OBJECTIVE—Peroxisome proliferator–activated receptor-γ (PPARγ) agonists (thiazolidinediones [TZDs]) are used for the treatment of diabetes. Bone marrow–derived endothelial progenitor cells (EPCs) improve vascular function and predict cardiovascular risk. The effect of pioglitazone therapy on EPCs was examined.

RESEARCH DESIGN AND METHODS AND RESULTS—We performed a prospective, randomized, double-blind study on patients with documented stable coronary artery disease and normal glucose tolerance. Of 54 patients with normal fasting glucose levels, 18 showed impaired glucose tolerance and 36 patients with normal glucose tolerance were randomized to 30-day treatment with pioglitazone (45 mg) or placebo in addition to optimal medical therapy. All patients in the TZD group showed an increase of adiponectin levels as an indicator of compliance (11.4 ± 1.1 to 36.8 ± 2.1 μg/ml; P < 0.001). TZD, but not placebo, decreased mean high-sensitivity C-reactive protein to 43 ± 19% (P < 0.05). Pioglitazone increased CD34+/kinase insert domain receptor+ EPCs to 142 ± 9% and cultured 1,1′-dioctadecyl-3,3,3′,3′-tetramethylindocarbocyanine–labeled acetylated LDL+/lectin+ EPCs to 180 ± 3% (P < 0.05). EPC numbers were not changed in the placebo group. TZD increased the SDF-1–induced migratory capacity to 146 ± 9% per EPC number (P < 0.05) and upregulated the clonogenic potential of EPCs, increasing the colony-forming units to 172 ± 12% (P < 0.001). In cultured human EPCs, TZD increased EPC numbers and migration and reduced NADPH-oxidase activity. The TZD effect was reversed by the PPARγ antagonist GW9662 and mimicked by treatment with adiponectin.

CONCLUSIONS—The PPARγ agonist pioglitazone increases the number and function of EPCs in patients with coronary artery disease. The effect represents a potential regenerative mechanism in atherosclerosis and is observed in normoglycemic individuals with stable coronary artery disease.

Peroxisome proliferator–activated receptor-γ (PPARγ) agonists (thiazolidinediones [TZDs]) lower serum glucose levels in patients with type 2 diabetes (1). In addition to their insulin-sensitizing effects, increasing evidence suggests that these drugs improve endothelium-dependent vascular function and prevent atherosclerotic disease progression (27). Furthermore, experiments on cultured vascular cells support a direct and beneficial modulation of key regulators of atherosclerosis, such as cellular adhesion molecules, tissue factor, plasminogen activator inhibitor, and matrix metalloproteinases (rev. in 810). These findings have led to the hypothesis that TZDs may exert vasculoprotective effects independently of their metabolic action (9).

Cardiovascular function and angiogenesis have been shown to be significantly modulated by circulating premature cells derived from the bone marrow (11). A subset of these stem cells named endothelial progenitor cells (EPCs) (12) enhances angiogenesis, promotes vascular repair, and improves endothelial function (1319). Recently, it was shown that reduced levels of circulating EPCs represent a cellular marker that independently predicts outcome in patients with vascular disease (20,21). The circulating numbers and function of EPCs are regulated. Vascular risk factors, and especially type 2 diabetes, have been shown to reduce EPC numbers and impair EPC function (14,2227). On the other hand, lipid lowering with statins or physical activity are interventions capable of raising EPC numbers and improving their function (13,15,24,28,29). Similarly, glucose-lowering treatment increases EPC numbers in diabetic individuals (30,31). Interestingly, experiments in cultured cells and in mice have suggested that treatment with TZD upregulates EPCs (3234).

On the basis of these preclinical data, we hypothesized that the PPARγ agonist pioglitazone may modulate the number and function of EPCs. We further speculated that these effects may be independent of serum glucose lowering. The effect of treatment with pioglitazone on EPCs was tested in a prospective, randomized, double-blind clinical trial on patients with stable coronary artery disease and documented normal glucose tolerance. We examined the level of CD34+/kinase insert domain receptor (KDR)+ mononuclear cells because this cell population was shown to predict the occurrence of cardiovascular events and death from cardiovascular causes (20). Furthermore, the effects of TZD on parameters of EPC function were characterized.

The study was designed as a prospective, randomized, double-blind clinical trial on patients with stable coronary artery disease and normal glucose tolerance. The trial was conducted with approval of the local ethics committee (Ethikvotum 130/05) and the German Federal Institute for Drugs and Medical Devices (Bfarm no. 4030855; Eudora-CT no. 2005-003939-42). Informed consent was obtained from all subjects.

All patients had angiographically documented and clinically stable coronary artery disease. Medication was not changed during the course of the study. Exclusion criteria included functional New York Heart Association class III or IV heart failure, left ventricular dysfunction measured as left ventricular ejection fraction <40%, elevated serum creatinine level, active liver disease, alanine transaminase levels of ≥2.5 times the upper limit of normal, regular use of nonsteroidal anti-inflammatory drugs, psychiatric diseases, and pregnancy.

Fifty-four eligible patients with normal fasting glucose underwent an oral glucose tolerance test (Dextro O.G-T.; Roche). Eighteen patients showed impaired glucose tolerance, and 36 patients with normal glucose tolerance were enrolled in the study. They were randomized to a 30-day treatment with pioglitazone (45 mg) or matching placebo in addition to an optimized cardiovascular medication. Randomization was performed by the pharmacy of the Universitätsklinikum des Saarlandes independently from the investigators. Venous blood samples were taken on the day of enrolment in the study and after 30 days of placebo/TZD therapy. Adiponectin serum levels were quantified by the human adiponectin radio immunoassay (Linco). High-sensitivity C-reactive protein (hsCRP) was measured by a turbidimetric latex test (CRP Dynamik; Biomed).

Flow cytometry analysis of EPCs.

Fluorescence-activated cell sorter (FACS) analyses were used to characterize mononuclear cells (MNCs) as previously described (20). MNCs were selected using Ficoll density gradient centrifugation (Biocoll Separating Solution; Biochrom) from 20 ml human blood drawn in sodium citrate and resuspended in 1 ml endothelial cell basal medium (EBM; CellSystems) with supplements (1 μg/ml hydrocortisone, 3 μg/ml bovine brain extract, 30 μg/ml gentamicin, 50 μg/ml amphotericin B, 10 μg/ml human endothelial growth factor, and 20% FCS). One hundred microliters was diluted in FACS buffer (PBS, 0.1% bovine albumin, and 15 units/l aprotinine) and immediately used for assessment of isotype controls and CD34-fluorescein isothiocyanate (CD34-FITC; Becton Dickinson)/goat anti-human KDR (R&D Systems) with the conjugated secondary antibody anti-goat streptavidin-phycoerythrin (PE) (DAKO). All antibody incubations were kept on ice in the dark. Isotype identical antibodies (IgG 2a-FITC and IgG 2a-PE; Becton Dickinson) served as controls in every experiment, and unspecific binding was blocked by addition of human serum. Cells were washed twice in FACS buffer and fixed with 2% paraformaldehyde. Each measurement was performed in two separate tubes by assessment of 105 MNCs in the lymphocyte gate using the Becton Dickinson FACSCalibur and Cell Quest Pro software. The intra-assay variability (coefficient of variation) was similar between groups and assays: FACS analysis of CD34+/KDR+ EPCs, placebo 28% and TZD 29%; 1,1′-dioctadecyl-3,3,3′,3′-tetramethylindocarbocyanine–labeled acetylated LDL (DiLDL)+/lectin+ EPCs, placebo 29% and TZD 30%. The interassay correlation between CD34+/KDR+ EPCs and DiLDL+/lectin+ EPCs was R2 = 0.54 (P < 0.01).

Cell culture of circulating EPCs.

MNCs were isolated by Ficoll density gradient centrifugation, the cell pellet was resuspended in 5 ml EBM (Cell Systems), and 4 × 106 mononuclear cells were cultured on fibronectin-coated dishes in EBM (CellSystems) (18,35). After 4 days in culture, adherent cells were incubated with DiLDL (2.4 μg/ml; CellSystems) and stained using FITC-labeled Ulex europaeus agglutinin I (lectin, 10 μg/ml) (Sigma). All measurements were performed in triplicates. Double-positive cells were counted manually by two independent observers blinded to the study (Nikon Eclipse E600; magnification ×10). Expression of PPARγ in human EPCs was examined by RT-PCR: PPAR1 forward, 5′-CTGGCCTCCTTGATGAATAA; PPAR1 reverse, 5′-GGCGGTCTCCACTGAGAATA; PPAR2 forward, 5′-AGGGCGATCTTGACAGGAAAG; PPAR2 reverse, 5′-CCCATCATTAAGGAATTCATGTCA. To test the effects of pioglitazone in vitro, EPCs isolated and cultured from healthy volunteers were treated with pioglitazone (10 μmol/l; Takeda Pharmaceuticals), the PPARγ antagonist GW9662 (1 μmol/l per l; Alexis Biochemicals), or adiponectin (20 μg/ml; Alexis Biochemicals) for 24 h.

Migration assay.

Modified Boyden chambers were used to assess the migratory capacity of EPCs (25,35). On day 1, 4 × 106 MNCs were plated on noncoated six-well culture dishes and cultured for 4 days. Culture medium was removed, and cells were harvested by trypsination and centrifugation. The pellet was resuspended in 300 μl EBM and counted. Cells (1 × 105) in 500 μl EBM without supplements were placed in the upper chamber (HTS Fluoroblock, 8 μm pore size, in triplicate; BD Biosciences), and the chamber was placed in a 24-well plate containing EBM without supplements and 100 ng/ml SDF-1 (R&D Systems). After 24 h, the filter was carefully removed, washed, fixed, and incubated with labeled DiLDL as described above. SDF-1–stimulated migratory capacity was then quantified by counting the migrated EPCs on the lower surface of the filter using fluorescence microscopy (magnification ×40, in triplicate).

Colony-forming units.

Colony-forming units (CFUs) were counted to examine the clonal expansion capacity of EPCs. After density gradient centrifugation, 5 × 106 MNCs were plated in a fibronectin-coated six-well plate for 48 h (in triplicate). Then, the nonadherent cells in the supernatant were centrifuged and resuspended, and 106 cells were plated into 24-well plates. After 5 more days in culture, EPC colonies (defined as clusters of more than 15 cells) were counted under a light microscope. Values are expressed as absolute colony number per well.

NADPH oxidase activity.

NADPH oxidase activity was measured by a lucigenin-enhanced chemiluminescence assay in buffer B containing 50 mmol/l phosphate (pH 7.0), 1 mmol/l EGTA, protease inhibitors (Complete; Roche), 150 mmol/l sucrose, 0.005 mmol/l lucigenin, and 0.1 mmol/l NADPH, as described previously (36). EPCs were lysed after washing with PBS in ice-cold buffer B, lacking lucigenin and substrate. Basal and phorbol myristate acetate (PMA)-stimulated NADPH oxidase activity was determined in 100 μg aliquots of the sample measured over 10 min in quadruplicates using NADPH as substrate in a scintillation counter (Berthold Lumat LB 9501) in 1-min intervals.

Statistical analysis.

Results are represented as mean ± SE. Statistical analysis was performed using the GraphPad Prism software (version 3.02). ANOVA and Student's t test were applied where applicable. Post hoc comparisons were made by using the Bonferroni test. Values of P < 0.05 were considered statistically significant.

Patient characteristics.

The baseline characteristics of the patients are summarized in Table 1 and did not differ between groups. There was no difference in medication between groups. The medication was not changed during the course of the study. Serum glucose and lipid concentrations were not different between groups and were not affected by treatment with pioglitazone. TZD treatment significantly reduced insulin concentrations and the homeostasis model assessment (HOMA) index (Table 1). None of the smokers stopped smoking during the course of the trial. Nine TZD and 10 placebo patients underwent coronary angioplasty or stenting (one drug-eluting stent in each group). The effect of TZD on EPC parameters did not differ between percutaneous coronary intervention (PCI) and non-PCI patients. Six individuals in the placebo group and four in the TZD group met the current International Diabetes Federation criteria for metabolic syndrome.

Adiponectin and hsCRP serum levels.

Adiponectin is a peptide hormone regulator produced in adipocytes, which is known to be increased by TZD treatment (1,37). The mean adiponectin level in the placebo group was 12.2 ± 1.2 μg/ml before and 12.2 ± 1.4 μg/ml after the study. In contrast, the adiponectin serum concentrations of the TZD group showed an increase to 322% after 4 weeks (11.4 ± 1.1 vs. 36.8 ± 2.1 μg/ml; P < 0.001) (Fig. 1A). Importantly, in every individual TZD patient, but no patient of the placebo group, the adiponectin level increased by more than twofold.

hsCRP serum concentration is a marker of vascular inflammation and independent predictor of vascular events. Treatment with the PPARγ agonist reduced hsCRP levels from 3.9 ± 0.7 to 1.69 ± 0.3 mg/l (P < 0.05; Fig. 1B), whereas placebo treatment had no effect (3.8 ± 0.7 vs. 2.9 ± 0.7 mg/l).

Number of EPCs in normoglycemic patients.

CD34+/KDR+ EPCs have been shown to predict cardiovascular events (20). Figure 2A depicts a representative FACS scan for CD34-FITC/KDR-PE in peripheral blood. Treatment with TZD for 30 days resulted in upregulation of CD34+/KDR+ EPC numbers to 142% (55 ± 5 vs. 78 ± 6/105 cells; P < 0.01; Fig. 2B). In the placebo group, the number of CD34+/KDR+ EPCs did not change.

Number of cultured DiLDL+/lectin+ EPCs.

Quantification of DiLDL uptake and lectin staining was used as an established second method to quantify and characterize EPCs (Fig. 3A) (11). Placebo treatment had no effect on the number of DiLDL+/lectin+ double-positive cells per microscopic field (122 ± 5 before treatment vs. 103 ± 4 after treatment; P > 0.05). In contrast, DiLDL+/lectin+ EPCs increased from 86 ± 4 to 154 ± 5 cells/microscopic field after TZD treatment (P < 0.05).

Migratory capacity.

Migration is an important functional property of EPCs that can be regulated independent of EPC numbers (11,25). After 1 month of therapy, there was no change in placebo controls, whereas TZD treatment resulted in an increase of EPC migration to 146 ± 9% per EPC number (P < 0.05; Fig. 4).

Colony-forming potential.

The ability to clonally expand and to create colonies in an endothelial-specific medium is a key functional feature of EPCs (14). There was no significant change in CFUs in patients treated with placebo (99 ± 9 CFUs at baseline vs. 119 ± 8 after 1 month; P > 0.05). After treatment with TZD, however, the development of CFUs per number of EPCs was upregulated to 172 ± 12%; P < 0.001; Fig. 5A).

NADPH oxidase activity.

The NADPH oxidase is an important source of endothelial superoxide anions (38). Reactive oxygen species impair the function of both mature and immature endothelial cells (16,39,40). Treatment of cultured EPCs from healthy volunteers (n = 4) with pioglitazone (10 μmol/l, 24 h) decreased basal NADPH oxidase activity from 40 ± 3.29 to 16 ± 8.34 relative light units (RLU)/μg. Similarly, TZD was able to prevent the PMA-induced increase of NADPH oxidase activity (from 72 ± 5.14 to 44 ± 5.45 RLU/μg) (Fig. 5B).

Regulation by PPARγ and adiponectin.

The RT-PCR analysis showed that PPARγ is expressed in human EPCs. To test whether upregulation of EPCs is mediated via PPARγ, the EPCs were treated with pioglitazone (10 μmol/l 24 h) alone and in the presence of the PPARγ antagonist GW9662 (1 μmol/l, 24 h). TZD treatment increased EPC numbers to 189 ± 15% (P < 0.05). Figure 6A shows that the increase of EPCs in the presence of TZD is completely inhibited by the PPARγ antagonist (n = 4; P < 0.05). In vitro treatment with pioglitazone (10 μmol/l, 24 h) increased the migratory capacity per number of EPCs to 158 ± 10%; the effect was completely prevented by cotreatment with GW9662 (1 μmol/l, 24 h). The upregulation of EPC numbers and the improvement of migratory function by TZD were mimicked by treatment of the cultured EPCs with adiponectin at a concentration similar to that observed in patients after TZD treatment (20 μg/ml, 24 h) (Fig. 6B).

The main finding of this prospective, randomized trial is the demonstration of a novel vascular effect of TZDs in normoglycemic individuals with coronary artery disease. Treatment with pioglitazone increased the number and the function of circulating EPCs. The effects of TZD on EPCs occurred on top of the treatment with aspirin, β-blockers, and inhibitors of the renin-angiotensin system and, importantly, in the presence of statin treatment.

To evaluate the adherence to the study medication, serum adiponectin concentrations were measured. Adiponectin has been shown to be increased by TZD treatment mediated by an effect on adipocytes contributing to improved insulin sensitivity (1,37). Adiponectin levels at least doubled in all of the pioglitazone-treated individuals, but no placebo-administered patient exhibited a similar increase. These findings suggest a very good compliance. To test whether adiponectin contributes to the regulation of EPCs by TZD, cultured human EPCs were exposed to adiponectin at concentrations similar to those observed in the patients taking TZD. Adiponectin potently increased both EPC numbers and function, suggesting that adiponectin contributes as a mediator to the effects of TZD on EPCs.

In agreement with previous observations, TZD treatment significantly lowered the established marker of vascular inflammation hsCRP (8,41). Our data show that this effect can be observed in normoglycemic patients with coronary artery disease after 30 days of treatment. These data are in agreement with recent data showing that treatment of isolated EPCs with recombinant CRP inhibits EPC differentiation, survival, and function and caused a concentration-dependent increase in reactive oxygen species production and apoptosis. Interestingly, the PPARγ agonist rosiglitazone was able to inhibit the negative effects of CRP on EPC biology (42,43). Most of these effects, however, were observed at a CRP concentration of 15 μg/ml, which is significantly higher than in our study population with mean hsCRP values of ≤5 μg/ml.

Several markers have been used to identify EPCs. Here, CD34+/KDR+ EPCs were quantified because they have been shown to predict cardiovascular outcomes in patients with coronary artery disease. The recent EPCAD (Endothelial Progenitor Cells Coronary Artery Disease) study showed an association between this population of EPCs and death from cardiovascular causes that is independent of the severity of coronary artery disease, cardiovascular risk factors, and medication known to influence cardiovascular outcomes (20). In addition to FACS analysis, we used a second, independent method of EPC characterization by culturing mononuclear cells and selection of EPCs by endothelial growth factors, adhesion to fibronectin, the ability to uptake LDL, and staining for lectin (11,14). As a third established method, we quantitated EPC CFUs, which have been shown to predict endothelial function in humans (14). All three methods of EPC quantification showed a robust increase of EPC numbers in patients taking the PPARγ agonist.

In addition to EPC numbers, functional properties of EPCs have been shown to determine cardiovascular disease (14,20). Recent data suggest that EPC function may be impaired by cardiovascular risk factors independent of the EPC number (26,27). In mice and in cultured human EPCs, pioglitazone prevents apoptotic cell death of EPCs by a mechanism involving phosphatidylinositol 3-kinase (34). Intracellular reactive oxygen species, such as superoxide and H2O2, impair the function of mature and immature endothelial cells, e.g., by promoting cell death (16,39,40). Of the several sources within vascular cells, the multi-subunit NADPH oxidase is a predominant contributor of endothelial superoxide free radicals (38). Here, cell culture experiments show that TZD treatment reduces basal and PMA-stimulated NADPH oxidase activity in EPCs, pinpointing a mechanism by which TZD treatment reduces EPC apoptosis and increases EPC numbers. Two widely studied functional characteristics of EPCs are their potential to migrate and their capacity to replicate. These features are likely to be of great importance for the improvement of endothelial function, neoangiogenesis, and inhibition of atherogenesis (44). The data show that TZD treatment increased both the migratory and the colony forming capacity per number of EPCs in patients with normal glucose tolerance, suggesting that the biological effect of TZD on EPCs may be significantly greater than the extent of the increase of EPC numbers. These effects are mediated by PPARγ because GW9662 reversed the effects of pioglitazone on EPC numbers and migration.

Cardiovascular disease accounts for ∼70% of mortality in patients with diabetes. Prospective studies show that compared with their nondiabetic counterparts, the relative risk of cardiovascular mortality for men with diabetes is two to three and for women with diabetes is three to four. Several large trials have demonstrated that optimal control of blood pressure and LDL cholesterol level can substantially reduce excess cardiovascular risk in patients with diabetes (45,46). Metabolic control alone is not sufficient for the prevention of cardiovascular events in patients with diabetes (47). However, even with optimal control of the potent cardiovascular risk factors blood pressure and LDL cholesterol, incremental risk for cardiovascular events remains high compared with individuals without diabetes (45,46). Beneficial effects of TZDs on vascular inflammation and function seem to be possibly independent of glucose lowering and have been demonstrated in nondiabetic, healthy individuals (2,4,48,49). EPCs are independent predictors of endothelial function, cardiovascular events, and cardiovascular mortality (14,20,21). Patients with diabetes are characterized by an impairment of EPC function that is observed independently of other cardiovascular risk factors (22,25,27). On the basis of our study, it is interesting to speculate that patients with diabetes may benefit from PPARγ agonists in addition to insulin sensitization by upregulation of EPCs. This needs to be confirmed in further trials.

Here, we show that treatment with pioglitazone powerfully improves number and function of EPCs in patients with vascular disease and normal glucose tolerance. Serum glucose and lipid concentrations were not affected by treatment with pioglitazone. TZD treatment reduced insulin concentrations and the HOMA index, suggesting that TZDs exert metabolic effects that occur in normoglycemic individuals and that do not result in significant changes of serum glucose (such as regulation of adiponectin and insulin). The effects of TZDs in mice and in isolated cultured EPCs suggest that some of these effects are effective on the level of progenitor cells. Until now, TZDs are only approved and used for individuals with elevated serum glucose levels. These clinical data viewed together with preclinical studies therefore evoke the provocative hypothesis that these agents may be also beneficial for patients with vascular disease despite normal glucose tolerance, such as normoglycemic individuals with stable coronary artery disease.

The design of the study has limitations. It does not allow answering the question of whether TZD therapy confers a prognostic benefit to nondiabetic coronary artery disease patients reducing cardiovascular events at long-term follow-up. Furthermore, subgroups of patients that may especially benefit from TZD treatment and the kinetics and time course of the effects of TZD on EPCs are not known. Finally, the intracellular signaling pathways mediating the effect of TZD on number and function of EPCs remain to be elucidated. These issues need to be addressed in further studies. However, this is a proof-of-concept study providing the first evidence for a new mechanism of TZD action and setting the stage for large outcome trials using risk factor stratification by EPC numbers and function.

In summary, EPCs have emerged as a new dimension of vascular biology. Increasing evidence suggests that bone marrow–derived adult stem cells significantly contribute to vascular and cardiac function. Improvement of EPC function may represent a novel and relevant protective mechanism of TZDs that could potentially benefit patients with vascular diseases in the absence of manifest diabetes.

FIG. 1.

Effect of TZD treatment for 30 days on serum concentrations of adiponectin (A) and hsCRP (B).

FIG. 1.

Effect of TZD treatment for 30 days on serum concentrations of adiponectin (A) and hsCRP (B).

Close modal
FIG. 2.

A: Example of a FACS scan depicting the gating (top row) and scatter graph (bottom row) for a representative patient from the TZD group showing CD34+/KDR+ EPCs at baseline (pre, left column) and after 1 month (post, right column) of TZD treatment. B: Quantification of the CD34+/KDR+ EPCs before and after therapy. The whiskers comprise minimum and maximum values and the box shows the range of 25–75% quartiles and the median in between. *P < 0.01 in the TZD group. Each measurement was in duplicate. SSC, sideward scatter; FSC, forward scatter.

FIG. 2.

A: Example of a FACS scan depicting the gating (top row) and scatter graph (bottom row) for a representative patient from the TZD group showing CD34+/KDR+ EPCs at baseline (pre, left column) and after 1 month (post, right column) of TZD treatment. B: Quantification of the CD34+/KDR+ EPCs before and after therapy. The whiskers comprise minimum and maximum values and the box shows the range of 25–75% quartiles and the median in between. *P < 0.01 in the TZD group. Each measurement was in duplicate. SSC, sideward scatter; FSC, forward scatter.

Close modal
FIG. 3.

A: Representative fluorescence microscopy showing DiLDL+/lectin+ EPCs in a patient from the placebo versus one of the TZD group (magnification ×10). Blue, DAPI (nuclei); green, FITC-lectin; red, DiLDL. B: Quantification of DiLDL+/lectin+ EPCs shown as mean ± SE. Each assay was in triplicate, with three random fields counted from each coverslip. Lectin, U. europaeus agglutinin I. (Please see http://dx.doi.org/10.2337/db07-0069 for a high-quality digital representation of this figure.)

FIG. 3.

A: Representative fluorescence microscopy showing DiLDL+/lectin+ EPCs in a patient from the placebo versus one of the TZD group (magnification ×10). Blue, DAPI (nuclei); green, FITC-lectin; red, DiLDL. B: Quantification of DiLDL+/lectin+ EPCs shown as mean ± SE. Each assay was in triplicate, with three random fields counted from each coverslip. Lectin, U. europaeus agglutinin I. (Please see http://dx.doi.org/10.2337/db07-0069 for a high-quality digital representation of this figure.)

Close modal
FIG. 4.

A: Representative fluorescence microscopic images (magnification ×40) showing the effects of TZD and placebo on EPC migration stimulated by 100 ng/ml SDF-1 in a Boyden chamber. B: Quantification. Each assay was performed in triplicate.

FIG. 4.

A: Representative fluorescence microscopic images (magnification ×40) showing the effects of TZD and placebo on EPC migration stimulated by 100 ng/ml SDF-1 in a Boyden chamber. B: Quantification. Each assay was performed in triplicate.

Close modal
FIG. 5.

A: Effect of TZD or placebo on the number of CFUs. Six wells were counted for each patient. B: Cultured human EPCs. NADPH oxidase activity measured by a lucigenin-enhanced chemiluminescence assay after treatment of cultured human EPCs with TZD (10 μmol/l pioglitazone). Basal NADPH oxidase activity was compared with PMA-stimulated NADPH oxidase activity (n = 4, *P < 0.05).

FIG. 5.

A: Effect of TZD or placebo on the number of CFUs. Six wells were counted for each patient. B: Cultured human EPCs. NADPH oxidase activity measured by a lucigenin-enhanced chemiluminescence assay after treatment of cultured human EPCs with TZD (10 μmol/l pioglitazone). Basal NADPH oxidase activity was compared with PMA-stimulated NADPH oxidase activity (n = 4, *P < 0.05).

Close modal
FIG. 6.

Effect of 24-h treatment with pioglitazone (TZD; 10 μmol/l), the PPARγ antagonist GW9662 (GW) (1 μmol/l), and adiponectin (Adi) (20 μg/ml) in cultured human EPCs (n = 4, mean ± SE, *P < 0.05). A: Quantification of DiLDL+/lectin+ EPCs. Lectin, U. europaeus agglutinin I. B: EPCs migration was stimulated by 100 ng/ml SDF-1 and quantified in modified Boyden chambers.

FIG. 6.

Effect of 24-h treatment with pioglitazone (TZD; 10 μmol/l), the PPARγ antagonist GW9662 (GW) (1 μmol/l), and adiponectin (Adi) (20 μg/ml) in cultured human EPCs (n = 4, mean ± SE, *P < 0.05). A: Quantification of DiLDL+/lectin+ EPCs. Lectin, U. europaeus agglutinin I. B: EPCs migration was stimulated by 100 ng/ml SDF-1 and quantified in modified Boyden chambers.

Close modal
TABLE 1

Patient characteristics

PlaceboTZDP value
n 17  18  NS 
Sex (male/female) 13/4  12/6  NS 
Age (years) 63 ± 2.48  55 ± 2.14  NS 
BMI (kg/m227 ± 0.94  27 ± 0.77  NS 
Hypertension 100  78  NS 
Cigarette smoking (active) 24  39  NS 
Hyperlipidemia 88  88  NS 
Positive family history 35  61  NS 
Aspirin 100  100  NS 
β-Blockers 88  89  NS 
ACEI/ARB 94  83  NS 
Statins 94  94  NS 
Nitrates 12  11  NS 
 Before After Before After  
Systolic blood pressure (mmHg) 141 ± 5.4 135 ± 5.1 136 ± 4.8 134 ± 3.9 NS 
Diastolic blood pressure (mmHg) 73 ± 2.5 74 ± 3.1 74 ± 2.5 75 ± 2.4 NS 
Heart rate (min−164 ± 2.9 63 ± 2.3 63 ± 2.0 63 ± 2.1 NS 
Fasting glucose (mg/dl) 100 ± 3.4 103 ± 6.2 97 ± 1.7 90 ± 5.2 NS 
Serum insulin (μIU/ml) 10.7 ± 2.1 11.6 ± 2.9 9.6 ± 1.7 5.8 ± 1.3 <0.05 
HOMA index 2.3 ± 0.56 2.4 ± 0.54 2.4 ± 0.39 1.2 ± 0.27 <0.05 
Cholesterol (mg/dl) 191 ± 10.2 183 ± 7.5 199 ± 6.8 179 ± 6.7 NS 
HDL cholesterol (mg/dl) 51 ± 3.5 43 ± 2.3 57 ± 3.4 47 ± 3.0 NS 
LDL cholesterol (mg/dl) 116 ± 9.4 109 ± 6.7 119 ± 8.8 116 ± 5.9 NS 
Triglycerides (mg/dl) 142 ± 12.8 162 ± 20.2 146 ± 16.4 133 ± 10.3 NS 
PlaceboTZDP value
n 17  18  NS 
Sex (male/female) 13/4  12/6  NS 
Age (years) 63 ± 2.48  55 ± 2.14  NS 
BMI (kg/m227 ± 0.94  27 ± 0.77  NS 
Hypertension 100  78  NS 
Cigarette smoking (active) 24  39  NS 
Hyperlipidemia 88  88  NS 
Positive family history 35  61  NS 
Aspirin 100  100  NS 
β-Blockers 88  89  NS 
ACEI/ARB 94  83  NS 
Statins 94  94  NS 
Nitrates 12  11  NS 
 Before After Before After  
Systolic blood pressure (mmHg) 141 ± 5.4 135 ± 5.1 136 ± 4.8 134 ± 3.9 NS 
Diastolic blood pressure (mmHg) 73 ± 2.5 74 ± 3.1 74 ± 2.5 75 ± 2.4 NS 
Heart rate (min−164 ± 2.9 63 ± 2.3 63 ± 2.0 63 ± 2.1 NS 
Fasting glucose (mg/dl) 100 ± 3.4 103 ± 6.2 97 ± 1.7 90 ± 5.2 NS 
Serum insulin (μIU/ml) 10.7 ± 2.1 11.6 ± 2.9 9.6 ± 1.7 5.8 ± 1.3 <0.05 
HOMA index 2.3 ± 0.56 2.4 ± 0.54 2.4 ± 0.39 1.2 ± 0.27 <0.05 
Cholesterol (mg/dl) 191 ± 10.2 183 ± 7.5 199 ± 6.8 179 ± 6.7 NS 
HDL cholesterol (mg/dl) 51 ± 3.5 43 ± 2.3 57 ± 3.4 47 ± 3.0 NS 
LDL cholesterol (mg/dl) 116 ± 9.4 109 ± 6.7 119 ± 8.8 116 ± 5.9 NS 
Triglycerides (mg/dl) 142 ± 12.8 162 ± 20.2 146 ± 16.4 133 ± 10.3 NS 

Data are n and means ± SE. ACEI, angiotensin-converting enzyme inhibitor; ARB, AT1-receptor blocker.

Published ahead of print at http://diabetes.diabetesjournals.org on 10 July 2007. DOI: 10.2337/db07-0069.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked “advertisement” in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

U.L. has received support from the Deutsche Forschungsgemeinschaft. This work has received support from Universität des Saarlandes. The Klinik für Innere Medizin III, Universitätsklinikum des Saarlandes, has received an unrestricted grant from Takeda (Aachen, Germany).

We thank Simone Jäger and Ellen Becker for their excellent technical assistance.

1.
Schoonjans K, Auwerx J: Thiazolidinediones: an update.
Lancet
355
:
1008
–1010,
2000
2.
Hetzel J, Balletshofer B, Rittig K, et al.: Rapid effects of rosiglitazone treatment on endothelial function and inflammatory biomarkers.
Arterioscler Thromb Vasc Biol
25
:
1804
–1809,
2005
3.
Pfutzner A, Marx N, Lubben G, et al.: Improvement of cardiovascular risk markers by pioglitazone is independent from glycemic control: results from the pioneer study.
J Am Coll Cardiol
45
:
1925
–1931,
2005
4.
Pistrosch F, Passauer J, Fischer S, et al.: In type 2 diabetes, rosiglitazone therapy for insulin resistance ameliorates endothelial dysfunction independent of glucose control.
Diabetes Care
27
:
484
–490,
2004
5.
Chen Z, Ishibashi S, Perrey S, et al.: Troglitazone inhibits atherosclerosis in apolipoprotein E-knockout mice: pleiotropic effects on CD36 expression and HDL.
Arterioscler Thromb Vasc Biol
21
:
372
–377,
2001
6.
Li AC, Brown KK, Silvestre MJ, et al.: Peroxisome proliferator-activated receptor gamma ligands inhibit development of atherosclerosis in LDL receptor-deficient mice.
J Clin Invest
106
:
523
–531,
2000
7.
Mazzone T, Meyer PM, Feinstein SB, et al.: Effect of pioglitazone compared with glimepiride on carotid intima-media thickness in type 2 diabetes: a randomized trial.
JAMA
296
:
2572
–2581,
2006
8.
Marx N: Peroxisome proliferator-activated receptor gamma and atherosclerosis.
Curr Hypertens Rep
4
:
71
–77,
2002
9.
Marx N, Duez H, Fruchart JC, et al.: Peroxisome proliferator-activated receptors and atherogenesis: regulators of gene expression in vascular cells.
Circ Res
94
:
1168
–1178,
2004
10.
Plutzky J: Medicine. PPARs as therapeutic targets: reverse cardiology?
Science
302
:
406
–407,
2003
11.
Asahara T, Murohara T, Sullivan A, et al.: Isolation of putative progenitor endothelial cells for angiogenesis.
Science
275
:
964
–967,
1997
12.
Rafii S, Lyden D: Therapeutic stem and progenitor cell transplantation for organ vascularization and regeneration.
Nat Med
9
:
702
–712,
2003
13.
Dimmeler S, Aicher A, Vasa M, et al.: HMG-CoA reductase inhibitors (statins) increase endothelial progenitor cells via the PI 3-kinase/Akt pathway.
J Clin Invest
108
:
391
–397,
2001
14.
Hill JM, Zalos G, Halcox JP, et al.: Circulating endothelial progenitor cells, vascular function, and cardiovascular risk.
N Engl J Med
348
:
593
–600,
2003
15.
Walter DH, Rittig K, Bahlmann FH, et al.: Statin therapy accelerates reendothelialization: a novel effect involving mobilization and incorporation of bone marrow-derived endothelial progenitor cells.
Circulation
105
:
3017
–3024,
2002
16.
Aicher A, Heeschen C, Mildner-Rihm C, et al.: Essential role of endothelial nitric oxide synthase for mobilization of stem and progenitor cells.
Nat Med
9
:
1370
–1376,
2003
17.
Strehlow K, Werner N, Berweiler J, et al.: Estrogen increases bone marrow-derived endothelial progenitor cell production and diminishes neointima formation.
Circulation
107
:
3059
–3065,
2003
18.
Werner N, Junk S, Laufs U, et al.: Intravenous transfusion of endothelial progenitor cells reduces neointima formation after vascular injury.
Circ Res
93
:
e17
–e24,
2003
19.
Schachinger V, Assmus B, Honold J, et al.: Normalization of coronary blood flow in the infarct-related artery after intracoronary progenitor cell therapy: intracoronary Doppler substudy of the TOPCARE-AMI trial.
Clin Res Cardiol
95
:
13
–22,
2006
20.
Werner N, Kosiol S, Schiegl T, et al.: Circulating endothelial progenitor cells and cardiovascular outcomes.
N Engl J Med
353
:
999
–1007,
2005
21.
Schmidt-Lucke C, Rossig L, Fichtlscherer S, et al.: Reduced number of circulating endothelial progenitor cells predicts future cardiovascular events: proof of concept for the clinical importance of endogenous vascular repair.
Circulation
111
:
2981
–2987,
2005
22.
Tepper OM, Galiano RD, Capla JM, et al.: Human endothelial progenitor cells from type II diabetics exhibit impaired proliferation, adhesion, and incorporation into vascular structures.
Circulation
106
:
2781
–2786,
2002
23.
Shintani S, Murohara T, Ikeda H, et al.: Mobilization of endothelial progenitor cells in patients with acute myocardial infarction.
Circulation
103
:
2776
–2779,
2001
24.
Werner N, Priller J, Laufs U, et al.: Bone marrow-derived progenitor cells modulate vascular reendothelialization and neointimal formation: effect of 3-hydroxy-3-methylglutaryl coenzyme a reductase inhibition.
Arterioscler Thromb Vasc Biol
22
:
1567
–1572,
2002
25.
Vasa M, Fichtlscherer S, Aicher A, et al.: Number and migratory activity of circulating endothelial progenitor cells inversely correlate with risk factors for coronary artery disease.
Circ Res
89
:
E1
–E7,
2001
26.
Fadini GP, Sartore S, Albiero M, et al.: Number and function of endothelial progenitor cells as a marker of severity for diabetic vasculopathy.
Arterioscler Thromb Vasc Biol
26
:
2140
–2146,
2006
27.
Ii M, Takenaka H, Asai J, et al.: Endothelial progenitor thrombospondin-1 mediates diabetes-induced delay in reendothelialization following arterial injury.
Circ Res
98
:
697
–704,
2006
28.
Adams V, Lenk K, Linke A, et al.: Increase of circulating endothelial progenitor cells in patients with coronary artery disease after exercise-induced ischemia.
Arterioscler Thromb Vasc Biol
24
:
684
–690,
2004
29.
Laufs U, Werner N, Link A, et al.: Physical training increases endothelial progenitor cells, inhibits neointima formation, and enhances angiogenesis.
Circulation
109
:
220
–226,
2004
30.
Kusuyama T, Omura T, Nishiya D, et al.: Effects of treatment for diabetes mellitus on circulating vascular progenitor cells.
J Pharmacol Sci
102
:
96
–102,
2006
31.
Wang CH, Ting MK, Verma S, et al.: Pioglitazone increases the numbers and improves the functional capacity of endothelial progenitor cells in patients with diabetes mellitus.
Am Heart J
152
:
1051
–1058,
2006
32.
Wang CH, Ciliberti N, Li SH, et al.: Rosiglitazone facilitates angiogenic progenitor cell differentiation toward endothelial lineage: a new paradigm in glitazone pleiotropy.
Circulation
109
:
1392
–1400,
2004
33.
Pistrosch F, Herbrig K, Oelschlaegel U, et al.: PPARgamma-agonist rosiglitazone increases number and migratory activity of cultured endothelial progenitor cells.
Atherosclerosis
183
:
163
–167,
2005
34.
Gensch C, Clever YP, Werner C, et al.: The PPA: R-gamma agonist pioglitazone increases neoangiogenesis and prevents apoptosis of endothelial progenitor cells.
Atherosclerosis
192
:
67
–74,
2007
35.
Laufs U, Urhausen A, Werner N, et al.: Running exercise of different duration and intensity: effect on endothelial progenitor cells in healthy subjects.
Eur J Cardiovasc Prev Rehabil
12
:
407
–414,
2005
36.
Maack C, Kartes T, Kilter H, et al.: Oxygen free radical release in human failing myocardium is associated with increased activity of Rac1-GTPase and represents a target for statin treatment.
Circulation
108
:
1567
–1574,
2003
37.
Semple RK, Chatterjee VK, O'Rahilly S: PPAR gamma and human metabolic disease.
J Clin Invest
116
:
581
–589,
2006
38.
Ray R, Shah AM: NADPH oxidase and endothelial cell function.
Clin Sci
(Lond) 
109
:
217
–226,
2005
39.
Imanishi T, Hano T, Nishio I: Angiotensin II accelerates endothelial progenitor cell senescence through induction of oxidative stress.
J Hypertens
23
:
97
–104,
2005
40.
Thum T, Fraccarollo D, Thum S, et al.: Differential effects of organic nitrates on endothelial progenitor cells are determined by oxidative stress.
Arterioscler Thromb Vasc Biol
27
:
748
–754,
2007
41.
Marx N: PPARgamma and vascular inflammation: adding another piece to the puzzle.
Circ Res
91
:
373
–374,
2002
42.
Fujii H, Li SH, Szmitko PE, et al.: C-reactive protein alters antioxidant defenses and promotes apoptosis in endothelial progenitor cells.
Arterioscler Thromb Vasc Biol
26
:
2476
–2482,
2006
43.
Verma S, Kuliszewski MA, Li SH, et al.: C-reactive protein attenuates endothelial progenitor cell survival, differentiation, and function: further evidence of a mechanistic link between C-reactive protein and cardiovascular disease.
Circulation
109
:
2058
–2067,
2004
44.
Urbich C, Dimmeler S: Endothelial progenitor cells functional characterization.
Trends Cardiovasc Med
14
:
318
–322,
2004
45.
Turnbull F, Neal B, Algert C, et al.: Effects of different blood pressure-lowering regimens on major cardiovascular events in individuals with and without diabetes mellitus: results of prospectively designed overviews of randomized trials.
Arch Intern Med
165
:
1410
–1419,
2005
46.
Costa J, Borges M, David C, et al.: Efficacy of lipid lowering drug treatment for diabetic and non-diabetic patients: meta-analysis of randomised controlled trials.
BMJ
332
:
1115
–1124,
2006
47.
Richter B, Bandeira-Echtler E, Bergerhoff K, et al.: Pioglitazone for type 2 diabetes mellitus.
Cochrane Database Syst Rev
CD006060,
2006
48.
Marx N, Wohrle J, Nusser T, et al.: Pioglitazone reduces neointima volume after coronary stent implantation: a randomized, placebo-controlled, double-blind trial in nondiabetic patients.
Circulation
112
:
2792
–2798,
2005
49.
Horio T, Suzuki M, Takamisawa I, et al.: Pioglitazone-induced insulin sensitization improves vascular endothelial function in nondiabetic patients with essential hypertension.
Am J Hypertens
18
:
1626
–1630,
2005