Ang-(1-7) protects HUVECs from high glucose-induced injury and inflammation via inhibition of the JAK2/STAT3 pathway

  • Authors:
    • Jianfang Chen
    • Wei Zhang
    • Qing Xu
    • Jihua Zhang
    • Wei Chen
    • Zhengrong Xu
    • Chaosheng Li
    • Zhenhua Wang
    • Yao Zhang
    • Yulan Zhen
    • Jianqiang Feng
    • Jun Chen
    • Jingfu Chen
  • View Affiliations

  • Published online on: February 22, 2018     https://doi.org/10.3892/ijmm.2018.3507
  • Pages: 2865-2878
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Angiotensin (Ang)‑1‑7, which is catalyzed by angiotensin‑converting enzyme 2 (ACE2) from angiotensin‑II (Ang‑II), exerts multiple biological and pharmacological effects, including cardioprotective effects and endothelial protection. The Janus kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3) pathway has been demonstrated to be involved in diabetes‑associated cardiovascular complications. The present study hypothesized that Ang‑(1‑7) protects against high glucose (HG)‑induced endothelial cell injury and inflammation by inhibiting the JAK2/STAT3 pathway in human umbilical vein endothelial cells (HUVECs). HUVECs were treated with 40 mmol/l glucose (HG) for 24 h to establish a model of HG‑induced endothelial cell injury and inflammation. Protein expression levels of p‑JAK2, t‑JAK2, p‑STAT3, t‑STAT3, NOX‑4, eNOS and cleaved caspase‑3 were tested by western blotting. CCK‑8 assay was performed to assess cell viability of HUVECs. Apoptotic cell death was analyzed by Hoechst 33258 staining. Mitochondrial membrane potential (MMP) was obtained using JC‑1. Superoxide dismutase (SOD) activity was tested by SOD assay kit. Interleukin (IL)‑1β, IL‑10, IL‑12 and TNF‑α levels in culture media were tested by ELISA. The findings demonstrated that exposure of HUVECs to HG for 24 h induced injury and inflammation. This injury and inflammation were significantly ameliorated by pre‑treatment of cells with either Ang‑(1‑7) or AG490, an inhibitor of the JAK2/STAT3 pathway, prior to exposure of the cells to HG. Exposure of the cells to HG also increased the phosphorylation of JAK2/STAT3 (p‑JAK2 and p‑STAT3). Increased activation of the JAK2/STAT3 pathway was attenuated by pre‑treatment with Ang‑(1‑7). To the best of our knowledge, the findings from the present study provided the first evidence that Ang‑(1‑7) protects against HG‑induced injury and inflammation by inhibiting activation of the JAK2/STAT3 pathway in HUVECs.

Introduction

The incidence of diabetes mellitus (DM) is estimated to rise to 7.7% by 2030 globally (1) and is predicted to affect 591,900,000 individuals by 2035 (2). DM is a major and increasing health problem worldwide due to its increasing incidence, which can lead to a variety of complications, including diabetic retinopathy, diabetic nephropathy, diabetic neuropathy, diabetic foot and cardiovascular complications. Of note, cardiovascular complications are the major cause of diabetes-associated mortality (3). The vascular endothelium is central to the pathogenesis of diabetic complications, with vascular endothelial cells mainly involved in maintaining endothelial dysfunction; cardiovascular homeostasis is considered an important factor in the pathogenesis of diabetes-associated vascular complications (4,5). Studies have revealed that hyperglycemia induces numerous pathological changes in vascular endothelial cell injury, including oxidative stress (6), inflammation (7), increased endothelial cell apoptosis (6,8) and mitochondrial membrane permeabilization (9). However, the associated molecular mechanism by which hyperglycemia results in vascular endothelial cell injury in DM remains to be elucidated.

As one of four protein-tyrosine kinases, Janus kinase (JAK)1, JAK2, JAK3 and Tyk2, JAK2 is an essential factor in cellular proliferation, differentiation, survival and senescence. JAK2 also regulates other signaling molecules, including the RAS, signal transducer and activator of transcription (STAT)5, STAT3 and phosphoinositide 3-kinase/Protein kinase B (PI3K/AKT) pathways (10). STAT3 is a member of the STAT family (STAT1, STAT2, STAT3, STAT4, STAT5a, STAT5b and STAT6), and its phosphorylation is induced by the activation of JAK2; phosphorylated (p-)STAT3 forms a dimer and translocates into the nucleus from the cytoplasm, where it binds to related sequences and alters the expression of various target genes. Therefore, the JAK2/STAT3 pathway is gradually being recognized as a membrane-to-nucleus pathway for a variety of stimulating responses in vitro and in vivo (11-13). Accumulating evidence has shown that the JAK2/STAT3 pathway is a cell survival signal, which contributes to cell proliferation, differentiation, growth and apoptosis (14-16). The JAK2/STAT3 pathway is also important in the progress of cardiovascular diseases. Manea et al (17) demonstrated that the JAK2/STAT3 pathway is a crucial regulator of the response of EAhy926 endothelial cells to diabetes-associated cardiovascular dysfunction. Hyperglycemia increases angiotensin (Ang)-II-induced vascular smooth muscle cell proliferation by increasing JAK2/STAT3 pathway transduction (18). These findings indicate that the JAK2/STAT3 pathway is important in cardiovascular diseases and endothelial dysfunction. However, the effects of the JAK2/STAT3 pathway on high glucose (HG)-induced endothelial dysfunction in vivo, and the associated mechanisms, remain to be elucidated.

There has been increasing focus on the protective effects of Ang-(1-7) against hyperglycemia-induced cardiovascular complication. The secretory level of plasma Ang-(1-7) was demonstrated to be low in patients with diabetic-induced cardiac dysfunction (19). The expression level of angiotensin-converting enzyme 2 (ACE2), which is responsible for the tissue degradation of Ang-II into Ang-(1-7), was also observed to be decreased in patients with diabetes (20). An increase in the expression levels of ACE2 markedly improved control of blood glucose levels and alleviated glomerular injury in streptozotocin (STZ)-induced diabetic mice (21), and Ang-(1-7) was reported to exert protective effects against diabetes-induced cardiovascular events (22,23). In addition, the Ang-(1-7) and ACE2/Ang-(1-7)/Mas receptor axis was revealed to have additional beneficial effects in preventing diabetes-induced cardiac dysfunction (24). These findings indicate that Ang-(1-7) is closely associated with hyperglycemia-induced cardiovascular dysfunction. However, the underlying mechanisms between Ang-(1-7) and hyperglycemia-induced cardiovascular dysfunction remain to be fully elucidated.

A previous study reported that Ang-(1-7) produced an inhibitory effect on the activation of JAK2/STAT3 (25). In diabetic nephropathy, Ang-(1-7) exerts renoprotective effects on diabetic nephropathy via inhibiting the STAT3 pathway, apparent as a reduction in inflammation, fibrosis, oxidative stress and lipotoxicity (26). The present study tested the hypothesis that exogenous Ang-(1-7) protects human umbilical vein endothelial cells (HUVECs) against HG-induced injury and inflammation by inhibiting the JAK2/STAT3 pathway.

Materials and methods

Materials

Ang-(1-7) was purchased from Sigma; Merck KGaA (Darmstadt, Germany) and stored at −20°C. The following 2′,7′-dichlorofluorescein diacetate (DCFH-DH), Hoechst 33258, 5′,6,6′-tetrachloro-1,1′,3,3′-tetraethyl-imida-carbocyanine iodide (JC-1) and AG490 (an inhibitor of the STAT3/JAK2 pathway) were obtained from Sigma-Aldrich; Merck KGaA). The Cell Counting Kit-8 (CCK-8) was supplied by Dojindo Molecular Technologies, Inc. (Kumamoto, Japan). Fetal bovine serum (FBS) and Dulbecco's modified Eagle's medium (DMEM) were purchased from Gibco; Thermo Fisher Scientific, Inc. (Waltham, MA, USA). Anti-p-STAT3 antibody (cat. no. SAB4300033), anti-total (t-)STAT3 antibody (cat. no. SAB4300708), anti-p-JAK2 antibody (cat. no. SAB4300124), anti-t-JAK2 antibody (cat. no. SAB4501599), anti-caspase-3 antibody (cat. no. C5737) anti-NADPH oxidase 4 (Nox4) antibody (cat. no. SAB4503153) and anti-endothelial nitric oxide synthase (eNOS) antibody (cat. no. N2643) were supplied by Cell Signaling Technology, Inc. (Boston, MA, USA), horseradish peroxidase (HRP)-conjugated secondary antibody (cat. no. KC-5A08) and a BCA protein assay kit were obtained from KangChen Biotech, Inc. (Shanghai, China). Enhanced chemiluminescence (ECL) solution was purchased from Nanjing KeyGen Biotech Co., Ltd. (Nanjing, China). The reagents for reverse transcription-quantitative polymerase chain reaction (RT-qPCR) analysis was obtained from Invitrogen; Thermo Fisher Scientific, Inc.).

Cell culture and treatments

The HUVECs were supplied by Sun Yat-sen University Experimental Animal Center (Guangzhou, China). The HUVECs were cultured in DMEM supplemented with 10% FBS under an atmosphere of 5% CO2 and 37°C with 95% air.

To establish a model of HG-induced HUVEC injury, the cells were cultured in DMEM (5.5 mM glucose) for 12 h prior to the administration of 40 mM glucose (final concentration) for 24 h. The glucose concentration of the control group was 5.5 mM. To investigate the protective effect of exogenous Ang-(1-7) against HG (40 mM glucose)-induced injury, the cells were seeded at a density of 1×104/ml and treated with HG in the presence or absence of Ang-(1-7) for 24 h at 37°C. In order to examine whether the STAT3/JAK2 pathway contributed to the protective effects of Ang-(1-7), and to further determine the mechanisms underlying the protective effects of Ang-(1-7), the HUVECs at a density of 1×104/ml were co-treated with HG, 2 µmol/l Ang-(1-7) and 20 µmol/l AG490 (an inhibitor of the STAT3/JAK2 pathway), and incubated at 37°C.

RNA interference

HUVECs were transfected at 70% confluency using Lipofectamine transfection reagent (Life Technologies; Thermo Fisher Scientific, Inc.), with siRNA against NLRP3 (Ribo Biotechnology, Shanghai, China) or a physiologically irrelevant negative control siRNA. The siRNA sequences used in the present study were as follows: Sense, 5′-GCU UCA GCC ACA UGA CUU UTT-3′; and antisense, 5′-AAA GUC AUG UGG CUG AAG CTT-3′. Each siRNA was dissolved in nuclease-free water to achieve a final concentration of 20 µM. A total of 5 µl siRNA (20 µM) and 5 µl Lipofectamine were added to a 500 µl buffer system. The mixtures were kept at room temperature for 30 min to form complexes, and equal amounts were added into wells of a 6-well plate. The cultures were incubated at 37°C in a 5% CO2 incubator. The medium was replaced after 12 h with DMEM without the presence of siRNA or transfection reagent. Cells were collected at 12 h for analyses.

Western blot analysis

Following the indicated treatments, the HUVECs were harvested using a cell scraper and lysed with cell lysis solution (Beyotime Institute of Biotechnology, Haimen, China), at 4°C for 30 min. The total proteins were quantified using the BCA protein assay kit. Loading buffer was added to the cytosolic extracts and boiled for 5 min. Equal quantities of supernatant from each sample (20 µg were fractionated by 10% sodium dodecyl sulphate-polyacrylamide gel electrophoresis, following which the total proteins were transferred onto polyvinylidene difluoride membranes. The membranes were blocked with 5% fat-free milk for 60 min in fresh blocking buffer containing 0.1% Tween20 in Tris-buffered saline (TBS-T) at room temperature, and incubated with either anti-p-STAT3 (1:1,000 dilution), anti-t-STAT3 (1:1,000 dilution), anti-Nox4 (1:1,000 dilution), anti-p-JAK2 (1:1,000 dilution), anti-t-JAK2 (1:1,000 dilution), anti-caspase-3 (1:1,000 dilution) and anti-eNOS (1:1,000 dilution) in freshly prepared TBS-T with 3% fat-free milk, overnight with gentle agitation at 4°C. The membranes were then washed for 15 min with TBS-T and incubated with horseradish peroxidase (HRP)-conjugated goat anti-rabbit secondary antibody (Kangchen Biotech, Inc.), at a 1:3,000 dilution, in TBS-T with 3% fat-free milk for 90 min at room temperature. The membranes were then washed three times with TBS-T for 15 min and the immunoreactive signals were visualized using an ECL detection. In order to quantify protein expression, the X-ray films were scanned and analyzed with ImageJ 1.47i software (National Institutes of Health, Bethesda, MA, USA). The experiment was repeated three times.

Measurement of cell viability

The HUVECs were seeded in 96-well plates at a concentration of 1×104/ml, and incubated at 37°C, following which a CCK-8 assay was used to assess the cell viability of HUVECs. Following the indicated treatments, 10 µl CCK-8 solution at a 1/10 dilution was added to each well and the plate was incubated for 1.5 h in the incubator. The absorbance at 450 nm was determined using a microplate reader (Molecular Devices LLC, Sunnyvale, CA, USA). The mean optical density (OD) of three wells in the indicated groups was used to calculate the percentage of cell viability according to the following formula: Cell viability (%) = (OD treatment group/OD control group) x 100%. The experiment was performed five times.

Hoechst 33258 nuclear staining for the analysis of apoptosis

Apoptotic cell death was analyzed using Hoechst 33258 staining followed by photofluorography. Firstly, the HUVECs were plated in 35 mm dishes at a density of 1×106 cells/well. Following the indicated treatments, the cells were fixed with 4% paraformaldehyde in 0.1 mol/l phosphate-buffered saline (PBS; pH 7.4) for 10 min at 4°C. The slides were washed 5 times with PBS. Following staining by incubation with 5 mg/ml Hoechst 33258 for 30 min, the cells were washed 5 times with PBS. Finally, the cells were visualized under a fluorescence microscope (Bx50-FLA; Olympus Corporation, Tokyo, Japan). Viable HUVECs exhibited a uniform blue fluorescence throughout the nucleus and normal nuclear size, whereas apoptotic HUVECs demonstrated condensed, distorted or fractured nuclei. The experiment was repeated 5 times.

Examination of intracellular reactive oxygen species (ROS) generation

Intracellular ROS generation was determined by the oxidative conversion of cell-permeable oxidation of DCF-DH to fluorescent DCF. The HUVECs were cultured on a slide with DMEM. Following the above treatments, the slides were washed twice with PBS. Subsequently, 10 µmol/l DCFH-DA solution in serum-free medium was added to the slides, and the cells were incubated at 37°C for a further 30 min. The slides were washed 5 times with PBS, and DCF fluorescence was measured over the entire field of vision using a fluorescence microscope connected to an imaging system (BX50-FLA; Olympus Corporation). The mean fluorescence intensity (MFI) from five randomly selected fields was measured using ImageJ 1.47i software and the MFI was used as an index of the level of ROS. The experiment was repeated 5 times.

Measurement of the mitochondrial membrane potential (MMP)

The MMP was determined using the fluorescent dye, JC-1, a cell-permeable cationic dye, which preferentially enters mitochondria based on the highly negative MMP. Depolarization of MMP results in a loss of MMP from the mitochondria and a decrease in green fluorescence. The HUVECs were cultured on a slide with DMEM at a density of 1×106 cells/well. Following the indicated treatments, the slides were washed 3 times with PBS; and the cells were incubated with 1 mg/l JC-1 at 37°C for 30 min in the incubator, washed briefly 3 times with PBS, and air-dried. The fluorescence was measured over the entire field of vision using a fluorescent microscope connected to an imaging system (BX50-FLA). The MFI of JC-1 from three randomly selected fields was analyzed using ImageJ 1.47i software, and the MFI was measured as an index of the levels of MMP. The experiment was repeated 3 times.

Measurement of superoxide dismutase (SOD) activity

SOD activity was analyzed using an SOD assay kit. Following the indicated treatments, the HUVECs were washed using PBS and lysed in ice-cold 0.1 M Tris/HCl (pH 7.4) containing 0.5% Triton, 5 mmol/lβ-mercaptoethanol and 0.1 mg/ml phenylmethylsulfonyl fluoride. The lysates were clarified by centrifugation at 14, 000 × g at 4°C for 5 min and cell debris was discarded. SOD activity was detected using a commercial SOD Assay kit according to the manufacturer's protocol (Sigma-Aldrich; Merck KGaA). The absorbance values at 450 nm were measured using a microplate reader. The experiment was repeated 3 times.

Enzyme-linked immunosorbent assay (ELISA) for the detection of interleukin (IL)-1β, IL-10, IL-12 and tumor necrosis factor (TNF)-α in culture supernatant

The HUVECs were seeded at 1×104 cells/well and cultured in 96-well plates. Following the indicated treatments, the levels of IL-1β, IL-10, IL-12 and TNF-α in culture media were analyzed using ELISA according to the manufacturer's protocol. The experiments were repeated 5 times.

RT-qPCR analysis

Total cellular RNA was extracted from cell cultures using TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's protocol. The extracted RNAs were DNase-treated with RQ1 RNase-Free DNase (Promega Corporation, Madison, WI, USA). First strand cDNA was prepared using Moloney Murine Leukemia Virus Reverse Transcriptase (M-MLV RT; cat. no. 1701; Promega Corporation). The RT-qPCR analysis was performed using the Rotor-Gene™ SYBR Green PCR kit (Qiagen GmbH, Dusseldorf, Germany) and the QuantiNova SYBR Green PCR kit (Qiagen GmbH) on a Rotor-Gene 6000 Rotary Analyzer (Qiagen GmbH), and determined using Rotor-Gene 6000 software version 2.3.3 (Qiagen GmbH). For each assay, a total of 8 ng cDNA was added to a final reaction volume of 25 µl containing 1X Rotor-Gene SYBR Green PCR master mix or QuantiNova SYBR Green PCR master mix and 1 µM of each forward and reverse primer. Quantitative gene amplifications were performed using the following thermocycling conditions: Initial denaturation for 5 min at 95°C, 40 cycles of denaturation at 95°C for 5 sec, and annealing and extension at 60°C for 20 sec. The glyceraldehyde-3-phosphate dehydrogenase (GAPDH) gene was used as endogenous control (or reference gene). A non-template reaction was used as a negative control. Three replicates were performed for all analysis.

The selection of the threshold intensity was set at a fixed intensity on the log-linear phase of the amplification curve for all the samples tested. Validation experiments, which included the generation of standard curves using a series of diluted cDNA samples, were performed to ensure primer efficiency, and target and reference gene amplification compatibility. Melt curve analysis and conventional agarose gel analysis were used alongside to verify the presence of a single amplicon. An interassay calibration scheme was used to minimize loading variation and to detect possible contamination with the inclusion of duplicate reactions and 'no-template' control, respectively, in each qPCR assay. Relative expression levels of the gene of interest were calculated using 2−ΔΔCq method (27). All samples were normalized to GAPDH as the endogenous control (28).

The primers used were as follows: STAT3, forward 5′-CTT TGA GAC CGA GGT GTA TCA CC-3′; and reverse 5′-GGT CAG CAT GTT GTA CCA CAG G-3′; JAK2, forward 5′-CCG GAA TTC GCT TTG AGT CGG TTT CTC CGG TTC C-3′; and reverse 5′-TGC TCT AGA CCT CAT GCA GTC GCT GAA TAA GTC C-3′. GAPDH, forward, 5′CCA CCC ATG GCA AAT TCC ATG GCA3′; and reverse 5′TCT AGA CGG CAG GTC AGG TCC ACC3′.

Statistical analysis

All data are presented as the mean ± standard error of the mean. Differences between groups were analyzed using one-way analysis of variance with SPSS 13.0 (SPSS, Inc., Chicago, IL, USA) software, and followed by an LSD post hoc comparison test. P<0.05 was considered to indicate a statistically significant difference.

Results

Ang-(1-7) and AG490 attenuate HG-induced decreased cell viability in HUVECs

The HUVECs were treated with different concentrations of glucose (10, 20, 30, 40, 50 and 60 mmol/l glucose) for 24 h, and it was found that glucose induced cell cytotoxicity (Fig. 1A). A glucose concentration of 40 mmol/l was considered a suitable concentration for use in the following experiments. To examine the cytoprotective effect of Ang-(1-7) against HG-induced cytotoxicity in HUVECs, a dose-response study with varying doses of Ang-(1-7) (0.5, 1, 2, 4, 6, 8 and 10 µM) was performed to calculate the cardioprotection dose of Ang-(1-7). As shown in Fig. 1B, exposure of the HUVECs to 40 mM glucose (HG) for 24 h induced cytotoxicity, as indicated by the decrease in cell viability. However, the cytotoxic effect of HG on HUVECs was markedly inhibited by pre-treatment of the cells with Ang-(1-7) for 30 min. The maximum inhibitory effect was observed with 2 µM Ang-(1-7). Alone, 10 µM Ang-(1-7) did not significantly alter the viability of the HUVECs. For this reason, the HUVECs were pre-treated with 2 µM Ang-(1-7) for 30 min prior to exposure to HG in all subsequent experiments.

As shown in Fig. 1C, exposure of the HUVECs to HG for 24 h induced cytotoxicity, which led to a decrease in cell viability. However, this decreased cell viability was markedly repressed by pre-treatment with 2 µM Ang-(1-7) or 20 µM AG490. Alone, neither 2 µM Ang-(1-7) or 20 µM AG490 affected the viability of HUVECs.

JAK2 siRNA inhibits expression of the JAK2/STAT3 pathway and attenuates the HG-induced decrease in cell viability of HUVECs

To observe the effects of JAK2 siRNA on the expression of the JAK2/STAT3 pathway, the HUVECs were treated with JAK2 siRNA. As shown in Fig. 2, JAK2 siRNA significantly inhibited the expression levels of p-JAK2 (Fig. 2A and B) and p-STAT3 (Fig. 2C and D). In addition, JAK2 siRNA attenuated the HG-induced decrease in cell viability in the HUVECs, as shown in Fig. 2E.

HG activates the JAK2/STAT3 pathway in HUVECs

The present study also examined the effects of HG on the STAT3/JAK2 pathway, including the phosphorylation of STAT3 and JAK2. As shown in Fig. 3A–D, the effect of glucose on the HUVECs were examined. The cells were exposed to the indicated concentrations (20, 30, 40, 50 and 60 mM) of glucose for 24 h, and exposure to glucose significantly upregulated the expression levels of (Fig. 3A and C) pSTAT3, peaking at 60 mM glucose. However, the expression of t-STAT3 was not affected by the indicated concentrations of glucose. Based on these results, the effect of time on the expression of p-STAT3 was examined. As shown in Fig. 3B and D, following exposure of HUVECs to 40 mM glucose for the indicated times (3, 6, 12, 18, 24, 36 and 48 h), the expression levels of p-STAT3 were significantly upregulated, reaching a peak at 36 h, whereas the expression of t-STAT3 remained unchanged. Similarly, exposure of the cells to 40 mM glucose increased the expression levels of p-JAK2, as shown in the dose-response experiment (Fig. 3E) and time-response experiment (Fig. 3F), respectively. The mRNA expression levels of STAT3 (Fig. 3G) and JAK2 (Fig. 3H) were also markedly increased.

Ang-(1-7) downregulates HG-induced activation of the JAK2/STAT3 pathway in HUVECs

To observe effects of Ang-(1-7) on the activation of the JAK2/STAT3 pathway induced by HG, the HUVECs were pre-treated with 2 µM Ang-(1-7) for 30 min, prior to exposure to HG for 24 h. As shown in Fig. 4, exposure of the cells to 40 mM glucose significantly increased the expression levels of p-STAT3 (Fig. 4A and B) and p-JAK2 (Fig. 4A and C). However, the increased phosphorylation of the JAK2/STAT3 pathway was reduced by pre-treatment with 2 µM Ang-(1-7).

HG upregulates the expression level of caspase-3 and downregulates the expression level of eNOS in HUVECs

The present study also examined the effects of HG on the expression levels of caspase-3 and eNOS in HUVECs. As shown in Fig. 5A and E, The HUVECs were exposed to (A) the indicated concentrations (20, 30, 40, 50 and 60 mM) of glucose for 24 h, or (B) with 40 mM glucose for the indicated durations (3, 6, 12, 18, 24, 36 and 48 h). (C) Exposure to different glucose concentrations markedly increased the expression levels of caspase-3, peaking at 60 mM glucose. Based on these results, the effect of time on the expression was examined. As shown in Fig. 5D, when the HUVECs were exposed to 40 mM glucose for the indicated durations (3, 6, 12, 18, 24, 36 and 48 h), the expression levels of caspase-3 were significantly upregulated, reaching a peak at 12 and 18 h. By contrast, exposure of the cells to HG decreased the expression levels of eNOS, as shown in the dose-response experiment (Fig. 5E) and time-response experiment (Fig. 5F), respectively.

Ang-(1-7) and AG490 downregulate the increased expression of caspase-3 and upregulate the decreased expression of eNOS induced by HG in HUVECs. To observe the effects of Ang-(1-7) and AG490 on the increased expression level of caspase-3 and upregulating the decreased expression level of eNOS induced by HG, the HUVECs were pre-treated with 2 µM Ang-(1-7), as shown in Fig. 6A–C, or 20 µM AG490 for 30 min (Fig. 6D–F), prior to exposure to HG for 24 h. As shown in Fig. 6, exposure of the cells to 40 mM glucose significantly increased the expression levels of caspase-3 (Fig. 6A, C, D and F). However, the increased expression level of caspase-3 was reduced by pre-treatment with 2 µM Ang-(1-7) (Fig. 6A and B) or 20 µM AG490 (Fig. 6D and F) for 30 min prior to exposure to HG for 24 h. Secondly, the exposure of cells to 40 mM glucose significantly decreased the expression levels of eNOS (Fig. 6A, B, D and E). However, the decreased expression levels of eNOS were upregulated following pre-treatment with 2 µM Ang-(1-7) (Fig. 6A and B) or 20 µM AG490 (Fig. 6D and E) for 30 min prior to exposure to HG for 24 h.

Ang-(1-7) and AG490 suppress HG-induced apoptosis in HUVECs

As shown in Fig. 7Aa and b, exposure of HUVECs to 40 mM glucose for 24 h induced typical apoptosis, which was manifested as the nuclear condensation and fragmentation condensation of chromatin, and the shrinkage of nuclei and apoptotic bodies. However, pre-treatment of the cells with 2 µM Ang-(1-7) for 30 min prior to exposure to HG for 24 h mitigated the HG-induced increase in the number of cells undergoing apoptosis (Fig. 7Ac). In addition, pre-conditioning of the cells with 20 µM AG490 for 30 min prior to exposure to HG for 24 h also ameliorated the HG-induced apoptosis of cardiac cells (Fig. 7Ad). Alone, 2 µM Ang-(1-7) (Fig. 7Ae) or 20 µM AG490 (Fig. 7Af) did not significantly alter the number of apoptotic cells. Quantification of results is shown in Fig. 7B.

Ang-(1-7) and AG490 reduce the oxidative stress induced by HG in HUVECs

The results demonstrated that oxidative stress contributed to HG-induced HUVEC injury. As shown in Figs. 8 and 9, exposure of the HUVECs to 40 mM glucose for 24 h resulted in oxidative stress, as evidenced by an increase in the generation of ROS (Fig. 8Ab and B), a decrease in SOD activity (Fig. 9A) and increases in the expression level of Nox4 in the dose-response experiment (20, 30, 40, 50 and 60 mM glucose; Fig. 9B and C) and time-response experiment (3, 6, 12, 18, 24, 36 and 48 h; Fig. 9D and E). However, pre-treatment of the cells with 2 µM Ang-(1-7) for 30 min prior to exposure to HG for 24 h mitigated the HG-induced increase in ROS generation (Fig. 8Ac and B), increased the HG-induced decrease in SOD activity (Fig. 9A) and decreased the expression level of Nox4 (Fig. 9F and G). To determine whether the JAK2/STAT3 pathway was involved in HG-induced oxidative stress, the HUVECs were pre-treated cells with 20 µM AG490 for 30 min prior to exposure to HG for 24 h. The resulting data showed that pre-treatment of the cells with 20 µM AG490 for 30 min prior to exposure to HG for 24 h decreased the generation of ROS (Fig. 8Ad and B), upregulated SOD activity (Fig. 9A) and reduced the expression levels of Nox4 (Fig. 9F and G) in the HUVECs. Treatment with Ang-(1-7) or AG490 alone did not alter the basal level of ROS, activity of SOD or expression level of Nox4 in the HUVECs.

Ang-(1-7) and AG490 inhibit the HG-induced dissipation of MMP in HUVECs

It was shown that the exposure of HUVECs to 40 mM glucose for 24 h elicited mitochondrial damage, as manifested by the dissipation of MMP (Fig. 10Aa and b). The dissipation of MMP was reduced by pre-treatment of the cells with 2 µM Ang-(1-7) for 30 min prior to exposure to HG for 24 h (Fig. 10Ac), which demonstrated that Ang-(1-7) protected the HUVECs against HG-induced mitochondrial damage. Similarly, pre-treatment of HUVECs with 20 µM AG490 for 30 min prior to exposure to HG for 24 h attenuated the HG-induced dissipation of MMP (Fig. 10Ad). The quantitative results are shown in Fig. 10B.

Ang-(1-7) and AG490 suppress the HG-induced increased production of pro-inflammatory cytokines in HUVECs

As shown in Fig. 11, the levels of IL-1β (Fig. 11A), IL-10 (Fig. 11B), IL-12 (Fig. 11C) and TNF-α (Fig. 11D) were markedly increased in the HG-induced HUVECs, compared with those in the control group (P<0.01). However, these increased levels of IL-1β, IL-10, IL-12 and TNF-α were significantly suppressed by pre-treatment of the HUVECs with 2 µM Ang-(1-7) for 30 min prior to exposure to HG for 24 h. This suggested an inhibitory effect of Ang-(1-7) on the production of pro-inflammatory cytokines, including IL-1β, IL-10, IL-12 and TNF-α, induced by HG. Similarly, pre-treatment of the HUVECs with 20 µM AG490 for 30 min prior to exposure to HG for 24 h decreased the enhanced production of IL-1β, IL-10, IL-12 and TNF-α.

Discussion

In the present study, the data revealed several novel findings indicating that the JAK2/STAT3 signaling pathway is relevant to the potential mechanisms responsible for HG-induced HUVEC injury and inflammation, and the effect of Ang-(1-7) on protecting vascular endothelium. First, in the HUVECs, the JAK2/STAT3 signaling pathway was involved in the HG-induced HUVEC injury and inflammation. Secondly, Ang-(1-7) exerted endothelial protection against HG-induced injury and inflammation. Finally, the protective effects on the vascular endothelium by Ang-(1-7) were associated with inhibition of the JAK2/STAT3 pathway.

The data obtained demonstrated that the exposure of HUVECs to HG for 24 h led to injury and inflammation, as characterized by an increase in apoptotic cells, expression levels of caspase-3 (a death effector domain), oxidative stress (demonstrated by increased ROS production), decreased activation of SOD, increased expression of Nox4 (an important component of the NADPH oxidase family), increased expression level of eNOS, decreased cell viability and dissipation of MMP, and the upregulation of secretion of inflammatory cytokines (IL-1β, ll-6, IL-12 and TNF-α). These results are consistent with those of previous studies (5,6-9,29-31) and demonstrated that the damage in HG-induced HUVEC injury and inflammation was extensive. However, the associated mechanism remains to be fully elucidated.

The JAK2/STAT3 pathway is known to mediate survival signals, which contribute to cell proliferation, differentiation, growth and apoptosis (14-16). Similarly, the effects of the JAK2/STAT3 pathway on the cardiovascular system are also important and have been widely examined. Accumulating evidence has demonstrated that the JAK2/STAT3 pathway is involved in the progress of various stimulation-induced cardiovascular complication (32-36), including apoptosis (32,33), reticulum stress (32), ROS (35,36), contractile dysfunction (36) and inflammation (37). Currently, the potential roles of the JAK2/STAT3 pathway in hyperglycemia-induced cardiovascular complication remain to be fully elucidated. Fiaschi et al identified a novel role for STAT3 as a crucial signaling molecule of collagen I production in cardiac fibroblasts induced by a diabetic environment (38). In addition, in streptozotocin (STZ)-induced diabetic rats, wortmannin (an inhibitor of PI3K) and AG490 (an inhibitor of JAK2), synergistically mitigate myocardial ischemia reperfusion injuries (39). This indicates that PI3K/Akt and JAK2/STAT3 are synergistically involved in myocardial injury in diabetes.

As the effects of the JAK2/STAT3 pathway in HG-induced HUVEC injury and inflammation remain to be fully elucidated, the present study aimed to elucidate the mechanism. Firstly, the role of HG on activation of the JAK2/STAT3 pathway in HUVECs was investigated. The results showed that exposure of the HUVECs to HG upregulated the expression levels of p-JAK2 and p-STAT3, indicating that HG activated the JAK2/STAT3 pathway in HUVECs. Secondly, the associated roles of JAK2/STAT3 pathway activation were examined in HG-stimulated injury. The data indicated that co-treatment of the HUVECs with HG and AG490, an inhibitor of the JAK2 pathway, significantly alleviated HG-induced injuries, including apoptosis, cytotoxicity, mitochondrial damage and oxidative stress, as evidenced by a decrease in the number of apoptotic cells, decreased expression levels of caspase-3 and Nox4, and an increase in the activation of SOD, cell viability, expression of eNOS and dissipation of MMP. These results suggested that JAK2/STAT3 activation was involved in HG-stimulated injury in HUVECs. In addition, as it has been demonstrated that hyperglycemia is involved in vascular endothelium inflammation in vitro and in vivo (29,40-43), the present study further examined the role of JAK2/STAT3 activation on the HG-induced inflammatory response in HUVECs. Similar to the results of previous studies (29,40-43), it was found that exposure of the HUVECs to HG promoted inflammatory responses, as indicated by the upregulated production of IL-1β, IL-6, IL-12 and TNF-α. However, the increased production of IL-1β, IL-6, IL-12 and TNF-α was decreased by AG490. This suggested that the JAK2/STAT3 pathway was involved in the HG-induced production of pro-inflammatory factors (IL-1β, IL-6, IL-12 and TNF-α). The above data provide definitive and novel evidence that activation of the JAK2/STAT3 pathway contributed to HG-induced injury and inflammation in HUVECs.

An important finding of the present study relates to the various endothelial protective effects of Ang-(1-7) on HG-induced injury and inflammation in HUVECs. Firstly, it was found that Ang-(1-7) markedly alleviated HG-induced cytotoxicity, as characterized by increased cell viability. These results are supported by previous findings that toxicity induced by various factors, including lipopolysaccharide, long-term hypoxia and STZ-induced diabetes is alleviated by Ang-(1-7) (44-46). Secondly, Ang-(1-7) can exert anti-apoptotic effects against HG-induced apoptosis in HUVECs. According to previous studies, the anti-apoptotic effect of Ang-(1-7) is widely recognized, and is associated with various systems and organs, including the endocrine system (46), reproductive system (47), circulatory system (48), respiratory system (49), urinary system (50) and motor system (51). These data demonstrate that Ang-(1-7) protects cells against the injury induced by various factors by exerting anti-apoptotic effects. In the present study, data indicated that Ang-(1-7) protected the HUVECs against HG-induced apoptosis, as indicated by decreases in cell apoptosis and the expression of caspase-3. Ang-(1-7) is also involved in the oxidative stress induced by HG in HUVECs. In the circulatory system, Ang-(1-7) exerts cardiovascular protective effects by attenuating oxidative stress in cardiac (53), cardiomyocyte autophagy (52), hypertension (53) and vascular remodeling (54). Additionally, AVE 0991, an analog of Ang-(1-7), attenuates cardiac hypertrophy via inhibiting oxidative stress (55). Ang-(1-7) also mediates the endothelial protection of signaling by reducing the oxidative stress induced by diabetes (56). Consistent with these previous findings, the results of the present study demonstrated that Ang-(1-7) protected HUVECs against HG-induced apoptosis by reducing oxidative stress, as characterized by a decrease in the production of ROS and expression level of Nox4, an important component of the NADPH oxidase family, and an increase in the activation of SOD. The findings of the present study also revealed that Ang-(1-7) had mitochondrial protective effects against HG-induced mitochondrial injury (a loss of MMP), which was comparable with a previous study showing that Ang-(1-7) alleviated the loss of MMP during H2O2-induced in pancreatic β cells (57). The endothelial protective effect of Ang-(1-7) is also associated with its anti-inflammatory effect. In previous studies, Ang-(1-7) exerted anti-inflammatory effects in cardiomyocytes (58) and pulmonary microvascular endothelial cells (59) of the circulatory system. Similarly, the anti-inflammatory effect of Ang-(1-7) has been shown to contribute to endothelial protection in endothelial cells (60,61). However, whether Ang-(1-7) inhibits HG-induced inflammation in HUVECs remains to be elucidated. In the present study, Ang-(1-7) significantly inhibited the HG-induced expression of inflammatory factors (IL-1β, IL-6, IL-12 and TNF-α). This indicates that the endothelial protective effect of Ang-(1-7) was involved in its anti-inflammatory effect. It was also found that Ang-(1-7) ameliorated the expression level of eNOS induced by HG in the HUVECs. Exposure of the HUVECs to HG for 24 h upregulated the expression level of eNOS. These data were supported by previous studies (62,63). Co-treatment of the HUVECs with HG and Ang-(1-7) considerably elevated the expression level of eNOS. However, the association between Ang-(1-7) and eNOS remains to be elucidated and requires further investigation in vitro and in vivo.

Another important finding of the present study involves the effects of inhibition of the JAK2/STAT3 pathway on the endothelial protective effects of exogenous Ang-(1-7) against HG-induced multifarious endothelial cell injury and inflammation. Several studies have reported that Ang-(1-7) exerts endothelial protective effects (44,47,55,58,59). The present study examined whether Ang-(1-7) protected HUVECs against HG-induced injury and inflammation by suppressing the JAK2/STAT3 pathway. The results demonstrated that exogenous Ang-(1-7) widely antagonized JAK2/STAT3 pathway activation and inflammatory factors (IL-1β, IL-6, IL-12 and TNF-α). In addition, similar to the inhibitory effects of AG490, an inhibitor of the JAK2 pathway, as indicated above, co-treatment of the HUVECs with HG and Ang-(1-7) mitigated HG-induced endothelial cell injury and inflammation. These results showed that the inhibitory effect of the JAK2/STAT3 pathway may be a crucial mechanism responsible for the endothelial protective effects of exogenous Ang-(1-7) against HG-induced endothelial cell injury and inflammation. Similarly, previous evidence demonstrates that exogenous Ang-(1-7) reduced Ang II-induced (25) or monocrotaline-induced (64) cell injury through inhibition of the JAK2/STAT3 pathway. Previous findings have also indicated that Ang-(1-7) exerts cardioprotective protection against HG-induced injury (65). These studies support the results of the present study.

In conclusion, the present study provided novel evidence that the JAK2/STAT3 pathway contributed to HG-induced endothelial cell injury and inflammation, and that exogenous Ang-(1-7) exerted endothelial protection against HG-induced endothelial cell injury and inflammation via the inhibitory effect on the JAK2/STAT3 pathways. These findings may assist in the development of novel therapeutic methods for the prevention and treatment of hyperglycemia-associated endothelial cell injury and inflammation.

Acknowledgments

The present study was supported by grants from the Guangdong Medical Research Foundation (grant no. A2012172), the Technology Planning Project of Huangpu District (grant no. 201544-01), the Science and Technology Planning Project of Guangdong Province, China (grant nos. 2012B031800358, 2012B031800365 and 2010B08071044), Medical Scientific Research Foundation of Guangdong Province (A2015287) and the Guangdong Natural Science Foundation (grant no. S2011010004381).

Notes

[1] Competing interests

The authors declare that they have no competing interests.

References

1 

Shaw JE, Sicree RA and Zimmet PZ: Global estimates of the prevalence of diabetes for 2010 and 2030. Diabetes Res Clin Pract. 87:4–14. 2010. View Article : Google Scholar

2 

Guariguata L, Whiting DR, Hambleton I, Beagley J, Linnenkamp U and Shaw JE: Global estimates of diabetes prevalence for 2013 and projections for 2035. Diabetes Res Clin Pract. 103:137–149. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Bachmann KN and Wang TJ: Biomarkers of cardiovascular disease: Contributions to risk prediction in individuals with diabetes. Diabetologia. Sep 28–2017.Epub ahead of print. View Article : Google Scholar : PubMed/NCBI

4 

Capellini VK, Celotto AC, Baldo CF, Olivon VC, Viaro F, Rodrigues AJ and Evora PR: Diabetes and vascular disease: Basic concepts of nitric oxide physiology, endothelial dysfunction, oxidative stress and therapeutic possibilities. Curr Vasc Pharmacol. 8:526–544. 2010. View Article : Google Scholar

5 

Taguchi K, Sakata K, Ohashi W, Imaizumi T, Imura J and Hattori Y: Tonic inhibition by G protein-coupled receptor kinase 2 of Akt/endothelial nitric-oxide synthase signaling in human vascular endothelial cells under conditions of hyperglycemia with high insulin levels. J Pharmacol Exp Ther. 349:199–208. 2014. View Article : Google Scholar : PubMed/NCBI

6 

Zhao H, Ma T, Fan B, Han C, Luo J and Kong L: Protective effect of trans-δ-viniferin against high glucose-induced oxidative stress in human umbilical vein endothelial cells through the SIRT1 pathway. Free Radic Res. 50:68–83. 2016. View Article : Google Scholar

7 

Zhang Y, Liu T, Chen Y, Dong Z, Zhang J, Sun Y, Jin B, Gao F, Guo S and Zhuang R: CD226 reduces endothelial cell glucose uptake under hyperglycemic conditions with inflammation in type 2 diabetes mellitus. Oncotarget. 7:12010–12023. 2016.PubMed/NCBI

8 

Bhatt MP, Lim YC, Hwang J, Na S, Kim YM and Ha KS: C-peptide prevents hyperglycemia-induced endothelial apoptosis through inhibition of reactive oxygen species-mediated transglutaminase 2 activation. Diabetes. 62:243–253. 2013. View Article : Google Scholar

9 

Surico D, Farruggio S, Marotta P, Raina G, Mary D, Surico N, Vacca G and Grossini E: Human chorionic gonadotropin protects vascular endothelial cells from oxidative stress by apoptosis inhibition, cell survival signalling activation and mitochondrial function protection. Cell Physiol Biochem. 36:2108–2120. 2015. View Article : Google Scholar : PubMed/NCBI

10 

Samanta A, Perazzona B, Chakraborty S, Sun X, Modi H, Bhatia R, Priebe W and Arlinghaus R: Janus kinase 2 regulates Bcr-Abl signaling in chronic myeloid leukemia. Leukemia. 25:463–472. 2011. View Article : Google Scholar

11 

Kiu H and Nicholson SE: Biology and significance of the JAK/STAT signalling pathways. Growth Factors. 30:88–106. 2012. View Article : Google Scholar : PubMed/NCBI

12 

Copf T, Goguel V, Lampin-Saint-Amaux A, Scaplehorn N and Preat T: Cytokine signaling through the JAK/STAT pathway is required for long-term memory in Drosophila. Proc Natl Acad Sci USA. 108:8059–8064. 2011. View Article : Google Scholar : PubMed/NCBI

13 

Hiroi T, Wajima T, Kaneko Y, Kiuchi Y and Shimizu S: An important role of increase intetrahydrobiopterin via H2O2-JAK2 signaling pathway in late phase of ischaemic preconditioning. Exp Physiol. 95:609–621. 2010. View Article : Google Scholar : PubMed/NCBI

14 

Qi QR and Yang ZM: Regulation and function of signal transducer and activator of transcription 3. World J Biol Chem. 5:231–239. 2014.PubMed/NCBI

15 

Corcoran RB, Contino G, Deshpande V, Tzatsos A, Conrad C, Benes CH, Levy DE, Settleman J, Engelman JA and Bardeesy N: STAT3 plays a critical role in KRAS-induced pancreatic tumorigenesis. Cancer Res. 71:5020–5029. 2011. View Article : Google Scholar : PubMed/NCBI

16 

Aittomäki S and Pesu M: Therapeutic targeting of the Jak/STAT pathway. Basic Clin Pharmacol Toxicol. 114:18–23. 2014. View Article : Google Scholar

17 

Manea SA, Manea A and Heltianu C: Inhibition of JAK/STAT signaling pathway prevents high-glucose-induced increase in endothelin-1 synthesis in human endothelial cells. Cell Tissue Res. 340:71–79. 2010. View Article : Google Scholar : PubMed/NCBI

18 

Amiri F, Venema VJ, Wang X, Ju H, Venema RC and Marrero MB: Hyperglycemia enhances angiotensin II-induced janus-activated kinase/STAT signaling in vascular smooth muscle cells. J Biol Chem. 274:32382–32386. 1999. View Article : Google Scholar : PubMed/NCBI

19 

Hao PP, Chen YG, Liu YP, Zhang MX, Yang JM, Gao F, Zhang Y and Zhang C: Association of plasma angiotensin-(1-7) level and left ventricular function in patients with type 2 diabetes mellitus. PLoS One. 8:e627882013. View Article : Google Scholar

20 

Reich HN, Oudit GY, Penninger JM, Scholey JW and Herzenberg AM: Decreased glomerular and tubular expression of ACE2 in patients with type 2 diabetes and kidney disease. Kidney Int. 74:1610–1616. 2008. View Article : Google Scholar : PubMed/NCBI

21 

Liu CX, Hu Q, Wang Y, Zhang W, Ma ZY, Feng JB, Wang R, Wang XP, Dong B, Gao F, et al: Angiotensin-converting enzyme (ACE) 2 overexpression ameliorates glomerular injury in a rat model of diabetic nephropathy: A comparison with ACE inhibition. Mol Med. 17:59–69. 2011.

22 

Benter IF, Yousif MH, Cojocel C, Al-Maghrebi M and Diz DI: Angiotensin-(1-7) prevents diabetes-induced cardiovascular dysfunction. Am J Physiol Heart Circ Physiol. 292:H666–H672. 2007. View Article : Google Scholar : PubMed/NCBI

23 

Yousif MH, Dhaunsi GS, Makki BM, Qabazard BA, Akhtar S and Benter IF: Characterization of Angiotensin-(1-7) effects on the cardiovascular system in an experimental model of type-1 diabetes. Pharmacol Res. 66:269–275. 2012. View Article : Google Scholar : PubMed/NCBI

24 

Hao PP, Yang JM, Zhang MX, Zhang K, Chen YG, Zhang C and Zhang Y: Angiotensin-(1-7) treatment mitigates right ventricular fibrosis as a distinctive feature of diabetic cardiomyopathy. Am J Physiol Heart Circ Physiol. 308:H1007–H1019. 2015. View Article : Google Scholar : PubMed/NCBI

25 

Kandalam U and Clark MA: Angiotensin II activates JAK2/STAT3 pathway and induces interleukin-6 production in cultured rat brainstem astrocytes. Regul Pept. 159:110–106. 2010. View Article : Google Scholar

26 

Mori J, Patel VB, Ramprasath T, Alrob OA, DesAulniers J, Scholey JW, Lopaschuk GD and Oudit GY: Angiotensin 1-7 mediates renoprotection against diabetic nephropathy by reducing oxidative stress, inflammation, and lipotoxicity. Am J Physiol Renal Physiol. 306:F812–F821. 2014. View Article : Google Scholar : PubMed/NCBI

27 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2−ΔΔCT method. Methods. 25:402–408. 2001. View Article : Google Scholar

28 

Huanga Z, Zhuanga X, Xieb C, Hua X, Donga X, Guoa Y, Lic S and Liao X: Exogenous hydrogen sulfide attenuates high glucose-induced cardiotoxicity by inhibiting NLRP3 inflammasome activation by suppressing TLR4/NF-κB pathway in H9c2 Cells. Cell Physiol Biochem. 40:v1578–v1590. 2016. View Article : Google Scholar

29 

Ku SK and Bae JS: Baicalin, baicalein and wogonin inhibits high glucose-induced vascular inflammation in vitro and in vivo. BMB Rep. 48:519–524. 2015. View Article : Google Scholar : PubMed/NCBI

30 

Chu P, Han G, Ahsan A, Sun Z, Liu S, Zhang Z, Sun B, Song Y, Lin Y, Peng J and Tang Z: Phosphocreatine protects endothelial cells from Methylglyoxal induced oxidative stress and apoptosis via the regulation of PI3K/Akt/eNOS and NF-κB pathway. Vascul Pharmacol. 91:26–35. 2017. View Article : Google Scholar

31 

Wang XM, Song SS, Xiao H, Gao P, Li XJ and Si LY: Fibroblast growth factor 21 protects against high glucose induced cellular damage and dysfunction of endothelial nitric-oxide synthase in endothelial cells. Cell Physiol Biochem. 34:658–671. 2014. View Article : Google Scholar : PubMed/NCBI

32 

Cui ZT, Liu JP and Wei WL: The effects of tanshinone IIA on hypoxia/reoxygenation-induced myocardial microvascular endothelial cell apoptosis in rats via the JAK2/STAT3 signaling pathway. Biomed Pharmacother. 83:1116–1126. 2016. View Article : Google Scholar : PubMed/NCBI

33 

Jiang X, Guo CX, Zeng XJ, Li HH, Chen BX and Du FH: A soluble receptor for advanced glycation end-products inhibits myocardial apoptosis induced by ischemia/reperfusion via the JAK2/STAT3 pathway. Apoptosis. 20:1033–1047. 2015. View Article : Google Scholar : PubMed/NCBI

34 

Zhao GL, Yu LM, Gao WL, Duan WX, Jiang B, Liu XD, Zhang B, Liu ZH, Zhai ME, Jin ZX, et al: Berberine protects rat heart from ischemia/reperfusion injury via activating JAK2/STAT3 signaling and attenuating endoplasmic reticulum stress. Acta Pharmacol Sin. 37:354–367. 2016. View Article : Google Scholar : PubMed/NCBI

35 

Li L, Li M, Li Y, Sun W, Wang Y, Bai S, Li H, Wu B, Yang G, Wang R, et al: Exogenous H2S contributes to recovery of isch-emic post-conditioning-induced cardioprotection by decrease of ROS level via down-regulation of NF-κB and JAK2-STAT3 pathways in the aging cardiomyocytes. Cell Biosci. 6:262016. View Article : Google Scholar

36 

Wu L, Tan JL, Wang ZH, Chen YX, Gao L, Liu JL, Shi YH, Endoh M and Yang HT: ROS generated during early reperfusion contribute to intermittent hypobaric hypoxia-afforded cardioprotection against postischemia-induced Ca2+ overload and contractile dysfunction via the JAK2/STAT3 pathway. J Mol Cell Cardiol. 81:150–161. 2015. View Article : Google Scholar : PubMed/NCBI

37 

Ni CW, Hsieh HJ, Chao YJ and Wang DL: Interleukin-6-induced JAK2/STAT3 signaling pathway in endothelial cells is suppressed by hemodynamic flow. Am J Physiol Cell Physiol. 287:C771–C780. 2004. View Article : Google Scholar : PubMed/NCBI

38 

Fiaschi T, Magherini F, Gamberi T, Lucchese G2, Faggian G2, Modesti A and Modesti PA: Hyperglycemia and angiotensin II cooperate to enhance collagen I deposition by cardiac fibroblasts through a ROS-STAT3-dependent mechanism. Biochim Biophys Acta. 1843:2603–2610. 2014. View Article : Google Scholar : PubMed/NCBI

39 

Wang T, Mao X, Li H, Qiao S, Xu A, Wang J, Lei S, Liu Z, Ng KF, Wong GT, et al: N-Acetylcysteine and allopurinol up-regulated the Jak/STAT3 and PI3K/Akt pathways via adiponectin and attenuated myocardial postischemic injury in diabetes. Free Radic Biol Med. 63:291–303. 2013. View Article : Google Scholar : PubMed/NCBI

40 

Hein TW, Xu W, Xu X and Kuo L: Acute and chronic hyperglycemia elicit JIP1/JNK-mediated endothelial vasodilator dysfunction of retinal arterioles. Invest Ophthalmol Vis Sci. 57:4333–4340. 2016. View Article : Google Scholar : PubMed/NCBI

41 

Wu N, Shen H, Liu H, Wang Y, Bai Y and Han P: Acute blood glucose fluctuation enhances rat aorta endothelial cell apoptosis, oxidative stress and pro-inflammatory cytokine expression in vivo. Cardiovasc Diabetol. 15:1092016. View Article : Google Scholar : PubMed/NCBI

42 

Pollack RM, Donath MY, LeRoith D and Leibowitz G: Anti-inflammatory agents in the treatment of diabetes and its vascular complications. Diabetes Care. 39(Suppl 2): S244–S252. 2016. View Article : Google Scholar : PubMed/NCBI

43 

Halvorsen B, Santilli F, Scholz H, Sahraoui A, Gulseth HL, Wium C, Lattanzio S, Formoso G, Di Fulvio P, Otterdal K, et al: LIGHT/TNFSF14 is increased in patients with type 2 diabetes mellitus and promotes islet cell dysfunction and endothelial cell inflammation in vitro. Diabetologia. 59:2134–2144. 2016. View Article : Google Scholar : PubMed/NCBI

44 

Li Y, Zeng Z, Li Y, Huang W, Zhou M, Zhang X and Jiang W: Angiotensin-converting enzyme inhibition attenuates lipopoly-saccharide-induced lung injury by regulating the balance between angiotensin-converting enzyme and angiotensin-converting enzyme 2 and inhibiting mitogen-activated protein kinase activation. Shock. 43:395–404. 2015. View Article : Google Scholar : PubMed/NCBI

45 

Gopallawa I and Uhal BD: Angiotensin-(1-7)/mas inhibits apoptosis in alveolar epithelial cells through upregulation of MAP kinase phosphatase-2. Am J Physiol Lung Cell Mol Physiol. 310:L240–L248. 2016. View Article : Google Scholar

46 

He J, Yang Z, Yang H, Wang L, Wu H, Fan Y, Wang W, Fan X and Li X: Regulation of insulin sensitivity, insulin production, and pancreatic β cell survival by angiotensin-(1-7) in a rat model of streptozotocin-induced diabetes mellitus. Peptides. 64:49–54. 2015. View Article : Google Scholar : PubMed/NCBI

47 

Al-Maghrebi M and Renno WM: The ACE/Angiotensin (1-7)/mas axis protects against testicular ischemia reperfusion injury. Urology. 94:312.e1–8. 2016. View Article : Google Scholar

48 

Yang HY, Bian YF, Zhang HP, Gao F, Xiao CS, Liang B, Li J, Zhang NN and Yang ZM: Angiotensin-(1-7) treatment ameliorates angiotensin II-induced apoptosis of human umbilical vein endothelial cells. Clin Exp Pharmacol Physiol. 39:1004–1010. 2012. View Article : Google Scholar : PubMed/NCBI

49 

Ma X, Xu D, Ai Y, Zhao S, Zhang L, Ming G and Liu Z: Angiotensin-(1-7)/mas signaling inhibits lipopolysaccharide-induced ADAM17 shedding activity andapoptosis in alveolar epithelial cells. Pharmacology. 97:63–71. 2016. View Article : Google Scholar

50 

Kim CS, Kim IJ, Bae EH, Ma SK, Lee J and Kim SW: Angiotensin-(1-7) attenuates kidney injury due to obstructive nephropathy in rats. PLoS One. 10:e01426642015. View Article : Google Scholar : PubMed/NCBI

51 

Meneses C, Morales MG, Abrigo J, Simon F, Brandan E and Cabello-Verrugio C: The angiotensin-(1-7)/Mas axis reduces myonuclear apoptosis during recovery from angiotensin II-induced skeletal muscle atrophy in mice. Pflugers Arch. 46:1975–1984. 2015. View Article : Google Scholar

52 

Lin L, Liu X, Xu J, Weng L, Ren J, Ge J and Zou Y: Mas receptor mediates cardioprotection of angiotensin-(1-7) against Angiotensin II-induced cardiomyocyte autophagy and cardiac remodelling through inhibition of oxidative stress. J Cell Mol Med. 20:48–57. 2016. View Article : Google Scholar

53 

Shi Y, Lo CS, Padda R, Abdo S, Chenier I, Filep JG, Ingelfinger JR, Zhang SL and Chan JS: Angiotensin-(1-7) prevents systemic hypertension, attenuates oxidative stress and tubulointerstitial fibrosis, and normalizes renal angiotensin-converting enzyme 2 and Mas receptor expression in diabetic mice. Clin Sci. 128:649–663. 2015. View Article : Google Scholar

54 

McKinney CA, Fattah C, Loughrey CM, Milligan G and Nicklin SA: Angiotensin-(1-7) and angiotensin-(1-9): Function in cardiac and vascular remodelling. Clin Sci. 126:815–827. 2014. View Article : Google Scholar : PubMed/NCBI

55 

Ma Y, Huang H, Jiang J, Wu L, Lin C, Tang A, Dai G, He J and Chen Y: AVE 0991 attenuates cardiac hypertrophy through reducing oxidative stress. Biochem Biophys Res Commun. 474:621–625. 2016. View Article : Google Scholar

56 

Zhang Y, Liu J, Luo JY, Tian XY, Cheang WS, Xu J, Lau CW, Wang L, Wong WT, Wong CM, et al: Upregulation of angiotensin (1-7)-mediated signaling preserves endothelial function through reducing oxidative stress in diabetes. Antioxid Redox Signal. 23:880–892. 2015. View Article : Google Scholar : PubMed/NCBI

57 

Zhang F, Liu C, Wang L, Cao X, Wang YY and Yang JK: Antioxidant effect of angiotensin (1-7) in the protection of pancreatic β cell function. Mol Med Rep. 14:1963–1969. 2016. View Article : Google Scholar : PubMed/NCBI

58 

Papinska AM, Mordwinkin NM, Meeks CJ, Jadhav SS1 and Rodgers KE: Angiotensin-(1-7) administration benefits cardiac, renal and progenitor cell function in db/db mice. Br J Pharmacol. Jun 15–2015.Epub ahead of print. View Article : Google Scholar : PubMed/NCBI

59 

Li Y, Cao Y, Zeng Z, Liang M, Xue Y, Xi C, Zhou M and Jiang W: Angiotensin-converting enzyme 2/angiotensin-(1-7)/Mas axis prevents lipopolysaccharide-induced apoptosis of pulmonary microvascular endothelial cells by inhibiting JNK/NF-κB pathways. Sci Rep. 5:82092015. View Article : Google Scholar

60 

Zhang YH, Zhang YH, Dong XF, Hao QQ, Zhou XM, Yu QT, Li SY, Chen X, Tengbeh AF, Dong B and Zhang Y: ACE2 and Ang-(1-7) protect endothelial cell function and prevent early atherosclerosis by inhibiting inflammatory response. Inflamm Res. 64:253–260. 2015. View Article : Google Scholar : PubMed/NCBI

61 

Wang L, Hu X, Zhang W and Tian F: Angiotensin (1-7) ameliorates angiotensin II-induced inflammation by inhibiting LOX-1 expression. Inflamm Res. 62:219–228. 2013. View Article : Google Scholar

62 

Yu JW, Deng YP, Han X, Ren GF, Cai J and Jiang GJ: Metformin improves the angiogenic functions of endothelial progenitor cells via activating AMPK/eNOS pathway in diabetic mice. Cardiovasc Diabetol. 15:882016. View Article : Google Scholar : PubMed/NCBI

63 

Bretón-Romero R, Feng B, Holbrook M, Farb MG, Fetterman JL, Linder EA, Berk BD, Masaki N, Weisbrod RM, Inagaki E, et al: Endothelial dysfunction in human diabetes is mediated by Wnt5a-JNK signaling. Arterioscler Thromb Vasc Biol. 36:561–569. 2016. View Article : Google Scholar : PubMed/NCBI

64 

Haga S, Tsuchiya H, Hirai T, Hamano T, Mimori A and Ishizaka Y: A novel ACE2 activator reduces monocrotaline-induced pulmonary hypertension by suppressing the JAK/STAT and TGF-β cascades with restored caveolin-1 expression. Exp Lung Res. 41:21–31. 2015. View Article : Google Scholar

65 

Lei Y, Xu Q, Zeng B, Zhang W, Zhen Y, Zhai Y, Cheng F, Mei W, Zheng D, Feng J, et al: Angiotensin-(1-7) protects cardiomyo-cytes against high glucose-induced injuries through inhibiting reactive oxygen species-activated leptin-p38 mitogen-activated protein kinase/extracellular signal-regulated protein kinase 1/2 pathways, but not the leptin-c-Jun N-terminal kinase pathway in vitro. J Diabetes Investig. 8:434–445. 2017. View Article : Google Scholar :

Related Articles

Journal Cover

May-2018
Volume 41 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
Chen J, Zhang W, Xu Q, Zhang J, Chen W, Xu Z, Li C, Wang Z, Zhang Y, Zhen Y, Zhen Y, et al: Ang-(1-7) protects HUVECs from high glucose-induced injury and inflammation via inhibition of the JAK2/STAT3 pathway. Int J Mol Med 41: 2865-2878, 2018
APA
Chen, J., Zhang, W., Xu, Q., Zhang, J., Chen, W., Xu, Z. ... Chen, J. (2018). Ang-(1-7) protects HUVECs from high glucose-induced injury and inflammation via inhibition of the JAK2/STAT3 pathway. International Journal of Molecular Medicine, 41, 2865-2878. https://doi.org/10.3892/ijmm.2018.3507
MLA
Chen, J., Zhang, W., Xu, Q., Zhang, J., Chen, W., Xu, Z., Li, C., Wang, Z., Zhang, Y., Zhen, Y., Feng, J., Chen, J., Chen, J."Ang-(1-7) protects HUVECs from high glucose-induced injury and inflammation via inhibition of the JAK2/STAT3 pathway". International Journal of Molecular Medicine 41.5 (2018): 2865-2878.
Chicago
Chen, J., Zhang, W., Xu, Q., Zhang, J., Chen, W., Xu, Z., Li, C., Wang, Z., Zhang, Y., Zhen, Y., Feng, J., Chen, J., Chen, J."Ang-(1-7) protects HUVECs from high glucose-induced injury and inflammation via inhibition of the JAK2/STAT3 pathway". International Journal of Molecular Medicine 41, no. 5 (2018): 2865-2878. https://doi.org/10.3892/ijmm.2018.3507