August 2005
Volume 46, Issue 8
Free
Retinal Cell Biology  |   August 2005
Effects of Oxidized and Glycated LDL on Gene Expression in Human Retinal Capillary Pericytes
Author Affiliations
  • Weiwei Song
    From the Section of Endocrinology and Diabetes, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma; and the
  • Jeremy L. Barth
    Department of Cell Biology and Anatomy and the
  • Yongxin Yu
    From the Section of Endocrinology and Diabetes, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma; and the
  • Kangmo Lu
    From the Section of Endocrinology and Diabetes, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma; and the
  • Azar Dashti
    From the Section of Endocrinology and Diabetes, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma; and the
  • Yan Huang
    Division of Endocrinology, Diabetes, and Medical Genetics, Medical University of South Carolina, Charleston, South Carolina.
  • Cynthia K. Gittinger
    Department of Cell Biology and Anatomy and the
  • W. Scott Argraves
    Department of Cell Biology and Anatomy and the
  • Timothy J. Lyons
    From the Section of Endocrinology and Diabetes, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma; and the
Investigative Ophthalmology & Visual Science August 2005, Vol.46, 2974-2982. doi:https://doi.org/10.1167/iovs.04-1505
  • Views
  • PDF
  • Share
  • Tools
    • Alerts
      ×
      This feature is available to authenticated users only.
      Sign In or Create an Account ×
    • Get Citation

      Weiwei Song, Jeremy L. Barth, Yongxin Yu, Kangmo Lu, Azar Dashti, Yan Huang, Cynthia K. Gittinger, W. Scott Argraves, Timothy J. Lyons; Effects of Oxidized and Glycated LDL on Gene Expression in Human Retinal Capillary Pericytes. Invest. Ophthalmol. Vis. Sci. 2005;46(8):2974-2982. https://doi.org/10.1167/iovs.04-1505.

      Download citation file:


      © ARVO (1962-2015); The Authors (2016-present)

      ×
  • Supplements
Abstract

purpose. Modified (oxidized and/or glycated) low-density lipoproteins (LDLs) have been implicated in retinal pericyte loss, one of the major pathologic features of early-stage diabetic retinopathy. To delineate underlying molecular mechanisms, the present study was designed to explore the global effects of modified LDL on pericyte gene expression.

methods. Quiescent human retinal pericytes were exposed to native LDL (N-LDL), glycated LDL (G-LDL), and heavily oxidized-glycated LDL (HOG-LDL) for 24 hours, and gene expression was evaluated by DNA microarray analysis. Several of the gene responses were checked, and in each case confirmed by reverse-transcription real-time PCR.

results. HOG-LDL induced a gene expression pattern markedly distinct from that of N-LDL or G-LDL, whereas G-LDL elicited gene expression similar to that of N-LDL. A comparison of responses to HOG-LDL versus N-LDL revealed 60 genes with expression that varied by ≥1.7-fold. The HOG-LDL-responsive genes included members of functional pathways, such as fatty acid, eicosanoid, and cholesterol metabolism; fibrinolytic regulation; cell growth and proliferation; cell stress responses; the kinin system; and angiogenesis.

conclusions. HOG-LDL elicits gene expression in retinal pericytes that may contribute to pericyte loss and other retinal abnormalities in diabetic retinopathy. Observed proapoptotic and proangiogenic responses to HOG-LDL may be of particular importance in this regard. The genes identified through these studies provide potential therapeutic targets for the prevention and treatment of diabetic retinopathy.

Hyperglycemia enhances nonenzymatic glycation and increases oxidative stress, resulting in modification of proteins, lipids, and DNA by covalent adducts and oxygen free radicals. 1 2 Extravasated low-density lipoproteins (LDL) are subject to such modification, and in the arterial intima, their role in the promotion of atherosclerosis is now established. In diabetic retinopathy, a leading cause of blindness among adults, 3 capillary leakage occurs early and results in plasma extravasation. Therefore, the possibility that extravasated, modified LDL may affect retinal pericytes merits investigation. In a prior study, we demonstrated a reduction in the number of cultured pericytes after exposure to glycated and mildly oxidized LDL versus native LDL. 4 We also observed that glycated-oxidized LDL stimulates apoptosis in cultured retinal pericytes. 5 These findings are consistent with the observation that pericytes are lost by apoptosis in vivo in diabetic retinopathy. 6  
In the present study, by using DNA microarray analysis, we investigated the effects of in vitro modified human LDL on gene expression in cultured human retinal pericytes. Native LDL (N-LDL) represented normal LDL as found in plasma in healthy people; glycated LDL (G-LDL) represented mildly modified LDL that has not been damaged by oxidation and is present in plasma of diabetic patients; and heavily oxidized-glycated LDL (HOG-LDL) simulated LDL that has been severely modified by prolonged extravasation. We identified altered expression of genes from several functional pathways, particularly in response to HOG-LDL. These findings may help identify new therapeutic targets for prevention and treatment of diabetic retinopathy. 
Methods
Preparation, Modification, and Characterization of LDL
LDL was isolated from three separate groups of human subjects. Each group comprised four healthy, normolipemic, nondiabetic subjects aged 20 to 40 years who were taking neither prescribed medications nor antioxidant vitamins. The study was approved by the Institutional Review Board at the Medical University of South Carolina (MUSC), and informed consent was obtained from all volunteers, in accordance with the provisions of the Declaration of Helsinki. N-LDL density ([d] = 1.019–1.063 g/mL) was prepared by sequential ultracentrifugation, pooled, and modified in vitro, as described previously. 7 Briefly, G-LDL was prepared by incubating N-LDL in 50 mM glucose for 72 hours at 37°C under antioxidant conditions. HOG-LDL was prepared from G-LDL by incubation in 10 μM CuCl2 for 24 hours at 37°C in air. The protein content of LDL was quantified with a bicinchoninic acid (BCA) assay kit (Pierce, Rockford, IL). Characterization of native and modified LDL by gel electrophoresis (Lipoepg; Beckman, Fullerton, CA), fluorescence at 360 excitation/430 emission (Fluorometer IV; Gilford, Oberlin, OH), and absorbance at 234 nm in a spectrophotometer (model DU650; Beckman) indicated that these products were similar to those previously reported. 7  
Cell Culture and Modified LDL Treatment
Human retinal pericytes (Clonetics, Walkersville, MD) were grown at 37°C with 5% CO2 in medium containing 5% fetal bovine serum, 0.04% hydrocortisone, 0.4% hFGF-B, 0.1% VEGF, 0.1% R3-IGF-1 (insulin-like growth factor), 0.1% ascorbic acid, 0.1% hEGF (human epidermal growth factor) and 0.1% GA-1000. At 85% confluence, cells were treated in serum-free medium (SFM) for 24 hours to induce quiescence and then for 24 hours with 100 mg/L N-LDL, G-LDL, or HOG-LDL, or they remained in SFM alone. Total RNA was collected from each sample for gene array analysis. This process was repeated four separate times to create a four replicates with each treatment. For additional experiments using RT-PCR to test the combined effects of HOG- and N-LDL, an identical protocol was used, except that HOG- and N-LDL were added together at 100 mg/L each. 
Total RNA Preparation and Microarray Hybridization
Probes for microarray hybridization were prepared as recommended by Affymetrix (Santa Clara, CA). Briefly, total RNA was isolated and purified (RNeasy Mini kit; Qiagen, Valencia, CA). RNA quality was assured by the A260/280 absorbance ratio and the 28s/18s rRNA ratio. Total RNA (10 μg) was converted into double-stranded cDNA with a T7-(dT)24 primer (Genset, San Diego, CA) and a cDNA synthesis kit (Custom SuperScript; Invitrogen, Carlsbad, CA). Biotin-labeled cRNA was synthesized from cDNA by in vitro transcription (Enzo BioArray HighYield RNA Transcript Labeling Kit; Enzo Life Sciences, Farmingdale, NY). After purification (RNeasy columns; Qiagen), labeled cRNA was fragmented and evaluated by agarose gel electrophoresis, to ensure appropriate size distribution (∼35–200 nt). Hybridization of cRNA samples to (HG-U95Av2 GeneChips; Affymetrix), posthybridization washing, fluorescence staining, and scanning were performed at the MUSC DNA Microarray and Bioinformatics Facility. DNA microarray data (raw and normalized) generated by this project are available online through the MUSC DNA Microarray Database (http://proteogenomics.musc.edu/quicksite/projectmanage.php?page=home). 
Real-Time RT-PCR
Triplicate RT-PCR was performed in a thermocycler (SmartCycler II; Cepheid, Sunnyvale, CA), with a PCR kit and master mix (GeneAmp and SYBR Green; Applied Biosystem, Inc. [ABI], Foster City, CA). Primers were selected so that the forward and reverse primer sequences were present on adjacent exons. Single-stranded cDNA was prepared from total RNA (0.5–1.0 μg) with oligo dT primer and MuLV reverse transcriptase. Two-step PCR was conducted with cDNA template (2 μL) and primers (3 picomoles) in a final volume of 25 μL (denaturation and hot start: 95°C for 10 minutes; 45 cycles with melting: 95°C, 15 seconds; and an extension: 60°C, 60 seconds). Melting-curve analysis (0.5°C/s increase from 55°C to 95°C) and agarose gel electrophoresis were performed to ensure correct amplicon size and reaction specificity. Quantification was determined from the threshold cycle (C T) of the target relative to that of glyceraldehyde-3-phosphate dehydrogenase (GAPDH) in the same cDNA sample. Gene expression was calculated by x-fold change = 2−Δ(ΔC T ), where ΔCT = CT,target − CT,GAPDH and Δ(ΔCT) = ΔCT,HOG LDL − ΔCT,N-LDL
Data Analysis
Microarray results were analyzed using the DNA-Chip Analyzer “dChip” (http://www.dchip.org/, a collaboration of the Wong Laboratory, Stanford University, Stanford, CA, and the Cheng Li Group, Harvard School of Public Health and Dana Farber Cancer Institute, Boston MA). Hybridization intensities were normalized by using the invariant gene set expression model. Model-based expression indices were calculated based on the perfect-match-only model. For genes differentially expressed among all treatments, gene expression levels were filtered with the following criteria: (1) variation across samples between 0.35 and 10; (2) genes scored present in ≥20% arrays; (3) replicate variation between 0 and 0.4; and (4) hybridization intensity ≥30 units in ≥20% of the samples. To identify gene expression differentially induced by HOG- and N-LDL, a change in hybridization intensity of ≥30 units (12.5% of GeneChip mean hybridization values [ABI]) was applied as a threshold to eliminate genes having small proportional changes and/or genes having overall low hybridization intensities. The change (x-fold) cutoffs between 1.5 and 2.0 were tested empirically to determine how they affected the number of genes discovered and the corresponding false discovery rate (FDR), estimated by 100 iterative comparisons of randomized sample groupings (dChip User’s Manual, http://biosun1.harvard.edu/complab/dchip/manual.htm). 8 9 As a function of increasing change, the number of genes discovered was significantly reduced (149 genes at 1.5-fold versus 32 genes at 2-fold), whereas FDR was only moderately improved (20% at 1.5-fold versus 12% at twofold). Therefore, the intermediate cutoff of 1.7-fold was selected, identifying 60 genes with an estimated FDR of ∼17% (Tables 1 2) . Unpaired t-tests performed on these genes identified a subset of 41 significant genes (P < 0.05), for which FDR approximated 0.0%, when taking into account all differential expression criteria. 
For RT-PCR, values were expressed as the mean ± SD. Statistical significance was determined by one-way ANOVA followed by the Dunnett test. P < 0.05 was considered significant. 
Results
Overall Gene Expression Profile
Four replicate DNA microarray experiments were conducted in which quiescent human retinal pericytes were treated in four different ways—that is, for 24 hours with N-LDL, G-LDL, HOG-LDL, or SFM. Analysis of hybridization data indicated that 152 genes were differentially expressed among treatment groups. Hierarchical clustering of the gene expression profiles revealed similarities and differences among the responses to LDL preparations (Fig. 1) . First, N-LDL caused significant changes in pericyte gene expression, inducing upregulation of 58 genes and downregulation of 50 genes by ≥1.7-fold (Supplementary Table S1). Second, G-LDL and N-LDL response profiles were largely the same, suggesting that under these conditions, glycation per se did not act as a gene regulatory stimulus. Third, HOG-LDL induced responses that were either similar or distinct from N-LDL. In instances in which the HOG-LDL response was distinct (Fig. 1 , black bars) the effect was sometimes similar to the SFM state, but in other instances was different from either SFM or N-LDL (Fig. 1 , green bar). Nineteen genes were affected by HOG-LDL ≥1.7-fold in relation to SFM and N-LDL (Supplementary Table S1 and the next section). These results demonstrate the importance of N-LDL in affecting gene regulation, and further indicate that HOG-LDL can cause responses that are distinct from those of either N-LDL or SFM. 
HOG-LDL Induces Specific Changes in Gene Expression
To understand better the effects of HOG-LDL on pericytes, microarray data were analyzed to identify genes differentially regulated by HOG- versus N-LDL. After genes that had low absolute differences (<30 units) were eliminated, 60 genes were found to have expression differing by ≥1.7-fold, 41 of which were significant by unpaired t-test (P < 0.05). Among the 60 genes, 21 were upregulated by HOG-LDL, with mean changes ranging up to 3.3-fold (Table 1) . Two fatty acid desaturases, FADS1 and FADS2, which are involved in the synthesis of long-chain polyunsaturated fatty acids (PUFAs), showed the greatest upregulation. Other lipid-related genes, including the LDL receptor (LDLR) and a fatty acid binding protein (FABP4), also were considerably upregulated. Thirty-nine genes were downregulated by HOG-LDL, with changes ranging to 6.8-fold (Table 2) . Included among the 60 HOG-LDL-regulated genes were the 19 that were also affected by ≥1.7-fold compared with SFM (Tables 1 2 , asterisks), again suggesting that some responses to HOG-LDL are unique and distinct from those to N-LDL or SFM. Overall categorization of the 60 genes is shown in Figure 2 . These genes are implicated in several pathways that can be related to diabetic retinopathy. 
Validation of Microarray Results
To verify the gene expression changes revealed by microarray analysis, 12 responding genes were tested by real-time RT-PCR, confirming the microarray findings in each case. Results for four of these genes are shown in Figure 3 . Of these, FABP4 and LDLR were upregulated by HOG- versus N-LDL, whereas bradykinin receptor B2 (BDKRB2) and insulin-like growth factor binding protein 3 (IGFBP3) were downregulated. Microarray findings were further confirmed for plasminogen activator inhibitor-1 (PAI-1, increased 1.7-fold by microarray) using both RT-PCR (2.8-fold increase) and Northern blot analysis (1.8-fold increase; data not shown). Two lipid-related genes, SDR1 and SC4MOL, which showed a more than 1.7-fold change but were not significantly changed by t-test, were also confirmed by RT-PCR (data not shown). 
Effects of Coincubation of N-LDL on HOG-LDL-Induced Gene Alterations
Gene expression responses to HOG-LDL that are distinct from N-LDL could be caused by lipoprotein deprivation (i.e., if modification inactivates the LDL) or as a reaction to a unique signal conferred by oxidation and glycation of LDL. To investigate these possibilities, experiments were performed to test the effects of combined treatment with N-LDL and HOG-LDL. As shown in Figure 3 , HOG-LDL-induced changes in LDLR, BDKRB2, and IGFBP3 expression were reversed by addition of N-LDL, but the increased expression of FABP4 was not altered by adding N-LDL. 
Discussion
Selective loss of pericytes is a major feature of early diabetic retinopathy, but the underlying mechanisms are unclear. Although the causes of vascular changes in the diabetic retina are probably multifactorial, the occurrence of antipericyte autoantibodies in type 2 diabetes suggests that autoimmunity may mediate pericyte loss. 10 However, the limited incidence of autoantibodies (∼55% of type 2 diabetes with retinopathy at grades from 10 to 53) 10 indicates that other mechanisms must also contribute to this process. In previous work with cultured bovine retinal pericytes, we reported that mildly glycated and/or oxidized LDL might contribute to pericyte loss, with an effect proportional to the extent of LDL modification. 4 Recently, we observed that a heavily modified form of LDL, HOG-LDL, exerts greater toxicity toward human retinal pericytes, stimulating apoptosis. 5 In those and the present experiments, HOG-LDL was used to simulate LDL that is severely modified in vivo after prolonged extravasation. This work therefore builds on the hypothesis that severe modification of LDL occurs not only in arteries, where it promotes and accelerates atherosclerosis in diabetes, but also in the diabetic retina. Consistent with this, we found that HOG-LDL induced many changes in pericyte gene expression that were distinct from that caused by N-LDL or G-LDL. Further, many HOG-LDL responsive genes are implicated in key metabolic and signaling functions that influence the pathologic course of diabetic retinopathy. 
The strengths of this study include the use of human cells and lipoproteins, the performance of quadruplicate microarray experiments, the use of three separate LDL pools, and, to date, the confirmation by alternative methods—that is, RT-PCR and Northern blot analysis—of all responses tested. Further, the degree of in vitro modification of LDL and the concentration (100 mg/L) used reflect a conservative estimate of conditions in vivo. Although it is difficult to select a period of exposure to modified LDL that simulates conditions in vivo, 24-hour exposure was chosen, because it does not cause the significant cell loss observed with longer exposure periods and/or higher doses of HOG-LDL. Such treatment aims to induce minor degrees of injury that would be present very early in the development of retinopathy. Therefore, it is not surprising that many of the genes significantly altered by HOG-LDL do not directly contribute to apoptosis. However, it is expected that these changes may eventually lead to this process. Ultimately, there are limitations inherent in using cultured cells to simulate conditions in vivo. 
Effects of HOG-LDL Distinct from Those of N-LDL
Oxidation and glycation of LDL may sufficiently alter the lipoprotein so that it no longer acts as cognate LDL. Alternatively, modification of LDL may confer a novel characteristic to the lipoprotein so that it now acts as a unique signaling entity. The differential expression responses to HOG- versus N-LDL suggest the presence of both mechanisms. For some genes, the HOG-LDL response was unlike that to N-LDL but was similar to the SFM state, indicating that the response is equivalent to that in the absence of N-LDL. However, in other cases, the HOG-LDL response was distinct from responses to either N-LDL or SFM. The existence of both types of genetic responses was confirmed when N-LDL and HOG-LDL were combined. N-LDL eliminated the HOG-LDL effect on LDLR, BDKRB2, and IGFBP3, but not on FABP4. Therefore, the observed changes in gene regulation include at least two components: a specific response to modified LDL and a response to N-LDL deprivation. Both components may be relevant in vivo in diabetes when retinal capillary leakage is present and extravasation of plasma occurs. Pericytes, which are normally outside the blood–retinal barrier, may then be exposed to any combination of native and modified LDL. 
Genes Involved in Fatty Acid, Eicosanoids, and Inflammation
The genes most upregulated by HOG-LDL are involved in fatty acid metabolism. Among these, FADS1 and FADS2 govern rate-limiting steps in long-chain PUFA synthesis. 11 FABP4 enhances fatty acid turnover, and a knockout of FABP4 inhibits atherosclerosis in animals. 12 Enhanced long-chain PUFA synthesis could reflect compensatory replacement of PUFAs (perhaps lost by free radical oxidation), or could reflect injurious changes in the synthesis of eicosanoids, mediators of inflammation, and thrombosis. 13 Other evidence indicates that oxidized LDL modulates arachidonate and eicosanoid pathways. Oxidized LDL activates cytosolic phospholipase A2, releases arachidonate, and induces apoptosis in macrophages and fibroblasts. The apoptosis is attenuated by phospholipase A2 inhibition. 14 Oxidized LDL also stimulates phospholipase A2 activity and prostaglandin E2 and thromboxane B2 production in rat mesangial cells. 15 In addition, oxidized LDL induces cyclooxygenase-2 protein expression and prostaglandin E2 release in endothelial cells. 16 In the present study, HOG-LDL decreased expression of prostaglandin E synthase by 1.9-fold. Therefore, further delineation of the eicosanoid pathway in mediation of HOG-LDL toxicity is required, especially since chronic inflammation has been implicated in diabetic retinopathy. 17 18 19  
Additional evidence that HOG-LDL induces inflammatory responses in pericytes is the upregulation of the acute-phase gene, PTX3, consistent with induction of PTX3 by oxidized LDL in human vascular smooth muscle cells 20 and its strong expression in atherosclerotic lesions. 21 Further, TNFAIP6, which is secreted by various cells on proinflammatory cytokine stimulation and which exhibits anti-inflammatory effects, 22 was downregulated. A knockout of TNFAIP6 exacerbates inflammation in mice 23 ; and, hence, reduced expression may indicate impaired ability of pericytes to combat local inflammation. 
Genes Involved in Cholesterol Metabolism
HOG-LDL modulated several genes involved in cholesterol metabolism. IDI1 and SC4MOL, key enzymes in cholesterol biosynthesis, 24 25 were both upregulated. These changes may increase cholesterol synthesis in pericytes exposed to HOG-LDL. Also, cholesterol accumulation is suggested by the upregulation of LDLR 26 and the downregulation of ABCA6, a transporter mediating cholesterol efflux. 27 Consistent with this, by direct measurements, modified LDL is known to increase intracellular cholesterol accumulation in human macrophages and smooth muscle cells. 28 29 LDL from diabetic patients compared with that of healthy subjects induces an increase in intracellular cholesteryl ester in monocyte-macrophages 30 and aortic smooth muscle cells. 31  
Genes in the Fibrinolytic Pathway
PAI-1, a primary regulator of fibrinolysis, is increased in serum, vitreous, subretinal fluid, and retinal microvasculature of diabetic patients (Brignole F, et al. IOVS 1994;35:ARVO Abstract 2041). 32 Overexpression in mouse retinal microvessels thickens basal membranes and increases the endothelial cell/pericyte ratio, reminiscent of diabetic retinopathy. 33 PAI-1 upregulation by HOG-LDL could partially explain the elevation of PAI-1 in diabetes and is in agreement with findings that UV-oxidized LDL enhances PAI-1 expression, both mRNA and protein, in human umbilical vein endothelial cells. 34 Apart from modulating fibrinolysis and extracellular matrix proteolysis, 35 PAI-1 induces apoptosis, 36 although antiapoptotic effects have also been observed. 37  
Genes Involved in Cell Growth and Proliferation
HOG-LDL generally upregulated pericyte growth inhibitors while downregulating growth-promoting genes. Among upregulated genes, P8 mediates growth arrest and apoptosis 38 39 and is over-expressed in response to apoptotic signals. 40 DDK1 antagonizes cell growth induced by WNT proteins, 41 interference with which is implicated in apoptotic degeneration in retinitis pigmentosa. 42  
Of downregulated genes, IGFs and IGFBPs play key roles in the balance between cell proliferation and apoptosis. 43 Reduced expression of IGFBP3/5 after HOG-LDL treatment suggests a role for IGF signaling in pericyte loss. This agrees with findings that oxidized LDL modulates IGF1, IGF1 receptors, and IGFBPs in rat aortic vascular smooth muscle cells, contributing to apoptosis. 44 Decreased serum IGFBP3 has been found in patients with diabetic retinopathy. 45 IGFBP3 is thought to sensitize cells to mitogenic IGF1 stimuli, whereas IGFBP5 stimulates growth of human retinal endothelial cells. 46  
Other downregulated survival-promoting genes that are of interest include AREG (growth factor); TGFB1 (growth factor); and ALDH1A2, RARRES1, and RAI3 (retinoic acid synthesis and response genes). 
Genes Related to Stress Responses
Several stress-responsive genes were modulated by HOG-LDL. ATF3, a stress-inducible transcriptional repressor highly expressed in cells undergoing apoptosis in atherosclerotic lesions, 47 48 was upregulated. It has been found that oxidized LDL induces ATF3 and increases its DNA binding, and that inhibition of ATF3 suppresses oxidized LDL-induced cell death. 48 This implicates ATF3 as an important mediator of apoptosis. 
Thioredoxin-interacting protein (TXNIP) is an endogenous inhibitor of the redox protein thioredoxin. 49 Overexpression of this gene increases oxidative stress, while its silencing restores thioredoxin activity in hyperglycemia. 50 Consequently, a reduced expression of TXNIP may reflect enhanced thioredoxin antioxidative responses. This finding differs from that observed in aorta smooth muscle cells where hyperglycemia inhibits thioredoxin by inducing TXNIP. 51 TXNIP is also induced by glucose incubation in fibroblasts 51 and mesangial cells. 52  
Metallothioneins (MTs) are ubiquitous metal-binding proteins involved in metal homeostasis, free radical scavenging, cell proliferation, and stress-induced apoptosis. 53 Decreased retinal MTs are associated with senescence 54 and macular degeneration, 55 whereas increased MTs protect against oxidative stress and apoptosis. 56 A reduction in expression of MT1B and MT2A in pericytes may reflect an impaired antioxidant defensive mechanism. 
Bradykinin Receptors
Bradykinin’s activity (vasodilation, blood flow, inflammatory responses) is mainly mediated by B2 kinin receptors in the retina. 57 Reduced expression of B2 receptors in pericytes by HOG-LDL may disturb retinal hemodynamics. Recent studies have shown a likely shift of vasodilatory functions from B2 to B1 receptors in retinal microvessels in streptozotocin-diabetic rats, 58 implicating the kinin system in diabetic retinopathy development. 
Genes Related to Angiogenesis
HOG-LDL altered the expression of diverse genes related to angiogenesis. Cysteine-rich angiogenic inducer 61 (CYR61) and angiomotin-like 2 (AMOTL2), both with proangiogenic activity, were upregulated. CYR61 stimulates the migration of human microvascular endothelial cells, induces neovascularization in rat cornea, and regulates genes participating in wound healing. 59 60 AMOTL2 is related to angiomotin and belongs to the motin family. 61 Angiomotin increases endothelial cell migration by binding to angiostatin, an inhibitor of angiogenesis. 62  
In contrast, antiangiogenic genes, including STC1, TIMP3, TGFB1, and COL15A1 were downregulated by HOG-LDL. STC1 is markedly upregulated in angiogenesis models, 63 64 where it inhibits hepatocyte growth factor-induced angiogenesis by interfering with pathways downstream of its receptor. 64 TIMP3 inhibits angiogenesis both in vitro 65 and in vivo in a mouse model of retinopathy 66 by inhibiting VEGF receptor binding and downstream signaling. 67 TGFB1, secreted by pericytes, inhibits endothelial cell growth and migration. 68 69 Its downregulation may promote endothelial cell proliferation and contribute to neovascularization in diabetic retinopathy. 70 Type XV collagen α1 (COL15A1) is the precursor of restin, which is homologous to endostatin and exhibits antiangiogenic effects. 71  
Thus, HOG-LDL appears to shift the balance in favor of angiogenesis by upregulating proangiogenic genes and downregulating antiangiogenic genes. This surprising finding goes against the notion that oxidized LDL impairs endothelial cell growth and generally inhibits angiogenesis. 72 73 However, studies have shown that oxidized LDL upregulates VEGF expression in endothelial cells. 74 The pericyte–endothelial cell relationship is complex and dynamic, in that pericytes may be both negative and positive regulators of endothelial cell proliferation and neovascularization. 75 76 77 78 79 80 Therefore, our findings suggest that, besides causing apoptosis, modified LDL at sublethal concentrations may have proangiogenic effects through alteration of gene expression in retinal pericytes. 
 
Table 1.
 
Genes Upregulated ≥1.7-fold by HOG- Versus N-LDL in Cultured Human Retinal Pericytes
Table 1.
 
Genes Upregulated ≥1.7-fold by HOG- Versus N-LDL in Cultured Human Retinal Pericytes
Gene Gene Symbol Accession Number Change (x-fold) P
Cholesterol, lipid, fatty acid
 Fatty acid desaturase 2 FADS2 AL050118 2.9 0.003
 Fatty acid desaturase 1* FADS1 AF035284 2.8 0.048
 Low-density lipoprotein receptor (familial hypercholesterolemia) LDLR L00352 2.7 0.002
 Fatty acid-binding protein 4, adipocyte* FABP4 AA128249 2.5 0.012
 Sterol C4-methyl oxidase-like* SC4MOL AI535653 2.0 0.091
 Isopentenyl-diphosphate delta isomerase* IDI1 X17025 1.7 0.018
Transcription
 p8 protein (candidate of metastasis 1) P8 AI557295 1.9 0.005
 Activating transcription factor 3* ATF3 L19871 1.7 0.020
Signal transduction
 Regulator of G-protein signaling 20* RGS20 AF060877 1.9 0.028
 Related RAS viral (r-ras) oncogene homolog 2 RRAS2 AI365215 1.7 0.003
Atherosclerosis
 Cysteine-rich, angiogenic inducer, 61 CYR61 Y11307 2.6 0.029
 Pentaxin-related gene, rapidly induced by IL-1 beta PTX3 M31166 1.9 <0.001
 Plasminogen activator inhibitor-1 PAI-1 J03764 1.7 0.008
Other
 Asparagine synthetase* ASNS M27396 2.7 0.045
 Actin, gamma 2, smooth muscle, enteric* ACTG2 D00654 2.0 0.083
 Angiomotin like 2 AMOTL2 AB023206 1.8 0.017
 Poliovirus receptor-related 3 PVRL3 AL050071 1.8 0.022
 Dickkopf homolog 1 (Xenopus laevis) DKK1 AB020315 1.8 0.118
 Deafness, autosomal dominant 5 DFNA5 AF073308 1.7 0.016
 DnaJ (Hsp40) homolog, subfamily B, member 4 DNAJB4 U40992 1.7 0.044
Unknown
 Hypothetical protein LOC339005* LOC339005 AL046961 1.8 0.180
Table 2.
 
Genes Downregulated ≥1.7-fold by HOG- Versus N-LDL in Cultured Human Retinal Pericytes
Table 2.
 
Genes Downregulated ≥1.7-fold by HOG- Versus N-LDL in Cultured Human Retinal Pericytes
Gene Gene Symbol Accession Number Change (x-fold) P
Signal transduction
 Retinoic acid receptor responder 1 RARRES1 AI887421 −6.8 0.013
 Bradykinin receptor B2 BDKRB2 X86163 −3.0 <0.001
 Insulin-like growth factor binding protein 3 IGFBP3 M35878 −2.9 0.001
 Chemokine (C-X-C motif) ligand 12 CXCL12 L36033 −2.2 0.001
 Insulin-like growth factor binding protein 5* IGFBP5 L27560 −2.0 0.020
 Tumor necrosis factor, alpha-induced protein 6* TNFAIP6 M31165 −2.0 0.058
 Stanniocalcin 1 STC1 U25997 −2.0 0.012
 Prostaglandin E synthase PTGES AF010316 −1.9 <0.001
 Protein kinase, cAMP-dependent, regulatory, type II, beta PRKAR2B M31158 −1.8 0.003
Transcription
 Inhibitor of DNA binding 1* ID1 X77956 −4.2 0.040
 Basic transcription element binding protein 1 BTEB1 D31716 −1.8 0.124
 Nuclear transcription factor Y, beta NFYB AA621555 −1.8 0.153
 Thioredoxin interacting protein TXNIP S73591 −1.8 0.036
 Jun B proto-oncogene JUNB X51345 −1.7 0.010
Stress response
 Metallothionein 1F (functional)* MT1F H68340 −1.7 0.044
 Metallothionein 1B (functional) MT1B M13485 −1.7 0.043
Cell cycle
 Retinoic acid induced 3 RAI3 AF095448 −2.0 0.073
 Transforming growth factor, beta-induced, 68kDa TGFB1 M77349 −1.9 0.005
 CDC28 protein kinase regulatory subunit 2 CKS2 X54942 −1.7 0.181
 Kinetochore associated 2 KNTC2 AF017790 −1.7 0.198
Extracellular matrix
 Tissue inhibitor of metalloproteinase 3* TIMP3 U14394 −2.6 0.004
 Collagen, type XV, alpha 1 COL15A1 L25286 −2.1 0.006
Enzyme
 Proprotein convertase subtilisin/kexin type 1* PCSK1 X64810 −3.3 0.001
 Short-chain dehydrogenase/reductase 1 SDR1 AF061741 −2.3 0.134
 Sulfatase 1 SULF1 AB029000 −2.2 0.002
 Aldehyde dehydrogenase 1 family, member A2 ALDH1A2 AB015228 −2.1 0.013
 Flavin containing monooxygenase 3* FMO3 M83772 −2.0 0.215
 Transglutaminase 2 TGM2 M55153 −1.9 0.022
 Sulfotransferase family, cytosolic, 1B, member 1 SULT1B1 D89479 −1.7 0.001
Transporter
 ATP-binding cassette, sub-family A (ABC1), member 6* ABCA6 AI651024 −2.2 0.002
 Aquaporin 9 AQP9 AB008775 −1.9 0.009
Other
 Topoisomerase (DNA) II alpha 170kDa* TOP2A L47276 −1.8 0.050
 Secretogranin II (chromogranin C) SCG2 M25756 −2.5 0.092
 Amphiregulin (schwannoma-derived growth factor) AREG M30704 −2.4 0.019
 Statherin STATH M18078 −2.0 0.157
 Vascular cell adhesion molecule 1* VCAM1 M30257 −1.9 0.111
 ZW10 interactor ZWINT AF067656 −1.7 0.266
Unknown
 PN001_AH_H08.r yodnorm mRNA sequence AI547258 −1.9 0.108
 KIAA0101 KIAA0101 D14657 −1.7 0.229
Figure 1.
 
Hierarchical clustering of genes differentially regulated in response to experimental LDL treatments. With the dChip analysis tool, hierarchical clustering was performed on the 152 genes differentially regulated between the control (SFM) and experimental LDL samples. Expression levels among replicate treatments were averaged before standardization and subsequent distance calculations. Red: relative high expression; blue: relative low expression; white: intermediate expression (see scale bar at the bottom of the cluster). Black vertical bars: sets of genes for which the response to N-LDL and G-LDL was similar but for which the response to HOG-LDL was distinct; green vertical bar: a group of genes that appeared to be specifically affected by HOG-LDL. Genes presented in the heat diagram are listed in Supplementary Table S1.
Figure 1.
 
Hierarchical clustering of genes differentially regulated in response to experimental LDL treatments. With the dChip analysis tool, hierarchical clustering was performed on the 152 genes differentially regulated between the control (SFM) and experimental LDL samples. Expression levels among replicate treatments were averaged before standardization and subsequent distance calculations. Red: relative high expression; blue: relative low expression; white: intermediate expression (see scale bar at the bottom of the cluster). Black vertical bars: sets of genes for which the response to N-LDL and G-LDL was similar but for which the response to HOG-LDL was distinct; green vertical bar: a group of genes that appeared to be specifically affected by HOG-LDL. Genes presented in the heat diagram are listed in Supplementary Table S1.
Figure 2.
 
General categorization of the 60 genes with expression that was significantly altered by HOG- versus N-LDL (100 mg/L, 24 hours) in cultured human retinal pericytes. Genes were identified as differentially regulated when average expression levels changed by ≥1.7-fold compared with N-LDL treatment. Categorization was based on information gathered from a literature search and online databases (i.e., OMIM; Online Mendelian Inheritance in Man; http://www.ncbi.nlm.nih.gov/Omim/ provided in the public domain by the National Center for Biotechnology Information, Bethesda, MD; and The Nucleotide Database, a collaboration among the tripartite, international collaboration of sequence databases: GenBank, the National Institutes of Health [NIH], Bethesda, MD; European Molecular Biology Laboratory [EMBL], Hinxton Hall, UK; and DNA Database of Japan [DDBJ], Mishima, Japan).
Figure 2.
 
General categorization of the 60 genes with expression that was significantly altered by HOG- versus N-LDL (100 mg/L, 24 hours) in cultured human retinal pericytes. Genes were identified as differentially regulated when average expression levels changed by ≥1.7-fold compared with N-LDL treatment. Categorization was based on information gathered from a literature search and online databases (i.e., OMIM; Online Mendelian Inheritance in Man; http://www.ncbi.nlm.nih.gov/Omim/ provided in the public domain by the National Center for Biotechnology Information, Bethesda, MD; and The Nucleotide Database, a collaboration among the tripartite, international collaboration of sequence databases: GenBank, the National Institutes of Health [NIH], Bethesda, MD; European Molecular Biology Laboratory [EMBL], Hinxton Hall, UK; and DNA Database of Japan [DDBJ], Mishima, Japan).
Figure 3.
 
Verification of gene expression in response to HOG-LDL by real-time RT-PCR and evaluation of the effect of N-LDL on HOG-LDL-elicited alterations of gene expression in cultured human retinal pericytes. Data represent the mean ± SD of results in triplicate experiments. *P < 0.05; **P < 0.01, versus N-LDL. Consistent with gene array findings, HOG-LDL (100 mg/L, 24 hours) significantly upregulated FABP4 and LDLR, and downregulated BDKRB2 and IGFBP3. Co-incubation with N-LDL (100 mg/L, 24 hours) reversed the HOG-LDL-induced changes in expression of LDLR, BDKRB2, and IGFBP3, but not of FABP4.
Figure 3.
 
Verification of gene expression in response to HOG-LDL by real-time RT-PCR and evaluation of the effect of N-LDL on HOG-LDL-elicited alterations of gene expression in cultured human retinal pericytes. Data represent the mean ± SD of results in triplicate experiments. *P < 0.05; **P < 0.01, versus N-LDL. Consistent with gene array findings, HOG-LDL (100 mg/L, 24 hours) significantly upregulated FABP4 and LDLR, and downregulated BDKRB2 and IGFBP3. Co-incubation with N-LDL (100 mg/L, 24 hours) reversed the HOG-LDL-induced changes in expression of LDLR, BDKRB2, and IGFBP3, but not of FABP4.
Supplementary Materials
The authors thank the MUSC DNA Microarray Facility, a shared-resources facility, for services provided during the study. 
BrownleeM. Advanced protein glycosylation in diabetes and aging. Annu Rev Med. 1995;46:223–234. [CrossRef] [PubMed]
KennedyAL, LyonsTJ. Glycation, oxidation, and lipoxidation in the development of diabetic complications. Metabolism. 1997;46(suppl 1)14–21. [CrossRef] [PubMed]
BarberAJ. A new view of diabetic retinopathy: a neurodegenerative disease of the eye. Prog Neuropsychopharmacol Biol Psychiatry. 2003;27:283–290. [CrossRef] [PubMed]
LyonsTJ, LiW, Wells-KnechtMC, JoklR. Toxicity of mildly modified low-density lipoproteins to cultured retinal capillary endothelial cells and pericytes. Diabetes. 1994;43:1090–1095. [CrossRef] [PubMed]
SongW, BarthJL, LuK, et al. Effects of modified LDL on human retinal pericyte survival. Ann NY Acad Sci. .In press.
MizutaniM, KernTS, LorenziM. Accelerated death of retinal microvascular cells in human and experimental diabetic retinopathy. J Clin Invest. 1996;97:2883–2890. [CrossRef] [PubMed]
JenkinsAJ, VelardeV, KleinRL, et al. Native and modified LDL activate extracellular signal-regulated kinases in mesangial cells. Diabetes. 2000;49:2160–2169. [CrossRef] [PubMed]
GolubTR, SlonimDK, TamayoP, et al. Molecular classification of cancer: class discovery and class prediction by gene expression monitoring. Science. 1999;286:531–537. [CrossRef] [PubMed]
TusherVG, TibshiraniR, ChuG. Significance analysis of microarrays applied to the ionizing radiation response. Proc Natl Acad Sci. 2001;98:5116–5121. [CrossRef] [PubMed]
NayakRC, AgardhCD, KwokMG, et al. Circulating anti-pericyte autoantibodies are present in Type 2 diabetic patients and are associated with non-proliferative retinopathy. Diabetologia. 2003;46:511–513. [PubMed]
MarquardtA, StohrH, WhiteK, WeberBH. cDNA cloning, genomic structure, and chromosomal localization of three members of the human fatty acid desaturase family. Genomics. 2000;66:175–183. [CrossRef] [PubMed]
BoordJB, MaedaK, MakowskiL, et al. Adipocyte fatty acid-binding protein, aP2, alters late atherosclerotic lesion formation in severe hypercholesterolemia. Arterioscler Thromb Vasc Biol. 2002;22:1686–1691. [CrossRef] [PubMed]
FunkCD. Prostaglandins and leukotrienes: advances in eicosanoid biology. Science. 2001;294:1871–1875. [CrossRef] [PubMed]
PaniniSR, YangL, RusinolAE, SinenskyMS, BonventreJV, LeslieCC. Arachidonate metabolism and the signaling pathway of induction of apoptosis by oxidized LDL/oxysterol. J Lipid Res. 2001;42:1678–1686. [PubMed]
OzakiM, YamadaY, MatobaK, et al. Phospholipase A2 activity in ox-LDL-stimulated mesangial cells and modulation by alpha-tocopherol. Kidney Int. 1999;71:S171–S173.
NorataGD, PirilloA, PellegattaF, InoueH, CatapanoAL. Native LDL and oxidized LDL modulate cyclooxygenase-2 expression in HUVECs through a p38-MAPK, NF-kappaB, CRE dependent pathway and affect PGE2 synthesis. Int J Mol Med. 2004;14:353–359. [PubMed]
JoussenAM, PoulakiV, LeML, et al. A central role for inflammation in the pathogenesis of diabetic retinopathy. FASEB J. 2004;18:1450–1452. [PubMed]
MohrS. Potential new strategies to prevent the development of diabetic retinopathy. Expert Opin Investig Drugs. 2004;13:189–198. [CrossRef] [PubMed]
TongPC, LeeKF, SoWY, et al. White blood cell count is associated with macro- and microvascular complications in chinese patients with type 2 diabetes. Diabetes Care. 2004;27:216–222. [CrossRef] [PubMed]
KloucheM, PeriG, KnabbeC, et al. Modified atherogenic lipoproteins induce expression of pentraxin-3 by human vascular smooth muscle cells. Atherosclerosis. 2004;175:221–228. [CrossRef] [PubMed]
RolphMS, ZimmerS, BottazziB, GarlandaC, MantovaniA, HanssonGK. Production of the long pentraxin PTX3 in advanced atherosclerotic plaques. Arterioscler Thromb Vasc Biol. 2002;22:E10–E14. [CrossRef] [PubMed]
WisniewskiHG, VilcekJ. TSG-6: an IL-1/TNF-inducible protein with anti-inflammatory activity. Cytokine Growth Factor Rev. 1997;8:143–156. [CrossRef] [PubMed]
SzantoS, BardosT, GalI, GlantTT, MikeczK. Enhanced neutrophil extravasation and rapid progression of proteoglycan-induced arthritis in TSG-6-knockout mice. Arthritis Rheum. 2004;50:3012–3022. [CrossRef] [PubMed]
SteinerS, GatlinCL, LennonJJ, et al. Proteomics to display lovastatin-induced protein and pathway regulation in rat liver. Electrophoresis. 2000;21:2129–2137. [CrossRef] [PubMed]
RodriguezC, RaposoB, Martinez-GonzalezJ, Llorente-CortesV, VilahurG, BadimonL. Modulation of ERG25 expression by LDL in vascular cells. Cardiovasc Res. 2003;58:178–185. [CrossRef] [PubMed]
HofmannSL, RussellDW, BrownMS, GoldsteinJL, HammerRE. Overexpression of low density lipoprotein (LDL) receptor eliminates LDL from plasma in transgenic mice. Science. 1988;239:1277–1281. [CrossRef] [PubMed]
KaminskiWE, WenzelJJ, PiehlerA, LangmannT, SchmitzG. ABCA6, a novel a subclass ABC transporter. Biochem Biophys Res Commun. 2001;285:1295–1301. [CrossRef] [PubMed]
TraoreM, JialingH, ChunlingX. Influence of Angelicae sinensis extract on lipid accumulation in rabbit aortic smooth muscle cells induced by oxidized LDL. Biorheology. 2003;40:389–394. [PubMed]
Lopes-VirellaMF, KleinRL, LyonsTJ, StevensonHC, WitztumJL. Glycosylation of low-density lipoprotein enhances cholesteryl ester synthesis in human monocyte-derived macrophages. Diabetes. 1988;37:550–557. [CrossRef] [PubMed]
LyonsTJ, KleinRL, BaynesJW, StevensonHC, Lopes-VirellaMF. Stimulation of cholesteryl ester synthesis in human monocyte-derived macrophages by low-density lipoproteins from type 1 (insulin-dependent) diabetic patients: the influence of non-enzymatic glycosylation of low-density lipoproteins. Diabetologia. 1987;30:916–923. [CrossRef] [PubMed]
TertovVV, SobeninIA, GabbasovZA, et al. Three types of naturally occurring modified lipoproteins induce intracellular lipid accumulation in human aortic intimal cells: the role of lipoprotein aggregation. Eur J Clin Chem Clin Biochem. 1992;30:171–178. [PubMed]
GrantMB, EllisEA, CaballeroS, MamesRN. Plasminogen activator inhibitor-1 overexpression in nonproliferative diabetic retinopathy. Exp Eye Res. 1996;63:233–244. [CrossRef] [PubMed]
GrantMB, SpoerriPE, PlayerDW, et al. Plasminogen activator inhibitor (PAI)-1 overexpression in retinal microvessels of PAI-1 transgenic mice. Invest Ophthalmol Vis Sci. 2000;41:2296–2302. [PubMed]
AllisonBA, NilssonL, KarpeF, HamstenA, ErikssonP. Effects of native, triglyceride-enriched, and oxidatively modified LDL on plasminogen activator inhibitor-1 expression in human endothelial cells. Arterioscler Thromb Vasc Biol. 1999;19:1354–1360. [CrossRef] [PubMed]
SakselaO, RifkinDB. Cell-associated plasminogen activation: regulation and physiological functions. Annu Rev Cell Biol. 1988;4:93–126. [CrossRef] [PubMed]
Al-FakhriN, ChavakisT, Schmidt-WollT, et al. Induction of apoptosis in vascular cells by plasminogen activator inhibitor-1 and high molecular weight kininogen correlates with their anti-adhesive properties. Biol Chem. 2003;384:423–435. [PubMed]
ChenY, KelmRJ, Jr, BuddRC, SobelBE, SchneiderDJ. Inhibition of apoptosis and caspase-3 in vascular smooth muscle cells by plasminogen activator inhibitor type-1. J Cell Biochem. 2004;92:178–188. [CrossRef] [PubMed]
SuSB, MotooY, IovannaJL, et al. Overexpression of p8 is inversely correlated with apoptosis in pancreatic cancer. Clin Cancer Res. 2001;7:1320–1324. [PubMed]
VasseurS, HoffmeisterA, Garcia-MonteroA, et al. p8-deficient fibroblasts grow more rapidly and are more resistant to Adriamycin-induced apoptosis. Oncogene. 2002;21:1685–1694. [CrossRef] [PubMed]
MalloGV, FiedlerF, CalvoEL, et al. Cloning and expression of the rat p8 cDNA, a new gene activated in pancreas during the acute phase of pancreatitis, pancreatic development, and regeneration, and which promotes cellular growth. J Biol Chem. 1997;272:32360–32369. [CrossRef] [PubMed]
FediP, BaficoA, Nieto SoriaA, AaronsonSA, et al. Isolation and biochemical characterization of the human Dkk-1 homologue, a novel inhibitor of mammalian Wnt signaling. J Biol Chem. 1999;274:19465–19472. [CrossRef] [PubMed]
JonesSE, JomaryC, GristJ, StewartHJ, NealMJ. Modulated expression of secreted frizzled-related proteins in human retinal degeneration. Neuroreport. 2000;11:3963–3967. [CrossRef] [PubMed]
KelleyKM, OhY, GargoskySE, et al. Insulin-like growth factor-binding proteins (IGFBPs) and their regulatory dynamics. Int J Biochem Cell Biol. 1996;28:619–637. [CrossRef] [PubMed]
ScheideggerKJ, JamesRW, DelafontaineP. Differential effects of low density lipoproteins on insulin-like growth factor-1 (IGF-1) and IGF-1 receptor expression in vascular smooth muscle cells. J Biol Chem. 2000;275:26864–26869. [PubMed]
FeldmannB, LangGE, ArnavazA, JehlePM, BohmBO, LangGK. Decreased serum level of free bioavailable IGF-I in patients with diabetic retinopathy. Ophthalmologe. 1999;96:300–305. [CrossRef] [PubMed]
GianniniS, CresciB, PalaL, et al. IGFBPs modulate IGF-I- and high glucose-controlled growth of human retinal endothelial cells. J Endocrinol. 2001;171:273–284. [CrossRef] [PubMed]
HaiT, WolfgangCD, MarseeDK, AllenAE, SivaprasadU. ATF3 and stress responses. Gene Expr. 1999;7:321–335. [PubMed]
NawaT, NawaMT, AdachiMT, et al. Expression of transcriptional repressor ATF3/LRF1 in human atherosclerosis: colocalization and possible involvement in cell death of vascular endothelial cells. Atherosclerosis. 2002;161:281–291. [CrossRef] [PubMed]
NishiyamaA, MatsuiM, IwataS, et al. Identification of thioredoxin-binding protein-2/vitamin D(3) up-regulated protein 1 as a negative regulator of thioredoxin function and expression. J Biol Chem. 1999;274:21645–21650. [CrossRef] [PubMed]
SchulzePC, YoshiokaJ, TakahashiT, HeZ, KingGL, LeeRT. Hyperglycemia promotes oxidative stress through inhibition of thioredoxin function by thioredoxin-interacting protein. J Biol Chem. 2004;279:30369–30374. [CrossRef] [PubMed]
HirotaT, OkanoT, KokameK, Shirotani-IkejimaH, MiyataT, FukadaY. Glucose down-regulates Per1 and Per2 mRNA levels and induces circadian gene expression in cultured Rat-1 fibroblasts. J Biol Chem. 2002;277:44244–44251. [CrossRef] [PubMed]
KobayashiT, UeharaS, IkedaT, ItadaniH, KotaniH. Vitamin D3 up-regulated protein-1 regulates collagen expression in mesangial cells. Kidney Int. 2003;64:1632–1642. [CrossRef] [PubMed]
MilesAT, HawksworthGM, BeattieJH, RodillaV. Induction, regulation, degradation, and biological significance of mammalian metallothioneins. Crit Rev Biochem Mol Biol. 2000;35:35–70. [CrossRef] [PubMed]
TateDJ, Jr, NewsomeDA, OliverPD. Metallothionein shows an age-related decrease in human macular retinal pigment epithelium. Invest Ophthalmol Vis Sci. 1993;34:2348–2351. [PubMed]
NicolasMG, FujikiK, MurayamaK, et al. Studies on the mechanism of early onset macular degeneration in cynomolgus monkeys. II. Suppression of metallothionein synthesis in the retina in oxidative stress. Exp Eye Res. 1996;62:399–408. [CrossRef] [PubMed]
LuH, HuntDM, GantiR, et al. Metallothionein protects retinal pigment epithelial cells against apoptosis and oxidative stress. Exp Eye Res. 2002;74:83–92. [CrossRef] [PubMed]
BhoolaKD, FigueroaCD, WorthyK. Bioregulation of kinins: kallikreins, kininogens, and kininases. Pharmacol Rev. 1992;44:1–80. [PubMed]
AbdouhM, KhanjariA, AbdelazzizN, OngaliB, CoutureR, HassessianHM. Early upregulation of kinin B1 receptors in retinal microvessels of the streptozotocin-diabetic rat. Br J Pharmacol. 2003;140:33–40. [CrossRef] [PubMed]
BabicAM, KireevaML, KolesnikovaTV, LauLF. CYR61, a product of a growth factor-inducible immediate early gene, promotes angiogenesis and tumor growth. Proc Natl Acad Sci. 1998;95:6355–6360. [CrossRef] [PubMed]
ChenCC, MoFE, LauLF. The angiogenic factor Cyr61 activates a genetic program for wound healing in human skin fibroblasts. J Biol Chem. 2001;276:47329–47337. [CrossRef] [PubMed]
BrattA, WilsonWJ, TroyanovskyB, et al. Angiomotin belongs to a novel protein family with conserved coiled-coil and PDZ binding domains. Gene. 2002;298:69–77. [CrossRef] [PubMed]
TroyanovskyB, LevchenkoT, ManssonG, MatvijenkoO, HolmgrenL. Angiomotin: an angiostatin binding protein that regulates endothelial cell migration and tube formation. J Cell Biol. 2001;152:1247–1254. [CrossRef] [PubMed]
GerritsenME, SorianoR, YangS, et al. In silico data filtering to identify new angiogenesis targets from a large in vitro gene profiling data set. Physiol Genomics. 2002;10:13–20. [CrossRef] [PubMed]
ZlotC, IngleG, HongoJ, et al. Stanniocalcin 1 is an autocrine modulator of endothelial angiogenic responses to hepatocyte growth factor. J Biol Chem. 2003;278:47654–47659. [CrossRef] [PubMed]
SunJX, ZhangCX, ChenZZ, WuXF. Cloning and expression of the cDNA encoding human tissue inhibitor of metalloproteinase-3 and its inhibition on angiogenesis [in Chinese]. Sheng Wu Hua Xue Yu Sheng Wu Wu Li Xue Bao. 1998;30:220–224. [PubMed]
AuricchioA, BehlingKC, MaguireAM, et al. Inhibition of retinal neovascularization by intraocular viral-mediated delivery of anti-angiogenic agents. Mol Ther. 2002;6:490–494. [CrossRef] [PubMed]
QiJH, EbrahemQ, MooreN, et al. A novel function for tissue inhibitor of metalloproteinases-3 (TIMP3): inhibition of angiogenesis by blockage of VEGF binding to VEGF receptor-2. Nat Med. 2003;9:407–415. [CrossRef] [PubMed]
Antonelli-OrlidgeA, SaundersKB, SmithSR, D’AmorePA. An activated form of transforming growth factor beta is produced by cocultures of endothelial cells and pericytes. Proc Natl Acad Sci. 1989;86:4544–4548. [CrossRef] [PubMed]
SatoY, RifkinDB. Inhibition of endothelial cell movement by pericytes and smooth muscle cells: activation of a latent transforming growth factor-beta 1-like molecule by plasmin during co-culture. J Cell Biol. 1989;109:309–315. [CrossRef] [PubMed]
SprangerJ, Meyer-SchwickerathR, KleinM, SchatzH, PfeifferA. Deficient activation and different expression of transforming growth factor-beta isoforms in active proliferative diabetic retinopathy and neovascular eye disease. Exp Clin Endocrinol Diabetes. 1999;107:21–28. [CrossRef] [PubMed]
RamchandranR, DhanabalM, VolkR, et al. Antiangiogenic activity of restin, NC10 domain of human collagen XV: comparison to endostatin. Biochem Biophys Res Commun. 1999;255:735–739. [CrossRef] [PubMed]
ChenCH, JiangW, ViaDP, et al. Oxidized low-density lipoproteins inhibit endothelial cell proliferation by suppressing basic fibroblast growth factor expression. Circulation. 2000;101:171–177. [CrossRef] [PubMed]
ImanishiT, HanoT, MatsuoY, NishioI. Oxidized low-density lipoprotein inhibits vascular endothelial growth factor-induced endothelial progenitor cell differentiation. Clin Exp Pharmacol Physiol. 2003;30:665–670. [CrossRef] [PubMed]
InoueM, ItohH, TanakaT, et al. Oxidized LDL regulates vascular endothelial growth factor expression in human macrophages and endothelial cells through activation of peroxisome proliferator-activated receptor-gamma. Arterioscler Thromb Vasc Biol. 2001;21:560–566. [CrossRef] [PubMed]
KuwabaraT, CoganDG. Retinal vascular patterns. VI. Mural cells of the retinal capillaries. Arch Ophthalmol. 1963;69:492–502. [CrossRef] [PubMed]
De OliveiraF. Pericytes in diabetic retinopathy. Br J Ophthalmol. 1966;50:134–143. [CrossRef] [PubMed]
CrockerDJ, MuradTM, GeerJC. Role of the pericyte in wound healing: an ultrastructural study. Exp Mol Pathol. 1970;13:51–65. [CrossRef] [PubMed]
OrlidgeA, D’AmorePA. Inhibition of capillary endothelial cell growth by pericytes and smooth muscle cells. J Cell Biol. 1987;105:1455–1462. [CrossRef] [PubMed]
EngeM, BjarnegardM, GerhardtH, et al. Endothelium-specific platelet-derived growth factor-B ablation mimics diabetic retinopathy. EMBO J. 2002;21:4307–4316. [CrossRef] [PubMed]
GerhardtH, BetsholtzC. Endothelial-pericyte interactions in angiogenesis. Cell Tissue Res. 2003;314:15–23. [CrossRef] [PubMed]
Figure 1.
 
Hierarchical clustering of genes differentially regulated in response to experimental LDL treatments. With the dChip analysis tool, hierarchical clustering was performed on the 152 genes differentially regulated between the control (SFM) and experimental LDL samples. Expression levels among replicate treatments were averaged before standardization and subsequent distance calculations. Red: relative high expression; blue: relative low expression; white: intermediate expression (see scale bar at the bottom of the cluster). Black vertical bars: sets of genes for which the response to N-LDL and G-LDL was similar but for which the response to HOG-LDL was distinct; green vertical bar: a group of genes that appeared to be specifically affected by HOG-LDL. Genes presented in the heat diagram are listed in Supplementary Table S1.
Figure 1.
 
Hierarchical clustering of genes differentially regulated in response to experimental LDL treatments. With the dChip analysis tool, hierarchical clustering was performed on the 152 genes differentially regulated between the control (SFM) and experimental LDL samples. Expression levels among replicate treatments were averaged before standardization and subsequent distance calculations. Red: relative high expression; blue: relative low expression; white: intermediate expression (see scale bar at the bottom of the cluster). Black vertical bars: sets of genes for which the response to N-LDL and G-LDL was similar but for which the response to HOG-LDL was distinct; green vertical bar: a group of genes that appeared to be specifically affected by HOG-LDL. Genes presented in the heat diagram are listed in Supplementary Table S1.
Figure 2.
 
General categorization of the 60 genes with expression that was significantly altered by HOG- versus N-LDL (100 mg/L, 24 hours) in cultured human retinal pericytes. Genes were identified as differentially regulated when average expression levels changed by ≥1.7-fold compared with N-LDL treatment. Categorization was based on information gathered from a literature search and online databases (i.e., OMIM; Online Mendelian Inheritance in Man; http://www.ncbi.nlm.nih.gov/Omim/ provided in the public domain by the National Center for Biotechnology Information, Bethesda, MD; and The Nucleotide Database, a collaboration among the tripartite, international collaboration of sequence databases: GenBank, the National Institutes of Health [NIH], Bethesda, MD; European Molecular Biology Laboratory [EMBL], Hinxton Hall, UK; and DNA Database of Japan [DDBJ], Mishima, Japan).
Figure 2.
 
General categorization of the 60 genes with expression that was significantly altered by HOG- versus N-LDL (100 mg/L, 24 hours) in cultured human retinal pericytes. Genes were identified as differentially regulated when average expression levels changed by ≥1.7-fold compared with N-LDL treatment. Categorization was based on information gathered from a literature search and online databases (i.e., OMIM; Online Mendelian Inheritance in Man; http://www.ncbi.nlm.nih.gov/Omim/ provided in the public domain by the National Center for Biotechnology Information, Bethesda, MD; and The Nucleotide Database, a collaboration among the tripartite, international collaboration of sequence databases: GenBank, the National Institutes of Health [NIH], Bethesda, MD; European Molecular Biology Laboratory [EMBL], Hinxton Hall, UK; and DNA Database of Japan [DDBJ], Mishima, Japan).
Figure 3.
 
Verification of gene expression in response to HOG-LDL by real-time RT-PCR and evaluation of the effect of N-LDL on HOG-LDL-elicited alterations of gene expression in cultured human retinal pericytes. Data represent the mean ± SD of results in triplicate experiments. *P < 0.05; **P < 0.01, versus N-LDL. Consistent with gene array findings, HOG-LDL (100 mg/L, 24 hours) significantly upregulated FABP4 and LDLR, and downregulated BDKRB2 and IGFBP3. Co-incubation with N-LDL (100 mg/L, 24 hours) reversed the HOG-LDL-induced changes in expression of LDLR, BDKRB2, and IGFBP3, but not of FABP4.
Figure 3.
 
Verification of gene expression in response to HOG-LDL by real-time RT-PCR and evaluation of the effect of N-LDL on HOG-LDL-elicited alterations of gene expression in cultured human retinal pericytes. Data represent the mean ± SD of results in triplicate experiments. *P < 0.05; **P < 0.01, versus N-LDL. Consistent with gene array findings, HOG-LDL (100 mg/L, 24 hours) significantly upregulated FABP4 and LDLR, and downregulated BDKRB2 and IGFBP3. Co-incubation with N-LDL (100 mg/L, 24 hours) reversed the HOG-LDL-induced changes in expression of LDLR, BDKRB2, and IGFBP3, but not of FABP4.
Table 1.
 
Genes Upregulated ≥1.7-fold by HOG- Versus N-LDL in Cultured Human Retinal Pericytes
Table 1.
 
Genes Upregulated ≥1.7-fold by HOG- Versus N-LDL in Cultured Human Retinal Pericytes
Gene Gene Symbol Accession Number Change (x-fold) P
Cholesterol, lipid, fatty acid
 Fatty acid desaturase 2 FADS2 AL050118 2.9 0.003
 Fatty acid desaturase 1* FADS1 AF035284 2.8 0.048
 Low-density lipoprotein receptor (familial hypercholesterolemia) LDLR L00352 2.7 0.002
 Fatty acid-binding protein 4, adipocyte* FABP4 AA128249 2.5 0.012
 Sterol C4-methyl oxidase-like* SC4MOL AI535653 2.0 0.091
 Isopentenyl-diphosphate delta isomerase* IDI1 X17025 1.7 0.018
Transcription
 p8 protein (candidate of metastasis 1) P8 AI557295 1.9 0.005
 Activating transcription factor 3* ATF3 L19871 1.7 0.020
Signal transduction
 Regulator of G-protein signaling 20* RGS20 AF060877 1.9 0.028
 Related RAS viral (r-ras) oncogene homolog 2 RRAS2 AI365215 1.7 0.003
Atherosclerosis
 Cysteine-rich, angiogenic inducer, 61 CYR61 Y11307 2.6 0.029
 Pentaxin-related gene, rapidly induced by IL-1 beta PTX3 M31166 1.9 <0.001
 Plasminogen activator inhibitor-1 PAI-1 J03764 1.7 0.008
Other
 Asparagine synthetase* ASNS M27396 2.7 0.045
 Actin, gamma 2, smooth muscle, enteric* ACTG2 D00654 2.0 0.083
 Angiomotin like 2 AMOTL2 AB023206 1.8 0.017
 Poliovirus receptor-related 3 PVRL3 AL050071 1.8 0.022
 Dickkopf homolog 1 (Xenopus laevis) DKK1 AB020315 1.8 0.118
 Deafness, autosomal dominant 5 DFNA5 AF073308 1.7 0.016
 DnaJ (Hsp40) homolog, subfamily B, member 4 DNAJB4 U40992 1.7 0.044
Unknown
 Hypothetical protein LOC339005* LOC339005 AL046961 1.8 0.180
Table 2.
 
Genes Downregulated ≥1.7-fold by HOG- Versus N-LDL in Cultured Human Retinal Pericytes
Table 2.
 
Genes Downregulated ≥1.7-fold by HOG- Versus N-LDL in Cultured Human Retinal Pericytes
Gene Gene Symbol Accession Number Change (x-fold) P
Signal transduction
 Retinoic acid receptor responder 1 RARRES1 AI887421 −6.8 0.013
 Bradykinin receptor B2 BDKRB2 X86163 −3.0 <0.001
 Insulin-like growth factor binding protein 3 IGFBP3 M35878 −2.9 0.001
 Chemokine (C-X-C motif) ligand 12 CXCL12 L36033 −2.2 0.001
 Insulin-like growth factor binding protein 5* IGFBP5 L27560 −2.0 0.020
 Tumor necrosis factor, alpha-induced protein 6* TNFAIP6 M31165 −2.0 0.058
 Stanniocalcin 1 STC1 U25997 −2.0 0.012
 Prostaglandin E synthase PTGES AF010316 −1.9 <0.001
 Protein kinase, cAMP-dependent, regulatory, type II, beta PRKAR2B M31158 −1.8 0.003
Transcription
 Inhibitor of DNA binding 1* ID1 X77956 −4.2 0.040
 Basic transcription element binding protein 1 BTEB1 D31716 −1.8 0.124
 Nuclear transcription factor Y, beta NFYB AA621555 −1.8 0.153
 Thioredoxin interacting protein TXNIP S73591 −1.8 0.036
 Jun B proto-oncogene JUNB X51345 −1.7 0.010
Stress response
 Metallothionein 1F (functional)* MT1F H68340 −1.7 0.044
 Metallothionein 1B (functional) MT1B M13485 −1.7 0.043
Cell cycle
 Retinoic acid induced 3 RAI3 AF095448 −2.0 0.073
 Transforming growth factor, beta-induced, 68kDa TGFB1 M77349 −1.9 0.005
 CDC28 protein kinase regulatory subunit 2 CKS2 X54942 −1.7 0.181
 Kinetochore associated 2 KNTC2 AF017790 −1.7 0.198
Extracellular matrix
 Tissue inhibitor of metalloproteinase 3* TIMP3 U14394 −2.6 0.004
 Collagen, type XV, alpha 1 COL15A1 L25286 −2.1 0.006
Enzyme
 Proprotein convertase subtilisin/kexin type 1* PCSK1 X64810 −3.3 0.001
 Short-chain dehydrogenase/reductase 1 SDR1 AF061741 −2.3 0.134
 Sulfatase 1 SULF1 AB029000 −2.2 0.002
 Aldehyde dehydrogenase 1 family, member A2 ALDH1A2 AB015228 −2.1 0.013
 Flavin containing monooxygenase 3* FMO3 M83772 −2.0 0.215
 Transglutaminase 2 TGM2 M55153 −1.9 0.022
 Sulfotransferase family, cytosolic, 1B, member 1 SULT1B1 D89479 −1.7 0.001
Transporter
 ATP-binding cassette, sub-family A (ABC1), member 6* ABCA6 AI651024 −2.2 0.002
 Aquaporin 9 AQP9 AB008775 −1.9 0.009
Other
 Topoisomerase (DNA) II alpha 170kDa* TOP2A L47276 −1.8 0.050
 Secretogranin II (chromogranin C) SCG2 M25756 −2.5 0.092
 Amphiregulin (schwannoma-derived growth factor) AREG M30704 −2.4 0.019
 Statherin STATH M18078 −2.0 0.157
 Vascular cell adhesion molecule 1* VCAM1 M30257 −1.9 0.111
 ZW10 interactor ZWINT AF067656 −1.7 0.266
Unknown
 PN001_AH_H08.r yodnorm mRNA sequence AI547258 −1.9 0.108
 KIAA0101 KIAA0101 D14657 −1.7 0.229
Supplementary Table S1
×
×

This PDF is available to Subscribers Only

Sign in or purchase a subscription to access this content. ×

You must be signed into an individual account to use this feature.

×