Next Article in Journal
Protective Effect of Tomato-Oleoresin Supplementation on Oxidative Injury Recoveries Cardiac Function by Improving β-Adrenergic Response in a Diet-Obesity Induced Model
Next Article in Special Issue
Antioxidant and Anti-Inflammatory Effects of Curcumin Nanoparticles on Drug-Induced Acute Myocardial Infarction in Diabetic Rats
Previous Article in Journal
Development of a Method Suitable for High-Throughput Screening to Measure Antioxidant Activity in a Linoleic Acid Emulsion
Previous Article in Special Issue
Inhibition of Heme Oxygenase Antioxidant Activity Exacerbates Hepatic Steatosis and Fibrosis In Vitro
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Oxidative Stress in Peripheral Arterial Disease (PAD) Mechanism and Biomarkers

by
Salvatore Santo Signorelli
*,
Salvatore Scuto
,
Elisa Marino
,
Anastasia Xourafa
and
Agostino Gaudio
Department of Clinical and Experimental Medicine, University of Catania, 95125 Catania, Italy
*
Author to whom correspondence should be addressed.
Antioxidants 2019, 8(9), 367; https://doi.org/10.3390/antiox8090367
Submission received: 31 July 2019 / Revised: 29 August 2019 / Accepted: 30 August 2019 / Published: 2 September 2019
(This article belongs to the Special Issue Oxidative Stress in Vascular Disease )

Abstract

:
Hemodynamic dysfunction mainly characterizes pathophysiology of peripheral arterial disease (PAD) leading to chronic ischemia. Hemodynamic dysfunction is the origin of intermittent claudication (chronic PAD) or of critical limb ischemia (very severe PAD). Notably, it is well known that oxidative stress (OxS) plays a pathophysiological role in PAD. The higher production of reactive oxygen species (ROS) from OxS and reduced redox capability are two crucial players in initiating and progressing PAD. A number of biomarkers highlight OxS and monitor it in PAD. The present review summarizes data on OxS, on biomarkers available to mark OxS occurrence and to monitor on PAD progression, as well as to evaluate the effects treatments in PAD patients. In conclusion, by detailing OxS and its biomarkers, we hope to encourage more studies to focus on drugs which combat OxS and inflammation.

1. Introduction

Peripheral arterial disease (PAD) is one clinical aspect of atherosclerotic disease, presenting impaired blood flow in the lower limbs and where intermittent claudication is the most significant symptom (Table 1) [1,2]. PAD patients have a high risk of cardiovascular disease (CVD) morbidity [3,4], because they suffer from several arterial co-morbidities (i.e., coronary artery disease, carotid ischemic diseases causing a risk of ischemic stroke) [5,6]. Atherosclerosis can begin in young adults with the development of fatty streaks [5]. Smoking and type 2 diabetes mellitus represent the two major risk factors predisposing to PAD [1,2,3,4,5,6,7,8]. Moreover, dyslipidemia, hypertension, endothelial dysfunction [9], and pro-inflammatory molecules [10,11] contribute to atherosclerotic diseases. Epidemiology of PAD ranges to more than 200 million of subject worldwide, it affects a mainly affects 60–65 year-old individuals thus affecting more than 20 million patients in Europe and North America [12,13,14,15,16]. PAD mainly affects 60–65 year-old individuals thus affecting more than 20 million patients in Europe and North America. Atherosclerosis is an inflammatory disease so inflammation has a crucial role in promoting, initiating and progressing PAD [13,14,15,16,17]. A number of inflammatory biomarkers (i.e., acute phase proteins, C reactive protein, fibrinogen, pro-inflammatory cytokines) have been found to be associated with PAD. Moreover, oxidative stress (OxS) seems to play a pathophysiological role in PAD [18,19,20]. OxS leads to the accumulation of post-translational bio-molecules (e.g., protein carbonylation or aldehyde/ketone adducts, nitration and sulfoxidation, DNA lesions such as 8-oxodG), furthermore interfering with physiological redox capability. Reactive oxygen species (ROS) and nitrogen species (RNS) multiply in many chronic diseases whereas redox regulation lowers them. There is a close association between OxS and cardiovascular diseases, oxidative stress biomarkers marking their progression.

1.1. Oxidative Stress and PAD

The close relationship between OxS and the atherosclerotic process have been widely investigated [21,22,23,24]. On OxS in PAD, it has been proposed since 1977 that oxidation processes may play a role in PAD pathophysiology [25]. A number of surrogate (indirect) oxidative markers were sought for PAD. Results of studies highlighted on several markers of oxidative stress such as beta 2 microglobulin [26], high sensitivity C-reactive protein [27] and cystatin C [28]. Most recently, other biochemical indices as the chemochil ligand 2, subclasses of isoprostanes [29] were proposed as useful in responding about role played by oxidative stress and inflammation in PAD pathophysiology and in diagnosing more favorable biochemical pathway for PAD. On OxS as a pathophysiological key in PAD, it enhances the inflammatory pathways responsible for the atherosclerotic process [30,31,32,33,34,35,36], which may be summarized as the downstream outcome of the inflammatory cytokines which promote lipid and protein oxidation in artery walls. Low-density lipoprotein (LDL) oxidation is the initial step for the atherosclerotic process. OxS is closely related to impaired nitric oxide (NO) synthesis [31]. The lack of vasodilation in the narrowed atherosclerotic arteries of the lower limbs [37,38] is the precursor to ischemic injury to tissues (Figure 1) and cells leading to the multiplication in the reactive oxygen species (ROS) caused by the dysfunction of NO bio-availability [39,40]. The bio-availability of ROS is known to result from the rate of ROS formation, above all by the mitochondria, and from its rate of clearance through the antioxidant defense system [40]. ROS cooperates with OxS leading to the progressive dysfunction of the antioxidant defense system as shown in studies where there is a significant decrease in the activity of the principal enzymatic antioxidants [40,41,42,43,44]. Additionally, OxS promotes endothelial dysfunction, which in turn decreases NO production impairing its protective role. This initiates a dangerous mechanism [45] which leads to accelerating atherosclerosis (Figure 2). Reperfusion injury is a crucial issue in the chronic ischemia of PAD. PAD patients experience a severe reduction in blood flow during muscular activity, followed by reperfusion when the muscular stress ceases. Reperfusion increases the ROS production by mitochondria, and the increased OxS in turn causes injury to ischemic muscles [45,46]. PAD patients suffer from the myopathy characterized by a decrease in myofibre cross-sectional area [47]. Indeed, metabolic myopathy and OxS plays a crucial role in the pathophysiology of PAD operating as mechanisms behind the structural and metabolic changes occurring in ischemic muscles. [48] To learn more about OxS, several biomarkers have been proposed as helpful tools in evaluating more prone individuals and in monitoring PAD outcomes. These OxS biomarkers have proven to be closely correlated to the progression and severity of arterial disease [39,47,48,49]. A number of clinical trials have been carried out to evaluate the therapeutic effects of anti-oxidant therapies (glutathione, vitamins E and C, carnitines) against OxS damage in PAD [48,49,50,51].

1.2. Surrogate Biomarkers in PAD

OxS has been closely associated with cardiovascular diseases, oxidative biomarkers correlating with disease progression. OxS biomarkers have proven helpful in improving the early diagnosis of PAD, particularly in asymptomatic patients [52]. Currently, imaging techniques (US, CT and MRI) are the preferred diagnostic instrument for PAD and its monitoring [53,54,55]. The ankle–brachial index (ABI) measurement is widely agreed as an easy and repeatable method for PAD diagnosis as well as management [53]. Having completed the research on reliable biomarkers, it may be interesting to identify asymptomatic PAD patients or those at higher-risk to seek for PAD. Although the role of OxS biomarkers in clinical practice has been evaluated, it is still under discussion. We have summarized the findings on surrogate OxS biomarkers in PAD in the following sections.

1.2.1. Malondyaldheide (MDA)

Antioxidants are classified into two groups according to the mechanism by which they prevent or retard oxidation. The primary (chain-breaking) antioxidant group (e.g., c*-tocopherols) acts by interrupting oxidation and converting free radicals to stable species. The secondary antioxidant (e.g., ascorbic acids) group reacts with oxygen before the start of oxidation. The oxidation rate may be evaluated by generating hydroperoxides (per minute and per milligram of low density lipoprotein). Another surrogate biomarker of OxS is malondialdehyde concentration. MDA plasma levels differ between PAD and healthy controls as well as MDA levels being higher in PADs than in non-PAD individual before starting a treadmill test [56,57]. Maximal muscle stress enhances differences in MDA levels. However, anti-oxidant drug treatment (by propionil-L-carnitine i.v.) initiates reductions in the plasma levels of these oxidative makers both at rest and after treadmill tests. There is reasonable agreement on the helpful role of oxidative biomarkers in highlighting OxS in PAD, so OxS must be targeted in treating PAD patients [58,59,60].

1.2.2. 4-Hydroxynonenale (4-HNE)

4-HNE (4-Hydroxy-2-nonenal) is generated by the peroxidation of n-6 polyunsaturated fatty acid. HNE has a number of biological properties mainly represented by HNE-adduct generation on free amino groups and thiol groups on proteins. HNE adduct accumulation initiates cellular dysfunction, such tissue damage being a huge factor in atherosclerosis and arterial atherosclerotic diseases. HNE contributes to the atherogenicity of oxidized LDL by deviating the LDL metabolism and leading it to generate foam cells through the scavenger receptor pathway of macrophage cells. HNE de-regulates cellular responses at higher concentrations causing inflammatory responses and apoptosis. HNE is involved in re-modelling atherosclerotic plaque through the progressive modification of smooth muscle cells, also effecting proliferation, angiogenesis, and cell apoptosis. HNE adducts in the core of atherosclerotic plaque contribute to macrophage and smooth muscle cell apoptosis which in turn lead to a high risk of athero–thrombotic events. There is data on the accumulation of biogenic toxic aldehydes in critical ischemic tissues such as in ischemia-reperfusion injury. Notably, 4-hydroxy-2-nonenal (4-HNE) is a highly reactive aldehyde with abundant adduct formation which negatively effects contractility and the mitochondrial energy of muscle cells. High levels of 4-HNE modified proteins were found in PAD patient muscles such that the accumulation of the derived aldehyde product (4-HNE) is now considered as a marker for skeletal muscle injury initiated by OxS [61,62].

1.2.3. Micro RNAs (mRNAs)

While the peculiar role played by endothelial cells on correct function of circulation is clearly known, on the other hand, it is known that vascular smooth cells communicate with endothelial cells via mRNAs. Such mRNAs (i.e.,143 and 145) promote cell-to-cell interplay, finally favoring specific effects. Transport of delivered mRNAs to endothelial cells has been shown by using a high resolution imaging method [63].
mRNAs 143/145 play a key role for maintaining contractile phenotype of vascular smooth cells (VSMC) dysregulation of vascular tone and of arterial pressure were demonstrated in mRNAs-143/145−/−deficient mice. It is clear as the mRNAs show different capabilities both in contrasting or in maintaining functions of endothelial cells leading to equilibrium of vascular tone [64].
Capability of endothelial cells on circulatory equilibrium must achieve aiming against haemodynamic dysfunction and its unbalance is directly favorable to provoking arterial damage. Such mRNAs as mRNA-126 released by the endothelial is internalized by white cells (monocytes) and vascular smooth cells leading them to play a role to against endothelial dysfunction. There is a clear explanation of the crucial protective capability of mRNAs 126 from study on diabetes having highly prevalence of arterial consequences also linked to endothelial cell dysfunction [65,66,67,68]. Cell-to-cell talk between VSMC with endothelial cells promotes activation of mRNAs, its transcription thus mRNAs are transferred to endothelial cells. In patients with ischemic coronary diseases there is a raised circulating level of mRNA 126 but it is noteworthy to prevent the deleterious effects of endothelial cells [69]. mRNAs are emerging biomarkers for assessing oxidative stress. We have shown that miRNA values significantly increase in patients with PAD compared to controls [70]. These results confirm those of previous studies which show increased MiRNAs values in patients with progressive atherosclerosis compared to controls [71]. mRNAs may play a key role in the adaptive mechanism of maintaining cellular homeostasis during ischemic injury [68,69]. Their production is stimulated by Nrf2, a fundamental nucleic factor involved in intracellular redox balance [71,72].

1.2.4. Isoprostanes

Markers for the oxidation of LDL and eicosanoid formation (e.g., isoprostanes) were among the first to be analyzed. Isoprostanes derive from the metabolism of arachidonic acid induced by free-radical catalyzed processes [73]. Several previous studies have proven the reliability of 8-iso-prostaglandin F (a subtype of F2-isoprostanes, the most plentiful subclass) as a dependable method for evaluating oxidative stress in PAD patients [73,74]. The plasma values of F2-isoprostanes were higher in PAD patients than in controls. Our group reported a contradictory result on F2-isoprostane values in PAD patients which were lower compared to controls [74]. This outcome is associated with a significant increase in micro-RNAs suggesting an adaptive response to reduce oxidative stress.

1.2.5. Paraoxonase

Among the most significant biomarkers of oxidative stress with the role of antioxidants is Paraoxonase (PON): this family of enzymes protects lipoprotein from peroxidation [72].
Particularly in PAD patients compared to no PAD individuals were demonstrated lower paroxonase-1 (PON-1) concentration whereas PON-1 activity was raised. As we know, PON-1 activity is strongly dependent on the oxidation of lipoproteins, and in PAD patients there is a compensatory mechanism in favoring protective of PON family to against peroxidation [73]. PON is most abundant in the circulation, and may be considered as a novel marker of arteries status, and as a helpful marker of oxidative stress in PAD with multiple atherosclerotic diseases and in PAD patients [75,76,77,78].

1.2.6. Nrf2/Heme-oxygenase 1 (HO-1)

Transcriptional factor Nrf2 is one of the most important regulator molecules in the antioxidant system, appearing to protect against reperfusion injury. It promotes the transcriptional activation of several antioxidant (ARE) binding responses [30]. Nrf2 is also a fundamental regulator of heme oxygenase-1 (HO-1), an enzyme that promotes protection against oxidative stress [79] and also contributes to angiogenesis. It catalyzes the breakdown of heme to CO, ferrous iron and biliverdin [79]. Biliverdin is reduced to bilirubin which protects against the oxidation of lipids. The exact role of HO-1 in PADs remains to be investigated. It seems to protect endothelial cells by the effect of hypertension, oxidative stress and inflammation [80]. In particular, the increased production of CO intensifies its effects of vasodilatation and anti-inflammatory activity [81]. Values of HO-1 are lower in PAD patients, and it has been suggested as an independent predictor of PAD [81]. It is likely that lower HO-1 is part of a compensatory mechanism to maintain cellular redox balance, and HO-1 may be a reliable marker in the diagnosis and prognosis of PAD. Among the biochemical responses to OxS, the heme oxygenase (HO) system has been suggested as key [82]. It is known that HO-1 is an intracellular enzyme that catalyzes the breakdown of heme to carbon monoxide, ferrous iron and biliverdin [82,83,84]. Numerous effects of potential significance to the cardiovascular system have been reported, including protection against ischemia/reperfusion [79], blood pressure regulation [23,85,86,87], inflammation [80,81] and in angiogenesis [83]. HO-1 may be released into the plasma of smooth muscle cells, cardiomyocytes, leukocytes, monocytes/macrophages and/or endothelial cells that are damaged by the effect of hypertension, oxidative stress and/or chronic inflammation [23,82,83,84,85,86,87]. We demonstrated low HO-1 plasma in PAD patients but no relationship was found between HO-1 plasma levels with the progressive stages of PAD [79]. Interestingly, in regression analysis we found a close relationship between low levels of glutathione as a redox marker for the severity of PAD. Reduced HO-1 plasma levels demonstrated an impairment in the protective properties of HO-1 in PAD patients [74,79].

2. Conclusions

It is known that OxS plays a role in atherosclerotic processes as well as in PAD, thus it is reasonable that OxS must be targeted by using multiple approaches. High levels of OxS biomarkers were found in PAD patients with chronic ischemia causing intermittent claudication and in critical limb ischemia patients showing more severe ischemia of the peripheral arteries. OxS and pro-inflammatory conditions increased in PADs both of which are crucial in determining endothelial dysfunction. It is widely accepted that all the aforementioned conditions accelerate the atherosclerotic process. To date, endothelial dysfunction is considered an independent risk factor for morbidity and mortality in PAD patients. It is worth remembering that PAD is a predictive clinical marker for extended atherosclerotic processes in other arteries. From a therapeutic point of view, physical and pharmacological options are mandatory to reduce or counteract OxS and inflammation. On the other hand, to date we note the positive effect originated by no pharmacological strategy for PAD as the supervised physical training. It reduces OxS as well as the plasma levels of inflammatory biomarkers in parallel with improved walking distances measured by the treadmill test. These findings are very intriguing because improved muscle and physical performance may be crucial issues for PAD patients. It is clearly accepted that a well-structured rehabilitation program, may improve both performance, quality of life furthermore walking capability effects on the functional balance of patients with PAD [88,89,90,91,92]. However authors highly endorse the evaluation of the efficacy of muscle exercise in threatening PAD, suggesting awareness of potential damage to calf muscle fibers derived by strenuous physical exercise during treadmill training. A number of drugs have demonstrated their anti-oxidant capability in PAD patients in results from studies on their clinical efficacy in PAD. However, we still want to call for action more studies focusing on anti-oxidant and anti-inflammatory agents to achieve the aforementioned targets as well as to ameliorate walking distance, lower cardiovascular morbidity and mortality in PAD patients. A combined approach including anti-oxidant drugs, physical training, and re-vascularization may be effective against the multiple pathological modifications induced by PAD [92,93].

Author Contributions

Conceptualization, S.S.S.; supervision, S.S.S.; writing—review and editing, S.S.S., S.S., E.M., A.X. and A.G.

Funding

This study was funded by the 2016/2018 Research Plan of University of Catania, Department of Clinical and Experimental Medicine (project #A grant).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Fontaine, R.; Kim, M.; Kieny, R. Surgical treatment of peripheral circulation disorders. Helv. Chir. Acta 1954, 21, 499–533. [Google Scholar] [PubMed]
  2. Rutherford, R.B.; Baker, J.D.; Ernst, C.; Johnston, K.W.; Porter, J.M.; Ahn, S.; Jones, D.N. Recommended standards for reports dealing with lower extremity ischemia: revised version. J. Vasc. Surg. 1997, 26, 517–538. [Google Scholar] [CrossRef]
  3. Fowkes, F.G.; Rudan, D.; Rudan, I.; Denenberg, V.A.O.; McDermott, M.M.; Norman, P.E.; Sampson, U.K.A.; Williams, L.J.; Mensah, G.A.; Criqui, M.H. Comparison of global estimates of prevalence and risk factors for peripheral artery disease in 2000 and 2010: A systematic review and analysis. Lancet 2013, 382, 1329–1340. [Google Scholar] [CrossRef]
  4. Aronow, H.; Hiatt, W.R. The burden of peripheral artery disease and the role of antiplatelet therapy. Postgrad. Med. 2009, 121, 123–135. [Google Scholar] [CrossRef] [PubMed]
  5. Herrington, W.; Lacey, B.; Sherliker, P.; Armitage, J.; Lewington, S. Epidemiology of Atherosclerosis and the Potential to Reduce the Global Burden of Atherothrombotic Disease. Circ. Res. 2016, 118, 535–546. [Google Scholar] [CrossRef] [PubMed]
  6. Criqui, M.H.; Langer, R.D.; Fronek, A.; Feigelson, H.S. Coronary disease and stroke in patients with large-vessel peripheral arterial disease. Drugs 1991, 42 (Suppl. S5), 16–21. [Google Scholar] [CrossRef]
  7. Tuzcu, E.M.; Kapadia, S.R.; Tutar, E.; Ziada, K.M.; Hobbs, R.E.; McCarthy, P.M.; Young, J.B.; Nissen, S.E. High prevalence of coronary atherosclerosis in asymptomatic teenagers and young adults: Evidence from intravascular ultrasound. Circulation 2001, 103, 2705. [Google Scholar] [CrossRef] [PubMed]
  8. Katsiki, N.; Athyros, V.G.; Karagiannis, A.; Mikhailidis, D.P. Peripheral artery disease in patients with type 2 diabetes. J. Diabetes Complicat. 2014, 28, 912. [Google Scholar] [CrossRef]
  9. Lu, L.; Mackay, D.F.; Pell, J.P. Meta-analysis of the association between cigarette smoking and peripheral arterial disease. Heart 2014, 100, 414–423. [Google Scholar] [CrossRef]
  10. Brevetti, G.; Schiano, V.; Chiariello, M. Endothelial dysfunction: A key to the pathophysiology and natural history of peripheral arterial disease? Atherosclerosis 2008, 197, 1–11. [Google Scholar] [CrossRef]
  11. Ridker, P.M.; Stampfer, M.J.; Rifai, N. Novel risk factors for systemic atherosclerosis: a comparison of C-reactive protein, fibrinogen, homocysteine, lipoprotein (a) and standard cholesterol screening as predictors of peripheral arterial disease. JAMA 2001, 285, 2481–2485. [Google Scholar] [CrossRef] [PubMed]
  12. Shu, J.; Santulli, G. Update on peripheral artery disease: Epidemiology and evidence-based facts. Atherosclerosis 2018, 275, 379–381. [Google Scholar] [CrossRef] [PubMed]
  13. Libby, P.; Ridker, P.M.; Maseri, A. Inflammation and atherosclerosis. Circulation 2002, 105, 1135–1143. [Google Scholar] [CrossRef] [PubMed]
  14. Fowkes, F.G.; Housley, E.; Cawood, E.H.; Macintyre, C.C.; Ruckley, C.V.; Prescott, R.J. Edinburgh Artery Study: Prevalence of asymptomatic and symptomatic peripheral arterial disease in the general population. Int. J. Epidemiol. 1991, 20, 384–392. [Google Scholar] [CrossRef] [PubMed]
  15. Signorelli, S.; Anzaldi, M.; Fiore, V.; Catanzaro, S.; Simili, M.; Torrisi, B.; Neri, S. Study on unrecognized peripheral arterial disease (PAD) by ankle/brachial index and arterial co-morbidity in Catania (Sicily, Italy). Angiology 2010, 61, 524–529. [Google Scholar] [CrossRef] [PubMed]
  16. Norgern, L.; Hiatt, W.R.; Dormandy, J.A.; Nehler, M.R.; Harris, K.A.; Fowkes, F.G. TASC II Working Group: Inter-society consensus for management of peripheral arterial diseases (TASCII). J. Vasc. Surg. 2007, 45 (Suppl. S), S5–S67. [Google Scholar] [CrossRef]
  17. Ross, R. Atherosclerosis as an inflammatory disease. N. Engl. J. Med. 1999, 340, 115–126. [Google Scholar] [CrossRef] [PubMed]
  18. Wildman, R.P.; Muntner, P.; Chen, J.; Sutton-Tyrrell, K.; He, J. Relation of inflammation to peripheral arterial disease in the National Health and Nutrition Examination Survey, 1999–2002. Am. J. Cardiol. 2005, 96, 1579–1583. [Google Scholar] [CrossRef] [PubMed]
  19. Ridker, P.M.; Cushman, M.; Stampfer, M.J.; Tracy, R.P.; Hennekens, C.H. Plasma concentration of C-reactive protein and risk of developing peripheral vascular disease. Circulation 1998, 97, 425–428. [Google Scholar] [CrossRef]
  20. Ellulu, M.S.; Patimah, I.; Khaza’ai, H.; Rahmat, A.; Abed, Y.; Ali, F. Atherosclerotic cardiovascular disease: A review of initiators and protective factors. Inflammopharmacology 2016, 24, 1–10. [Google Scholar] [CrossRef]
  21. Husain, K.; Hernandez, W.; Ansari, R.A.; Ferder, L. Inflammation, oxidative stress and renin angiotensin system in atherosclerosis. World J. Biol. Chem. 2015, 6, 209–217. [Google Scholar] [CrossRef] [PubMed]
  22. Siti, H.N.; Kamisah, Y.; Kamsiah, J. The role of oxidative stress, antioxidants and vascular inflammation in cardiovascular disease (a review). Vascul. Pharmacol. 2015, 71, 40–56. [Google Scholar] [CrossRef] [PubMed]
  23. Barbagallo, I.; Galvano, F.; Frigiola, A.; Cappello, F.; Riccioni, G.; Murabito, P.; D’Orazio, N.; Torella, M.; Gazzolo, D.; Volti, G.L. Potential therapeutic effects of natural heme oxygenase-1 inducers in cardiovascular diseases. Antioxid. Redox Signal. 2013, 18, 507–521. [Google Scholar] [CrossRef] [PubMed]
  24. Novo, G.; Cappello, F.; Rizzo, M.; Fazio, G.; Zambuto, S.; Tortorici, E.; Gammazza, A.M.; Corrao, S.; Zummo, G.; de Macario, E.C.; et al. Hsp60 and heme oxygenase- 1 (Hsp32) in acute myocardial infarction. Transl. Res. 2011, 157, 285–292. [Google Scholar] [CrossRef] [PubMed]
  25. Brevetti, G.; Giugliano, G.; Brevetti, L.; Hiatt, W.R. Inflammation in peripheral artery disease. Circulation 2010, 122, 862–1875. [Google Scholar] [CrossRef] [PubMed]
  26. Nanson, E.M. Autoxidation in peripheral vascular disease. Lancet 1977, 1, 1057. [Google Scholar] [CrossRef]
  27. Cooke, J.P.; Wilson, A.M. Biomarkers of peripheral arterial disease. J. Am. Coll. Cardiol. 2010, 55, 2017–2023. [Google Scholar] [CrossRef] [PubMed]
  28. Mc Dermott, M.M.; Lloyd-Jones, S. The role of biomarkers and genetic in peripheral arterial disease. J. Am. Coll. Cardiol. 2009, 54, 1228–1237. [Google Scholar] [CrossRef] [PubMed]
  29. Hiatt, W.R.; Zakharyan, A.; Fung, E.T.; Crutcher, G.; Smith, A.; Stanford, C.; Cooke, J. A validated biomarker panel to identify peripheral artery disease. Vasc. Med. 2012, 17, 386–393. [Google Scholar] [CrossRef] [PubMed]
  30. Milne, G.L.; Dai, Q.; Roberts, L.J. The isoprostanes 25 years later. Biochim. Biophys. Acta 2015, 1851, 433–445. [Google Scholar] [CrossRef] [PubMed]
  31. Fort-Gallifa, I.; García-Heredia, A.; Hernández-Aguilera, A.; Simó, J.M.; Ju Sepúlveda, J.; Martín-Paredero, V.; Camps, J.; Joven, J. Biochemical indices of oxidative stress and inflammation in the evaluation of peripheral artery disease. Free Radic. Biol. Med. 2016, 97, 568–576. [Google Scholar] [CrossRef] [PubMed]
  32. Koutakis, P.; Weiss, J.; Miserlis, D.; Shostrom, V.K.; Papoutsi, E.; Ha, D.M.; Carpenter, L.A.; McComb, R.D.; Casale, G.P.; Pipinos, I.I. Oxidative damage in the gastrocnemius of patients with peripheral artery disease is myofiber type selective. Redox Biol. 2014, 2, 921–928. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Weiss, D.J.; Casale, G.P.; Koutakis, P.; Nella, A.A.; Swanson, S.A.; Zhu, Z.; Miserlis, D.; Johanning, J.M.; Pipinos, I.I. Oxidative damage and myofiber degeneration in the gastrocnemius of patients with peripheral arterial disease. J. Transl. Med. 2013, 11, 230. [Google Scholar] [CrossRef] [PubMed]
  34. Krishna, S.M.; Moxon, J.V.; Golledge, J. A review of the pathophysiology and potential biomarkers for peripheral artery disease. Int. J. Mol. Sci. 2015, 16, 11294–11322. [Google Scholar] [CrossRef]
  35. Boger, R.H.; Bode-Boger, S.M.; Thiele, W.; Junker, W.; Alexander, K.; Frolich, J.C. Biochemical evidence for impaired nitric oxide synthesis in patients with peripheral arterial disease. Circulation 1997, 95, 2068–2074. [Google Scholar] [CrossRef] [PubMed]
  36. McDermott, M.M. Lower extremity manifestations of peripheral arterial disease. The pathophysiologic and functional implications of leg ischemia. Circ. Res. 2015, 116, 1540–1550. [Google Scholar] [CrossRef] [PubMed]
  37. Pastori, D.; Carnevale, R.; Pignatelli, P. Is there a clinical role for oxidative stress biomarkers in atherosclerotic diseases? Intern. Emerg. Med. 2014, 9, 123–131. [Google Scholar] [CrossRef]
  38. Suarez Munguia, P.J. Release of nitric oxide induced by blood flow. Update and perspectives of research. Arch. Inst. Cardiol. Mex. 2000, 70, 197–202. [Google Scholar] [PubMed]
  39. Loffredo, L.; Carnevale, R.; Cangemi, R.; Angelico, F.; Augelletti, T.; di Santo, S.; Calabrese, C.M.; della Volpe, L.; Pignatelli, P.; Perri, L.; et al. NOX2 up-regulation is associated with artery dysfunction in patients with peripheral artery disease. Int. J. Cardiol. 2013, 165, 184–192. [Google Scholar] [CrossRef] [PubMed]
  40. Signorelli, S.; Neri, S.; Di Pino, L.; Costa, M.P.; Pennisi, G.; Digrandi, D.; Ierna, D. Oxidative stress and endothelial damage in patients with asymptomatic carotid atherosclerosis. Clin. Exp. Med. 2001, 1, 9–12. [Google Scholar] [CrossRef]
  41. Gliemann, L.; Nyberg, M.; Hellsten, Y. Nitric oxide and reactive oxygen species in limb vascular function: What is the effect of physical activity? Free Radic. Res. 2014, 48, 71–83. [Google Scholar] [CrossRef] [PubMed]
  42. Pipinos, I.I.; Judge, A.R.; Zhu, Z.; Selsby, J.T.; Swanson, S.A.; Johanning, J.M.; Baxter, B.T.; Lynch, T.G.; Dodd, S.L. Mitochondrial defects and oxidative damage in patients with peripheral arterial disease. Free Radic. Biol. Med. 2006, 41, 262–269. [Google Scholar] [CrossRef] [PubMed]
  43. Pessah-Rasmussen, H.; Seidegård, J.; Stavenow, L.; Solem, J.O.; Lindblad, B.; Xu, C.B. Glutathione transferase activity in human vessels and in cultured arterial smooth muscle cells. Int. Angiol. 1993, 12, 348–354. [Google Scholar] [PubMed]
  44. Edwards, A.T.; Blann, A.D.; Suarez-Mendez, V.J.; Lardi, A.M.; McCollum, C.N. Systemic responses in patients with intermittent claudication after treadmill exercise. Br. J. Surg. 1994, 81, 1738–1741. [Google Scholar] [CrossRef]
  45. Frei, B. Reactive oxygen species and antioxidant vitamins: Mechanisms of action. Am. J. Med. 1994, 97, S5–S13. [Google Scholar] [CrossRef]
  46. Langlois, M.D.; Duprez, J.; Delanghe, M.; De Buyzere Clement, D.L. Serum vitamin C concentration is low in peripheral arterial disease and is associated with inflammation and severity of atherosclerosis. Circulation 2001, 103, 1863–1868. [Google Scholar] [CrossRef]
  47. Judge, A.R.; Dodd, S.L. Oxidative damage to skeletal muscle following an acute bout of contractile claudication. Atherosclerosis 2003, 171, 219–224. [Google Scholar] [CrossRef]
  48. Spark, J.I.; Robinson, J.M.; Gallavin, L.; Gough, M.J.; Homer-Vanniasinkam, S.; Kester, R.C.; Scott, D.J.A. Patients with chronic critical limb ischaemia have reduced total antioxidant capacity and impaired nutritional status. Eur. J. Vasc. Endovasc. Surg. 2002, 24, 535–539. [Google Scholar] [Green Version]
  49. Braunwald, E.R.; Kloner, A. Myocardial reperfusion: A double-edged sword? J. Clin. Investig. 1985, 76, 1713–1719. [Google Scholar] [CrossRef]
  50. Koutakis, P.; Ismaeel, A.; Farmer, S.; Purcell, R.S.; Smith, J.L.; Eidson, W.T. Bohannon: Oxidative stress and antioxidant treatment in patients with peripheral artery disease. Physiol. Rep. 2018, 6, e1350. [Google Scholar] [CrossRef]
  51. Dopheide, J.F.; Doppler, C.; Scheer, M.; Obst, V.; Radmacher, M.-C.; Radsak, M.P.; Gori, T.; Warnholtz, A.; Fottner, C.; Münzel, T.; et al. Critical limb ischemia is characterized by an increased production of whole blood reactive oxygen species and expression of TREM-1 on neutrophils. Atherosclerosis 2013, 229, 396–403. [Google Scholar] [CrossRef] [PubMed]
  52. Williams, H.T.; Fenna, D.; Macbeth, R.A. Alpha tocopherol in the treatment of intermittent claudication. Surg. Gynecol. Obstet. 1971, 132, 662–666. [Google Scholar]
  53. Wijnen, M.H.; Coolen, S.A.; Vader, H.L.; Reijenga, J.C.; Huf, F.A.; Roumen, R.M. Antioxidants reduce oxidative stress in claudicants. J. Surg. Res. 2001, 96, 183–187. [Google Scholar] [CrossRef]
  54. Silvestro, A.; Scopacasa, F.; Oliva, G.; de Cristofaro, T.; Iuliano, L.; Brevetti, G. Vitamin C prevents endothelial dysfunction induced by acute exercise in patients with intermittent claudication. Atherosclerosis 2002, 165, 277–283. [Google Scholar] [CrossRef]
  55. Arosio, E.; de Marchi, S.; Zannoni, M.; Prior, M.; Lechi, A. Effect of glutathione infusion on leg arterial circulation, cutaneous microcirculation, and pain-free walking distance in patients with peripheral obstructive arterial disease: A randomized, double blind, placebo controlled trial. Mayo Clin. Proc. 2002, 77, 754–759. [Google Scholar] [CrossRef] [PubMed]
  56. Doobay, A.V.; Anand, S.S. Sensitivity and specificity of the ankle–brachial index to predict future cardiovascular outcomes: A systematic review. Arterioscler. Thromb. Vasc. Biol. 2005, 25, 1463–1469. [Google Scholar] [CrossRef] [PubMed]
  57. Holland-Letz, T.; Endres, H.G.; Biedermann, S.; Mahn, M.; Kunert, J.; Groh, S.; Pittrow, D.; von Bilderling, P.; Sternitzky, R.; Diehm, C. Reproducibility and reliability of the ankle-brachial index as assessed by vascular experts, family physicians and nurses. Vasc. Med. 2007, 12, 105–112. [Google Scholar] [CrossRef] [PubMed]
  58. Kalines, D.F.; Omary, R.A.; Dix, E.; Evans, A.J.; Hillman, B.J. Specificity of MR angiography as a confirmatory test of carotid artery stenosis. AJNR Am. J. Neuroradiol. 1996, 17, 1501–1506. [Google Scholar]
  59. Polak, J.F.; Bajakian, R.L.; O’Leary, D.H.; Anderson, M.R.; Donaldson, M.C.; Jolesz, F.A. Detection of internal carotid artery stenosis: Comparison of MR angiography, color Doppler sonography, and arteriography. Radiology 1992, 182, 35–40. [Google Scholar] [CrossRef]
  60. Cimminiello, C.; Kownator, S.; Wautrecht, J.C.; Carvounis, C.P.; Kranendonk, S.E.; Kindler, B.; Mangrella, M.; Borghi, C. PANDORA Study Investigators. The PANDORA study: peripheral arterial disease in patients with non-high cardiovascular risk. Intern. Emerg. Med. 2011, 6, 509–519. [Google Scholar] [CrossRef]
  61. Nègre-Salvayre, A.; Garoby-Salom, S.; Swiader, A.; Rouahi, M.; Pucelle, M.; Salvayre, R. Proatherogenic effects of 4-hydroxynonenal. Free Radic. Biol. Med. 2017, 111, 127–139. [Google Scholar] [CrossRef] [PubMed]
  62. Gomes, K.M.; Campos, J.C.; Bechara, L.R.; Queliconi, B.; Lima, V.M.; Disatnik, M.H.; Magno, P.; Chen, Ch.; Brum, P.C.; Kowaltowski, A.J.; et al. Aldehyde dehydrogenase 2 activation in heart failure restores mitochondrial function and improves ventricular function and remodeling. Cardiovasc. Res. 2014, 103, 498–508. [Google Scholar] [CrossRef] [PubMed]
  63. Santo, S.S.; Sergio, N.; Luigi, D.; Giuseppe, M.; Margherita, F.; Gea, O.C.; Roberto, F.; Gabriella, C.; Giuseppe, P.; Massimiliano, A. Effect of PLC on functional parameters and oxidative profile in type 2 diabetes-associated PAD. Diabetes Res. Clin. Pract. 2006, 72, 231–237. [Google Scholar] [CrossRef] [PubMed]
  64. Brass, E.P.; Hoppel, C.L. Relationship between acid-soluble carnitine and coenzyme A pools in vivo. Biochem. J. 1980, 190, 495–504. [Google Scholar] [CrossRef] [PubMed]
  65. Signorelli, S.S.; Fatuzzo, P.; Rapisarda, F.; Neri, S.; Ferrante, M.; Oliveri Conti, G.; Roberto Fallico, L.D.; et al. A randomised, controlled clinical trial evaluating changes in therapeutic efficacy and oxidative parameters after treatment with propionyl L-carnitine in patients with peripheral arterial disease requiring haemodialysis. Drugs Aging 2006, 23, 263–270. [Google Scholar] [CrossRef] [PubMed]
  66. Pennisi, G.; Marchese, G.; Costa, M.P.; Digrandi, D.; Celotta, G.; Virgilio, V. High circulating levels of cytokines (IL-6 and TNFalpha), adhesion molecules (VCAM-1 and ICAM-1) and selectins in patients with peripheral arterial disease at rest and after a treadmill test. Vasc. Med. 2003, 8, 15–19. [Google Scholar]
  67. Signorelli, S.S.; Katsiki, N. Oxidative Stress and Inflammation: Their Role in the Pathogenesis of Peripheral Artery Disease with or Without Type 2 Diabetes Mellitus. Curr. Vasc. Pharmacol. 2018, 16, 547–554. [Google Scholar] [CrossRef]
  68. Signorelli, S.S. Pathophysiology of the Peripheral Arterial Disease: Where Is the Paradox? Angiology 2018, 69, 183. [Google Scholar] [CrossRef]
  69. Climent, M.; Quintavalle, M.; Miragoli, M.; Chen, J.; Condorelli, G.; Elia, L. TGF beta triggers miR-143/145 transfer from smooth muscle cells to endothelial cells, thereby modulating vessel stabilization. Circ. Res. 2015, 116, 1753–1764. [Google Scholar] [CrossRef]
  70. Sala, F.; Aranda, J.F.; Rotllan, N.; Ramirez, C.M.; Aryal, B.; Elia, L. Condorelli, G.; Catapano, A.L.; Fernández-Hernando, C.; Norata, G.D. MiR-143/145 deficiency attenuates the progression of atherosclerosis in Ldlr−/−mice. Thromb. Haemost. 2014, 112, 796–802. [Google Scholar] [CrossRef]
  71. Wang, S.; Aurora, A.B.; Johnson, B.A.; Qi, X.; McAnally, J.; Hill, J.A.; Richardson, J.A.; Bassel-Duby, R.; Olson, E.N. The endothelial-specific microRNA miR-126 governs vascular integrity and angiogenesis. Dev. Cell 2008, 15, 261–271. [Google Scholar] [CrossRef] [PubMed]
  72. Tian, H.S.; Zhou, Q.G.; Shao, F. Relationship between arterial atheromatous plaque morphology and platelet-associated miR-126 and miR-223 expressions. Asian Pac. J. Trop. Med. 2015, 8, 309–314. [Google Scholar] [CrossRef]
  73. Leeper, N.J.; Raiesdana, A.; Kojima, Y.; Chun, H.J.; Azuma, J.; Maegdefessel, L.; Kundu, R.K.; Quertermous, T.; Tsao, P.S.; Spin, J.M. MicroRNA-126a is a novel regulator of vascular smooth muscle cell function. J. Cell. Physiol. 2011, 226, 1035–1043. [Google Scholar] [CrossRef] [PubMed]
  74. Prattichizzo, F.; Giuliani, A.; Ceka, A.; Rippo, M.R.; Bonfigli, A.R.; Testa, R.; Procopio, A.D.; Olivieri, F. Epigenetic mechanisms of endothelial dysfunction in type 2 diabetes. Clin. Epigenet. 2015, 7, 56. [Google Scholar] [CrossRef] [PubMed]
  75. Santulli, G. microRNAs Distinctively Regulate Vascular Smooth Muscle and Endothelial Cells: Functional Implications in Angiogenesis, Atherosclerosis, and In-Stent Restenosis. Adv. Exp. Med. Biol. 2015, 887, 53–77. [Google Scholar] [PubMed] [Green Version]
  76. Signorelli, S.S.; Li Volsi, G.; Pitruzzella, A.; Fiore, V.; Mangiafico, M.; Vanella, L.; Parenti, R.; Rizzo, M.; Volti, G.L. Circulating miR-130a, miR-27b, and miR-210 in patients with peripheral artery disease and their potential relationship with oxidative stress: A pilot study. Angiology 2016, 67, 945–950. [Google Scholar] [CrossRef] [PubMed]
  77. Santulli, G.; Wronska, A.; Uryu, K.; Diacovo, T.G.; Gao, M.; Marx, S.O.; Kitajewski, J.; Chilton, J.M.; Akat, K.M.; Tuschl, T.; et al. A selective microRNA-based strategy inhibits restenosis while preserving endothelial function. J. Clin. Investig. 2014, 124, 4102–4147. [Google Scholar] [CrossRef] [PubMed]
  78. Li, T.; Cao, H.; Zhuang, J.; Wan, J.; Guan, M.; Yu, B.; Li, X.; Zhang, W. Identification of miR-130a, miR-27b and miR-210 as serum biomarkers for atherosclerosis obliterans. Clin. Chim. Acta 2011, 412, 66–70. [Google Scholar] [CrossRef] [PubMed]
  79. Signorelli, S.S.; Li Volsi, G.; Fiore, V.; Mangiafico, M.; IParenti, R.B.; Rizzo, M.; Volti, G.L. Plasma heme oxygenase-1 is decreased in peripheral artery disease patients. Mol. Med. Rep. 2016, 14, 3459–3463. [Google Scholar] [CrossRef] [PubMed]
  80. Camps, J.; Marsillach, J.; Joven, J. The paraoxonases: Role in human diseases and methodological difficulties in measurement. Crit. Rev. Clin. Sci. 2009, 46, 83–106. [Google Scholar] [CrossRef] [PubMed]
  81. Pasqualini, C.; Cortese, S.; Marchesi, D.; Siepi, M.; Pirro, G.; Liberatoscioli, L.; Gnasso, A.; Schillaci, G.; Mannarino, E. Paraoxonase-1activity modulates endothelial function in patients with peripheral arterial disease. Atherosclerosis 2005, 83, 349–354. [Google Scholar] [CrossRef] [PubMed]
  82. Loffredo, L.; Pignatelli, P.; Cangemi, R.; Andreozzi, P.; Panico, M.A.; Meloni, V.; Violi, F. Imbalance between nitricoxide generation ando xidative stress in patients with peripheral arterial isease: Effect of anantioxidan ttreatment. J. Vasc. Surg. 2006, 44, 525–530. [Google Scholar] [CrossRef] [PubMed]
  83. Hernández-Aguilera, A.; Sepúlveda, J.; Rodríguez-Gallego, E.; Guirro, M.; García-Heredia, A.; Cabré, N.; Luciano-Mateo, F.; Fort-Gallifa, I.; Martín-Paredero, V.; Joven, J.; et al. Immuno histochemical analysis of paraoxonase sand chemokines in arteries of patients with peripheral artery disease. Int. J. Mol. Sci. 2015, 16, 11323–11338. [Google Scholar] [CrossRef] [PubMed]
  84. Mikhed, Y.; Gorlach, A.; Knaus, U.G.; Daiber, A. Redox regulation of genome stability by effects on gene expression, epigenetic pathways and DNA damage/repair. Redox Biol. 2015, 5, 275–289. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Li Volti, G.; Sorrenti, V.; Murabito, P.; Galvano, F.; Veroux, M.; Gullo, A.; Acquaviva, R.; Stacchiotti, A.; Bonomini, F.; Vanella, L.; et al. Pharmacological induction of heme oxygenase-1 inhibits iNOS and oxidative stress in renal ischemia-reperfusion injury. Transplant. Proc. 2007, 39, 2986–2991. [Google Scholar] [CrossRef] [PubMed]
  86. Li Volti, G.; Sacerdoti, D.; Sangras, B.; Vanella, A.; Mezentsev, A.; Scapagnini, G.; Falck, J.R.; Abraham, N.G. Carbon monoxide signaling in promoting angiogenesis in human microvessel endo thelial cells. Antioxid. Redox Signal. 2005, 7, 704–710. [Google Scholar] [CrossRef]
  87. Barbagallo, I.; Nicolosi, A.; Calabrese, G.; David, S.; Cimino, S.; Madonia, M.; Cappello, F. The role of the heme oxygenase system in the metabolic syndrome. Curr. Pharm. Des. 2014, 20, 4970–4974. [Google Scholar] [CrossRef] [PubMed]
  88. Van den Houten, M.M.L.; Hageman, D.; Gommans, L.N.M.; Kleijnen, J.; Scheltinga, M.R.M.; Teijink, J.A.W. The Effect of supervised exercise, home based exercise and endovascular revascularization on physical activity in patients with intermittent claudication: A network meta-analysis. Eur. J. Vasc. Endovasc. Surg. 2019, 1078, 30923–30927. [Google Scholar]
  89. Gardner, A.W.; Parker, D.E.; Montgomery, P.S. Changes in vascular and inflammatory biomarkers after exercise rehabilitation in patients with symptomatic peripheral artery disease. J. Vasc. Surg. 2019, in press. [Google Scholar] [CrossRef]
  90. Ismaeel, A.; Brumberg, R.S.; Kirk, J.S.; Papoutsi, E.; Farmer, P.J.; Bohannon, W.T.; Smith, R.S.; Eidson, J.L.; Sawicki, I.; Koutakis, P. Oxidative Stress and Arterial Dysfunction in Peripheral Artery Disease. Antioxidants (Basel) 2018, 19, 7. [Google Scholar] [CrossRef]
  91. Sacerdoti, D.; Colombrita, C.; Ghattas, M.H.; Ismaeil, E.F.; Scapagnini, G.; Bolognesi, M.; Li Volti, G.; Abraham, N.G. Heme oxygenase-1 transduction in endothelial cells causes downregulation of monocyte chemoattractant protein-1 and of genes involved in inflammation and growth. Cell. Mol. Biol. 2005, 51, 363–370. [Google Scholar] [PubMed]
  92. Brass, E.P. Intermittent claudication: New targets for drug development. Drugs 2013, 73, 999–1014. [Google Scholar] [CrossRef] [PubMed]
  93. Stevens, S.; Daibe, A.; Dopheide, J.F.; Thomas Münze, T.; Espinola-Klein, C. Peripheral artery disease, redox signaling, oxidative stress—Basic and clinical aspects. Redox Biol. 2017, 12, 787–797. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Macrocirculatory and microcirculatory mechanisms in compromising walking capability in peripheral chronic ischemia.
Figure 1. Macrocirculatory and microcirculatory mechanisms in compromising walking capability in peripheral chronic ischemia.
Antioxidants 08 00367 g001
Figure 2. Pathophysiology of peripheral arterial disease.
Figure 2. Pathophysiology of peripheral arterial disease.
Antioxidants 08 00367 g002
Table 1. Progression and staging of peripheral arterial disease (PAD) by based on two most applied classifications.
Table 1. Progression and staging of peripheral arterial disease (PAD) by based on two most applied classifications.
Fontaine’s Classification [1].Rutherford’s Classification [2].
StageClinical SymptomsPathophysiologyClinicalGrade
1stno symptomsoccasional
discovery of
aortic and iliac
calcifications
ats plaque
risk plaque
inflammation
asymptomatic0/0
2nd Aclaudicationabsolute
claudication
distance > 200 mt
recovery t. < 2 min
discrepancy
oxygen request
arterial supply
mild
claudication
moderate
claudication
I/1
I/2
2nd BclaudicationACD < 200 m
recovery time > 2 min
ACD < 100 m
recovery time > 2 min
discrepancy
oxygen request
arterial supply
Highest
discrepancy
and acidosis
severe
claudication
I/3
3rdischaemic
rest pain
ischaemic
rest pain
skin hypoxia
acidosis
ischaemic
rest pain
II/4
4thulceration
or gangrene
Skinn necrosis
gangrene
severe skin
hypoxia
acidosis
minor
tissue
loss
major
tissue loss
III/5
III/6

Share and Cite

MDPI and ACS Style

Signorelli, S.S.; Scuto, S.; Marino, E.; Xourafa, A.; Gaudio, A. Oxidative Stress in Peripheral Arterial Disease (PAD) Mechanism and Biomarkers. Antioxidants 2019, 8, 367. https://doi.org/10.3390/antiox8090367

AMA Style

Signorelli SS, Scuto S, Marino E, Xourafa A, Gaudio A. Oxidative Stress in Peripheral Arterial Disease (PAD) Mechanism and Biomarkers. Antioxidants. 2019; 8(9):367. https://doi.org/10.3390/antiox8090367

Chicago/Turabian Style

Signorelli, Salvatore Santo, Salvatore Scuto, Elisa Marino, Anastasia Xourafa, and Agostino Gaudio. 2019. "Oxidative Stress in Peripheral Arterial Disease (PAD) Mechanism and Biomarkers" Antioxidants 8, no. 9: 367. https://doi.org/10.3390/antiox8090367

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop