Open Access

MicroRNA‑379‑5p suppresses renal fibrosis by regulating the LIN28/let‑7 axis in diabetic nephropathy

  • Authors:
    • Nan Li
    • Li‑Juan Wang
    • Wei‑Long Xu
    • Su Liu
    • Jiang‑Yi Yu
  • View Affiliations

  • Published online on: August 30, 2019     https://doi.org/10.3892/ijmm.2019.4325
  • Pages: 1619-1628
  • Copyright: © Li 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

MicroRNAs (miRNAs or miRs) play an important role in pathological processes in diabetic nephropathy (DN). This study aimed to explore whether miR‑379‑5p is associated with renal fibrosis in DN and to elucidate the underlying mechanisms. In vitro experiments indicated that miR‑379‑5p expression was downregulated by high glucose (HG) treatment in mouse mesangial cells (MMCs). Transfection with miR‑379‑5p mimics suppressed the proliferation and the accumulation of extracellular matrix (ECM) components, which were promoted by HG treatment. LIN28B was proven to be a direct target of miR‑379‑5p by luciferase report assay. In addition, the loss of expression of LIN28B, as well as the decrease in cell proliferation and in the accumulation of ECM components, which were induced by the knockdown of LIN28B, were attenuated in the MMCs following transfection with miR‑379‑5p inhibitors. Furthermore, type 2 diabetic db/db mice were used to examine the efficiency of miR‑379‑5p agomir in the alleviation of renal fibrosis. Consistent with the results of the in vitro experiments, miR‑379‑5p agomir suppressed mesangial cell proliferation and the accumulation of ECM components by regulating the LIN28B/let‑7 pathway. Taken together, the findings of this study suggest that miR‑379‑5p is highly involved in renal fibrosis in DN, and that it may be a potential effective therapeutic target for DN.

Introduction

As a main complication of diabetes mellitus, diabetic nephropathy (DN) is a leading cause of end-stage renal disease, accounting for >35% of all new cases requiring dialysis therapy globally (1). Clinically, DN is characterized by the development of proteinuria followed by decreased glomerular filtration (2). Pathologically, DN is characterized by the thickening of the basement membrane, renal tubal epithelial-mesenchymal transition (EMT), the accumulation of extracellular matrix (ECM) components, glomerular sclerosis and tubulointerstitial fibrosis (3). However, there is limited acknowledge available on the pathological mechanisms involved in DN, and thus there is also a lack of effective treatments for DN. Therefore, it is of utmost importance to study its precise molecular mechanisms and to develop novel effective therapeutic strategies for DN.

MicroRNAs (miRNAs or miRs) are small non-coding single-stranded RNAs which have been confirmed to play critical roles in cell proliferation, apoptosis and differentiation, contributing to the pathogenesis of a number of human deceases, such as cancer and diabetes (4,5). Researches have indicated that miRNAs are highly involved in DN, which suggests the great potential of miRNAs in DN detection, as well as therapy (6,7). It has been found that urinary miRNAs related to histopathological lesions and kidney damage can be applied to the sensitive, specific and non-invasive detection of DN (8). Furthermore, miRNAs have been successfully applied to DN therapy. The addition of the agomir of the downregulated miRNAs (i.e., miR-455-3p) or antagomir of the commonly upregulated miRNAs (i.e., miR-192 and miR-377) in DN has been shown to induce a decrease in albuminuria and mesangial matrix accumulation in animal models (9-11).

miR-379-5p has been reported to be downregulated in spontaneous type 2 diabetic KKay mice (12). In this study, we investigated the role of miR-379-5p in DN both in vitro and in vivo. Glomerular hypertrophy, mainly caused by glomerular mesangial cell (MC) proliferation and the accumulation of ECM components, is considered to be one of the earliest pathological characteristics of DN. Of the three types of constitutive cells in the glomerulus, MCs are the main source of ECM proteins. Therefore, we isolated and cultured mouse glomerular MCs (MMCs) for experiments in vitro. LIN28B, the direct target of miR-379-5p, was identified by the database, TargetScan. LIN28B has been shown to be closely related to various types of cancer (13). It has been reported that in MMCs treated with transforming growth factor (TGF)-β, the LIN28/let-7 pathway can affect the accumulation of ECM components by targeting collagen type 1-α2 (Col1a2) and collagen type 4-α1 (Col4a1) (14). In this study, we attempted to investigate whether miR-379-5p regulates the LIN28/let-7 axis in DN, in an aim to provide a promising approach for the clinical treatment of DN.

Materials and methods

Cell culture and transfection

MMCs were isolated from 8-week-old male C57BL/6 mice (n=6) as previously described (15). The mice were euthanized by an intraperitoneal injection of sodium pentobarbital (100 mg/kg). All experiments were approved by the Animal Care and Use Committee of the Nanjing University of Chinese Medicine and complied with the Declaration of the National Institutes of Health Guide for the Care and Use of Laboratory Animals. MMCs were obtained from the renal cortex of the 8-week-old male mice using sterile techniques. After washing with RPMI-1640 medium (Beyotime Biotechnology), the cortices were separated from the medullas, pooled, minced and the glomeruli were isolated. The glomeruli were then pre-treated with collagenase for 15 min and plated in RPMI-1640 medium supplemented with 10% fetal bovine serum (FBS) and 20 µg/ml insulin at 37°C with 5% CO2. Cells at passages between 3 and 7 were used in all the experiments.

The MMCs were treated with normal glucose (5 mmol/l) plus mannitol (25 mmol/l) for the control group and high glucose (30 mmol/l) for the high glucose (HG) group. miR-379-5p mimics, inhibitors, agomir and siRNA against LIN28B and the negative controls were purchased from RiboBio (Guangzhou). All of the transient transfections were conducted using Lipofectamine 2000 reagent (Invitrogen; Thermo Fisher Scientific). 293T cells were acquired from the Chinese Academy of Sciences Cell Bank and were cultured in Dulbecco's modified Eagle's medium (DMEM) containing 10% FBS.

Luciferase reporter assay

The LIN28B transcript sequences were acquired from NCBI resources (gene ID, 380669). The 3′ UTR of LIN28B with predicted target sites for miR-379-5p seed sequence was amplified and cloned into the PGL-3 vector (Promega) using KpnI/HindIII sites. The primers used were as follows: Sense, 5′-GGTACCGCCTTTGATTCAGAAACGG-3′ and antisense, 5′-AAGCTTCTATAAAACATGACACCCGC-3′. The mutated 3′ UTR of LIN28B was generated using the QuikChange II Site-Directed Mutagenesis kit (Stratagene). At 48 h following the co-transfection of miR-379-5p mimics (100 nM) and the wild-type or mutated 3′ UTR of LIN28B, the luciferase activity of the 293T cells was measured using the dual-luciferase assay kit (Promega) with Renilla luciferase as the internal control.

Reverse transcription-quantitative PCR (RT-qPCR)

Total RNA was extracted from the MMCs and renal tissues (see below) using TRIzol reagent (Invitrogen; Thermo Fisher Scientific). cDNA was synthesized using the PrimeScript® RT reagent kit (Takara). qPCR was performed using SYBR Premix ExTaq™ (Takara) on the platform of Applied Biosystems 7500. U6 was used as an endogenous normalization control. The total reaction system of 20 μl volume was as follows: 1 μl cDNA, 10 μl SYBR Premix EX Taq, 1 μl each of the primers (10 μM) and 7 μl ddH2O. The PCR cycling conditions were as follows: 95°C for 5 min, followed by 40 cycles of 95°C for 15 sec and 60°C for 30 sec. The primers used were as follows: miR-379-5p forward, 5′-GCGCTGGTAGACTATGGAA-3′ and reverse, 5′-GTG CAG GGT CCG AGG T-3′; U6 forward, 5′-CTC GCT TCG GCA GCA CAT ATA CT-3′ and reverse, 5′-ACG CTT CAC GAA TTT GCG TGT C-3′; Lin28B forward, 5′-GTC AAT ACG GGT AAC AGG AC-3′ and reverse, 5′-TTC TTT GGC TGA GGA GGT AG-3′; Let-7b forward, 5′-TGA GGT AGT AGG TTG TGT-3′ and reverse, 5′-GTC GTA TCC AGT GCA GGG TCC GAG GT-3′; Col-1 forward, 5′-CCA CCC CAG CCG CAA AGA GTC-3′ and reverse, 5′-GTC ATC GCA CAC AGC CGT GC-3′; Col-4 forward, 5′-ATC TCT GCC AGG ACC AAG TG-3′ and reverse, 5′-CGG GCT GAC ATT CCA CAA T-3′; GAPDH forward, 5′-GAA ATC CCA TCA CCA TCT TCC AGG-3′ and reverse, 5′-GAG CCC CAG CCT TCT CCA TG-3′. Data analyses were performed using the comparative Cq (ΔΔCq) method for calculating relative gene expression (16).

Western blot analysis

Renal tissue (see below) and MMCs were lysed in radioimmunoprecipitation buffer. Approximately 80 μg of total proteins were separated by 12% or 10% SDS-PAGE gel and then transferred onto PVDF membranes (Millipore Corp.) by electroblotting. After being blocked for 60 min with 5% non-fat milk, the membranes were incubated at 4°C overnight with primary antibodies against Col4 (ab6586; 1:1,000), fibronectin (FN; ab2413; 1:1,000), p21 (ab109199; 1:1,000), cyclin D1 (ab134175; 1:1,000) (all from Abcam) and LIN28B (#5422; 1:1,000; Cell Signaling Technology). GAPDH (ab9485; 1:2,000; Abcam) was served as the loading control. The membranes were then incubated with the secondary antibodies anti-rabbit IgG, HRP-linked antibody (1:1,000, #7074, Cell Signaling Technology) for 1 h at room temperature. Subsequently, they were visualized with ECL detection reagent (Millipore), and the gray values of the bands were calculated automatically (ImageJ software, version 4.3).

Cell proliferation

Cell viability was assayed using the Cell Counting kit-8 (Dojindo) following the manufacturer's instructions. The cells were seeded in 96-well plates at a density of 2×103 cells per well. CCK-8 solution (10 μl) was added to each well for 1 h following 48 h of incubation, and OD450 was determined using a microplate reader (ELx×808, BioTek). Moreover, 5-ethynyl-2′-deoxyuridine (EdU) assay (Beyotime Biotechnology) was performed to detect the proliferation of the MMCs. Following incubation with 10 μM EdU for 2 h at 37°C, the cells were fixed in 4% paraformaldehyde. Subsequently, Hoechst 33342 (Beyotime Biotechnology) was used to stain the nuclei at room temperature for 30 min. Finally, the cells were visualized under a fluorescence microscopy (Leica).

Cell cycle analysis

For cell cycle analysis, the MMCs were harvested, suspended in 500 μl of phosphate-buffered saline (PBS), fixed with 70% ethanol and subjected to PI/RNase (Beyotime Biotechnology) staining for 30 min at 4°C. The MMCs were then analyzed by flow cytometry (FACSCanto II, BD Biosciences).

Animal experiments

A total of 18 C57BL/KsJ type 2 diabetic db/db male mice (aged 4 weeks, weighing 18-22 g) and a total of 6 heterozygote db/m male mice (aged 4 weeks, weighing 18-22 g) used in this study were purchased from the Model Animal Research Center of Nanjing University. The mice were housed in a clean environment with 50-60% humidity, 20-22°C and a 12-h dark/light cycle, and fed a standard diet during a 1-week adaptation period. When the mice were 8 weeks old, they were randomly divided into the control group (untreated db/m mice, n=6); the DN group (untreated db/db mice, n=6); an agomir NC (agomir NC-treated db/db, n=6) group and an agomir-379-5p (agomir-379-5p-treated db/db, n=6) group. Every 2 days, the db/db mice were injected with miR-379-5p agomir or agomir NC (10 mg/kg) (GenePharma) via the tail vein. Four weeks later, the mice were sacrificed by an intraperitoneal injection of sodium pentobarbital (100 mg/kg) and the kidneys were harvested for analysis. During the experiments, animal health and behavior were monitored every 2 days. All experiments were approved by the Animal Care and Use Committee of the Nanjing University of Chinese Medicine and complied with the Declaration of the National Institutes of Health Guide for the Care and Use of Laboratory Animals.

Measurement of urine protein levels

Metabolic cages were used to collect urine samples in a 24-h period, and urinary protein levels were evaluated using the mouse albumin ELISA quantitation set (Bethyl Laboratories).

Histology and immunostaining assays

Fresh renal tissues were fixed with 4% paraformaldehyde and embedded in paraffin. Paraffin-embedded specimens were sectioned (4 μm thicknesses) and stained with PAS, Masson's trichrome staining (Beyotime Biotechnology) at room temperature for 15 min and immunohistochemistry. The antibodies included LIN28B (ab71415; 1:50), Ki-67 (ab15580; 1:500) and normal rabbit IgG (ab6728, 1:1,000) (all from Abcam) were incubated for 2 h at 37°C.

In situ hybridization (ISH)

To detect the presence of miR-379-5p in the renal tissues, in situ hybridization was performed. The frozen renal tissues sections of mice from control group (untreated db/m mice), DN group (untreated db/db mice) and agomir-379-5p (agomir-379-5p-treated db/db mice) group were fixed in paraformaldehyde followed by rinsing in PBS. A digoxin-labeled oligonucleotide probe (5′-TGG TAG ACT ATG GAA CGT AGG-3′) (0.5 μg/ml) was synthesized by Dingguo BioTechnology Co. Ltd. In situ hybridization was performed with the ISH kit according to the protocol (Dingguo BioTech). The slides were treated with 3,3′-diaminobenzidine (DAB) for 5 min, and then counterstained with hematoxylin for 1-2 min at room temperature. All images were acquired by using a light microscope (Olympus) at the magnification of ×400.

Bioinformatics analysis

TargetScan (http://www.targetscan.org/) was used to predict the putative target genes for miR-379-5p.

Statistical analysis

GraphPad Prism (version 5.0; GraphPad Software, Inc.) was used to conduct all statistical analyses. Measurement data are presented as the means ± standard deviation. One-way ANOVA analysis followed by a Tukey's post hoc test were used to compare the difference between multiple groups. Values of P<0.05 and P<0.01 were considered to indicate statistically significant and highly statistically significant differences, respectively.

Results

miR-379-5p is downregulated by high glucose treatment in MMCs

To explore the potential role of miR-379-5p in DN, we analyzed the expression of miR-379-5p in MMCs stimulated with HG. As shown in Fig. 1A and B, miR-379-5p expression was significantly decreased in the cells treated with high glucose in a dose- and time-dependent manner, and its expression was approximately one third of that of the control following stimulation of the cells with 30 mM glucose for 48 h.

miR-379-5p suppresses the proliferation and accumulation of ECM components in MMCs

Mesangial cell proliferation and the accumulation of ECM components are important pathological features of DN (17). In this study, to explore the effects of miR-379-5p on cell proliferation and the accumulation of ECM components, the MMCs were transfected with miR-379-5p mimics. The results revealed that miR-379-5p expression was markedly increased in the MMCs transfected with miR-379-5p mimics (Fig. 1C). The results of CCK-8 assay also revealed that HG stimulation markedly promoted the viability of the MMCs. However, the promoting effects of HG stimulation on cell viability were reversed following transfection of the MMCs with miR-379-5p mimics (Fig. 2A). The results of EdU assays yielded similar conclusions, in that transfection with miR-379-5p mimics attenuated the effects of HG stimulation on cell proliferation (Fig. 2B). Furthermore, cell cycle analysis indicated that there was a decrease in the percentage of cells in the G0/G1 phase, as well as an increase in the percentage of cells in the S phase in the HG group compared with the control group. MMCs transfected with miR-379-5p mimics exhibited an increase in the percentage of cells in the G0/G1 phase and a decrease in the percentage of cells in the S phase compared with the HG group (Fig. 2C). This suggested that the overexpression of miR-379-5p prevented MMCs cell cycle progression.

p21, a cyclin-dependent kinase inhibitor, almost inhibits all cyclin/CDK complexes. The expression level of p21 and cyclin D1 can reflect the proliferation of MMCs (18). In this study, the results of western blot analysis revealed that transfection with miR-379-5p mimics attenuated the decrease and increase in the expression level of p21 and cyclin D1, respectively induced by HG stimulation (Fig. 2D). The cell cycle analysis and the related protein experimental data suggested that the overexpression of miR-379-5p suppressed the proliferation of MMCs following HG stimulation. Tubulointerstitial fibrosis is always involved with the increased deposition of ECM proteins, such as collagens and FN (19,20). In this study, we detected Col4 and FN expression by western blot analysis. As shown in Fig. 2E, Col4 and FN expression levels were markedly promoted in the HG group and the mimics-NC group compared to the control group, and decreased in the mimics group compared to the mimics-NC group. Taken together, these results indicated that miR-379-5p suppressed the proliferation and accumulation of ECM components in MMCs.

LIN28B is a target of miR-379-5p

The TargetScan database was used to identify the targets of miR-379-5p. It was suggested that miR-379-5p had a conserved binding site in the 3′ UTR of LIN28B (Fig. 3A). To verify the association between miR-379-5p and LIN28B, luciferase reporter assays were performed. The results revealed that the luciferase activity was significantly decreased in the MMCs co-transfected with miR-379-5p mimics and the wild-type 3′ UTR reporter gene other than the mutated 3′ UTR reporter gene, indicating that miR-379-5p directly binds to the 3′ UTR of LIN28B (Fig. 3B).

LIN28B, a RNA-binding protein belongs to LIN28 family, mediates diverse biological functions (21). In this study, to examine the regulatory association between miR-379-5p and LIN28B, the expression levels of LIN28B and let-7b were detected in MMCs transfected with miR-379-5p mimics. As shown in Fig. 3C, the overexpression of miR-379-5p markedly suppressed the LIN28B expression level, which was induced by HG. The results of western blot analysis also revealed that miR-379-5p inactivated LIN28B expression in a HG-independent manner (Fig. 3D).

miR-379-5p suppresses the proliferation and accumulation of ECM components by regulating LIN28B expression

Previously, Kim et al demonstrated that LIN28B increased the expression of ECM proteins, such as Col1a2 and Col4a1, by decreasing let-7 levels (22). In this study, firstly, the transfection efficiency of miR-379-5p inhibitor was evaluated by RT-qPCR. The results revealed that the cells transfected with miR-379-5p inhibitor exhibited lower levels of miR-379-5p expression compared with the negative control group, indicating that miR-379-5p inhibitor was transfected into the cells successfully (Fig. 4A). To explore whether miR-379-5p suppresses the proliferation and accumulation of ECM components by regulating the LIN28/let-7 axis, a si-LIN28B vector was constructed and transfected into the MMCs (Fig. 4B and C). Transfection with si-LIN28B decreased LIN28B expression; however, miR-379-5p inhibitor partly reversed the decrease in LIN28B induced by si-LIN28B in the cells treated with HG (Fig. 4B and C). CCK-8 assay was performed in the HG group, si-NC group, inhibitors-NC group, si-LIN28B group and si-LIN28B + miR-379-5p inhibitors group to examine the effects of LIN28B on MMC viability. The results revealed that the knockdown of LIN28B suppressed MMC viability (Fig. 4D). Transfection with si-LIN28B decreased the mRNA expression levels of Col1 and Col4, and increased the let-7 expression level, as shown by RT-qPCR (Fig. 4E). Western blot analysis for Col-1 and Col-4 revealed that si-LIN28B decreased the accumulation of the ECM components, Col-1 and Col-4, compared to si-NC group (Fig. 4F). However, MMCs co-transfected with miR-379-5p inhibitors and si-LIN28B exhibited a higher proliferation rate and a greater accumulation of ECM components compared to the corresponding si-LIN28B group (Fig. 4D-F). Taken together, these results suggested that miR-379-5p suppressed the proliferation and accumulation of ECM components by regulating the LIN28/let-7 axis.

miR-379-5p suppresses renal fibrosis in vivo

To examine the effects of miR-379-5p on renal fibrosis in DN, we used type 2 diabetic db/db mice for research in vivo with db/m mice as a normal control. miR-379-5p expression levels in the kidney tissues were significantly downregulated in the db/db mice, as detected by RT-qPCR and in situ hybridization assays. miR-379-5p agomir injection markedly promoted miR-379-5p expression (Fig. 5A and B). Consequently, LIN28B expression was upregulated in the kidney tissues of the db/db mice and downregulated after the injection of miR-379-5p agomir (Fig. 5C-E). Moreover, glomerular hypertrophy with an increased glomerular area was observed in the db/db mice. However, miR-379-5p agomir attenuated glomerular hypertrophy. As is well-known, the earliest pathological characteristic of DN is glomerular hypertrophy, which is mainly caused by glomerular MC proliferation and ECM accumulation. In this study, we wished to determine the role of the miR-379-5p/LIN28/let-7 axis in glomerular MCs during DN. Physiological markers, such as urine protein and blood glucose were also measured. It was found that urine protein levels were significantly increased in the db/db mice, and urine protein levels were decreased in the miR-379-5p agomir group (Fig. 5F).

In order to examine the effects of miR-379-5p on renal fibrosis in DN, some pathological markers, such as PAS and Masson's trichrome staining were used (Fig. 6A). The results indicated that miR-379-5p alleviated pathological changes, such as glomerular hypertrophy, mesangial amplification and renal fibrosis in the diabetic mice. Furthermore, mesangial cell proliferation detected by Ki-67 staining is presented in Fig. 6B, and ECM accumulation was measured by RT-qPCR and western blot analysis for let-7b, Col-4 and Col-1 expression (Fig. 6C and D). The results revealed that miR-379-5p agomir decreased mesangial cell proliferation and accumulation of ECM components that were increased in the db/db mice, which was consistent with the data obtained from the in vitro assays.

Discussion

Recently, increasing evidence has indicated that the dysregulation of miRNAs, such as miR-214, miR-15a, miR-29a and miR-34a plays an important role in the pathogenesis of DN (23-31). In this study, we found that miR-379-5p was downregulated and acted as a suppressor of renal fibrosis by regulating LIN28B in diabetic nephropathy.

miR-379 has been found to participate in a number of biological processes. It acts as a tumor suppressor and is downregulated in human cancers (32-34). In hepatocel-lular carcinoma and non-small-cell lung cancer, miR-379 is associated with chemosensitivity (35,36). Moreover, miR-379 has been shown to be downregulated in patients with athero-sclerotic coronary artery disease and acute myocardial infraction (37,38).

LIN28B is an oncogene that has been reported to be overexpressed in numerous malignant tumors (39-42). LIN28A AND LIN28B selectively block the expression of let-7 miRNAs and promote tumorigenesis (43). On the other hand, the LIN28B/let-7 axis has been reported to be involved in controlling TGF-β-induced collagen accumulation in DN (14). In addition, LIN28B has been reported to be associated with fibrosis lesions. For instance, LIN28B can participate in the progression of organ damage and fibrosis in metabolic diseases, such as diabetes (14). LIN28B can ameliorate liver fibrosis by inhibiting mesenchymal signaling pathways (44). The upregulation of LIN28B has been shown to contribute to idiopathic pulmonary fibrosis (45). It is well known that transforming growth factor-β1 (TGF-β1) expression is increased in glomerular MCs that lead to an increased accumulation of ECM components and hypertrophy during the progression of DN (46). TGF-β1 can induce LIN28B expression which results in the suppression of let-7 and the upregulation of collagen expression in glomerular mesangial cells under diabetic conditions (14). Moreover, the LIN28/let-7 axis plays an important role in regulating glucose metabolism (47). Let-7a regulates glucose metabolism and insulin synthesis/secretion in type 2 diabetes mellitus by targeting the LIN28 pathway (48).

However, there are some limitations to this study. Mesangial cells have been used to study glomerular fibrosis in a number of studies (49,50). The LIN28B/let-7 signaling pathway has been reported to be involved in cell energy metabolism in some cells. Further, we aim to examine the effects of miR-379-5p on podocytes in metabolism, not through fibrosis, particularly the influence of miR-379-5p in the process of podocyte diminution and disappearance in nephropathy in future studies.

In this study, another miRNA, miR-379-5p, was found to suppress renal fibrosis by regulating the LIN28B/let-7 axis in DN. miR-379-5p expression was downregulated and that of LIN28B was upregulated both in MMCs treated with HG and in glomeruli of db/db mice. However, miR-379-5p mimics and agomir suppressed the expression of LIN28B in vitro and in vivo, respectively. The TargetScan database and luciferase reporter assay indicated that miR-379-5p directly bound to the 3′ UTR of LIN28B and suppressed the expression of LIN28B. We verified that miR-379-5p regulated LIN28B in a HG-independent manner. In addition, let-7b, a target of LIN28B, was upregulated when the expression of miR-379-5p was increased. As the results of the overexpression of miR-379-5p, MMC proliferation and collagen protein accumulation were alleviated. It is thus suggested that miR-379-5p suppressed renal fibrosis by the regulating LIN28/let-7 axis in DN, which may lay the foundation for clinical treatment in the future.

In conclusion, the findings of this study suggested a coherent mechanism for the role of miR-379-5p in renal fibrosis during DN, and provide new insight into the mechanisms of renal protection associated with miR-379-5p and LIN28/let-7, which may lead to a novel therapeutic strategy for DN.

Acknowledgements

Not applicable.

Funding

This study was supported by grants from the General Program of National Natural Science Foundation of China (grant no. 81774117); Postgraduate Research and Practice Innovation Program of Jiangsu Province (Grant no. KYCX18_1574) and National Youth Foundation of China (grant no. 81804027).

Availability of data and materials

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

Authors' contributions

JYY designed research and revised the manuscript; NL and LJW performed the research analysis and wrote the manuscript; NL, LJW, WLX and SL analyzed the data. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

All experiments were approved by the Animal Care and Use Committee of the Nanjing University of Chinese Medicine and complied with the Declaration of the National Institutes of Health Guide for the Care and Use of Laboratory Animals.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Incidence and prevalence of ESRD: United States Renal Data System. Am J Kidney Dis. 32(2 Suppl 1): pp. S38–S49. 1998, View Article : Google Scholar : PubMed/NCBI

2 

Sun YM, Su Y, Li J and Wang LF: Recent advances in understanding the biochemical and molecular mechanism of diabetic nephropathy. Biochem Biophys Res Commun. 433:359–361. 2013. View Article : Google Scholar : PubMed/NCBI

3 

Herbach N: Pathogenesis of diabetes mellitus and diabetic complications. Studies on diabetic mouse models. Pathologe. 33(Suppl 2): pp. S318–S324. 2012, In German. View Article : Google Scholar

4 

Lagos-Quintana M, Rauhut R, Lendeckel W and Tuschl T: Identification of novel genes coding for small expressed RNAs. Science. 294:853–858. 2001. View Article : Google Scholar : PubMed/NCBI

5 

Gholaminejad A, Abdul Tehrani H and Gholami Fesharaki M: Identification of candidate microRNA biomarkers in diabetic nephropathy: A meta-analysis of profiling studies. J Nephrol. 31:813–831. 2018. View Article : Google Scholar : PubMed/NCBI

6 

Allison SJ: Diabetic nephropathy: A lncRNA and miRNA mega-cluster in diabetic nephropathy. Nat Rev Nephrol. 12:7132016. View Article : Google Scholar

7 

Alvarez ML and DiStefano JK: Towards microRNA-based therapeutics for diabetic nephropathy. Diabetologia. 56:444–456. 2013. View Article : Google Scholar

8 

Cardenas-Gonzalez M, Srivastava A, Pavkovic M, Bijol V, Rennke HG, Stillman IE, Zhang X, Parikh S, Rovin BH, Afkarian M, et al: Identification, confirmation, and replication of novel urinary MicroRNA biomarkers in lupus nephritis and diabetic nephropathy. Clin Chem. 63:1515–1526. 2017. View Article : Google Scholar : PubMed/NCBI

9 

Liu F, Zhang ZP, Xin GD, Guo LH, Jiang Q and Wang ZX: miR-192 prevents renal tubulointerstitial fibrosis in diabetic nephropathy by targeting Egr1. Eur Rev Med Pharmacol Sci. 22:4252–4260. 2018.PubMed/NCBI

10 

Peng J, Wu Y, Deng Z, Zhou Y, Song T, Yang Y, Zhang X, Xu T, Xia M, Cai A, et al: miR-377 promotes white adipose tissue inflammation and decreases insulin sensitivity in obesity via suppression of sirtuin-1 (SIRT1). Oncotarget. 8:70550–70563. 2017. View Article : Google Scholar : PubMed/NCBI

11 

Liu Wu J, Ding J, Zhu Y, Lu M, Zhou K, Xie J, Xu X, Shen Y, Chen XY, et al: miR-455-3p suppresses renal fibrosis through repression of ROCK2 expression in diabetic nephropathy. Biochem Biophys Res Commun. 503:977–983. 2018. View Article : Google Scholar : PubMed/NCBI

12 

Zhu X, Zhang C, Fan Q, Liu X, Yang G, Jiang Y and Wang L: Inhibiting microRNA-503 and microRNA-181d with losartan ameliorates diabetic nephropathy in KKAy mice. Med Sci Monit. 22:3902–3909. 2016. View Article : Google Scholar : PubMed/NCBI

13 

Balzeau J, Menezes MR, Cao S and Hagan JP: The LIN28/let-7 pathway in cancer. Front Genet. 8:312017. View Article : Google Scholar : PubMed/NCBI

14 

Park JT, Kato M, Lanting L, Castro N, Nam BY, Wang M, Kang SW and Natarajan R: Repression of let-7 by transforming growth factor-β1-induced Lin28 up-regulates collagen expression in glomerular mesangial cells under diabetic conditions. Am J Physiol Renal Physiol. 307:F1390–F1403. 2014. View Article : Google Scholar : PubMed/NCBI

15 

Kim YS, Reddy MA, Lanting L, Adler SG and Natarajan R: Differential behavior of mesangial cells derived from 12/15-lipoxygenase knockout mice relative to control mice. Kidney Int. 64:1702–1714. 2003. View Article : Google Scholar : PubMed/NCBI

16 

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

17 

Shao Y, Lv C, Wu C, Zhou Y and Wang Q: Mir-217 promotes inflammation and fibrosis in high glucose cultured rat glomerular mesangial cells via Sirt1/HIF-1α signaling pathway. Diabetes Metab Res Rev. 32:534–543. 2016. View Article : Google Scholar : PubMed/NCBI

18 

Zhang Y, Wang S, Qian W, Ji D, Wang Q, Zhang Z, Wang S, Ji B, Fu Z and Sun Y: uc.338 targets p21 and cyclin D1 via PI3K/AKT pathway activation to promote cell proliferation in colorectal cancer. Oncol Rep. 40:1119–1128. 2018.PubMed/NCBI

19 

Kolset SO, Reinholt FP and Jenssen T: Diabetic nephropathy and extracellular matrix. J Histochem Cytochem. 60:976–986. 2012. View Article : Google Scholar : PubMed/NCBI

20 

Zhou SX, Huo DM, He XY, Yu P, Xiao YH, Ou CL, Jiang RM, Li D and Li H: High glucose/lysophosphatidylcholine levels stimulate extracellular matrix deposition in diabetic nephropathy via plateletactivating factor receptor. Mol Med Rep. 17:2366–2372. 2018.

21 

McWhorter ES, West RC, Russ JE, Ali A, Winger QA and Bouma GJ: LIN28B regulates androgen receptor in human trophoblast cells through Let-7c. Mol Reprod Dev. Jun 19–2019, Epub ahead of print. View Article : Google Scholar : 2019, PubMed/NCBI

22 

Kim CW, Vo MT, Kim HK, Lee HH, Yoon NA, Lee BJ, Min YJ, Joo WD, Cha HJ, Park JW and Cho WJ: Ectopic over-expression of tristetraprolin in human cancer cells promotes biogenesis of let-7 by down-regulation of Lin28. Nucleic Acids Res. 40:3856–3869. 2012. View Article : Google Scholar : PubMed/NCBI

23 

Bera A, Das F, Ghosh-Choudhury N, Mariappan MM, Kasinath BS and Ghosh Choudhury G: Reciprocal regulation of miR-214 and PTEN by high glucose regulates renal glomerular mesangial and proximal tubular epithelial cell hypertrophy and matrix expansion. Am J Physiol Cell Physiol. 313:C430–C447. 2017. View Article : Google Scholar : PubMed/NCBI

24 

Chen D, Li Y, Mei Y, Geng W, Yang J, Hong Q, Feng Z, Cai G, Zhu H, Shi S, et al: miR-34a regulates mesangial cell proliferation via the PDGFR-β/Ras-MAPK signaling pathway. Cell Mol Life Sci. 71:4027–4042. 2014. View Article : Google Scholar : PubMed/NCBI

25 

Jee YH, Wang J, Yue S, Jennings M, Clokie SJ, Nilsson O, Lui JC and Baron J: Mir-374-5p, mir-379-5p, and mir-503-5p regulate proliferation and hypertrophic differentiation of growth plate chondrocytes in male rats. Endocrinology. 159:1469–1478. 2018. View Article : Google Scholar : PubMed/NCBI

26 

Liang Y, Zhao G, Tang L, Zhang J, Li T and Liu Z: miR-1003p and miR-8773pregulate overproduction of IL-8 and IL-1β in mesangial cells activated by secretory IgA from IgA nephropathy patients. Exp Cell Res. 347:312–321. 2016. View Article : Google Scholar : PubMed/NCBI

27 

Sun T, Yang J, Dong W, Wang R, Ma P, Kang P, Zhang H, Xie C, Du J and Zhao L: Down-regulated miR-15a mediates the epithelial-mesenchymal transition in renal tubular epithelial cells promoted by high glucose. Biosci Biotechnol Biochem. 78:1363–1370. 2014. View Article : Google Scholar : PubMed/NCBI

28 

Wang B, Yao K, Wise AF, Lau R, Shen HH, Tesch GH and Ricardo SD: miR-378 reduces mesangial hypertrophy and kidney tubular fibrosis via MAPK signalling. Clin Sci (Lond). 131:411–423. 2017. View Article : Google Scholar

29 

Wang X, Shen E, Wang Y, Jiang Z, Gui D, Cheng D, Chen T and Wang N: miR-196a regulates high glucose-induced mesangial cell hypertrophy by targeting p27kip1. J Lab Autom. 20:491–499. 2015. View Article : Google Scholar : PubMed/NCBI

30 

Du B, Ma LM, Huang MB, Zhou H, Huang HL, Shao P, Chen YQ and Qu LH: High glucose down-regulates miR-29a to increase collagen IV production in HK-2 cells. FEBS Lett. 584:811–816. 2010. View Article : Google Scholar : PubMed/NCBI

31 

Liu H, Zhao J and Lv J: Inhibitory effects of miR-101 overexpression on cervical cancer SiHa cells. Eur J Gynaecol Oncol. 38:236–240. 2017.PubMed/NCBI

32 

Laddha SV, Nayak S, Paul D, Reddy R, Sharma C, Jha P, Hariharan M, Agrawal A, Chowdhury S, Sarkar C and Mukhopadhyay A: Genome-wide analysis reveals downregulation of miR-379/miR-656 cluster in human cancers. Biol Direct. 8:102013. View Article : Google Scholar : PubMed/NCBI

33 

Khan S, Brougham CL, Ryan J, Sahrudin A, O'Neill G, Wall D, Curran C, Newell J, Kerin MJ and Dwyer RM: miR-379 regulates cyclin B1 expression and is decreased in breast cancer. PLoS One. 8:pp. e687532013, View Article : Google Scholar : PubMed/NCBI

34 

Nayak S, Aich M, Kumar A, Sengupta S, Bajad P, Dhapola P, Paul D, Narta K, Purkrait S, Mehani B, et al: Novel internal regulators and candidate miRNAs within miR-379/miR-656 miRNA cluster can alter cellular phenotype of human glioblastoma. Sci Rep. 8:76732018. View Article : Google Scholar : PubMed/NCBI

35 

Huang DJ, Huang JZ, Yang J, Li YH, Luo YC, He HY and Huang HJ: Bioinformatic identification of IGF1 as a hub gene in hepatocellular carcinoma (HCC) and in-vitro analysis of the chemosensitizing effect of miR-379 via suppressing the IGF1/IGF1R signaling pathway. Eur Rev Med Pharmacol Sci. 20:5098–5106. 2016.

36 

Hao GJ, Hao HJ, Ding YH, Wen H, Li XF, Wang QR and Zhang BB: Suppression of EIF4G2 by miR-379 potentiates the cisplatin chemosensitivity in nonsmall cell lung cancer cells. FEBS Lett. 591:636–645. 2017. View Article : Google Scholar : PubMed/NCBI

37 

Yi J and An Y: Circulating miR-379 as a potential novel biomarker for diagnosis of acute myocardial infarction. Eur Rev Med Pharmacol Sci. 22:540–546. 2018.PubMed/NCBI

38 

Xu Z, Han Y, Liu J, Jiang F, Hu H, Wang Y, Liu Q, Gong Y and Li X: miR-135b-5p and miR-499a-3ppromote cell proliferation and migration in atherosclerosis by directly targeting MEF2C. Sci Rep. 5:122762015. View Article : Google Scholar

39 

Cheng SW, Tsai HW, Lin YJ, Cheng PN, Chang YC, Yen CJ, Huang HP, Chuang YP, Chang TT, Lee CT, et al: Lin28B is an oncofetal circulating cancer stem cell-like marker associated with recurrence of hepatocellular carcinoma. PLoS One. 8:pp. e800532013, View Article : Google Scholar : PubMed/NCBI

40 

King CE, Cuatrecasas M, Castells A, Sepulveda AR, Lee JS and Rustgi AK: LIN28B promotes colon cancer progression and metastasis. Cancer Res. 71:4260–4268. 2011. View Article : Google Scholar : PubMed/NCBI

41 

Alajez NM, Shi W, Wong D, Lenarduzzi M, Waldron J, Weinreb I and Liu FF: Lin28b promotes head and neck cancer progression via modulation of the insulin-like growth factor survival pathway. Oncotarget. 3:1641–1652. 2012. View Article : Google Scholar

42 

Kugel S, Sebastian C, Fitamant J, Ross KN, Saha SK, Jain E, Gladden A, Arora KS, Kato Y, Rivera MN, et al: SIRT6 suppresses pancreatic cancer through control of lin28b. Cell. 165:1401–1415. 2016. View Article : Google Scholar : PubMed/NCBI

43 

Piskounova E, Polytarchou C, Thornton JE, LaPierre RJ, Pothoulakis C, Hagan JP, Iliopoulos D and Gregory RI: Lin28A and Lin28B inhibit let-7 microRNA biogenesis by distinct mechanisms. Cell. 147:1066–1079. 2011. View Article : Google Scholar : PubMed/NCBI

44 

McDaniel K, Huang L, Sato K, Wu N, Annable T, Zhou T, Ramos-Lorenzo S, Wan Y, Huang Q, Francis H, et al: The let-7/Lin28 axis regulates activation of hepatic stellate cells in alcoholic liver injury. J Biol Chem. 292:11336–11347. 2017. View Article : Google Scholar : PubMed/NCBI

45 

Liang H, Liu S, Chen Y, Bai X, Liu L, Dong Y, Hu M, Su X, Chen Y, Huangfu L, et al: miR-26a suppresses EMT by disrupting the Lin28B/let-7d axis: Potential cross-talks among miRNAs in IPF. J Mol Med (Berl). 94:pp. 655–665. 2016, View Article : Google Scholar

46 

Hills CE and Squires PE: TGF-beta1-induced epithelial-to-mesenchymal transition and therapeutic intervention in diabetic nephropathy. Am J Nephrol. 31:68–74. 2010. View Article : Google Scholar

47 

Zhu H, Shyh-Chang N, Segre AV, Shinoda G, Shah SP, Einhorn WS, Takeuchi A, Engreitz JM, Hagan JP, Kharas MG, et al: The Lin28/let-7 axis regulates glucose metabolism. Cell. 147:81–94. 2011. View Article : Google Scholar : PubMed/NCBI

48 

Frost RJ and Olson EN: Control of glucose homeostasis and insulin sensitivity by the Let-7 family of microRNAs. Proc Natl Acad Sci USA. 108:21075–21080. 2011. View Article : Google Scholar : PubMed/NCBI

49 

Ji J, Tao P and He L: Kangxianling decoction prevents renal fibrosis in rats with 5/6 nephrectomy and inhibits Ang II-induced ECM production in glomerular mesangial cells. J Pharmacol Sci. 139:367–372. 2019. View Article : Google Scholar : PubMed/NCBI

50 

Lee EJ, Kang MK, Kim DY, Kim YH, Oh H and Kang YH: Chrysin inhibits advanced glycation end products-induced kidney fibrosis in renal mesangial cells and diabetic kidneys. Nutrients. 10:E8822018. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2019
Volume 44 Issue 5

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
Li N, Wang LJ, Xu WL, Liu S and Yu JY: MicroRNA‑379‑5p suppresses renal fibrosis by regulating the LIN28/let‑7 axis in diabetic nephropathy. Int J Mol Med 44: 1619-1628, 2019.
APA
Li, N., Wang, L., Xu, W., Liu , S., & Yu, J. (2019). MicroRNA‑379‑5p suppresses renal fibrosis by regulating the LIN28/let‑7 axis in diabetic nephropathy. International Journal of Molecular Medicine, 44, 1619-1628. https://doi.org/10.3892/ijmm.2019.4325
MLA
Li, N., Wang, L., Xu, W., Liu , S., Yu, J."MicroRNA‑379‑5p suppresses renal fibrosis by regulating the LIN28/let‑7 axis in diabetic nephropathy". International Journal of Molecular Medicine 44.5 (2019): 1619-1628.
Chicago
Li, N., Wang, L., Xu, W., Liu , S., Yu, J."MicroRNA‑379‑5p suppresses renal fibrosis by regulating the LIN28/let‑7 axis in diabetic nephropathy". International Journal of Molecular Medicine 44, no. 5 (2019): 1619-1628. https://doi.org/10.3892/ijmm.2019.4325