International Journal of Cardiology xxx (2012) xxx–xxx Contents lists available at SciVerse ScienceDirect International Journal of Cardiology journal homepage: www.elsevier.com/locate/ijcard Reduced number and impaired function of circulating endothelial progenitor cells in patients with abdominal aortic aneurysm Shih-Hsien Sung Chun-Che Shih b,c,d,1 a,d,e , Tao-Cheng Wu a,c,d d,f g a,c,d , Jia-Shiong Chen , Yung-Hsiang Chen , Po-Hsun Huang , Shing-Jong Lin a,c,d , a,d,f, , Jaw-Wen Chen ⁎ a Division of Cardiology, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei, Taiwan Division of b Cardiovascular Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan Institute of Clinical Medicine, National Yang-Ming University School of Medicine, Taipei, Taiwan d Cardiovascular Research Center, National Yang-Ming University School of Medicine, Taipei, Taiwan e Institute of Pharmacology, National Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Institute of Public Health, National Yang-Ming University School of Medicine, Taipei, Taiwan Yang-Ming University School of Medicine, Taipei, Taiwan g f Medical University, Taichung, Taiwan article info Article history: Received 16 July 2011 Received in revised form 29 May 2012 Accepted 1 November 2012 Available online xxxx Keywords: Abdominal aortic aneurysm Endothelial function Endothelial progenitor cells abstract different, it shares some risk factors with CAD. Therefore, the correlation between EPCs and AAA was investigated. Methods and results: Seventy-eight subjects (age 77.2±7.8 years) with suspected AAA were prospectively enrolled. Cut-off values (men, 3.5–5.5 cm; women, 3–5 cm) were used to define normal aorta, small AAA, and large AAA on thoraco-abdominal computer tomography. Endothelial function was measured by flow-mediated vasodilation (FMD). Flow cytometry and colony-forming units (CFUs) were used to evaluate circulating EPC numbers. Circulating EPCs were defined as mononuclear cells with low CD45 staining and double-positive staining for KDR, CD34, or CD133. Late out-growth EPCs were cultured from six patients with large AAAs and six age-and sex-matched controls to evaluate proliferation, adhesion, migration, tube formation, and senescence. FMD was significantly lower with large (5.26%±3.11%) and small AAAs (6.31%±3.66%) than in controls (8.88%±4.83%, P=0.008). Both CFUs (normal 38.39±12.99, small AAA 21.22±7.14, large AAA 6.98±1.97; P=0.026) and circulating EPCs (CD34 /KDR and + Aim: Circulating endothelial progenitor cells (EPCs) are associated with coronary artery disease (CAD) and predict its outcome. Although the pathophysiology of abdominal aortic aneurysm (AAA) is + CD133 /KDR )weresignificantly fewer in AAA patients than in controls. On multivariate analysis, CFUs and circulating + + EPCs (CD34 /KDR ) were independently, inversely correlated to AAA diameter. Proliferation, adhesion, migration, tube formation, and senescence of late EPCs were significantly impaired in AAA patients. Conclusion: The number and function of EPCs were impaired in AAA patients, suggesting their potential role in AAA. © 2012 Elsevier Ireland Ltd. All rights reserved. + + 1. Introduction Abdominal aortic aneurysm (AAA) accounts for more than 15,000 annual deaths in the US and is one of the leading causes of mortality in men aged more than 50 year-old [1]. Its prevalence increases with increasing age, and the risk of rupture increases with increasing aneurysm diameter. Once the aneurysm ruptures, only 18% of the patients survive. The presence of AAA shares similar risks, such as smoking, hypertension, and hypercholesterolemia, with atherosclerotic cardiovascular ⁎ Corresponding author at: Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, No. 201, Sec. 2, Shih-Pai Road, Taipei 112, Taiwan. Tel.: +886 2 2875 7511; fax: +886 2 2871 1601. E-mail address: jwchen@vghtpe.gov.tw (J.-W. Chen). Co-corresponder. 0167-5273/$ – see front matter © 2012 Elsevier Ireland Ltd. All rights reserved. http://dx.doi.org/10.1016/j.ijcard.2012.11.002 disease [2,3]. However, the managements of these risk factors failed to prevent the progression of AAA. AAA development and progression appear to be related to an imbalance between destructive and restorative vascular wall processes. Aneurysmal dilatation characteristically shows destruction of elastin and collagen in the media and adventitia, loss of medial smooth muscle cells, vessel wall thinning, and transmural lymphocyte and macrophage infiltration [4,5]. In addition, the loss of endothelial integrity was also noted in AAA from histological examination, which was also the fundamental cause of mural thrombosis [6].Moreover, in a small series study of 30 patients with AAA, the endothelial function reflected by endothelium dependent vasodilation has a negative and linear correlation with the aneurysm diameter [7]. In an experimental AAA model, creation of high aortic flow status by creating AV fistula promoted re-endothelization of AAA and reduced its progression, when comparing with normal or low aortic flow status [8].It Please cite this article as: Sung S-H, et al, Reduced number and impaired function of circulating endothelial progenitor cells in patients with abdominal aortic aneurysm, Int J Cardiol (2012), http://dx.doi.org/10.1016/j.ijcard.2012.11.002 seems that the endothelial injury was associated with the occurrence of AAA and the recovery of endothelial integrity correlated with the progression of AAA. Since the circulating endothelial progenitor cells (EPCs) are a cellular reservoir to repair or replace dysfunctional endothelium at vascular injury sites and restore endothelial function [9–11]. The number of circulating EPCs may predict adverse events and mortality in patients with different cardiovascular diseases [12,13]. It is believed that the circulating EPCs might be associated with the presence and progression of AAA. However, the only published data demonstrated an unexpected result. The circulating EPCs defined by positive surface markers of CD133 and/or CD34 increased in 25 subjects with AAA, comparing with 18 age-matched controls [14].Therefore, we would like to investigate the associations of circulating EPCs as well as endothelial function with the progression of AAA in the present study. 2.6. EPC culture Peripheral blood samples (20 mL) were obtained in heparin-coated tubes from six patients with large AAAs and six age-and sex-matched patients with normal aortas to study late EPCs in culture. MNCs were isolated by density gradient centrifugation with Histopaque-1077 (Sigma), and the serum was preserved [17]. Briefly, 6 MNCs (5×10 ) were plated in 2-mL of endothelial growth medium (EGM-2 MV Cambrex, East Rutherford, NJ, USA), with 15% individual serum on fibronectin-coated, 6-well plates. After 4 days of culturing, the medium was changed, and nonadherent cells were removed; attached early EPCs appeared elongated with spindle shapes. Some MNCs were allowed to grow into colonies of late (out-growth) EPCs, which emerged 2–3 weeks after the start of MNC culture. The late EPCs exhibited a ‘cobblestone’ morphology and monolayer growth pattern typical of mature endothelial cells at confluence. Late EPCs were collected for the functional assays. 2.7. EPC characterization Early EPCs were characterized as adherent cells, double-positive for acetylated LDL uptake and lectin binding by direct fluorescent staining, as previously described [17]. Briefly, the adherent cells were first 2. Methods incubated 2.1. Study population Subjects referred for a thoraco-abdominal computer tomogram (CT) with suspected AAA were eligible. Patients with unstable angina, decompensated heart failure, inflammatory disease, ongoing infections, severe renal failure, and who were allergic to contrast medium were excluded. Written, informed consent, as approved by our Institutional Review Board, was obtained from each patient before enrolment. After overnight fasting, patients were studied in a quiet, temperature-controlled room. Blood pressures (BPs) were obtained by averaging three different measurements taken after a 15-minute rest. Serum and plasma samples were acquired for plasma creatinine (PCr), fasting glucose, lipid profiles, and high-sensitivity C-reactive protein (hsCRP). Height, weight, history, demographics, and prescribed medications were recorded. Estimated glomerular filtration rate (eGFR) was calculated from plasma creatinine (PCr) using a modified Modification of Diet in Renal Disease equation for the Chinese population: eGFR (mL/min/1.73 m 2 )= ×age ×(0.79 if − 1.234 − 0.179 175×PCr female) [15]. Serum hsCRP was determined by particle-enhanced immunoturbidimetry using latex agglutination (Toshiba, Tokyo, Japan). with 2.4 mg/mL 1,1 ′ -dioctadecyl-3,3, -tetramethylindocarbocyanine 2.8.1. EPC adhesion assay 4 EPCs' ability to adhere to the injured site to initiate the repair process was evaluated by plating 1×10 late EPCs onto a fibronectin-coated, 6-well plate and incubating for 30 minutes. Gentle washing with PBS three times was performed after a 30 min adhesion, and adherent cells in six random, high-power (×100) microscopic fields (HPF) of each well were counted by independent, blinded investigators. 2.8.2. EPC proliferation assay The proliferation of EPCs was determined by 3-(4,5-dimethylthiazol-2-yl)-2,5, diphenyltetrazolium bromide 4 The maximal abdominal aorta diameter was measured by reviewing each coronal CT section. Cut-off values (men, 3.5–5.5 cm; women, 3–5 cm) were used to define normal aorta, small AAA, and large AAA. ,3 ′ 2.8. EPC functional assays (MTT) assay [17]. After being cultured with 1×10 2.2. AAA definitions 3 ′ perchlorate-acetylated LDL (DiI-acLDL; Molecular Probes, Eugene, OR, USA) for 1 h, then fixed in 2% paraformaldehyde and counterstained with 10 mg/mL FITC-labeled lectin from Ulex europaeus (UEA-1; Sigma). The late EPC-derived outgrowth endothelial cell population was characterized by immunofluorescence staining for expressions of vascular endothelial (VE)-cadherin, platelet/endothelial cell adhesion molecule-1 (PECAM-1, CD-31), and CD34 (Santa Cruz). Fluorescent images were recorded under a laser scanning confocal microscope. late EPCs in fibronectin-coated 24-well plates for 3 days, the EPCs were supplemented with MTT (0.5 mg/mL; Sigma) and incubated for 4 h for the proliferation assay. The blue formazen was dissolved with dimethyl sulfoxide and measured at 550/650 nm. In addition, simultaneous culturing with human aortic endothelial cells (HAECs) served as the internal control [19]. The EPC proliferation activity was presented as the ratio to HAECs. 2.3. Determination of endothelial function 2.8.3. EPC migration assay EPC migration was evaluated by a modified Boyden chamber assay (Transwell, Coster; Corning Inc., Acton, Endothelial function was measured by endothelium-dependent flow-mediated vasodilatation (FMD) using a 7.5-MHz linear array transducer (Sonos 5500; Hewlett-Packard, Andover, MA, USA) to scan the brachial artery [16]. The procedure was performed in a quiet, air-conditioned room (22–25 °C) by an experienced technician blinded to the clinical data. The left arm was stabilized with a cushion, and a sphygmomanometric cuff was placed on the forearm. A baseline image was acquired, and blood flow was estimated by time-averaging the pulsed Doppler velocity signals from a mid-artery sample volume. Then, the cuff was MA, USA) [17]. Isolated EPCs were detached as described above with trypsin/EDTA, and 3×10 inflated to ≥ 50 mm Hg above systolic pressure to occlude the arteries for 5 min and released abruptly. A mid-artery pulsed Doppler signal was obtained immediately upon cuff release, and brachial artery diameters were obtained at 30, 60, 90, 120, and 150 s after deflation. FMD was calculated as maximal post-occlusion brachial artery diameter relative to averaged baseline diameters. 2.4. Circulating EPC assay + + for + CD34 + low + + low and low 2.5. EPC colony-forming assay Isolated mononuclear cells (MNCs) were resuspended in growth medium (EndoCult; StemCell Technologies, 6 Vancouver, Canada), and 5×10 MNCs were preplated onto a fibronectin-coated six-well plate in duplicate [18]. After 48 h, nonadherent cells were collected by pipetting the medium in each well up and down three 6 times, and 1×10 2.8.4. EPC tube-formation assay In vitro tube formation was assayed using the In Vitro Angiogenesis Assay Kit (Chemicon) [17]. ECMatrix gel solution was thawed at 4 °C overnight, mixed with ECMatrix diluent buffer, and placed in a 96-well plate at 37 °C for 1 h to allow the matrix solution to solidify. EPCs were harvested as described above with 4 KDR CD45 , CD34 CD133 CD45 , KDR CD133 CD45 , respectively. (Fig. 1). defined cells were replated onto a fibronectin-coated 24-well plate. On day 5 of the assay, the colony-forming units (CFUs) per well for each sample were counted manually in a minimum of three wells by two independent observers. EPCs were placed in the upper chambers of 24-well transwell plates with polycarbonate membranes (8-mm pores) that contained serum-free EGM; vascular endothelial growth factor (VEGF, 50 ng/mL) was added to the medium in the lower chambers. After incubation for 24 h, the membrane was washed briefly with PBS and fixed with 4% paraformaldehyde. The upper side of the membrane was wiped gently with a cotton ball. The membrane was stained using hematoxylin solution and carefully removed. The number of migratory late EPCs was evaluated by counting the migrated cells in six random HPFs. trypsin/ EDTA, then 1×10 The method for assessing the number of circulating EPCs has been previously described [17,18]. Briefly, a 10-mL volume of peripheral blood was incubated for 30 min in the dark with monoclonal antibodies against human kinase insert domain-conjugating receptor (KDR; R&D, Minneapolis, MN, USA) followed by phycoerythrin (PE)-conjugated secondary antibody, with fluorescein isothiocyanate (FITC)-labeled monoclonal antibodies against human CD45 (Becton Dickinson, Franklin Lakes, NJ, USA), with PE-conjugated monoclonal antibody against human CD133 (Miltenyi Biotec, Bergisch Gladbach, Germany), and FITC-conjugated or PE-conjugated monoclonal antibodies against human CD34 (Serotec, Raleigh, NC, USA) and KDR (Sigma, St Louis, MI, USA). Isotype-identical antibodies served as controls (Becton Dickinson). After incubation, cells were lysed, washed with phosphate-buffered saline (PBS), and fixed in 2% paraformaldehyde before analysis. Each analysis included 100,000 events. The numbers of circulating EPCs were 4 EPCs were placed on matrix solution with EGM-2 MV medium with glucose or mannitol, and incubated at 37 °C for 16 h. Tubule formation was inspected under an inverted light microscope (×100). Four representative fields were taken, and the average total area of complete tubes formed by cells was compared using Image-Pro Plus software (Media Cybernetics, Inc., MD, USA). Please cite this article as: Sung S-H, et al, Reduced number and impaired function of circulating endothelial progenitor cells in patients with abdominal aortic aneurysm, Int J Cardiol (2012), http://dx.doi.org/10.1016/j.ijcard.2012.11.002 Fig. 1. Representative flow-cytometry analysis for quantifying the number of circulating endothelial and KDR CD133 CD45 , respectively. + + low progenitor cells (EPCs). Mononuclear cells (MNCs) were gated by forward/ sideward scatter (FSC/SSC). The + numbers of circulating EPCs were defined for CD34 CD133 CD45 , CD34 KDR CD45 , + low + + low 2.8.5. EPC senescence assay Cellular aging was quantified using a Senescence Cell Staining Kit (Sigma) [17]. EPCs were plated in 12-well plates and cultured for 4 days. After washing with PBS, EPCs were fixed for 6 min in 2% formaldehyde and 0.2% glutaraldehyde in PBS, and then incubated for 12 h at 37 °C without CO2 with fresh X-gal staining solution. Green-stained cells and total cells were counted in six random HPFs, and the percentage of β -galactosidase-positive cells was calculated. 2.9. Statistical analysis A total of seventy-eight patients (72 men; mean age, 77.2± 7.8 years) were enrolled; 15 had normal aortas, 27 had small AAAs, and 36 had large AAAs. Baseline characteristics are shown in Table 1. In brief, while systolic and diastolic BPs were similar in the three groups, large AAA patients had the lowest pulse pressures. Compared to controls, AAA patients more frequently received beta-blockers, and large AAA patients had lower eGFR. Other characteristics did not differ among the groups. Means, standard deviations, and percentages were used to describe the participants' characteristics. Comparisons of continuous variables among three groups were performed by analysis of variance using oneway ANOVA and Bonferroni's post-hoc test. Subgroup comparisons of categorical variables were assessed by the chi-square or Fisher's exact test. Correlations of various factors with maximum AAA diameter were determined with bivariate Pearson correlation. Independent risk factors associated with maximum AAA diameters were identified using a multivariate linear regression model. The late Table 1 Subjects' baseline characteristics. EPCs' functions were compared between AAA and normal aorta subjects, using Student's t-test. Pb0.05 was considered significant; all analyses were carried out with SPSS 15.0 (SPSS Inc. Chicago, IL, USA). 3. Results Normal aorta N=15 Small AAA N=27 Large AAA N=36 P value Age, years Male sex, N (%) BMI, kg/m2 SBP, mm Hg DBP, mm Hg 78.13±7.02 15 (100) 22.88±3.92 140.7±16.8 77.7±8.6 77.22±6.40 25 (92.6) 27.91±22.09 136.3±21.4 75.3±10.2 76.83±9.16 32 (88.9) 23.90±3.38 129.6±15.8 76.2±11.2 0.867 0.397 0.427 0.152 0.795 Pulse pressure, mm Hg 63.0±10.7 61.0±18.0 53.4±10.38⁎,† 26 (72.2) 4 (11.1) 19 (52.8) 12 (33.3) 0.051 8 (53.3) 3 (20) 5 (33.3) 3 (20) Hypertension, N (%) Diabetes, N (%) CAD, N (%) Smoker, N (%) 2.99±0.35 20 (74.1) 2 (7.4) 10 (37.0) 8 (29.6) 4.70±0.60⁎ WBC,/cm3 Hemoglobin, mg/dL Creatinine, mg/dL eGFR, mL/min/1.73 m2 Lipid profiles, mg/dL 6307±1634 13.27±1.35 1.03±0.24 82.85±23.14 6470±1511 13.16±1.97 1.27±0.41 66.58±23.60 6.76±1.02⁎,† 6578±2620 12.32±2.12 1.59±1.52 63.64±27.84⁎ b0.001 0.916 0.142 0.196 0.052 Total cholesterol Triglyceride HDL LDL Fasting glucose, mg/dL HsCRP, mg/dL LVEF, % Medications, N (%) 179.00±37.80 96.69±38.15 59.00±29.40 95.90±40.29 106.58±21.99 0.44±0.44 54.8±8.9 171.72±30.77 108.64±55.97 52.36±24.05 104.36±29.65 101.92±24.93 1.03±2.33 53.4±8.2 172.25±34.63 108.92±62.67 45.97±14.63 109.50±30.33 94.97±17.72 0.92±1.65 51.9±8.6 0.797 0.785 0.164 0.470 0.197 0.528 0.678 Anti-platelet Beta-blocker 12 (80) 0 (0) 23 (85.2) 10 (37) 27 (75) 10 (27.8) ACEI/ARB Statin Normal aorta 12 (44.4) 6 (22.2) 14 (38.9) 5 (13.9) Small AAA 9 (60) 1 (6.7) Large AAA N=15 N=27 Diameter, cm 0.328 0.470 0.309 0.636 0.611 0.029 0.385 0.385 P value N=36 FMD, % 8.88±4.83 6.31±3.66⁎ 5.26±3.11⁎ 0.008 Data are displayed as means±standard deviation or number (%). AAA: abdominal aortic aneurysm; ACEI: angiotensin-converting enzyme inhibitor; ARB: angiotensin II receptor blocker; BMI: body mass index; CAD: coronary Progenitor cells,% artery disease; DBP: diastolic blood pressure; eGFR: estimated glomerular filtration rate; HDL: high-density lipoprotein; HsCRP: highsensitivity C-reactive protein; LDL: low-density lipoprotein; LVEF: left ventricular ejection fraction; pressure; WBC: white blood cell. CD34 SBP: systolic blood2.1±0.9 2.3±1.5 2.2±0.9 0.890 ⁎CD34/CD133 Pb 0.05 compared to controls in post hoc analysis. 0.510±0.878 0.272±0.345 †CD34/KDR 0.246±0.256⁎ 0.500±0.590 0.064±0.099⁎ CD133/KDR 0.208±0.342 Pb 0.05 compared to small AAA group in post hoc analysis. 24.78±12.12⁎ EPC CFU/well 34.67±13.36 0.250±0.213 0.174±0.255⁎ 0.011 0.164 0.045±0.074⁎ 0.007 12.58±8.01⁎,† b0.00 Please cite this article as: Sung S-H, et al, Reduced number and impaired 1function of circulating endothelial progenitor cells in patients with abdominal aortic aneurysm, Systolic blood pressure, mm Hg − 0.256 0.035 Pulse pressure, mm Hg − 0.297 0.014 Int J Cardiol (2012), http://dx.doi.org/10.1016/j.ijcard.2012.11.002 3.1. Endothelial function and EPCs Normal aorta N=15 Controls had better endothelial function, reflected by FMD, than AAA patients (Table 2). The level of circulating CD34+ cells was similar among the 3 groups. However, AAA patients had significantly fewer circulating EPCs, reflected by double-positive stainings of CD34/KDR and CD133/KDR, but not CD34/CD133. In addition, AAA patients had fewer EPC CFUs. 3.2. Factors associated with maximal AAA diameter Systolic BP, pulse pressure, hemoglobin, and eGFR were negatively associated, and serum creatinine positively associated, with maximal aortic diameter (Table 3). FMD was negatively correlated to aneurysm diameter (Fig. 2A). Both the numbers of circulating CD34 /KDR EPCs and EPC CFUs were negatively associated with AAA diameter (Fig. 2B, C). After adjusting for systolic BP, pulse pressure, hemoglobin, and eGFR in a multivariate linear regression + + model, the numbers of CD34 /KDR EPCs and EPC CFUs remained independently associated with aortic diameter (Table 3). Standardized coefficients + + were − 0.231 (P=0.048) and − 0.443 (Pb0.001), respectively. 3.3. Characterization of late EPCs Small AAA N=27 Age, years Male sex, N (%) BMI, kg/m2 SBP, mm Hg DBP, mm Hg 78.13±7.02 15 (100) 22.88±3.92 140.7±16.8 77.7±8.6 77.22±6.40 25 (92.6) 27.91±22.09 136.3±21.4 75.3±10.2 Pulse pressure, mm Hg 63.0±10.7 61.0±18.0 Hypertension, N (%) 8 (53.3) Diabetes, N (%) 3 (20) CAD, N (%) 5 (33.3) Smoker, N (%) 3 (20) Table 3 Diameter, cm 2.99±0.35 Correlation coefficients of the maximum abdominal aortic aneurysm diameters and other variables. WBC,/cm3 6307±1634 Hemoglobin, mg/dL 13.27±1.35 Coefficients P value Creatinine, mg/dL 1.03±0.24 eGFR, mL/min/1.73 m2 82.85±23.14 Lipid profiles, mg/dL 20 (74.1) 2 (7.4) 10 (37.0) 8 (29.6) 4.70±0.60⁎ 6470±1511 13.16±1.97 1.27±0.41 66.58±23.60 Total 179.00±37.80 CFU:cholesterol colony-forming unit; eGFR: estimated glomerular filtration rate; EPC: endothelial progenitor cell; FMD: 171.72±30.77 Triglyceride 96.69±38.15 108.64±55.97 flow-mediated vasodilation. HDL 59.00±29.40 52.36±24.05 LDL 95.90±40.29 104.36±29.65 Fasting glucose, mg/dL 106.58±21.99 101.92±24.93 HsCRP, mg/dL 0.44±0.44 1.03±2.33 3.4.4. Tube formation LVEF, % 54.8±8.9 53.4±8.2 Late EPC tube formation on ECMatrix gel was lower in AAA patients (22.15±7.63 Medications, N (%) tubes/well) than in controls (41.49±17.74 tubes/well; P =0.042) (Fig. 4D). The peripheral blood MNCs seeded on fibronectin-coated wells were initially round (Fig. 3A) [17]. After changing the medium on day 4, attached early EPCs appeared elongated with spindle shapes (Fig. 3B). Late EPCs with cobblestone-like morphology similar to mature endothelial cells were grown to confluence (Fig. 3C). Late EPC characterization was performed by immunohistochemical staining; most cells expressed VE-cadherin, PECAM-1, and CD34; critical markers of outgrowth endothelial cell-producing late EPCs (Fig. 3D, E, F). Anti-platelet Beta-blocker 3.4.5. Senescence ACEI/ARB Statin AAA patients tended Normal aorta 12 (80) 0 (0) 23 (85.2) 10 (37) 9 (60) 12 (44.4) 6 (22.2) 1 (6.7) of senescence-associated, β to have higher percentages Small AAA Large AAA P -galactosidase-positive EPCs (63.32%±13.72%) than controls (48.77%±8.52%; P value N=15 N=27 N=36 =0.052) (Fig. 4E). FMD, % 3.4.6. Senescence 8.88±4.83 6.31±3.66⁎ 5.26±3.11⁎ 0.008 Progenitor cells,% AAA patients also have higher percentages of senescence-associated, β 2.3±1.5 2.2±0.9 0.890 (65.6%±10.2%) than controls (48.8%±8.5%; P=0.011) 0.272±0.345 0.250±0.213 0.164 (Fig. 4E). 0.246±0.256⁎ 0.174±0.255⁎ CD34/KDR 0.500±0.590 0.011 CD34 2.1±0.9 -galactosidase-positive EPCs CD34/CD133 0.510±0.878 3.4. In vitro AAA EPC functions 3.4.1. Adhesion After a 30-min incubation, patients with normal aorta had more adherent cells than large AAA patients (49.8±13.9 cells/HPF and 31.1±9.4 cells/HPF, respectively; P =0.021) (Fig. 4A). CD133/KDR 0.208±0.342 4. Discussion EPC CFU/well 34.67±13.36 0.064±0.099⁎ 0.045±0.074⁎ 0.007 24.78±12.12⁎ 12.58±8.01⁎,† b0.00 1 0.035 comprehensively− 0.256 the endothelial function, circulating Pulse mm Hg − 0.297 0.014 EPCpressure, number by 2 distinct methods, and biological functions of late outgrowth Hemoglobin, − 0.245 0.032 function, EPCs. Themg/dL data showed that patients with AAA had impaired endothelial 3.4.2. Proliferation Creatinine, mg/dL 0.001 measured by flow-mediated vasodilation and 0.362 reduced number of circulating EPCs, After a 3 day culture, significantly attenuated EPC proliferation activity was eGFR, mL/min/1.73 m2 − 0.303 measured by both flow cytometry and colony-forming units. In 0.007 addition, the noted in patients with AAA group than in patients with normal aorta (Fig. 4B). FMD, % functions of late EPCs cultured from − 0.319 0.005impaired, in-vitro AAA patients were also CD34 +/CD133+ EPCs, % 0.088 − 0.196 comparing with those from subjects with normal aorta. Furthermore, the numbers 3.4.3. Migration CD34 +/KDR+ EPCs, % − 0.230 of circulating EPCs and EPC CFUs were independently associated 0.045 with maximal After a 24 h incubation, less EPCs cultured from AAA patients appeared to CD133 + EPCs, % 0.092 0.193pulse pressure, hemoglobin, AAA+/KDR diameters, after accounting for systolic− BP, and migrate to the lower chambers (37.1±18.6 cells/HPF) than those from controls EPC-CFU/well b0.001 eGFR. Although this is a cross-sectional− 0.542 study, it firstly demonstrated the (65.2±22.9 cells/HPF; P =0.042) (Fig. 4C). consistent and independent associations of circulating EPCs with clinical AAA, to the best of our knowledge. These findings may explain the potential impact of circulating EPCs in AAA development and progression, and support the future target of endothelial function for the prevention and potential adjunct treatment of Normal aorta SmallAAA. AAA Large AAA P value N=15 N=27 Age, years Male sex, N (%) BMI, kg/m2 SBP, mm Hg DBP, mm Hg 78.13±7.02 15 (100) 22.88±3.92 140.7±16.8 77.7±8.6 Pulse pressure, mm Hg 63.0±10.7 Hypertension, N (%) 8 (53.3) Diabetes, N (%) 3 (20) TableN2(%) CAD, 5 (33.3) Smoker, N (%) 3 (20) Endothelial function, endothelial progenitor cells, and systemic inflammation by group. Diameter, cm 2.99±0.35 WBC,/cm3 6307±1634 Hemoglobin, mg/dL 13.27±1.35 Creatinine, mg/dL aortic aneurysm; CFU: colony-forming unit; 1.03±0.24 AAA: abdominal EPC: endothelial progenitor cell; FMD: eGFR, mL/min/1.73 m2 82.85±23.14 flow-mediated vasodilation. Lipid profiles, mg/dL to controls in post hoc analysis. ⁎ Pb 0.05 compared † Systolic blood pressure, Hg The present studymmaccessed Total cholesterol 179.00±37.80 Triglyceride 96.69±38.15 Pb0.05 compared to small AAA group in post hoc analysis. HDL 59.00±29.40 LDL 95.90±40.29 N=36 4.1. Endothelial and vascular damage in AAA 76.83±9.16 77.22±6.40 25 (92.6) 27.91±22.09 Different, and 136.3±21.4 found in AAA 75.3±10.2 32 (88.9) risk23.90±3.38 factors 129.6±15.8 may76.2±11.2 include 0.867 0.397 0.427 to those of atherosclerosis have been 0.152 old age, male gender, current smoking, 0.795 even opposite, patients, which hypertension, and dyslipidemia,53.4±10.38⁎,† but not diabetes mellitus [20]. Moreover, while 61.0±18.0 0.051 smoking and aneurysm diameter 26are(72.2) risk factors for aneurysm expansion, low 20 (74.1) 0.328 2 (7.4) 4 (11.1) ankle-brachial index—amarkerof peripheral arterial disease—prevents0.470 progression, 10 (37.0) 19 (52.8) 0.309 suggesting that local factors such 12 as (33.3) regional hemodynamic and/or local vascular 8 (29.6) 0.636 injury rather than systemic pathology affect disease progressionb0.001 [21].Thus, a 4.70±0.60⁎ 6.76±1.02⁎,† complex interaction of different mechanisms, including vascular inflammation and 6470±1511 6578±2620 0.916 13.16±1.97 12.32±2.12 0.142 AAA is hemodynamic and genetic factors, may cause AAA [22]. Although 1.27±0.41 1.59±1.52 0.196 generally caused by a primary process involving the media, which is0.052 the cellular 66.58±23.60 63.64±27.84⁎ dysfunction of smooth muscle cells, 171.72±30.77 Please cite 108.64±55.97 function of 52.36±24.05 104.36±29.65 172.25±34.63 0.797 109.50±30.33 0.470 this article as: Sung S-H, et al, Reduced number and0.785 impaired 108.92±62.67 circulating endothelial progenitor cells in patients with 0.164 abdominal 45.97±14.63 aortic aneurysm, Int J http://dx.doi.org/10.1016/j.ijcard.2012.11.002 Cardiol (2012), Fig. 2. The association of abdominal aortic aneurysm diameter with endothelial function (flow-mediated vasodilation), the level of circulating endothelial progenitor cells (EPCs, CD34 /KDR ), + + and EPC colony-forming units (CFUs). The graphs show the distribution of maximum aneurysmal diameter and flow-mediated vasodilation (A), CD34 /KDR EPCs (B), and EPC CFUs (C) in 78 participants. + + fragmentation of elastic fibers, and collagen degradation through various proteolytic enzymes, particularly matrix metalloproteinase (MMP) [23], further hemodynamic effects of BP and mechanical wall stress are essential to the progressive enlargement. Therefore, the endothelial cells, which modulate the vasodilatation and the inflammation as well as the activity of proteolytic enzymes in the arterial walls [24,25], might be associated with the remodeling of arterial medial layer and the progression of aortic aneurysm. The aortic endothelium may also be injured not only due to traditional atherosclerosis risk factors, but also due to increased mechanical wall stress associated with the changing aortic wall structure [24,26]. While the integrity and functional activity of the endothelial monolayer are important in atherogenesis [27], a defective vascular endothelium leading to an uninhibited inflammatory responses appears to contribute to aneurysm formation [24].In addition, Knipp et al. demonstrated impaired vasoreactivity despite higher plasma nitrite in AAA patients than in peripheral arterial disease patients, suggesting desensitization of the systemic vasculature to normal vasodilatory stimuli [28]. In line with previous findings, the present study showed that forearm FMD, which reflects systemic vascular endothelial function, was inversely and linearly associated with AAA diameter, suggesting that vascular endothelial dysfunction and AAA development and/or progression are linked. 4.2. EPCs' potential role in AAA development and progression Circulating EPCs may play an important role in endothelial repair of vascular damage. Endothelial function and the level of circulating EPCs have been associated in various populations [9,29]. Although it has been showed that patients with AAA would have impaired endothelial function, the expected negative correlation of the present AAA and circulating CD34 /CD133 EPC number did not exist in the only published study [7,14]. However, CD34 and/or CD133 might not be adequate surface markers to identify + + EPCs [30]. In addition, Parietti et al. illustrated that the level of CD34 /KDR EPCs was inversely related to aneurysm diameter in 27 patients with thoracic aortic aneurysm [31]. But the pathologic features of thoracic aortic aneurysm might not be identical to AAA. In this study, in addition to FMD, the numbers of EPC CFUs and circulating EPCs were significantly inversely and linearly correlated to aortic diameter, and remained independent even after adjusting for BP, hemoglobin, and renal function. Since + + CD34 is a surface marker of stem cells and the circulating CD34 progenitor cells are similar between groups, it supports that the diminished EPC number in the patients with AAA is not due to bone marrow dysfunction and related poor production. Further, the present data also showed impaired function of late EPCs in AAA + Fig. 3. Morphology and characterization of human endothelial progenitor cells (EPCs) from peripheral blood. Mononuclear cells (MNCs) were plated on a fibronectin-coated culture dish on the first day (A). Four days after plating, adherent early EPCs with spindle shapes are seen (B). Three weeks after plating, late (outgrowth) EPCs with a cobblestone-like morphology were selected, reseeded, and grown to confluence (C). Immunofluorescence detection of 1,1′ -dioctadecyl-3,3,3′ ,3′ -tetramethylindocarbocyanine perchlorate-acetylated LDL (DiI-acLDL, red), VE-cadherin (green) (D), platelet/endothelial cell adhesion molecule-1 (CD31, green) (E), and CD34 (green) (F) for late EPCs. (For interpretation of the references to color in this figure legend, the reader is referred to the web version of this article.) Please cite this article as: Sung S-H, et al, Reduced number and impaired function of circulating endothelial progenitor cells in patients with abdominal aortic aneurysm, Int J Cardiol (2012), http://dx.doi.org/10.1016/j.ijcard.2012.11.002 Fig. 4. Functional assays (including in vitro migration, reactive oxygen species production, cellular senescence, and apoptosis) of late endothelial progenitor cells (EPCs) cultured from six patients with large abdominal aortic 4 aneurysms (AAAs) and from six age-and sex-matched controls. After incubation of 1×10 late EPCs for 30 min, the EPC adhesion in AAA patients is significantly fewer than in the normal aorta (A). After incubation of 1×10 late 4 EPCs for 3 days, the EPC proliferation activity is significantly less in AAA patients than in the normal aorta (B). A modified Boyden chamber assay with vascular endothelial growth factor (VEGF) as a chemoattractant factor was used to assess EPC migration. Representative photos are shown; the small dots are holes in the barrier membrane. The migrated cells are stained with hematoxylin and counted under the microscope. The migration function of EPCs from AAA patients is significantly less than in the normal aorta (C). An in vitro angiogenesis assay for late EPCs was used with ECMatrix gel. The average total numbers of complete tubes formed by cells were compared by computer software. The in vitro tube formation is significantly impaired in late EPCs cultured from AAA patients compared to normal aorta (D). Compared to patients with normal aorta, incubated EPCs from AAA patients have a significantly increased percentage of senescence-associated β -galactosidase-positive EPCs (E). (Data expressed as means±standard error). patients, suggesting that vascular and endothelial repair fails not only high flow conditions may stabilize aortic integrity and limit aneurysm due to fewer EPCs, but also due to inefficacious late EPCs in AAA pa-growth [26]. These findings highlight EPCs' potential protective role tients. In experimental aneurysm models, enhanced cellularities of in vascular repair during AAA development. Thus, a possible theraendothelial cells and smooth muscle cells in the arterial wall under peutic strategy with statins and other medications may improve Please cite this article as: Sung S-H, et al, Reduced number and impaired function of circulating endothelial progenitor cells in patients with abdominal aortic aneurysm, Int J Cardiol (2012), http://dx.doi.org/10.1016/j.ijcard.2012.11.002 endothelial cells or even EPC function and modulate AAA progression [32,33]. Conversely, decreased and/or dysfunctional smooth muscle cells in the arterial media might be the sentinel changes related to MMP activation and extracellular matrix reduction. Sporadic studies have demonstrated an overexpression of proangiogenic cytokine and abundant neovascularization in the media of aneurysm rupture edge, compared to the normal aorta [34]. Given the circulating EPC potential role in neovascularization with vascular occlusion, they might be involved in aortic aneurysm rupture. However, since no patients had ruptured AAAs in our study, its findings might not relate to patients with relatively stable AAA. Plaque neovascularization is thought not only to participate intimately in the growth and progression of human atherosclerosis but also to play a role in plaque destabilization, causing plaque rupture [35]. However, recent clinical trials in acute or chronic CAD patients showed no increased incidence of plaque progression and/or rupture after cell therapy with vascular progenitor cells including EPCs [35,36]. Given the similar trends of circulating EPCs for disease progression in CAD and in AAA, circulating EPCs may contribute to aortic endothelial repair and vascular protection rather than to vascular injury and medial neovascularization in clinical AAA. 4.3. BP, pulse pressure, and hemodynamics in AAA development and progression Hemodynamic conditions affect AAA progression clinically, suggesting the need for adequate BP control [8]. However, whether BP lowering therapy prevents aneurysm progression is unknown [21]. In the present study, there was no difference in the presence of hypertension, baseline systolic and diastolic BPs, and the use of antihypertensives among the three groups, except that beta-blockers were more frequently used in patients with AAAs. On the other hand, pulse pressure, a surrogate of arterial stiffness, is associated with the incidence and clinical outcomes of cardiovascular diseases [37]. Although a high pulse pressure is generally considered a risk factor, patients with large AAAs tended to have a lower pulse pressure than patients with small AAAs. Given that low pulse pressure may be associated with low shear stress, our findings are partially compatible with previous in vivo findings that flow conditions may affect CD34+ cell localization and differentiation in experimental AAA. In rat AAA models, low flow and shear stress reduced endothelial proliferation, luminal surface re-endothelialization, and mural smooth muscle proliferation, and increased transmural macrophage infiltration, while promoting AAA progression. However, high flow limits AAA progression [38]. Accordingly, in our patients, both the impaired EPCs and low pulse pressure might have contributed to the poor differentiation and maturation of EPCs, persistent vascular inflammation, and medial hypocellularity, causing AAA progression. 4.4. Study limitations Some study limitations should be addressed. First, the patient sample was relatively small, and the presence of co-morbidities might affect circulating EPC levels. However, there was no significant heterogeneity of baseline characteristics among the study population, except for beta-blocker use. Furthermore, circulating EPC levels were evaluated comprehensively by different methods that showed similar EPC reductions in AAA patients. Since the controversies remain to identify circulating EPCs using flow cytometry or CFUs, while vast majority of these cells are thought to be hematopoietic lineage cells, future works are needed to calculate the true vasculogenic late outgrowth EPCs in peripheral circulating. Although it would be more interesting to measure the circulating mesenchymal stem cells and its association with EPCs since AAA is primarily a medial pathology, numerous studies continue to struggle in the definition. Second, late EPC functions were examined in a limited number of patients. However, age-matched cohorts were compared, and the data consistently showed impaired late EPC function by distinct functional assays. Moreover, the success rate of the current method for culturing late EPCs is over 80% in our laboratory. Both of the above may minimize potential selection bias. Finally, given this study's cross-sectional nature, no causal relationship can be inferred between impaired circulating EPCs and AAA. However, the correlations of EPCs and AAA were consistently and independently demonstrated by using different assays in this study, which might be applied to a longitudinal follow-up and potential cell therapy. 5. Conclusions Circulating EPC levels were reduced and the function of late EPCs was impaired in AAA patients. Though the underlying mechanisms were not elucidated, the current findings are the first to demonstrate the clinical connections between injury and repair of the vascular endothelium and AAA development and progression. Our findings also support the hypothesis that vascular progenitor cells such as EPCs might be a novel therapeutic target for AAA [38]. Future clinical studies, particularly longitudinal cohort studies with or without EPC-targeted pharmacological intervention, may be warranted. Funding This work was partly supported by research grants from the National Science Council (95-2314-B-010-024-MY3 to Chen JW, and UST-UCSD International Center of Excellence in Advanced Bio-engineering NSC99-2911-I-009-101 to Chen JW & Lin SJ) and from Taipei Veterans General Hospital (V97ER2-003 and VN 9803 to Chen JW, and V99B1003 to Sung SH), and also by a grant from the Ministry of Education “Aim for the Top University” Plan. Conflict of interest statement None declared. Acknowledgments The authors of this manuscript have certified that they comply with the Principles of Ethical Publishing in the International Journal of Cardiology [39]. References [1] Lederle FA, Johnson GR, Wilson SE, et al. Prevalence and associations of abdominal aortic aneurysm detected through screening. Aneurysm Detection and Management (ADAM) Veterans Affairs Cooperative Study Group. Ann Intern Med 1997;126: 441-9. [2] Blanchard JF, Armenian HK, Friesen PP. Risk factors for abdominal aortic aneurysm: results of a case–control study. Am J Epidemiol 2000;151:575-83. [3] Forsdahl SH, Singh K, Solberg S, Jacobsen BK. Risk factors for abdominal aortic aneurysms: a 7-year prospective study: the Tromso Study, 1994–2001. Circulation 2009;119:2202-8. [4] Davies MJ. Aortic aneurysm formation: lessons from human studies and experimental models. Circulation 1998;98:193-5. [5] Freestone T, Turner RJ, Coady A, Higman DJ, Greenhalgh RM, Powell JT. Inflammation and matrix metalloproteinases in the enlarging abdominal aortic aneurysm. Arterioscler Thromb Vasc Biol 1995;15:1145-51. [6] Touat Z, Ollivier V, Dai J, et al. Renewal of mural thrombus releases plasma markers and is involved in aortic abdominal aneurysm evolution. Am J Pathol 2006;168:1022-30. [7] Medina F, de Haro J, Florez A, Acin F. Relationship between endothelial dependent vasodilation and size of abdominal aortic aneurysms. Ann Vasc Surg 2010;24: 752-7. [8] Sho E, Sho M, Nanjo H, Kawamura K, Masuda H, Dalman RL. Hemodynamic regulation of CD34+ cell localization and differentiation in experimental aneurysms. Arterioscler Thromb Vasc Biol 2004;24:1916-21. [9] Hill JM, Zalos G, Halcox JP, et al. Circulating endothelial progenitor cells, vascular function, and cardiovascular risk. N Engl J Med 2003;348:593-600. Please cite this article as: Sung S-H, et al, Reduced number and impaired function of circulating endothelial progenitor cells in patients with abdominal aortic aneurysm, Int J Cardiol (2012), http://dx.doi.org/10.1016/j.ijcard.2012.11.002 [10] Kaushal S, Amiel GE, Guleserian KJ, et al. Functional small-diameter neovessels created using endothelial progenitor cells expanded ex vivo. Nat Med 2001;7:1035-40. [11] 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 2002;105:3017-24. [12] Michowitz Y, Goldstein E, Wexler D, Sheps D, Keren G, George J. Circulating endothelial progenitor cells and clinical outcome in patients with congestive heart failure. Heart 2007;93:1046-50. [13] Werner N, Kosiol S, Schiegl T, et al. Circulating endothelial progenitor cells and cardiovascular outcomes. N Engl J Med 2005;353:999–1007. [14] Dawson J, Tooze J, Cockerill G, Choke E, Loftus I, Thompson MM. Endothelial progenitor cells and abdominal aortic aneurysms. Ann N Y Acad Sci 2006;1085: 327-30. [15] Ma YC, Zuo L, Chen JH, et al. Modified glomerular filtration rate estimating equation for Chinese patients with chronic kidney disease. J Am Soc Nephrol 2006;17: 2937-44. [16] Corretti MC, Anderson TJ, Benjamin EJ, et al. Guidelines for the ultrasound assessment of endothelial-dependent flow-mediated vasodilation of the brachial artery: a report of the International Brachial Artery Reactivity Task Force. J Am Coll Cardiol 2002;39:257-65. [17] Chen YH, Lin SJ, Lin FY, et al. High glucose impairs early and late endothelial progenitor cells by modifying nitric oxide-related but not oxidative stress-mediated mechanisms. Diabetes 2007;56:1559-68. [18] Huang PH, Huang SS, Chen YH, et al. Increased circulating CD31+/annexin V+ apoptotic microparticles and decreased circulating endothelial progenitor cell levels in hypertensive patients with microalbuminuria. J Hypertens 2010;28: 1655-65. [19] Chen JW, Chen YH, Lin FY, Chen YL, Lin SJ. Ginkgo biloba extract inhibits tumor necrosis factor-alpha-induced reactive oxygen species generation, transcription factor activation, and cell adhesion molecule expression in human aortic endothelial cells. Arterioscler Thromb Vasc Biol 2003;23:1559-66. [20] Golledge J, Muller J, Daugherty A, Norman P. Abdominal aortic aneurysm: pathogenesis and implications for management. Arterioscler Thromb Vasc Biol 2006;26: 2605-13. [21] Brady AR, Thompson SG, Fowkes FG, Greenhalgh RM, Powell JT. Abdominal aortic aneurysm expansion: risk factors and time intervals for surveillance. Circulation 2004;110:16-21. [22] Shibamura H, Olson JM, van Vlijmen-Van Keulen C, et al. Genome scan for familial abdominal aortic aneurysm using sex and family history as covariates suggests genetic heterogeneity and identifies linkage to chromosome 19q13. Circulation 2004;109:2103-8. [23] van Laake LW, Vainas T, Dammers R, Kitslaar PJ, Hoeks AP, Schurink GW. Systemic dilation diathesis in patients with abdominal aortic aneurysms: a role for matrix metalloproteinase-9? Eur J Vasc Endovasc Surg 2005;29:371-7. [24] Jamous MA, Nagahiro S, Kitazato KT, et al. Endothelial injury and inflammatory response induced by hemodynamic changes preceding intracranial aneurysm formation: experimental study in rats. J Neurosurg 2007;107:405-11. [25] Roan JN, Yeh CY, Chiu WC, et al. Functional dilatation and medial remodeling of the renal artery in response to chronic increased blood flow. Kidney Blood Press Res 2011;34:447-56. [26] Hoshina K, Sho E, Sho M, Nakahashi TK, Dalman RL. Wall shear stress and strain modulate experimental aneurysm cellularity. J Vasc Surg 2003;37:1067-74. [27] Fuster V, Badimon L, Badimon JJ, Chesebro JH. The pathogenesis of coronary artery disease and the acute coronary syndromes (2). N Engl J Med 1992;326:310-8. [28] Knipp BS, Peterson DA, Rajagopalan S, et al. Impaired vasoreactivity despite an increase in plasma nitrite in patients with abdominal aortic aneurysms. J Vasc Surg 2002;35:363-7. [29] Huang PH, Chen YH, Chen YL, Wu TC, Chen JW, Lin SJ. Vascular endothelial function and circulating endothelial progenitor cells in patients with cardiac syndrome X. Heart 2007;93:1064-70. [30] Bertolini F, Shaked Y, Mancuso P, Kerbel RS. The multifaceted circulating endothelial cell in cancer: towards marker and target identification. Nat Rev Cancer 2006;6: 835-45. [31] Parietti E, Pallandre JR, Deschaseaux F, et al. Presence of circulating endothelial progenitor cells and levels of stromal-derived factor-1alpha are associated with ascending aorta aneurysm size. Eur J Cardiothorac Surg 2011;40:e6-12. [32] Schouten O, van Laanen JH, Boersma E, et al. Statins are associated with a reduced infrarenal abdominal aortic aneurysm growth. Eur J Vasc Endovasc Surg 2006;32: 21-6. [33] Sukhija R, Aronow WS, Sandhu R, Kakar P, Babu S. Mortality and size of abdominal aortic aneurysm at long-term follow-up of patients not treated surgically and treated with and without statins. Am J Cardiol 2006;97:279-80. [34] Choke E, Thompson MM, Dawson J, et al. Abdominal aortic aneurysm rupture is associated with increased medial neovascularization and overexpression of proangiogenic cytokines. Arterioscler Thromb Vasc Biol 2006;26:2077-82. [35] Finn AV, Jain RK. Coronary plaque neovascularization and hemorrhage: a potential target for plaque stabilization? JACC Cardiovasc Imaging 2010;3:41-4. [36] Ma ZL, Mai XL, Sun JH, et al. Inhibited atherosclerotic plaque formation by local administration of magnetically labeled endothelial progenitor cells (EPCs) in a rabbit model. Atherosclerosis 2009;205:80-6. [37] Franklin SS, Khan SA, Wong ND, Larson MG, Levy D. Is pulse pressure useful in predicting risk for coronary heart disease? The Framingham heart study. Circulation 1999;100:354-60. [38] Sho E, Sho M, Nanjo H, Kawamura K, Masuda H, Dalman RL. Hemodynamic regulation of CD34+ cell localization and differentiation in experimental aneurysms. Arterioscler Thromb Vasc Biol 2004;24:1916-21. [39] Coats AJ, Shewan LG. Ethics in the authorship and publishing of scientific articles. Int J Cardiol 2011;153:239-40. Please cite this article as: Sung S-H, et al, Reduced number and impaired function of circulating endothelial progenitor cells in patients with abdominal aortic aneurysm, Int J Cardiol (2012), http://dx.doi.org/10.1016/j.ijcard.2012.11.002