Xiaomei Ren's research while affiliated with Southeast University (China) and other places

What is this page?


This page lists the scientific contributions of an author, who either does not have a ResearchGate profile, or has not yet added these contributions to their profile.

It was automatically created by ResearchGate to create a record of this author's body of work. We create such pages to advance our goal of creating and maintaining the most comprehensive scientific repository possible. In doing so, we process publicly available (personal) data relating to the author as a member of the scientific community.

If you're a ResearchGate member, you can follow this page to keep up with this author's work.

If you are this author, and you don't want us to display this page anymore, please let us know.

Publications (4)


Correlations between plasma level of AGE-p and FMD in type 2 diabetic patients with or without coronary artery disease. AGE-p: advanced glycation end product peptides (U/ml); FMD: flow-mediated vasodilatation (%)
Effects of AGEs on eNOS mRNA in HCAECs. HCAECs were cultured with different concentrations of AGEs for different periods of time. The mRNA levels of eNOS and glyceraldehyde-3-phosphatede-hydrogenase (GAPDH) were determined by real-time PCR analysis. a Concentration-dependent study. Cells were treated with different concentrations of AGEs (50, 100, or 200 μg/ml) for 24 h. b Time-dependent study. Cells were treated with AGEs (100 μg/ml) for different times (12, 24 and 48 h). c Effect of anti-RAGE antiboday. Cells were treated with 100 μg/ml AGEs and different concentrations of anti-RAGE antiboday for 30 min and followed with AGEs treatment for 24 h. Isotype IgG was used for a negative control. d eNOS mRNA stability. Cells were treated with 5 μg/ml actinomycin D in the presence or absence of AGEs (100 μg/ml) for indicated time points (0, 2, 4, 8, or 16 h), and eNOS mRNA levels were determined by real-time PCR. *P < 0.05, compare with control, #P < 0.05, compare with AGEs treatment, n = 3 experiments. Data are means and SE of multiple experiments (n)
Effects of AGEs on eNOS protein levels and NOS activity in HCAECs. a Western blot analysis. Cells were treated with 50, 100 or 200 μg/ml AGEs for 24 h. Representative bands of eNOS and β-actin staining and quantitation of band density ratios (eNOS and β-actin) Full-length blots are presented. *P < 0.05 compare with control, n = 3 experiments. Data are means and SE of multiple experiments (n). b NOS activity. Cells were treated withs AGEs for 24 h. The NOS activity was determined by a commercial eNOS fluorimetric assay kit. c Cellular NO levels. Cells were treated with AGEs (100 μg/l) for 24 h and cellular NO levels were determined by DAF-FM DA (10 μM) staining and flow cytometric analysis. d Nitric oxide (NO) levels in the supernatant of cell culture (Griess assay). Cells were treated with AGEs (100 μg/ml) in the presence or absence of LY-83,583 (3 μM) or l-NAME (100 μM) for 24 h. Basal and bradykinin-stimulated levels of NO-derived nitrite in the culture supernatant were detected. *P < 0.05, compare with control, n = 3 experiments. Data are means and SE of multiple experiments (n)
Effects of AGEs on oxidative stress in HCAECs. a O2⁻ levels. Cells were treated with 50, 100 or 200 μg/ml of AGEs for 24 h, and intracellular O2⁻ levels were determined by DHE (5 μM) staining and flow cytometric analysis. b ROS production (glutathione assay). HCAECs were treated with AGEs (100 μg/ml) and/or other molecules for 24 h. RLU, relative light units; TFA, thenoyltrifluoroacetone. c Mitochondrial membrane potential (JC-1 staining and flow cytometry). Cells were treated with AGEs for 24 h, and mitochondrial membrane potential was determined by JC-1 staining and flow cytometric analysis. d ATP content. Cells were treated with 100 μg/ml AGEs for 24 h, and ATP content was determined by an ATPLite kit. *P < 0.05, compare with control. e Effect of mitochondrial complexII inhibitor TTFA on eNOS mRNA levels. HCAECs were treated with AGEs and/or TFA (10 μM) for 24 h, and eNOS mRNA levels were determined by real-time PCR analysis. F: Effect of mitochondrial complexII inhibitor TTFA on eNOS protein levels. HCAECs were treated with AGEs and/or TTFA (10 μM) for 24 h, and eNOS protein levels were determined by Western blot. Full-length blots are presented. *P < 0.05, compare with control, #P < 0.05, compare with AGEs treatment, n = 3. Data are means and SE of multiple experiments (n)
Effects of AGEs, SeMet and MnTBAP on activities of NOX, CAT, and SOD and eNOS mRNA levels in HCAECs. a Effect of SeMet on NADPH oxidase (NOX) activity. Cells were treated with AGEs (100 μg/ml) for 24 h, and NOX activities were determined by lucigenin-enhanced chemiluminescence with the presence of its substrate-NADPH. O2⁻ scavenger Tiron or flavoprotein inhibitor DPI was included in the assay to confirm the specificity of NOX activity. b Effect of SeMet on CAT activity. Cells were treated with AGEs (100 μg/ml) and/or antioxidant SeMet (20 μM) for 24 h and CAT activity was determined with acommercial kit. c Effect of SeMet on SOD activity. Cells were treated with AGEs (100 μg/ml) and/or antioxidant SeMet (20 μM) for 24 h and SOD activity was determined with acommercial kit. d Effect of SeMet on eNOS mRNA levels. Cells were treated with AGEs (100 μg/ml) and/or antioxidant SeMet (20 μM) for 24 h, and eNOS mRNA levels were determined by real-time PCR analysis. e Effect of MnTBAP on eNOS mRNA levels. Cells were treated with AGEs (100 μg/ml) and/or MnTBAP (2 μM) for 24 h, and eNOS mRNA levels were determined by real-time PCR analysis. f Effect of MnTBAP on eNOS protein levels. Cells were treated with AGEs (100 μg/ml) and/or MnTBAP (2 μM) for 24 h, and eNOS protein were determined by western blot. Full-length blots are presented. *P < 0.05, compare with control, #P < 0.05, compare with AGEs treatment, n = 3. Data are means and SE of multiple experiments (n)

+1

Advanced glycation end-products decreases expression of endothelial nitric oxide synthase through oxidative stress in human coronary artery endothelial cells
  • Article
  • Full-text available

April 2017

·

171 Reads

·

124 Citations

Cardiovascular Diabetology

Xiaomei Ren

·

Liqun Ren

·

·

[...]

·

Naifeng Liu

Background Advanced glycation end-products (AGEs) are elevated under diabetic conditions and associated with insulin resistance, endothelial dysfunction and vascular inflammation in humans. It has been demonstrated that AGEs evoke oxidative and inflammatory reactions in endothelial cells through the interaction with a receptor for AGEs (RAGE). Here, we aimed to identify the cellular mechanisms by which AGEs exacerbate the endothelial dysfunction in human coronary artery endothelial cells (HCAECs). Methods 30 type 2 diabetic patients with or without coronary artery atherosclerosis were recruited for this study. Plasma levels of AGE peptides (AGE-p) were analyzed using flow injection assay. Endothelial function was tested by brachial artery flow-mediated vasodilatation (FMD). Further investigations were performed to determine the effects and mechanisms of AGEs on endothelial dysfunction in HCAECs. Results AGE-p was inversely associated with FMD in diabetic patients with coronary artery atherosclerosis in our study. After treated with AGEs, HCAECs showed significant reductions of eNOS mRNA and protein levels including eNOS and phospho-eNOS Ser1177, eNOS mRNA stability, eNOS enzyme activity, and cellular nitric oxide (NO) levels, whereas superoxide anion production was significantly increased. In addition, AGEs significantly decreased mitochondrial membrane potential, ATP content and catalase and superoxyde dismutase (SOD) activities, whereas it increased NADPH oxidase activity. Treatment of the cells with antioxidants SeMet, SOD mimetic MnTBAP and mitochondrial inhibitor thenoyltrifluoroacetone (TTFA) effectively blocked these effects induced by AGEs. AGEs also increased phosphorylation of the mitogen-activated protein kinases p38 and ERK1/2, whereas the specific inhibitors of p38, ERK1/2, and TTFA effectively blocked AGEs-induced reactive oxygen species production and eNOS downregulation. Conclusions AGEs cause endothelial dysfunction by a mechanism associated with decreased eNOS expression and increased oxidative stress in HCAECs through activation of p38 and ERK1/2. Electronic supplementary material The online version of this article (doi:10.1186/s12933-017-0531-9) contains supplementary material, which is available to authorized users.

Download
Share

Chronic intermittent hypoxia induces cardiac inflammation and dysfunction in a rat obstructive sleep apnea model

November 2016

·

18 Reads

·

52 Citations

Journal of Biomedical Research

Chronic intermittent hypoxia is considered to play an important role in cardiovascular pathogenesis during the development of obstructive sleep apnea (OSA). We used a well-described OSA rat model induced with simultaneous intermittent hypoxia. Male Sprague Dawley rats were individually placed into plexiglass chambers with air pressure and components were electronically controlled. The rats were exposed to intermittent hypoxia 8 hours daily for 5 weeks. The changes of cardiac structure and function were examined by ultrasound. The cardiac pathology, apoptosis, and fibrosis were analyzed by H&E staining, TUNNEL assay, and picosirius staining, respectively. The expression of inflammation and fibrosis marker genes was analyzed by quantitative real-time PCR and Western blot. Chronic intermittent hypoxia/low pressure resulted in significant increase of left ventricular internal diameters (LVIDs), end-systolic volume (ESV), end-diastolic volume (EDV), and blood lactate level and marked reduction in ejection fraction and fractional shortening. Chronic intermittent hypoxia increased TUNNEL-positive myocytes, disrupted normal arrangement of cardiac fibers, and increased Sirius stained collagen fibers. The expression levels of hypoxia induced factor (HIF)-1α, NF-kB, IL-6, and matrix metallopeptidase 2 (MMP-2) were significantly increased in the heart of rats exposed to chronic intermittent hypoxia. In conclusion, the left ventricular function was adversely affected by chronic intermittent hypoxia, which is associated with increased expression of HIF-1α and NF-kB signaling molecules and development of cardiac inflammation, apoptosis and fibrosis.


Advanced glycation end products accelerate rat vascular calcification through RAGE/oxidative stress

March 2013

·

250 Reads

·

136 Citations

BMC Cardiovascular Disorders

Background Arterial media calcification (AMC) is highly prevalent and is a major cause of morbidity, mortality, stroke and amputation in patients with diabetes mellitus (DM). Previous research suggests that advanced glycation end products (AGEs) are responsible for vascular calcification in diabetic patients. The potential link between oxidative stress and AGEs-induced vascular calcification, however, has not been examined. Methods Male Wistar rats received a high fat diet for 8 weeks followed by a single dose of streptozotocin to induce DM (DM). Calcification was induced with Vitamin D3 and nicotine (VDN). We started VDN treatment at 1 week after the initial streptozotocin injection (DM+VDN). Age-matched rats were used as controls (CON). Metabolic parameters, aortic calcium content, alkaline phosphatase (ALP) protein, malondialdehyde (MDA) content, Cu/Zn superoxide dismutase (SOD) activity, aorta receptor for advanced glycation end products (RAGE) and aorta AGEs levels were measured. In vitro, vascular smooth muscle cells (VSMCs) were cultured with AGEs in DMEM containing 10 mmol·L-1 ß -glycerophosphate (ß-GP). Calcium content and ALP activity were used to identify osteoblastic differentiation and mineralization. Western blots were used to examine protein expression of Cu/Zn SOD, NADPH oxidase Nox1 and RAGE. In addition, the intracellular reactive oxygen species (ROS) generation was evaluated using fluorescent techniques with dihydroethidine (DHE) method. Results The DM+VDN group showed a significant increase in aortic calcium content, levels of aorta AGEs, MDA content, ALP protein levels and RAGE expression, although Cu/Zn SOD activity decreased significantly. In vitro, enhanced Nox1, RAGE expression as well as the production of intracellular superoxide anions, and reduced expression of Cu/Zn SOD induced by AGEs were attenuated by the anti-RAGE antibody or a ROS inhibitor. Furthermore, the AGEs-stimulated ROS increase was also significantly inhibited by a SOD mimetic. Increased ALP activity and calcium deposition were also inhibited markedly by the ROS inhibitor and the anti-RAGE antibody. Conclusions These results suggest that AGEs enhance vascular calcification partly through a RAGE/oxidative stress pathway.


Citations (3)


... Persistent hyperglycemia can cause inadequate blood flow to the penis via penile arteries (Fig. 2). In this regard, hyperglycemia increases the deposition of advanced glycated end-products and free radicals within testicular tissues with subsequent oxidative injury and reduced nitric oxide production [65]. Also, hyperglycemia may damage endothelial cells that produce nitric oxide in the penile arteries, compromise guanylyl cyclase activity with reduced cGMP production, and inhibit syncytial relaxation of the erectile muscle [66]. ...

Reference:

Cardiometabolic Disorder and Erectile Dysfunction
Advanced glycation end-products decreases expression of endothelial nitric oxide synthase through oxidative stress in human coronary artery endothelial cells

Cardiovascular Diabetology

... Intermittent hypoxia/reoxygenation is also a strong inducer of systemic inflammatory signaling (for example, NF-KB-dependent pathways [72,73]). In a rat model of chronic intermittent hypoxia/reoxygenation, the myocardial levels of inflammation markers (e.g., tumor necrosis factor-α and interleukin-6) correlated with myocardial hypertrophy. ...

Chronic intermittent hypoxia induces cardiac inflammation and dysfunction in a rat obstructive sleep apnea model
  • Citing Article
  • November 2016

Journal of Biomedical Research

... An increasing body of evidence suggests that AGE-RAGE enables bidirectional regulation of osteogenic differentiation. The AGE-RAGE signaling axis promotes vascular calcification but inhibits bone mineralization by targeting multiple pro-osteogenic signaling pathways 42,43 . A similar meniscal bone paradox may exist because meniscal calcification and osteoporosis are commonly observed in elderly individuals 44 . ...

Advanced glycation end products accelerate rat vascular calcification through RAGE/oxidative stress

BMC Cardiovascular Disorders