Quercetin protects against high glucose-induced damage in bone marrow-derived endothelial progenitor cells

  • Authors:
    • Li-Rong Zhao
    • Yu-Jun Du
    • Lei Chen
    • Zhi-Gang Liu
    • Yue-Hai Pan
    • Jian-Feng Liu
    • Bin Liu
  • View Affiliations

  • Published online on: July 14, 2014     https://doi.org/10.3892/ijmm.2014.1852
  • Pages: 1025-1031
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Endothelial progenitor cells (EPCs), a group of bone marrow-derived pro-angiogenic cells, contribute to vascular repair after damage. EPC dysfunction exists in diabetes and results in poor wound healing in diabetic patients with trauma or surgery. The aim of the present study was to determine the effect of quercetin, a natural flavonoid on high glucose‑induced damage in EPCs. Treatment with high glucose (40 mM) decreased cell viability and migration, and increased oxidant stress, as was evidenced by the elevated levels of reactive oxygen species (ROS), malondialdehyde (MDA) and superoxide dismutase in bone marrow-derived EPCs. Moreover, high glucose reduced the levels of endothelial nitric oxide synthase (eNOS) phosphorylation, nitric oxide (NO) production and intracellular cyclic guanosine monophosphate (cGMP). Quercetin supplement protected against high glucose‑induced impairment in cell viability, migration, oxidant stress, eNOS phosphorylation, NO production and cGMP levels. Quercetin also increased Sirt1 expression in EPCs. Inhibition of Sirt1 by a chemical antagonist sirtinol abolished the protective effect of quercetin on eNOS phosphorylation, NO production and cGMP levels following high glucose stress. To the best of our knowledge, the results provide the first evidence that quercetin protects against high glucose‑induced damage by inducing Sirt1-dependent eNOS upregulation in EPCs, and suggest that quercetin is a promising therapeutic agent for diabetic patients undergoing surgery or other invasive procedures.

Introduction

Vascular endothelial cells (ECs) are a dynamic border between circulating blood and the surrounding tissue. If ECs were impaired, endothelial progenitor cells (EPCs), a group of bone marrow-derived cells identified in 1997 (1), would contribute to postnatal vasculogenesis (2). Generally, EPCs are mobilized from the bone marrow into peripheral blood and are home to damaged areas for neo-angiogenesis (2). EPC dysfunction is known to be a critical event of diabetes (3). Circulating EPCs are reduced in peripheral vascular complications of type 1 and 2 diabetes mellitus (45). Many factors involved in the pathophysiology of diabetes, such as hyperglycemia, lead to EPC dysfunctions. In vitro studies also demonstrated that high glucose resulted in cellular injury in EPCs by inducing oxidant stress (6), apoptosis (7) and the downregulation of endothelial nitric oxide synthase (eNOS) (8). Since EPC dysfunction may result in poor wound healing in diabetic patients with trauma or surgery, restoring the biological functions of EPCs for these patients is crucial.

Flavonoids have recently received much attention as potential preventive and therapeutic agents for diabetes (910). Quercetin, a ubiquitous flavonol abundantly found in plant products such as capers, lovage, apples, onions and grapes, possesses antioxidative and anti-inflammatory properties (11). Recently, the protective and therapeutic value of quercetin in diabetes has been recognized. Oral administration of quercetin in rats with type 1 diabetes significantly decreased incremental plasma glucose levels (12). Quercetin had beneficial effects on cell membranes altered in diabetic conditions, by restoring transmembrane potential (13). Moreover, quercetin protected against diabetes-induced exaggerated vasoconstriction, adventitial leukocyte infiltration, endothelial pyknosis and increased collagen deposition in rats (14). Thus, the ability of quercetin to protect against the deleterious effects of hyperglycemia and to improve glucose metabolism and intake must be considered as it affects treatment strategy. Notably, quercetin has been tested in a double-blind randomized controlled clinical trial (15). In women with type 2 diabetes, quercetin reduced systolic blood pressure, serum concentration of tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) (15).

Based on the fact that quercetin has significant beneficial effects for diabetes, we hypothesized that there are likely more molecular mechanisms underlying the protective effects of quercetin in diabetes. In this study, we investigated the effect of quercetin on high glucose-induced damage in EPCs. To the best of our knowledge, the results provided the first evidence that quercetin protects against high glucose-induced damage by inducing Sirt1-dependent eNOS upregulation in EPCs.

Materials and methods

Reagents

Quercetin (catalogue no: Q4951) and antibodies against phosphor-eNOS (p-eNOS), and total-eNOS were purchased from Cell Signaling Biotechnology (Danvers, MA, USA). The antibody against Sirt1 and the Sirt1 inhibitor sirtinol, was purchased from Santa Cruz Biotechnology, Inc. (Santa Cruz, CA, USA). The antibody against β-actin was purchased from Sigma (St. Louis, MO, USA). The cell viability assay [cell counting kit-8 (CCK-8)] was purchased from Dojindo Molecular Technologies, Inc. (Kumamoto, Japan). Dichlorodihydrofluorescein diacetate (DCFH-DA) was purchased from Invitrogen Life Technologies (Carslbad, CA, USA). Nitric oxide (NO) assay was purchased from Abcam (Cambridge, MA, USA). Malondialdehyde (MDA) and cyclic guanosine monophosphate (cGMP) ELISA kits were purchased from Cell Biolab, Inc. (San Diego, CA, USA).

Bone marrow cell isolation and culture

Bone marrow-derived EPCs were prepared as previously described (16). Bone marrow was harvested from the femurs and tibiae of 2-month-old male C57BL/6J mice (Changchun Institute of Biological Products, Changchun, China). The mononuclear cell fraction was isolated with Ficoll (Amersham Pharmacia Biotech, Inc., Piscataway, NJ, USA). Cells were seeded in fibronectin-coated six-well plates (Corning, Lowell, MA, USA) and cultured in Dulbecco’s modified Eagle’s medium (DMEM) supplemented with a specific mixture of vascular endothelial growth factor, fibroblast growth factor-2, insulin-like growth factor-1, epidermal growth factor, hydrocortisone, gentamicin, amphotericin-B, 5% fetal bovine serum and ascorbic acid (EGM2-MV; Lonza, Walkersville, MD, USA). The concentration of glucose in this medium was 5.5 mM. To produce a high glucose challenge in EPCs, the EPCs were cultured in high glucose (40 mM).

Cell viability assay

Viability of EPCs was evaluated using a non-radioactive CCK-8 assay as previously described (1718). High glucose incubated EPCs were treated with quercetin (20 or 100 μM) for 48 h. The control cells were cultured in normal glucose medium. After discarding the medium, the cells were incubated with 10 μl of CCK-8 solution for 3 h at 37°C. The optical density at 450 nm was analyzed in a microplate reader (Thermo Fisher Scientific Inc., Waltham, MA USA). Experiments were performed in duplicate.

Transwell migration assay

EPCs (1×105) were placed in the upper chamber of a 24-well transwell migration insert (pore size, 5 μm) with DMEM in the presence or absence of high glucose and quercetin (20 and 100 μM, respectively). The lower chamber contained migration medium (EBM-2, 0.5% BSA, 10% FBS, 100 IU/ml penicillin, and 100 μg/ml streptomycin) (19). After migration for 24 h, the non-migrating cells on the upper side of the membrane were wiped away, while the cells on the lower side of the membrane were fixed, excised, mounted with DAPI containing mounting medium and counted on a fluorescent microscope (IX71; Olympus, Tokyo, Japan).

Reactive oxygen species (ROS) assay

Intracellular ROS production was evaluated by incubating the cells with 30 μM DCFH-DA at 37°C for 1 h. The fluorescence was revealed by fluorescence microscopy, using the same exposure conditions in each experiment (20). The cells were counted by two blinded readers obtaining similar results. The DCFH-DA fluorescence was calculated using Image Pro Plus software (Media Cybernetics Inc., Silver Spring, MD, USA).

Superoxide dismutase and MDA activity assays

Intracellular superoxide dismutase and MDA assays were performed as previously described (2123). EPCs were treated with high glucose or high glucose plus quercetin (100 μM) for 6 h and lysed using RIPA buffer. The superoxide dismutase level and MDA concentration of each sample were detected according to the manufacturer’s instructions using a microplate reader.

Western blotting

Western blotting was performed as previously described (2425). Briefly, cells were lysed in lysis buffer (50 mM Tris, pH 7.4, 150 mM sodium chloride, 1% Nonidet P-40, 1 mM EDTA, 1 mM sodium orthovanate, 1 mM sodium fluoride, 1 mg/ml leupeptin, 1 mg/ml aprotinin, 1 mg/ml pepstatin A, and 1 mM PMSF) (26). Cell lysates were centrifuged at 10,000 × g for 10 min and the supernatants were collected. The samples were boiled for 10 min. The protein concentration was determined using a BCA assay. Equal amounts of protein were separated by 10% SDS-PAGE and transferred to nitrocellulose by standard procedures (27). Membranes were incubated overnight at 4°C with antibodies against p-eNOS (1:200), t-eNOS (1:500), Sirt1 (1:250) and β-actin (1:1,000). After incubation with the corresponding secondary antibodies (1:5,000), the membranes were washed three times and bands were detected by the enhanced chemoluminescence kit (ECL; Amersham Pharmacia Biotech, Inc.) (28).

NO production measurement

NO concentrations in EPCs were determined by detecting the concentration of nitrite, the stable product of NO. EPCs were seeded in 24-well plates at a density of 1×105 cells/well. Following treatment with glucose or glucose + quercetin or glucose + quercetin + sirtinol for 12 h, the medium was collected and NO content in the medium (NO production) was measured using a microplate procedure based on the Griess reaction by a NO assay kit. The optical densities at a wavelength of 560 nm were obtained using a microplate reader and concentrations of NO were calculated according to the calibration curve.

Intracellular cGMP measurement

EPCs were treated with glucose or glucose + quercetin or glucose + quercetin + sirtinol for 12 h. The cells were then lysed using a RIPA buffer. The protein concentration was determined by a BCA assay. The cGMP level was measured using a commercial cGMP ELISA kit according to the manufacturer’s instructions.

Statistical analysis

Statistical calculations were performed using the GraphPad Prism 5 software program (GraphPad Software, Inc., La Jolla, CA, USA). The data are expressed as the means ± SEM and compared using ANOVA with Tukey correction. Statistical significance was set at P<0.05.

Results

Quercetin protects against high glucose-induced damage in EPCs

First, we studied whether quercetin exerted protective effects on EPCs. As shown in Fig. 1A, high glucose significantly decreased the cell viability at Day 2 in culture EPCs. The two concentrations of quercetin (20 and 100 μM) attenuated this detrimental effect of high glucose. The effect of quercetin on the cell migration of EPCs was also investigated. Quercetin (20 and 100 μM) rescued the migration of EPCs impaired by high glucose (Fig. 1B). These results clearly suggested that quercetin protects against high glucose-induced damage in EPCs.

Quercetin decreases high glucose-induced oxidant stress in EPCs

The effects of quercetin on high glucose-induced damage oxidant stress in EPCs were examined. Results of the DCFH-DA assay revealed that high glucose treatment triggered fluorescence in EPCs, suggesting that the ROS level was markedly increased by high glucose (Fig. 2A). Quercetin supplement (100 μM) significantly decreased DCFH-DA fluorescence (Fig. 2A). MDA level is an index of endogenous lipid peroxidation. Notably, high glucose also enhanced the MDA level in EPCs (Fig. 2B). Quercetin successfully inhibited the increase of MDA level (Fig. 2B). Total superoxide dismutase levels, major scavengers of ROS, were also determined. High glucose treatment decreased the superoxide level in EPCs, which was partly rescued by quercetin (Fig. 2C). These results suggested that quercetin decreased high glucose-induced oxidant stress in EPCs.

Quercetin prevents high glucose-induced inhibition of eNOS phosphorylation and NO production in EPCs

NO is, not only the key endothelium-derived relaxing factor that plays pivotal roles in maintaining vascular function, but also a potent positive regulator of EPCs (2930). Thus, we studied the effects of quercetin on NO production in EPCs following high glucose stress. First, we measured eNOS expression, the major enzyme responsible for NO production in EPCs. High glucose did not alter the eNOS expression (Fig. 3A). However, high glucose decreased eNOS phosphorylation, which is critical for eNOS enzymatic activity (Fig. 3A). Quercetin partly inhibited the high glucose-induced decrease of eNOS phosphorylation in EPCs (Fig. 3A). Moreover, we determined NO production in the medium of cultured EPCs. High glucose treatment significantly reduced the NO production of EPCs (Fig. 3B), which was partly blocked by the quercetin supplement (Fig. 3B). To identify the underlying molecular mechanisms, we assayed intracellular cGMP, which is produced by soluble guanylyl cyclases in responding to NO. High glucose decreased the intracellular cGMP level, which was prevented by quercetin (Fig. 3C). These results emphasized that quercetin prevented the high glucose-induced inhibition of eNOS phosphorylation and NO production in EPCs.

Quercetin protects high glucose-induced inhibition of eNOS phosphorylation in a Sirt1-dependent manner in EPCs

Sirt1, a nicotinamide adenosine dinucleotide (NAD+)-dependent histone deacetylase, deacetylates eNOS, stimulating eNOS activity and increasing endothelial NO production (31). We studied the alteration of Sirt1 in EPCs following treatments of high glucose and quercetin. High glucose treatment decreased Sirt1 expression, whereas quercetin partly rescued the Sirt1 expression (Fig. 4). To elucidate whether this modulation on Sirt1 of quercetin is essential for the activation of eNOS, we used sirtinol, a specific inhibitor of Sirt1, to block Sirt1 deacetylase activity. As shown in Fig. 5A, the sirtinol supplement completely blocked the rescuing effect of quercetin on eNOS phosphorylation. Furthermore, sirtinol blocked the increase of NO production (Fig. 5B) and intracellular cGMP (Fig. 5C) induced by quercetin treatment following high glucose stress.

Discussion

In the present study, we have demonstrated that quercetin protects against the high glucose-induced decrease of cell viability and migration in EPCs. Further experiments showed that quercetin decreased high glucose-induced oxidant stress, as evidenced by levels of ROS, MDA and superoxide dismutase. Quercetin prevented the high glucose-induced decrease of eNOS phosphorylation, NO production and intracellular cGMP level. Inhibition of Sirt1 by sirtinol also markedly blocked the rescuing effect of quercetin on eNOS phosphorylation, NO production and the intracellular cGMP level. These data suggest that quercetin protects against high glucose-induced damage in EPCs by restoring eNOS in a Sirt1-dependent manner.

In the present study, quercetin protects against high glucose-induced cell injury in EPCs. Previously, quercetin was reported to protect against high glucose-induced apoptosis in human umbilical vein ECs via its anti-oxidant effects (32). Bournival et al (33) and Shi et al (34) demonstrated a similar phenomenon in neuronal cells respectively. However, to the best of our knowledge, there is no report on the effects of quercetin on EPCs following high-glucose stress. Accumulating evidence suggests that oxidant stress-caused EPCs dysfunction is involved in the pathogenesis of diabetes (45). The number of EPCs obtained from type 1 diabetic patients in culture was 44% lower compared with age- and sex-matched control subjects (4). Similarly, EPCs from type 2 diabetic patients were reduced by 40% compared with healthy subjects (5). Additionally, conditioned media from patient EPCs were significantly reduced in their capacity to support endothelial tube formation in comparison to control EPCs (4). High glucose was shown to impair early and late EPCs by modifying NO-mediated mechanisms (35). Besides this mechanism, modulation of oxidant stress (6), apoptosis (7) and autophagy (36) also contribute to high glucose-induced cell damage. To the best of our knowledge, this is the first study on the protective effect of quercetin in high glucose-challenged EPCs, emphasizing the potential therapeutic value of quercetin in diabetic patients with trauma or surgery.

A novel mechanism underlying the protective effect of quercetin on EPCs under high glucose was identified in this study. To the best of our knowledge, this is the first study showing that quercetin prevented high glucose-induced inhibition of eNOS phosphorylation, NO production and intracellular cGMP in EPCs in a Sirt1-dependent manner. High glucose decreased eNOS and thereby NO production (35). NO donor although not antioxidants reversed the impairments, suggesting the role of NO-related mechanisms in high glucose-caused EPC downregulation (35). Moreover, uncoupling of eNOS in glucose-treated EPCs resulted in production of eNOS-mediated O2− (37), which is detrimental to EPCs. In the present study, high glucose significantly decreased the phosphorylation of eNOS in EPCs and NO production. Of note, quercetin reversed this detrimental effect, suggesting that the improved NO signaling may account for the protective effect of quercetin in EPCs.

Of note, we found that quercetin upregulated Sirt1 in EPCs. Sirt1 is a well-known key regulator of cell defenses, metabolism, aging and cell survival (38). High glucose has been reported to downregulate the number of EPCs by reducing Sirt1 expression levels and enzyme activity (39). Certain chemicals, such as stachydrine, may ameliorate high-glucose induced endothelial cell senescence and SIRT1 downregulation (40). The results of the present study demonstrate the enhancing effect on Sirt1 of quercetin in EPCs, which is in conconrdance with previous studies reporting that quercetin increases Sirt1 expression in brain and skeletal muscle (4142). Moreover, we used sirtinol, an inhibitor of Sirt1, to investigate the effect of Sirt1 deacetylase activity inhibition on quercetin-induced eNOS upregulation and NO production. As expected, sirtinol efficiently inhibited the promoting effect of quercetin on eNOS upregulation and thereby NO production and cGMP concentration, suggesting that the beneficial effects of quercetin on EPCs is Sirt1-dependent.

In conclusion, to the best of our knowledge, we have demonstrated for the first time that quercetin protects against high glucose-induced damage by inducing Sirt1-dependent eNOS upregulation in EPCs. Thus, our results support the hypothesis that dietary supplement of quercetin may promote EPCs biological function and improve vascular repair, which is crucial for diabetic patients undergoing surgery or other invasive procedures.

Acknowledgements

This study was supported by the National Natural Science Foundation of China (no. 81272999).

References

1 

Asahara T, Murohara T, Sullivan A, et al: Isolation of putative progenitor endothelial cells for angiogenesis. Science. 275:964–967. 1997. View Article : Google Scholar : PubMed/NCBI

2 

Hristov M, Erl W and Weber PC: Endothelial progenitor cells: mobilization, differentiation, and homing. Arterioscler Thromb Vasc Biol. 23:1185–1189. 2003. View Article : Google Scholar : PubMed/NCBI

3 

Hristov M and Weber C: Endothelial progenitor cells: characterization, pathophysiology, and possible clinical relevance. J Cell Mol Med. 8:498–508. 2004. View Article : Google Scholar : PubMed/NCBI

4 

Loomans CJ, de Koning EJ, Staal FJ, et al: Endothelial progenitor cell dysfunction: a novel concept in the pathogenesis of vascular complications of type 1 diabetes. Diabetes. 53:195–199. 2004. View Article : Google Scholar : PubMed/NCBI

5 

Fadini GP, Miorin M, Facco M, et al: Circulating endothelial progenitor cells are reduced in peripheral vascular complications of type 2 diabetes mellitus. J Am Coll Cardiol. 45:1449–1457. 2005. View Article : Google Scholar : PubMed/NCBI

6 

Sorrentino SA, Bahlmann FH, Besler C, et al: Oxidant stress impairs in vivo reendothelialization capacity of endothelial progenitor cells from patients with type 2 diabetes mellitus: restoration by the peroxisome proliferator-activated receptor-gamma agonist rosiglitazone. Circulation. 116:163–173. 2007. View Article : Google Scholar

7 

Rosso A, Balsamo A, Gambino R, et al: p53 Mediates the accelerated onset of senescence of endothelial progenitor cells in diabetes. J Biol Chem. 281:4339–4347. 2006. View Article : Google Scholar : PubMed/NCBI

8 

Hamed S, Brenner B and Roguin A: Nitric oxide: a key factor behind the dysfunctionality of endothelial progenitor cells in diabetes mellitus type-2. Cardiovasc Res. 91:9–15. 2011. View Article : Google Scholar : PubMed/NCBI

9 

van Dam RM, Naidoo N and Landberg R: Dietary flavonoids and the development of type 2 diabetes and cardiovascular diseases: review of recent findings. Curr Opin Lipidol. 24:25–33. 2013.PubMed/NCBI

10 

Nicolle E, Souard F, Faure P and Boumendjel A: Flavonoids as promising lead compounds in type 2 diabetes mellitus: molecules of interest and structure-activity relationship. Curr Med Chem. 18:2661–2672. 2011. View Article : Google Scholar : PubMed/NCBI

11 

Formica JV and Regelson W: Review of the biology of Quercetin and related bioflavonoids. Food Chem Toxicol. 33:1061–1080. 1995. View Article : Google Scholar : PubMed/NCBI

12 

Kim JH, Kang MJ, Choi HN, Jeong SM, Lee YM and Kim JI: Quercetin attenuates fasting and postprandial hyperglycemia in animal models of diabetes mellitus. Nutr Res Pract. 5:107–111. 2011. View Article : Google Scholar : PubMed/NCBI

13 

Margina D, Gradinaru D, Manda G, Neagoe I and Ilie M: Membranar effects exerted in vitro by polyphenols - quercetin, epigallocatechin gallate and curcumin - on HUVEC and Jurkat cells, relevant for diabetes mellitus. Food Chem Toxicol. 61:86–93. 2013. View Article : Google Scholar

14 

Mahmoud MF, Hassan NA, El Bassossy HM and Fahmy A: Quercetin protects against diabetes-induced exaggerated vasoconstriction in rats: effect on low grade inflammation. PloS One. 8:e637842013. View Article : Google Scholar : PubMed/NCBI

15 

Zahedi M, Ghiasvand R, Feizi A, Asgari G and Darvish L: Does quercetin improve cardiovascular risk factors and inflammatory biomarkers in women with type 2 diabetes: a double-blind randomized controlled clinical trial. Int J Prev Med. 4:777–785. 2013.PubMed/NCBI

16 

Marrotte EJ, Chen DD, Hakim JS and Chen AF: Manganese superoxide dismutase expression in endothelial progenitor cells accelerates wound healing in diabetic mice. J Clin Invest. 120:4207–4219. 2010. View Article : Google Scholar : PubMed/NCBI

17 

Wang P, Xu TY, Guan YF, Su DF, Fan GR and Miao CY: Perivascular adipose tissue-derived visfatin is a vascular smooth muscle cell growth factor: role of nicotinamide mononucleotide. Cardiovasc Res. 81:370–380. 2009. View Article : Google Scholar : PubMed/NCBI

18 

Wang P, Guan YF, Du H, Zhai QW, Su DF and Miao CY: Induction of autophagy contributes to the neuroprotection of nicotinamide phosphoribosyltransferase in cerebral ischemia. Autophagy. 8:77–87. 2012. View Article : Google Scholar : PubMed/NCBI

19 

Liu BQ, Du ZX, Zong ZH, et al: BAG3-dependent noncanonical autophagy induced by proteasome inhibition in HepG2 cells. Autophagy. 9:905–916. 2013. View Article : Google Scholar : PubMed/NCBI

20 

Garg AD, Dudek AM, Ferreira GB, et al: ROS-induced autophagy in cancer cells assists in evasion from determinants of immunogenic cell death. Autophagy. 9:1292–1307. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Zhang XH, Lei H, Liu AJ, Zou YX, Shen FM and Su DF: Increased oxidative stress is responsible for severer cerebral infarction in stroke-prone spontaneously hypertensive rats. CNS Neurosci Ther. 17:590–598. 2011. View Article : Google Scholar : PubMed/NCBI

22 

Schmeisser H, Fey SB, Horowitz J, et al: Type I interferons induce autophagy in certain human cancer cell lines. Autophagy. 9:683–696. 2013. View Article : Google Scholar : PubMed/NCBI

23 

Luciani A, Villella VR, Esposito S, et al: Targeting autophagy as a novel strategy for facilitating the therapeutic action of potentiators on DeltaF508 cystic fibrosis transmembrane conductance regulator. Autophagy. 8:1657–1672. 2012. View Article : Google Scholar

24 

Wang P, Zhang RY, Song J, et al: Loss of AMP-activated protein kinase-alpha2 impairs the insulin-sensitizing effect of calorie restriction in skeletal muscle. Diabetes. 61:1051–1061. 2012. View Article : Google Scholar : PubMed/NCBI

25 

Wang P, Xu TY, Guan YF, et al: Nicotinamide phosphoribosyltransferase protects against ischemic stroke through SIRT1-dependent adenosine monophosphate-activated kinase pathway. Ann Neurol. 69:360–374. 2011. View Article : Google Scholar

26 

Oh JM, Choi EK, Carp RI and Kim YS: Oxidative stress impairs autophagic flux in prion protein-deficient hippocampal cells. Autophagy. 8:1448–1461. 2012. View Article : Google Scholar : PubMed/NCBI

27 

Zhang T, Li Y, Park KA, et al: Cucurbitacin induces autophagy through mitochondrial ROS production which counteracts to limit caspase-dependent apoptosis. Autophagy. 8:559–576. 2012. View Article : Google Scholar

28 

Song YM, Song SO, Jung YK, et al: Dimethyl sulfoxide reduces hepatocellular lipid accumulation through autophagy induction. Autophagy. 8:1085–1097. 2012. View Article : Google Scholar : PubMed/NCBI

29 

Zhao YD, Courtman DW, Deng Y, Kugathasan L, Zhang Q and Stewart DJ: Rescue of monocrotaline-induced pulmonary arterial hypertension using bone marrow-derived endothelial-like progenitor cells: efficacy of combined cell and eNOS gene therapy in established disease. Circ Res. 96:442–450. 2005. View Article : Google Scholar

30 

Duda DG, Fukumura D and Jain RK: Role of eNOS in neovascularization: NO for endothelial progenitor cells. Trends Mol Med. 10:143–145. 2004. View Article : Google Scholar : PubMed/NCBI

31 

Mattagajasingh I, Kim CS, Naqvi A, et al: SIRT1 promotes endothelium-dependent vascular relaxation by activating endothelial nitric oxide synthase. Proc Natl Acad Sci USA. 104:14855–14860. 2007. View Article : Google Scholar

32 

Chao CL, Hou YC, Chao PD, Weng CS and Ho FM: The antioxidant effects of quercetin metabolites on the prevention of high glucose-induced apoptosis of human umbilical vein endothelial cells. Br J Nutr. 101:1165–1170. 2009. View Article : Google Scholar : PubMed/NCBI

33 

Bournival J, Francoeur MA, Renaud J and Martinoli MG: Quercetin and sesamin protect neuronal PC12 cells from high-glucose-induced oxidation, nitrosative stress, and apoptosis. Rejuvenation Res. 15:322–333. 2012. View Article : Google Scholar : PubMed/NCBI

34 

Shi Y, Liang XC, Zhang H, Wu QL, Qu L and Sun Q: Quercetin protects rat dorsal root ganglion neurons against high glucose-induced injury in vitro through Nrf-2/HO-1 activation and NF-kappaB inhibition. Acta Pharmacol Sin. 34:1140–1148. 2013. View Article : Google Scholar : PubMed/NCBI

35 

Chen YH, Lin SJ, Lin FY, et al: High glucose impairs early and late endothelial progenitor cells by modifying nitric oxide-related but not oxidative stress-mediated mechanisms. Diabetes. 56:1559–1568. 2007. View Article : Google Scholar : PubMed/NCBI

36 

Kobayashi S, Xu X, Chen K and Liang Q: Suppression of autophagy is protective in high glucose-induced cardiomyocyte injury. Autophagy. 8:577–592. 2012. View Article : Google Scholar : PubMed/NCBI

37 

Thum T, Fraccarollo D, Schultheiss M, et al: Endothelial nitric oxide synthase uncoupling impairs endothelial progenitor cell mobilization and function in diabetes. Diabetes. 56:666–674. 2007. View Article : Google Scholar : PubMed/NCBI

38 

Herranz D and Serrano M: SIRT1: recent lessons from mouse models. Nat Rev Cancer. 10:819–823. 2010. View Article : Google Scholar : PubMed/NCBI

39 

Balestrieri ML, Rienzo M, Felice F, et al: High glucose downregulates endothelial progenitor cell number via SIRT1. Biochim Biophys Acta. 1784:936–945. 2008. View Article : Google Scholar : PubMed/NCBI

40 

Servillo L, D’Onofrio N, Longobardi L, et al: Stachydrine ameliorates high-glucose induced endothelial cell senescence and SIRT1 downregulation. J Cell Biochem. 114:2522–2530. 2013. View Article : Google Scholar : PubMed/NCBI

41 

Davis JM, Murphy EA, Carmichael MD and Davis B: Quercetin increases brain and muscle mitochondrial biogenesis and exercise tolerance. Am J Physiol Regul Integr Comp Physiol. 296:R1071–R1077. 2009. View Article : Google Scholar : PubMed/NCBI

42 

Casuso RA, Martinez-Lopez EJ, Nordsborg NB, et al: Oral quercetin supplementation hampers skeletal muscle adaptations in response to exercise training. Scand J Med Sci Sports. Oct 14–2013.(E-pub ahead of print).

Related Articles

Journal Cover

October 2014
Volume 34 Issue 4

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
Zhao L, Du Y, Chen L, Liu Z, Pan Y, Liu J and Liu B: Quercetin protects against high glucose-induced damage in bone marrow-derived endothelial progenitor cells. Int J Mol Med 34: 1025-1031, 2014
APA
Zhao, L., Du, Y., Chen, L., Liu, Z., Pan, Y., Liu, J., & Liu, B. (2014). Quercetin protects against high glucose-induced damage in bone marrow-derived endothelial progenitor cells. International Journal of Molecular Medicine, 34, 1025-1031. https://doi.org/10.3892/ijmm.2014.1852
MLA
Zhao, L., Du, Y., Chen, L., Liu, Z., Pan, Y., Liu, J., Liu, B."Quercetin protects against high glucose-induced damage in bone marrow-derived endothelial progenitor cells". International Journal of Molecular Medicine 34.4 (2014): 1025-1031.
Chicago
Zhao, L., Du, Y., Chen, L., Liu, Z., Pan, Y., Liu, J., Liu, B."Quercetin protects against high glucose-induced damage in bone marrow-derived endothelial progenitor cells". International Journal of Molecular Medicine 34, no. 4 (2014): 1025-1031. https://doi.org/10.3892/ijmm.2014.1852