* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project
Download Aberrant Circulating Levels of Purinergic Signaling Markers are
Survey
Document related concepts
Transcript
Aberrant Circulating Levels of Purinergic Signaling Markers are Associated with Several Key Aspects of Peripheral Atherosclerosis and Thrombosis Juho Jalkanen1, Gennady G. Yegutkin2, Maija Hollmén2, Kristiina Aalto2, Tuomas Kiviniemi3, Veikko Salomaa4, Sirpa Jalkanen2 and Harri Hakovirta1,2 1 Department of Vascular Surgery, Turku University Hospital, Turku, Finland; 2Medicity Research Laboratory, Department of Microbiology and Immunology, University of Turku, Turku, Finland; 3Heart Center, Turku University Hospital, and; 4National Institute for Health and Welfare, Helsinki, Finland Running title: Purinergic Signaling in Peripheral Artery Disease Downloaded from http://circres.ahajournals.org/ by guest on June 14, 2017 Subject code: [17] Peripheral artery disease [134] Atherosclerosis basic science pathophysiology [135] Risk factors [138] Cell signaling Address correspondence to: Dr. Sirpa Jalkanen MediCity Research Laboratory University of Turku Tykistökatu 6 A FIN-20520 Turku Finland Tel: +358 2 333 7007 Fax: +358 2 333 7000 [email protected] In December 2014, the average time from submission to first decision for all original research papers submitted to Circulation Research was 14.47 days. DOI: 10.1161/CIRCRESAHA.116.305715 1 ABSTRACT Rationale: Purinergic signaling plays an important role in inflammation and vascular integrity, but little is known about purinergic mechanisms during the pathogenesis of atherosclerosis in humans. Objective: To study markers of purinergic signaling in a cohort of patients with peripheral artery disease (PAD). Downloaded from http://circres.ahajournals.org/ by guest on June 14, 2017 Methods and Results: Plasma ATP and ADP levels and serum nucleoside triphosphate diphosphohydrolase-1 (NTPDase1/CD39) and ecto-5'-nucleotidase/CD73 activities were measured in 226 stable peripheral artery disease (PAD) patients admitted for non-urgent invasive imaging and/or treatment. The major findings were that ATP, ADP, and CD73 values were higher in atherosclerotic patients than in controls without clinically evident PAD (P < 0.0001). Low CD39 activity was associated with disease progression (P = 0.01). In multivariable linear regression models high CD73 activity was associated with chronic hypoxia (P = 0.001). Statin use was associated with lower ADP (P = 0.041) and tended to associate with higher CD73 (P = 0.054), while lower ATP was associated with the use of angiotensin receptor blockers (P = 0.015). Conclusions: Purinergic signaling plays an important role in PAD progression. Elevated levels of circulating ATP and ADP are especially associated with atherosclerotic diseases of younger age, and smoking. The anti-thrombotic and anti-inflammatory effects of statins may partly be explained by their ability to lower ADP. We suggest that the pro-thrombotic nature of smoking could be a cause of elevated ADP, and this may explain why cardiovascular patients who smoke benefit from platelet P2Y12 receptor antagonists more than their non-smoking peers. Keywords: ADP, ATP, atherosclerosis, CD39, CD73, purinergic signaling, thrombosis, smoking, adenosine, adenosine receptor. Nonstandard Abbreviations and Acronyms: ARB angiotensin receptor blocker CAD coronary artery disease NT5E ecto-5'-nucleotidase NTPDase1 nucleoside triphosphate diphosphohydrolase-1 PAD peripheral artery disease P-AFOS plasma alkaline phosphatase DOI: 10.1161/CIRCRESAHA.116.305715 2 INTRODUCTION Although atherosclerosis is a widely studied disease and extensive interventions are practiced, it still remains a major cause of death in Western societies.1 The contemporary view of the disease as an inflammatory process of the vascular wall is well-established and reviewed in several excellent articles.1-3 The importance of extracellular purinergic signaling or metabolic pathways (i.e., the conversion of circulating ATP and ADP to AMP, and then to adenosine, by cell surface-associated and soluble nucleotidases) in inflammation and cell trafficking is acknowledged.4-7 Moreover, based on preclinical findings, evidence suggests that purinergic signaling plays an important role in atherosclerosis.8,9 The first such findings from human samples were reported by Lecka et al. in 201010, and strikingly, recent human genetic studies indicate that NT5E (ecto-5'-nucleotidase/CD73) mutations are an important factor contributing to peripheral arterial calcification.11 Thus, there is compelling evidence for the role of extracellular ATP metabolism in vascular inflammation, but in a broader spectrum, very little is known about the contribution of purinergic signaling in atherosclerosis. Downloaded from http://circres.ahajournals.org/ by guest on June 14, 2017 Endogenous nucleotides, ATP and ADP, are released into circulation from cells under stress and injury. After release, they create an important signaling cascade to modulate vascular tone, platelet aggregation, and inflammation.12,13 Firstly, the endothelial cell surface enzyme nucleoside triphosphate diphosphohydrolase-1 (NTPDase1, also known as CD39) converts ATP to ADP, and further to AMP. ADP is the most significant mediator of platelet aggregation via activation of the P2Y12 receptor expressed on platelets, and the clearance of ADP via NTPDase1/CD39 is crucial for thromboregulation.14-16 Subsequent breakdown of ATP/ADP-derived AMP to adenosine is mediated by another enzyme, ecto-5'nucleotidase/CD73.12 Adenosine itself acts as a powerful local anti-inflammatory agent regulating vascular permeability and leukocyte trafficking via different adenosine receptor subtypes.5,17,18 This study was designed to investigate whether plasma concentrations of ATP and ADP, and serum CD39 and CD73 activities, can discriminate different patient groups suffering from PAD and thus explain the mechanisms underlying disease onset and progression. METHODS This study was approved by the local Ethical Committee of the Hospital District of Southwestern Finland. By approval of the Ethical Committee, a register under the name The Role of Purinergic Signaling in Atherosclerosis (PURE ASO) was formed and is held at the Department of Vascular Surgery, Turku University Hospital. PAD patient cohort. For one year, from February 2012 to March 2013, we enrolled every patient diagnosed with a stable state of PAD of the lower extremities who were admitted to the Department of Vascular Surgery at the Turku University Hospital (Finland) for elective invasive treatment and imaging, i.e., open surgery or angiography and endovascular treatment. Patients admitted for urgent treatment from the emergency unit and those with acute ischemia, infections, or major tissue loss were not included in the study. During our enrollment period, 227 suitable patients were screened. Only one patient declined, and 226 gave written informed consent. The study patients had earlier been seen and diagnosed with PAD by a vascular surgeon at the outpatient clinic of the department. Non-atherosclerotic control cohort. For non-atherosclerotic controls, 64 middle-aged (25–50 years of age; 41 males and 23 females) volunteers not on permanent medications and without clinically evident PAD were recruited. In addition, to match the older age groups of the PURE ASO patient cohort, 89 frozen (˗70°C) samples were obtained from the DOI: 10.1161/CIRCRESAHA.116.305715 3 general population cohort of FINRISK 199719 (see Online Supplement for further details). Controls had to be free of chronic heart diseases, PAD, stroke, diabetes, cancer, chronic obstructive pulmonary disease (COPD), and rheumatic illnesses both on the basis of a self-report and register linkage with national health care databases. Blood samples. Samples were drawn in the morning after at least 4 hr of fasting. Two 9 mL samples of whole blood were obtained, and each sample was placed in a serum sample tube or EDTA sample tube and handled as described in the Online Supplement. Quantification of ATP and ADP levels in human plasma. Plasma ATP and ADP levels were determined using an ATPlite enzyme-coupled assay kit with a long-lived luminescent signal (Perkin Elmer, Groningen, The Netherlands) as described previously20, with further details in the Online Supplement. Downloaded from http://circres.ahajournals.org/ by guest on June 14, 2017 Measurement of soluble nucleotidase activity in human serum. Soluble ADPase/NTPDase and 5’-nucleotidase activities were assayed radiochemically, as described previously21, with further details in the Online Supplement. Statistical analyses. Statistical analyses were performed in association with a professional statistical provider, 4Pharma Ltd. (Turku, Finland). First, simple distributional characteristics of purinergic signaling markers were compared between the controls and the patient cohort. Markers with skewed distributions were log-transformed to better fit to normality, which was tested using a Shapiro-Wilk test. The patient cohort was analyzed according to different subgroups and background variables, and compared against other subgroups or the controls using a t-test for normally distributed variables (including log-transformed data). Pearson correlations were used to study the associations of continuous variables. Linear regression models were used to study the possible trends in the means of log-transformed marker values across PAD localization and the association of markers to age as a continuous variable. After comparing the controls and the patient cohort, further explorative statistical modeling was performed using only the patient cohort. First, based on univariate analysis the associations of suspected cardiovascular risk factors (for CD39: smoking, COPD, hypoxia, uremia, renal insufficiency, and hypertension; for CD73: hypoxia, renal insufficiency, uremia; for ATP and ADP: smoking and hypertension) to log-transformed markers were tested individually in linear regression models containing gender, age group and PAD localization as fixed effects, and P-AFOS as a covariate. In addition, the associations of used medications (aspirin, beta-blockers, angiotensin converting enzyme (ACE) blockers, calcium channel blockers, furosemide, warfarin, nitroglycerin, metformin, oral or inhaled cortisone, angiotensin receptor blockers (ARBs), bisphosphonates and gliptins) to log-transformed markers were tested individually in linear regression models containing gender, age group, PAD classification, statin use, and clopidogrel use as fixed effects, and P-AFOS as a covariate. A stepwise backward elimination method was applied for all the above-mentioned models. The variable under question was retained in the model even when fulfilling the elimination criterion to examine its significance in the final reduced model. Second, the identified factors appearing to be related to the marker were then selected for the final multivariate model incorporating all the selected factors, and gender, age group, PAD localization and P-AFOS. This full combined model was then simplified to a reduced model using the backward elimination approach leaving only statistically significant factors into the model. In summary, we first tested cardiovascular risk factors and medications separately in the linear regression model. From these factors, a combined model was formed encompassing all factors that had shown significance. Finally, a backward elimination method was applied to the combined model to determine the most decisive factors affecting marker levels. DOI: 10.1161/CIRCRESAHA.116.305715 4 No P-value adjustments were performed. A P-value of 0.05 was used as the threshold for statistical significance. All statistical analyses were performed using SAS Software for Windows version 9.3. RESULTS Description of the patient cohort and controls. For a comprehensive description of the patient cohort and controls, see the Online Supplement including Tables I, II, and III. Elevated P-AFOS levels can interfere with interpretations of purinergic signaling, especially CD73. Downloaded from http://circres.ahajournals.org/ by guest on June 14, 2017 Before further analysis, the cohort was screened for increased P-AFOS levels. The reference values of the local hospital laboratory were used with an upper threshold of 105 U/L (international units per liter). As P-AFOS is elevated in acute hepatobiliar diseases, and bone-related disorders, it was our assumption (and unpublished data) that these diseases, especially hepatobiliar diseases, may have a significant effect on purinergic signaling without contributing significantly to atherosclerosis. Thus, the markers of purinergic signaling could be misinterpreted as a result of another underlying illness. Altogether, 12 patients with elevated P-AFOS were noted. In six of these patients, P-AFOS was only marginally over the threshold, and no underlying condition explaining this could be seen in the patient files. All these six patients were included in the final analysis. Six patients with clearly elevated P-AFOS levels (one patient with liver cirrhosis, two patients with chronic pancreatitis, and three patients with prostate carcinoma and bone metastases) were identified and excluded from the final statistical analyses. P-AFOS concentrations positively correlated with CD39 levels (Figure 1A). An even stronger positive correlation was observed with CD73 (Figure 1B). A slightly negative correlation was noted for P-AFOS with ATP and ADP (Figure 1C and D). The correlations of CD39, ATP, and ADP with P-AFOS were abolished after exclusion of the six previously mentioned patients with marked background illnesses. A positive correlation persisted for only P-AFOS and CD73 (Pearson r = 0.226, P < 0.001). Markers of purinergic signaling are elevated in the patient cohort. Normal serum CD39 activity is 12–20 nmol/mL/hr, and CD73 activity is 120–280 nmol/mL/hr.20The control group displayed consistently normal levels of CD39 and CD73 activity (Table 1). CD39 activity did not significantly differ between the PAD patients and the control group (P = 0.09), but CD73 activity was significantly higher in the patient cohort (P < 0.0001) than in the controls. Normal plasma ATP and ADP values are generally within a 500–3000 nmol/L range.20,21 Again, the control group displayed consistently normal levels of ATP and ADP (Table 1). Both ATP and ADP were clearly higher in the patient cohort than in the controls (P < 0.0001). There were no significant differences between genders amongst patients (data not shown). CD39 was rather normally distributed, but for CD73, ATP and ADP some patients had extremely high values, which skewed the distribution; thus log-transformed values were used to calculate statistical significance. 23 For further insight and analysis, the patient cohort was divided into previously derived age groups (<60, 60–69, 70–79, and >80 years) and into distinct clinically relevant subgroups of PAD depending on the localization of the disease (aorta-iliac, femoropopliteal, crural, or pedal). More proximal disease localizations tended to associate with higher ATP and ADP levels, and higher CD39 activity (see Figure 2A). However, despite this prominent trend, using log-transformed values and linear regression only CD39 showed a declining trend towards distal PAD (P = 0.017). All PAD localization categories significantly differed from the control group for ATP and ADP, but not for CD39 (Figure 2A). CD73 activity did not DOI: 10.1161/CIRCRESAHA.116.305715 5 show any meaningful trend depending on PAD localization, but aorta-iliac (P = 0.0002), femoropopliteal (P < 0.0001) and crural (P = 0.042) groups significantly differed when compared to healthy controls (data not shown). For patients of a younger age, ATP/ADP levels and CD39 activity tended to be higher than those of older age groups (see Figure 2B). However, only CD39 showed declining linear trend towards older age in patients (P = 0.055) and in controls (P < 0.0001). Different age groups were compared directly against their age-matched controls, and all showed clear statistically significant differences (Figure 2B). CD73 activity did not show any meaningful trend across age groups, and only patients in age group 70–79 showed a significant increase (P = 0.003) when compared to its corresponding control group (data not shown). Correlation of markers of purinergic signaling with dyslipidemia, hypertension, and hyperglycemia. Downloaded from http://circres.ahajournals.org/ by guest on June 14, 2017 Since cardiovascular risk factors are known to associate with different PAD localizations, it was our assumption that certain risk factors could affect purinergic signaling and lead to an atherosclerotic disease. Thus, we tested potential correlations of cholesterol levels, triglycerides, systolic blood pressure, glycemia, and creatinine levels against markers of purinergic signaling and found that several known risk factor related elements positively correlated with ATP and ADP concentrations in PAD patients, but not in controls without clinically evident PAD. The most pronounced results of these tests amongst patients are presented in Figure 3. Systolic blood pressure (P = 0.043, Figure 3A) and triglycerides (P = 0.048, not shown) had a statistically significant positive correlation with ADP concentration. Total cholesterol had a strong positive correlation with ATP concentration (P = 0.013, Figure 3C) and CD39 activity (P = 0.003, Figure 3B). Also triglycerides had a strong positive correlation with ATP (P = 0.005, not shown). To illustrate the effect of hyperglycemia, the GHbA1c value was used because it reflects a more constant elevation in blood glucose levels than a single fasting glucose value. ATP and ADP levels correlated positively with increased glucose levels as a measure of GHbA1c, but did not reach statistical significance (Figure 3D). Creatinine levels did not positively correlate with ATP or ADP in the patient cohort. We also performed the same testing with the control group. Systolic blood pressure, LDL cholesterol, triglycerides, and creatinine were not significantly correlated with markers of purinergic signaling (data not shown). Only total cholesterol had a slight positive correlation with ATP (P = 0.026). Active smoking is associated with high ATP and ADP levels and increased CD39 activity in PAD patients. To examine the effect of smoking on purinergic signaling as a cardiovascular risk factor, we formed subgroups from PAD patients with only one distinct cardiovascular risk factor: smoking, hypertension, dyslipidemia, or diabetes (see Table IV in the Online Supplement for subgroup characteristics). Especially ADP tended to be higher in PAD patients with only smoking as a cardiovascular risk factor when compared to other PAD subgroups with one distinct risk factor (see Table 2A). Smoking did not elevate ATP and ADP levels in control subjects without clinically evident PAD (data not shown). ATP (P < 0.0001) and ADP (P < 0.0001) levels were clearly higher and CD39 to some extent higher (P = 0.043) in smoking PAD patients when compared to smoking controls (Table 2B). Similarly, we also tested PAD patients who had only dyslipidemia as a risk factor against control subjects with dyslipidemia. Control subjects with clear dyslipidemia had significantly lower ATP (P = 0.015) and ADP (P = 0.0475) when compared to PAD patients with only dyslipidemia as a risk factor (Table 2C). The entire patient cohort was further tested for the effect of smoking on all markers of purinergic signaling. CD39 activity steadily increased with the degree of smoking: patients who had never smoked (mean, 16.4; SD, 6.4 nmol/mL/hr), had quit smoking (mean, 17.9; SD, 5.5 nmol/mL/hr) or were active smokers (mean, 20.3; SD, 7.7 nmol/mL/hr). Active smokers had significantly higher CD39 activity than DOI: 10.1161/CIRCRESAHA.116.305715 6 those who had smoked but quit (P = 0.041) or those who had never smoked (P = 0.001), using the Student’s t-test. Low CD39 activity is associated with disease progression. CD39 activity was significantly lower in patients with critical ischemia (n = 118; mean, 17.3; SD, 6.5 nmol/mL/hr) than in patients with claudication (n = 97; mean, 19.3; SD, 6.9 nmol/mL/hr) using Student’s t-test (P = 0.025). This suggests that higher CD39 activity could protect from disease progression to critical ischemia. However, this could also be associated with an increasing severity of ischemia because CD39 activity steadily decreased from Rutherford values of 1 to 6 (i.e., from mild claudication to major tissue loss) giving a correlation (Spearman) of ˗0.174 (P = 0.01). Other markers did not show any correlations. Similarly, patients with severe coronary artery disease (CAD) (mean, 17.7; SD, 7.6 nmol/mL/hr) had lower CD39 than patients with mild CAD (mean, 19.1; SD, 6.3 nmol/mL/hr), but this was not statistically significant (P = 0.264). Downloaded from http://circres.ahajournals.org/ by guest on June 14, 2017 Explorative statistical modeling of multiple background variables related to high or low levels of ATP and ADP, or CD39 and CD73 activity. In accordance with the previously presented trends of increased ATP and ADP levels, and CD39 and CD73 activity in the patient cohort, explorative statistical modeling of multiple background variables was performed (by linear regression) to gain more insight on which variables affect marker levels. For this, only the patient cohort was used, with log-transformed values for purinergic markers. All of the statistically significant findings are summarized in Table 3. The first column, “Full combined model”, indicates all variables, which showed some significance in their respective group analysis (demographic factors, cardiovascular risk factors, and medication) and were used in the final combined reduction model. The second column, “Reduced combined model”, shows variables that remained significant. For ATP, only high P-AFOS levels and the use of angiotensin receptor blockers (ARBs) were associated with low levels of ATP. Similarly for ADP, high P-AFOS was associated with low values, but the relationship was not as strong as that of ATP. For ADP, younger age and hypertension were associated with high values, and the use of statin medication was associated with low values. Despite this finding, no association between the use of statins and CD39 activity was detected. No associations between CD39 and medications were detected at all. For CD39 only smoking and uremia were significant in the final combined model. Out of all of the markers, CD73 had the strongest and most diverse associations with different factors. The most pronounced phenomenon was the association of high CD73 activity with chronic hypoxia, i.e., severe COPD, but not to milder COPD or smoking. In addition, higher P-AFOS levels strongly associated with increased CD73 activity, as already noted in Figure 1. Surprisingly, the use of warfarin was associated with increased CD73 activity, as was female gender. When medications alone were tested, the use of statins had the strongest association with increased CD73 activity (data not shown), but this effect was to some extent diminished in the final combined model, in which warfarin exhibited a stronger association. DOI: 10.1161/CIRCRESAHA.116.305715 7 DISCUSSION We hypothesized that malfunctions of CD73-derived extracellular adenosine production would contribute to atherosclerosis and that this effect would be observed in our PAD patient cohort. Instead, throughout our patient cohort, we observed high CD73 activity, and to some extent CD39 activity as well. Elevated CD73 was especially associated with hypoxemic atherosclerotic disease, while elevated CD39 was associated with smoking and ATP/ADP-dependent entities. All markers of purinergic signaling were normal in the controls, and values of ATP, ADP, and CD73 were significantly higher in atherosclerotic patients. CD39 activity was not significantly increased in the patient cohort as a result of the high CD39 activity in the youngest control population (controls <40 years, Figure 2B). The youngest control group with the highest CD39 activity also had the lowest ATP and ADP levels (see Figure 2B). Downloaded from http://circres.ahajournals.org/ by guest on June 14, 2017 Our findings are in line with previous studies of the role of CD39 and ATP/ADP interplay in thrombosis and vascular inflammation.24,25 It is also well-documented from lung tissue and bronchial fluid samples that purinergic markers, especially CD73, are elevated in the presence of COPD and hypoxia,26,27 and in in vivo animal studies demonstrating the effects of CD73 in hypoxia transduction.17 In the present study, the same effects were observed in blood samples from PAD patients. However, CD73 activity was clearly more associated with a chronically hypoxic state than CD39 activity, and CD39 activity had a stronger relationship with active smoking, which could be interpreted as intermittent hypoxia. Both CD73 and CD39 expression and activity can be driven by hypoxia,28 but as seen here and before,29 CD39 is somewhat unaffected by chronic hypoxia, but instead intermittent hypoxia can have an effect. However, the increased ATP and ADP levels in systemic circulation of the studied patients may reflect diminished endothelial bound ecto-nucleotidase levels in the vascular wall. Similar experimental results were reported previously both in young atherosclerosis-prone apolipoprotein E-deficient mice26 and in chronically hypoxic bovine vasa vasorum endothelial cells.29 A similar underlying pathological mechanism may occur in the atherosclerotic hypoxic vessels observed in the patient cohort of this study. Lost ecto-nucleotidases from the vascular wall could be reflected in the high circulating levels observed herein. Notably, NTPDase1/CD39 and other nucleotidases may circulate in the bloodstream either as "true" soluble enzymes16,21 or in the form of microparticle-embedded enzymes30. Similar patterns of nucleotide metabolism were observed in human serum and heparinized plasma, and also after additional ultracentrifugation of serum samples for 1 hr at 100,000 g21 (and our unpublished data). These data exclude the potential release of membrane-bound nucleotidases from the blood elements in the course of serum preparation and provide further evidence for the predominant contribution of soluble rather than microparticle-associated enzymes to the measured NTPDase/ADPase and ecto-5'-nucleotidase/CD73 activities. The results of this study indicate that in a general population of patients with PAD, it is not the impairment of CD39 and CD73 that contributes to atherosclerosis, but it is the burden of circulating ATP and ADP that plays a more determinant role in the pathogenesis of the disease. This mechanism is especially notable in young patients. Old age, which is a known major independent risk factor for PAD, does not seem to play a significant role within this mechanism. Alternatively, age-related impairments (e.g., of CD39), even in the absence of increased levels of ATP and ADP, could be a contributing factor. It is a well-known clinical observation amongst practitioners of vascular surgery, as also demonstrated by Diehm et al.,31 that smoking is associated with proximal PAD in young patients, while old age is often associated with a distal disease. Now, for the first time, our findings demonstrate that this clinical phenomenon is connected to changes at a molecular level. DOI: 10.1161/CIRCRESAHA.116.305715 8 Downloaded from http://circres.ahajournals.org/ by guest on June 14, 2017 All atherosclerotic patients in this cohort displayed relatively high levels of ATP and ADP when compared to those of the controls and known normal levels derived from past experiments.20,21 Distinct cardiovascular risk factors were shown to drive the increase in ATP and ADP levels amongst subjects with a clinically significant atherosclerotic disease, but not amongst the controls. However, in the multivariable modeling, high ATP levels could not be clearly appointed to any specific cardiovascular risk factor, and ADP could only be appointed to hypertension. ATP and ADP appeared to be especially high in proximal PAD and younger age, but to some extent this phenomenon lacked strength after log-transformation. This observation could, however, strongly associate with smoking. ADP was clearly higher in patients who had only smoking as a cardiovascular risk factor when compared to smoking controls and other risk factorbased subgroups. PAD patients with only diabetes as a risk factor, on the other hand, had the lowest ATP and ADP levels and amongst the cardiovascular risk factor-related laboratory values, hyperglycemia had the weakest correlation with ATP and ADP. An explanation to this weak association could be that, in practice, diabetes is mostly associated with distal PAD, while smoking, dyslipidemia, and hypertension are associated with proximal PAD.31 Thus, ATP and ADP could be a common denominator of molecular pathophysiology amongst several cardiovascular risk factors, although smoking may be the strongest driver for both of these factors, especially ADP. In addition to high levels of ATP and ADP, smoking patients exhibited significant CD39 activity. This could indicate that CD39 activity is required to clear the elevated levels of ATP and ADP caused by smoking. It would be logical to assume that without an elevated level of CD39 activity, ATP and ADP levels would be even higher in smoking patients. Lower CD39 activity could contribute to disease progression amongst PAD patients. Such an effect was observed when comparing patients with claudication to those with critical ischemia. Lower CD39 activity was observed in patients with critical ischemia, and CD39 activity decreased steadily as the disease progressed according to the Rutherford classification. In previous studies, soluble CD39 exhibited significant antithrombotic effects.15,16,32 Building on clinical knowledge and past findings, it is known that smoking has a highly damaging effect on lower extremity vascular bypass grafts. Smoking independently increases the risk of graft failure by up to 4-fold.33 It was recently shown that smokers with atherosclerotic diseases benefit more from antiplatelet therapy targeting the P2Y12-receptor than non-smoking atherosclerotic patients. However, the mechanism remains unknown.34-36 The effect of smoking on cytochrome P450 (CYP)1A2, which converts clopidogrel into its active metabolite, was provided as a rationale,37 but this does not explain a similar smoker’s paradox observed with prasugrel and ticagrelor, which have different mechanisms of action.38 On the basis of our findings, we suggest that the beneficial effect is specifically due to the inhibition of ADP via the P2Y12 receptor expressed on platelets because circulating ADP levels are elevated in smokers. Moreover, statins are known to have anti-inflammatory and antithrombotic effects. A suggested mechanism of action could be an improvement in CD39 activity.39 In the present study, the use of statins was not associated with CD39 activity, but the use of statins was clearly associated with lower ADP levels. We already observed a similar phenomenon in a prospective setting in which young diabetic patients exhibited low ADP values after statin therapy, but no change in CD39 activity.40 As Kaneider et al. showed in an in vitro study, statins restored impaired CD39 function.39 They also did not report increased levels of expression. Thus, it could be that the beneficial effects of statins are directed to CD39 on the vascular wall and are only observed as changes in nucleotide, but not NTPDase, levels when measuring circulating values. However, as noted in the present study, and as shown earlier, statins improve CD73 expression, although the effect is only transient.40,41 An explanation could be that statins inhibit Rho-GTPase-dependent endocytosis of ecto-nucleotidases41 but lack the ability to induce new protein synthesis. The use of antihypertensive ARBs, however, was clearly associated with low ATP values amongst the patient cohort. This could to some extent explain the beneficial effects in cardiovascular events associated with the use of ARBs.42,43 However, on the basis of our results, this is currently an entirely theoretical interpretation. DOI: 10.1161/CIRCRESAHA.116.305715 9 In conclusion, we showed, for the first time, changes in intravascular nucleotide turnover in different subgroups of patients suffering from PAD and related cardiovascular risk factors. We believe that our findings indicate an important role for purinergic signaling during the atherosclerotic process, particularly contributing to atherosclerotic diseases associated with a younger age, tobacco smoking, and thrombosis. ACKNOWLEDGMENTS We thank Dr. Jan-Erik Wickström for helping to recruit patients, Tommi Pesonen, M.Sc., for professional assistance and guidance in statistical analyses, and Sari Mäki, Anne Meyer, and Teija Kanasuo for technical assistance. Downloaded from http://circres.ahajournals.org/ by guest on June 14, 2017 SOURCES OF FUNDING The study was supported by the Academy of Finland, the Sigrid Juselius Foundation, and the Clinical Research Fund (EVO) of Turku University Hospital. DISCLOSURES The authors have nothing to disclose. REFERENCES 1. Weber C, Noels H. Atherosclerosis: current pathogenesis and therapeutic options. Nat Med. 2011;17:1410–1422. doi:10.1038/nm.2538. 2. Hansson GK. Inflammation, atherosclerosis, and coronary artery disease. N Engl J Med. 2005;352:1685–1695. doi:10.1056/NEJMra043430. 3. Libby P. Inflammation in atherosclerosis. Arterioscler Thromb Vasc Biol. 2012;32:2045–2051. doi:10.1161/ATVBAHA.108.179705. 4. Colgan SP, Eltzschig HK, Eckle T, Thompson LF. Physiological roles for ecto-5'-nucleotidase (CD73). Purinergic Signal. 2006;2:351–360. doi:10.1007/s11302-005-5302-5. 5. Grünewald JK, Ridley AJ. CD73 represses pro-inflammatory responses in human endothelial cells. J Inflamm (Lond). 2010;7:10. doi:10.1186/1476-9255-7-10. 6. Antonioli L, Pacher P, Vizi ES, Haskó G. CD39 and CD73 in immunity and inflammation. Trends Mol Med. 2013;19:355–367. doi:10.1016/j.molmed.2013.03.005. 7. Eltzschig HK, Sitkovsky MV, Robson SC. Purinergic signaling during inflammation. N Engl J Med. 2012;367:2322–2333. doi:10.1056/NEJMra1205750. 8. Zernecke A, Bidzhekov K, Ozüyaman B, Fraemohs L, Liehn EA, Lüscher-Firzlaff JM, Lüscher B, Schrader J, Weber C. CD73/ecto-5'-nucleotidase protects against vascular inflammation and neointima formation. Circulation. 2006;113:2120–2127. doi:10.1161/CIRCULATIONAHA.105.595249. DOI: 10.1161/CIRCRESAHA.116.305715 10 Downloaded from http://circres.ahajournals.org/ by guest on June 14, 2017 9. Buchheiser A, Ebner A, Burghoff S, Ding Z, Romio M, Viethen C, Lindecke A, Köhrer K, Fischer JW, Schrader J. Inactivation of CD73 promotes atherogenesis in apolipoprotein E-deficient mice. Cardiovasc Res. 2011;92:338–347. doi:10.1093/cvr/cvr218. 10. Lecka J, Bloch-Boguslawska E, Molski S, Komoszynski M. Extracellular purine metabolism in blood vessels (Part II): Activity of ecto-enzymes in blood vessels of patients with abdominal aortic aneurysm. Clin Appl Thromb Hemost. 2010;16:650–657. doi:10.1177/1076029609354329. 11. St Hilaire C, Ziegler SG, Markello TC, Brusco A, Groden C, Gill F, Carlson-Donohoe H, Lederman RJ, Chen MY, Yang D, Siegenthaler MP, Arduino C, Mancini C, Freudenthal B, Stanescu HC, Zdebik AA, Chaganti RK, Nussbaum RL, Kleta R, Gahl WA, Boehm M. NT5E mutations and arterial calcifications. N Engl J Med. 2011;364:432–442. doi:10.1056/NEJMoa0912923. 12. Gordon JL. Extracellular ATP: effects, sources and fate. Biochem J. 1986;233:309–319. 13. Zimmermann H, Zebisch M, Sträter N. Cellular function and molecular structure of ectonucleotidases. Purinergic Signal. 2012;8:437–502. doi:10.1007/s11302-012-9309-4. 14. Robson SC, Wu Y, Sun X, Knosalla C, Dwyer K, Enjyoji K. Ectonucleotidases of CD39 family modulate vascular inflammation and thrombosis in transplantation. Semin Thromb Hemost. 2005;31:217–233. doi:10.1055/s-2005-869527. 15. Huttinger ZM, Milks MW, Nickoli MS, Aurand WL, Long LC, Wheeler DG, Dwyer KM, d'Apice AJF, Robson SC, Cowan PJ, Gumina RJ. Ectonucleotide triphosphate diphosphohydrolase-1 (CD39) mediates resistance to occlusive arterial thrombus formation after vascular injury in mice. Am J Pathol. 2012;181:322–333. doi:10.1016/j.ajpath.2012.03.024. 16. Yegutkin GG, Wieringa B, Robson SC, Jalkanen S. Metabolism of circulating ADP in the bloodstream is mediated via integrated actions of soluble adenylate kinase-1 and NTPDase1/CD39 activities. FASEB J. 2012;26:3875–3883. doi:10.1096/fj.12-205658. 17. Thompson LF, Eltzschig HK, Ibla JC, Van De Wiele CJ, Resta R, Morote-Garcia JC, Colgan SP. Crucial role for ecto-5'-nucleotidase (CD73) in vascular leakage during hypoxia. J Exp Med. 2004;200:1395–1405. doi:10.1084/jem.20040915. 18. Thompson LF, Takedachi M, Ebisuno Y, Tanaka T, Miyasaka M, Mills JH, Bynoe MS. Regulation of leukocyte migration across endothelial barriers by ECTO-5'-nucleotidase-generated adenosine. Nucleosides Nucleotides Nucleic Acids. 2008;27:755–760. doi:10.1080/15257770802145678. 19. Vartiainen E, Jousilahti P, Alfthan G, Sundvall J, Pietinen P, Puska P. Cardiovascular risk factor changes in Finland, 1972-1997. Int J Epidemiol. 2000;29:49–56. 20. Helenius M, Jalkanen S, Yegutkin G. Enzyme-coupled assays for simultaneous detection of nanomolar ATP, ADP, AMP, adenosine, inosine and pyrophosphate concentrations in extracellular fluids. Biochim Biophys Acta. 2012;1823:1967–1975. doi:10.1016/j.bbamcr.2012.08.001. 21. Yegutkin GG, Samburski SS, Mortensen SP, Jalkanen S, González-Alonso J. Intravascular ADP and soluble nucleotidases contribute to acute prothrombotic state during vigorous exercise in DOI: 10.1161/CIRCRESAHA.116.305715 11 humans. J Physiol (Lond). 2007;579:553–564. doi:10.1113/jphysiol.2006.119453. Downloaded from http://circres.ahajournals.org/ by guest on June 14, 2017 22. Niemelä J, Ifergan I, Yegutkin GG, Jalkanen S, Prat A, Airas L. IFN‐β regulates CD73 and adenosine expression at the blood–brain barrier. Eur J Immunol. 2008;38:2718–2726. doi:10.1002/eji.200838437. 23. Bellingan G, Maksimow M, Howell DC, Stotz M, Beale R, Beatty M, Walsh T, Binning A, Davidson A, Kuper M, Shah S, Cooper J, Waris M, Yegutkin GG, Jalkanen J, Salmi M, Piippo I, Jalkanen M, Montgomery H, Jalkanen S. The effect of intravenous interferon-beta-1a (FP-1201) on lung CD73 expression and on acute respiratory distress syndrome mortality: an open-label study. The Lancet Respiratory Medicine. 2014;2:98–107. doi:10.1016/S2213-2600(13)70259-5. 24. Mercier N, Kiviniemi TO, Saraste A, Miiluniemi M, Silvola J, Jalkanen S, Yegutkin GG. Impaired ATP-induced coronary blood flow and diminished aortic NTPDase activity precede lesion formation in apolipoprotein E-deficient mice. Am J Pathol. 2012;180:419–428. doi:10.1016/j.ajpath.2011.10.002. 25. Burnstock G, Ralevic V. Purinergic signaling and blood vessels in health and disease. Pharmacol Rev. 2014;66:102–192. doi:10.1124/pr.113.008029. 26. Zhou Y, Murthy JN, Zeng D, Belardinelli L, Blackburn MR. Alterations in adenosine metabolism and signaling in patients with chronic obstructive pulmonary disease and idiopathic pulmonary fibrosis. PLoS ONE. 2010;5:e9224. doi:10.1371/journal.pone.0009224. 27. Esther CR, Lazaar AL, Bordonali E, Qaqish B, Boucher RC. Elevated airway purines in COPD. Chest. 2011;140:954–960. doi:10.1378/chest.10-2471. 28. Eltzschig HK, Ibla JC, Furuta GT, Leonard MO, Jacobson KA, Enjyoji K, Robson SC, Colgan SP. Coordinated adenine nucleotide phosphohydrolysis and nucleoside signaling in posthypoxic endothelium: role of ectonucleotidases and adenosine A2B receptors. J Exp Med. 2003;198:783– 796. doi:10.1084/jem.20030891. 29. Yegutkin GG, Helenius M, Kaczmarek E, Burns N, Jalkanen S, Stenmark K, Gerasimovskaya EV. Chronic hypoxia impairs extracellular nucleotide metabolism and barrier function in pulmonary artery vasa vasorum endothelial cells. Angiogenesis. 2011;14:503–513. doi:10.1007/s10456-0119234-0. 30. Banz Y, Beldi G, Wu Y, Atkinson B, Usheva A, Robson SC. CD39 is incorporated into plasma microparticles where it maintains functional properties and impacts endothelial activation. Br J Haematol. 2008;142:627–637. doi:10.1111/j.1365-2141.2008.07230.x. 31. Diehm N, Shang A, Silvestro A, Do D-D, Dick F, Schmidli J, Mahler F, Baumgartner I. Association of cardiovascular risk factors with pattern of lower limb atherosclerosis in 2659 patients undergoing angioplasty. Eur J Vasc Endovasc Surg. 2006;31:59–63. doi:10.1016/j.ejvs.2005.09.006. 32. Buergler JM, Maliszewski CR, Broekman MJ, Kaluza GL, Schulz DG, Marcus AJ, Raizner AE, Kleiman NS, Ali NM. Effects of SolCD39, a novel inhibitor of Platelet Aggregation, on Platelet Deposition and Aggregation after PTCA in a Porcine Model. J Thromb Thrombolysis. 2005;19:115–122. doi:10.1007/s11239-005-1381-y. DOI: 10.1161/CIRCRESAHA.116.305715 12 Downloaded from http://circres.ahajournals.org/ by guest on June 14, 2017 33. Willigendael EM, Teijink JAW, Bartelink M-L, Peters RJG, Büller HR, Prins MH. Smoking and the patency of lower extremity bypass grafts: a meta-analysis. J Vasc Surg. 2005;42:67–74. doi:10.1016/j.jvs.2005.03.024. 34. Dong A, Caicedo J, Han SG, Mueller P, Saha S, Smyth SS, Gairola CG. Enhanced platelet reactivity and thrombosis in Apoe-/- mice exposed to cigarette smoke is attenuated by P2Y12 antagonism. Thromb Res. 2010;126:e312–7. doi:10.1016/j.thromres.2010.03.010. 35. Rollini F, Franchi F, Cho JR, Degroat C, Bhatti M, Ferrante E, Patel R, Darlington A, TelloMontoliu A, Desai B, Ferreiro J, Muniz-Lozano A, Zenni MM, Guzman LA, Bass TA, Angiolillo DJ. Cigarette smoking and antiplatelet effects of aspirin monotherapy versus clopidogrel monotherapy in patients with atherosclerotic disease: results of a prospective pharmacodynamic study. J Cardiovasc Transl Res. 2014;7:53–63. doi:10.1007/s12265-013-9535-3. 36. Gurbel PA, Bliden KP, Logan DK, Kereiakes DJ, Lasseter KC, White A, Angiolillo DJ, Nolin TD, Maa J-F, Bailey WL, Jakubowski JA, Ojeh CK, Jeong Y-H, Tantry US, Baker BA. The influence of smoking status on the pharmacokinetics and pharmacodynamics of clopidogrel and prasugrel: the PARADOX study. J Am Coll Cardiol. 2013;62:505–512. doi:10.1016/j.jacc.2013.03.037. 37. Desai NR, Mega JL, Jiang S, Cannon CP, Sabatine MS. Interaction between cigarette smoking and clinical benefit of clopidogrel. J Am Coll Cardiol. 2009;53:1273–1278. doi:10.1016/j.jacc.2008.12.044. 38. Gagne JJ, Bykov K, Choudhry NK, Toomey TJ, Connolly JG, Avorn J. Effect of smoking on comparative efficacy of antiplatelet agents: systematic review, meta-analysis, and indirect comparison. BMJ. 2013;347:f5307. 39. Kaneider NC, Egger P, Dunzendorfer S, Noris P, Balduini CL, Gritti D, Ricevuti G, Wiedermann CJ. Reversal of thrombin-induced deactivation of CD39/ATPDase in endothelial cells by HMGCoA reductase inhibition: effects on Rho-GTPase and adenosine nucleotide metabolism. Arterioscler Thromb Vasc Biol. 2002;22:894–900. 40. Kiviniemi TO, Yegutkin GG, Toikka JO, Paul S, Aittokallio T, Janatuinen T, Knuuti J, Rönnemaa T, Koskenvuo JW, Hartiala JJ, Jalkanen S, Raitakari OT. Pravastatin-induced improvement in coronary reactivity and circulating ATP and ADP levels in young adults with type 1 diabetes. Front Physiol. 2012;3:338. doi:10.3389/fphys.2012.00338. 41. Ledoux S. Lovastatin Enhances Ecto-5'-Nucleotidase Activity and Cell Surface Expression in Endothelial Cells: Implication of Rho-Family GTPases. Circ Res. 2002;90:420–427. doi:10.1161/hh0402.105668. 42. Molnar MZ, Kalantar-Zadeh K, Lott EH, Lu JL, Malakauskas SM, Ma JZ, Quarles DL, Kovesdy CP. Angiotensin-converting enzyme inhibitor, angiotensin receptor blocker use, and mortality in patients with chronic kidney disease. J Am Coll Cardiol. 2014;63:650–658. doi:10.1016/j.jacc.2013.10.050. 43. Smink PA, Miao Y, Eijkemans MJC, Bakker SJL, Raz I, Parving H-H, Hoekman J, Grobbee DE, de Zeeuw D, Lambers Heerspink HJ. The importance of short-term off-target effects in estimating the long-term renal and cardiovascular protection of angiotensin receptor blockers. Clin Pharmacol Ther. 2014;95:208–215. doi:10.1038/clpt.2013.191. DOI: 10.1161/CIRCRESAHA.116.305715 13 TABLE 1. Differences in Purinergic Signaling Markers: Patient Cohort vs. Controls Patients Variable N CD39 220 18.01 CD73 Controls Mean SE Median N 0.45 17.0 220 253 10.8 ATP 201 5165 ADP 190 4654 Mean SE Median P-value* 123 17.63 0.82 15.0 0.09 212.0 123 195 9.36 182 <0.0001 256 4088 153 1805 137 1104 <0.0001 288 3627 151 886 85 508 <0.0001 Downloaded from http://circres.ahajournals.org/ by guest on June 14, 2017 CD39 and CD73 are expressed as enzymatic activity in nmol/mL/hr. ATP and ADP are expressed as nmol/L. *Student’s t-test for log-transformed values. DOI: 10.1161/CIRCRESAHA.116.305715 14 TABLE 2. Markers of Purinergic Signaling in Subgroups of Patients with Only One Cardiovascular Risk Factor and Comparison to the Corresponding Group of Controls ATP (nmol/L) A ADP (nmol/L) Downloaded from http://circres.ahajournals.org/ by guest on June 14, 2017 Patient subgroup Mean SE Mean SE Smoking (N = 17) 6615 1107 6686 1220 Hypertension (N = 12) 6648 1350 5190 1249 Dyslipidemia (N = 9) 5718 1059 4869 1553 Diabetes (N = 8) 5109 844 4144 971 B Patients: smoking Variable N Mean SE N Mean SE P-value ATP (nmol/L) 16 6615 1107 29 2044 335 <0.0001* ADP (nmol/L) 15 6686 1220 29 846 122 <0.0001* CD39 (nmol/mL/hr) 17 18.9 1.9 16 13 1.4 0.043** CD73 (nmol/mL/hr) 17 229 31 16 191 27 0.103* Controls: smoking C Patients with dyslipidemia Controls with Variable N Mean SE N Mean SE P-value ATP (nmol/L) 9 5718 1059 14 2404 378 0.014** ADP (nmol/L) 8 4869 1553 14 1133 242 0.048** CD39 (nmol/mL/hr) 9 18.7 1.9 12 16.3 1.6 0.343** CD73 (nmol/mL/hr) 9 190 25 12 238 34 0.759* dyslipidemia * Student’s t-test for log-transformed values ** Student’s t-test for normally distributed values DOI: 10.1161/CIRCRESAHA.116.305715 15 TABLE 3. Results of the Linear Regression Model on Factors Affecting Markers of Purinergic Signaling Using the Patient Cohort Full combined model ATP Downloaded from http://circres.ahajournals.org/ by guest on June 14, 2017 ADP CD39 CD73 Proximal PAD 0.79 (0.20; SE, 0.23) Male gender 0.44 (˗0.08; SE, 0.11) Young age 0.32 (0.36; SE, 0.19) Statins 0.21 (˗0.13; SE, 0.11) ARBs 0.032 (˗0.29; SE, 0.13) P-AFOS 0.031 (˗0.006; SE, 0.003) Proximal PAD 0.92 (0.19; SE, 0.32) Male gender 0.20 (˗0.19; SE, 0.15) Young age 0.038 (0.64; SE, 0.29) Hypertension 0.023 (0.40; SE, 0.18) ACE blockers 0.32 (0.15; SE, 0.15) Gliptins 0.21 (0.29; SE, 0.23) Statins 0.018 (˗0.36; SE, 0.15) P-AFOS 0.031 (˗0.006; SE, 0.003) Proximal PAD 0.44 (0.14; SE, 0.12) Male gender 0.57 (˗0.03; SE, 0.05) Young age 0.97 (0.03; SE, 0.10) Clopidogrel 0.13 (0.14; SE, 0.10) Cortisone 0.25 (˗0.08; SE, 0.07) Smoking 0.13 (0.12; SE, 0.08) COPD 0.34 (0.07; SE, 0.08) Hypoxia 0.37 (0.11; SE, 0.12) Uremia 0.03 (0.26; SE, 0.12) P-AFOS 0.17 (0.002; SE, 0.001) Male gender 0.04 (˗0.17; SE, 0.08) Statins 0.05 (0.16; SE, 0.08) Nitroglycerin 0.18 (˗0.14; SE, 0.10) Warfarin 0.35 (0.20; SE, 0.09) Hypoxia 0.002 (0.55; SE, 0.17) P-AFOS 0.0001 (0.007; SE, 0.002) Reduced combined model ARBs 0.015 (˗0.317; SE, 0.129) P-AFOS 0.027 (˗0.006; SE, 0.003) Young age 0.042 (0.62, SE, 0.28) Hypertension 0.007 (0.47; SE, 0.17) Statins 0.041 (˗0.03; SE, 0.15) P-AFOS 0.07 (˗0.007; SE, 0.004) Smoking 0.045 (0.17; SE, 0.07) Uremia 0.023 (0.26; SE, 0.12) Male gender 0.03 (˗0.17; SE, 0.08) Hypoxia 0.001 (0.56; SE, 0.17) Warfarin 0.046 (0.19; SE, 0.09) Statins 0.054 (0.15; SE, 0.08) P-AFOS 0.0001 (0.007; SE, 0.002) The subsequent numerical value represents P-values with beta coefficients and standard errors in brackets for log-transformed variables. DOI: 10.1161/CIRCRESAHA.116.305715 16 FIGURE LEGENDS Figure 1. Correlations of P-AFOS and markers of purinergic signaling. (A) CD39, (B) CD73, (C) ATP, and (D) ADP. P-AFOS is expressed as U/L, CD39 and CD73 as nmol/mL/hr, and ATP and ADP as nmol/L. Downloaded from http://circres.ahajournals.org/ by guest on June 14, 2017 Figure 2. ATP, ADP, and CD39 values vary according to the localization of atherosclerotic lesions in PAD (A) and by age group (B). A, ATP, ADP, and CD39 values were high in proximal PAD and decline in distal PAD. The effect was strongest with CD39 (P = 0.017), calculated as a linear trend between groups by linear regression (linear trends between groups for ATP and ADP were not statistically significant). Bottom row: P-values represent a comparison to the control cohort using the Student’s t-test for logtransformed values. B, Similarly, ATP, ADP, and CD39 values were high in young age groups and steadily declined in the older age groups. Amongst patients the linear trend was strongest again with CD39 (P = 0.055) but does not reach statistical significance with CD39, nor with ATP and ADP. Amongst controls CD39 had a strong declining trend towards older age (P < 0.0001). Age groups are also compared directly with their age-matched controls using the Student’s t-test for log-transformed values (bottom row). CD39 is expressed as nmol/mL/hr, and ATP and ADP as nmol/L. Figure 3. Correlation of purinergic signaling markers with measurable numeric values relating to hypertension, hypercholesterolemia, and hyperglycemia. A, ADP positively correlates with systolic blood pressure. B and C, CD39 and ATP positively correlate with cholesterol levels. D, ATP tends to positively correlate with blood glucose levels measured by GHbA1c, but this did not reach statistical significance. CD39 is expressed as nmol/mL/hr, and ATP and ADP as nmol/L. DOI: 10.1161/CIRCRESAHA.116.305715 17 Novelty and Significance What Is Known? Purinergic signaling plays an important role in inflammation and vascular wall integrity. Impairment of the signaling cascade may lead to atherosclerosis. Smoking is the leading cause of lower extremity vascular bypass graft failure. What New Information Does This Article Contribute? Downloaded from http://circres.ahajournals.org/ by guest on June 14, 2017 Purinergic signaling, i.e., ATP metabolism, plays an important role in the development of peripheral artery disease at a young age and affects large arteries (aorta-iliac or femoropopliteal arteries). Smoking is associated with elevated ATP/ADP levels and disease burden. Cardiovascular patients who continue to smoke could benefit from permanent ADP/P2Y12 antiplatelet therapy, especially if they have a vascular bypass. In general, little is known about purinergic signaling in atherosclerosis, especially in peripheral artery disease (PAD). In this study, we provide evidence that the pro-thrombotic effect of tobacco smoke is due to elevated plasma ADP levels. Our findings provide evidence for a mechanism underlying the greater beneficial effects of clopidogrel-like antiplatelet therapy in smokers than non-smoking patients. The use of P2Y12 antiplatelet therapy for vascular bypass graft protection in PAD patients who smoke should be tested in a prospective randomized study. DOI: 10.1161/CIRCRESAHA.116.305715 18 Downloaded from http://circres.ahajournals.org/ by guest on June 14, 2017 Downloaded from http://circres.ahajournals.org/ by guest on June 14, 2017 Downloaded from http://circres.ahajournals.org/ by guest on June 14, 2017 Downloaded from http://circres.ahajournals.org/ by guest on June 14, 2017 Aberrant Circulating Levels of Purinergic Signaling Markers are Associated with Several Key Aspects of Peripheral Atherosclerosis and Thrombosis Juho Jalkanen, Gennady G Yegutkin, Maija Hollmén, Kristiina Aalto, Tuomas O Kiviniemi, Veikko Salomaa, Sirpa Jalkanen and Harri H Hakovirta Circ Res. published online February 2, 2015; Circulation Research is published by the American Heart Association, 7272 Greenville Avenue, Dallas, TX 75231 Copyright © 2015 American Heart Association, Inc. All rights reserved. Print ISSN: 0009-7330. Online ISSN: 1524-4571 The online version of this article, along with updated information and services, is located on the World Wide Web at: http://circres.ahajournals.org/content/early/2015/02/02/CIRCRESAHA.116.305715 Data Supplement (unedited) at: http://circres.ahajournals.org/content/suppl/2015/02/02/CIRCRESAHA.116.305715.DC1 Permissions: Requests for permissions to reproduce figures, tables, or portions of articles originally published in Circulation Research can be obtained via RightsLink, a service of the Copyright Clearance Center, not the Editorial Office. Once the online version of the published article for which permission is being requested is located, click Request Permissions in the middle column of the Web page under Services. Further information about this process is available in the Permissions and Rights Question and Answer document. Reprints: Information about reprints can be found online at: http://www.lww.com/reprints Subscriptions: Information about subscribing to Circulation Research is online at: http://circres.ahajournals.org//subscriptions/ ONLINE SUPPLEMENT Methods Patient Cohort For the 226 subjects forming the patient cohort, all major demographic factors, baseline characteristics, cardiovascular risk factors, and medications were recorded, in addition to specific data on the severity and distribution of PAD. Disease severity was classified according to the Rutherford classification and then converted to the usual clinically relevant three categories: no symptoms (Rutherford 0), claudication (Rutherford 1–3), and critical ischemia (Rutherford 4–6). Elective patients with minor ulcers or dry peripheral gangrene of a small area, e.g., one to two toes, were included in the study, but patients with infections or tissue loss leading to transmetatarsal amputation were not included. Baseline characteristics and cardiovascular or risk factors were determined on a yes/no basis, if recorded in the patient file. Type I and type II diabetic patients were evaluated separately, but also together as a single variable of three categories: no diabetes, diabetics on oral medication, and diabetic patients treated with insulin. Patients with coronary artery disease (CAD) were also assigned in a variable of three categories: i) no diagnosis or symptoms, ii) diagnosed and only mildly symptomatic (no invasive treatment or history of acute myocardial infarction, AMI), and iii) symptomatic (previous AMI or invasive treatment received). All autoimmune and rheumatic diseases were reported (e.g., systemic lupus erythematosus, psoriasis, inflammatory bowel disease, etc.), but the prevalence was so small that an overall “rheumatic disease” variable was formed. Patients with dyslipidemia were either already diagnosed and on statin therapy, or were newly diagnosed according to the current national guidelines (based on cholesterol and triglyceride levels) and were prescribed statin therapy during the index hospital stay. Patients were divided into the following age groups (<60, 60–69, 70–79, and >80 years) and categorized according to the distribution of disease burden in the lower extremities (aorta-iliac, femoropopliteal crural, and pedal). Patients could exhibit calcification at several levels, but a distinct category was assigned if one area was proven to be clearly affected according to the Inter-Society Consensus on the Management of Peripheral Artery Disease (TASC II) -classification and the practicing vascular surgeon. Only one patient was excluded from categorization as a result of several levels of significant atherosclerotic lesions. Non-atherosclerotic Control Cohort Sixty-four newly recruited volunteers were all screened by a practicing medical doctor of the research group for symptoms and signs of PAD. A palpable pedal pulse was used to determine whether subjects were without evidence of clinically evident PAD. Eighty-nine subjects (73 males, 16 females) were selected from the FINRISK 1997 study based on age (35–74 years), health status, and residence in either Southern Finland (Helsinki and Vantaa) or Eastern Finland (Province of North Karelia). A member of the research group did not see these subjects. The subjects had to be free of CHD, PAD, stroke, diabetes, cancer, COPD, and rheumatic illnesses on the basis of both a self-report and register linkage and national health care databases. These 89 subjects from FINRISK 1997 and the 64 new volunteers without clinically evident PAD formed the control pool (n = 153) for the PURE ASO patient cohort and were divided into age groups to match the patient cohort: <40 (n = 81), 40–59 (n = 16), 60–69 (n = 40), and 70–79 (n = 16) years. The oldest age group (>80) could not be matched. Blood Samples Samples were drawn in the morning after at least 4 hr of fasting. Nine mL of whole blood was drawn to a serum sample tube and 9 mL to an EDTA sample tube. The serum sample tube was left to clot at room temperature while it was transported to the Medicity Research Laboratory, University of Turku. On arrival, it was centrifuged at 2000 g for 10 min, after which the serum was extracted for analysis. For EDTA plasma samples, the accredited University Hospital Central Laboratory conducted standard creatinine, lipid, glucose, and alkaline phosphatase (P-AFOS) analyses according to a photometric International Federation of Clinical Chemistry and Laboratory Medicine (IFCC) procedure (values expressed as international units per liter, U/L), and the remaining plasma was transported to the Medicity Research Laboratory for ATP and ADP analysis. Venous blood samples from the FINRISK 1997 cohort were drawn in 1997. All blood sample aliquots were stored at ˗70°C prior to analysis. Quantification of ATP and ADP Levels in Human Plasma Briefly, 10 µL aliquots of EDTA plasma were transferred into two parallel wells of a white non-phosphorescent 96-well microplate containing 100 µL of PBS with (A) or without (B) a solution of 200 µM UTP and 5 U/mL NDP kinase from baker’s yeast S. cerevisiae (Sigma). Following the addition of 50 µL of ATP-monitoring reagent, sample luminescence was measured using a Tecan Infinite M200 microplate reader (Salzburg, Austria). The difference in luminescence signals between well “A” (ATP + ADP) and “B” (only ATP) enabled the quantification of ADP concentration, which was converted into ATP through an NDP kinase-mediated reaction in the presence of exogenous UTP. This approach allows simultaneous measurement of both ATP and ADP content within the same sample. Measurement of Soluble Nucleotidase Activities in Human Serum For ADPase/NTPDase activity, serum (10 µL) was incubated for 60 min at 37°C in 80 µL of RPMI-1640 medium containing 5 mM β-glycerophosphate, 80 µM adenylate kinase inhibitor Ap5A, and 50 µM ADP with a [2,8-3H]ADP tracer (Perkin Elmer, Boston, USA). Likewise, 5’-nucleotidase activity was assayed by incubating 10 µL of serum for 60 min with 300 µM [2-3H]AMP (Quotient Bioresearch, GE Healthcare, Rushden, UK). Radiolabelled substrates and their dephosphorylated products were separated by thin-layer chromatography and quantified by scintillation β-counting. Enzymatic activities were expressed as nanomoles of 3H-substrate metabolized per hr by 1 mL of serum. Results Description of the Patient Cohort The patient cohort consisted of 226 participants of which 128 (56.6%) were male. All were of Caucasian origin. Overall mean age was 69.8 years (SD ± 11.44, 46–93 years) with a small tendency for males to be younger (males, 67.9 years [SD ± 11.3], and females, 72.3 years [SD ± 11.2]). Registered illnesses and cardiovascular risk factors together with their prevalence are presented in Online Table I, along with basic laboratory values in Online Table II, and PAD-related descriptive variables and subgroup characteristics are presented in Online Table III. High blood pressure was very common in the patient cohort, up to 75% (Online Table I). The prevalence of rheumatic diseases was also somewhat high (16%). From these 35 rheumatic patients, only one was diagnosed with vasculitis, and all others had PAD accompanied by rheumatoid arthritis (20/35), psoriasis (8/35), systemic lupus erythematosus (3/35), or inflammatory bowel disease (3/35). Smoking, diabetes, and dyslipidemia were more prevalent in young age groups, while hypertension, renal failure, and rheumatic diseases were more abundant in older age groups. In a similar fashion, smoking and dyslipidemia were more prevalent in proximal PAD, and renal failure, rheumatic diseases, but also diabetes, were more prevalent in distal PAD. Amongst all cardiovascular risk factors, renal insufficiency and diabetes were also more prevalent in critical ischemia than in claudication (see Online Table III). Statin therapy was the most commonly used medication upon index hospitalization and blood sampling (64%). Following statins, the most commonly used medications were as follows: aspirin (59%), beta-blockers (56%), angiotensin converting enzyme blockers (44%), calcium channel blockers (31%), diuretic medications (27%, mostly furosemide), warfarin (22%), nitroglycerin (20%), metformin (19%), oral or inhaled cortisone (19%, topical use excluded), and angiotensin receptor blockers (ARBs) (18%), gliptins (11%), bisphosphonates (7%). Only 7% of patients were on clopidogrel at index hospitalization, and only 1% were on dipyridamole. Description of the Control Cohort The non-atherosclerotic control cohort consisted of 153 subjects of which 70.6% were male. The mean age was 45.9 years (SD ± 18.5, 20–74 years). The percentage of active smokers amongst the controls was 19%, of which 76% were male. In the laboratory values of control subjects, we identified three subjects with different degrees of renal insufficiency (creatinine, 212, 220, and 433 µmol/L). All had normal levels of purinergic signaling markers and remained in the study. We also identified 14 control subjects with clear dyslipidemia (total cholesterol >6.5 and LDL >3.5). They remained in the study and performed as controls to PAD patients with only dyslipidemia as a cardiovascular risk factor (see Online Table IV). Online Table II presents laboratory values for the patient cohort and controls. An interesting finding was that total cholesterol and LDL levels were statistically higher in the controls than in the patient cohort reflecting the fact that the patient cohort was under medication. Triglycerides, however, were higher in the patient cohort, as was systolic blood pressure. No difference was observed with HDL cholesterol or creatinine. Division of Subjects to Subgroups with One Distinct Cardiovascular Risk Factor For further insight and analysis, both patients and control subjects were divided into subgroups according to one single cardiovascular risk factor. No other risk factor was allowed. The groups formed, and their descriptions are presented in Online Table IV. We were able to form groups for smoking, hypertension, dyslipidemia, and diabetes for PAD patients, and groups for smoking and dyslipidemia for control subjects. No groups could be formed for uremia or renal insufficiency and rheumatic diseases, because they were always accompanied by another risk factor. There were also no PAD patients without at least one cardiovascular risk factor. Online Table I. Prevalence of Background Illnesses and Cardiovascular Risk Factors in the Patient Cohort (n = 226) and Control Cohort (n = 153) Prevalence in Patient Cohort Prevalence in Control Cohort COPD 21.2% (67% male) 0% Chronic hypoxia 5.8% (69% male) 0% Hypertension 74.8% (56% male) 0% Sleep apnea 4.9% (82% male) 0% Type I diabetes 6.2% (50% male) 0% Type II diabetes 28.8% (71% male) 0% Dyslipidemia 31.9% (57% male) 13% (81% male) Renal insufficiency 23.9% (65% male) 2% (100% male) Uremia 5.3% (83% male) 0% Rheumatic disease 15.9% (39% male) 0% Smoking Patient cohort Control cohort Coronary artery disease Patient cohort Control cohort Diabetes Patient cohort Control cohort Never smoked Quit smoking Current smoker 41.2% 27.4% 31.4% (41% male) (71% male) (65% male) 81% 0% No Mild 19% (76% male) Severe 58.0% 12.4% 29.6% (51% male) (57% male) (65% male) 100% 0% 0% No diabetes Oral treatment Insulin treatment 65% 14.2% 20.8% (51% male) (72% male) (64% male) 100% 0% 0% Online Table II. Basic Laboratory Values for the Patient Cohort (n = 226) and Control Cohort (n = 153) Patient Cohort Control Cohort P-value Systolic blood pressure (mmHg) 149 (SD, 27) 138 (SD, 22) <0.0001* Total cholesterol (mmol/L) 4.3 (SD, 1.15) 5.1 (SD, 1.1) <0.0001* LDL cholesterol (mmol/L) 2.23 (SD, 0.93) 3.1 (0.96) <0.0001* HDL cholesterol (mmol/L) 1.41 (SD, 0.51) 1.46 (SD, 0.42) ns Triglycerides (mmol/L) 1.4 (SD, 0.82) 0.87 (SD, 0.40) <0.0001* 95 (SD, 65) 88 (SD, 42) ns Creatinine (umol/L) GHbA1c (%) 6.89 (SD, 1.3) Fasting plasma glucose (mmol/L) *Student’s t-test for normally distributed values NA 5.2 (SD, 0.67) NA Online Table III. Characteristics of the PAD Patient Cohort According to Age, Disease Localization, and Severity (n = 226) <60 years 60–69 years 70–79 years >80 years 10.6% 32.7% 30.2% 26.5% 63%/37% 67%/33% 59%/41% 35%/65% History of smoking 75% 78% 64% 23% Hypertension 50% 74% 78% 86% Dyslipidemia 38% 40% 21% 33% Diabetes 42% 40% 38% 22% Renal insufficiency 13% 14% 25% 37% Rheumatic disease 8% 10% 24% 18% Aorta-iliac Femoropopliteal Crural Pedal Localization 26.1% 43.8% 23.5% 6.2% Male/female 56%/44% 58%/42% (53%/47%) 50%/50% Mean age (years) 66 (SD, 9.3) 71 (SD, 9.3) 78 (SD, 11.8) 74 (SD, 12.6) History of smoking 86% 69% 26% 7% Hypertension 63% 81% 83% 64% Dyslipidemia 44% 34% 25% 7% Diabetes 33% 43% 49% 50% Renal insufficiency 9% 16% 42% 64% Rheumatic disease 7% 13% 25% 43% No symptoms /missing Claudication Critical ischemia 2.3% 44.5% 53.2% 55%/45% 58%/42% 68 (SD, 9.3) 74 (SD, 11.5) Smoking 72% 50% Hypertension 76% 77% Dyslipidemia 42% 25% Diabetes 25% 45% Renal insufficiency 10% 35% Rheumatic disease 11% 20% Age group Male/female Disease severity Male/female Mean age (years) Online Table IV. Descriptive Information on Patient Subgroups with Only One Distinct Cardiovascular Risk Factor N Age Male % Syst BP Chol LDL HDL Trig Smoking (PAD) 17 57 (10) 48 144 (15) 4.5 (1.2) 2.4 (0.9) 1.5 (0.4) 1.4 (0.8) Hypertension (PAD) 12 77 (10) 33 164 (30) 3.9 (0.7) 2.3 (0.7) 1.2 (0.3) 1.1 (0.2) Dyslipidemia (PAD) 9 82 (5) 33 171 (25) 5.0 (1.2) 3.0 (1.2) 1.5 (0.2) 1.1 (0.3) Diabetes (PAD) 8 72 (15) 63 173 (24) 4.2 (0.9) 2.1 (0.7) 1.4 (0.5) 1.4 (0.6) Smoking (control) 29 50 (14) 76 135 (21) 5.3 (0.9) 3.3 (0.9) 1.4 (0.4) 1.5 (0.5) Dyslipidemia (control) 14 67 (5) 81 149 (22) 7.0 (0.6) 4.7 (0.6) 1.5 (0.6) 2.0 (0.7) Standard deviation in parentheses. Lipid levels are expressed as mmol/L, systolic blood pressure (Syst BP) in mmHg and age in years. Chol, total cholesterol; Trig, triglycerides.