Disordered neovascularization and impaired wound healing are important contributors to diabetic vascular complications. We recently showed that high-density lipoproteins (HDLs) enhance ischemia-mediated neovascularization, and mounting evidence suggests HDL have antidiabetic properties. We therefore hypothesized that HDL rescue diabetes-impaired neovascularization. Streptozotocin-induced diabetic mice had reduced blood flow recovery and neovessel formation in a hindlimb ischemia model compared with nondiabetic mice. Reconstituted HDL (rHDL) infusions in diabetic mice restored blood flow recovery and capillary density to nondiabetic levels. Topical rHDL application rescued diabetes-impaired wound closure, wound angiogenesis, and capillary density. In vitro, rHDL increased key mediators involved in hypoxia-inducible factor-1α (HIF-1α) stabilization, including the phosphoinositide 3-kinase/Akt pathway, Siah1, and Siah2, and suppressed the prolyl hydroxylases (PHD) 2 and PHD3. rHDL rescued high glucose–induced impairment of tubulogenesis and vascular endothelial growth factor (VEGF) A protein production, a finding associated with enhanced phosphorylation of proangiogenic mediators VEGF receptor 2 and endothelial nitric oxide synthase. Siah1/2 small interfering RNA knockdown confirmed the importance of HIF-1α stability in mediating rHDL action. Lentiviral short hairpin RNA knockdown of scavenger receptor class B type I (SR-BI) in vitro and SR-BI−/− diabetic mice in vivo attenuated rHDL rescue of diabetes-impaired angiogenesis, indicating a key role for SR-BI. These findings provide a greater understanding of the vascular biological effects of HDL, with potential therapeutic implications for diabetic vascular complications.

The vascular complications of diabetes are characterized by disordered angiogenesis and impairment of ischemia-induced neovascularization. Sufferers of diabetes have reduced coronary collateral formation following vascular occlusion (1), impaired wound healing, and increased rates of amputation (2). Despite advances in the treatment of athero-occlusive disease and the demonstration that intensive blood glucose control attenuates some vascular complications, many patients with diabetes and vasculopathy remain refractory to current treatment approaches.

Diabetes-impaired ischemia-induced neovascularization is associated with decreased hypoxia inducible factor-1α (HIF-1α) stability (3,4), reduced vascular endothelial growth factor (VEGF; specifically VEGFA) production and signaling via VEGF receptor 2 (VEGFR2) (5), and inhibition of endothelial nitric oxide synthase (eNOS) activity (6). However, the key triggers for these events are incompletely understood, as are potential therapies to minimize these abnormalities.

To date, preclinical studies have consistently demonstrated that high-density lipoprotein (HDL) and its main protein constituent, apolipoprotein (apo)A-I, exert antiatherogenic effects (7). Despite this, randomized trials of HDL-raising therapies have not demonstrated clinical benefit. However, long-term mortality follow-up of the Helsinki Heart study found that the incidence of coronary heart disease was significantly reduced when HDL levels were raised by only 5–10% (8), indicating that the biology of HDL is yet to be fully elucidated, and perhaps aggressive attempts to raise HDL have other effects at the cellular and molecular level. Increasing evidence demonstrates that HDL exerts endothelial protective and antidiabetic effects. Infusions of reconstituted HDL (rHDL) reduce plasma glucose levels, restore impaired endothelial function (9), and promote endothelial progenitor cell mobilization (10) in patients with type 2 diabetes. HDL interacts with the cholesterol transporters ABCA1 and ABCG1 and scavenger receptor class B type I (SR-BI). Although ABCA1 and ABCG1 are predominantly involved in HDL-mediated cholesterol efflux, SR-BI is known to mediate the vasculoprotective effects of HDL, including increasing re-endothelialization and endothelial cell migration (11,12). Despite a significant amount of work, the vascular biological effects of HDL remain incompletely understood and further efforts to translate HDL into a potential therapeutic agent require a fuller understanding of its properties.

We therefore sought to investigate the effect of rHDL on diabetes-impaired angiogenesis in two murine models of diabetic vascular complications and to elucidate the mechanisms of action. We report that rHDL rescues diabetes-related impairment of ischemia-driven angiogenesis and wound healing. This occurs via the receptor SR-BI and by HIF-1α stabilization, enhanced VEGFA/VEGFR2 production and signaling, and increased eNOS activity. These findings may have implications for therapeutic modulation of diabetic vascular complications.

Preparation of Discoidal rHDL

apoA-I was isolated from plasma obtained from a pool of multiple healthy donors (>5) by ultracentrifugation and anion-exchange chromatography, as described previously (13). Discoidal rHDL was prepared by complexing apoA-I with 1-palmitoyl-2-linoleoyl-phosphatidylcholine.

Animal Studies

All experimental procedures were conducted with approval from the Sydney Local Health District Animal Welfare Committee. Male 8-week-old C57Bl/6J, SR-BI−/−, and wild-type (WT) littermates were rendered diabetic 2 weeks prior to surgery by a bolus intraperitoneal injection of streptozotocin (165 µg/g).

Murine Hindlimb Ischemia Model

The hindlimb ischemia model was conducted as described previously (14). The left femoral artery and vein were ligated and excised from the hindlimb of mice (n = 8–12/group). A sham procedure was performed on the opposite hindlimb. Mice received intravenous injections of PBS (vehicle control) or rHDL (200 μg/mouse) via the tail vein every second day following surgery. Hindlimb blood reperfusion was determined by laser Doppler perfusion imaging prior to and immediately following surgery and then at days 2 to 3, 7, and 10 postsurgery.

Murine Wound Healing Model

The wound healing model was conducted as previously described (15). Two full-thickness excisions were created on the dorsum and a silicone splint secured around the wound. For each mouse, one wound received rHDL (50 µg/wound/day) and the other PBS topically applied directly on the wound. A transparent occlusive dressing (Opsite) was applied. Digital images and wound area were measured daily. Wound blood perfusion was determined using laser Doppler.

Plasma Lipid and Glucose Concentrations

Total, HDL, and LDL cholesterol concentrations on mouse plasma were determined enzymatically (Roche Diagnostics). HDL cholesterol concentrations were determined following polyethylene glycol precipitation of apoB-containing lipoproteins. Glucose concentrations were measured using a glucometer (Accu-Chek Performa; Roche).

Immunocytochemistry

Fresh frozen 5-µm sections of gastrocnemius muscle from ischemic and nonischemic hindlimbs were stained to detect the number of new capillaries (CD31+; DakoCytomation) per myocyte (laminin; Abcam). The 5-µm sections were taken from the midpoint of paraffin-embedded wound tissues and assessed for CD31+ neovessels (Abcam).

Cell Culture

Human coronary artery endothelial cells (HCAECs; Cell Applications) were cultured in MesoEndo media and used at passages 4 to 5. Cells were seeded at 8 × 104 cells/well, cultured for 8 h, and then treated for 18 h with rHDL (20 μmol/L; final apoA-I concentration) or PBS. Cells were replaced with fresh DMEM media in glucose conditions for 48 h. For high glucose conditions, media was supplemented with d-glucose to a final concentration of 25 mmol/L. For the measurement of phosphorylated proteins, cells were stimulated with 10 ng/mL recombinant human VEGF protein (R&D Systems). Each experiment was performed three times independently and in triplicate.

Siah1/2 Knockdown in HCAECs

HCAECs were transfected for 6 h with 60 nmol/L small interfering RNA (siRNA) for Siah1, Siah2, or control scrambled (Santa Cruz Biotechnology), then treated with rHDL (20 μmol/L) or PBS for 18 h, and exposed to high glucose (25 mmol/L, 48 h).

SR-BI Knockdown in HCAECs

Lentiviruses containing either short hairpin RNA (shRNA) for SR-BI (shSR-BI) or the empty vector (shControl) were generated in 293T17 cells as described previously (16,17). Viral titers were quantified using Lenti-X qRT-PCR Titration Kit (Clontech).

HCAECs were seeded and grown overnight to 50% confluency and then exposed for 24 h to 1 × 107 lentiviral particles/mL containing either shSR-BI or shControl. Transduced cells were seeded at 8 × 104 cells/well. Cells were treated with rHDL (20 μmol/L) or PBS for 18 h then exposed to high glucose (25 mmol/L, 48 h).

RNA Expression

Quantitative real-time PCR was performed for: 1) Vegfa (F: 5′-GGCTGCTGTAACGATGAAG-3′; R: 5′-CTCTCTATGTGCTGGCTTTG-3′), murine Glut1 (forward [F]: 5′-TCAACACGGCCTTCACTG-3′; reverse [R]: 5′-CACGATGCTCAGATAGGACATC-3′), Ppargc1a (F: 5′-TGGAGTGACATAGAGTGTGCTG-3′; R: 5′-TGTTCGCAGGCTCATTGTTG-3′), Hif-2α (F: 5′-AGGTCTGCAAAGGACTTCGG-3′; R: 5′-CAAGTGTGAACTGCTGGTGC-3′), prolyl hydroxylase (Phd) 1, (F: 5′-TAAGGTGCATGGCGGCCTGC-3′; R: 5′-TGGCTGCTGCCCGTTCCTTG-3′), pyruvate dehydrogenase lipoamide kinase isozyme 4 (Pdk4) (F: 5′-CACGTACTCCACTGCTCCAA-3′; R: 5′-AGCGTCTGTCCCATAACCTG-3′), Scarb1 (F: 5′-CTGAGCACGTTCTACACGCA-3′; R: 5′-GGCCTGAATGGCCTCCTTAT-3′), Siah1a (F: 5′-GACTGCTACAGCATTACCCACT-3′; R: 5′-GTTGGATGCAGTTGTGCCG-3′), Siah2 (F: 5′-CTAACGCCCAGCATCAGGAA-3′; R: 5′-GAACAGCCCGTGGTAGCATA-3′), Hif-1α (F: 5′-TCCCTTGCTCTTTGTGGTTGGGT-3′; R: 5′-AACGTAAGCGCTGACCCAGG-3′), Vegfr2 (F: 5′-GCCCAGACTGTGTCCCGCAG-3′; R: 5′-AGCGCAAGACCGGGGAGAGC-3′), and 36B4 (F: 5′-CAACGGCAGCATTTATAACCC-3′; R: 5′-CCCATTGATGATGGAGTGTGG-3′) in murine hindlimbs and wound tissue, and 2) human SIAH1, SIAH2, and β2-microglobulin (B2M) in cultured HCAECs using primers designed previously (16). Relative changes in gene expression were normalized using the ΔΔ threshold cycle method to murine 36B4 or human B2M.

Protein Expression

Whole-cell and nuclear protein extracts were subjected to Western blot analysis and probed with antibodies for phosphoinositide 3-kinase (PI3K; p85) (Abcam), phosphorylated Akt (Ser473), total Akt (Cell Signaling Technology), PHD2, PHD3, HIF-1α (Novus Biologicals), VEGFA (Abcam), VEGFB, VEGFC, VEGFD (R&D Systems), phosphorylated VEGFR2 (Tyr1175), total VEGFR2, phosphorylated eNOS (Ser1177), total eNOS (Cell Signaling Technology), and SR-BI (Novus Biologicals). Even protein loading was confirmed by α-tubulin (Abcam) for whole-cell lysates or lamin B1 (Abcam) for nuclear fractions. Secreted and cytoplasmic VEGFR1 expression were measured in the media and cytoplasmic fractions by ELISA (R&D Systems).

Matrigel Tubulogenesis Assay

Pretreated HCAECs were seeded at 8 × 103 cells/well on polymerized growth factor–reduced Matrigel and incubated for 4 h. Tubules were photographed at ×40 magnification under light microscopy and total number tubules formed determined using ImageJ (National Institutes of Health).

Statistical Analyses

Data are expressed as mean ± SEM. Differences between treatment groups were calculated using a one-way ANOVA (Bonferroni comparison test post hoc) or Student t test. A two-way ANOVA (Bonferroni comparison test post hoc) was performed when comparing data at multiple time points. Significance was set at a two-sided P < 0.05.

rHDL Rescues Diabetes-Impaired Angiogenesis In Vivo

We first studied the effects of rHDL in two models of diabetes-impaired vascular complications including: 1) ischemia-mediated neovascularization, and 2) wound healing and angiogenesis. In the hindlimb ischemia model, femoral artery ligation reduced blood flow equally in all mice at day 0 (Fig. 1A). In the nondiabetic mice, rHDL infusions promoted blood flow recovery compared with PBS-infused mice, reaching significance at day 7 (P < 0.05). Diabetes severely impaired blood flow recovery in PBS-infused mice. However, this was rescued by rHDL infusions (P < 0.01). Consistent with this, rHDL increased capillary density in the gastrocnemius muscle of ischemic hindlimbs in both the nondiabetic (P < 0.01) and diabetic mice (P < 0.001) (Fig. 1B). The impact of diabetes on wound healing and angiogenesis was more striking. In nondiabetic mice, topical rHDL application increased wound closure compared with PBS-treated wounds at day 10 (Fig. 1C). As expected, diabetic mice exhibited delayed wound closure. Topical rHDL rescued diabetes-related impairment in wound closure to the level of nondiabetic controls. Furthermore, in nondiabetic and diabetic mice, blood perfusion was elevated in rHDL-treated wounds in the important early stages of wound recovery (days 2–6) (Fig. 1D). Diabetic mice had reduced wound capillary density in PBS-treated wounds (Fig. 1E). However, topical rHDL enhanced capillary density in diabetic wounds (P < 0.05). In both models, we found that diabetes suppressed Vegfa expression, which was rescued by rHDL treatment (Supplementary Fig. 1A and B). Taken together, we show that rHDL rescues diabetes-impaired angiogenesis. These effects were independent of changes in glucose and lipid levels (Supplementary Tables 1 and 2). Furthermore, rHDL had no effect on markers of glucose metabolism, including the glucose transporter Glut1, the metabolic regulator Ppargc1a and three genes involved in metabolic cellular programming: Hif-2α, Phd1, and Pdk4 (Supplementary Fig. 2). This indicates that rHDL does not mediate its effects on angiogenesis via changes in glucose metabolism. Finally, we found there were no differences in Scarb1 (SR-BI) levels between diabetic and nondiabetic animals in both studies (Supplementary Fig. 3A). However, rHDL significantly augmented hindlimb Scarb1 expression in diabetic mice.

Figure 1

rHDL rescues diabetes-impaired angiogenesis in vivo. Ischemia-mediated neovascularization: femoral artery ligation was performed on nondiabetic and diabetic C57Bl/6J mice (n = 11/group). Mice received i.v. injections of rHDL (200 μg/mouse) or PBS (vehicle) on alternate days following ligation until sacrifice. A: Blood flow perfusion was determined using laser Doppler; images show high (red) to low (blue) blood flow at day 10. Laser Doppler perfusion index (LDPI) was determined based on the ratio of ischemic (ISC) to nonischemic (NON) hindlimb. White circles, nondiabetic PBS-infused mice; gray triangles, nondiabetic rHDL-infused mice; black circles, diabetic PBS-infused mice; and blue squares, diabetic rHDL-infused mice. B: Capillaries were identified using immunocytochemistry for CD31+ and quantified as number of vessels per myocyte. Photomicrographs represent ischemic gastrocnemius muscle stained for capillaries (CD31+, stained red, denoted by arrows) and myocytes (laminin, stained blue). Wound healing and angiogenesis: two full-thickness wounds were created on nondiabetic and diabetic C57Bl/6J mice (n = 11/group). Mice received daily topical applications of rHDL (50 μg/wound) or PBS (vehicle). C: Wound area was calculated from the average of three daily diameter measurements along the x-, y-, and z-axes. Wound closure is expressed as a percentage of initial wound area at day 0. White circles, nondiabetic PBS-treated wound; gray triangles, nondiabetic rHDL-treated wound; black circles, diabetic PBS-treated wound; and red squares, diabetic rHDL-treated wound. D: rHDL:PBS wound blood flow perfusion ratio was determined using laser Doppler imaging; images represent high (red) to low (blue) blood flow at day 10 in nondiabetic (gray triangles) and diabetic (red squares) mice. E: Capillaries were identified in wound sections using immunohistochemistry for CD31+. Photomicrographs represent wounds stained for CD31+ (stained brown, denoted by arrows). Scale bars, 200 μm. Results are expressed as mean ± SEM. *P < 0.05, **P < 0.01, ***P < 0.001 vs. nondiabetic PBS mice, #P < 0.05, ##P < 0.01, ###P < 0.001 vs. diabetic PBS mice.

Figure 1

rHDL rescues diabetes-impaired angiogenesis in vivo. Ischemia-mediated neovascularization: femoral artery ligation was performed on nondiabetic and diabetic C57Bl/6J mice (n = 11/group). Mice received i.v. injections of rHDL (200 μg/mouse) or PBS (vehicle) on alternate days following ligation until sacrifice. A: Blood flow perfusion was determined using laser Doppler; images show high (red) to low (blue) blood flow at day 10. Laser Doppler perfusion index (LDPI) was determined based on the ratio of ischemic (ISC) to nonischemic (NON) hindlimb. White circles, nondiabetic PBS-infused mice; gray triangles, nondiabetic rHDL-infused mice; black circles, diabetic PBS-infused mice; and blue squares, diabetic rHDL-infused mice. B: Capillaries were identified using immunocytochemistry for CD31+ and quantified as number of vessels per myocyte. Photomicrographs represent ischemic gastrocnemius muscle stained for capillaries (CD31+, stained red, denoted by arrows) and myocytes (laminin, stained blue). Wound healing and angiogenesis: two full-thickness wounds were created on nondiabetic and diabetic C57Bl/6J mice (n = 11/group). Mice received daily topical applications of rHDL (50 μg/wound) or PBS (vehicle). C: Wound area was calculated from the average of three daily diameter measurements along the x-, y-, and z-axes. Wound closure is expressed as a percentage of initial wound area at day 0. White circles, nondiabetic PBS-treated wound; gray triangles, nondiabetic rHDL-treated wound; black circles, diabetic PBS-treated wound; and red squares, diabetic rHDL-treated wound. D: rHDL:PBS wound blood flow perfusion ratio was determined using laser Doppler imaging; images represent high (red) to low (blue) blood flow at day 10 in nondiabetic (gray triangles) and diabetic (red squares) mice. E: Capillaries were identified in wound sections using immunohistochemistry for CD31+. Photomicrographs represent wounds stained for CD31+ (stained brown, denoted by arrows). Scale bars, 200 μm. Results are expressed as mean ± SEM. *P < 0.05, **P < 0.01, ***P < 0.001 vs. nondiabetic PBS mice, #P < 0.05, ##P < 0.01, ###P < 0.001 vs. diabetic PBS mice.

Close modal

rHDL Rescues High Glucose–Impaired HIF-1α Stabilization In Vitro

We then studied the effects of rHDL on key angiogenic pathways in vitro in high-glucose conditions. Decreased HIF-1α stability, impaired VEGFA production, and eNOS inhibition are strongly implicated in the pathogenesis of diabetes-impaired angiogenesis (36). HIF-1α is posttranslationally modulated, beginning with the PI3K/Akt signaling pathway, which induces the E3 ubiquitin ligases Siah1 and Siah2 (18). Under high-glucose conditions, rHDL increased PI3K protein expression (Fig. 2A) (P < 0.05) and induced an increase in phosphorylated Akt (pAkt) (Fig. 2B) (P < 0.001). rHDL increased SIAH1 and SIAH2 mRNA levels, irrespective of glucose conditions (Fig. 2C and D) (P < 0.001). The Siahs suppress PHD2 and PHD3 that ubiquitinate and target HIF-1α for degradation. Exposure to high glucose increased PHD2 and PHD3 protein levels (Fig. 2E and F). However, rHDL suppressed both PHD2 (P < 0.01) and PHD3 (P < 0.05) levels. Consistent with the decreases in PHDs, rHDL rescued high glucose–induced reductions in both total and nuclear HIF-1α protein levels (Figs. 2G and H) (P < 0.05). These results show that rHDL rescues high glucose–impaired HIF-1α stabilization in vitro.

Figure 2

rHDL rescues high glucose–impaired HIF-1α stabilization in vitro. HCAECs were treated with rHDL (20 μmol/L, white bars) or PBS (vehicle, black bars) for 18 h prior to 48-h glucose exposure (5–25 mmol/L) and then used for RNA or protein analysis. PI3K (A) and pAkt relative to total Akt (AktT) (B) protein levels; SIAH1 (C) and SIAH2 mRNA levels (D), normalized to B2M; and PHD2 (E), PHD3 (F), total HIF-1α (G), and nuclear HIF-1α (H) protein levels. Black lines separate noncontiguous lanes from the same gel. Results are expressed as mean ± SEM. *P < 0.05, **P < 0.01, ***P < 0.001.

Figure 2

rHDL rescues high glucose–impaired HIF-1α stabilization in vitro. HCAECs were treated with rHDL (20 μmol/L, white bars) or PBS (vehicle, black bars) for 18 h prior to 48-h glucose exposure (5–25 mmol/L) and then used for RNA or protein analysis. PI3K (A) and pAkt relative to total Akt (AktT) (B) protein levels; SIAH1 (C) and SIAH2 mRNA levels (D), normalized to B2M; and PHD2 (E), PHD3 (F), total HIF-1α (G), and nuclear HIF-1α (H) protein levels. Black lines separate noncontiguous lanes from the same gel. Results are expressed as mean ± SEM. *P < 0.05, **P < 0.01, ***P < 0.001.

Close modal

rHDL Rescues High Glucose–Impaired VEGFA/VEGFR2 Production/Activation In Vitro

Although the VEGFA/VEGFR2 signaling axis has been strongly implicated in diabetic vascular complications (5), other members of the VEGF ligand-receptor family have also been implicated in diabetes (1923). We determined the effects of rHDL on the VEGF ligand-receptor family. rHDL prevented high glucose–induced inhibition of VEGFA and VEGFB protein (Fig. 3A and B) (P < 0.05), but did not change VEGFC or VEGFD levels (Fig. 3C and D). Although high glucose did not impact on VEGFR1 expression, rHDL significantly decreased both soluble and cytoplasmic VEGFR1 protein irrespective of glucose conditions (Fig. 3E and F). rHDL promoted VEGFR2 phosphorylation (pVEGFR2) (Fig. 3G) and augmented eNOS phosphorylation in high glucose (Fig. 3H). Finally, consistent with increases in HIF-1α stabilization, VEGFA/VEGFR2 signaling, and eNOS activation, rHDL augmented endothelial cell tubule formation (Fig. 3I) (P < 0.05).

Figure 3

rHDL rescues high glucose–impaired VEGFA/VEGFR2 production/activation in vitro. HCAECs were treated with rHDL (20 μmol/L, white bars) or PBS (vehicle, black bars) for 18 h prior to 48-h glucose exposure (5–25 mmol/L) and then used for protein analysis or Matrigel tubulogenesis assay. VEGFA (A), VEGFB (B), VEGFC (C), VEGFD (D), secreted VEGFR1 (E), cytoplasmic VEGFR1 (F), pVEGFR2 relative to total VEGFR2 (VEGFR2T) (G), and phosphorylated eNOS (peNOS) relative to total eNOS (eNOST) (H) protein levels. Black lines separate noncontiguous lanes from the same gel. I: Representative images of tubule formation photographed at ×40 magnification under light microscopy. Tubule branches were counted using ImageJ (National Institutes of Health). Results are expressed as mean ± SEM. *P < 0.05, **P < 0.01, ***P < 0.001.

Figure 3

rHDL rescues high glucose–impaired VEGFA/VEGFR2 production/activation in vitro. HCAECs were treated with rHDL (20 μmol/L, white bars) or PBS (vehicle, black bars) for 18 h prior to 48-h glucose exposure (5–25 mmol/L) and then used for protein analysis or Matrigel tubulogenesis assay. VEGFA (A), VEGFB (B), VEGFC (C), VEGFD (D), secreted VEGFR1 (E), cytoplasmic VEGFR1 (F), pVEGFR2 relative to total VEGFR2 (VEGFR2T) (G), and phosphorylated eNOS (peNOS) relative to total eNOS (eNOST) (H) protein levels. Black lines separate noncontiguous lanes from the same gel. I: Representative images of tubule formation photographed at ×40 magnification under light microscopy. Tubule branches were counted using ImageJ (National Institutes of Health). Results are expressed as mean ± SEM. *P < 0.05, **P < 0.01, ***P < 0.001.

Close modal

Posttranslational HIF-1α Modulators Siahs Mediate rHDL Rescue of High Glucose–Impaired Angiogenesis In Vitro

To elucidate the importance of Siah1 and Siah2 in rHDL-induced HIF-1α stabilization and VEGFA augmentation, an siRNA knockdown approach was used. In the scrambled siRNA (siScr) cells, rHDL augmented HIF-1α, VEGFA, pVEGFR2, and tubulogenesis (Fig. 4A–D) in high glucose. However, knockdown of Siah1 and Siah2 abrogated several steps in the angiogenic pathway, including rHDL-induced increases in HIF-1α, VEGFA, pVEGFR2, and tubulogenesis, highlighting an important role for Siahs in mediating the proangiogenic effects of rHDL in high glucose.

Figure 4

Posttranslational HIF-1α modulators Siahs mediate rHDL rescue of high glucose–impaired angiogenesis in vitro. HCAECs were transfected with scrambled (siScr), Siah1 (siSiah1), or Siah2 (siSiah2) siRNA for 6 h, then incubated with rHDL (20 μmol/L, white bars) or PBS (vehicle, black bars) for 18 h prior to 48 h in high glucose (25 mmol/L), and used for protein analysis or Matrigel tubulogenesis assay. HIF-1α (A), VEGFA (B), and pVEGFR2 relative to total VEGFR2 (VEGFR2T) (C) protein levels. D: Representative images of tubule formation photographed at ×40 magnification under light microscopy. Tubule branches were counted using ImageJ (National Institutes of Health). Results are expressed as mean ± SEM. *P < 0.05, **P < 0.01 vs. relative PBS controls. Rel., relative.

Figure 4

Posttranslational HIF-1α modulators Siahs mediate rHDL rescue of high glucose–impaired angiogenesis in vitro. HCAECs were transfected with scrambled (siScr), Siah1 (siSiah1), or Siah2 (siSiah2) siRNA for 6 h, then incubated with rHDL (20 μmol/L, white bars) or PBS (vehicle, black bars) for 18 h prior to 48 h in high glucose (25 mmol/L), and used for protein analysis or Matrigel tubulogenesis assay. HIF-1α (A), VEGFA (B), and pVEGFR2 relative to total VEGFR2 (VEGFR2T) (C) protein levels. D: Representative images of tubule formation photographed at ×40 magnification under light microscopy. Tubule branches were counted using ImageJ (National Institutes of Health). Results are expressed as mean ± SEM. *P < 0.05, **P < 0.01 vs. relative PBS controls. Rel., relative.

Close modal

SR-BI Mediates rHDL-Induced Rescue of High Glucose–Impaired Angiogenesis In Vitro

In HCAECs, high glucose induced a 50% decrease in SR-BI protein (Supplementary Fig. 3B) (P < 0.05), consistent with previous studies (2426). Incubation with rHDL prevented high glucose–induced SR-BI inhibition (P < 0.05). The role of SR-BI in mediating the proangiogenic effects of rHDL in high glucose conditions was next assessed in vitro using a lentiviral shSR-BI. In the lentiviral control shRNA (shControl) cells, rHDL augmented pAkt; however, this was abrogated in shSR-BI cells (Fig. 5A). rHDL also failed to augment HIF-1α expression in shSR-BI cells (Fig. 5B). Although rHDL increased VEGFA protein in shControl cells, this induction was abrogated by shSR-BI (Fig. 5C). Finally, rHDL-induced tubule formation was attenuated in shSR-BI cells (Fig. 5D).

Figure 5

SR-BI mediates rHDL-induced rescue of high glucose–impaired angiogenesis in vitro. HCAECs were transduced with lentivirus (1 × 107 viral particles) expressing shSR-BI or the empty vector (shControl). Transduced cells were treated with rHDL (20 μmol/L, white bars) or PBS (vehicle, black bars) for 18 h prior to 48 h in high glucose (25 mmol/L) and then used for protein analysis or Matrigel tubulogenesis assay. pAkt relative to total Akt (AktT) (A), HIF-1α (B), and VEGFA (C) protein levels. D: Representative images of tubule formation photographed at ×40 magnification under light microscopy. Tubule branches were counted using ImageJ (National Institutes of Health). Results are expressed as mean ± SEM. *P < 0.05 vs. relative PBS controls. Rel., relative.

Figure 5

SR-BI mediates rHDL-induced rescue of high glucose–impaired angiogenesis in vitro. HCAECs were transduced with lentivirus (1 × 107 viral particles) expressing shSR-BI or the empty vector (shControl). Transduced cells were treated with rHDL (20 μmol/L, white bars) or PBS (vehicle, black bars) for 18 h prior to 48 h in high glucose (25 mmol/L) and then used for protein analysis or Matrigel tubulogenesis assay. pAkt relative to total Akt (AktT) (A), HIF-1α (B), and VEGFA (C) protein levels. D: Representative images of tubule formation photographed at ×40 magnification under light microscopy. Tubule branches were counted using ImageJ (National Institutes of Health). Results are expressed as mean ± SEM. *P < 0.05 vs. relative PBS controls. Rel., relative.

Close modal

SR-BI Mediates rHDL Rescue of Diabetes-Impaired Ischemia-Induced Neovascularization In Vivo

We then investigated the role of SR-BI in mediating the effects of rHDL on ischemia-mediated neovascularization in vivo using SR-BI−/− mice. In nondiabetic WT mice, rHDL augmented hindlimb blood perfusion, which was attenuated in nondiabetic SR-BI−/− mice (Supplementary Fig. 4A). In diabetic WT littermates, rHDL infusions promoted blood flow recovery, capillary density, and augmented mRNA levels of key angiogenic mediators including Siah1a, Siah2, Hif-1α, and Vegfa (Fig. 6A–F) (P < 0.05). However, the proangiogenic ability of rHDL was completely abrogated in SR-BI−/− diabetic mice, suggesting that rHDL augments ischemia-induced angiogenesis in diabetes via SR-BI.

Figure 6

SR-BI mediates rHDL rescue of diabetes-impaired ischemia-induced neovascularization in vivo. Femoral artery ligation was performed on diabetic WT and SR-BI−/− mice (n = 9 to 10/group). Mice received i.v. injections of rHDL (200 μg/mouse) or PBS (vehicle) on alternate days following ligation. A: Laser Doppler perfusion index (LDPI) was determined based on the ischemic (ISC):nonischemic (NON) hindlimb ratio. White circles, diabetic WT PBS-infused mice; gray triangles, diabetic WT rHDL-infused mice; black circles, diabetic SR-BI−/− PBS-infused mice; blue squares, diabetic SR-BI−/− rHDL-infused mice. B: Capillaries were identified using immunocytochemistry for CD31+ and quantified as number of vessels per myocyte. Photomicrographs represent ischemic gastrocnemius muscle stained for capillaries (CD31+, stained red, denoted by arrows) and myocytes (blue). Scale bars, 200 μm. Siah1a (C), Siah2 (D), Hif-1α (E), Vegfa (F), and Vegfr2 (G) mRNA levels, expressed as a ratio of ISC:NON, normalized to 36B4. Results are expressed as mean ± SEM. *P < 0.05.

Figure 6

SR-BI mediates rHDL rescue of diabetes-impaired ischemia-induced neovascularization in vivo. Femoral artery ligation was performed on diabetic WT and SR-BI−/− mice (n = 9 to 10/group). Mice received i.v. injections of rHDL (200 μg/mouse) or PBS (vehicle) on alternate days following ligation. A: Laser Doppler perfusion index (LDPI) was determined based on the ischemic (ISC):nonischemic (NON) hindlimb ratio. White circles, diabetic WT PBS-infused mice; gray triangles, diabetic WT rHDL-infused mice; black circles, diabetic SR-BI−/− PBS-infused mice; blue squares, diabetic SR-BI−/− rHDL-infused mice. B: Capillaries were identified using immunocytochemistry for CD31+ and quantified as number of vessels per myocyte. Photomicrographs represent ischemic gastrocnemius muscle stained for capillaries (CD31+, stained red, denoted by arrows) and myocytes (blue). Scale bars, 200 μm. Siah1a (C), Siah2 (D), Hif-1α (E), Vegfa (F), and Vegfr2 (G) mRNA levels, expressed as a ratio of ISC:NON, normalized to 36B4. Results are expressed as mean ± SEM. *P < 0.05.

Close modal

SR-BI Mediates rHDL Rescue of Diabetes-Impaired Wound Closure/Angiogenesis

In nondiabetic WT mice, rHDL promoted the rate of wound closure, which did not occur in nondiabetic SR-BI−/− mice (Supplementary Fig. 4B). In diabetic WT littermates, rHDL promoted wound closure and wound angiogenesis and induced Siah1a, Siah2, Hif-1α, Vegfa, and Vegfr2 mRNA levels (Fig. 7A–H) (P < 0.05). The ability of rHDL to promote wound healing and angiogenesis was attenuated in diabetic SR-BI−/− mice. Taken together with the in vitro studies reported in Fig. 5 and the in vivo hindlimb ischemia studies described in Fig. 6, this highlights an important role for SR-BI in the induction of angiogenesis by rHDL in diabetes.

Figure 7

SR-BI mediates rHDL rescue of diabetes-impaired wound closure/angiogenesis. Two full-thickness wounds were created on diabetic WT and SR-BI−/− mice (n = 7 to 8/group). Mice received daily topical applications of rHDL (50 μg/wound) or PBS (vehicle). A: Wound area was calculated from the average of three daily diameter measurements along the x-, y-, and z-axes. Wound closure is expressed as a percentage of initial wound area at day 0. White circles, diabetic WT PBS-treated wound; gray triangles, diabetic WT rHDL-treated wound; black circles, diabetic SR-BI−/− PBS-treated wound; red squares, diabetic SR-BI−/− rHDL-treated wound. B: Blood flow perfusion was determined using laser Doppler imaging; images represent high (red) to low (blue) blood flow at day 10 in diabetic WT (gray triangles) and SR-BI−/− (red squares) mice. C: Capillaries were identified using immunohistochemistry for CD31+ (stained brown, denoted by arrows) and expressed relative to wound area. Scale bars, 200 μm. Siah1a (D), Siah2 (E), Hif-1α (F), Vegfa (G), and Vegfr2 (H) mRNA levels, normalized to 36B4. Results are expressed as mean ± SEM. *P < 0.05.

Figure 7

SR-BI mediates rHDL rescue of diabetes-impaired wound closure/angiogenesis. Two full-thickness wounds were created on diabetic WT and SR-BI−/− mice (n = 7 to 8/group). Mice received daily topical applications of rHDL (50 μg/wound) or PBS (vehicle). A: Wound area was calculated from the average of three daily diameter measurements along the x-, y-, and z-axes. Wound closure is expressed as a percentage of initial wound area at day 0. White circles, diabetic WT PBS-treated wound; gray triangles, diabetic WT rHDL-treated wound; black circles, diabetic SR-BI−/− PBS-treated wound; red squares, diabetic SR-BI−/− rHDL-treated wound. B: Blood flow perfusion was determined using laser Doppler imaging; images represent high (red) to low (blue) blood flow at day 10 in diabetic WT (gray triangles) and SR-BI−/− (red squares) mice. C: Capillaries were identified using immunohistochemistry for CD31+ (stained brown, denoted by arrows) and expressed relative to wound area. Scale bars, 200 μm. Siah1a (D), Siah2 (E), Hif-1α (F), Vegfa (G), and Vegfr2 (H) mRNA levels, normalized to 36B4. Results are expressed as mean ± SEM. *P < 0.05.

Close modal

We report for the first time that rHDL rescues diabetes-impaired angiogenesis by enhancement of ischemia-mediated neovascularization and acceleration of wound closure and wound angiogenesis. In vitro studies indicate that these effects may, at least in part, be via enhanced posttranslational HIF-1α modulation and nuclear translocation, increased VEGFA/VEGFR2 production and signaling, and augmented eNOS activity. Siah siRNA knockdown in vitro confirmed the importance of posttranslational HIF-1α modulation in mediating the proangiogenic effects of rHDL in high glucose. Furthermore, in vitro studies using lentiviral shSR-BI knockdown and in vivo studies with SR-BI−/− mice indicate that these effects of rHDL are mediated by the receptor SR-BI. In summary, we have demonstrated a key role for rHDL in the attenuation of diabetes-related impairment of angiogenesis with implications for the therapeutic modulation of diabetic vascular complications.

Collateral vessel network development is an important response to tissue ischemia following vascular occlusion (27). Similarly, the extent of neovascularization in the early stages following wounding is a key determinant of wound closure rate (28). Diabetes is associated with poor outcomes following vascular occlusion and impaired coronary collateral development (29), and patients with peripheral vascular disease manifest increased peripheral limb ulceration and amputation, with cutaneous wounds more prone to amputation (2,30). Consistent with this, our in vivo studies found that diabetes caused impairment of ischemia-induced neovascularization in hindlimbs and in wound closure/angiogenesis, with a more striking impact on wound healing. These are two distinctly different models; therefore, the timing for capturing changes may vary. Compared with the hindlimb ischemia model, which is primarily driven by ischemia-mediated angiogenesis, wound healing is more complex and not solely dependent on angiogenesis but involves other cellular processes, including epithelialization and cellular proliferation. Additionally, a chronic inflammatory state as is commonly seen in patients with diabetes, significantly impairs wound healing and can lead to severe unfavorable outcomes such as amputation. HDL also exhibits anti-inflammatory effects (31,32) and may assist in the more robust impact of HDL. Regardless, rHDL promoted neovascularization in both angiogenic models, rescuing both ischemia-mediated neovascularization and wound healing and angiogenesis, potentially highlighting a new role for HDL in attenuating vascular complications associated with diabetes-impaired neovascularization.

Our in vitro studies found that high glucose suppressed posttranslational HIF-1α modulation (via augmentation of PHDs), VEGFA/VEGFR2 production and signaling, and tubulogenesis. HIF-1α is the pivotal transcription factor involved in ischemia-mediated neovascularization and governed by a complex orchestration of posttranslational regulation. Previous studies have reported that hyperglycemia inhibits hypoxia-induced HIF-1α stabilization and suggest that mechanisms involving proline hydroxylation are important (33). The current study, however, is the first to directly show that high glucose decreases HIF-1α stability via posttranslational effects including an increase in PHD (PHD2 and PHD3) expression and a decrease in both total and nuclear HIF-1α protein. More importantly, rHDL rescued HIF-1α stabilization at each step in its posttranslational regulation by: 1) activating the PI3K/Akt signaling pathway, which 2) triggered an increase in the expression of the E3 ubiquitin ligases Siahs that 3) suppressed PHD expression and ultimately 4) rescued hyperglycemia-induced reductions in HIF-1α. These observations complement a recent study that found rHDL increased posttranslational HIF-1α stabilization, but this study was not conducted in high-glucose conditions (17). Nuclear localization studies found rHDL augmented nuclear HIF-1α protein levels, demonstrating strong evidence of the impact of rHDL and the Siah/PHD axis on promoting nuclear HIF-1α translocation. Furthermore, siRNA knockdown of Siahs confirmed the importance of posttranslational HIF-1α modulation in the effects of rHDL in high glucose, as silencing of the Siahs abrogated key steps in the angiogenic pathway in vitro.

As expected, high glucose suppressed VEGFA protein levels in vitro. rHDL treatment prevented this decrease, most likely via the stabilization of HIF-1α, the critical transcription factor mediating VEGFA expression. Our in vitro analysis of the VEGF ligand-receptor family found that rHDL augmented VEGFB but had no effect on VEGFC or VEGFD. Both VEGFA and VEGFB bind to VEGFR1, which is also implicated in diabetes-impaired neovascularization (19,34). However, we found that rHDL decreased VEGFR1 expression, suggesting that VEGFR1 is not mediating rHDL-induced rescue of angiogenesis in high glucose. VEGFR2 is the receptor that regulates the proangiogenic effects of VEGFA. Following binding of VEGFA to VEGFR2, the receptor dimerizes and causes the activation of receptor-kinase activity, leading to the phosphorylation of the receptor. pVEGFR2 induces the activation of an array of angiogenic signaling pathways (35). We found that rHDL increased the phosphorylation (activation) of VEGFR2 in high glucose. This is particularly important, as hyperglycemia reduces VEGFA sensitivity via suppression of VEGFR2 activation (5). Previous studies have found rHDL augments hypoxia-induced VEGFR2 total protein levels (14), but this is the first study to show that in high glucose, rHDL promotes both VEGFA protein expression and signaling via VEGFR2 phosphorylation/activation. The current study also found that rHDL promoted eNOS phosphorylation in high glucose, which is likely to be due to the increase in VEGFA/VEGFR2 and the subsequent increase in Akt phosphorylation (downstream of VEGFR2). Elevated eNOS phosphorylation leads to nitric oxide release, promoting angiogenic functions including endothelial cell migration, proliferation, and vessel growth (36,37). Consistent with this, other studies have found that rHDL increases eNOS activity in vitro and in vivo (38,39). Finally, using a functional Matrigel assay for endothelial tubule formation, we found that rHDL augmented high glucose–impaired tubule formation, suggesting that the effects of rHDL on the key angiogenic proteins is translated into critical cellular processes involved in angiogenic functions.

It is becoming increasingly recognized that HDL exhibits antidiabetic effects (9,40). We show in this study that rHDL rescues diabetes-mediated impairment of hindlimb and wound angiogenesis. This is supported by previous work showing that apoA-I/rHDL augment ischemia-driven angiogenesis and promote angiogenesis-related functions including migration and re-endothelialization (14,17,41). However, these studies were not done in the clinically relevant setting of hyperglycemia, one of the key contributors to diabetes-impaired vascular complications. In the current study, the effects of rHDL were independent of changes in glucose and lipid levels. Our in vivo gene analysis of both hindlimb and wound tissues found that rHDL had no effect on the expression of the glucose transporter GLUT1 and the transcriptional coactivator peroxisome proliferator-activated receptor γ coactivator 1-α, a powerful regulator in metabolism that is involved in HIF-independent regulation of VEGF and angiogenesis in diabetes (42). Hypoxia tolerance and HIF-1α stabilization are central to a hypometabolic state characterized by reduced oxygen consumption, such as that seen in diabetes-impaired neovascularization. At a cellular level, metabolic cellular reprogramming involves increased efficiency of energy-producing pathways, via increased anaerobic glycolysis activity, and decreased energy-consuming processes and is mediated via HIF-2α, PHD1, and PDK4 (43). rHDL did not have any effect on gene expression of any of these markers. Taken together, this confirms that the action of rHDL on diabetes-impaired angiogenesis is not due to glucose stress dependent effects but via a number of mechanisms previously found to contribute to diabetes-impaired neovascularization including: 1) promotion of posttranslational HIF-1α stabilization, 2) VEGFA/VEGFR2 production and signaling, and 3) eNOS activation.

The scavenger receptor SR-BI has been implicated in mediating a number of the endothelial protective effects of HDL such as migration, tubulogenesis, and re-endothelialization (11,12,17,44,45). This is the first study to date that has provided a direct link between SR-BI and the proangiogenic effects of HDL in diabetes. We found that diabetic animals had slightly lower levels of Scarb1 mRNA, although this did not reach significance, whereas SR-BI protein levels were significantly reduced under high-glucose conditions in vitro. Currently, there is no consensus on what happens to SR-BI expression in patients with diabetes, with one study reporting elevated SR-BI mRNA levels in patients with diabetes (46) whereas two studies reported no differences (47,48). Data from in vitro studies are more robust. Exposure to high glucose is shown to reduce SR-BI levels across several cell types, including intestinal Caco-2/15 cells (24), hepatic HepG2 cells (25), and human monocyte-derived macrophages (26). More importantly, rHDL increased SR-BI expression in vivo and in vitro. Our in vitro and in vivo data show that SR-BI is critical in mediating the ability of rHDL to rescue diabetes-impaired neovascularization. rHDL was unable to restore high glucose–impaired angiogenesis in shSR-BI cells in vitro or rescue diabetes-impaired hindlimb and wound angiogenesis/closure in SR-BI−/− mice in vivo. Furthermore, compared with WT littermates of both the hindlimb ischemia and wound-healing models, we found that the ability of rHDL to augment Siah1a, Siah2, Hif-1α, Vegfa, and Vegfr2 mRNA levels were attenuated in SR-BI−/− mice. Interestingly, SR-BI is the preferred cholesterol acceptor for spherical HDL particles rather than discoidal rHDL (49). However, it would be expected that following incubation in vitro, systemic injection in vivo, or topical application to wounds, rHDL would rapidly acquire lipid, forming a spherical particle that is able to interact with SR-BI (50). In support of our findings, a recent study found that SR-BI acts as a cholesterol sensor triggering intracellular signaling and is important for the actions of HDL on endothelial cells (45). SR-BI was also important in mediating ischemia-mediated neovascularization and wound healing/angiogenesis in nondiabetic mice. The role of SR-BI in angiogenesis is further supported by reports that show important signaling pathways including PI3K/Akt are downstream of SR-BI and are associated with angiogenesis-related functions such as migration and proliferation (12).

Despite the vast number of reports demonstrating the therapeutic benefits of HDL on the cardiovascular system, to date, there is no translated use of HDL-targeted treatments. There is, however, increasing evidence for the antidiabetic effects of HDL, which may present an alternative translation pathway. We found that rHDL rescues both high glucose–related impairment of tubulogenesis in vitro and diabetes-impaired neovascularization in vivo. The mechanisms for these effects are via increased posttranslational HIF-1α stabilization, VEGFA/VEGFR2 production/activation and signaling, and eNOS activation (Fig. 8). Furthermore, SR-BI is important in mediating rHDL-induced rescue of diabetes-impaired angiogenesis. The current study provides a greater understanding into the vascular biological effects of HDL in the context of diabetic vascular complications. This may ultimately facilitate the translation of HDL, not only for cardiovascular disease but also diseases associated with impaired angiogenesis and the vascular complications of diabetes.

Figure 8

Proposed mechanism of action of rHDL rescue of diabetes-impaired angiogenesis. Under high-glucose conditions, rHDL activates the PI3K/Akt pathway, inducing the expression of the E3 ubiquitin ligases, Siah1 and Siah2. Increases in Siahs result in the inhibition of two members of the prolyl hydroxylase domain protein family, PHD2 and PHD3. Suppression of PHD2/3 prevents HIF-1α from degradation, allowing it to translocate to the nucleus and bind to the hypoxia response element (HRE), activating transcription of proangiogenic mediators including VEGFA. VEGFA is released into the circulation, where it binds and phosphorylates VEGFR2, further augmenting angiogenesis via the PI3K/Akt pathway and eNOS phosphorylation. These effects of rHDL are mediated via SR-BI. ↑ and ↓ denotes the effects of rHDL.

Figure 8

Proposed mechanism of action of rHDL rescue of diabetes-impaired angiogenesis. Under high-glucose conditions, rHDL activates the PI3K/Akt pathway, inducing the expression of the E3 ubiquitin ligases, Siah1 and Siah2. Increases in Siahs result in the inhibition of two members of the prolyl hydroxylase domain protein family, PHD2 and PHD3. Suppression of PHD2/3 prevents HIF-1α from degradation, allowing it to translocate to the nucleus and bind to the hypoxia response element (HRE), activating transcription of proangiogenic mediators including VEGFA. VEGFA is released into the circulation, where it binds and phosphorylates VEGFR2, further augmenting angiogenesis via the PI3K/Akt pathway and eNOS phosphorylation. These effects of rHDL are mediated via SR-BI. ↑ and ↓ denotes the effects of rHDL.

Close modal

J.T.M.T. and H.C.G.P. made equal author contributions to this publication.

M.K.C.N. and C.A.B. made equal author contributions to this publication.

See accompanying article, p. 2826.

Acknowledgments. The authors thank Pat Pisansarakit, from the Heart Research Institute, for the maintenance of HCAECs and Emily King, from the Heart Research Institute, for the use of the murine Hif-2α, Phd1, and Pdk4 primers.

Funding. This work was supported by the National Health and Medical Research Council of Australia Project Grant (632512 to M.K.C.N. and C.A.B.), Early Career Fellowship (537537 to L.L.D.), PhD Scholarship (APP1038394 to S.C.G.Y.), National Heart Foundation Career Development Fellowship (CR07S3331 to C.A.B.), and PhD Scholarship (PB12S6959 to L.Z.V.).

Duality of Interest. No potential conflicts of interest relevant to this article were reported.

Author Contributions. J.T.M.T., H.C.G.P., and C.A.B. developed the study design, designed the experiments, interpreted the data, performed the experiments, and wrote the manuscript. L.L.D. and S.R. performed the experiments and reviewed and edited the manuscript. L.Z.V., A.R., T.T., L.L., Z.E.C., S.C.G.Y., and Y.T.L. performed the experiments. D.S.C. reviewed and edited the manuscript. M.K.C.N. developed the study design, designed the experiments, interpreted the data, and reviewed and edited the manuscript. C.A.B. is the guarantor of this work and, as such, had full access to all the data in the study and takes responsibility for the integrity of the data and the accuracy of the data analysis.

Prior Presentation. Parts of this study were presented in abstract form at the 75th Scientific Sessions of the American Diabetes Association, Boston, MA, 5–9 June 2015.

1.
Abaci
A
,
Oğuzhan
A
,
Kahraman
S
, et al
.
Effect of diabetes mellitus on formation of coronary collateral vessels
.
Circulation
1999
;
99
:
2239
2242
[PubMed]
2.
Willyard
C
.
Limb-saving medicines sought to prevent amputations
.
Nat Med
2012
;
18
:
328
[PubMed]
3.
Botusan
IR
,
Sunkari
VG
,
Savu
O
, et al
.
Stabilization of HIF-1alpha is critical to improve wound healing in diabetic mice
.
Proc Natl Acad Sci U S A
2008
;
105
:
19426
19431
[PubMed]
4.
Mace
KA
,
Yu
DH
,
Paydar
KZ
,
Boudreau
N
,
Young
DM
.
Sustained expression of Hif-1alpha in the diabetic environment promotes angiogenesis and cutaneous wound repair
.
Wound Repair Regen
2007
;
15
:
636
645
[PubMed]
5.
Rivard
A
,
Silver
M
,
Chen
D
, et al
.
Rescue of diabetes-related impairment of angiogenesis by intramuscular gene therapy with adeno-VEGF
.
Am J Pathol
1999
;
154
:
355
363
[PubMed]
6.
Lin
KY
,
Ito
A
,
Asagami
T
, et al
.
Impaired nitric oxide synthase pathway in diabetes mellitus: role of asymmetric dimethylarginine and dimethylarginine dimethylaminohydrolase
.
Circulation
2002
;
106
:
987
992
[PubMed]
7.
Barter
PJ
.
Cardioprotective effects of high-density lipoproteins: the evidence strengthens
.
Arterioscler Thromb Vasc Biol
2005
;
25
:
1305
1306
[PubMed]
8.
Tenkanen
L
,
Mänttäri
M
,
Kovanen
PT
,
Virkkunen
H
,
Manninen
V
.
Gemfibrozil in the treatment of dyslipidemia: an 18-year mortality follow-up of the Helsinki Heart Study
.
Arch Intern Med
2006
;
166
:
743
748
[PubMed]
9.
Nieuwdorp
M
,
Vergeer
M
,
Bisoendial
RJ
, et al
.
Reconstituted HDL infusion restores endothelial function in patients with type 2 diabetes mellitus
.
Diabetologia
2008
;
51
:
1081
1084
[PubMed]
10.
van Oostrom
O
,
Nieuwdorp
M
,
Westerweel
PE
, et al
.
Reconstituted HDL increases circulating endothelial progenitor cells in patients with type 2 diabetes
.
Arterioscler Thromb Vasc Biol
2007
;
27
:
1864
1865
[PubMed]
11.
Mineo
C
,
Shaul
PW
.
Role of high-density lipoprotein and scavenger receptor B type I in the promotion of endothelial repair
.
Trends Cardiovasc Med
2007
;
17
:
156
161
[PubMed]
12.
Seetharam
D
,
Mineo
C
,
Gormley
AK
, et al
.
High-density lipoprotein promotes endothelial cell migration and reendothelialization via scavenger receptor-B type I
.
Circ Res
2006
;
98
:
63
72
[PubMed]
13.
Weisweiler
P
.
Isolation and quantitation of apolipoproteins A-I and A-II from human high-density lipoproteins by fast-protein liquid chromatography
.
Clin Chim Acta
1987
;
169
:
249
254
[PubMed]
14.
Prosser
HC
,
Tan
JT
,
Dunn
LL
, et al
.
Multifunctional regulation of angiogenesis by high-density lipoproteins
.
Cardiovasc Res
2014
;
101
:
145
154
[PubMed]
15.
Dunn
L
,
Prosser
HC
,
Tan
JT
,
Vanags
LZ
,
Ng
MK
,
Bursill
CA
.
Murine model of wound healing
.
J Vis Exp
2013
;
75
:
e50265
[PubMed]
16.
Bursill
CA
,
McNeill
E
,
Wang
L
, et al
.
Lentiviral gene transfer to reduce atherosclerosis progression by long-term CC-chemokine inhibition
.
Gene Ther
2009
;
16
:
93
102
[PubMed]
17.
Tan
JT
,
Prosser
HC
,
Vanags
LZ
,
Monger
SA
,
Ng
MK
,
Bursill
CA
.
High-density lipoproteins augment hypoxia-induced angiogenesis via regulation of post-translational modulation of hypoxia-inducible factor 1α
.
FASEB J
2014
;
28
:
206
217
[PubMed]
18.
Bruick
RK
,
McKnight
SL
.
A conserved family of prolyl-4-hydroxylases that modify HIF
.
Science
2001
;
294
:
1337
1340
[PubMed]
19.
Cai
J
,
Ahmad
S
,
Jiang
WG
, et al
.
Activation of vascular endothelial growth factor receptor-1 sustains angiogenesis and Bcl-2 expression via the phosphatidylinositol 3-kinase pathway in endothelial cells
.
Diabetes
2003
;
52
:
2959
2968
[PubMed]
20.
Hagberg
CE
,
Mehlem
A
,
Falkevall
A
, et al
.
Targeting VEGF-B as a novel treatment for insulin resistance and type 2 diabetes
.
Nature
2012
;
490
:
426
430
[PubMed]
21.
Hazarika
S
,
Dokun
AO
,
Li
Y
,
Popel
AS
,
Kontos
CD
,
Annex
BH
.
Impaired angiogenesis after hindlimb ischemia in type 2 diabetes mellitus: differential regulation of vascular endothelial growth factor receptor 1 and soluble vascular endothelial growth factor receptor 1
.
Circ Res
2007
;
101
:
948
956
[PubMed]
22.
Mehlem
A
,
Palombo
I
,
Wang
X
,
Hagberg
CE
,
Eriksson
U
,
Falkevall
A
.
PGC-1alpha coordinates mitochondrial respiratory capacity and muscular fatty acid uptake via regulation of VEGF-B
.
Diabetes
2016
;
65
:
861
873
[PubMed]
23.
Witmer
AN
,
Blaauwgeers
HG
,
Weich
HA
,
Alitalo
K
,
Vrensen
GF
,
Schlingemann
RO
.
Altered expression patterns of VEGF receptors in human diabetic retina and in experimental VEGF-induced retinopathy in monkey
.
Invest Ophthalmol Vis Sci
2002
;
43
:
849
857
[PubMed]
24.
Ravid
Z
,
Bendayan
M
,
Delvin
E
, et al
.
Modulation of intestinal cholesterol absorption by high glucose levels: impact on cholesterol transporters, regulatory enzymes, and transcription factors
.
Am J Physiol Gastrointest Liver Physiol
2008
;
295
:
G873
G885
[PubMed]
25.
Murao
K
,
Yu
X
,
Imachi
H
, et al
.
Hyperglycemia suppresses hepatic scavenger receptor class B type I expression
.
Am J Physiol Endocrinol Metab
2008
;
294
:
E78
E87
[PubMed]
26.
Moheimani
F
,
Tan
JT
,
Brown
BE
, et al
.
Effect of exposure of human monocyte-derived macrophages to high, versus normal, glucose on subsequent lipid accumulation from glycated and acetylated low-density lipoproteins
.
Exp Diabetes Res
2011
;
2011
:
851280
27.
Kannel
WB
,
McGee
DL
.
Diabetes and cardiovascular disease. The Framingham study
.
JAMA
1979
;
241
:
2035
2038
[PubMed]
28.
Gurtner
GC
,
Werner
S
,
Barrandon
Y
,
Longaker
MT
.
Wound repair and regeneration
.
Nature
2008
;
453
:
314
321
[PubMed]
29.
Waltenberger
J
,
Lange
J
,
Kranz
A
.
Vascular endothelial growth factor-A-induced chemotaxis of monocytes is attenuated in patients with diabetes mellitus: A potential predictor for the individual capacity to develop collaterals
.
Circulation
2000
;
102
:
185
190
[PubMed]
30.
Jonasson
JM
,
Ye
W
,
Sparén
P
,
Apelqvist
J
,
Nyrén
O
,
Brismar
K
.
Risks of nontraumatic lower-extremity amputations in patients with type 1 diabetes: a population-based cohort study in Sweden
.
Diabetes Care
2008
;
31
:
1536
1540
[PubMed]
31.
van der Vorst
EP
,
Vanags
LZ
,
Dunn
LL
,
Prosser
HC
,
Rye
KA
,
Bursill
CA
.
High-density lipoproteins suppress chemokine expression and proliferation in human vascular smooth muscle cells
.
FASEB J
2013
;
27
:
1413
1425
[PubMed]
32.
Wu
BJ
,
Chen
K
,
Shrestha
S
,
Ong
KL
,
Barter
PJ
,
Rye
KA
.
High-density lipoproteins inhibit vascular endothelial inflammation by increasing 3β-hydroxysteroid-Δ24 reductase expression and inducing heme oxygenase-1
.
Circ Res
2013
;
112
:
278
288
[PubMed]
33.
Catrina
SB
,
Okamoto
K
,
Pereira
T
,
Brismar
K
,
Poellinger
L
.
Hyperglycemia regulates hypoxia-inducible factor-1alpha protein stability and function
.
Diabetes
2004
;
53
:
3226
3232
[PubMed]
34.
Ferrara
N
,
Gerber
HP
,
LeCouter
J
.
The biology of VEGF and its receptors
.
Nat Med
2003
;
9
:
669
676
[PubMed]
35.
Olsson
AK
,
Dimberg
A
,
Kreuger
J
,
Claesson-Welsh
L
.
VEGF receptor signalling - in control of vascular function
.
Nat Rev Mol Cell Biol
2006
;
7
:
359
371
[PubMed]
36.
Cooke
JP
,
Losordo
DW
.
Nitric oxide and angiogenesis
.
Circulation
2002
;
105
:
2133
2135
[PubMed]
37.
Jacobi
J
,
Sydow
K
,
von Degenfeld
G
, et al
.
Overexpression of dimethylarginine dimethylaminohydrolase reduces tissue asymmetric dimethylarginine levels and enhances angiogenesis
.
Circulation
2005
;
111
:
1431
1438
[PubMed]
38.
Drew
BG
,
Fidge
NH
,
Gallon-Beaumier
G
,
Kemp
BE
,
Kingwell
BA
.
High-density lipoprotein and apolipoprotein AI increase endothelial NO synthase activity by protein association and multisite phosphorylation
.
Proc Natl Acad Sci U S A
2004
;
101
:
6999
7004
[PubMed]
39.
Yuhanna
IS
,
Zhu
Y
,
Cox
BE
, et al
.
High-density lipoprotein binding to scavenger receptor-BI activates endothelial nitric oxide synthase
.
Nat Med
2001
;
7
:
853
857
[PubMed]
40.
Drew
BG
,
Duffy
SJ
,
Formosa
MF
, et al
.
High-density lipoprotein modulates glucose metabolism in patients with type 2 diabetes mellitus
.
Circulation
2009
;
119
:
2103
2111
[PubMed]
41.
Sumi
M
,
Sata
M
,
Miura
S
, et al
.
Reconstituted high-density lipoprotein stimulates differentiation of endothelial progenitor cells and enhances ischemia-induced angiogenesis
.
Arterioscler Thromb Vasc Biol
2007
;
27
:
813
818
[PubMed]
42.
Arany
Z
,
Foo
SY
,
Ma
Y
, et al
.
HIF-independent regulation of VEGF and angiogenesis by the transcriptional coactivator PGC-1alpha
.
Nature
2008
;
451
:
1008
1012
[PubMed]
43.
Aragonés
J
,
Schneider
M
,
Van Geyte
K
, et al
.
Deficiency or inhibition of oxygen sensor Phd1 induces hypoxia tolerance by reprogramming basal metabolism
.
Nat Genet
2008
;
40
:
170
180
[PubMed]
44.
Feng
Y
,
van Eck
M
,
Van Craeyveld
E
, et al
.
Critical role of scavenger receptor-BI-expressing bone marrow-derived endothelial progenitor cells in the attenuation of allograft vasculopathy after human apo A-I transfer
.
Blood
2009
;
113
:
755
764
[PubMed]
45.
Saddar
S
,
Carriere
V
,
Lee
WR
, et al
.
Scavenger receptor class B type I is a plasma membrane cholesterol sensor
.
Circ Res
2013
;
112
:
140
151
[PubMed]
46.
Rein-Fischboeck
L
,
Krautbauer
S
,
Eisinger
K
, et al
.
Hepatic scavenger receptor BI is associated with type 2 diabetes but unrelated to human and murine non-alcoholic fatty liver disease
.
Biochem Biophys Res Commun
2015
;
467
:
377
382
[PubMed]
47.
Forcheron
F
,
Cachefo
A
,
Thevenon
S
,
Pinteur
C
,
Beylot
M
.
Mechanisms of the triglyceride- and cholesterol-lowering effect of fenofibrate in hyperlipidemic type 2 diabetic patients
.
Diabetes
2002
;
51
:
3486
3491
[PubMed]
48.
Zhou
H
,
Tan
KC
,
Shiu
SW
,
Wong
Y
.
Determinants of leukocyte adenosine triphosphate-binding cassette transporter G1 gene expression in type 2 diabetes mellitus
.
Metabolism
2008
;
57
:
1135
1140
[PubMed]
49.
Tall
AR
.
Cholesterol efflux pathways and other potential mechanisms involved in the athero-protective effect of high density lipoproteins
.
J Intern Med
2008
;
263
:
256
273
[PubMed]
50.
Kee
P
,
Rye
KA
,
Taylor
JL
,
Barrett
PH
,
Barter
PJ
.
Metabolism of apoA-I as lipid-free protein or as component of discoidal and spherical reconstituted HDLs: studies in wild-type and hepatic lipase transgenic rabbits
.
Arterioscler Thromb Vasc Biol
2002
;
22
:
1912
1917
[PubMed]
Readers may use this article as long as the work is properly cited, the use is educational and not for profit, and the work is not altered. More information is available at http://www.diabetesjournals.org/content/license.

Supplementary data