Diabetic endothelial dysfunction associated with diminished endothelial nitric oxide (NO) synthase (eNOS) activity accelerates the development of atherosclerosis and cardiomyopathy. However, the approaches to restore eNOS activity and endothelial function in diabetes remain limited. The current study shows that enhanced expression of Krüppel-like factor 2 (KLF2), a shear stress-inducible transcription factor, effectively improves endothelial function through increasing NO bioavailability. KLF2 expression is suppressed in diabetic mouse aortic endothelium. Running exercise and simvastatin treatment induce endothelial KLF2 expression in db/db mice. Adenovirus-mediated endothelium-specific KLF2 overexpression enhances both endothelium-dependent relaxation and flow-mediated dilatation, while it attenuates oxidative stress in diabetic mouse arteries. KLF2 overexpression increases the phosphorylation of eNOS at serine 1177 and eNOS dimerization. RNA-sequencing analysis reveals that KLF2 transcriptionally upregulates genes that are enriched in the cyclic guanosine monophosphate–protein kinase G–signaling pathway, cAMP-signaling pathway, and insulin-signaling pathway, all of which are the upstream regulators of eNOS activity. Activation of the phosphoinositide 3-kinase–Akt pathway and Hsp90 contributes to KLF2-induced increase of eNOS activity. The present results suggest that approaches inducing KLF2 activation, such as physical exercise, are effective to restore eNOS activity against diabetic endothelial dysfunction.

Article Highlights

  • Exercise and statins restore the endothelial expression of Krüppel-like factor 2 (KLF2), which is diminished in diabetic db/db mice.

  • Endothelium-specific overexpression of KLF2 improves endothelium-dependent relaxation and flow-mediated dilation through increasing nitric oxide bioavailability.

  • KLF2 promotes endothelial nitric oxide synthase (eNOS) coupling and phosphorylation in addition to its known role in eNOS transcription.

  • KLF2 upregulates the expression of several panels of genes that regulate eNOS activity.

Vascular complications of diabetes predispose the development of atherosclerosis that leads to disability and death (1). Endothelial dysfunction, an initial pathological event during atherogenesis, is closely associated with diabetes (2). The current recommended interventions to improve endothelial function in diabetes include physical exercise and drug treatment with statins, metformin, and angiotensin type 1 receptor antagonists (36).

Krüppel-like factor 2 (KLF2) is a transcription factor induced by laminar flow and statins in endothelial cells (ECs) (7,8). By contrast, KLF2 expression is inhibited by proinflammatory cytokines and atheroprone disturbed flow (7,9). A large number of vasoprotective genes induced by laminar flow are reported to be KLF2-dependent (10,11), thus explaining an antiatherosclerotic property of KLF2 via its anti-inflammatory and antioxidant effects in ECs (12,13). Of note, risk factors that inhibit KLF2 expression, such as proinflammatory cytokines and disturbed flow, also impair endothelial function, suggesting that KLF2 downregulation is likely to be a mechanism underlying endothelial dysfunction in diabetes. However, the precise role of KLF2 in the development of diabetic endothelial dysfunction is largely unclear, and whether augmenting KLF2 expression/activity improves endothelium-dependent relaxation (EDR) in diabetic mice remains underexplored.

Endothelial nitric oxide (NO) synthase (eNOS) mediates NO production, endothelial function, and vascular tone regulation. In diabetic rodent vasculature, increased oxidative stress and inflammation cause eNOS dysfunction manifested by reduced phosphorylation of eNOS at serine 1177 (p-eNOS S1177) and uncoupled eNOS (14,15). Instead of producing NO, the uncoupled eNOS releases superoxide anion (16). The fully functional eNOS requires its coupling with cofactors such as tetrahydrobiopterin (BH4) (16). Sufficient cellular level of BH4, mainly synthesized de novo through the rate-limiting guanosine triphosphate cyclohydrolase I (GCH-1), promotes eNOS dimerization for efficient NO production (17). The association with Hsp90 facilitates agonist- and fluid shear stress-induced eNOS activation (18), and Akt-mediated p-eNOS S1177 greatly enhances eNOS activity and NO generation (19,20). The transcription of the eNOS gene can be increased by KLF2, and fluid shear stress-mediated upregulation of eNOS expression is KLF2-dependent (11,21). Nevertheless, whether KLF2 is protective in preserving eNOS function under diabetic conditions remains basically unknown.

The current study shows that KLF2 overexpression improved endothelial function through increasing NO bioavailability in diabetic db/db mice. Mechanistically, the KLF2-mediated transcriptional program creates a signaling network to restore eNOS activity, which is partially mediated by phosphoinositide 3-kinase (PI3K)–Akt signaling and Hsp90. The present results suggest that upregulation of the KLF2 level is effective to rescue eNOS activity and thus preserve endothelial function in diabetic mice.

Animals

The animal use and experimental protocols were approved by the Laboratory Animal Experimentation Ethical Committee (Ref no. 16-029-MIS), Chinese University of Hong Kong. Male and female C57BL/6J, db/m+ and db/db mice (10–16 weeks old) were provided by Chinese University of Hong Kong Laboratory Animal Service Centre, housed in a temperature-controlled room (22–24°C) with a 12-h light/dark cycle, fed with standard research diet, and had free access to water.

Exercise Protocol and Simvastatin Treatment

Male and female mice were both trained to run on a motorized treadmill. For C57BL/6J mice, the running speed was initially set at 8 m/min for 30 min and then gradually increased to the target speed at 12 m/min for 30 min, for 7 days. For db/db mice, the initial running speed was set at 5 m/min for 30 min and then increased to 8 m/min for 30 min, 6 days per week, for 4 weeks (5). The sedentary mice were placed on a nonmoving treadmill for the same duration as the exercising mice. For simvastatin treatment, 12-week-old male db/db mice were orally administered with the drug (40 mg/kg/day) for 4 weeks before sacrifice for collection of blood vessels for examination.

Cell Culture

The culture of human umbilical vein ECs (HUVECs) from Lonza (CC-2519), human aortic ECs (HAECs) from Thermo Fisher Scientific (C0065C), and HEK293A/T cells from ATCC were performed using previously reported method (22).

Adenoviral Vectors and Adenovirus Packaging

Full-length human KLF2 coding sequence was cloned into a pAdTrack-cytomegalovirus vector and packaged to recombinant adenovirus using an AdEasy system (23). Briefly, the recombinant construct was transfected to HEK293A cells for virus package. The condensation of adenovirus (Ad) was achieved by polyethylene glycol 6000 precipitation before use in cell culture. Purification of Ad for animal studies was achieved by using the commercial kit Adeno-X Maxi Purification Kit (Clontech, Mountain View, CA). Ad-KLF2, Ad-Cdh5-Klf2, or Ad-green fluorescent protein (GFP) (1010 plaque forming units/mouse) were administered to mice via tail vein injection for 7 days. The EC-specific Klf2 overexpression virus Ad-Cdh5-Klf2 (Mouse Klf2 coding sequence) and human KLF2 knockdown virus Ad-shKLF2 were generated using the same method.

Blood Vessel Preparation and Isometric Force Measurement

The mice were sacrificed, and the vessels were rapidly removed and placed in oxygenated ice-cold Krebs-Henseleit solution. Aortas and main mesenteric arteries were carefully dissected out and cut into ∼2-mm-long segments. Two segments from each mouse were amounted to a Multi Wire Myograph System (Danish Myo Technology, East Jutland, Denmark) to measure isometric tension. Each arterial ring was first precontracted by 60 mmol/L KCl to test viability. Phenylephrine (Phe) at 1 μmol/L was used to contract the rings, followed by cumulative addition of acetylcholine (ACh) to cause endothelium-dependent relaxations (24). Endothelium-independent relaxations to sodium nitroprusside (SNP), an NO donor, were determined in rings pretreated with a NOS inhibitor l-NG-nitro-l-arginine methyl ester (l-NAME; 100 μmol/L).

Flow-Mediated Dilatation

The segments of second-order mouse mesenteric arteries were dissected and cannulated between two glass cannulas. The intraluminal pressure and vessel diameter were monitored by a light-inverted microscopy with a video camera and the MyoVIEW software (Danish Myo Technology), as previously described (24). Briefly, Phe at 3 μmol/L was added to induce constriction after the vessel’s diameter stabilized, and flow-mediated dilatation (FMD) was triggered by pressure change that equals 15 dynes/cm2 shear stress. Passive dilatation was obtained at the end of the experiment by changing the bathing solution to Ca2+-free Krebs solution with 2 mmol/L EGTA. FMD was calculated as the percentage of diameter changes: (flow-induced dilatation-Phe tone)/(passive dilatation-Phe tone).

Ex Vivo Culture of Mouse Aortas

The mice were euthanized, and aortas were dissected and placed in ice-cold Krebs solution. Each aorta was cut into several ring segments before being cultured in DMEM supplemented with 40% serum collected from db/m+ and db/db mice (10). Ad-GFP and Ad-Cdh5-Klf2 were added to the culture plate to infect the aortic rings for 16 h before mounting to a wire myograph for isometric force measurement.

Laminar Shear Stress

Laminar shear stress (12 dynes/cm2) was generated using a previously reported method (25). Briefly, 5 × 105 HAECs were seeded onto fibronectin (50 μg/mL)-coated glass slides (75 mm × 38 mm; Corning) and cultured in Medium 200 supplemented with 10% FBS. During the laminar shear stress experiment, the culture medium was replaced with Medium 200 supplemented with 2% FBS, and the cells were exposed to shear force for 24 h before harvest.

Real-Time PCR

The quantitative PCR analysis was performed on the ViiA7 Real-Time PCR system (Applied Biosystems) (22). Briefly, samples were homogenized and lysed in TRIzol reagent, and 1 μg total RNA was reversely transcribed using RevertAid Reverse Transcriptase (Thermo Scientific, no. EP0442) to synthesize cDNA. Gene expression was analyzed by the comparative CT method and expressed as the value relative to GAPDH. Please refer to Supplementary Table 1 for the DNA sequences of primers.

Immunoprecipitation and Western Blotting

For immunoprecipitation, cells were homogenized in NP-40 lysis buffer containing protease inhibitors and phosSTOP phosphatase inhibitors (Roche Life Sciences, 04906845001). The lysate was precleared by Sepharose beads (GE Healthcare, 17088601) before incubation overnight with anti-Hsp90 or anti-eNOS antibodies. After incubation with Sepharose beads at 4°C for 2 h the next day, the immune complex was precipitated, washed, and boiled in 1 × denaturing Laemmli sample buffer for 10 min before electrophoresis. For Western blotting, cells or tissues were homogenized in radioimmunoprecipitation assay lysis buffer. The proteins were separated by SDS-PAGE and probed with primary antibodies at 4°C overnight. The polyvinylidene fluoride membranes were washed and subsequently incubated with corresponding secondary antibodies conjugated with horseradish peroxidase (DakoCytomation California, Inc.) before being developed with enhanced chemiluminescence detection on X-ray films. The antibody information is provided in the Supplementary Table 2.

Low-Temperature SDS-PAGE

Low-temperature SDS-PAGE was used for detection of eNOS dimer and monomer. Briefly, protein samples were incubated with loading dye without β-mercaptoethanol at 37°C for 5 min before loading to 7.5% SDS-PAGE gel preequilibrated in 4°C electrophoresis buffer. The blots were probed with anti-eNOS and anti-GAPDH antibodies, followed by horseradish peroxidase-conjugated secondary antibody.

Detection of Reactive Oxygen Species by Dihydroethidium Staining

En face staining of reactive oxygen species (ROS) generation using dihydroethidium (DHE, Ex515/Em585 nm) was performed in thoracic aortas from db/db mice according to the previously described method (26). Briefly, freshly isolated aortic rings were incubated with 5 μmol/L DHE for 15 min at 37°C in extracellular medium. The aortic rings were cut open longitudinally, and the endothelium was placed upside down between two coverslips before being captured on a confocal microscope (FV1000, Olympus, Tokyo, Japan).

Immunohistochemical Staining

Frozen sections of mouse aortas were used for the immunohistochemistry for KLF2. Antigen retrieval was performed in 0.01 mol/L citrate buffer at pH 6.0. After blocking by 5% BSA for 2 h at room temperature, the tissue sections were incubated with the primary antibody anti-KLF2 (1:500; Millipore, 09-820) at 4°C overnight. The signals were developed by using UltraSensitive SP kit (Fuzhou MAXIM Biological Technology Development Co., Ltd., Fujan, China) according to the manufacturer’s instructions. The nuclei were counterstained in hematoxylin.

Nitrite Assay

NO content was presented as its metabolite, and the nitrite level was measured by a Griess assay kit (Life Technologies) according to manufacturer’s instructions. Briefly, HUVECs were pretreated with Ad-GFP and Ad-KLF2 for 24 h, and culture medium was replaced with serum-free endothelial growth medium containing A23187 (1 μmol/L) for 16 h. Absorbance was measured in a spectrophotometric microplate reader at wavelength of 548 nm, and the data were analyzed and converted to nitrite concentrations by comparing calibration readings. For measurement of nitrite level in aortas, the db/db mouse aortas were treated with ACh (10 μmol/L) for 10 min, followed by incubation with nitrate reductase to reduce nitrate to nitrite. Aortas were thereafter homogenized, and the supernatants were collected for quantification of total nitrite level by using the kit. The protein content of tissue homogenates was determined by the Bradford method and used to normalize the nitrite values.

Statistical Analysis

Results represent means ± SEM of n separate experiments. Gene and protein expression were normalized to the expression level of GAPDH and then expressed as relative to the control. Concentration-response curves were constructed using GraphPad Prism 6 software (GraphPad Software, San Diego, CA). Unpaired t test was used to compare differences between two groups, and one-way ANOVA was used to analyze dose-response curves, followed by the Dunnett posttest. P < 0.05 was considered as statistically different.

Data and Resource Availability

All analyzed data in this study are included in this article (and its online supplementary files). Resources available upon request.

KLF2 Expression Is Inhibited by Diabetic Risk Factors While Induced by Running Exercise and Simvastatin Treatment

We isolated aortas from db/m+ and db/db mice and determined levels of KLF2 along with phosphorylated and total eNOS. The results showed that the protein level of eNOS was not changed, but the levels of KLF2 protein and p-eNOS S1176 were reduced in aortas from db/db mice (Fig. 1A). We then mechanically denuded the endothelium of aortas of db/m+ and db/db mice, as evidenced by a marked reduction of the Nos3 gene, which encodes eNOS (Supplementary Fig. 1A). The results showed that the level of Klf2 mRNA was significantly lower in endothelium-intact aortas from db/db mice compared with those from db/m+ mice (Fig. 1B). However, the Klf2 mRNA levels in endothelium-denuded aortas from both strains of mice were comparable (Fig. 1B), suggesting that downregulation of Klf2 mRNA in db/db mouse aortas is likely caused by reduced endothelial expression of KLF2. Diabetes increases circulating proinflammatory mediators such as interleukin 1-β (IL-1β) and advanced glycation end products (AGEs) (27). To test whether diabetic risk factors inhibit KLF2 expression, we treated HAECs with oxidized LDL (Ox-LDL), lysophosphatidylcholine (LPC), IL-1β, and AGEs and showed that KLF2 mRNA expression was suppressed by these risk factors (Fig. 1C). KLF2 expression in ECs is responsive to shear stress, but whether its expression can be upregulated by running exercise remains unclear. We next subjected C57BL/6J mice to treadmill exercise and found, as compared with sedentary mice, 1-week running exercise induced mRNA expression of Klf2 and Nos3 (a known KLF2 downstream target) but not Cnn1 (a smooth muscle marker) in mouse aortic endothelium (Fig. 1D and E and Supplementary Fig. 1B), suggesting that endothelial KLF2 expression and KLF2-mediated transcription is exercise-responsive. Additionally, db/db mice receiving 4-week exercise or simvastatin treatment showed increased expression of Klf2 mRNA in their aortas (Fig. 1F and G), and exercise also increased p-eNOS S1176 and KLF2 protein levels in db/db mouse aortas (Fig. 1H and K). These results demonstrate that reduced endothelial KLF2 expression in diabetes can be reversed by exercise and simvastatin, indicating that the upstream pathway controlling KLF2 expression remains responsive to vasoprotective signals under diabetic conditions.

Figure 1

Endothelial KLF2 expression is inhibited by diabetes and upregulated by exercise and simvastatin treatment. A: Reduced protein level of p-eNOSS1176 and KLF2 in aortas from diabetic db/db mice compared with that from db/m+ mice. B: Reduced Klf2 mRNA level in aortic endothelium from diabetic db/db mice compared with that from db/m+ mice. EC+, with endothelium; EC−, endothelium denuded. *P < 0.05 vs. db/m+ (EC+), two-way ANOVA with the Tukey multiple comparison test. C: KLF2 mRNA expression is suppressed by diabetic risk factors Ox-LDL (100 μg/mL), LPC (100 μmol/L), IL-1β (10 ng/mL), and AGEs (100 μg/mL) in HAECs. *P < 0.05 vs. vehicle, unpaired two-tailed t test. Aortic endothelium from C57BL/6J mice that underwent 1-week treadmill exercise expresses elevated levels of Klf2 (D) and Nos3 (E) mRNA. #P < 0.05 vs. Ex-EC−, ##P < 0.01 vs. Ex-EC−. Ex, exercise; Sed, sedentary. **P < 0.01 vs. sedentary-EC+, #P < 0.05 vs. Ex-EC+, two-way ANOVA with the Tukey multiple comparison test. Upregulated Klf2 mRNA level in mouse aortas from db/db mice that underwent 4-week treadmill exercise (F) and db/db mice treated with simvastatin (SMV) for 4 weeks (G). Representative Western blotting images (H) and summarized results (IK) showing increased protein level of p-eNOSS1176 and KLF2 in aortas from exercised db/db mice compared with those kept sedentary. The results are means ± SEM of n experiments. *P < 0.05 vs. vehicle, **P < 0.01 vs. Sed; N.S., not significant. Unpaired two-tailed t test.

Figure 1

Endothelial KLF2 expression is inhibited by diabetes and upregulated by exercise and simvastatin treatment. A: Reduced protein level of p-eNOSS1176 and KLF2 in aortas from diabetic db/db mice compared with that from db/m+ mice. B: Reduced Klf2 mRNA level in aortic endothelium from diabetic db/db mice compared with that from db/m+ mice. EC+, with endothelium; EC−, endothelium denuded. *P < 0.05 vs. db/m+ (EC+), two-way ANOVA with the Tukey multiple comparison test. C: KLF2 mRNA expression is suppressed by diabetic risk factors Ox-LDL (100 μg/mL), LPC (100 μmol/L), IL-1β (10 ng/mL), and AGEs (100 μg/mL) in HAECs. *P < 0.05 vs. vehicle, unpaired two-tailed t test. Aortic endothelium from C57BL/6J mice that underwent 1-week treadmill exercise expresses elevated levels of Klf2 (D) and Nos3 (E) mRNA. #P < 0.05 vs. Ex-EC−, ##P < 0.01 vs. Ex-EC−. Ex, exercise; Sed, sedentary. **P < 0.01 vs. sedentary-EC+, #P < 0.05 vs. Ex-EC+, two-way ANOVA with the Tukey multiple comparison test. Upregulated Klf2 mRNA level in mouse aortas from db/db mice that underwent 4-week treadmill exercise (F) and db/db mice treated with simvastatin (SMV) for 4 weeks (G). Representative Western blotting images (H) and summarized results (IK) showing increased protein level of p-eNOSS1176 and KLF2 in aortas from exercised db/db mice compared with those kept sedentary. The results are means ± SEM of n experiments. *P < 0.05 vs. vehicle, **P < 0.01 vs. Sed; N.S., not significant. Unpaired two-tailed t test.

Close modal

KLF2 Overexpression Improves Endothelial Function Through Enhancing NO Bioavailability in db/db Mice

Given that KLF2 expression is reduced in diabetic mouse aortic endothelium, we used Ad-mediated overexpression of KLF2 (Ad-KLF2) to determine whether KLF2 upregulation is sufficient to improve the impaired EDR in diabetic mice. Ad-KLF2 administration to db/db mice for 7 days does not affect body weight, insulin sensitivity, and lipid profile (Supplementary Fig. 2). Compared with the Ad-GFP–treated group, Ad-KLF2 increased KLF2 mRNA expression (Supplementary Fig. 3A) and protein level of p-eNOS S1176, eNOS, and KLF2 in db/db mouse aortas (Fig. 2A and Supplementary Fig. 3CE), indicating the successful overexpression of KLF2. Ad-KLF2 also improved ACh-induced EDR in db/db mouse aortas (Fig. 2B), while SNP-induced endothelium-independent relaxations were similar in aortas from both groups (Fig. 2C).

Figure 2

KLF2 overexpression improves endothelium-dependent relaxation through enhancing NO bioavailability in the aorta and main mesenteric arteries from db/db mice. A: Western blotting images show increased protein level of p-eNOSS1176, eNOS, and KLF2 in aortas from db/db mice injected with Ad-KLF2. Concentration-response curves showing ACh-induced endothelium-dependent relaxation in aortas from Ad-GFP– and Ad-KLF2–treated db/db mice (B) and SNP-induced endothelium-independent relaxation (C). *P < 0.05 vs. Ad-GFP, one-way ANOVA with the Tukey multiple comparison test. D: Concentration-response curves show ACh-induced endothelium-dependent relaxation in main mesenteric arteries from Ad-GFP– and Ad-KLF2–treated db/db mice. *P < 0.05 vs. Ad-GFP, one-way ANOVA with the Tukey multiple comparison test. ACh concentration-response curves after incubation with indomethacin plus 20 mmol/L KCl (E) and l-NAME (F) in main mesenteric arteries from Ad-GFP– and Ad-KLF2–treated db/db mice (n = 5). *P < 0.05 vs. Ad-GFP, one-way ANOVA with the Tukey multiple comparison test. G: Immunohistochemistry images showing increased KLF2 expression in aortic endothelium from db/db mice injected with Ad-Cdh5-Klf2. Scale bar: 50 μm. Concentration-response curves showing ACh-induced endothelium-dependent relaxation in aortas from db/db mice treated with Ad-GFP and Ad-Cdh5-Klf2 (H); and SNP-induced endothelium-independent relaxation (I). *P < 0.05 vs. Ad-GFP, one-way ANOVA with the Tukey multiple comparison test. The results are means ± SEΜ of five to six experiments.

Figure 2

KLF2 overexpression improves endothelium-dependent relaxation through enhancing NO bioavailability in the aorta and main mesenteric arteries from db/db mice. A: Western blotting images show increased protein level of p-eNOSS1176, eNOS, and KLF2 in aortas from db/db mice injected with Ad-KLF2. Concentration-response curves showing ACh-induced endothelium-dependent relaxation in aortas from Ad-GFP– and Ad-KLF2–treated db/db mice (B) and SNP-induced endothelium-independent relaxation (C). *P < 0.05 vs. Ad-GFP, one-way ANOVA with the Tukey multiple comparison test. D: Concentration-response curves show ACh-induced endothelium-dependent relaxation in main mesenteric arteries from Ad-GFP– and Ad-KLF2–treated db/db mice. *P < 0.05 vs. Ad-GFP, one-way ANOVA with the Tukey multiple comparison test. ACh concentration-response curves after incubation with indomethacin plus 20 mmol/L KCl (E) and l-NAME (F) in main mesenteric arteries from Ad-GFP– and Ad-KLF2–treated db/db mice (n = 5). *P < 0.05 vs. Ad-GFP, one-way ANOVA with the Tukey multiple comparison test. G: Immunohistochemistry images showing increased KLF2 expression in aortic endothelium from db/db mice injected with Ad-Cdh5-Klf2. Scale bar: 50 μm. Concentration-response curves showing ACh-induced endothelium-dependent relaxation in aortas from db/db mice treated with Ad-GFP and Ad-Cdh5-Klf2 (H); and SNP-induced endothelium-independent relaxation (I). *P < 0.05 vs. Ad-GFP, one-way ANOVA with the Tukey multiple comparison test. The results are means ± SEΜ of five to six experiments.

Close modal

Endothelium-derived relaxing factors (EDRFs) include NO, prostacyclin I2, and endothelium-derived hyperpolarizing factor (EDHF) in smaller arteries (2830). To dissect the role of these EDRFs in contributing to ACh-induced relaxation in Ad-KLF2–treated main mesenteric arteries, we used l-NAME, indomethacin, and 20 mmol/L KCl as inhibitors for NOS, cyclooxygenase, and EDHF, respectively. Ad-KLF2 treatment produced an improvement of EDR in the main mesenteric arteries (Fig. 2D), which was not affected by indomethacin or 20 mmol/L KCl but was reversed by l-NAME treatment (Fig. 2E and F). These results show that NO is the primary form of EDRFs in response to KLF2 overexpression to contribute to the improved EDR in mesenteric arteries of db/db mice.

To exclude a possible endothelium-independent effect of KLF2 overexpression using Ad-KLF2, we generated an endothelium-specific overexpression vector Ad-Cdh5-Klf2 (31) and delivered it to db/db mice for 7 days (Fig. 2G and Supplementary Fig. 3B). Similar to the results observed in the Ad-KLF2 experiment, Ad-Cdh5-Klf2 also improved EDR in aortas from db/db mice (Fig. 2H). SNP-induced relaxation was similar in both Ad-GFP– and Ad-Cdh5-Klf2–treated groups (Fig. 2I). To show a direct effect of endothelial KLF2 overexpression on improvement of EDR, ex vivo Ad-Cdh5-Klf2 transduction was performed in aortic rings isolated from db/db mice, and the results showed a marked improvement of EDR in Ad-Cdh5-Klf2–transduced mouse aortas, again without affecting SNP-induced relaxations (Fig. 4H and I). Taken together, these results demonstrate that augmenting endothelial KLF2 level is effective to improve endothelial function, most likely through increasing NO bioavailability in diabetic mouse arteries.

FMD and Flow-Induced p-eNOS by KLF2

FMD is one of the fundamental mechanisms controlling vascular tone, with NO as an important mediator of flow-induced vasodilatation (31). The impaired FMD in db/db mice was significantly improved by Ad-KLF2 treatment (Fig. 3A–C). Flow-induced p-eNOS S1177 is critical for flow-induced NO production. To determine whether KLF2 is required for flow-induced p-eNOS S1177, we silenced KLF2 using shRNA in HAECs (Fig. 4G and Supplementary Fig. 4A). The Western blotting result showed that KLF2 is required for both the basal and laminar flow-induced p-eNOS S1177 and eNOS in HAECs (Fig. 3D–F).

Figure 3

KLF2 overexpression improves FMD in second-order mesenteric arteries from db/db mice. Representative traces and images of FMD in small mesenteric arteries from db/db mice with Ad-GFP (A) and Ad-KLF2 (B) treatment. C: Summarized data showing FMD was improved by Ad-KLF2 treatment. *P < 0.05 vs. Ad-GFP, unpaired two-tailed t test. Representative Western blotting images (D) and summarized data (E–G) showing KLF2 is required for flow-induced elevation of p-eNOS S1177 level and eNOS expression in human aortic ECs. *P < 0.05, **P < 0.01, ***P < 0.001 vs. SCR at static; ##P < 0.01, ###P < 0.001 vs. SCR at flow, two-way ANOVA with the Tukey multiple comparison test.

Figure 3

KLF2 overexpression improves FMD in second-order mesenteric arteries from db/db mice. Representative traces and images of FMD in small mesenteric arteries from db/db mice with Ad-GFP (A) and Ad-KLF2 (B) treatment. C: Summarized data showing FMD was improved by Ad-KLF2 treatment. *P < 0.05 vs. Ad-GFP, unpaired two-tailed t test. Representative Western blotting images (D) and summarized data (E–G) showing KLF2 is required for flow-induced elevation of p-eNOS S1177 level and eNOS expression in human aortic ECs. *P < 0.05, **P < 0.01, ***P < 0.001 vs. SCR at static; ##P < 0.01, ###P < 0.001 vs. SCR at flow, two-way ANOVA with the Tukey multiple comparison test.

Close modal
Figure 4

Reduced oxidative stress and increased eNOS dimerization in aortas from db/db mice treated with Ad-KLF2. Representative DHE fluorescence images (A) and summarized data of DHE fluorescent intensity (B) indicative of en face ROS levels in aortas from Ad-GFP– and Ad-KLF2–treated db/db mice. *P < 0.05 vs. Ad-GFP, unpaired two-tailed t test. (C) Relative chloromethyl–2′,7′-dichlorodihydrofluorescein diacetate (CM-H2DCFDA) fluorescent intensity indicative of ROS levels in HUVECs exposed to normal glucose (NG, 5.5 mmol/L) and HG (30 mmol/L) and followed by Ad-GFP and Ad-KLF2 treatment. *P < 0.05 vs. Ad-GFP at NG, #P < 0.05 vs. Ad-GFP at HG, two-way ANOVA with the Tukey multiple comparison test. D: The level of eNOS dimer and eNOS monomer in aortas from db/m+ and db/db mice treated with Ad-GFP or Ad-KLF2. E: Ad-KLF2 transduction in HUVECs for 24 h, followed by A23187 (1 μmol/L) stimulation for 16 h, increased NO production indicated by extracellular nitrite levels. *P < 0.05 vs. DMSO, #P < 0.05 vs. A23187, two-way ANOVA with the Tukey multiple comparison test. F: Increased nitrite level in db/db mouse aortas transduced with Ad-Cdh5-Klf2 vector. *P < 0.05 vs. Ad-GFP, unpaired two-tailed t test. G: Western blotting data showing decreased protein nitrotyrosine level by Ad-Cdh5-Klf2 transduction in db/db mouse aortas. H: Ad-Cdh5-Klf2 transduction ex vivo to aortic rings isolated from db/db mice for 16 h improved EDR in response to ACh. *P < 0.05 vs. Ad-GFP, one-way ANOVA with the Tukey multiple comparison test. I: Ad-Cdh5-Klf2 transduction ex vivo did not affect endothelium-independent relaxation in aortic rings isolated from db/db mice in response to sodium nitroprusside.

Figure 4

Reduced oxidative stress and increased eNOS dimerization in aortas from db/db mice treated with Ad-KLF2. Representative DHE fluorescence images (A) and summarized data of DHE fluorescent intensity (B) indicative of en face ROS levels in aortas from Ad-GFP– and Ad-KLF2–treated db/db mice. *P < 0.05 vs. Ad-GFP, unpaired two-tailed t test. (C) Relative chloromethyl–2′,7′-dichlorodihydrofluorescein diacetate (CM-H2DCFDA) fluorescent intensity indicative of ROS levels in HUVECs exposed to normal glucose (NG, 5.5 mmol/L) and HG (30 mmol/L) and followed by Ad-GFP and Ad-KLF2 treatment. *P < 0.05 vs. Ad-GFP at NG, #P < 0.05 vs. Ad-GFP at HG, two-way ANOVA with the Tukey multiple comparison test. D: The level of eNOS dimer and eNOS monomer in aortas from db/m+ and db/db mice treated with Ad-GFP or Ad-KLF2. E: Ad-KLF2 transduction in HUVECs for 24 h, followed by A23187 (1 μmol/L) stimulation for 16 h, increased NO production indicated by extracellular nitrite levels. *P < 0.05 vs. DMSO, #P < 0.05 vs. A23187, two-way ANOVA with the Tukey multiple comparison test. F: Increased nitrite level in db/db mouse aortas transduced with Ad-Cdh5-Klf2 vector. *P < 0.05 vs. Ad-GFP, unpaired two-tailed t test. G: Western blotting data showing decreased protein nitrotyrosine level by Ad-Cdh5-Klf2 transduction in db/db mouse aortas. H: Ad-Cdh5-Klf2 transduction ex vivo to aortic rings isolated from db/db mice for 16 h improved EDR in response to ACh. *P < 0.05 vs. Ad-GFP, one-way ANOVA with the Tukey multiple comparison test. I: Ad-Cdh5-Klf2 transduction ex vivo did not affect endothelium-independent relaxation in aortic rings isolated from db/db mice in response to sodium nitroprusside.

Close modal

KLF2 Attenuates Vascular Oxidative Stress and Increases eNOS Dimerization

The ROS level indicated by dihydroethidium staining in the endothelium of db/db mouse aortas was reduced following Ad-KLF2 treatment (Fig. 4A and B). Exposure of HUVECs to high glucose (HG, 30 mmol/L) for 48 h increased cellular ROS level indicated by chloromethyl–2′,7′-dichlorodihydrofluorescein diacetate fluorescence intensity, and 24 h Ad-KLF2 treatment reduced ROS levels in both normal glucose (5.5 mmol/L) and HG conditions (Fig. 4C). Additionally, KLF2 decreased levels of tyrosine nitrated proteins in db/db mouse aortas (Fig. 4G), suggesting attenuated oxidative stress by KLF2 overexpression. To test whether KLF2 promotes eNOS coupling, Western blotting with low-temperature SDS-PAGE under nonreduced condition was performed. The results showed that KLF2 overexpression for 24 h increased levels of both eNOS dimer and monomer in cultured HUVECs (Supplementary Fig. 4B). To determine whether KLF2 promotes eNOS dimerization in vivo, protein samples from db/m+ mouse aortas treated with Ad-GFP and db/db mouse aortas treated with Ad-GFP or Ad-Cdh5-Klf2 were detected. Compared with db/m+ mouse aortas, the eNOS dimer level was reduced in db/db mouse aortas, which was restored by Ad-Cdh5-Klf2 (Fig. 4D). To examine whether KLF2 increases NO release, A23187 as the calcium ionophore was used as an eNOS activator to induce NO biosynthesis in HUVECs. Ad-KLF2 treatment increased NO production reflected by elevated extracellular nitrite level (Fig. 4E). Consistently, measurement of the nitrite level in KLF2-overexpressed mouse aortas showed that KLF2 increases the nitrite level in these tissues (Fig. 4F).

Transcriptional Regulation of the Upstream Regulators of eNOS Activity by KLF2

To reveal the mechanism by which KLF2 enhances eNOS activity and NO production, we analyzed the RNA-sequencing data showing the changes of mRNA expression in HUVECs overexpressed with KLF2 (22). Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis of KLF2-upregulated genes showed that KLF2 is critical for the regulation of cyclic guanosine monophosphate–protein kinase (PKG)/cAMP-dependent protein kinase (PKA)/insulin pathways (Fig. 5A). Analysis of KLF2-suppresed genes revealed that KLF2 inhibits p53/transforming growth factor-β/FoxO signaling pathways (Fig. 5B), which are activated in diabetic conditions (3234). Further analysis of KLF2-upregulated genes showed that KLF2 increased the expression of genes (PLCB2, PLCB3, PLCB4, PLCD3, PLCG2) encoding phospholipase C (PLC) isotypes (Fig. 5C, D, and G), whose activation is critical for intracellular Ca2+ signaling and diacylglycerol formation (35). KLF2 also induced the expression of INSR (insulin receptor), TEK (angiopoietin-1 receptor), and FLT1 (vascular endothelial growth factor receptor 1) (Fig. 5C, E, and G), whose upregulation can lead to activation of the PI3K-Akt cascade. In addition, KLF2 induces upregulation of genes in the integrin family and genes coding for signaling transducers that lead to activation of PKA (Fig. 5C and F). At the translational and posttranslational level, Ad-KLF2 increased the phosphorylated levels of Akt at serine 473/threonine 308 and eNOS at serine 1177, together with the increased total contents of Akt1 and eNOS (Fig. 5G), thus confirming a stimulatory regulation of Akt-eNOS activity by KLF2 in human ECs.

Figure 5

KLF2 transcriptionally upregulates the expression of upstream regulators critical for eNOS activation. Enriched KEGG pathways for upregulated genes (A) and downregulated genes (B) in KLF2-overexpressed HUVECs. TGF, transforming growth factor. C: Heat map showing key KLF2-induced upregulation of genes involved in eNOS activation. Quantitative PCR data showing KLF2 induces upregulation of genes in PLC family (D), growth factor receptors and integrin family (E), and signaling transducers (F). G: Western blots data showing increased protein expression level of PLCB3, PLCD3, INSR, and activated Akt-eNOS signaling axis in HUVECs overexpressed with KLF2 for 24 h. *P < 0.05 vs. Ad-GFP.

Figure 5

KLF2 transcriptionally upregulates the expression of upstream regulators critical for eNOS activation. Enriched KEGG pathways for upregulated genes (A) and downregulated genes (B) in KLF2-overexpressed HUVECs. TGF, transforming growth factor. C: Heat map showing key KLF2-induced upregulation of genes involved in eNOS activation. Quantitative PCR data showing KLF2 induces upregulation of genes in PLC family (D), growth factor receptors and integrin family (E), and signaling transducers (F). G: Western blots data showing increased protein expression level of PLCB3, PLCD3, INSR, and activated Akt-eNOS signaling axis in HUVECs overexpressed with KLF2 for 24 h. *P < 0.05 vs. Ad-GFP.

Close modal

PI3K-Akt Pathway and Hsp90 Are Critical for KLF2-Induced eNOS Activation

To determine whether PI3K-Akt activation participates in KLF2-induced p-eNOS S1177, HUVECs pretransduced by Ad-KLF2 were treated for 12 h individually with PI3K inhibitors wortmannin (1 μmol/L) and LY294002 (25 μmol/L) as well as a selective Akt inhibitor triciribine (10 μmol/L). The results showed that KLF2-induced an increase in p-eNOS S1177 but not in the total eNOS level, which was reduced by these inhibitors (Fig. 6A and B and Supplementary Fig. 4C), suggesting the involvement of PI3K-Akt activation in KLF2-induced p-eNOS S1177. The abolishment of p-eNOS at serine 615, a site mainly regulated by activated Akt, by triciribine treatment further supports that Akt is critically involved in KLF2-induced eNOS activation in ECs (Fig. 6B).

Figure 6

Required role of PI3K-Akt pathway and Hsp90 in KLF2-induced activation of eNOS. A: Inhibition of PI3K by wortmannin (1 μmol/L) and LY294002 (25 μmol/L) attenuated KLF2-induced phosphorylation of p-eNOSS1177 in HUVECs. B: Inhibition of Akt by triciribine (5 μmol/L) abolished KLF2-induced phosphorylation of p-eNOSS615 and p-eNOSS1177. Immunoprecipitation (IP) of Hsp90 (C) and eNOS (D) in KLF2-overexpressed lysates from HUVECs showed that KLF2 promotes the association between Hsp90 and eNOS. WB, Western blot. E: Inhibition of Hsp90 by 17-AAG (1 μmol/L) attenuated activity of Akt-eNOS signaling and GCH-1 expression induced by KLF2. F: Schematic diagram showing KLF2-mediated signaling events play a critical role in eNOS function and NO production in diabetic ECs. In diabetic endothelium, KLF2 is inhibited but can be increased by exercise-induced laminar flow and statins treatment. KLF2-mediated transcriptional program increases eNOS activity through the PI3K-Akt pathway and Hsp90.

Figure 6

Required role of PI3K-Akt pathway and Hsp90 in KLF2-induced activation of eNOS. A: Inhibition of PI3K by wortmannin (1 μmol/L) and LY294002 (25 μmol/L) attenuated KLF2-induced phosphorylation of p-eNOSS1177 in HUVECs. B: Inhibition of Akt by triciribine (5 μmol/L) abolished KLF2-induced phosphorylation of p-eNOSS615 and p-eNOSS1177. Immunoprecipitation (IP) of Hsp90 (C) and eNOS (D) in KLF2-overexpressed lysates from HUVECs showed that KLF2 promotes the association between Hsp90 and eNOS. WB, Western blot. E: Inhibition of Hsp90 by 17-AAG (1 μmol/L) attenuated activity of Akt-eNOS signaling and GCH-1 expression induced by KLF2. F: Schematic diagram showing KLF2-mediated signaling events play a critical role in eNOS function and NO production in diabetic ECs. In diabetic endothelium, KLF2 is inhibited but can be increased by exercise-induced laminar flow and statins treatment. KLF2-mediated transcriptional program increases eNOS activity through the PI3K-Akt pathway and Hsp90.

Close modal

The activation of eNOS by extracellular stimuli, such as fluid shear stress, requires the binding of Hsp90 to facilitate eNOS dimer formation (18). To examine whether Hsp90 plays a role in KLF2-induced eNOS activation, we performed Hsp90 immunoprecipitation in KLF2-overexpressed lysate from HUVECs and found that KLF2 promoted the association of eNOS with Hsp90, which can be disrupted by Hsp90 chaperoning function inhibitor 17-N-allylamino-17-demethoxygeldanamycin (17-AAG) (Fig. 6C). Moreover, eNOS immunoprecipitation in KLF2-overexpressed HUVEC lysate also showed that increased association of Hsp90 with eNOS was inhibited by 17-AAG (Fig. 6D). Further analysis using Western blotting showed that KLF2-induced Akt-eNOS activation, GCH-1 expression, and eNOS dimerization were all attenuated by 17-AAG (Fig. 6E and Supplementary Fig. 6), thus supporting a required role of Hsp90 in KLF2-mediated activation of Akt-eNOS signaling.

The current study provides several lines of novel and important findings. First, the endothelial KLF2 level is inducible by running exercise and simvastatin treatment, indicating that targeting KLF2 against diabetic endothelial dysfunction is achievable. Second, KLF2 overexpression improves the impaired EDR and FMD in arteries from db/db mice, thereby revealing a vasoprotective role of KLF2 in diabetic conditions. Third, the correction of eNOS dysfunction, a key abnormality in diabetes, reflected by KLF2-induced increase in both phosphorylation and dimerization of eNOS, is likely the major mechanism responsible for vascular benefits of KLF2 activation. Moreover, KLF2 attenuates oxidative stress in aortic ECs in db/db mice and in HUVECs exposed to HG, suggesting an additional pleiotropic effect of KLF2 in protecting endothelial function in diabetes. Finally, the mechanistic study uncovered that KLF2 regulates the transcription of the genes coding for upstream regulators of eNOS activity, thereby revealing a previously unrecognized role of KLF2 in the regulation of PI3K-Akt and phospholipase C-inositol trisphosphate signaling. Taken together, the protective role of KLF2 in endothelial function in db/db mice indicates the effectiveness of targeting KLF2 for restoration of endothelial function in diabetes.

We first observed KLF2 downregulation in diabetic aortic endothelium and in HAECs in response to diabetes risk factors, showing the likely involvement of KLF2 downregulation in diabetic endothelial dysfunction. KLF2 is a flow-responsive transcription factor that is crucially involved in the maintenance of flow-dependent EC phenotype (11). One key upstream regulator of KLF2 under laminar flow is extracellular signal–regulated kinase 5 (ERK5), whose suppression has been identified as a critical mediator of endothelial dysfunction in diabetes and atherogenesis (36). The involvement of ERK5 in diabetic endothelial dysfunction suggests that KLF2 as a key downstream transcriptional effector of ERK5 might also play important roles in pathogenesis of diabetic endothelial dysfunction.

To investigate whether reduced KLF2 can be corrected by vasoprotective strategies, db/db mice were subjected to running exercise on treadmill and simvastatin treatment. The present results show that endothelial Klf2 mRNA level is increased by exercise and simvastatin and that exercise also upregulates aortic KLF2 protein expression in db/db mice, indicating that enhancement of the KLF2 level is a feasible therapeutic option to rescue endothelial function in diabetes. An unclear question in the current study is whether exercise and statin treatment activate KLF2 through a common mechanism. Previous study found that human cardiac ECs treated with statin show increased activity of mitogen/extracellular signal-regulated kinase-5 (MEK5)-ERK5 signaling (37), which is also activated by exercise in skeletal muscle (38). It is possible that the downstream signaling of these two distinct approaches converges on the MEK5-ERK5 signaling cascade to promote KLF2 expression and activity.

A noticeable abnormality in diabetic vasculatures is the impaired EDR and FMD in arteries. The central enzyme controlling EDR and FMD is eNOS, which is inhibited in diabetic situations, although levels of other EDRFs, such as prostacyclin I2, were also decreased in diabetic vasculature (39). The present results show that KLF2 overexpression improved both EDR and FMD in the arteries of db/db mice. The eNOS activity is regulated via multiple mechanisms, including phosphorylation (20), S-glutathionylation (40), subcellular localization (18), and coupling (41). The mechanism underpinning KLF2-increased eNOS activity remains elusive. The current study shows that the improved EDR was inhibited by l-NAME, suggesting that KLF2 preserves EDR through enhancing NO bioavailability in diabetes.

Increased oxidative stress in the vascular wall was reported to quench NO and damage vascular function (16). The expression of KLF2 is required for flow-mediated resistance to oxidative stress, and KLF2 promotes nuclear translocation of Nrf2 (11,42). These findings indicate that KLF2 might be able to reduce overproduction of ROS in diabetic endothelium. In diabetes, increased vascular wall oxidative stress uncouples eNOS and induces vascular dysfunction (43). An important finding on eNOS regulation in the current study is that KLF2 increases eNOS dimerization in vitro and in vivo. We found that KLF2 overexpression increased the expression of GCH-1 (Supplementary Fig. 5AC), the rate-limiting enzyme for de novo synthesis of BH4, which is critical for eNOS dimerization (44,45). This result suggests that KLF2 is a previously unrecognized nuclear effector in the regulation of GCH-1 expression. The Western blotting results showed a difference in KLF2-promoted eNOS dimerization in vitro and in vivo. In cultured human ECs, KLF2 also increased eNOS monomer while KLF2 decreased eNOS monomer in db/db mouse aortas. It is likely that in the presence of other promoting factors of eNOS coupling, such as shear stress, KLF2 in vivo is a determinant mediator of eNOS dimer formation. However, in a cultured condition, no flow is present to facilitate eNOS dimerization. Hence, it is interesting to study whether KLF2 is required for eNOS dimer formation under flow.

To investigate the molecular basis by which KLF2 improves eNOS function, we analyzed the RNA-sequencing data in HUVECs overexpressed with KLF2. The results showed that KLF2-upregulated genes are enriched in the cGMP-PKG–signaling pathway, cAMP-signaling pathway, and insulin-signaling pathway, which are critical upstream regulators of eNOS activity. Further analysis revealed that genes encoding PLC isotypes (PLC-β2–4, PLC-γ2, PLC-δ3) are upregulated by KLF2. Increased expression of PLC facilitates the generation of intracellular inositol 1,4,5-trisphosphate (IP3) and diacylglycerol (DAG), resulting in Ca2+ release and PKC activation (46). IP3 triggers intracellular Ca2+ mobilization from endoplasmic reticulum via interaction with the IP3 receptor, which plays an essential role in Ca2+-dependent eNOS activation in ECs and agonist-induced EDR (47,48). The ability of KLF2 to strongly induce expression of PLC isotypes suggests that KLF2 enhancement is likely to provide an augmented response of vascular endothelium to agonists to improve endothelial function. The other arm of the PLC signaling leads to the activation of PKC by the membrane-retained DAG. Increased PKC activity promotes p-eNOS at threonine 495 (p-eNOS T495) to reduce overall eNOS enzymatic activity (49). We detected p-eNOS T495 level in KLF2-overexpressed HUVECs and found that although KLF2 increased the p-eNOS T495 level, such increase was proportional to the elevated total eNOS level (Supplementary Fig. 5D), suggesting that KLF2 does not increase PKC activity to cause eNOS inhibition, which was confirmed by Western blotting results showing that KLF2 does not affect phosphorylation of PKC-β (Supplementary Fig. 5D) in ECs.

The present results show that KLF2 potently activates Akt to increase p-eNOS S1177, a required step toward eNOS activation. Although the detailed mechanism by which KLF2 activates Akt is not fully resolved in the current study, our results suggest that the KLF2-induced upregulation of growth factor receptors, such as INSR, FLT1, and TEK, is most likely responsible for KLF2-induced Akt activation in ECs. We confirmed that KLF2 increases INSR expression at mRNA and protein levels (Fig. 5E and G), indicating that through regulating insulin receptor expression, KLF2 level is likely critical to endothelial insulin signal transduction. The interaction between Hsp90 and eNOS increases overall enzymatic activity of eNOS. Hsp90 functions as a scaffold for the optimal regulation of eNOS by protein kinases such as Akt (50). Inhibition of Hsp90 reversed KLF2-induced phosphorylation of Akt and eNOS, placing Hsp90 activation at the upstream of the Akt-eNOS axis in response to KLF2 activation. We attempted to understand how KLF2 promotes the Hsp90 activity and showed that KLF2 upregulated genes coding for Hsp90α and Hsp90β without affecting the mRNA level of heat shock factor 1 (HSF1) (Supplementary Fig. 7). The transcription of Hsp90 is mainly controlled by HSF1, and statins were found to activate HSF1 (51,52). The simultaneous activation of HSF1 and KLF2 by statins suggests that posttranslational mechanisms, such as protein-protein interaction between KLF2 and HSF1, are likely to explain the increased Hsp90 activity by KLF2.

In summary, the current study provides several lines of novel and important insights into the regulation of eNOS activity and endothelial function by KLF2 in diabetic mice (Fig. 6F). It is conceivable that pharmacologically targeting KLF2 has potential beneficial effects on preservation of endothelial function in diabetes.

This article contains supplementary material online at https://doi.org/10.2337/figshare.23550072.

J.-Y.L. and C.K.C. contributed equally to this manuscript.

Acknowledgments. The authors thank Dr. Yujie Pu (City University of Hong Kong) for help in obtaining research materials for this project. The authors are also grateful to Dr. Jinghui Dong (Hebei Medical University) for critical comments on the manuscript.

Funding. This study was supported by Research Grants Council of Hong Kong (SRFS2021-4S04, AoE/M/707-18, 14112919, 14109618) and National Natural Science Foundation of China (32241016, 91939302).

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

Author Contributions. J.-Y.L. conceived the project, performed most of the animal and cellular experiments and data analysis, and drafted and revised the manuscript. C.K.C. and L.G. performed mechanistic studies, assisted in the cellular and animal experiments, and made revisions of the manuscript. L.H., L.Z., and L.W. contributed to the study design and performed flow experiments. Y.Z., C.W.L., A.X., and A.F.C. provided crucial reagents and made critical comments on the manuscript. Y.H. supervised the study, provided materials, and contributed to the experimental design and the manuscript preparation and revision. Y.H. 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.

1.
Benjamin
EJ
,
Blaha
MJ
,
Chiuve
SE
, et al.;
American Heart Association Statistics Committee and Stroke Statistics Subcommittee
.
Heart disease and stroke statistics-2017 update: a report from the American Heart Association
.
Circulation
2017
;
135
:
e146
e603
2.
Sena
CM
,
Pereira
AM
,
Seiça
R
.
Endothelial dysfunction—a major mediator of diabetic vascular disease
.
Biochim Biophys Acta
2013
;
1832
:
2216
2231
3.
Economides
PA
,
Caselli
A
,
Tiani
E
,
Khaodhiar
L
,
Horton
ES
,
Veves
A
.
The effects of atorvastatin on endothelial function in diabetic patients and subjects at risk for type 2 diabetes
.
J Clin Endocrinol Metab
2004
;
89
:
740
747
4.
Tousoulis
D
,
Simopoulou
C
,
Papageorgiou
N
, et al
.
Endothelial dysfunction in conduit arteries and in microcirculation. Novel therapeutic approaches
.
Pharmacol Ther
2014
;
144
:
253
267
5.
Cheang
WS
,
Wong
WT
,
Zhao
L
, et al
.
PPARδ is required for exercise to attenuate endoplasmic reticulum stress and endothelial dysfunction in diabetic mice
.
Diabetes
2017
;
66
:
519
528
6.
Cheang
WS
,
Tian
XY
,
Wong
WT
, et al
.
Metformin protects endothelial function in diet-induced obese mice by inhibition of endoplasmic reticulum stress through 5′ adenosine monophosphate-activated protein kinase-peroxisome proliferator-activated receptor δ pathway
.
Arterioscler Thromb Vasc Biol
2014
;
34
:
830
836
7.
SenBanerjee
S
,
Lin
Z
,
Atkins
GB
, et al
.
KLF2 Is a novel transcriptional regulator of endothelial proinflammatory activation
.
J Exp Med
2004
;
199
:
1305
1315
8.
Sen-Banerjee
S
,
Mir
S
,
Lin
Z
, et al
.
Kruppel-like factor 2 as a novel mediator of statin effects in endothelial cells
.
Circulation
2005
;
112
:
720
726
9.
Wang
N
,
Miao
H
,
Li
Y-S
, et al
.
Shear stress regulation of Krüppel-like factor 2 expression is flow pattern-specific
.
Biochem Biophys Res Commun
2006
;
341
:
1244
1251
10.
Song
W
,
Zhang
C-L
,
Gou
L
, et al
.
Endothelial TFEB (transcription factor EB) restrains IKK (IκB Kinase)-p65 pathway to attenuate vascular inflammation in diabetic db/db mice
.
Arterioscler Thromb Vasc Biol
2019
;
39
:
719
730
11.
Parmar
KM
,
Larman
HB
,
Dai
G
, et al
.
Integration of flow-dependent endothelial phenotypes by Kruppel-like factor 2
.
J Clin Invest
2006
;
116
:
49
58
12.
Luo
J-Y
,
Cheng
CK
,
He
L
, et al
.
Endothelial UCP2 is a mechanosensitive suppressor of atherosclerosis
.
Circ Res
2022
;
131
:
424
441
13.
Gimbrone
MA
Jr
,
García-Cardeña
G
.
Endothelial cell dysfunction and the pathobiology of atherosclerosis
.
Circ Res
2016
;
118
:
620
636
14.
Li
Q
,
Atochin
D
,
Kashiwagi
S
, et al
.
Deficient eNOS phosphorylation is a mechanism for diabetic vascular dysfunction contributing to increased stroke size
.
Stroke
2013
;
44
:
3183
3188
15.
Thum
T
,
Fraccarollo
D
,
Schultheiss
M
, et al
.
Endothelial nitric oxide synthase uncoupling impairs endothelial progenitor cell mobilization and function in diabetes
.
Diabetes
2007
;
56
:
666
674
16.
Förstermann
U
,
Sessa
WC
.
Nitric oxide synthases: regulation and function
.
Eur Heart J
2012
;
33
:
829
837
,
837a
837d
17.
Alp
NJ
,
Channon
KM
.
Regulation of endothelial nitric oxide synthase by tetrahydrobiopterin in vascular disease
.
Arterioscler Thromb Vasc Biol
2004
;
24
:
413
420
18.
García-Cardeña
G
,
Fan
R
,
Shah
V
, et al
.
Dynamic activation of endothelial nitric oxide synthase by Hsp90
.
Nature
1998
;
392
:
821
824
19.
Dimmeler
S
,
Fleming
I
,
Fisslthaler
B
,
Hermann
C
,
Busse
R
,
Zeiher
AM
.
Activation of nitric oxide synthase in endothelial cells by Akt-dependent phosphorylation
.
Nature
1999
;
399
:
601
605
20.
Fulton
D
,
Gratton
JP
,
McCabe
TJ
, et al
.
Regulation of endothelium-derived nitric oxide production by the protein kinase Akt
.
Nature
1999
;
399
:
597
601
21.
Lin
Z
,
Kumar
A
,
SenBanerjee
S
, et al
.
Kruppel-like factor 2 (KLF2) regulates endothelial thrombotic function
.
Circ Res
2005
;
96
:
e48
e57
22.
Huang
J
,
Pu
Y
,
Zhang
H
, et al
.
KLF2 mediates the suppressive effect of laminar flow on vascular calcification by inhibiting endothelial BMP/SMAD1/5 signaling
.
Circ Res
2021
;
129
:
e87
e100
23.
Luo
J
,
Deng
Z-L
,
Luo
X
, et al
.
A protocol for rapid generation of recombinant adenoviruses using the AdEasy system
.
Nat Protoc
2007
;
2
:
1236
1247
24.
Tian
XY
,
Wong
WT
,
Wang
N
, et al
.
PPARδ activation protects endothelial function in diabetic mice
.
Diabetes
2012
;
61
:
3285
3293
25.
Wang
L
,
Luo
J-Y
,
Li
B
, et al
.
Integrin-YAP/TAZ-JNK cascade mediates atheroprotective effect of unidirectional shear flow
.
Nature
2016
;
540
:
579
582
26.
Zhang
Y
,
Liu
J
,
Luo
J-Y
, et al
.
Upregulation of angiotensin (1-7)-mediated signaling preserves endothelial function through reducing oxidative stress in diabetes
.
Antioxid Redox Signal
2015
;
23
:
880
892
27.
Donath
MY
,
Shoelson
SE
.
Type 2 diabetes as an inflammatory disease
.
Nat Rev Immunol
2011
;
11
:
98
107
28.
Loscalzo
J
.
The identification of nitric oxide as endothelium-derived relaxing factor
.
Circ Res
2013
;
113
:
100
103
29.
Félétou
M
,
Huang
Y
,
Vanhoutte
PM
.
Endothelium-mediated control of vascular tone: COX-1 and COX-2 products
.
Br J Pharmacol
2011
;
164
:
894
912
30.
Mombouli
JV
,
Vanhoutte
PM
.
Endothelium-derived hyperpolarizing factor(s): updating the unknown
.
Trends Pharmacol Sci
1997
;
18
:
252
256
31.
Green
DJ
,
Dawson
EA
,
Groenewoud
HMM
,
Jones
H
,
Thijssen
DHJ
.
Is flow-mediated dilation nitric oxide mediated?: a meta-analysis
.
Hypertension
2014
;
63
:
376
382
32.
O’Neill
BT
,
Bhardwaj
G
,
Penniman
CM
, et al
.
FoxO transcription factors are critical regulators of diabetes-related muscle atrophy
.
Diabetes
2019
;
68
:
556
570
33.
Yamamoto
T
,
Nakamura
T
,
Noble
NA
,
Ruoslahti
E
,
Border
WA
.
Expression of transforming growth factor beta is elevated in human and experimental diabetic nephropathy
.
Proc Natl Acad Sci U S A
1993
;
90
:
1814
1818
34.
Fiordaliso
F
,
Leri
A
,
Cesselli
D
, et al
.
Hyperglycemia activates p53 and p53-regulated genes leading to myocyte cell death
.
Diabetes
2001
;
50
:
2363
2375
35.
Putney
JW
,
Tomita
T
.
Phospholipase C signaling and calcium influx
.
Adv Biol Regul
2012
;
52
:
152
164
36.
Le
N-T
,
Heo
K-S
,
Takei
Y
, et al
.
A crucial role for p90RSK-mediated reduction of ERK5 transcriptional activity in endothelial dysfunction and atherosclerosis
.
Circulation
2013
;
127
:
486
499
37.
Wilkinson
EL
,
Sidaway
JE
,
Cross
MJ
.
Statin regulated ERK5 stimulates tight junction formation and reduces permeability in human cardiac endothelial cells
.
J Cell Physiol
2018
;
233
:
186
200
38.
Widegren
U
,
Ryder
JW
,
Zierath
JR
.
Mitogen-activated protein kinase signal transduction in skeletal muscle: effects of exercise and muscle contraction
.
Acta Physiol Scand
2001
;
172
:
227
238
39.
Carreras
LO
,
Chamone
DA
,
Klerckx
P
,
Vermylen
J
.
Decreased vascular prostacyclin (PGI2) in diabetic rats. Stimulation of PGI2 release in normal and diabetic rats by the antithrombotic compound Bay g 6575
.
Thromb Res
1980
;
19
:
663
670
40.
Chen
C-A
,
Wang
T-Y
,
Varadharaj
S
, et al
.
S-glutathionylation uncouples eNOS and regulates its cellular and vascular function
.
Nature
2010
;
468
:
1115
1118
41.
Landmesser
U
,
Dikalov
S
,
Price
SR
, et al
.
Oxidation of tetrahydrobiopterin leads to uncoupling of endothelial cell nitric oxide synthase in hypertension
.
J Clin Invest
2003
;
111
:
1201
1209
42.
Fledderus
JO
,
Boon
RA
,
Volger
OL
, et al
.
KLF2 primes the antioxidant transcription factor Nrf2 for activation in endothelial cells
.
Arterioscler Thromb Vasc Biol
2008
;
28
:
1339
1346
43.
Yuan
T
,
Yang
T
,
Chen
H
, et al
.
New insights into oxidative stress and inflammation during diabetes mellitus-accelerated atherosclerosis
.
Redox Biol
2019
;
20
:
247
260
44.
Hattori
Y
,
Nakanishi
N
,
Akimoto
K
,
Yoshida
M
,
Kasai
K
.
HMG-CoA reductase inhibitor increases GTP cyclohydrolase I mRNA and tetrahydrobiopterin in vascular endothelial cells
.
Arterioscler Thromb Vasc Biol
2003
;
23
:
176
182
45.
Antoniades
C
,
Bakogiannis
C
,
Leeson
P
, et al
.
Rapid, direct effects of statin treatment on arterial redox state and nitric oxide bioavailability in human atherosclerosis via tetrahydrobiopterin-mediated endothelial nitric oxide synthase coupling
.
Circulation
2011
;
124
:
335
345
46.
Berridge
MJ
.
The inositol trisphosphate/calcium signaling pathway in health and disease
.
Physiol Rev
2016
;
96
:
1261
1296
47.
Lin
Q
,
Zhao
L
,
Jing
R
, et al
.
Inositol 1,4,5-trisphosphate receptors in endothelial cells play an essential role in vasodilation and blood pressure regulation
.
J Am Heart Assoc
2019
;
8
:
e011704
48.
Yuan
Q
,
Yang
J
,
Santulli
G
, et al
.
Maintenance of normal blood pressure is dependent on IP3R1-mediated regulation of eNOS
.
Proc Natl Acad Sci U S A
2016
;
113
:
8532
8537
49.
Matsubara
M
,
Hayashi
N
,
Jing
T
,
Titani
K
.
Regulation of endothelial nitric oxide synthase by protein kinase C
.
J Biochem
2003
;
133
:
773
781
50.
Qian
J
,
Fulton
D
.
Post-translational regulation of endothelial nitric oxide synthase in vascular endothelium
.
Front Physiol
2013
;
4
:
347
51.
Gomez-Pastor
R
,
Burchfiel
ET
,
Thiele
DJ
.
Regulation of heat shock transcription factors and their roles in physiology and disease
.
Nat Rev Mol Cell Biol
2018
;
19
:
4
19
52.
Fu
Q
,
Wang
J
,
Boerma
M
, et al
.
Involvement of heat shock factor 1 in statin-induced transcriptional upregulation of endothelial thrombomodulin
.
Circ Res
2008
;
103
:
369
377
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 https://www.diabetesjournals.org/journals/pages/license.