Open Access

Rosiglitazone prevents acute pancreatitis through inhibiting microRNA‑26a expression

  • Authors:
    • Yan Chen
    • Wei Xiang
    • Xiang Li
    • Daming Wang
    • Chunyan Qian
  • View Affiliations

  • Published online on: June 26, 2019     https://doi.org/10.3892/etm.2019.7711
  • Pages: 1246-1252
  • Copyright: © Chen et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

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


Abstract

The aim of the present study was to investigate the regulatory effect of rosiglitazone on the progression of acute pancreatitis (AP) and pancreas injury, and the underlying mechanism. An AP rat model was established using caerulein and validated by detection of amylase, lipase, tumor necrosis factor‑α (TNF‑α), interleukin‑6 (IL‑6) and transforming growth factor‑β (TGF‑β) serum levels. Pancreatic injury was assessed by pathological examination. The expression levels of microRNA (miR)‑26a in AP rats and AR42J cells were analyzed using reverse transcription‑quantitative PCR (RT‑qPCR). Luciferase reporter gene assay was applied for detecting whether miR‑26a bound to the target gene phosphatase and tensin homolog (PTEN). The regulatory effect of rosiglitazone on the PI3K/AKT signaling pathway was analyzed by western blot analysis. Results demonstrated that establishment of an AP model was successful with severe pancreas injury and classic AP phenotypes observed in rats. Increased serum expression of amylase, lipase, TNF‑α, IL‑6 and TGF‑β were observed in AP rats. Rosiglitazone pretreatment prevented AP progression through suppression of miR‑26a expression via binding to and degrading PTEN. Western blot analysis demonstrated that rosiglitazone blocked the PI3K/AKT signaling pathway through PTEN. In conclusion, it was determined that rosiglitazone prevented AP by downregulating miR‑26a via the PI3K/AKT signaling pathway.

Introduction

Acute pancreatitis (AP) is an inflammatory disease that affects the pancreas. The pathogenesis of AP is complex and involves multiple factors, such as hyperlipidemia, alcoholism and biliary diseases (1). The prognosis of mild AP is typically good, with a very low mortality rate; however, severe AP may cause serious consequences and have a poor prognosis (2). AP progression is associated with the activation of pancreatin, cytokines and chemokines (3,4). Therefore, it is of great clinical significance to explore the inflammation mechanism and to find a potential treatment that targets the inflammation process.

Rosiglitazone, currently the most effective thiazolidinedione drug, is principally used for the treatment of diabetes (5,6). Numerous studies have demonstrated that rosiglitazone can increase insulin sensitivity and decrease insulin resistance (57). However, in recent years, due to better understanding of peroxisome proliferator activated receptor (PPAR)γ and its ligands, rosiglitazone has been determined to have significant effects on the inflammatory response, cell differentiation and cell metabolism (810). Previous research suggested that rosiglitazone exhibits anti-inflammatory effects on osteoporosis, acute or chronic gastrointestinal diseases and other systemic inflammatory response syndromes (7,1112).

MicroRNA (miRNA) is a non-coding, single-chain RNA (18–25 nucleotides in length), which can bind to the 3′-untranslated region (UTR) of target genes and suppress the translation or promote the degradation of genes (13). Although miRNA only accounts for ~1% of the human genome, it regulates ~60% protein expression (14). Present studies determined that miRNAs are involved in various cellular functions, such as proliferation, differentiation and the inflammation response (13,14). miRNA (miR)-26 is located in chromosome 19q14.12, which is closely associated with tumor development by regulation of tumor cell proliferation and apoptosis (15). In addition, miR-26a is involved in the allergic inflammatory reaction and the toll like receptor 4 (TLR-4)-mediated inflammatory response (16,17). However, the specific role of miR-26a in AP has not been fully elucidated.

The aim of the present study was to investigate the regulatory effect of rosiglitazone on the progression of AP and pancreas injury, and its underlying mechanism.

Materials and methods

Animal model

A total of 40 male Sprague Dawley rats (age, 6–8 weeks; weight, 180–200 g) were purchased from Model Animal Research Center of Nanjing University (Nanjing, China). The rats were housed in a temperature-controlled room (21±2°C) with 60–70% relative humidity on a 12-h light: dark cycle (lights on at 06:00). All rats had free access to water and food. The rats of specific pathogen free level were randomly divided into three groups: The control group (n=10), the AP model group (n=15) and the rosiglitazone-treated group (n=15). AP model rats were anesthetized by intraperitoneal injection of pentobarbital sodium (30 mg/kg) then injected intraperitoneally with 50 µg/kg caerulein (MedChemExpress) five times at 1-h intervals. Rats in the control group were given the same volume (1 ml each time) of 0.9% NaCl. Rats in the rosiglitazone-treated group were administered with rosiglitazone (4 mg/kg, Sigma-Aldrich; Merck KGaA) by intraperitoneal injection 1 h before the first injection of caerulein. Peripheral blood was collected 6, 12 and 24 h after the last injection of caerulein then the rats were sacrificed for pancreatic tissues. Parts of the pancreatic tissues were stored in liquid nitrogen, whilst the remainder were fixed for histological analysis. All animals used in the experiment were obtained from the Model Animal Research Center of Nanjing University. This experiment was approved by Soochow University Ethics Committee (Soochow, China).

ELISA

Blood samples were maintained at room temperature for 20 min then centrifuged at 1,000 × g at 4°C for 15 min for serum sample preparation. Serum contents of amylase (cat. no. SL-B23234), lipase (cat. no. LM04412B), tumor necrosis factor-α (TNF-α; cat. no. QY-R2813), interleukin-6 (IL-6; cat. no. HS2102) and transforming growth factor-β (TGF-β; cat. no. FS-E6931) were determined using ELISA kits purchased from Guidechem. In brief, the standard solution was added to each well and incubated for 2 h at 20°C. Then the liquid was removed and an anti-biotin antibody was added for a 1-h incubation at 20°C. Each well was washed then horseradish peroxidase-labeled streptavidin work solution was added. Following a 1-h incubation at 20°C, substrates were added for color development in the dark. Termination solution was added 15–30 min later, prior to detection of optical density.

AP assessment in rats

Pancreas tissues were fixed in 10% neutral buffered formaldehyde for >24 h at room temperature. Tissue samples were embedded in paraffin blocks and sliced into 5 µm sections for hematoxylin and eosin (H&E) staining (hematoxylin, 5 min at room temperature and eosin, 3 min at room temperature). Histological analysis was performed using a light microscope (magnification, ×400).

Histological scoring of pancreatic tissue was performed to grade the severity and extent of acinar edema (0, no edema; 1, inter lobular edema; 2, intralobular edema; and 3, inter acinar edema), inflammation [0, no inflammation; 1, inflammatory cells in ducts; 2, inflammatory cells in the parenchyma (<50% of the lobules); and 3, inflammatory cells in the parenchyma (>50% of the lobules)] and finally acinar cell necrosis (0, no necrosis; 1, <5% necrosis; 2, 5–20% necrosis; and 3, 20–50% necrosis).

Bioinformatic analysis

Bioinformatic analysis predicted that PTEN was the potential target gene of miR-26a (http://www.targetscan.org). In detail, rat was selected as species in the first search box and miR-26a-5p was put into the microRNA name search box. PTEN was found in the table of results.

Cell culture and transfection

Rat pancreatic AR42J cells (Type Culture Collection of the Chinese Academy of Sciences) were cultured in DMEM-F12 containing 20% fetal bovine serum (both Gibco; Thermo Fisher Scientific, Inc.), 100 U/ml penicillin and 100 g/l streptomycin and maintained in a 5% CO2 incubator. The culture medium was changed every 2 days.

AR42J cells were inoculated into 24-well plates at 1×105 cells/well. When the cell density reached 70–80%, miR-26a inhibitor (hsa-miR-26a-in; Hanbio Biotechnology Co., Ltd.), miR-26a mimics (hsa-miR-26a-mi; Hanbio Biotechnology Co., Ltd), phosphatase and tensin homolog (PTEN) small interfering (si)-RNA and related non-targeting control (NC) siRNA (both GenePharma) were transfected into cells using Lipofectamine® 2000 (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions. The sequences were as follows: PTEN siRNA forward, 5′-AACCCACCACAGCUAGAACTT-3′ and reverse, 5′-AAGUUCUAGCUGUGGUGGGTT-3′ and NC siRNA forward, 5′-UUCUCCGAACGUGUCACGUTT-3′ and reverse, 5′-ACGUGACACGUUCGGAGAATT-3′. Following incubation for 24–48 h, cells were collected for the following experiments.

In vitro AP model

One day prior to model establishment, AR42J cells were inoculated into 6-well plates with 1×105/ml and treated with 10 nmol/l caerulein. Following caerulein treatment for 8 h at 37°C cells were then treated with 0, 0.01, 0.1 or 1 µM rosiglitazone for 24 h.

RNA extraction and reverse transcription-quantitative (RT-q)PCR

Total RNA from cells was extracted with TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc.), and then mixed with radioimmunoprecipitation assay (RIPA) lysis buffer (Beyotime Institute of Biotechnology). The same volume of chloroform was added into the mixture and centrifuged at 10,000 × g at 4°C for 5 min. The aqueous phase was collected into Eppendorf tubes. Following addition of 0.5 ml 99% isopropanol to the mixture, the Eppendorf tubes were rinsed with ethanol to remove the residue then samples were air dried at the room temperature. Total RNA was reverse transcribed into cDNA using Takara PrimeScript™ RT Master Mix kit (Takara Biotechnology Co., Ltd.). qPCR was performed using SYBR® Green Master Mix (Takara Bio, Inc.). The thermocycling conditions as: 40 cycles of 95°C for 30 sec, 95°C for 5 sec and 60°C for 31 sec. GAPDH served as the internal control for PTEN, while U6 served as the internal control for miR-204. Primers sequences were as follows: miR-26a-5p forward, 5′-GGATCCGCAGAAACTCCAGAGAGAAGGA-3′ and reverse, 5′-AAGCTTGCCTTTAGCAGAAAGGAGGTT-3′; PTEN forward, 5′-GTTTACCGGCAGCATCAAAT-3′ and reverse, 5′-CCCCCACTTTAGTGCACAGT-3′; GAPDH forward, 5′-CGGAGTCAACGGATTTGGTCGTAT-3′ and reverse, 5′-AGCCTTCTCCATGGTGGTGAAGAC-3′; and U6 forward, 5′-CTCGCTTCGGCAGCACA-3′ and reverse, 5′-AACGCTTCACGAATTTGCGT-3′. The 2−ΔΔCq method was used to determine the relative expression levels (18). mRNA expression levels were normalized to GAPDH, whereas miRNA expression levels were normalized to U6.

Western blot analysis

Cells in the logarithmic growth phase were collected and digested by pancreatin, subsequently rinsed by PBS and fully lysed by RIPA buffer (Beyotime Institute of Biotechnology). Following oscillating incubation at 4°C for 10 min, cells were decomposed using sonication at 20 kHz at 4°C for 2 min then centrifuged at 5,000 × g for 10 min at 4°C for supernatant preparation. Protein concentration was quantified using bicinchoninic acid protein assay (Pierce; Thermo Fisher Scientific, Inc.). Proteins (10 µg) were separated by 12% SDS-PAGE, followed by an electrophoretic transfer onto polyvinylidene fluoride membranes (EMD Millipore). Membranes were placed into 5% non-fat milk to block non-specific binding at 25°C for 1 h. Membranes were then incubated with primary antibodies including PTEN (1:500; cat. no. ab32199; Abcam), phosphorylated (p)-PI3K (1:500; cat. no. ab182651; Abcam), PI3K (1:500; cat. no. ab151549; Abcam), p-AKT (1:500; cat. no. ab38449; Abcam), AKT (1:500; cat. no. ab8805; Abcam), GAPDH (1:500; cat. no. ab8245; Abcam) overnight at 4°C. Finally, the membranes were rinsed with PBS and incubated with goat horseradish peroxidase-conjugated goat anti-rabbit IgG H&L secondary antibody (1:1,000; cat. no. ab7090; Abcam) at 25°C for 1 h. Protein bands were visualized following a 3-min incubation with enhanced chemiluminescence reagent (Thermo Fisher Scientific, Inc.). Quantity One (version 4.0; Bio-Rad Laboratories, Inc.) was used for densitometric analysis.

Luciferase reporter gene assay

Cells were seeded and cultured into 24-well plates at a density of 5×104 cells/well. The cells were transfected with Lipofectamine 2000 (Thermo Fisher Scientific, Inc.). The pGL3-PTEN-3′UTR wild-type or mutant plasmid (0.5 µg/ml; Hanbio Biotechnology Co., Ltd) was co-transfected with miR-26a mimic (0.2 µg/ml) or miR-26a NC (0.2 µg/ml) and pRL-TK Renilla plasmid (0.02 µg/ml; Promega Corporation) into the cells. Following incubation for 48 h at room temperature, cells were collected for analysis of the luciferase activities of both firefly and Renilla using a Dual Luciferase® Reporter Assay System (Promega Corporation). Firefly luciferase activity was normalized by comparing the activity levels to pRL-TK Renilla.

Statistical analysis

Statistical analysis was performed by Statistical Product and Service Solutions v.19.0 (IBM Corp.). All data were expressed as mean ± standard deviation. Statistical difference was assessed using two-tailed Student t-test for comparisons amongst two groups. Comparisons between multiple groups was performed using one-way analysis of variance test followed by Least Significant Difference post hoc test. P<0.05 was considered to indicate statistical significance.

Results

Rosiglitazone reduces serum levels of amylase and cytokines

Rats were intraperitoneally injected with caerulein for the establishment of an AP rat model. Results demonstrated that serum expressions of amylase, lipase, TNF-α, IL-6 and TGF-β were significantly increased in the AP model group compared with control group, whilst the levels decreased in the rosiglitazone-treated group (Fig. 1A-E). Pathological examination of the pancreas indicated that caerulein induced infiltration of immune cells and pancreas injury (Fig. 1F). Rosiglitazone pretreatment remarkably and significantly reduced the level of pancreas injury at all timepoints compared with caerulein treatment (Fig. 1G).

Rosiglitazone suppresses miR-26a expression

AR42J cells were pretreated with different concentrations of rosiglitazone (0, 0.01, 0.1 and 1 µM). Results revealed that miR-26a expression in AR42J cells was significantly decreased in what appears to be a dose-dependent manner by rosiglitazone compared with the untreated group (Fig. 2A) whilst PTEN mRNA expression was significantly increased in what appears to be a dose-dependent manner compared with the untreated group (Fig. 2B). In addition, expression levels of miR-26a in pancreatic tissues and serum of rats was determined. miR-26a expression was significantly increased in the AP model group but decreased with rosiglitazone pretreatment compared with control group (Fig. 2C and D). Results indicated that rosiglitazone may regulate AP via miR-26a.

miR-26a regulates PTEN expression whilst PTEN has no effect on miR-26a

Bioinformatic analysis predicted that PTEN was the potential target gene of miR-26a (http://www.targetscan.org). Transfection efficacy of constructed plasmids was first verified by RT-qPCR as miR-26a mimics significantly increased and miR-26a inhibitor significantly decreased expression compared with the miR-26a NC group (Fig. 3A). It was identified that miR-26a overexpression significantly suppressed PTEN expression and miR-26a inhibition significantly increased PTEN levels compared with the miR-26a NC group (Fig. 3B). Similar results were produced when detecting PTEN protein levels (Fig. 3C and D). The luciferase reporter gene assay demonstrated that miR-26a mimics significantly decreased the luciferase activity of cells co-transfected with PTEN-WT compared with those co-transected with miR-26a NC, which suggested that miR-26a could directly bind to PTEN and inhibit its expression (Fig. 3E). The expression of PTEN was significantly decreased in cells transfected with si-PTEN compared with those transfected with NC siRNA (Fig. 3F). By contrast, the expression of miR-26a was not changed significantly following PTEN knockdown (Fig. 3G).

Rosiglitazone suppresses the PI3K/AKT signaling pathway

A previous study demonstrated that the biological functions of PTEN were mediated by the PI3K/AKT signaling pathway (19). Therefore, the effect of rosiglitazone on the PI3K/AKT signaling pathway were investigated. Rosiglitazone pretreatment significantly suppressed the phosphorylation of key proteins involved in the PI3K/AKT signaling pathway compared with the control group (Fig. 4A). The serum levels of amylase and lipase were significantly increased in the PTEN knockdown group compared with the rosiglitazone + NC-siRNA group, indicating that PTEN knockdown reversed the beneficial effect of rosiglitazone on serum levels of amylase and lipase (Fig. 4B). In addition, the ratios of p-PI3K/PI3K and p-AKT/AKT were significantly increased in the PTEN knockdown group compared with the rosiglitazone + NC-siRNA group, indicating that PTEN knockdown reversed the inhibitory effect of rosiglitazone on the PI3K/AKT signaling pathway (Fig. 4C). These results suggest that rosiglitazone regulated the PI3K/AKT signaling pathway via PTEN.

Discussion

During the initial stage of AP, TNF-α is the main regulatory factor responsible for triggering the inflammatory cascade. Activation of the immune system elevates inflammatory signaling, and further leads to cell injury and necrosis. The TLR-4-mediated inflammatory reaction can active multiple cytokines (20,21). The present study demonstrated that expression levels of amylase, lipase, TNF-α, IL-6 and TGF-β were significantly increased following caerulein treatment, which suggested successful establishment of the AP model and pancreas injury. Rats pretreated with intraperitoneal injection of rosiglitazone significantly attenuated the inflammatory response and pancreas injury. Therefore, the present study next explored the underlying mechanism of rosiglitazone.

Previous studies have identified that miRNAs serve important roles in the progression of inflammatory diseases. Wu et al (22) reported that miRNA regulates macrophage polarity and thus controls the inflammatory reaction. In addition, miRNA is associated with various inflammatory diseases. For example, miR-365 directly suppresses the expression of histone deacetylase 4 and contributes to the development of rheumatoid arthritis (23). Sorbin and SH3 domain containing 2-mediated cardiac dysfunction during sepsis is regulated by miR-21-3p (24). Since miRNA can regulate the expressions of several critical components and cytokines, it has become an important diagnostic and therapeutic target for rheumatoid arthritis (25). In the present study, rosiglitazone suppressed miR-26a expression, thus resulting in the elevated expression of the target gene PTEN.

PTEN and the PTEN-mediated pathway are involved in the occurrence and development of various diseases (26). Previous studies have demonstrated that the biological function of PTEN involved regulation of cell survival, cell proliferation and inflammation via the P13K/AKT signaling pathway (27,28). Inflammatory mediators can lead to the activation and chemotaxis of immune cells via the PI3K pathway (29). The present study demonstrated that decreased expression of PTEN reduced the inhibitory effect of miR-26a on the PI3K/AKT pathway, thereby regulating inflammation. However, the underlying mechanism of rosiglitazone suppression on the PI3K/AKT pathway remains poorly understood. Future work will use the PI3K/AKT inhibitor wortmannin to further investigate the underlying mechanism

In conclusion, rosiglitazone prevented AP progression through suppressing miR-26a expression, which elevated expression of PTEN. PTEN has been implicated in the development of various diseases therefore research into the gene can provide potential novel strategies for treatment.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

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

Authors' contribution

YC and CQ designed the study and performed the experiments. YC, WX and XL established the animal models. YC and DW collected the data. YC and WX analyzed the data. YC and CQ prepared the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

This study was approved by the Soochow University Ethics Committee (Soochow, China).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Banks PA and Freeman ML; Practice Parameters Committee of the American College of Gastroenterology, : Practice guidelines in acute pancreatitis. Am J Gastroenterol. 101:2379–2400. 2006. View Article : Google Scholar : PubMed/NCBI

2 

Banks PA, Bollen TL, Dervenis C, Gooszen HG, Johnson CD, Sarr MG, Tsiotos GG and Vege SS; AcutePancreatitis Classification Working Group, : Classification of acute pancreatitis-2012: Revision of the Atlanta classification and definitions by international consensus. Gut. 62:102–111. 2013. View Article : Google Scholar : PubMed/NCBI

3 

Kylanpaa ML, Repo H and Puolakkainen PA: Inflammation and immunosuppression in severe acute pancreatitis. World J Gastroenterol. 16:2867–2872. 2010. View Article : Google Scholar : PubMed/NCBI

4 

Petrov M: Nutrition, inflammation, and acute pancreatitis. ISRN Inflamm. 2013:3414102013. View Article : Google Scholar : PubMed/NCBI

5 

Kahn SE, Haffner SM, Heise MA, Herman WH, Holman RR, Jones NP, Kravitz BG, Lachin JM, O'Neill MC, Zinman B, et al: Glycemic durability of rosiglitazone, metformin, or glyburide monotherapy. N Engl J Med. 355:2427–2443. 2006. View Article : Google Scholar : PubMed/NCBI

6 

Fryer LG, Parbu-Patel A and Carling D: The anti-diabetic drugs rosiglitazone and metformin stimulate amp-activated protein kinase through distinct signaling pathways. J Biol Chem. 277:25226–25232. 2002. View Article : Google Scholar : PubMed/NCBI

7 

Ramakers JD, Verstege MI, Thuijls G, Te VA, Mensink RP and Plat J: The ppargamma agonist rosiglitazone impairs colonic inflammation in mice with experimental colitis. J Clin Immunol. 27:275–283. 2007. View Article : Google Scholar : PubMed/NCBI

8 

Ji XX, Ji XJ, Li QQ, Lu XX and Luo L: Rosiglitazone reduces apoptosis and inflammation in lipopolysaccharide-induced human umbilical vein endothelial cells. Med Sci Monit. 24:6200–6207. 2018. View Article : Google Scholar : PubMed/NCBI

9 

Ding F, Qiu J, Li Q, Hu J, Song C, Han C, He H and Wang J: Effects of rosiglitazone on proliferation and differentiation of duck preadipocytes. In Vitro Cell Dev Biol Anim. 52:174–181. 2016. View Article : Google Scholar : PubMed/NCBI

10 

Cheng Y, Li S, Wang M, Cheng C and Liu R: Peroxisome proliferator activated receptor gamma (PPARgamma) agonist rosiglitazone ameliorate airway inflammation by inhibiting toll-like receptor 2 (TLR2)/Nod-like receptor with pyrin domain containing 3 (NLRP3) inflammatory corpuscle activation in asthmatic Mice. Med Sci Monit. 24:9045–9053. 2018. View Article : Google Scholar : PubMed/NCBI

11 

Levi Z, Shaish A, Yacov N, Levkovitz H, Trestman S, Gerber Y, Cohen H, Dvir A, Rhachmani R, Ravid M and Harats D: Rosiglitazone (PPARgamma-agonist) attenuates atherogenesis with no effect on hyperglycaemia in a combined diabetes-atherosclerosis mouse model. Diabetes Obes Metab. 5:45–50. 2003. View Article : Google Scholar : PubMed/NCBI

12 

Hassumi MY, Silva-Filho VJ, Campos-Junior JC, Vieira SM, Cunha FQ, Alves PM, Alves JB, Kawai T, Gonçalves RB and Napimoga MH: PPAR-gamma agonist rosiglitazone prevents inflammatory periodontal bone loss by inhibiting osteoclastogenesis. Int Immunopharmacol. 9:1150–1158. 2009. View Article : Google Scholar : PubMed/NCBI

13 

Lin S and Gregory RI: Microrna biogenesis pathways in cancer. Nat Rev Cancer. 15:321–333. 2015. View Article : Google Scholar : PubMed/NCBI

14 

Iorio MV, Visone R, Di Leva G, Donati V, Petrocca F, Casalini P, Taccioli C, Volinia S, Liu CG, Alder H, et al: Microrna signatures in human ovarian cancer. Cancer Res. 67:8699–8707. 2007. View Article : Google Scholar : PubMed/NCBI

15 

Lu J, He ML, Wang L, Chen Y, Liu X, Dong Q, Chen YC, Peng Y, Yao KT, Kung HF and Li XP: Mir-26a inhibits cell growth and tumorigenesis of nasopharyngeal carcinoma through repression of ezh2. Cancer Res. 71:225–233. 2011. View Article : Google Scholar : PubMed/NCBI

16 

Kwon Y, Kim Y, Eom S, Kim M, Park D, Kim H, Noh K, Lee H, Lee YS, Choe J, et al: Microrna-26a/-26b-cox-2-mip-2 loop regulates allergic inflammation and allergic inflammation-promoted enhanced tumorigenic and metastatic potential of cancer cells. J Biol Chem. 290:14245–14266. 2015. View Article : Google Scholar : PubMed/NCBI

17 

Kumar A, Bhatia HS, de Oliveira AC and Fiebich BL: Microrna-26a modulates inflammatory response induced by toll-like receptor 4 stimulation in microglia. J Neurochem. 135:1189–1202. 2015. View Article : Google Scholar : PubMed/NCBI

18 

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 : PubMed/NCBI

19 

Georgescu MM: PTEN tumor suppressor network in PI3K-AKT pathway control. Genes Cancer. 1:1170–1177. 2010. View Article : Google Scholar : PubMed/NCBI

20 

Chen L, Yu CX, Song B, Cai W, Liu C and Guan QB: Free fatty acids mediates human umbilical vein endothelial cells inflammation through toll-like receptor-4. Eur Rev Med Pharmacol Sci. 22:2421–2431. 2018.PubMed/NCBI

21 

Sah RP, Garg P and Saluja AK: Pathogenic mechanisms of acute pancreatitis. Curr Opin Gastroenterol. 28:507–515. 2012. View Article : Google Scholar : PubMed/NCBI

22 

Wu XQ, Dai Y, Yang Y, Huang C, Meng XM, Wu BM and Li J: Emerging role of microRNAS in regulating macrophage activation and polarization in immune response and inflammation. Immunology. 148:237–248. 2016. View Article : Google Scholar : PubMed/NCBI

23 

Yang X, Guan Y, Tian S, Wang Y, Sun K and Chen Q: Mechanical and IL-1β responsive miR-365 contributes to osteoarthritis development by targeting histone deacetylase 4. Int J Mol Sci. 17:4362016. View Article : Google Scholar : PubMed/NCBI

24 

Wang H, Bei Y, Shen S, Huang P, Shi J, Zhang J, Sun Q, Chen Y, Yang Y, Xu T, et al: miR-21-3p controls sepsis-associated cardiac dysfunction via regulating SORBS2. J Mol Cell Cardiol. 94:43–53. 2016. View Article : Google Scholar : PubMed/NCBI

25 

Sharma AR, Sharma G, Lee SS and Chakraborty C: Mirna-regulated key components of cytokine signaling pathways and inflammation in rheumatoid arthritis. Med Res Rev. 36:425–439. 2016. View Article : Google Scholar : PubMed/NCBI

26 

Chalhoub N and Baker SJ: PTEN and the PI3-kinase pathway in cancer. Annu Rev Pathol. 4:127–150. 2009. View Article : Google Scholar : PubMed/NCBI

27 

Suzuki A, Yamaguchi MT, Ohteki T, Sasaki T, Kaisho T, Kimura Y, Yoshida R, Wakeham A, Higuchi T, Fukumoto M, et al: T cell-specific loss of Pten leads to defects in central and peripheral tolerance. Immunity. 14:523–534. 2001. View Article : Google Scholar : PubMed/NCBI

28 

Paez J and Sellers WR: PI3K/PTEN/AKT pathway. A critical mediator of oncogenic signaling. Cancer Treat Res. 115:145–167. 2003. View Article : Google Scholar : PubMed/NCBI

29 

Weichhart T and Saemann MD: The PI3K/AKT/MTOR pathway in innate immune cells: Emerging therapeutic applications. Ann Rheum Dis. 67 (Suppl 3):iii70–iii74. 2008. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2019
Volume 18 Issue 2

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

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Chen Y, Xiang W, Li X, Wang D and Qian C: Rosiglitazone prevents acute pancreatitis through inhibiting microRNA‑26a expression. Exp Ther Med 18: 1246-1252, 2019
APA
Chen, Y., Xiang, W., Li, X., Wang, D., & Qian, C. (2019). Rosiglitazone prevents acute pancreatitis through inhibiting microRNA‑26a expression. Experimental and Therapeutic Medicine, 18, 1246-1252. https://doi.org/10.3892/etm.2019.7711
MLA
Chen, Y., Xiang, W., Li, X., Wang, D., Qian, C."Rosiglitazone prevents acute pancreatitis through inhibiting microRNA‑26a expression". Experimental and Therapeutic Medicine 18.2 (2019): 1246-1252.
Chicago
Chen, Y., Xiang, W., Li, X., Wang, D., Qian, C."Rosiglitazone prevents acute pancreatitis through inhibiting microRNA‑26a expression". Experimental and Therapeutic Medicine 18, no. 2 (2019): 1246-1252. https://doi.org/10.3892/etm.2019.7711