Open Access

Antagonist targeting miR‑106b‑5p attenuates acute renal injury by regulating renal function, apoptosis and autophagy via the upregulation of TCF4

Retraction in: /10.3892/ijmm.2024.5353

  • Authors:
    • Jing-Meng Hu
    • Li-Jie He
    • Peng-Bo Wang
    • Yan Yu
    • Ya-Ping Ye
    • Li Liang
  • View Affiliations

  • Published online on: July 9, 2021     https://doi.org/10.3892/ijmm.2021.5002
  • Article Number: 169
  • Copyright: © Hu 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

Acute renal injury (ARI) is a life‑threatening condition and a main contributor to end‑stage renal disease, which is mainly caused by ischemia‑reperfusion (I/R). miR‑106b‑5p is a kidney function‑related miRNA; however, whether miR‑106b‑5p regulates the progression of ARI remains unclear. The present study thus aimed to examine the effects of miR‑106b‑5p antagonist on the regulation of ARI progression. It was found that miR‑106b‑5p expression was upregulated in the renal tissue of rats with I/R‑induced ARI and in NRK‑52E rat renal proximal tubular epithelial cells subjected to hypoxia‑reoxygenation (H/R). In vitro, H/R induction suppressed the proliferation, and promoted the apoptosis and autophagy of NRK‑52E cells, whereas miR‑106b‑5p antagonist (inhibition of miR‑106b‑5p) promoted the proliferation, and attenuated the apoptosis and autophagy of NRK‑52E cells under the H/R condition. Dual luciferase reporter gene assay validated that transcription factor 4 (TCF4) was a target of miR‑106b‑5p. It was further found that TCF4 overexpression promoted the proliferation, and inhibited the apoptosis and autophagy of NRK‑52E cells subjected to H/R. Moreover, the effects of miR‑106b‑5p antagonist on NRK‑52E cell proliferation, apoptosis and autophagy were mediated through the regulation of TCF4. In vivo, miR‑106b‑5p antagonist reduced the severity of renal injury, decreased cell proliferation in renal tissues and lowered the serum creatinine (Scr) and blood urea nitrogen (BUN) levels in the blood samples from rats with I/R‑induced ARI. On the whole, the findings presented herein demonstrate that miR‑106b‑5p antagonist attenuates ARI by promoting the proliferation, and suppressing the apoptosis and autophagy of renal cells via upregulating TCF4.

Introduction

Acute renal injury (ARI) is a life-threatening condition and a main contributor to end-stage renal disease with a high incidence of 13.5 million individuals annually worldwide. ARI can be caused by ischemia-reperfusion (I/R), nephrotoxicity and sepsis (1). I/R is considered a major cause of ARI (2,3), and can lead to the loss of renal function, inflammatory responses, renal proximal tubular epithelial cell apoptosis and autophagy (4-6). Although some therapies have been shown to ameliorate the condition by reducing the inflammatory responses, and suppressing apoptosis and autophagy following I/R-induced ARI (7,8), satisfactory therapies for the treatment of I/R-induced ARI are limited; thus, novel strategies that can be used to attenuate ARI are urgently required.

MicroRNA (miRNA/miR)-106b-5p, a member of the miR-106b~25 cluster, has been reported to exhibit an upregulated expression in hepatocellular carcinoma cells, renal cell carcinoma cells and pancreatic islet cells (9-11), and a high expression of miR-106b-5p has been shown to promote the radioresistance of colorectal cancer cells and post-injury β-cell proliferation (11). In addition, the silencing of miR-106b-5p has been shown to suppress the proliferation of glioma tumor cells and to promote cell apoptosis by targeting its downstream molecules, retinoblastoma-like (RBL)1, RBL2 and caspase-8 (12). miR-106b-5p can also activate fibroblast autophagy by inhibiting the expression of extracellular signal-regulated kinase 1/2 (ERK1/2) (13). Of note, researchers have found that miR-106b-5p from the plasma of patients undergoing coronary angiography is closely related to the estimated glomerular filtration rate (eGFR), which indicates that this miRNA is associated with kidney function in patients undergoing coronary angiography (14). However, whether miR-106b-5p regulates the proliferation, apoptosis and autophagy of renal proximal tubular epithelial cells in ARI remains unclear.

Transcription factor 4 (TCF4) is a basic helix-loop-helix transcription factor that plays a critical role in a variety of cancers, Pitt-Hopkins syndrome and eye formation (15-17). TCF4 has been shown to be closely related to renal injury in I/R-induced ARI via regulating the β-catenin protective pathway (18,19). The downregulation of TCF4 expression can increase the severity of renal injury and can contribute to the apoptosis of NRK-52E renal proximal tubular epithelial cells (20). According to a previous study, the β-catenin/TCF4 pathway can play a positive role in the regulation of renal cell carcinoma cell proliferation (21). The inhibition of the β-catenin/TCF4 pathway can increase the autophagic flux in glioblastoma (22), and this pathway is also involved in the modulation of 1,25-dihydroxyvitamin-D3-induced autophagy in diabetic cardiomyopathy in type 1 diabetic rats (23). However, the role of TCF4 in the regulation of renal proximal tubular epithelial cell proliferation and autophagy in ARI has not yet been elucidated.

In the present study, the binding sites between miR-106b-5p and TCF4 were predicted using bioinformatics software. The present study also aimed to investigate whether miR-106b-5p plays a role in regulating the proliferation, apoptosis and autophagy of renal proximal tubular epithelial cells in I/R-induced ARI through TCF4.

Materials and methods

Establishment of rat model of ARI

Sprague-Dawley rats (male, 8 weeks old, weighing 200-220 g) were obtained from the Laboratory Animal Center of Southern Medical University. All rats were acclimated for 7 days prior to surgery and were housed in a standard animal room (humidity, 50-70%; temperature, 21-25°C) with a 12-h light/dark cycle with no limitations to food and water. A total of 24 rats were randomly divided into the sham-operated (sham), I/R, I/R + scrambled version of the antagonist (NC) and I/R + antagonist groups, with 6 rats in each group. Firstly, the rats were intraperitoneally injected with 30 mg/kg sodium pentobarbital for anesthesia. A surgical incision (3-4 cm) was made along the midline of the abdomen. For the rats in the I/R group, a microvascular clamp was used to occlude the bilateral renal pedicles (30 min) for renal ischemia (24). An additional 10 mg/kg sodium pentobarbital was used to maintain anesthesia. The clamps were then removed for reperfusion. For the rats in the sham group, the microvascular clamp was not used to occlude the bilateral renal pedicles, while the other procedures were the same as those in the I/R group. For the rats in I/R + antagonist group, they were pre-treated with an intravenous injection of miR-106b-5p antagonist (2 mg/kg body weight; Shanghai GenePharma Co., Ltd.) via the tail vein. According to preliminary experiments, various doses of miR-106b-5p antagonist (0.08, 0.4, 2 and 10 mg/kg) were used to treat rats, and the results revealed that 2 mg/kg miR-106b-5p antagonist achieved the optimal alleviating effect (data not shown). Briefly, miR-106b-5p antagonist in 0.1 ml of saline was injected into the tail vein of the rats 48 h prior to ischemic surgery at 20-40 µl/sec. During anesthesia and recovery, the body temperature of rats was maintained at 36-37°C in a temperature-controlled apparatus. After 24 h, all rats were euthanized by an overdose of 100 mg/kg sodium pentobarbital via intraperitoneal injection. Blood and kidney tissues were collected for use in the following experiments. The animal experiment was approved by the Ethics Committee of The Southern Hospital of Southern Medical University (SYXK2020-0056).

Hematoxylin and eosin (H&E) staining assay

Following dissection, the whole kidney was washed with iso-osmotic saline and pressed between two filter papers. The dried kidney was fixed using 10% formaldehyde for 24 h, dehydrated with a gradient concentration of ethanol, embedded in paraffin and cut into 4-µm-thick sections. The renal tissue sections were then deparaffinized by xylene, hydrated by gradient ethanol, and stained with H&E (Beyotime Institute of Biotechnology; hematoxylin for 10 min and eosin for 30 sec at room temperature). The sections were observed under an optical microscope (Olympus Corporation) at a magnification of ×400. The scores of the sections were evaluated by two experienced pathologists in a blinded manner. The score of each section was quantified using the following criteria by the degree of tubular injury (0 to 4): 0, no damage; 1, <25%; 2, 25-50%; 3, 50-75%; 4, >75%, which was evaluated by the loss of brush border, tubular dilation, tubular necrosis, intertubular hemorrhaging, vacuolar degeneration and cast formation.

TUNEL assay

Cell apoptosis in renal tissue was detected using a TUNEL staining kit (Roche Diagnostics). Whole renal tissue was immersed in 30% sucrose solution for 12 h and embedded in optimal cutting temperature (OCT) compound (Tissue-Tek; Sakura Finetek USA, Inc.). Subsequently, the frozen renal tissue sections (7-µm-thick) were thawed, dehydrated with a gradient concentration of ethanol, washed with phosphate-buffered saline (PBS), and treated with 15 µg/ml proteinase K (Beyotime Institute of Biotechnology) without DNase. The renal tissue sections were then stained with 500 µl TUNEL reaction mixture from the TUNEL staining kit (Roche Diagnostics) for 60 min at 37°C in the dark, and TUNEL-positive cells were observed under an optical microscope (Olympus Corporation). The rate of apoptosis was calculated as follows: (number of TUNEL-positive cells/total number of cells) ×100%.

Detection of serum creatinine (Scr) and blood urea nitrogen (BUN) levels

The blood obtained from the rats was allowed to stand for 30 min, and serum was then separated by centrifugation at 3,000 x g for 10 min at 4°C. Scr and BUN levels in the blood samples were measured using a Creatinine Assay kit (cat. no. ab65340, Abcam) and a Urea Nitrogen (BUN) Colorimetric Detection kit (cat. no. EIABUN, Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions.

Cells, cell culture and transfection

NRK-52E rat renal proximal tubular epithelial cell line was obtained from The Cell Bank of Type Culture Collection of the Chinese Academy of Sciences. NRK-52E cells were cultured in Dulbecco's modified Eagle's medium (DMEM; Invitrogen; Thermo Fisher Scientific, Inc.) supplemented with 4 mM L-glutamine (Gibco; Thermo Fisher Scientific, Inc.), 1.5 g/l sodium bicarbonate (Sigma-Aldrich; Merck KGaA), 4.5 g/l glucose (Sigma-Aldrich; Merck KGaA) and 5% bovine calf serum (Gibco; Thermo Fisher Scientific, Inc.) in a 5% CO2 incubator at 37°C.

The NRK-52E cells (3×105 cells/well) were transfected with 100 nM miR-106b-5p antagonizing sequence (antagonist) and 100 nM of a scrambled version of the antagonist (NC) (synthesized by Shanghai GenePharma Co., Ltd.); 1 µg TCF4 overexpression vector (pcDNA-TCF4; synthesized by Shanghai GenePharma Co., Ltd.) and 1 µg its negative control (pcDNA-NC; synthesized by Shanghai GenePharma Co., Ltd.), 1 µg small interfering RNA (siRNA) targeting TCF4 (si-TCF4; synthesized by Shanghai GenePharma Co., Ltd.; sequence, 5′-CCG GAA CAG ACA GTA TAA TG-3′) and 1 µg siRNA targeting negative control (si-NC) (synthesized by Shanghai GenePharma Co., Ltd.; sequence, 5′-CAC TAC CGT TGT TAT AGG TG-3′), 1 µg miR-106b-5p mimic (synthesized by Shanghai GenePharma Co., Ltd.) and 1 µg its negative control (miR-NC; synthesized by Shanghai GenePharma Co., Ltd.) using Lipofectamine 2000® reagent (Invitrogen; Thermo Fisher Scientific, Inc.) for 48 h at 37°C. After 48 h, cells were collected and used for in vitro experiments.

Hypoxia-reoxygenation (H/R) cell model

NRK-52E cells were cultured in serum-free medium and exposed to 6 h of hypoxia (5% CO2, 1% O2, 94% N2) followed by 12 h of reoxygenation (5% CO2, 21% O2, and 74% N2), as previously described (25). Control cells were cultured in normal DMEM and exposed to a mixture of 5% CO2 and 95% air for the same time.

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

Total RNA was isolated from the renal tissues or NRK-52E cells using TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.). The renal tissues were ground into a suspension using a homogenizer TRIzol® reagent was added to the suspension, and the supernatant was obtained by centrifugation at 3,000 x g for 15 min at 4°C. The NRK-52E cells were digested with TRIzol® reagent for 5 min at 25°C, and the supernatant was obtained by centrifugation at 3,000 x g for 15 min at 4°C. Isopropyl alcohol (50%) was added to the supernatant, mixed and centrifuged at 3,000 x g for 10 min at 4°C. The supernatant was discarded, and the precipitate was mixed with diethyl pyrocarbonate water to measure the RNA concentration. RNA was reverse transcribed into cDNA using the RNeasy Mini kit (cat. no. 74004; Qiagen, Inc.). qPCR was conducted using a QuantStudio 1 Real-Time PCR System (cat. no. A40426; Applied Biosystems; Thermo Fisher Scientific, Inc.). The PCR cycling conditions were as follows: Initial denaturation at 95°C for 30 sec; followed by 40 cycles of 95°C for 5 sec, 60°C for 35 sec and 72°C for 30 sec. Primers used for RT-qPCR were synthesized by Invitrogen; Thermo Fisher Scientific, Inc. The gene expression levels of miR-106b-5p and TCF4 were calculated using the 2−ΔΔCq method (26). The expression levels of miR-106b-5p and TCF4 were normalized to the internal controls, U6 or GAPDH. The primers used were the following: MiR-106b-5p forward, 5′-CTG CTG GGA CTA AAG TGC TGA C-3′ and reverse, 5′-GCA GCA AGT ACC CAC AGT GC-3′; TCF4 forward, 5′-GCA AGT TGG ACG CCC GCA AGA TC-3′ and reverse, 5′-TAG TCA GCC ATG GGG CGG AGA-3′; U6 forward, 5′-GCT TCG GCA GCA CAT ATA CTA AAA T-3′ and reverse, 5′-CGC TTC AGA ATT TGC GTG TCA T-3′; GAPDH forward, 5′-TGC ACC ACC AAC TGC TTA GC-3′ and reverse, 5′-GGC ATG GAC TGT GGT CAT GAG-3′.

EdU staining assay

EdU staining assay was conducted to detect the proliferation of the NRK-52E cells in the different groups according to a previous report (27). NRK-52E cells (5×105 cells/well) were seeded into 96-well plates, and cell transfection was conducted 48 h later. After 48 h, the NRK-52E cells were incubated with 10 µM EdU (Beyotime Institute of Biotechnology) for 2 h at 37°C. The NRK-52E cells were then fixed with 4% paraformaldehyde for 15 min at room temperature. The NRK-52E cells were then counterstained with 4′,6-diamidino-2-phenylindole (DAPI; 1 µg/ml; Beyotime Institute of Biotechnology) for 10 min at room temperature. Images were acquired using a fluorescence microscope (Nikon Corporation) at a magnification of ×200.

Flow cytometry

NRK-52E cells from the different groups were harvested and washed with PBS three times. The NRK-52E cells (5×104 cells) were re-suspended in 195 µl Annexin V-FITC binding buffer (Beyotime Institute of Biotechnology). Annexin V-FITC reagent (5 µl; Beyotime Institute of Biotechnology) was added into the cell suspension, and the NRK-52E cells were incubated in the dark for 10 min at room temperature. Subsequently, 10 µl propidium iodide (PI; Beyotime Institute of Biotechnology) were added to the NRK-52E cells and apoptotic cells were distinguished by a fluorescence-activated cell sorting analyzer (FACS; BD Biosciences).

Western blot analysis

Proteins were extracted from renal tissues or NRK-52E cells using RIPA lysis buffer (Beyotime Institute of Biotechnology). The protein concentration was measured using the Pierce BCA Protein Assay kit (Thermo Fisher Scientific, Inc.). Protein (50 µg) was separated on 10% SDS-polyacrylamide gel electrophoresis (SDS-PAGE). Proteins were then transferred to polyvinylidene difluoride (PVDF) membrane and blocked with 5% skim milk for 1 h at room temperature. Primary antibodies against Bax (1:2,000, cat. no. ab182733; Abcam), Bcl-2 (1:2,000, cat. no. ab194583; Abcam), cleaved caspase-3 (1:1,000, cat. no. 9661; Cell Signaling Technology, Inc.), p62 (1:10,000, cat. no. ab109012; Abcam), LC3 (1:500, cat. no. ab63817; Abcam), TCF4 (1:25,000, cat. no. ab185736; Abcam) and GAPDH (1:2,000, cat. no. ab8245; Abcam) were incubated with the membranes at 4°C overnight. Peroxidase-conjugated secondary antibodies (1:3,000, cat. no. ab205718; Abcam) were then added to incubate with the membranes at room temperature for 1 h. The BeyoECL Plus kit (Beyotime Institute of Biotechnology) was used to develop the blots and for visualization. Finally, densitometric analysis was conducted using ImageLab software version 3.0 (Bio-Rad Laboratories, Inc.). GAPDH was used as the internal reference.

Luciferase reporter gene assay

The binding sites between TCF4 and miR-106b-5p were predicted using bioinformatics software (TargetScan; http://www.targetscan.org/vert_72/). A species was first selected, followed by the entry of a gene symbol, and then clicking on the 'submit' button. For validating the binding site between TCF4 and miR-106b-5p, the 3′ untranslated region (UTR) of TCF4 including wild-type (WT) or mutant-type (MUT) of the binding sites were cloned downstream of the Firefly luciferase gene in psiCHECK-2 reporter vector (Promega Corporation). NRK-52E cells were co-transfected with 100 nM miR-NC, 100 nM miR-106b-5p, 200 ng TCF4 3′ UTR-WT reporter construct, and 200 ng TCF4 3′UTR-MUT reporter construct (psiCHECK-2 vector; Promega Corporation). After 48 h, the NRK-52E cells were harvested and washed with PBS. The Dual Luciferase Reporter Assay System (Promega Corporation) was used to detect the luciferase activity, and the relative luciferase activity was normalized to Renilla luciferase activity.

Statistical analysis

All experiments were performed in triplicate. All data were normally distributed and presented as the mean ± standard deviation (SD). The Student's t-test was used to analyze statistical significance between 2 groups, and one-way analysis of variance (ANOVA) with the Bonferroni post hoc test was used to analyze statistical significance among ≥3 groups. A P-value <0.05 was considered to indicate a statistically significant difference.

Results

miR-106b-5p has an upregulated expression in renal tissue from the rat model of ARI

To investigate the role of miR-106b-5p in the progression of ARI, a rat model of ARI induced by I/R was established. The severity of renal injury was firstly evaluated in rats from the sham and I/R group by H&E staining, which revealed that inflammatory cell infiltration and injury were more severe in the I/R group than the sham group (Fig. 1A). Moreover, the number of TUNEL-positive cells in the renal tissues was significantly increased in the I/R group compared with the sham group, indicating that cell apoptosis was increased in the I/R group (Fig. 1B and C). In addition, the Scr and BUN levels were significantly elevated in the blood samples from the I/R group compared with the sham group (Fig. 1D and E). These findings thus indicated that the rat model of ARI was successfully established. The present study then detected miR-106b-5p expression in renal tissues, and it was found that miR-106b-5p expression was significantly upregulated in the I/R group compared with Sham group (Fig. 1F). In the NRK-52E cells subjected to H/R, miR-106b-5p expression was also found to be significantly upregulated compared with the control group (Fig. 1G).

miR-106b-5p antagonist promotes the proliferation, and inhibits the apoptosis and autophagy of NRK-52E cells subjected to H/R

To further investigate the role of miR-106b-5p in the proliferation, apoptosis and autophagy of NRK-52E cells subjected to H/R, miR-106b-5p antagonist was used to treat the NRK-52E cells subjected to H/R. No significant differences were found in proliferation and apoptosis between the normal NRK-52E cells and the miR-106b-5p antagonist-treated NRK-52E cells (Fig. S1). However, miR-106b-5p mimic significantly promoted NRK-52E cell apoptosis in the H/R + miR-106b-5p mimic group compared with the H/R + miR-NC group (Fig. S2). Furthermore, the apoptosis of NRK-52E cells subjected to H/R was significantly inhibited in the miR-106b-5p antagonist group compared with the DMSO group (Fig. S3). In addition, miR-106b-5p expression was significantly increased in the NRK-52E cells transfected with miR-106b-5p mimic compared with the NRK-52E cells transfected with miR-NC (Fig. S4A). As shown in Fig. 2A, H/R induction significantly upregulated miR-106b-5p expression, and miR-106b-5p antagonist reversed the promoting effect on miR-106b-5p expression. The number of EdU-positive cells was significantly reduced in the H/R group compared with the control group, indicating that H/R induction inhibited the proliferation of NRK-52E cells (Fig. 2B). However, miR-106b-5p antagonist reversed the inhibitory effect on NRK-52E cell proliferation (Fig. 2B). Moreover, H/R induction significantly promoted the apoptosis of NRK-52E cells, and miR-106b-5p antagonist reversed the promoting effect on NRK-52E cell apoptosis (Fig. 2C). In addition, the levels of the apoptosis-related proteins, Bax and cleaved caspase-3, and those of the anti-apoptotic protein, Bcl-2, were measured by western blot analysis. The results revealed that H/R induction significantly upregulated the Bax, cleaved caspase-3 protein levels and the ratio of cleaved caspase-3/caspase-3, and downregulated the Bcl-2 protein level; miR-106b-5p antagonist reversed the changes in the levels of apoptosis-related proteins (Fig. 2D). Moreover, H/R induction significantly downregulated the level of the autophagy-related protein, p62, and promoted the conversion of LC3I to LC3II. miR-106b-5p antagonist reversed these changes in the levels of autophagy-related protein (Fig. 2E).

Regulation of TCF4 by miR-106b-5p in ARI

According to the bioinformatics software. TargetScan, there were potential binding sites between miR-106b-5p and TCF4 (Fig. 3A). Luciferase reporter gene assay revealed that miR-106b-5p significantly reduced the luciferase activity of TCF4 3′UTR-WT, whereas miR-106b-5p did not affect the luciferase activity of TCF4 3′UTR-MUT (Fig. 3B). The protein level of TCF4 was also detected in renal tissues from the rats with ARI induced by I/R. The results revealed that the TCF4 protein level was downregulated in the I/R group compared with the sham group (Fig. 3C). In addition, the TCF4 protein level was downregulated in the NRK-52E cells subjected to H/R compared with the control cells (Fig. 3D). These findings indicate that miR-106b-5p may negatively regulate TCF4 expression in NRK-52E cells and renal tissue from rats with ARI.

Overexpression of TCF4 promotes the proliferation, and inhibits the apoptosis and autophagy of NRK-52E cells subjected to H/R

To explore whether TCF4 is involved in the proliferation, apoptosis and autophagy of NRK-52E cells injured by H/R, a TCF4 overexpression vector was transfected into the NRK-52E cells. Firstly, TCF4 mRNA/protein expression was found to be significantly increased in the NRK-52E cells transfected with pcDNA-TCF4 compared with the NRK-52E cells transfected with pcDNA-NC (Fig. 4B and C). As shown in Fig. 4A and B, H/R induction significantly downregulated TCF4 mRNA and protein expression, and pcDNA-TCF4 significantly upregulated TCF4 mRNA and protein expression under the H/R condition. H/R induction significantly decreased the percentage of EdU-positive cells, and pcDNA-TCF4 significantly increased the percentage of EdU-positive cells (Fig. 4C). In addition, H/R induction significantly promoted the apoptosis of NRK-52E cells, and pcDNA-TCF4 significantly inhibited the apoptosis of NRK-52E cells under the H/R condition (Fig. 4D). The changes in the levels of the apoptotic-related proteins, Bax, Bcl-2 and cleaved caspase-3, and the ratio of cleaved caspase-3/caspase-3 also indicated that pcDNA-TCF4 inhibited the apoptosis of NRK-52E cells under the H/R condition (Fig. 4E). In addition, H/R induction significantly downregulated the p62 protein level and promoted the conversion of LC3I to LC3II, whereas pcDNA-TCF4 significantly upregulated the p62 protein level and inhibited the conversion of LC3I to LC3II (Fig. 4F).

miR-106b-5p antagonist attenuates ARI by preventing the inhibition of TCF4

To further explore whether miR-106b-5p antagonist plays a positive role in attenuating ARI by preventing the inhibition of TCF4, the NRK-52E cells were transfected with si-TCF4. Firstly, TCF4 mRNA/protein expression was significantly decreased in the NRK-52E cells transfected with si-TCF4 compared with the NRK-52E cells transfected with si-NC (Fig. S4D and E). It was found that miR-106b-5p antagonist significantly upregulated TCF4 mRNA and protein expression in the NRK-52E cells subjected to H/R, whereas si-TCF4 reversed this promoting effect on TCF4 expression (Fig. 5A and B). Furthermore, miR-106b-5p antagonist significantly increased the percentage of EdU-positive cells, whereas si-TCF4 reversed the promoting effect on the proliferation of NRK-52E cells subjected to H/R (Fig. 5C). miR-106b-5p antagonist also significantly inhibited the apoptosis of NRK-52E cells subjected to H/R, whereas si-TCF4 reversed the inhibitory effect on cell apoptosis (Fig. 5D). In addition, the changes in the levels of the apoptosis-related proteins, Bax, Bcl-2 and cleaved caspase-3, and the ratio of cleaved caspase-3/caspase-3 also indicated that si-TCF4 promoted the apoptosis of NRK-52E cells subjected to H/R (Fig. 5E). miR-106b-5p antagonist also significantly upregulated the p62 protein level and inhibited the conversion of LC3I to LC3II, whereas si-TCF4 reversed the changes in the levels of autophagy-related proteins (Fig. 5F).

miR-106b-5p antagonist attenuates ARI in vivo

As shown in Fig. 6A, treatment of the rats with ARI with miR-106b-5p antagonist reduced the severity of renal injury in the rats with ARI induced by I/R. Moreover, the number of TUNEL-positive cells in the renal tissues was significantly reduced in the rats with ARI injected with miR-106b-5p antagonist (Fig. 6B and C). The Scr and BUN levels were significantly decreased in the blood samples of ARI rats injected with miR-106b-5p antagonist (Fig. 6D and E). These findings indicated that miR-106b-5p antagonist attenuated the progression of ARI. Moreover, miR-106b-5p expression was significantly downregulated in the renal tissues from rats with ARI injected with miR-106b-5p antagonist (Fig. 6F). The TCF4 protein level was significantly upregulated in the renal tissues from rats with ARI injected with miR-106b-5p antagonist (Fig. 6G).

Discussion

The present study used miR-106b-5p antagonist to treat rats and cells in order to explore the effects of miR-106b-5p inhibition on the regulation of I/R-induced ARI. The main findings were the following: i) In the rats with I/R-induced ARI and RK-52E cells subjected to H/R, miR-106b-5p expression was increased in the renal tissue from rats with ARI and in NRK-52E cells; ii) miR-106b-5p expression targeted the 3′UTR of TCF4 mRNA; iii) miR-106b-5p antagonist or TCF4 overexpression promoted cell proliferation, and inhibited cell apoptosis and autophagy; iv) miR-106b-5p antagonist attenuated ARI by preventing the inhibition of TCF4 in vivo. These findings identified a novel role of miR-106b-5p antagonist acting as an essential modulator for the treatment in ARI.

miRNAs have been identified to be crucial modulators in regulating the severity of renal injury in I/R-induced ARI (28,29). Aberrant miRNA expression is closely related to the severity of renal injury and cell apoptosis in kidneys in the progression of ARI (30,31). For example, the upregulation of miR-146a expression in I/R-injured C57BL/6 mouse kidneys has been shown to be significantly associated with more severe I/R-induced renal injury (30). miR-17-5p expression has been shown to be upregulated in the renal tissues of mice with I/R-induced ARI and in tubular cells subjected to H/R, and the upregulation of miR-17-5p significantly increases the apoptosis of tubular cells (31). miR-489 expression has also been shown to be elevated in kidneys from mice with I/R-induced ARI and in renal tubular cells subjected to H/R, and the inhibition of miR-489 promotes the apoptosis of renal tubular cells, indicating that miR-489 can protect renal tubular cells from I/R injury (32). Hence, focusing on miRNAs may provide new insight into understanding I/R-induced ARI and may aid in the development of novel treatment strategies for patients with ARI. In the present study, more severe renal injury was found in the rats with I/R-induced ARI, with increased apoptosis in renal tissues and elevated Scr and BUN levels in the blood samples, indicating the loss of renal function in rats with ARI. The results also revealed that miR-106b-5p expression was upregulated in renal tissues from rats with I/R-induced ARI and in NRK-52E cells subjected to H/R. These results suggest that aberrant miR-106-5p expression may be related to the severity of renal injury in I/R-induced ARI.

Increasing evidence has indicated that miRNAs perform important functions in a variety of cellular processes, such as cell proliferation, apoptosis and autophagy (33,34). For example, miR-381 expression has been shown to be decreased in rats with I/R-induced ARI, and miR-381 overexpression promotes the proliferation and inhibits the apoptosis of renal tubular epithelial cells (35). It has also been shown that H/R-induction promotes miR-424 expression in proximal kidney tubular cells, and the suppression of miR-424 promotes apoptosis and activates caspases (36). I/R induction has been shown to increase miR-34a expression in mice with ARI, and the silencing of miR-34a inhibits autophagy in tubular epithelial cells (37). As previously demonstrated, I/R induction increases miR-214 expression in renal tissues of mice with ARI and rat kidney proximal tubular cells, and miR-214 inhibition relieves mitochondrial fragmentation and suppresses apoptosis (38). miRNA-188 has been shown to be markedly upregulated in rats with ARI and HK-2 cells, and miRNA-188 overexpression facilitates cell apoptosis (39). In the present study, it was found that H/R induction upregulated miR-106b-5p expression, inhibited cell proliferation, and promoted the apoptosis and autophagy of NRK-52E cells. Further treatment with miR-106b-5p antagonist promoted cell proliferation, and inhibited the apoptosis and autophagy of NRK-52E cells subjected to H/R, indicating that miR-106b-5p antagonist played an effective role in protecting NRK-52E cells from H/R injury.

It has been identified that miRNAs play important roles in I/R-induced ARI by suppressing downstream targeted mRNAs (40). For example, miR-191 has been shown to negatively regulate downstream targeted mRNA cystathionine-β-synthase to modulate the progression of I/R-induced ARI (41). As previously demonstrated, miR-204 inhibits epithelial-mesenchymal transition through targeting SP1 to alleviate chronic fibrotic changes in I/R-induced ARI (42). miR-16 has also been shown to reduce renal function via inhibiting downstream targeted Bcl-2 to aggravate the progression of I/R-induced ARI (43). According to the prediction of the bioinformatics software, TargetScan, the present study identified binding sites between TCF4 and miR-106b-5p. Therefore, a dual luciferase reporter gene assay was conducted to confirm the regulatory effects of miR-106b-5p on TCF4 expression. The results revealed that miR-106b-5p significantly reduced the luciferase activity of TCF4 3′UTR-WT, indicating TCF4 was a target of miR-106b-5p. It was further identified that TCF4 expression was negatively modulated by miR-106b-5p in the renal tissues of rats with I/R-induced ARI and in NRK-52E cells subjected to H/R. Therefore, it was identified that miR-106b-5p targeted TCF4 in I/R-induced ARI.

TCF4, a E-box transcription factor, has been identified to play a protective role in I/R-induced ARI, and a decreased TCF4 expression has been shown to increase the severity of renal injury in vivo and promote the apoptosis of NRK-52E cells in vitro (19,20). According to previous reports, TCF4 can be directly targeted by several miRNAs, including miR-130a-3p, miR-129-5p and miR-155 in chondrocytes, osteoblasts and NRK-52E cells subjected to H/R (24,44,45). Since it was identified that TCF4 was a target of miR-106b-5p in I/R-induced ARI, the biological function of TCF4 was further investigated in NRK-52E cells subjected to H/R. In the present study, it was found that H/R induction inhibited NRK-52E cell proliferation, and promoted NRK-52E cell apoptosis and autophagy, whereas TCF4 overexpression reversed the effects of H/R induction on NRK-52E cells. This indicated that TCF4 played a protective role in NRK-52E cells subjected to H/R by promoting cell proliferation, and inhibiting cell apoptosis and autophagy.

Studies have demonstrated that miRNAs can negatively regulate gene expression via the translational suppression of target mRNAs, playing critical roles in regulating cell proliferation, apoptosis and autophagy (46,47). Various disease-specific miRNAs, such as miR-155, miR-194 and miR-182 have been identified to exert their biological functions in renal tubular epithelial cells subjected to H/R via suppressing their downstream molecules (20,24,48). The present study investigated whether miR-106b-5p regulates the proliferation, apoptosis and autophagy of NRK-52E cells by suppressing TCF4 under the H/R condition. miR-106b-5p antagonist upregulated the mRNA and protein levels of TCF4 in NRK-52E cells subjected to H/R. Further treatment with si-TCF4 abolished the promoting effect of miR-106b-5p antagonist on the proliferation of NRK-52E cells subjected to H/R. It also reversed the inhibitory effect of miR-106b-5p antagonist on the apoptosis and autophagy of NRK-52E cells subjected to H/R. These findings indicated that miR-106b-5p antagonist played an effective role in protecting the NRK-52E cells from H/R injury via negatively regulating TCF4. To the best of our knowledge, no other studies to date have identified the role of miR-106b-5p/TCF4 in modulating NRK-52E cell proliferation, apoptosis and autophagy in ARI, which may provide basic strategies for ARI treatment.

Studies have provided evidence that miRNA antagonist can effectively inhibit the progression of ARI in vivo (49,50). However, the underlying mechanisms of miR-106b-5p antagonist in ameliorating I/R-induced ARI in rats was not illustrated. In the present in vivo experiment, it was found that miR-106b-5p antagonist reduced the severity of renal injury in rats with I/R-induced ARI, decreased the number of TUNEL-positive cells in renal tissues from rats with I/R-induced ARI, and decreased the Scr and BUN levels in the blood samples from rats with I/R-induced ARI, which indicated that miR-106b-5p antagonist attenuated the progression of ARI. Moreover, miR-106b-5p antagonist downregulated miR-106b-5p expression and upregulated TCF4 protein level in renal tissues from rats with ARI, indicating that miR-106b-5p and TCF4 were dysregulated in the rat model of ARI.

In conclusion, the present study demonstrated that miR-106b-5p antagonist promoted the proliferation, and inhibited the apoptosis and autophagy of renal proximal tubular epithelial cells in ARI by upregulating TCF4 expression in vitro. Thus, miR-106b-5p antagonist attenuated ARI in rats and H/R injury in cells.

Supplementary Data

Availability of data and materials

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

Authors' contributions

JMH and LL designed the study. JMH, YPY and LJH performed the experiments. JMH wrote the manuscript. PBW and YY analyzed the data. JMH and LL confirm the authenticity of all the raw data. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

The animal experiment was approved by the Ethics Committee of The Southern Hospital of Southern Medical University (SYXK2020-0056).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

Not applicable.

References

1 

Fortrie G, de Geus HR and Betjes MG: The aftermath of acute kidney injury: A narrative review of long-term mortality and renal function. Crit Care. 23:242019. View Article : Google Scholar : PubMed/NCBI

2 

Kölling M, Genschel C, Kaucsar T, Hübner A, Rong S, Schmitt R, Sörensen-Zender I, Haddad G, Kistler A, Seeger H, et al: Hypoxia-induced long non-coding RNA Malat1 is dispensable for renal ischemia/reperfusion-injury. Sci Rep. 8:34382018. View Article : Google Scholar : PubMed/NCBI

3 

Diao C, Wang L, Liu H, Du Y and Liu X: Aged kidneys are refractory to autophagy activation in a rat model of renal ischemia-reperfusion injury. Clin Interv Aging. 14:525–534. 2019. View Article : Google Scholar : PubMed/NCBI

4 

Wang M, Deng J, Lai H, Lai Y, Meng G, Wang Z, Zhou Z, Chen H, Yu Z, Li S and Jiang H: Vagus nerve stimulation ameliorates renal ischemia-reperfusion injury through inhibiting NF-κB activation and iNOS protein expression. Oxid Med Cell Longev. 2020:71065252020.

5 

Liu H, Wang L, Weng X, Chen H, Du Y, Diao C, Chen Z and Liu X: Inhibition of Brd4 alleviates renal ischemia/reperfusion injury-induced apoptosis and endoplasmic reticulum stress by blocking FoxO4-mediated oxidative stress. Redox Biol. 24:1011952019. View Article : Google Scholar : PubMed/NCBI

6 

Xi X, Zou C, Ye Z, Huang Y, Chen T and Hu H: Pioglitazone protects tubular cells against hypoxia/reoxygenation injury through enhancing autophagy via AMPK-mTOR signaling pathway. Eur J Pharmacol. 863:1726952019. View Article : Google Scholar : PubMed/NCBI

7 

Prieto-Moure B, Lloris-Carsí JM, Belda-Antolí M, Toledo-Pereyra LH and Cejalvo-Lapeña D: Allopurinol protective effect of renal ischemia by downregulating TNF-α, IL-1β, and IL-6 response. J Invest Surg. 30:143–151. 2017. View Article : Google Scholar

8 

Yingjie K, Haihong Y, Lingwei C, Sen Z, Yuanting D, Shasha C, Liutong P, Ying W and Min Z: Apoptosis repressor with caspase recruitment domain deficiency accelerates ischemia/reperfusion (I/R)-induced acute kidney injury by suppressing inflammation and apoptosis: The role of AKT/mTOR signaling. Biomed Pharmacother. 112:1086812019. View Article : Google Scholar : PubMed/NCBI

9 

Gu H, Gu S, Zhang X, Zhang S, Zhang D, Lin J, Hasengbayi S and Han W: MiR-106b-5p promotes aggressive progression of hepatocellular carcinoma via targeting RUNX3. Cancer Med. 8:6756–6767. 2019. View Article : Google Scholar : PubMed/NCBI

10 

Sun K, Jia Z, Duan R, Yan Z, Jin Z, Yan L, Li Q and Yang J: Long non-coding RNA XIST regulates miR-106b-5p/P21 axis to suppress tumor progression in renal cell carcinoma. Biochem Biophys Res Commun. 510:416–420. 2019. View Article : Google Scholar : PubMed/NCBI

11 

Tsukita S, Yamada T, Takahashi K, Munakata Y, Hosaka S, Takahashi H, Gao J, Shirai Y, Kodama S, Asai Y, et al: MicroRNAs 106b and 222 improve hyperglycemia in a mouse model of insulin-deficient diabetes via pancreatic β-Cell proliferation. EBioMedicine. 15:163–172. 2017. View Article : Google Scholar

12 

Liu F, Gong J, Huang W, Wang Z, Wang M, Yang J, Wu C, Wu Z and Han B: MicroRNA-106b-5p boosts glioma tumorigensis by targeting multiple tumor suppressor genes. Oncogene. 33:4813–4822. 2014. View Article : Google Scholar

13 

Zeng T, Wang X, Wang W, Feng Q, Lao G, Liang Y, Wang C, Zhou J, Chen Y, Liu J, et al: Endothelial cell-derived small extracellular vesicles suppress cutaneous wound healing through regulating fibroblasts autophagy. Clin Sci (Lond). 133:CS201900082019. View Article : Google Scholar

14 

Muendlein A, Geiger K, Leiherer A, Saely CH, Fraunberger P and Drexel H: Evaluation of the associations between circulating microRNAs and kidney function in coronary angiography patients. Am J Physiol Renal Physiol. 318:F315–F321. 2020. View Article : Google Scholar

15 

In't Hout FEM, Gerritsen M, Bullinger L, Van der Reijden BA, Huls G, Vellenga E and Jansen JH: Transcription factor 4 (TCF4) expression predicts clinical outcome in RUNX1 mutated and translocated acute myeloid leukemia. Haematologica. 105:e454–e457. 2020. View Article : Google Scholar : PubMed/NCBI

16 

Wang Y, Lu Z, Zhang Y, Cai Y, Yun D, Tang T, Cai Z, Wang C, Zhang Y, Fang F, et al: Transcription factor 4 safeguards hippocampal dentate gyrus development by regulating neural progenitor migration. Cereb Cortex. 30:3102–3115. 2020. View Article : Google Scholar

17 

Young RM, Ewan KB, Ferrer VP, Allende ML, Godovac-Zimmermann J, Dale TC and Wilson S: Developmentally regulated Tcf7l2 splice variants mediate transcriptional repressor functions during eye formation. Elife. 8:e514472019. View Article : Google Scholar :

18 

Menon MC, Chuang PY, Li Z, Wei C, Zhang W, Luan Y, Yi Z, Xiong H, Woytovich C, Greene I, et al: Intronic locus determines SHROOM3 expression and potentiates renal allograft fibrosis. J Clin Invest. 125:208–221. 2015. View Article : Google Scholar :

19 

Al-bataineh MM, Kinlough CL, Poland PA, Pastor-Soler NM, Sutton TA, Mang HE, Bastacky SI, Gendler SJ, Madsen CS, Singh S, et al: Muc1 enhances the β-catenin protective pathway during ischemia-reperfusion injury. Am J Physiol Renal Physiol. 310:F569–F579. 2016. View Article : Google Scholar : PubMed/NCBI

20 

Li H, Ma Y, Chen B and Shi J: MiR-182 enhances acute kidney injury by promoting apoptosis involving the targeting and regulation of TCF7L2/Wnt/β-catenins pathway. Eur J Pharmacol. 831:20–27. 2018. View Article : Google Scholar : PubMed/NCBI

21 

Xu Z, Hong Z, Ma M, Liu X, Chen L, Zheng C, Xi X and Shao J: Rock2 promotes RCC proliferation by decreasing SCARA5 expression through β-catenin/TCF4 signaling. Biochem Biophys Res Commun. 480:586–593. 2016. View Article : Google Scholar : PubMed/NCBI

22 

Nàger M, Sallán MC, Visa A, Pushparaj C, Santacana M, Macià A, Yeramian A, Cantí C and Herreros J: Inhibition of WNT-CTNNB1 signaling upregulates SQSTM1 and sensitizes glioblastoma cells to autophagy blockers. Autophagy. 14:619–636. 2018. View Article : Google Scholar : PubMed/NCBI

23 

Wei H, Qu H, Wang H, Ji B, Ding Y, Liu D, Duan Y, Liang H, Peng C, Xiao X and Deng H: 1,25-Dihydroxyvitamin-D3 prevents the development of diabetic cardiomyopathy in type 1 diabetic rats by enhancing autophagy via inhibiting the beta-catenin/TCF4/GSK-3beta/mTOR pathway. J Steroid Biochem Mol Biol. 168:71–90. 2017. View Article : Google Scholar : PubMed/NCBI

24 

Zhang XB, Chen X, Li DJ, Qi GN, Dai YQ, Gu J, Chen MQ, Hu S, Liu ZY and Yang ZM: Inhibition of miR-155 ameliorates acute kidney injury by apoptosis involving the regulation on TCF4/Wnt/β-Catenin pathway. Nephron. 143:135–147. 2019. View Article : Google Scholar

25 

Shen B, Mei M, Pu Y, Zhang H, Liu H, Tang M, Pan Q, He Y, Wu X and Zhao H: Necrostatin-1 attenuates renal ischemia and reperfusion injury via meditation of HIF-1α/mir-26a/TRPC6/PARP1 Signaling. Mol Ther Nucleic Acids. 17:701–713. 2019. View Article : Google Scholar : PubMed/NCBI

26 

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. View Article : Google Scholar

27 

Hu Y, Yang C, Yang S, Cheng F, Rao J and Wang X: MiR-665 promotes hepatocellular carcinoma cell migration, invasion, and proliferation by decreasing Hippo signaling through targeting PTPRB. Cell Death Dis. 9:9542018. View Article : Google Scholar : PubMed/NCBI

28 

Wang X, Liu J, Yin W, Abdi F, Pang PD, Fucci QA, Abbott M, Chang SL, Steele G, Patel A, et al: MiR-218 expressed in endothelial progenitor cells contributes to the development and repair of the kidney microvasculature. Am J Pathol. 190:642–659. 2020. View Article : Google Scholar : PubMed/NCBI

29 

Jia P, Wu X, Dai Y, Teng J, Fang Y, Hu J, Zou J, Liang M and Ding X: MicroRNA-21 is required for local and remote ischemic preconditioning in multiple organ protection against sepsis. Crit Care Med. 45:e703–e710. 2017. View Article : Google Scholar : PubMed/NCBI

30 

Amrouche L, Desbuissons G, Rabant M, Sauvaget V, Nguyen C, Benon A, Barre P, Rabaté C, Lebreton X, Gallazzini M, et al: MicroRNA-146a in human and experimental ischemic AKI: CXCL8-dependent mechanism of action. J Am Soc Nephrol. 28:479–493. 2017. View Article : Google Scholar :

31 

Hao J, Wei Q, Mei S, Li L, Su Y, Mei C and Dong Z: Induction of microRNA-175p by p53 protects against renal ischemia-reperfusion injury by targeting death receptor 6. Kidney Int. 91:106–118. 2017. View Article : Google Scholar

32 

Wei Q, Liu Y, Liu P, Hao J, Liang M, Mi QS, Chen JK and Dong Z: MicroRNA-489 induction by hypoxia-inducible factor-1 protects against ischemic kidney injury. J Am Soc Nephrol. 27:2784–2796. 2016. View Article : Google Scholar : PubMed/NCBI

33 

Xu Z, Li Z, Wang W, Xia Y, He Z, Li B, Wang S, Huang X, Sun G, Xu J, et al: MIR-1265 regulates cellular proliferation and apoptosis by targeting calcium binding protein 39 in gastric cancer and, thereby, impairing oncogenic autophagy. Cancer Lett. 449:226–236. 2019. View Article : Google Scholar : PubMed/NCBI

34 

Ma L, Li Z, Li W, Ai J and Chen X: MicroRNA-142-3p suppresses endometriosis by regulating KLF9-mediated autophagy in vitro and in vivo. RNA Biol. 16:1733–1748. 2019. View Article : Google Scholar : PubMed/NCBI

35 

Zheng GH, Wen X, Wang YJ, Han XR, Shan Q, Li W, Zhao T, Wu DM, Lu J and Zheng YL: MicroRNA-381-induced down-regulation of CXCR4 promotes the proliferation of renal tubular epithelial cells in rat models of renal ischemia reperfusion injury. J Cell Biochem. 119:3149–3161. 2018. View Article : Google Scholar

36 

Chen S, Yao Y, Lin F, Bian F, Zhu C and Jiang G: MiR-424 is over-expressed and attenuates ischemia-reperfusion kidney injury via p53 and death receptor 6 pathway. Am J Transl Res. 11:1965–1979. 2019.PubMed/NCBI

37 

Liu XJ, Hong Q, Wang Z, Yu YY, Zou X and Xu LH: MicroRNA-34a suppresses autophagy in tubular epithelial cells in acute kidney injury. Am J Nephrol. 42:168–175. 2015. View Article : Google Scholar : PubMed/NCBI

38 

Yan Y, Ma Z, Zhu J, Zeng M, Liu H and Dong Z: MiR-214 represses mitofusin-2 to promote renal tubular apoptosis in ischemic acute kidney injury. Am J Physiol Renal Physiol. 318:F878–887. 2020. View Article : Google Scholar : PubMed/NCBI

39 

Liu B, Chai Y, Guo W, Lin K, Chen S, Liu J, Sun G, Chen G, Song F, He Y, et al: MicroRNA-188 aggravates contrast-induced apoptosis by targeting SRSF7 in novel isotonic contrast-induced acute kidney injury rat models and renal tubular epithelial cells. Ann Transl Med. 7:3782019. View Article : Google Scholar : PubMed/NCBI

40 

Xu X, Song N, Zhang X, Jiao X, Hu J, Liang M, Teng J and Ding X: Renal protection mediated by hypoxia inducible factor-1α depends on proangiogenesis function of miR-21 by targeting thrombospondin 1. Transplantation. 101:1811–1819. 2017. View Article : Google Scholar : PubMed/NCBI

41 

Wu XQ, Tian XY, Wang ZW, Wu X, Wang JP and Yan TZ: MiR-191 secreted by platelet-derived microvesicles induced apoptosis of renal tubular epithelial cells and participated in renal ischemia-reperfusion injury via inhibiting CBS. Cell Cycle. 18:119–129. 2019. View Article : Google Scholar :

42 

Chen SJ, Wu P, Sun LJ, Zhou B, Niu W, Liu S, Lin FJ and Jiang GR: MiR-204 regulates epithelial-mesenchymal transition by targeting SP1 in the tubular epithelial cells after acute kidney injury induced by ischemia-reperfusion. Oncol Rep. 37:1148–1158. 2017. View Article : Google Scholar

43 

Chen HH, Lan YF, Li HF, Cheng CF, Lai PF, Li WH and Lin H: Urinary miR-16 transactivated by C/EBPβ reduces kidney function after ischemia/reperfusion-induced injury. Sci Rep. 6:279452016. View Article : Google Scholar

44 

Luo X, Wang J, Wei X, Wang S and Wang A: Knockdown of lncRNA MFI2-AS1 inhibits lipopolysaccharide-induced osteoarthritis progression by miR-130a-3p/TCF4. Life Sci. 240:1170192020. View Article : Google Scholar

45 

Yin C, Tian Y, Yu Y, Yang C, Su P, Zhao Y, Wang X, Zhang K, Pei J, Li D, et al: MiR-129-5p inhibits bone formation through TCF4. Front Cell Dev Biol. 8:6006412020. View Article : Google Scholar : PubMed/NCBI

46 

Xu J, Cao D, Zhang D, Zhang Y and Yue Y: MicroRNA-1 facilitates hypoxia-induced injury by targeting NOTCH3. J Cell Biochem. 121:4458–4469. 2020. View Article : Google Scholar : PubMed/NCBI

47 

Li N, Guo X, Liu L, Wang L and Cheng R: Molecular mechanism of miR-204 regulates proliferation, apoptosis and autophagy of cervical cancer cells by targeting ATF2. Artif Cells Nanomed Biotechnol. 47:2529–2535. 2019. View Article : Google Scholar : PubMed/NCBI

48 

Shen Y, Zhao Y, Wang L, Zhang W, Liu C and Yin A: MicroRNA-194 overexpression protects against hypoxia/reperfusion-induced HK-2 cell injury through direct targeting Rheb. J Cell Biochem. Nov 28–2018.Epub ahead of print.

49 

Guo Y, Ni J, Chen S, Bai M, Lin J, Ding G, Zhang Y, Sun P, Jia Z, Huang S, et al: MicroRNA-709 mediates acute tubular injury through effects on mitochondrial function. J Am Soc Nephrol. 29:449–461. 2018. View Article : Google Scholar :

50 

Lorenzen JM, Kaucsar T, Schauerte C, Schmitt R, Rong S, Hübner A, Scherf K, Fiedler J, Martino F, Kumarswamy R, et al: MicroRNA-24 antagonism prevents renal ischemia reperfusion injury. J Am Soc Nephrol. 25:2717–2729. 2014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2021
Volume 48 Issue 3

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Hu J, He L, Wang P, Yu Y, Ye Y and Liang L: Antagonist targeting miR‑106b‑5p attenuates acute renal injury by regulating renal function, apoptosis and autophagy via the upregulation of TCF4 Retraction in /10.3892/ijmm.2024.5353. Int J Mol Med 48: 169, 2021
APA
Hu, J., He, L., Wang, P., Yu, Y., Ye, Y., & Liang, L. (2021). Antagonist targeting miR‑106b‑5p attenuates acute renal injury by regulating renal function, apoptosis and autophagy via the upregulation of TCF4 Retraction in /10.3892/ijmm.2024.5353. International Journal of Molecular Medicine, 48, 169. https://doi.org/10.3892/ijmm.2021.5002
MLA
Hu, J., He, L., Wang, P., Yu, Y., Ye, Y., Liang, L."Antagonist targeting miR‑106b‑5p attenuates acute renal injury by regulating renal function, apoptosis and autophagy via the upregulation of TCF4 Retraction in /10.3892/ijmm.2024.5353". International Journal of Molecular Medicine 48.3 (2021): 169.
Chicago
Hu, J., He, L., Wang, P., Yu, Y., Ye, Y., Liang, L."Antagonist targeting miR‑106b‑5p attenuates acute renal injury by regulating renal function, apoptosis and autophagy via the upregulation of TCF4 Retraction in /10.3892/ijmm.2024.5353". International Journal of Molecular Medicine 48, no. 3 (2021): 169. https://doi.org/10.3892/ijmm.2021.5002