIN28B overexpression overturned the suppressive influence of miR-23c mimic on HG-induced HMC injury. Moreover, MEG3 sponged miR-23c to regulate LIN28B expression. Therefore, we concluded that HG-induced MEG3 regulated proliferation, ECM accumulation, and inflammation via regulating LIN28B expression by sponging miR-23c.
In summary, HG-induced MEG3 adsorbed miR-23c to elevate LIN28B expression, resulting in promoting proliferation, ECM accumulation, and inflammation of HMCs. The study provided a novel mechanism responsible for HG-induced mesangial cell damage.

Notes

Conflicts of interest

All authors have no conflicts of interest to declare.

Data sharing statement

The analyzed data sets generated during the present study are available from the corresponding author on reasonable request.

Authors’ contributions

Conceptualization, Methodology: HX, JH

Data curation, Formal analysis: JH, JL, HX

Investigation, Validation: LR, HX

Writing–original draft preparation: LR, HX, JH

Writing–review & editing: LR, HX, JH

All authors read and approved the final manuscript.

Figure 1.

Expression patterns of MEG3 and miR-23c in DN and HG-stimulated HMCs.

(A, B) Expression levels of MEG3 and miR-23c in serums derived from DN patients (n = 25) and healthy controls (n = 25). (C) The correlation between MEG3 and miR-23c in the serum of DN patients (Pearson correlation analysis). (D, E) Expression levels of MEG3 and miR-23c in HG-stimulated HMCs and control group.
DN, diabetic nephropathy; HMC, human mesangial cell; HG, high glucose; MEG3, maternally expressed gene 3; miR, microRNA.
***p < 0.001 and ****p < 0.0001.
j-krcp-23-090f1.jpg
Figure 2.

Effects of MEG3 downregulation on HG-induced HMC injury.

(A–F) HMCs were transfected with si-MEG3 or si-NC and then treated with HG. (A) qRT-PCR analysis of MEG3 expression in HMCs. (B–D) MTT, EdU, and flow cytometry assays were performed to evaluate the proliferation and cell cycle progression of HMCs. (E, F) Western blotting analysis of fibronectin, collagen IV, E-cadherin, and N-cadherin protein levels in HMCs. (G) Levels of IL-6 and TNF-α in HMCs.
EdU, 5-ethynyl-2'-deoxyuridine; GAPDH, glyceraldehyde-3-phosphate dehydrogenase; HMC, human mesangial cell; HG, high glucose; IL, interleukin; MEG3, maternally expressed gene 3; MTT, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide; NC, negative control; NS, not significant; OD, optical density; qRT-PCR, quantitative reverse transcription polymerase chain reaction; si, small interfering; TNF, tumor necrosis factor.
*p < 0.05, **p < 0.01, ***p < 0.001, and ****p < 0.0001.
j-krcp-23-090f2.jpg
Figure 3.

MEG3 was a sponge for miR-23c in HMCs.

(A) The diagram showed the complementary binding sites of miR-23c and MEG3. (B) Dual-luciferase reporter assay was performed to verify the binding sites between miR-23c and MEG3. (C) Levels of miR-23c and MEG3 in RNA-induced silencing complexes were analyzed. (D) Expression of MEG3 in HMCs with pcDNA or MEG3 under HG treatment. (E) Expression of miR-23c in HMCs with si-NC, si-MEG3, pcDNA, or MEG3 under HG treatment.
HG, high glucose; HMC, human mesangial cell; IgG, immunoglobulin G; MEG3, maternally expressed gene 3; miR, microRNA; MUT, mutant; NC, negative control; pcDNA, plasmid DNA; si, small interfering; WT, wild type.
***p < 0.001 and ****p < 0.0001.
j-krcp-23-090f3.jpg
Figure 4.

MEG3 accelerated HG-induced HMC injury via sponging miR-23c.

(A–H) After si-NC, si-MEG3, si-MEG3 + anti-miR-NC, or si-MEG3 + anti-miR-23c transfection, HMCs were treated with HG. (A) qRT-PCR analysis of miR-23c expression in HMCs. (B–E) The proliferation and cell cycle progression of HMCs were determined (D, original magnification of ×200). (F, G) Protein levels of fibronectin, collagen IV, E-cadherin, and N-cadherin in HMCs. (H) Levels of IL-6 and TNF-α in HMCs were measured.
EdU, 5-ethynyl-2'-deoxyuridine; GAPDH, glyceraldehyde-3-phosphate dehydrogenase; HMC, human mesangial cell; HG, high glucose; IL, interleukin; MEG3, maternally expressed gene 3; miR, microRNA; NC, negative control; OD, optical density; qRT-PCR, quantitative reverse transcription polymerase chain reaction; si, small interfering; TNF, tumor necrosis factor.
*p < 0.05, **p < 0.01, ***p < 0.001, and ****p < 0.0001.
j-krcp-23-090f4.jpg
Figure 5.

LIN28B served as a target for miR-23c in HMCs.

(A) The diagram exhibited the complementary binding sites between LIN28B and miR-23c. (B) Dual-luciferase reporter assay was executed to check the binding sites between LIN28B and miR-23c. (C) Levels of LIN28B and miR-23c in RNA-induced silencing complexes were examined. (D) Expression of LIN28B mRNA in serums of DN patients. (E) The correlation between LIN28B and miR-23c in serums of DN patients (Pearson correlation analysis). (F) Western blotting analysis of LIN28B protein levels in HG-induced HMCs.
DN, diabetic nephropathy; GAPDH, glyceraldehyde 3-phosphate dehydrogenase; HG, high glucose; HMC, human mesangial cell; IgG, immunoglobulin G; LIN28B, lin-28 homolog B; mRNA, messenger RNA; MUT, mutant; miR, microRNA; UTR, untranslated region; WT, wild type.
***p < 0.001 and ****p < 0.0001.
j-krcp-23-090f5.jpg
Figure 6.

MiR-23c targeted LIN28B to regulate HG-induced HMC injury.

(A–H) After miR-NC, miR-23c, miR-23c + pcDNA, or miR-23c + LIN28B transfection, HMCs were treated with HG. (A) Protein levels of LIN28B protein in HMCs were analyzed. (B–E) The proliferation and cell cycle progression of HMCs were evaluated (D, original magnification of ×200). (F, G) Western blotting presented the levels of fibronectin, collagen IV, E-cadherin, and N-cadherin in HMCs. (H) Levels of IL-6 and TNF-α in HMCs.
EdU, 5-ethynyl-2'-deoxyuridine; GAPDH, glyceraldehyde 3-phosphate dehydrogenase; HG, high glucose; HMC, human mesangial cell; IL, interleukin; LIN28B, lin28 homolog B; miRc, microRNA; NC, negative control; OD, optical density; pcDNA, plasmid DNA; TNF, tumor necrosis factor.
*p < 0.05, **p < 0.01, ***p < 0.001, and ****p < 0.0001.
j-krcp-23-090f6.jpg
Figure 7.

MEG3 modulated LIN28B expression via sponging miR-23c.

(A, B) After HG treatment, LIN28B mRNA and protein levels in HMCs with si-NC, si-MEG3, si-MEG3 + anti-miR-NC, or si-MEG3 + anti-miR-23c were examined.
GAPDH, glyceraldehyde-3-phosphate dehydrogenase; HMC, human mesangial cell; HG, high glucose; LIN28B, lin28 homolog B; MEG3, maternally expressed gene 3; miR, microRNA; mRNA, messenger RNA; NC, negative control; si, small interfering.
**p < 0.01, ***p < 0.001, and ****p < 0.0001.
j-krcp-23-090f7.jpg
Table 1.
Clinical and biochemical characteristics of DN patients and healthy controls
Parameter Normal group DN group
No. of patients 25 25
Sex, male:female 12:13 11:14
Age (yr) 56.3 ± 6.3 58.7 ± 5.8
Duration of diabetes (yr) None 7.4 ± 1.2
Fasting plasma glucose (mmol/L) 4.2 ± 1.1 7.2 ± 2.2
Blood urea nitrogen (mmol/L) 4.3 ± 1.5 7.6 ± 4.1
HbA1c (mmol/mol) 41.2 ± 12.6 72.3 ± 9.6
Body mass index (kg/m2) 22.6 ± 1.8 24.2 ± 2.3
Creatinine (µmol/L) 82.1 ± 2.5 86.3 ± 5.6
eGFR (mL/min) 89.3 ± 9.6 62.7 ± 8.4
Albuminuria (mg/day) 4.7 ± 1.6 67.5 ± 22.5

Data are expressed as number only or mean ± standard deviation.

DN, diabetic nephropathy; eGFR, estimated glomerular filtration rate; HbA1c, glycated hemoglobin.

Table 2.
Primer sequences for quantitative real-time polymerase chain reaction
Gene name Forward primer (5’-3’) Reverse primer (5’-3’)
MEG3 CCGTCCACCTCCTTGTCTTC CTCGTGAGGTGTAGATGGGC
LIN28B CCTGTTTAGGAAGTGAAAGAAGAC CACTTCTTTGGCTGAGGAGGTAG
GAPDH GATTCCACCCATGGCAAATTCC TCGCTCCTGGAAGATGGTGAT
miR-23c CGCGATCACATTGCCAGTGA AGTGCAGGGTCCGAGGTATT
U6 GCTTCGGCAGCACATATACTAAAAT CGCTTCACGAATTTGCGTGTCAT

References

1. Umanath K, Lewis JB. Update on diabetic nephropathy: core curriculum 2018. Am J Kidney Dis 2018;71:884–895.
crossref pmid
2. Wolf G. New insights into the pathophysiology of diabetic nephropathy: from haemodynamics to molecular pathology. Eur J Clin Invest 2004;34:785–796.
crossref pmid
3. Bhattacharjee N, Barma S, Konwar N, Dewanjee S, Manna P. Mechanistic insight of diabetic nephropathy and its pharmacotherapeutic targets: an update. Eur J Pharmacol 2016;791:8–24.
crossref pmid
4. Magee C, Grieve DJ, Watson CJ, Brazil DP. Diabetic nephropathy: a tangled web to unweave. Cardiovasc Drugs Ther 2017;31:579–592.
crossref pmid pmc pdf
5. Huang H, Huang H, Li Y, et al. Gremlin induces cell proliferation and extra cellular matrix accumulation in mouse mesangial cells exposed to high glucose via the ERK1/2 pathway. BMC Nephrol 2013;14:33.
crossref pmid pmc pdf
6. Chen B, Li Y, Liu Y, Xu Z. circLRP6 regulates high glucose-induced proliferation, oxidative stress, ECM accumulation, and inflammation in mesangial cells. J Cell Physiol 2019;234:21249–21259.
crossref pmid pdf
7. Thiery JP, Acloque H, Huang RY, Nieto MA. Epithelial-mesenchymal transitions in development and disease. Cell 2009;139:871–890.
crossref pmid
8. Gonzalez DM, Medici D. Signaling mechanisms of the epithelial-mesenchymal transition. Sci Signal 2014;7:re8.
crossref pmid pmc
9. Gao P, Li L, Yang L, et al. Yin Yang 1 protein ameliorates diabetic nephropathy pathology through transcriptional repression of TGFβ1. Sci Transl Med 2019;11:eaaw2050.
crossref pmid
10. Kim D, Nam GY, Seo E, Jun HS. Inhibition of ChREBP ubiquitination via the ROS/Akt-dependent downregulation of Smurf2 contributes to lysophosphatidic acid-induced fibrosis in renal mesangial cells. J Biomed Sci 2022;29:31.
crossref pmid pmc pdf
11. Li A, Peng R, Sun Y, Liu H, Peng H, Zhang Z. LincRNA 1700020I14Rik alleviates cell proliferation and fibrosis in diabetic nephropathy via miR-34a-5p/Sirt1/HIF-1α signaling. Cell Death Dis 2018;9:461.
crossref pmid pmc pdf
12. Qiao S, Liu R, Lv C, et al. Bergenin impedes the generation of extracellular matrix in glomerular mesangial cells and ameliorates diabetic nephropathy in mice by inhibiting oxidative stress via the mTOR/β-TrcP/Nrf2 pathway. Free Radic Biol Med 2019;145:118–135.
crossref pmid
13. Shi X, Sun M, Liu H, Yao Y, Song Y. Long non-coding RNAs: a new frontier in the study of human diseases. Cancer Lett 2013;339:159–166.
crossref pmid
14. Li Y, Xu K, Xu K, Chen S, Cao Y, Zhan H. Roles of identified long noncoding RNA in diabetic nephropathy. J Diabetes Res 2019;2019:5383010.
crossref pmid pmc pdf
15. Gao J, Wang W, Wang F, Guo C. LncRNA-NR_033515 promotes proliferation, fibrogenesis and epithelial-to-mesenchymal transition by targeting miR-743b-5p in diabetic nephropathy. Biomed Pharmacother 2018;106:543–552.
crossref pmid
16. Liu DW, Zhang JH, Liu FX, et al. Silencing of long noncoding RNA PVT1 inhibits podocyte damage and apoptosis in diabetic nephropathy by upregulating FOXA1. Exp Mol Med 2019;51:1–15.
crossref pdf
17. Miyoshi N, Wagatsuma H, Wakana S, et al. Identification of an imprinted gene, Meg3/Gtl2 and its human homologue MEG3, first mapped on mouse distal chromosome 12 and human chromosome 14q. Genes Cells 2000;5:211–220.
crossref pmid pdf
18. Ghafouri-Fard S, Taheri M. Maternally expressed gene 3 (MEG3): a tumor suppressor long non coding RNA. Biomed Pharmacother 2019;118:109129.
crossref pmid
19. Shihabudeen Haider Ali MS, Cheng X, Moran M, et al. LncRNA Meg3 protects endothelial function by regulating the DNA damage response. Nucleic Acids Res 2019;47:1505–1522.
crossref pmid
20. Li G, Liu Y, Meng F, et al. LncRNA MEG3 inhibits rheumatoid arthritis through miR-141 and inactivation of AKT/mTOR signalling pathway. J Cell Mol Med 2019;23:7116–7120.
crossref pmid pmc pdf
21. Li J, Jiang X, Duan L, Wang W. Long non-coding RNA MEG3 impacts diabetic nephropathy progression through sponging miR-145. Am J Transl Res 2019;11:6691–6698.
pmid pmc
22. Zha F, Qu X, Tang B, et al. Long non-coding RNA MEG3 promotes fibrosis and inflammatory response in diabetic nephropathy via miR-181a/Egr-1/TLR4 axis. Aging (Albany NY) 2019;11:3716–3730.
crossref pmid pmc
23. Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 2004;116:281–297.
crossref pmid
24. Mendell JT, Olson EN. MicroRNAs in stress signaling and human disease. Cell 2012;148:1172–1187.
crossref pmid pmc
25. Lu M, Ding N, Zhuang S, Li Y. LINC01410/miR-23c/CHD7 functions as a ceRNA network to affect the prognosis of patients with endometrial cancer and strengthen the malignant properties of endometrial cancer cells. Mol Cell Biochem 2020;469:9–19.
crossref pmid pdf
26. Zhang L, Wang L, Wang Y, et al. LncRNA KTN1-AS1 promotes tumor growth of hepatocellular carcinoma by targeting miR-23c/ERBB2IP axis. Biomed Pharmacother 2019;109:1140–1147.
crossref pmid
27. Li X, Zeng L, Cao C, et al. Long noncoding RNA MALAT1 regulates renal tubular epithelial pyroptosis by modulated miR-23c targeting of ELAVL1 in diabetic nephropathy. Exp Cell Res 2017;350:327–335.
crossref pmid
28. Li N, Jia T, Li YR. LncRNA NEAT1 accelerates the occurrence and development of diabetic nephropathy by sponging miR-23c. Eur Rev Med Pharmacol Sci 2020;24:1325–1337.
pmid
29. Loganathan TS, Sulaiman SA, Abdul Murad NA, et al. Interactions among non-coding RNAs in diabetic nephropathy. Front Pharmacol 2020;11:191.
crossref pmid pmc
30. Xu J, Chen Y, Xing Y, Ye S. Metformin inhibits high glucose-induced mesangial cell proliferation, inflammation and ECM expression through the SIRT1-FOXO1-autophagy axis. Clin Exp Pharmacol Physiol 2019;46:813–820.
crossref pmid pdf
31. Wu J, Liu J, Ding Y, et al. MiR-455-3p suppresses renal fibrosis through repression of ROCK2 expression in diabetic nephropathy. Biochem Biophys Res Commun 2018;503:977–983.
crossref pmid
32. Carew RM, Wang B, Kantharidis P. The role of EMT in renal fibrosis. Cell Tissue Res 2012;347:103–116.
crossref pmid pdf
33. Salmena L, Poliseno L, Tay Y, Kats L, Pandolfi PP. A ceRNA hypothesis: the Rosetta Stone of a hidden RNA language? Cell 2011;146:353–358.
crossref pmid pmc
34. Zhang J, Ratanasirintrawoot S, Chandrasekaran S, et al. LIN28 regulates stem cell metabolism and conversion to primed pluripotency. Cell Stem Cell 2016;19:66–80.
crossref pmid
35. Guo W, Hu Z, Bao Y, et al. A LIN28B tumor-specific transcript in cancer. Cell Rep 2018;22:2016–2025.
crossref pmid
36. Fu X, Ou B. miR-152/LIN28B axis modulates high-glucose-induced angiogenesis in human retinal endothelial cells via VEGF signaling. J Cell Biochem 2020;121:954–962.
crossref pmid pdf
37. Park JT, Kato M, Lanting L, et al. Repression of let-7 by transforming growth factor-β1-induced Lin28 upregulates collagen expression in glomerular mesangial cells under diabetic conditions. Am J Physiol Renal Physiol 2014;307:F1390–F1403.
crossref pmid pmc
38. Li N, Wang LJ, Xu WL, Liu S, Yu JY. MicroRNA‑379‑5p suppresses renal fibrosis by regulating the LIN28/let‑7 axis in diabetic nephropathy. Int J Mol Med 2019;44:1619–1628.
crossref pmid pmc


ABOUT
BROWSE ARTICLES
EDITORIAL POLICY
FOR CONTRIBUTORS
Editorial Office
#301, (Miseung Bldg.) 23, Apgujenog-ro 30-gil, Gangnam-gu, Seoul 06022, Korea
Tel: +82-2-3486-8736    Fax: +82-2-3486-8737    E-mail: registry@ksn.or.kr                

Copyright © 2024 by The Korean Society of Nephrology.

Developed in M2PI

Close layer