Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Aldolase B Knockdown Prevents High Glucose-Induced Methylglyoxal Overproduction and Cellular Dysfunction in Endothelial Cells

  • Jianghai Liu,

    Affiliation Department of Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada

  • Timothy Chun-Ping Mak,

    Affiliation Department of Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada

  • Ali Banigesh,

    Affiliation Department of Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada

  • Kaushik Desai,

    Affiliation Department of Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada

  • Rui Wang,

    Affiliation Department of Biology, Lakehead University, Thunder Bay, Ontario, Canada

  • Lingyun Wu

    lwu@lakeheadu.ca

    Affiliations Department of Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada, Department of Health Sciences, Lakehead University and Thunder Bay Regional Research Institute, Thunder Bay, Ontario, Canada

Abstract

We used cultured endothelial cells as a model to examine whether up-regulation of aldolase B and enhanced methylglyoxal (MG) formation play an important role in high glucose-induced overproduction of advanced glycosylation endproducts (AGEs), oxidative stress and cellular dysfunction. High glucose (25 mM) incubation up-regulated mRNA levels of aldose reductase (an enzyme converting glucose to fructose) and aldolase B (a key enzyme that catalyzes MG formation from fructose) and enhanced MG formation in human umbilical vein endothelial cells (HUVECs) and HUVEC-derived EA. hy926 cells. High glucose-increased MG production in EA. hy926 cells was completely prevented by siRNA knockdown of aldolase B, but unaffected by siRNA knockdown of aldolase A, an enzyme responsible for MG formation during glycolysis. In addition, inhibition of cytochrome P450 2E1 or semicarbazide-sensitive amine oxidase which produces MG during the metabolism of lipid and proteins, respectively, did not alter MG production. Both high glucose (25 mM) and MG (30, 100 µM) increased the formation of N(ε)-carboxyethyl-lysine (CEL, a MG-induced AGE), oxidative stress (determined by the generation of oxidized DCF, H2O2, protein carbonyls and 8-oxo-dG), O-GlcNAc modification (product of the hexosamine pathway), membrane protein kinase C activity and nuclear translocation of NF-κB in EA. hy926 cells. However, the above metabolic and signaling alterations induced by high glucose were completely prevented by knockdown of aldolase B and partially by application of aminoguanidine (a MG scavenger) or alagebrium (an AGEs breaker). In conclusion, efficient inhibition of aldolase B can prevent high glucose-induced overproduction of MG and related cellular dysfunction in endothelial cells.

Introduction

Hyperglycemia in diabetes mellitus damages blood vessels and induces vascular complications in the retinal, renal, and cardiovascular tissues [1], [2]. Vascular endothelial cells are the early and primary targets of hyperglycemic damage in diabetes [3], [4]. Hyperglycemia-triggered endothelial dysfunction, including increased endothelial permeability and inflammation, decreased nitric oxide (NO) bioavailability and endothelium-dependent relaxation, and vascular remodeling, is considered a key event in the pathogenesis of diabetic vascular complications [5].

Methylglyoxal (MG) is a highly reactive metabolite of glucose [6], [7]. Increased MG levels were observed in vascular endothelial cells cultured in high glucose-containing media and in the aorta, kidney and retina of diabetic rats [8], [9], [10]. Accumulating evidence indicates that high glucose-increased MG production is an important molecular mechanism linking diabetes to endothelial damage. MG modifies lysine, arginine, and cysteine residues in peptides or proteins to yield irreversible advanced glycosylation end products (AGEs), leading to cross-linking and denaturation of proteins [11], [12], [13]. MG also increases the generation of reactive oxygen species (ROS) and oxidative stress in endothelial cells [8]. Moreover, indirect evidence implicates MG in the high glucose-activated protein kinase C (PKC), hexosamine, and nuclear factor κB (NF-κB) pathways. For example, AGEs activated PKC in cultured endothelial cells [14]; incubation with alagebrium, an AGEs breaker, reduced PKC activation in high glucose-treated vascular smooth muscle cells (VSMCs) [15]. Activation of the hexosamine pathway by hyperglycemia leads to O-linked N-acetyl glucosamine (O-GlcNAc) modification of various proteins on serine or threonine residues which impair the normal functions of proteins [1]. Overexpression of glyoxalase-1, an enzyme metabolizing MG, reduced high glucose-increased O-GlcNAc modification in endothelial cells [16]. NF-κB is activated by the elevation of glucose, leading to up-regulation of target genes relevant to endothelial inflammation and apoptosis [17], [18]. AGEs activated NF-κB in cultured endothelial cells [19]. A study in the cultured VSMCs showed that MG treatment induced the activation of NF-κB [20]. These data suggest that inhibition of MG production could be a strategy to prevent endothelial damage in diabetic vascular complications.

Several MG scavengers have been developed, but most of them, such as aminoguanidine, metformin and N-acetyl cysteine, are non-specific to MG and their utilization for scavenging MG and preventing diabetic damage is limited [21]. We recently identified aldolase B which converts glucose or fructose to MG as a primary enzyme responsible for MG overproduction in high glucose-treated VSMCs and in aorta of diabetic rats [6]. In this paper, the gene expression of aldolase B and its role in MG formation in high glucose-treated endothelial cells were evaluated and whether knockdown of aldolase B in endothelial cells prevented high glucose-induced MG overproduction and other metabolic and signaling abnormalities was investigated.

Materials and Methods

Cell Culture and Treatment

Human umbilical vein endothelial cells (HUVECs) from American Type Culture Collection were cultured in Kaighns F12K medium containing 10% fetal bovine serum (FBS), 0.1 mg/mL heparin and 0.03–0.05 mg/mL endothelial cell growth supplement. HUVECs between passage 3 and 6 were used for the experiments. EA. hy926 cells, a endothelial cell line derived from the fusion of HUVECs with A549 lung carcinoma cells [22] (a gift kindly provided by Dr. Cora-Jean Edgell, University of North Carolina at Chapel Hill), were cultured in Dulbecco’s modified Eagle’s medium (DMEM) supplemented with 10% FBS. EA. hy926 cells will retain endothelial phenotype and functions, such as expression of eNOS, and grow rapidly in culture without requirement for special growth factors, and thus are very often used as an in vitro model for endothelial cells [22], [23], [24], [25]. HUVECs and EA. hy926 cells were starved in DMEM containing 0.5% FBS for 24 hrs and then treated with glucose (25 mM) or MG (30 or 100 µM) in DMEM containing 10% FBS for 3 days.

Small Interfering RNA (siRNA)

Knockdown of aldolase A or aldolase B was established by 24-h transfection of cells with a siRNA pool (a mixture of 3 or 4 different siRNA duplexes) targeting aldolase A or B (Santa Cruz Biotechnology Inc., Santa Cruz, CA, USA) in DharmaFECT™ 4 Transfection Reagent (Thermo Fisher, Nepean, ON, Canada). The non-targeting control siRNA pool is purchased from Santa Cruz (CA, USA). Briefly, transfection complexes were formed by incubating 100 µL siRNA pool (10 µM) with 25 µL of DharmaFECT™ 4 Transfection Reagent in 1 mL of serum-free DMEM for 20 min at room temperature. Transfection complexes were mixed with 4 mL of serum-free DMEM and added to cells. After 6 h of incubation, 5 mL DMEM supplemented with 20% FBS was added for a final siRNA concentration at 100 nM. After another 18 h, the transfection medium was replaced by 10% FBS DMEM with or without MG or high glucose and incubated for 3 days. Aldolase B mRNA was determined by a real-time PCR assay using SYBR Green PCR Master Mix (Bio-Rad) with the primers for human aldolase B (forward 5′-AGCCTCGCTATCCAGGAAAACG-3′, reverse 5′-TGGCAGTGTTCCAGGTCATGGT-3′) and β-actin (forward 5′-ACTTAGTTGCGTTACACCCTT-3′, reverse 5′-GTCACCTTCACCGTTCCA-3′). Primers for human aldolase A (Catalog Number QT00082460) and aldose reductase (Catalog Number QT01668695) were purchased from Qiagen (Mississauga, ON, Canada). The mRNA expression of aldolase A, aldolase B or aldose reductase was normalized relative to reference gene β-actin using ΔCt calculations [26].

thumbnail
Figure 1. Knockdown of aldolase B prevented MG overproduction in high glucose-treated EA. hy926 cells.

Real-time PCR analysis of aldose reductase and aldolase B expression (A, C) and MG levels (B, D) in human umbilical vein endothelial cells (HUVECs) (A, B) and EA. hy926 cells (C, D) treated with glucose (Glu) in the presence or absence of aminoguanidine (AG, 1 mM) and alagebrium (ALA, 100 µM) for 3 days. **P<0.01 vs. 5 mM glucose; ##P<0.01 vs. 25 mM glucose. Levels of aldolase B mRNA (E) and levels of MG (F) in wide-type cells (WT) and cells transfected with control or aldolase B siRNA, or only transfection agent (Mock). **P<0.01 vs. 5 mM glucose; ##P<0.01 vs. control siRNA.

https://doi.org/10.1371/journal.pone.0041495.g001

MG Measurement

MG levels were determined using our recently modified method [27]. Briefly, cells were sonicated three times for 5 seconds each time and centrifuged at 12,000 rpm (10 min, 4°C). The supernatant of 180 µL was incubated with 180 µL of perchloric acid (PCA, 1 N) and 40 µL of o-phenylenediamine (o-PD, 100 mM) for 24 h at room temperature in the dark. The mixture was centrifuged at 12,000 rpm (10 min, 4°C). The supernatant of 180 µL was mixed with 20 µL of 5-methylquinoxaline (5-MQ, internal standard) and analyzed by high-performance liquid chromatography (HPLC) with mobile phase buffer containing 17% acetonitrile, 8% 50 mM NaH2PO4 (pH 4.5), and 75% water.

Confocal Imaging of AGEs

N(ε)-carboxyethyl-lysine (CEL) is a MG specific AGE formed by the reaction between MG and lysine residues in proteins [28]. CEL formation in EA. hy926 cells was visualized by confocal microscopy after immunofluorescent staining. Cells were cultured on glass coverslips, fixed and permeabilized with pre-cold methanol (20 min, −20°C) and blocked with goat serum in phosphate-buffered saline (PBS) (1∶30, 30 min), and then, incubated with mouse monoclonal CEL antibody (Cosmo bio, diluted 1∶250 in blocking solution, 3 h at room temperature or overnight at 4°C). Subsequently, the processed cell preparations were washed with PBS, and incubated with Alexa 488-conjugated secondary antibodies (Invitrogen, Burlington, ON, Canada, diluted 1∶300 in blocking solution, 2 h at room temperature). Finally, the prepared cells were washed again and mounted in mounting media with propidium iodide (Invitrogen, Burlington, ON, Canada). Thereafter, the slides were examined under a confocal microscope with the appropriate filters. The fluorescence intensity was determined using Image J by analyzing at least 50 random cells per sample.

Measurement of Oxidative Stress

After different treatments, cells were washed with PBS containing 0.9 mM calcium chloride and 0.5 mM magnesium chloride, and then stained with a non-specific ROS probe (DCF–DA, Invitrogen, Burlington, ON, Canada) or a specific cell-permeable fluorogenic H2O2 probe (Calbiochem, San Diego, CA, USA). The H2O2 probe is a monosulfonated non-fluorescent fluorescein ester compound which selectively and sensitively reacts with H2O2 through a non-oxidative mechanism to yield corresponding fluorescein [29]. The fluorescence intensities of these probes were analyzed with a Fluoroskan Ascent plate reader (Thermo LabSystem, Franklin, MA, USA) as previously described [29], [30]. Protein oxidation was assessed by measuring total protein carbonyls with an immunoblot kit (Cell Biolabs Inc., San Diego, CA, USA). DNA oxidation biomarker 8-oxo-dG was visualized by immunofluorescent staining and photographed under a fluorescent microscopy, using a specific mouse monoclonal 8-oxo-dG antibody (Trevigen, Gaithersburg, MD, USA), following the manufacturer’s instructions.

Western Blot Analysis

Cells were harvested and lysed in RIPA buffer (Santa Cruz Biotechnology Inc., Santa Cruz, CA, USA) supplemented with protease inhibitors. Total cellular proteins were fractionated by 10% SDS-PAGE and immunoblotted with antibodies as follows: O-GlcNAc (RL2) (1∶1000, Thermo Fisher, Nepean, ON, Canada), and α-tubulin (1∶500, Santa Cruz Biotechnology Inc., Santa Cruz, CA, USA). Nuclear proteins were extracted as previously described [31] and level of nuclear NF-κB was measured with antibodies against NF-κB (p65) (1∶500) and lamin B (1∶1000) purchased from Santa Cruz Biotechnology Inc., Santa Cruz, CA, USA.

Membrane PKC Activity

Cells cultured on 100-mm dish were washed, scrapped off, suspended in 1 mL of Tris-sucrose buffer (20 mM Tris-base, 2 mM EDTA, 0.5 mM EGTA and 0.3 M sucrose, pH 7.4), and then added with protease inhibitors and homogenized by being passed 15 times through a 27.5 gauge needle. After centrifugation at 2,500 g (10 min, 4°C) to remove nuclei and cell debris, cell membrane was fractionated by a high speed ultra-centrifugation at 105,000 g (30 min, 4°C). Membrane pellets were washed with Tris buffer (20 mM Tris-base, 2 mM EDTA and 0.5 mM EGTA, pH 7.4), ultra-centrifuged (105,000 g, 30 min, 4°C) and re-suspended in 0.3 mL of Tris buffer with 0.5% Triton X-100 on ice for 1 h. The supernatant after ultra-centrifugation (105,000 g, 30 min, 4°C) was collected and membrane PKC proteins were purified through a DEAE cellulose (DE-52) column previously equilibrated with the Tris buffer. After washing the column with 3 mL of Tris buffer, the bound PKC was eluted with 0.5 ml Tris buffer containing 0.2 M NaCl. Membrane PKC activity was assessed by a PKC activity assay kit according to the manufacturer’s instructions (Assay Designs, Ann Arbor, MI, USA).

thumbnail
Figure 2. Knockdown of aldolase B prevented AGEs overproduction in high glucose-treated EA. hy926 cells.

Levels of MG (A) and N(ε)-carboxyethyl-lysine (CEL) in green (B) in cells treated with exogenous MG for 3 days. **P<0.01 vs. control (5 mM glucose). (C) CEL levels in cells treated with glucose (Glu) in the presence or absence of aminoguanidine (AG, 1 mM) and alagebrium (ALA, 100 µM) for 3 days. *P<0.05, **P<0.01 vs. 5 mM glucose; ##P<0.01 vs. 25 mM glucose. (D) CEL levels in cells transfected with control or aldolase B siRNA, or only transfection agent (mock). **P<0.01 vs. 5 mM glucose; ##P<0.01 vs. control siRNA. Nuclear DNA (red) was stained with propidium iodide (PI). The summary of fluorescence intensity of CEL was measured using Image J.

https://doi.org/10.1371/journal.pone.0041495.g002

Materials

(E)-2-(4-fluorophenethyl)-3-fluoroallylamine (MDL-72974) was a generous gift from Dr. Peter Yu (Department of Pharmacology, University of Saskatchewan, Canada). Alagebrium was a generous gift from Synvista Therapeutics (Montvale, NJ). Diallyl disulfide (DADS) and aminoguanidine was purchased from Sigma-Aldrich, Oakville, ON, Canada.

Statistics

Data are expressed as mean ± SEM from at least five independent experiments (n≥5 in each group). Statistical analyses were performed using parametric Student’s t-test (two-tailed) or one-way ANOVA.

thumbnail
Figure 3. Knockdown of aldolase B prevented the increase of oxidized DCF and H2O2 levels in high glucose-treated EA. hy926 cells.

Levels of oxidized DCF (A) in cells treated with exogenous MG (**P<0.01 vs. control), (B) in cells treated with glucose in the presence or absence of aminoguanidine (AG, 1 mM) and alagebrium (ALA, 100 µM) (*P<0.05, **P<0.01 vs. 5 mM glucose; ##P<0.01 vs. 25 mM glucose), and (C) in cells transfected with control or aldolase B siRNA, or only transfection agent (mock) (*P<0.05, **P<0.01 vs. 5 mM glucose; ##P<0.01 vs. control siRNA). Levels of H2O2 (D) in cells treated with exogenous MG (*P<0.05, **P<0.01 vs. control), (E) in cells treated with glucose in the presence or absence of aminoguanidine (AG, 1 mM) and alagebrium (ALA, 100 µM) (**P<0.01 vs. 5 mM glucose; ##P<0.01 vs. 25 mM glucose), and (F) in cells transfected with control or aldolase B siRNA, or only transfection agent (mock) (**P<0.01 vs. 5 mM glucose; ##P<0.01 vs. control siRNA).

https://doi.org/10.1371/journal.pone.0041495.g003

Results

High Glucose Up-regulated Aldolase B and Increased MG Formation in HUVECs and EA. hy926 cells

High glucose (25 mM) treatment up-regulated aldose reductase (P<0.01) and aldolase B (P<0.01) mRNA expression and accelerated MG formation (P<0.01) in HUVECs (Figs. 1A, 1B). Cellular MG overproduction induced by high glucose was partially reduced by aminoguanidine (P<0.01), and not reduced by alagebrium (P = 0.78) (Fig. 1B).

To investigate the role of aldolase B in the high glucose-induced MG overproduction and endothelial cell dysfunction, HUVEC-derived EA. hy926 cells were used in this study. Similarly, high glucose up-regulated aldose reductase (P<0.01) and aldolase B (P<0.01) in EA. hy926 cells (Fig. 1C). High glucose increased MG production (P<0.01) in EA. hy926 cells, which was partially reduced by aminoguanidine (P<0.01) and unaltered by alagebrium (P = 0.19) (Fig. 1D).

Knockdown of Aldolase B Prevented High Glucose-increased MG Formation in EA. hy926 cells

Transfection with aldolase B siRNA reduced cellular mRNA levels of aldolase B by 74%, and completely prevented high glucose-elevated formation of MG in EA. hy926 cells (Figs. 1E, 1F). Aldolase A, cytochrome P450 2E1 (CYP 2E1) and semicarbazide-sensitive amine oxidase (SSAO) are responsible for MG generation in glycolysis and in the metabolism of fatty acids and proteins, respectively [6]. However, high glucose-increased MG production in EA. hy926 cells was not affected by application of DADS (an inhibitor of CYP 2E1, 100 µM, P = 0.51) or MDL-72974 (an inhibitor of SSAO, 5 µM, P = 0.84), and by transfection with aldolase A siRNA (P = 0.24) which reduced cellular mRNA levels of aldolase A by 70% (data not shown).

Knockdown of Aldolase B Prevented High Glucose-increased AGEs Formation in EA. hy926 cells

Since incubation of EA. hy926 cells with MG (100 µM) or glucose (25 mM) induced a similar increase in cellular MG levels (P = 0.88) (Fig. 2A), the direct effects of MG (30 or 100 µM) on endothelial cells were investigated. MG (30 or 100 µM, 3 days) elevated CEL levels (P<0.01) in EA. hy926 cells (Fig. 2B). Glucose (25 mM) treatment increased cellular CEL levels (P<0.01), which was similar with that induced by 100 µM MG (P = 0.71) (Figs. 2B, 2C). High glucose-induced CEL overproduction in endothelial cells was partially reduced by aminoguanidine (P<0.01) and completely prevented by alagebrium (P<0.01) or knockdown of aldolase B expression (P<0.01) (Figs. 2C, 2D).

Knockdown of Aldolase B Prevented High Glucose-induced ROS in EA. hy926 cells

High glucose-increased production of ROS is regarded as an important contributor to endothelial dysfunction in diabetic vascular complications [32]. MG (30 or 100 µM, 3 days) elevated levels of oxidized DCF (a marker of total cellular ROS) in EA. hy926 cells (P<0.01) (Fig. 3A). The increase in oxidized DCF levels induced by MG (100 µM) was similar with that induced by glucose (25 mM) (P = 0.79) (Figs. 3A, 3B). High glucose-increased cellular formation of oxidized DCF was totally abolished by aminoguanidine (P<0.01) or by transfection with aldolase B siRNA (P<0.01), and partially reduced by alagebrium (P<0.01) (Figs. 3B, 3C). Cellular levels of H2O2 were similarly elevated by MG (100 µM) and glucose (25 mM) (P = 0.54) (Figs. 3D, 3E). Application of aminoguanidine or alagebrium, or transfection of aldolase B siRNA prevented the formation of H2O2 (P<0.01) in high glucose-treated endothelial cells (Figs. 3E, 3F).

Knockdown of Aldolase B Prevented High Glucose-induced Protein and DNA Oxidation in EA. hy926 cells

MG (100 µM) and glucose (25 mM) induced similar increases in levels of protein carbonyls (a marker of protein oxidation) in EA. hy926 cells (P = 0.85) (Figs. 4A, 4B). Application of aminoguanidine or transfection of aldolase B siRNA totally abolished high glucose-increased levels of protein carbonyls (P<0.01). Application of alagebrium partially reduced the formation of protein carbonyls in high glucose-treated cells (P<0.01) (Figs. 4B, 4C).

thumbnail
Figure 4. Knockdown of aldolase B prevented protein and DNA oxidation in high glucose-treated EA. hy926 cells.

Levels of protein carbonyls (A) in cells treated with exogenous MG (**P<0.01 vs. control), (B) in cells treated with glucose in the presence or absence of aminoguanidine (AG, 1 mM) and alagebrium (ALA, 100 µM) (*P<0.05, **P<0.01 vs. 5 mM glucose; ##P<0.01 vs. 25 mM glucose), and (C) in cells transfected with control or aldolase B siRNA, or only transfection agent (mock) (**P<0.01 vs. 5 mM glucose; ##P<0.01 vs. control siRNA). Levels of 8-oxo-dG (D) in cells treated with exogenous MG (**P<0.01 vs. control), (E) in cells treated with glucose in the presence or absence of aminoguanidine (AG, 1 mM) and alagebrium (ALA, 100 µM) (*P<0.05, **P<0.01 vs. 5 mM glucose; ##P<0.01 vs. 25 mM glucose), and (F) in cells transfected with control or aldolase B siRNA, or only transfection agent (mock) (**P<0.01 vs. 5 mM glucose; ##P<0.01 vs. control siRNA). Fluorescence intensity of 8-oxo-dG per cell was measured using Image J.

https://doi.org/10.1371/journal.pone.0041495.g004

MG (30 or 100 µM, 3 days) elevated levels of 8-oxo-dG (a marker of DNA oxidation) in EA. hy926 cells (P<0.01) (Fig. 4D). The increase of 8-oxo-dG levels induced by MG (100 µM) was similar to that induced by 25 mM glucose (P = 0.26) (Figs. 4D, 4E). High glucose-increased 8-oxo-dG levels were prevented by aminoguanidine or by transfection with aldolase B siRNA, and partially reduced by alagebrium (P<0.01) (Figs. 4E, 4F).

Knockdown of Aldolase B Blocked High Glucose-activated Metabolic or Signalling Pathways in EA. hy926 cells

Activation of the hexosamine pathway by high glucose causes an elevated O-GlcNAc modification of nuclear and cytosolic proteins [1]. MG (30 or 100 µM) increased O-GlcNAc modification of proteins in EA. hy926 cells (P<0.01) (Fig. 5A). Glucose (25 mM)-elevated O-GlcNAc modification was similar to that induced by MG (100 µM) (P = 0.76), and totally abolished by alagebrium (P<0.01) or by transfection with aldolase B siRNA (P<0.01) but only partially reversed by aminoguanidine (P<0.01) (Figs. 5B, 5C).

thumbnail
Figure 5. Knockdown of aldolase B prevented high glucose-increased O-linked N-acetyl glucosamine (O-GlcNAc) modification in EA. hy926 cells.

(A) O-GlcNAc modification of total cellular proteins in cells treated with exogenous MG for 3 days. **P<0.01 vs. control (5 mM glucose). (B) O-GlcNAc modification of total cellular proteins in cells treated with glucose (Glu) in the presence or absence of aminoguanidine (AG, 1 mM) and alagebrium (ALA, 100 µM). *P<0.05, **P<0.01 vs. 5 mM glucose; ##P<0.01 vs. 25 mM glucose. (C) O-GlcNAc modification of total cellular proteins in cells transfected with control or aldolase B siRNA, or only transfection agent (mock). **P<0.01 vs. 5 mM glucose; ##P<0.01 vs. control siRNA.

https://doi.org/10.1371/journal.pone.0041495.g005

Activation of PKC leads to its translocation to the plasma membrane where it catalyzes the phosphorylation of various substrates and mediates a diverse variety of biological processes [33]. MG (30 or 100 µM) elevated plasma membrane PKC activities in EA. hy926 cells (P<0.01) (Fig. 6A). High glucose (25 mM) incubation induced a similar elevation in the plasma membrane PKC activities as did 100 µM MG (P = 0.69) (Figs. 6A, 6B). High glucose-elevated plasma membrane PKC activity was prevented by alagebrium (P<0.01) or by knockdown of aldolase B (P<0.01), but only partially by aminoguanidine (P<0.01) (Figs. 6B, 6C).

thumbnail
Figure 6. Knockdown of aldolase B prevented high glucose-increased membrane protein kinase C (PKC) activities in EA. hy926 cells.

(A) Membrane PKC activities in cells treated with exogenous MG for 3 days. **P<0.01 vs. control (5 mM glucose). (B) Membrane PKC activities in cells treated with glucose (Glu) in the presence or absence of aminoguanidine (AG, 1 mM) and alagebrium (ALA, 100 µM). *P<0.05, **P<0.01 vs. 5 mM glucose; ##P<0.01 vs. 25 mM glucose. (C) Membrane PKC activities in cells transfected with control or aldolase B siRNA, or only transfection agent (mock). **P<0.01 vs. 5 mM glucose; ##P<0.01 vs. control siRNA.

https://doi.org/10.1371/journal.pone.0041495.g006

Activated NF-κB (p50/p65 dimer) translocates into the nucleus and regulates the expression of a large number of genes involved in immune and inflammatory response, apoptosis, cell proliferation and differentiation [34]. MG (30 or 100 µM) increased nuclear p65 subunit of NF-κB in EA. hy926 cells (P<0.01) (Fig. 7A). High glucose-elevated nuclear amount of NF-κB p65 was similar with that induced by 100 µM MG (P = 0.82), and prevented by alagebrium (P<0.01), aminoguanidine (P<0.01), and aldolase B knockdown (P<0.01) (Figs. 7A, 7B, 7C).

thumbnail
Figure 7. Knockdown of aldolase B prevented high glucose-increased NF-κB nuclear translocation in EA. hy926 cells.

(A) Nuclear p65 subunit of NF-κB in cells treated with exogenous MG for 3 days. **P<0.01 vs. control (5 mM glucose). (B) Nuclear p65 subunit of NF-κB in cells treated with glucose (Glu) in the presence or absence of aminoguanidine (AG, 1 mM) and alagebrium (ALA, 100 µM). **P<0.01 vs. 5 mM glucose; ##P<0.01 vs. 25 mM glucose. (C) Nuclear p65 subunit of NF-κB in cells transfected with control or aldolase B siRNA, or only transfection agent (mock). **P<0.01 vs. 5 mM glucose; ##P<0.01 vs. control siRNA.

https://doi.org/10.1371/journal.pone.0041495.g007

Discussion

Our present work demonstrated that siRNA knockdown of aldolase B blocked high glucose-activated metabolic and signaling pathways by the normalization of MG production in endothelial cells. This revelation was substantiated by the following observations: 1) Treatment of EA. hy926 cells with MG (100 µM) and high glucose (25 mM) induced a similar increase in cellular MG levels and a similar activation of biochemical pathways involved in hyperglycemic damage; 2) Aldolase B is the major enzyme for high glucose-increased MG production in endothelial cells because aldolase B knockdown completely inhibited MG overproduction in high glucose-treated EA. hy926 cells; 3) Both knockdown of aldolase B and the application of aminoguanidine or alagebrium prevented high glucose-activated metabolic and signaling pathways in EA. hy926 cells.

Enhanced accumulation of MG is postulated to be one of the important molecular mechanisms leading to endothelial dysfunction and diabetic vascular complications [1], [5], [8]. We previously reported that incubation with MG (30 or 100 µM) and glucose (25 mM) similarly decreased NO synthase activity and NO production in cultured endothelial cells and reduced endothelium-dependent muscle relaxation in rat aorta [8]. Our present study further validates the role of MG as an upstream activator for hyperglycemia-induced metabolic and signaling changes. MG is the major precursor of AGEs in endothelial cells [10]. MG treatment elevated AGEs (as estimated by intracellular CEL) levels in EA. hy926 cells (Fig. 2). AGEs alter protein structures and functions. For example, MG-modified extracellular matrix molecules impaired matrix-matrix interactions and increased the stiffness of the vasculature [35]. In addition, AGEs can activate their specific receptors on endothelial cells and cause cellular perturbation, such as increased permeability, oxidative stress, activation of NF-κB and vascular inflammation [35], [36]. MG is also a pro-oxidant [30], [37]. A possible reason is a MG-induced decrease in mitochondrial complex III and SOD activities [38]. Here we found MG enhanced the formation of ROS and oxidation of protein and DNA in EA. hy926 cells (Figs. 3, 4). Moreover, we provided the first evidence that treatment with MG can directly stimulate O-GlcNAc modification and plasma membrane PKC activation (Figs. 5, 6). It is well known that abnormal activation of PKC by high glucose decreased NO production and increased vasoconstrictor endothelin-1 (ET-1) and ROS production in endothelial cells [1]. Increased O-GlcNAc modification of proteins appears to be important in the pathogenesis of endothelial dysfunction. For example, high glucose-increased O-GlcNAc modification of endothelial NO synthase decreased its activity in endothelial cells [39]. We also found that MG directly activated NF-κB by stimulating its nuclear translocation in endothelial cells (Fig. 7). High glucose-activated NF-κB via AGEs, ROS or PKC increased expression of genes contributing to endothelial dysfunction, such as ET-1, adhesion molecules and inflammatory cytokines [35], [40], [41].

Aldolase B is a major enzyme responsible for high glucose-induced MG overproduction in VSMCs and the aorta [6]. Triosephosphates glyceraldehyde 3-phosphate (GA3P) and dihydroxyacetone phosphate (DHAP) are considered major sources for endogenous MG formation and showed high efficiencies of non-enzymatic conversion to MG [6], [42], [43]. In cells, fructose is quickly phosphorylated to fructose 1-phosphate (F-1-P), which is cleaved by aldolase B to generate GA3P and DHAP [44]. Fructose is produced from glucose via the polyol pathway [1], [6], [45]. On the other hand, glucose is metabolized enzymatically through the glycolytic pathway into fructose-1,6-diphosphate, which subsequently forms GA3P and DHAP catalyzed by aldolase A [44]. Secondary sources of MG include the oxidation of aminoacetone by SSAO and the oxidation of acetone by CYP 2E1 [46]. We have recently reported that aldolase B, but not aldolase A, SSAO or CYP 2E1, was up-regulated and MG was over-produced in the aorta of diabetic rats; knockdown of aldolase B prevented high glucose-elevated MG formation in VSMCs [6]. In the present work, we observed that high glucose up-regulated aldose reductase (the first and rate-limiting enzyme of the polyol pathway [45]) and aldolase B gene expression and increased MG formation in endothelial cells (Fig. 1). siRNA knockdown of aldolase B completely inhibited the excess MG generation in glucose-treated endothelial cells (Fig. 1). However, siRNA knockdown of aldolase A or inhibition of SSAO or CYP 2E1 had no effect on glucose-increased cellular MG overproduction (data not shown). These data indicate that aldolase B is predominantly responsible for glucose-increased MG formation in endothelial cells and the inhibition of MG formation is solely responsible for the observed effects of aldolase B knockdown on high glucose-activated metabolic and signaling pathways.

Aminoguanidine and alagebrium are the most widely used MG scavenger and AGEs breaker, respectively [21]. The inhibitory effects of aminoguanidine or alagebrium on high glucose-induced endothelial abnormalities confirm the role of MG as a mediator for high glucose-activated cellular pathways (Figs. 2, 3, 4, 5, 6, 7). However, our work also reveals limitations of aminoguanidine or alagebrium in accurately evaluating MG’s contribution to endothelial dysfunction and diabetic complications, when compared with the knockdown of aldolase B which specifically prevents glucose-induced MG overproduction. Aminoguanidine is a non-specific MG scavenger. Its guanidine residue can react with the carbonyl in MG or in other carbonyl compounds, such as 3-deoxyglucosone and malondialdehyde [21]. Moreover, aminoguanidine can react directly with ROS, such as H2O2, hydroxyl radical and peroxynitrite [47]. We found that the application of aminoguanidine completely abolished oxidative stress, but only partially decreased MG and CEL production, O-GlcNAc modification and plasma membrane PKC activities in high glucose-treated cells (Figs. 1, 2, 3, 4, 5, 6). Alagebrium breaks the established AGE crosslinks [21]. The application of alagebrium completely inhibited the formation of CEL, but it did not change MG levels in high glucose-treated EA. hy926 cells (Figs. 1, 2). In addition to the formation of AGEs, MG also stimulates the formation of ROS [38]. Our studies showed that alagebrium only partially reduced the formation of oxidized DCF, protein carbonyls and 8-oxo-dG in high glucose-treated EA. hy926 cells (Figs. 3, 4), although it can react with H2O2 in the test tube [48] and completely reduced the glucose-increased H2O2 formation in our test cells (Fig. 3).

In conclusion, MG directly mediates high glucose-induced production of AGEs, oxidative stress, and increases in O-GlcNAc modification and protein levels or activities of protein kinase C and NF-κB in endothelial cells. More importantly, this study demonstrates that aldolase B is the major enzyme for high glucose-increased MG production in endothelial cells. Knockdown of aldolase B prevents MG overproduction and, by doing so, blocks high glucose-induced activation of multiple metabolic and signaling pathways in endothelial cells.

Acknowledgments

We are grateful to Mrs. Arlene Drimmie (Department of Pharmacology, University of Saskatchewan) for her excellent technical assistance.

Author Contributions

Conceived and designed the experiments: JL RW LW. Performed the experiments: JL TCM AB. Analyzed the data: JL TCM AB. Wrote the paper: JL LW KD RW. Obtained permission for use of cell line: RW.

References

  1. 1. Brownlee M (2001) Biochemistry and molecular cell biology of diabetic complications. Nature 414: 813–820.
  2. 2. Hanssen KF (1997) Blood glucose control and microvascular and macrovascular complications in diabetes. Diabetes 46: S101–103.
  3. 3. Scalia R, Gong Y, Berzins B, Zhao LJ, Sharma K (2007) Hyperglycemia is a major determinant of albumin permeability in diabetic microcirculation: the role of mu-calpain. Diabetes 56: 1842–1849.
  4. 4. Han J, Mandal AK, Hiebert LM (2005) Endothelial cell injury by high glucose and heparanase is prevented by insulin, heparin and basic fibroblast growth factor. Cardiovasc Diabetol 4: 12.
  5. 5. Schalkwijk CG, Stehouwer CD (2005) Vascular complications in diabetes mellitus: the role of endothelial dysfunction. Clin Sci (Lond) 109: 143–159.
  6. 6. Liu J, Wang R, Desai K, Wu L (2011) Upregulation of aldolase B and overproduction of methylglyoxal in vascular tissues from rats with metabolic syndrome. Cardiovasc Res 92: 494–503.
  7. 7. Dhar A, Desai K, Kazachmov M, Yu P, Wu L (2008) Methylglyoxal production in vascular smooth muscle cells from different metabolic precursors. Metabolism 57: 1211–1220.
  8. 8. Dhar A, Dhar I, Desai KM, Wu L (2010) Methylglyoxal scavengers attenuate endothelial dysfunction induced by methylglyoxal and high concentrations of glucose. Br J Pharmacol 161: 1843–1856.
  9. 9. Phillips SA, Mirrlees D, Thornalley PJ (1993) Modification of the glyoxalase system in streptozotocin-induced diabetic rats. Effect of the aldose reductase inhibitor Statil. Biochem Pharmacol 46: 805–811.
  10. 10. Shinohara M, Thornalley PJ, Giardino I, Beisswenger P, Thorpe SR, et al. (1998) Overexpression of glyoxalase-I in bovine endothelial cells inhibits intracellular advanced glycation endproduct formation and prevents hyperglycemia-induced increases in macromolecular endocytosis. J Clin Invest 101: 1142–1147.
  11. 11. Chang T, Wang R, Olson DJ, Mousseau DD, Ross AR, et al. (2011) Modification of Akt1 by methylglyoxal promotes the proliferation of vascular smooth muscle cells. FASEB J 25: 1746–1757.
  12. 12. Jia X, Olson DJ, Ross AR, Wu L (2006) Structural and functional changes in human insulin induced by methylglyoxal. FASEB J 20: 1555–1557.
  13. 13. Wang X, Desai K, Clausen JT, Wu L (2004) Increased methylglyoxal and advanced glycation end products in kidney from spontaneously hypertensive rats. Kidney Int 66: 2315–2321.
  14. 14. Mamputu JC, Renier G (2002) Advanced glycation end products increase, through a protein kinase C-dependent pathway, vascular endothelial growth factor expression in retinal endothelial cells. Inhibitory effect of gliclazide. J Diabetes Complications 16: 284–293.
  15. 15. Thallas-Bonke V, Lindschau C, Rizkalla B, Bach LA, Boner G, et al. (2004) Attenuation of extracellular matrix accumulation in diabetic nephropathy by the advanced glycation end product cross-link breaker ALT-711 via a protein kinase C-alpha-dependent pathway. Diabetes 53: 2921–2930.
  16. 16. Yao D, Taguchi T, Matsumura T, Pestell R, Edelstein D, et al. (2007) High glucose increases angiopoietin-2 transcription in microvascular endothelial cells through methylglyoxal modification of mSin3A. J Biol Chem 282: 31038–31045.
  17. 17. Morigi M, Angioletti S, Imberti B, Donadelli R, Micheletti G, et al. (1998) Leukocyte-endothelial interaction is augmented by high glucose concentrations and hyperglycemia in a NF-kB-dependent fashion. J Clin Invest 101: 1905–1915.
  18. 18. Ho FM, Lin WW, Chen BC, Chao CM, Yang CR, et al. (2006) High glucose-induced apoptosis in human vascular endothelial cells is mediated through NF-kappaB and c-Jun NH2-terminal kinase pathway and prevented by PI3K/Akt/eNOS pathway. Cell Signal 18: 391–399.
  19. 19. Bierhaus A, Chevion S, Chevion M, Hofmann M, Quehenberger P, et al. (1997) Advanced glycation end product-induced activation of NF-kappaB is suppressed by alpha-lipoic acid in cultured endothelial cells. Diabetes 46: 1481–1490.
  20. 20. Wu L, Juurlink BH (2002) Increased methylglyoxal and oxidative stress in hypertensive rat vascular smooth muscle cells. Hypertension 39: 809–814.
  21. 21. Desai K, Wu L (2007) Methylglyoxal and advanced glycation endproducts: new therapeutic horizons? Recent Pat Cardiovasc Drug Discov 2: 89–99.
  22. 22. Edgell CJ, McDonald CC, Graham JB (1983) Permanent cell line expressing human factor VIII-related antigen established by hybridization. Proc Natl Acad Sci U S A 80: 3734–3737.
  23. 23. Riederer I, Sievert W, Eissner G, Molls M, Multhoff G (2010) Irradiation-induced up-regulation of HLA-E on macrovascular endothelial cells confers protection against killing by activated natural killer cells. PLoS One 5: e15339.
  24. 24. O’Connor RP, Madison SD, Leveque P, Roderick HL, Bootman MD (2010) Exposure to GSM RF fields does not affect calcium homeostasis in human endothelial cells, rat pheocromocytoma cells or rat hippocampal neurons. PLoS One 5: e11828.
  25. 25. Lim EJ, Smart EJ, Toborek M, Hennig B (2007) The role of caveolin-1 in PCB77-induced eNOS phosphorylation in human-derived endothelial cells. Am J Physiol Heart Circ Physiol 293: H3340–3347.
  26. 26. Pfaffl MW (2001) A new mathematical model for relative quantification in real-time RT-PCR. Nucleic Acids Res 29: e45.
  27. 27. Dhar A, Desai K, Liu J, Wu L (2009) Methylglyoxal, protein binding and biological samples: are we getting the true measure? J Chromatogr B Analyt Technol Biomed Life Sci 877: 1093–1100.
  28. 28. Singh R, Barden A, Mori T, Beilin L (2001) Advanced glycation end-products: a review. Diabetologia 44: 129–146.
  29. 29. Maeda H, Fukuyasu Y, Yoshida S, Fukuda M, Saeki K, et al. (2004) Fluorescent probes for hydrogen peroxide based on a non-oxidative mechanism. Angew Chem Int Ed Engl 43: 2389–2391.
  30. 30. Chang T, Wang R, Wu L (2005) Methylglyoxal-induced nitric oxide and peroxynitrite production in vascular smooth muscle cells. Free Radic Biol Med 38: 286–293.
  31. 31. Li Y, Bor YC, Misawa Y, Xue Y, Rekosh D, et al. (2006) An intron with a constitutive transport element is retained in a Tap messenger RNA. Nature 443: 234–237.
  32. 32. Cai H, Harrison DG (2000) Endothelial dysfunction in cardiovascular diseases: the role of oxidant stress. Circ Res 87: 840–844.
  33. 33. Nishizuka Y (1992) Intracellular signaling by hydrolysis of phospholipids and activation of protein kinase C. Science 258: 607–614.
  34. 34. Barnes PJ, Karin M (1997) Nuclear factor-kappaB: a pivotal transcription factor in chronic inflammatory diseases. N Engl J Med 336: 1066–1071.
  35. 35. Goldin A, Beckman JA, Schmidt AM, Creager MA (2006) Advanced glycation end products: sparking the development of diabetic vascular injury. Circulation 114: 597–605.
  36. 36. Wautier JL, Zoukourian C, Chappey O, Wautier MP, Guillausseau PJ, et al. (1996) Receptor-mediated endothelial cell dysfunction in diabetic vasculopathy. Soluble receptor for advanced glycation end products blocks hyperpermeability in diabetic rats. J Clin Invest 97: 238–243.
  37. 37. Wu L (2005) The pro-oxidant role of methylglyoxal in mesenteric artery smooth muscle cells. Can J Physiol Pharmacol 83: 63–68.
  38. 38. Wang H, Liu J, Wu L (2009) Methylglyoxal-induced mitochondrial dysfunction in vascular smooth muscle cells. Biochem Pharmacol 77: 1709–1716.
  39. 39. Du XL, Edelstein D, Dimmeler S, Ju Q, Sui C, et al. (2001) Hyperglycemia inhibits endothelial nitric oxide synthase activity by posttranslational modification at the Akt site. J Clin Invest 108: 1341–1348.
  40. 40. Nishikawa T, Edelstein D, Du XL, Yamagishi S, Matsumura T, et al. (2000) Normalizing mitochondrial superoxide production blocks three pathways of hyperglycaemic damage. Nature 404: 787–790.
  41. 41. Pieper GM, Riaz ul H (1997) Activation of nuclear factor-kappaB in cultured endothelial cells by increased glucose concentration: prevention by calphostin C. J Cardiovasc Pharmacol 30: 528–532.
  42. 42. Phillips SA, Thornalley PJ (1993) The formation of methylglyoxal from triose phosphates. Investigation using a specific assay for methylglyoxal. Eur J Biochem 212: 101–105.
  43. 43. Richard JP (1993) Mechanism for the formation of methylglyoxal from triosephosphates. Biochem Soc Trans 21: 549–553.
  44. 44. Cox TM (1994) Aldolase B and fructose intolerance. FASEB J 8: 62–71.
  45. 45. Chung SS, Chung SK (2005) Aldose reductase in diabetic microvascular complications. Curr Drug Targets 6: 475–486.
  46. 46. Desai KM, Chang T, Wang H, Banigesh A, Dhar A, et al. (2010) Oxidative stress and aging: is methylglyoxal the hidden enemy? Can J Physiol Pharmacol 88: 273–284.
  47. 47. Yildiz G, Demiryurek AT, Sahin-Erdemli I, Kanzik I (1998) Comparison of antioxidant activities of aminoguanidine, methylguanidine and guanidine by luminol-enhanced chemiluminescence. Br J Pharmacol 124: 905–910.
  48. 48. Park J, Kwon MK, Huh JY, Choi WJ, Jeong LS, et al. (2011) Renoprotective antioxidant effect of alagebrium in experimental diabetes. Nephrol Dial Transplant.