Hostname: page-component-8448b6f56d-wq2xx Total loading time: 0 Render date: 2024-04-23T09:39:20.908Z Has data issue: false hasContentIssue false

New perspectives on bioactivity of olive oil: evidence from animal models, human interventions and the use of urinary proteomic biomarkers

Published online by Cambridge University Press:  17 July 2015

S. Silva
Affiliation:
iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal ITQB, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal Pharmacy Faculty, University of Lisbon, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
E. Combet
Affiliation:
Human Nutrition, School of Medicine, University of Glasgow, Glasgow G31 2ER, UK
M. E. Figueira
Affiliation:
Pharmacy Faculty, University of Lisbon, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal iMED.Ulisboa, Research Institute for Medicines, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
T. Koeck
Affiliation:
Mosaiques diagnostics GmbH, Mellendorfer Strabe 7-9, 30625 Hannover, Germany
W. Mullen
Affiliation:
Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow G12 8QQ, UK
M. R. Bronze*
Affiliation:
iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal ITQB, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal Pharmacy Faculty, University of Lisbon, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal iMED.Ulisboa, Research Institute for Medicines, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
*
*Corresponding author: M. R. Bronze, fax (+351) 217946470, email mrbronze@ff.ulisboa.pt
Rights & Permissions [Opens in a new window]

Abstract

Olive oil (OO) is the primary source of fat in the Mediterranean diet and has been associated with longevity and a lower incidence of chronic diseases, particularly CHD. Cardioprotective effects of OO consumption have been widely related with improved lipoprotein profile, endothelial function and inflammation, linked to health claims of oleic acid and phenolic content of OO. With CVD being a leading cause of death worldwide, a review of the potential mechanisms underpinning the impact of OO in the prevention of disease is warranted. The current body of evidence relies on mechanistic studies involving animal and cell-based models, epidemiological studies of OO intake and risk factor, small- and large-scale human interventions, and the emerging use of novel biomarker techniques associated with disease risk. Although model systems are important for mechanistic research nutrition, methodologies and experimental designs with strong translational value are still lacking. The present review critically appraises the available evidence to date, with particular focus on emerging novel biomarkers for disease risk assessment. New perspectives on OO research are outlined, especially those with scope to clarify key mechanisms by which OO consumption exerts health benefits. The use of urinary proteomic biomarkers, as highly specific disease biomarkers, is highlighted towards a higher translational approach involving OO in nutritional recommendations.

Type
Conference on ‘Carbohydrates in health: friends or foes’
Copyright
Copyright © The Authors 2015 

Relevance of the Mediterranean diet and olive oil to health

The olive tree, Olea europaea L., is one of the oldest agricultural tree crops and provides diversified products for human consumption such as table olives and olive oil (OO)( Reference Obied, Prenzler and Ryan 1 ). The analytical parameters to ascertain OO quality and classify OO are defined by European Union regulations( 2 ). Oils obtained only by mechanical extraction are virgin olive oils (VOO) and further quality assessment can lead to a classification as extra virgin olive oil (EVOO)( 3 ).

OO is the primary source of fat in the Mediterranean diet and has been associated with longevity and a lower incidence of chronic diseases, particularly CHD( Reference Tripoli, Giammanco and Tabacchi 4 Reference Urpi-Sarda, Casas and Chiva-Blanch 7 ). OO consumption is also associated with decreased rates of cancer, diabetes and neurodegenerative diseases( Reference Martinez-Lapiscina, Clavero and Toledo 8 ) as well as body weight reduction and obesity prevention( Reference Kastorini, Milionis and Goudevenos 9 , Reference Razquin, Martinez and Martinez-Gonzalez 10 ). The epidemiological evidence underpinning the relevance of the Mediterranean diet to health is strong with over seventeen studies including 2300 volunteers confirming that a Mediterranean diet decreases inflammation and improves endothelial function( Reference Schwingshackl and Hoffmann 11 ), and a meta-analysis of thirty-two cohort studies (>800 000 subjects) indicating that there is an inverse correlation between OO intake and CHD( Reference Schwingshackl and Hoffmann 12 ).

Olive oil bioactive components

The major components of OO are glycerols (saponifiable fraction) which represent more than 98 % of the total oil weight and are mainly TAG esters of oleic acid (55–83 %), palmitic acid (7·5–20 %), linoleic acid (3·5–21 %) and other fatty acids such as stearic acid (0·5–5 %)( Reference Harwood and Aparico 13 ). Minor components (the unsaponifiable fraction) include aliphatic and triterpenic alcohols, sterols, hydrocarbons as squalene, volatile compounds, tocopherols, carotenes, chlorophyll and phenolic compounds( Reference Harwood and Aparico 13 Reference Perez-Jimenez, Ruano and Perez-Martinez 15 ).

Special attention has been given to the phenolic compounds only found in VOO and EVOO. The agronomic and technological aspects of OO production have an impact on the concentration of phenolic compounds, as do the pedoclimatic conditions and agronomic techniques (e.g. irrigation)( Reference Tripoli, Giammanco and Tabacchi 4 , Reference Servili and Montedoro 14 ). The main classes of phenolic compounds present in VOO are phenolic acids, phenolic alcohols (hydroxytyrosol and tyrosol), flavonoids, lignans and secoiridoids; Table 1.

Table 1. Main classes of phenolic compounds in virgin olive oil

Oleuropein and ligstroside, the most significant secoiridoids in O. europaea L., are esters of elenolic acid glucoside with hydroxytyrosol and tyrosol, respectively. During the mechanical extraction of the oil, fruit endogenous β-glucosidases( Reference Servili and Montedoro 14 , Reference Di Maio, Esposto and Taticchi 16 ) are released leading to the secoiridoid aglycones formation, accounting for more than 50 % of phenolic content of the oil( Reference Brenes, Garcia and Garcia 17 , Reference Hrncirik and Fritsche 18 ). The most abundant secoiridoids of VOO are the oleuropein and ligstroside aglycons and dialdehydic forms of deacetoxy of oleuropein and ligstroside aglycons( Reference Servili and Montedoro 14 ) also named oleacein and oleocanthal, respectively( Reference Karkoula, Skantzari and Melliou 19 ).

Phenolic compounds bioavailability and bioactivity

Once OO has been ingested, it produces a micellar solution composed of a lipid and an aqueous phase. Chemical hydrolysis of secoiridoids can take place in the acidic medium of the stomach( Reference Corona, Tzounis and Assunta Dessi 20 ) or in alkaline conditions in the small intestine( Reference Pinto, Paiva-Martins and Corona 21 , Reference Aranzazu Soler, Alba and Shikha 22 ) leading to an increase of free phenolic alcohols released into the aqueous phase. As a result OO phenolic compounds are further absorbed in the small intestine( Reference Vissers, Zock and Roodenburg 23 ). Measuring the bioavailability of these compounds in plasma and urine reveals that OO phenolics undergo a conjugation process of methylation, glucuronidation and sulfation indicating that there is phase 2 metabolism involved during the absorption of these compounds( Reference Garcia-Villalba, Carrasco-Pancorbo and Nevedomskaya 24 Reference Miro Casas, Farre Albadalejo and Covas Planells 27 ). The between-subjects variability in human absorption and metabolism of OO phenolics may explain differences in proportion of methyl, glucuronide and sulphate conjugates reported( Reference Garcia-Villalba, Larrosa and Possemiers 28 Reference Preedy and Watson 30 ).

Bioavailability of OO phenolic compounds differs according to the intake matrix. OO as the intake vehicle promotes absorption of hydroxytyrosol: the corresponding bioavailability of hydroxytyrosol in rats for aqueous and OO solutions were reported as 75 and 99 %( Reference Tuck, Freeman and Hayball 31 ), respectively. When a supplement containing hydroxytyrosol as a single oral dose (2·5 mg/kg) was fed to human subjets, the bioavailability was below 10 %( Reference Gonzalez-Santiago, Fonolla and Lopez-Huertas 32 ), whereas previous studies showed higher bioavailability for hydroxytyrosol supplementation in lipid vehicles( Reference Visioli, Galli and Grande 33 ). The addition of hydroxytyrosol to low fat yoghurt and administered to human subjects was also associated with a lower excretion of hydroxytyrosol when compared with OO( Reference Visioli, Galli and Grande 33 ). As OO phenolic compounds are mainly absorbed in the small intestine( Reference Vissers, Zock and Roodenburg 23 ) the increase of hydroxytyrosol bioavailability, in OO, might be related to the rate of gastric emptying( Reference Gonzalez-Santiago, Fonolla and Lopez-Huertas 32 ) and slow release of hydroxytyrosol from the oil matrix( Reference Miro-Casas, Covas and Farre 26 , Reference Gonzalez-Santiago, Fonolla and Lopez-Huertas 32 ). The presence of other antioxidants in OO might prevent breakdown of hydroxytyrosol before absorption in the gastrointestinal tract( Reference Tuck, Freeman and Hayball 31 ).

Secoiridoids that are not absorbed in the small intestine are degraded by the colonic microbiota with oleuropein producing hydroxytyrosol as the major product( Reference Corona, Tzounis and Assunta Dessi 20 ). In vitro colonic metabolism was evaluated on tyrosol, hydroxytyrosol, hydroxytyrosol acetate and oleuropein showing an increase in phenolic acids, stability of hydroxytyrosol and tyrosol and degradation of hydroxytyrosol acetate and oleuropein mainly to hydroxytyrosol( Reference Mosele, Martin-Pelaez and Macia 34 ). To evaluate OO phenolic metabolites produced from colonic fermentation, faecal samples were analysed before and after mid-term consumption of phenol-rich OO( Reference Mosele, Martin-Pelaez and Macia 34 ). A significant increase in hydroxytyrosol concentration (P < 0·05) was observed after phenol-rich OO intake. Although absorption of OO phenolic compounds mainly occurs in the small intestine a small proportion of hydroxytyrosol and its derivatives still pass into the large intestine( Reference Vissers, Zock and Roodenburg 23 ). This highlights the need to study the impact of OO phenolics in the colon, either with gut microbiota interaction or local activity due to its antioxidant and anti-inflammatory properties.

When assessing the chemical and in vitro biological antioxidant activities of these compounds, it is the glucuronide conjugates of hydroxytyrosol and tyrosol that must be assessed. These were tested in the range 0·01–10 μm against the radical 1,1-diphenyl-2-picrylhydrazyl. None of the glucuronides displayed significant antioxidant activities at the concentrations tested, whereas the parent aglycones did display antioxidant activity at these concentrations( Reference Khymenets, Fito and Tourino 35 ). This conflicts with the results of others( Reference Tuck, Hayball and Stupans 36 ) with differences attributed to the fact that in one study reference standard material( Reference Khymenets, Fito and Tourino 35 ) was used and in the other the glucuronide conjugates were extracted from urine samples( Reference Tuck, Hayball and Stupans 36 ), and likely contained impurities that had antioxidant activity. Hydroxytyrosol metabolites might act as ‘sinks’ of hydroxytyrosol that could be locally released in the cells after enzymatic hydrolysis( Reference Kotronoulas, Pizarro and Serra 37 ), thereby explaining the proposed hydroxytyrosol biological effects observed in vivo. Moreover, in situ deconjugation of hydroxytyrosol metabolites (into their free form) in erythrocytes was observed in rats after oral administration of an OO phenolic extract obtained from olive cake (1·5 g/kg body weight, equivalent to 34·4 mg hydroxytyrosol and derivatives), highlighting a potential protective mechanism against cell oxidative damage( Reference Laura Rubió, Alba and Carme 38 ).

Although there are a number of biological effects for OO phenolic compounds, most cannot be achieved via normal dietary exposure to OO. This has led to development of enriched products with natural OO phenolic compounds. OO by-products such as olive mill wastewater( Reference Hamden, Allouche and Damak 39 ) and olive pomace( Reference Sanchez de Medina, Priego-Capote and Luque de Castro 40 , Reference Suarez, Romero and Motilva 41 ) are potential sources of natural bioactives which could be used to supplement OO. The development of new OO products such as pomace OO or refined olive oil enriched in natural bioactives opens new perspectives in the field.

Olive oil and inflammation

Inflammation involves a complex cascade of events partly related with the production of an excess of free radicals due to internal or environmental stress( Reference Pashkow 42 ). The inflammation process triggers signalling molecules such as NF-κB, which upregulates the production of inflammatory mediators, such as TNF-α( Reference Vlantis and Pasparakis 43 ), inducible NO synthase, cyclooxygenase (COX)-2 and IL-1β( Reference Pashkow 42 ).

A number of phenolic compounds present in OO have anti-inflammatory properties, including oleocanthal, a secoiridoid (dose-dependent inhibition of COX-1 and -2 activities, similar to the anti-inflammatory drug ibuprofen( Reference Beauchamp, Keast and Morel 44 )). However, to achieve comparable effect with the recommended daily dose of ibuprofen, 500 g EVOO would need to be consumed( Reference Beauchamp, Keast and Morel 45 , Reference Tulp, Bruhn and Bohlin 46 ) making the dose–effect relationship out with any (acute) inflammatory benefits due to typical OO consumption.

Chronic inflammation

Rheumatoid arthritis is a major inflammatory, autoimmune, disease characterised by chronic joint inflammation( Reference O'Connor 47 , Reference Waterman and Lockwood 48 ). Hydroxytyrosol has been studied for its anti-inflammatory effects in a rheumatoid arthritis animal model. We reported that it provided beneficial effects in the evolution of the disease( Reference Silva, Sepodes and Rocha 49 ), with 0·5 and 5 mg/kg doses in rats, after gavage administration, using refined olive oil as vehicle (human-equivalent of 4·9 and 49 mg/d, respectively, for a 60 kg adult), Fig. 1. Significant effects, on paw oedema reduction, were observed for a human-equivalent dose of 49 mg/d, a dose ten times higher than the approved European Food Safety Authority (EFSA) dose for phenolic compounds in relation to protection of lipid oxidation( 50 ). The same hydroxytyrosol dose was effective on colitis, another chronic inflammatory disease( Reference Sanchez-Fidalgo, Sanchez de Ibarguen and Cardeno 51 ). This dose would only be achievable through nutraceutical supplementation of OO with hydroxytyrosol, and the use of this functional food on a daily basis.

Fig. 1. Chronic inflammation model and impact on rats paw oedema. ANOVA, *P < 0·001 v. positive control – Rheumatoid Arthritis, + P < 0·01 v. Refined Olive Oil; OHTYR, hydroxytyrosol. Adapted from Silva et al. ( Reference Silva, Sepodes and Rocha 49 ).

To further evaluate the anti-inflammatory mechanisms involved with hydroxytyrosol, we studied COX-2 and inducible NO synthase expression( Reference Silva, Sepodes and Rocha 49 ). The treatment at 5 mg/kg dose significantly decreased histological damage, COX-2 and inducible NO synthase expression (P < 0·001 v. positive control), markedly reduced the degree of bone resorption, soft tissue swelling and osteophyte formation, improving articular function in the treated animals. Moreover, at the same dose there was a significant decrease (P < 0·005 v. positive control and refined olive oil) in TNF-α serum levels. These results are in line with others that reported benefits on rheumatoid arthritis, in animal models, after oral administration of an EVOO extract( Reference Rosillo, Alcaraz and Sanchez-Hidalgo 52 ), intraperitoneal administration of oleuropein aglycone( Reference Impellizzeri, Esposito and Mazzon 53 ) or polyphenol supplemented VOO diets( Reference Martinez-Dominguez, de la Puerta and Ruiz-Gutierrez 54 ). The reports highlight the effects on rheumatoid arthritis of OO phenolic compounds either administered as isolated compounds or as an extract. However, dose comparison between animal studies have to take in consideration not only differences in species (rats v. mice), but also routes of administration. Compared with intraperitoneal administration, an oral dose has an extra pass through the liver with consequent metabolism through the first-pass effect.

Acute inflammation

Acute inflammation has been commonly induced using carrageenan in animals to evaluate the effects of non-steroid anti-inflammatory drugs( Reference Bignotto, Rocha and Sepodes 55 ). We studied the effect of hydroxytyrosol-supplemented OO on acute inflammation, induced by carrageenan in rats, at 0·5 and 5 mg/kg( Reference Silva, Sepodes and Rocha 49 ) dose, after gavage administration which occurred 30 min before the challenge with carrageenan. Both doses significantly reduced paw oedema (P < 0·001 v. positive control) with the lowest effective dose being achievable through OO daily intake. Previous studies in rats( Reference Gong, Geng and Jiang 56 ) also showed inhibition of carrageenan; acute inflammation of an aqueous hydroxytyrosol formulation (HT-20, 22 % hydroxytyrosol), and significant effects were obtained at a 22 mg/kg hydroxytyrosol dose. Differences in dose effect might be related to the administration vehicle with refined olive oil or OO being better vehicles than water.

Cardioprotection of olive oil

Most of the interventional studies focusing on the benefit of VOO intake on CVD have investigated the effect of phenolic compounds on the prevention of oxidation of LDL and HDL( Reference Gimeno, de la Torre-Carbot and Lamuela-Raventos 57 Reference Katan, Zock and Mensink 64 ), two risk markers of CVD. A number of trials have also focused on cardioprotection against inflammation( Reference Chrysohoou, Panagiotakos and Pitsavos 65 ) mainly on antioxidant activity and inflammatory mediators.

Impact of olive oil constituents on lipoproteins and atherosclerosis

Fat content

LDL particles carry about two-thirds of plasma cholesterol and can infiltrate the arterial wall attracting macrophages, smooth muscle cells and endothelial cells( Reference Huang and Sumpio 66 ) thus driving atherosclerosis.

LDL particle size is influenced by type and amount of dietary fat consumed( Reference Covas, Konstantinidou and Fito 67 ): Low-fat diets lead to a decrease in the size of LDL particles compared with high-fat diets( Reference Krauss and Dreon 68 ). The type of fat ingested is also important: LDL particles are larger with high-MUFA diets (such as those based on OO), compared with diets with a high PUFA intake, where LDL particles are smaller( Reference Bos, Poortvliet and Scheffer 69 ). LDL particle size is especially relevant, since small-size particle are more prone to oxidation and can better enter into the arterial wall when compared with larger LDL particles( Reference Chait, Brazg and Tribble 70 ). Conversely, HDL particles are antiatherogenic, as their primarily role is to deliver cholesterol to the liver to be metabolised and excreted or reused. HDL may also be able to dislodge cholesterol molecules from atheromas in arterial walls( Reference Huang and Sumpio 66 ). It has been reported in patients with peripheral vascular disease( Reference Aguilera, Mesa and Ramirez-Tortosa 71 , Reference Kratz, Cullen and Kannenberg 72 ) that LDL particles are less susceptible to oxidation when the diet is enriched in VOO MUFA, compared with the PUFA of sunflower-oil enriched diets. Moreover, when compared with SFA intake, OO oleic acid reduces the level of LDL-cholesterol( Reference Ruiz-Canela and Martinez-Gonzalez 63 , Reference Katan, Zock and Mensink 64 ).

The health benefits associated with monounsaturated fat content in OO were recognised by the United States Food and Drug Administration in 2004, highlighting ‘the benefits on the risk of CHD of eating about two tablespoons (23 g) of OO daily’( 73 ). Health benefits were related with a decrease of total and LDL-cholesterol in serum( 73 ), diet improvement of endothelial dysfunction( Reference Fuentes, Lopez-Miranda and Perez-Martinez 74 ), coagulation activity( Reference Capurso, Massaro and Scoditti 75 ) and reduced LDL susceptibility to oxidation( Reference Kratz, Cullen and Kannenberg 72 ).

Phenolic content

Antioxidants that can prevent lipid peroxidation, such as phenolic compounds, could play an important role in preventing oxidative modification of LDL( Reference Tripoli, Giammanco and Tabacchi 4 ), with the oxidative process an initiating factor for atherosclerotic plaques( Reference Khurana, Venkataraman and Hollingsworth 76 ). Once monocytes differentiate in macrophages on the endothelium they scavenge oxidised LDL, then becoming foam cells, leading to plaque formation( Reference Covas 5 ).

The Effect of Olive Oil on Oxidative Damage in European Populations study was a cross-over fat replacement intervention( Reference Covas, Nyyssonen and Poulsen 58 ), using OO with different phenolic content in healthy male volunteers. Its findings led to the current EFSA recommendation (Opinion of the Scientific Committee/Scientific Panel, EFSA Journal ( 50 , 77 , 78 )). A linear increase in HDL-cholesterol levels after 3 weeks was observed after low-, medium- and high-polyphenol OO consumption: mean change from preintervention, 0·02 (95 % CI 0·00, 0·05) mm/l, 0·03 (95 % CI 0·00, 0·05) mm/l, and 0·04 (95 % CI 0·02, 0·06) mm/l, respectively. Total cholesterol:HDL-cholesterol ratio decreased linearly with the phenolic content of the OO. TAG levels decreased by an average of 0·05 mm/l for all OO( Reference Covas, Nyyssonen and Poulsen 58 ). Mean changes from preintervention for oxidised LDL levels (where U represents arbitrary units) were 1·21 (95 % CI −0·8, 3·6) U/l, −1·48 (95 % CI −3·6, 0·6) U/l and −3·21 (95 % CI −5·1, −0·8) U/l for the low-, medium- and high-polyphenol OO, respectively, showing a dose-dependent relation with VOO phenolic content( Reference Covas, Nyyssonen and Poulsen 58 ). The EFSA confirmed a cause–effect relationship between consumption of OO phenolics (standardised by the content of hydroxytyrosol and its derivatives) and protection of LDL-cholesterol particles against oxidative damage. To support the EFSA health claim, 5 mg hydroxytyrosol and its derivatives should be consumed daily in 20 g OO( 50 ), but concentrations in some OO may be too low to achieve this target in the context of a balanced diet. Moreover, the EFSA Panel reported study design limitations as most human interventions with OO have been conducted in more homogeneous male populations( 77 ) and not in general population.

The contribution of OO phenolics towards cardiovascular health benefits has been challenged with inconsistent results reported for ex vivo resistance of LDL to oxidation( Reference Vissers, Zock and Katan 79 , Reference Rietjens, Bast and Haenen 80 ). Seven human intervention studies with OO were compared for impact of phenolics on oxidised LDL, with no effect seen in five of them( Reference Vissers, Zock and Katan 79 ), possibly explained by artefacts generated during LDL isolation.

Since the approval of the EFSA claim, both terminology and analytical methodology supporting the dose calculation of hydroxytyrosol and derivatives have been appraised. Mastralexi et al. ( Reference Mastralexi, Nenadis and Tsimidou 81 ) commented on the weaknesses of the claim terminology namely the term ‘olive oil polyphenols’ is not entirely clear and accurate as ‘olive oil’ is a generic term for the type of oil, and the basic structure of OO phenolic compounds do not coincide with a ‘polyphenolic’ structure; accordingly ‘virgin olive oil bioactive phenols’ is a more appropriate term. Others also commented about the lack of robust and reliable methods for quantifying phenolic compounds in OO. A simple and robust method for routine analysis of hydroxytyrosol and tyrosol was proposed( Reference Mastralexi, Nenadis and Tsimidou 81 , Reference Romero and Brenes 82 ) based on hydrolysis of the polar fraction of OO. This was followed by development and validation of a 1H NMR method enabling direct measurement of tyrosol and hydroxytyrosol derivatives, as well as oleocanthal and oleacein in OO, overcoming analytical issues such as chromatographic peak broadening( Reference Karkoula, Skantzari and Melliou 19 ).

Cardioprotective mechanisms of oleic acid

OO intake has been related with a decrease on blood pressure with oleic acid regarded as being a major contributor to this effect, as evidenced in animal models( Reference Teres, Barcelo-Coblijn and Benet 83 ). Chronic oral administration of VOO (rich in oleic acid), triolein (a TAG with three oleic acid moieties) or oleic acid over 14 d significantly reduced systolic blood pressure in rats (−26 (sem 4) for VOO and −21 (sem 3) mm Hg for triolein, P < 0·001 and −17 (sem 1·9) mm Hg for oleic acid P < 0·05) when compared with the control group that received water. Similarly acute (2 h) treatments with either VOO or triolein also significantly reduced systolic blood pressure when compared with the control group (−20 (sem 0) mm Hg, P < 0·001 and −14 (sem 2) mm Hg, respectively; P < 0·05) with oleic acid again significantly reducing systolic blood pressure (−13·0 (sem 0·3) mm Hg; P < 0·001). In contrast, chronic treatment with the trans-MUFA elaidic (18 : 1n-9) or the SFA stearic acid (18 : 0) did not significantly affect blood pressure. Results show that saturation and cis/trans double-bond arrangement are implicated with the cardioprotective effect of the long-chain fatty acid in this animal model at high-dose levels( Reference Teres, Barcelo-Coblijn and Benet 83 ). Similar significant results were obtained after VOO and oleic acid intake in an animal model of hypertension using spontaneously hypertensive rats( Reference Teres, Barcelo-Coblijn and Benet 83 ).

The molecular mechanisms were evaluated by measuring signalling proteins involved in the control of blood pressure in the aorta. OO intake increases oleic acid levels in membranes, which regulate membrane lipid structure and impact on G-protein-mediated signalling, causing a reduction in blood pressure( Reference Yang, Alemany and Casas 84 ). Unlike its analogues elaidic and stearic acid, oleic acid, due to its cis-18 : 1n-9 structure, regulates cellular membrane lipid structure and the α2 receptor system involved in the control of blood pressure (α2A/D – adrenoreceptor/G protein/adenylyl cyclase-cAMP/PKA) as demonstrated in vitro ( Reference Yang, Alemany and Casas 84 ) and in vivo ( Reference Teres, Barcelo-Coblijn and Benet 83 ). Oleic acid can also contribute to heart health via intramyocardial TAG turnover( Reference Lahey, Wang and Carley 85 ), which is reduced in pressure-overloaded failing hearts. In this situation, oleate (derivative of oleic acid) upregulated TAG dynamics when compared with palmitate (derivative of palmitic acid and major SFA of palm oil). This result underscores the importance of the intracellular lipid storage type on nuclear receptor signalling and contractility( Reference Lahey, Wang and Carley 85 ) in diseased hearts.

An important driver of vasorelaxation is NO, a free radical that readily reacts with fats and proteins. Nitro-fatty acids are mediators of cardiovascular signalling actions( Reference Rudolph, Rudolph and Schopfer 86 ) as these compounds relax blood vessels, attenuate platelet activation and reduce inflammation( Reference Coles, Bloodsworth and Clark 87 , Reference Coles, Bloodsworth and Eiserich 88 ).

Both oleic acid and linoleic acid are unsaturated fatty acids that after reaction with nitrite may form nitro-fatty acids. Nitro-oleic acid-mediated antihypertensive signalling actions were shown in a mouse model( Reference Charles, Rudyk and Prysyazhna 89 ). The mechanism was attributed to the inhibition of soluble epoxide hydrolase by nitro-fatty acids, thus lowering blood pressure in an angiotensin II-induced hypertension( Reference Charles, Rudyk and Prysyazhna 89 ). It is however unclear how the extent of nitrite in the human diet may contribute to nitration of dietary fat, and the physiological relevance of this finding.

Role of phenolic compounds on endothelium protection

Oxidative stress and reactive oxygen species have been implicated in endothelial damage, progression to atherosclerosis, injury in sustained myocardial infarction and ischaemia reperfusion( Reference Khurana, Venkataraman and Hollingsworth 76 , Reference Dhalla, Temsah and Netticadan 90 Reference Raedschelders, Ansley and Chen 92 ). Monocytes and macrophages are critical cells that are involved in atherosclerosis. These cells produce proinflammatory cytokines, such as IL-1β, TNF-α and C-reactive protein, which induce the expression of adhesion molecules such as intercellular adhesion molecule-1, vascular-cell adhesion molecule-1 and E-selectin( Reference Ross 93 ).

Meanwhile, oxidative stress through reactive oxygen species production promotes the expression of the adhesion molecules on the endothelium( Reference Dell'Agli, Fagnani and Galli 94 ).

Expression of adhesion molecules attracts circulating monocytes, inducing their adherence to the endothelium. OO phenolic compounds have been shown to act on endothelium protection as evidenced in in vitro assays with typical OO phenolic compounds and less on in vivo circulating metabolites. OO phenolic extract, oleuropein aglycone or homovanillic alcohol (metabolite of hydroxytyrosol) had inhibitory effects on vascular-cell adhesion molecule-1, intercellular adhesion molecule-1 and E-selectin surface expression in human umbilical vascular endothelial cells, using TNF-α as pro-inflammatory stimulus( Reference Dell'Agli, Fagnani and Mitro 95 ).

Endothelium dysfunction refers to an impairment of endothelium-dependent vasorelaxation caused by a loss of NO bioactivity in the vessel wall. In animal models with rats oral hydroxytyrosol administration was tested on NO production and platetet function( Reference Gonzalez-Correa, Navas and Munoz-Marin 96 ). Results showed that hydroxytyrosol administration (100 mg/kg daily) increased vascular NO production by up to 34·2 % (P < 0·01) and inhibited platelet aggregation for 50 % inhibitory dose of 48·25 mg/d for hydroxytyrosol (P < 0·01) when compared with control group (treated with isotonic saline solution). Animal dose translation to human subjects allowed us to conclude that the effective hydroxytyrosol doses tested would be above the expected intake through OO daily. The reported benefits would only be achievable through nutraceutical supplementation.

Endothelium repair: matrix metalloproteinases and olive oil

Matrix metalloproteinases (MMP) play a role in endothelium repair. Macrophages resident in human and experimental atherosclerosis co-localise with and release active MMP, including the gelatinase MMP-9, which is specialised in the digestion of basement membrane collagens and elastin, and is implicated in atherogenesis, unstable coronary syndromes and in aortic aneurysms( Reference Dollery and Libby 97 ). Accumulating evidence points to the MMP as major molecular mediators of arterial diseases( Reference Dollery and Libby 97 ). Collagens, types 1 and 3, are the main proteins in arterial walls being also present in the thickened intima of atherosclerotic lesions( Reference Barderas, Vivanco and Alvarez-Llamas 98 , Reference von Zur Muhlen, Schiffer and Zuerbig 99 ). Fragments of collagens found in urine are present as a result of proteolytic activity in arterial walls and other vascular structures. Collagen type 1 or 3 fragments were upregulated in urine in coronary artery disease (CAD) patients( Reference Zimmerli, Schiffer and Zurbig 100 ). Increase in collagen degradation is related with an increase on collagenases circulation, such as MMP-9, as shown in patients with CAD( Reference Kalela, Koivu and Sisto 101 ).

In an in vitro study hydroxytyrosol (1–10 μm) reduced MMP-9 (IC50 = 10 μm/l, P < 0·05) and COX-2 induction in activated human monocytes, with phorbol myristate acetate( Reference Scoditti, Nestola and Massaro 102 ). These effects were mediated by inhibition of transcription factor NF-κB and protein kinase Cα and protein kinase C β1 activation( Reference Scoditti, Nestola and Massaro 102 ). Results are in line with previous in vitro reports that showed inhibition of MMP-9 on endothelial cells by OO phenolics namely hydroxytyrosol in phorbol myristate acetate-induced cells( Reference Scoditti, Calabriso and Massaro 103 ) and oleuropein aglycone in TNF-α-induced cells by acting on NF-κB( Reference Dell'Agli, Fagnani and Galli 94 ). No hydroxytyrosol activity on MMP-9 was found in TNF-α-induced cells( Reference Dell'Agli, Fagnani and Galli 94 ).

The discriminatory polypetides that increase in CAD includes collagen type 1 and 3 fragments with a C-terminal GxPGP motif( Reference Delles, Schiffer and von Zur Muhlen 104 ). Increase on these polypeptides would come from a protease decrease activity possibly related with chemical change of the substrate (e.g. oxidative damage) thus inhibiting it acting at a specific site, or a decrease in circulating levels by lack of enzyme activation. MMP-2 is secreted in an inactive form (pro-MMP-2) and several factors can promote its activation such as plasmin( Reference Monea, Lehti and Keski-Oja 105 ) and thrombin( Reference Lafleur, Hollenberg and Atkinson 106 ). Other mechanisms that involve proteinases or oxidative stress can also activate MMP-2( Reference Rajagopalan, Meng and Ramasamy 107 ). Therefore antioxidants, as phenolic compounds, might have a role on MMP-2 activation and published data indicate phenolic compounds from red wine( Reference Oak, El Bedoui and Anglard 108 ) and green tea( Reference El Bedoui, Oak and Anglard 109 ) as acting on prevention of thrombin-induced activation of MMP-2 in vascular smooth cells.

We evaluated the impact of a 6-week OO supplementation in healthy adults on urinary proteomic biomarkers of CAD in a randomised, parallel, controlled, double-blind study( Reference Silva, Bronze and Figueira 110 ). The present study was the first to describe the significant impact of daily OO supplementation on highly specific disease biomarkers for CAD. Analysis of urinary proteomic profiles at baseline and endpoint enabled the identification of twelve sequenced peptides that were significantly regulated towards healthy scoring. Eight of them included four collagen α−1(I) chain, one α−2 (1) chain, one α−2(V) chain and one α−2(VI) chain fragments. Changes in circulating concentrations of collagenases may mediate these changes in the urinary fingerprint. Therefore with more data or in future intervention studies with OO, it would be interesting to link urinary fragments to the proteases involved in their generation. This predictive analysis would enable looking at the peptide cleavage sites studying the MMP up or downregulated with OO intervention.

The majority of studies of dietary intake of proposed bioactive foods asses the activities of these foods based on the major risk factors of CVD. However, markers such as lipoprotein profile, blood pressure, endothelial function, inflammation and oxidative stress have no direct link to the disease itself but are merely associated with it. There is a great need for more biomarkers that appear as a direct result of the disease itself( Reference Ruiz-Canela and Martinez-Gonzalez 63 , Reference Covas, Konstantinidou and Fito 67 ).

Proteomics biomarkers as a mechanistic approach to explain olive oil health effects

The systems biology approach (encompassing genomics, transcriptomics, proteomics and metabolomics using urine, blood or saliva) could provide a greater understanding of disease development, treatment efficacy and evaluation of the influence of food bioactive compounds( Reference Tulp, Bruhn and Bohlin 46 , Reference Wang, Lamers and Korthout 111 ). There is a need for biomarkers of practical value for clinical intervention, allowing disease risk prediction and more importantly early diagnosis. Accuracy, reproducibility, availability, feasibility of implementation into the clinical settings, sensitivity and specificity are additional characteristics to be fulfilled, and panels of biomarkers are gaining acceptance instead of individual molecules( Reference Finley Austin and Babiss 112 ), as single biomarkers are often not available and lack the ability to adequately describe complex diseases( Reference Schanstra and Mischak 113 ). Candidate biomarkers should be carefully validated in a wide and different cohort of samples from those used in the discovery phase as often overfitting of the biomarker model has occurred( Reference Mischak, Allmaier and Apweiler 114 ).

The proteome, corresponding to a set of expressed proteins, informs the current ‘status’ of an organism, constantly changing according to endogenous and exogenous factors( Reference Schanstra and Mischak 113 ). Proteins are widely used in different clinical tests for both diagnosis and prognosis of diseases and to follow their evolutions( Reference Barderas, Vivanco and Alvarez-Llamas 98 ). They can be used to measure the extent of inflammation, calcification and the development of plaques on the arteries. Understanding what causes plaque rupture is of great importance. As previously mentioned, MMP could have a key role in this process( Reference Stegemann, Didangelos and Barallobre-Barreiro 115 ). The discovery of proteomic biomarkers may be useful in understanding the molecular mechanisms involved in the onset and progression of other vascular diseases( Reference Mischak and Rossing 116 ). Plasma, serum and urine are the most commonly used biological matrices in cardiovascular research, due to their perceived clinical relevance as a source of potential biomarkers( Reference Barderas, Vivanco and Alvarez-Llamas 98 ). However, proteomic studies have also been carried out on vascular tissues (arteries), artery layers, cells looking at proteomes and secretomes, exosomes, lipoproteins and metabolites( Reference Barderas, Vivanco and Alvarez-Llamas 98 ). Although sampling the tissue may seem an obvious method there are a number of difficulties, especially where the need for a biopsy would be required( Reference Lescuyer, Hochstrasser and Rabilloud 117 ). Recent advances in extraction processes and LC-MS/MS analysis has allowed the quantitative analysis of tissue samples in vascular research to be carried out( Reference Wisniewski, Zougman and Nagaraj 118 , Reference Husi, Van Agtmael and Mullen 119 ).

Urine as a sample source is now recognised as the source of choice for proteomic biomarker investigations. It has a number of advantages such as being non-invasive and can be collected by untrained personnel. Urine is produced by renal filtration of the plasma and approximately 70 % of proteins in the normal human urinary proteome are of kidney origin, whereas the remaining 30 % are derived from plasma proteins( Reference Thongboonkerd, McLeish and Arthur 120 , Reference Thongboonkerd and Malasit 121 ). It has high stability due to absence of proteolytic agents and the low dynamic range of analyte concentration facilitates the detection and quantification of peptides( Reference Schanstra and Mischak 113 , Reference Mischak, Kolch and Aivaliotis 122 ).

Using capillary electrophoresis coupled with MS( Reference Albalat, Franke and Gonzalez 123 ) urinary biomarker classifiers for the diagnosis of diseases such as chronic kidney disease (CKD)( Reference Good, Zurbig and Argiles 124 ), acute kidney injury( Reference Metzger, Kirsch and Schiffer 125 ), stroke( Reference Dawson, Walters and Delles 126 ) and CAD( Reference Delles, Schiffer and von Zur Muhlen 104 ), were already identified, allowing classification of case–control groups with good accuracy( Reference Mischak and Schanstra 127 ).

Urinary peptides and protein fragments are the end products of proteolytic processes. The different pattern of urinary excretion of peptides when comparing controls and disease patients might indicate their role in the pathophysiology of disease. Therefore changes in the normal urine ‘fingerprint’ (e.g. presence of collagen fragments) can be used as biomarkers of disease. Besides collagens, common blood proteins (e.g. α1-antitrypsin, haemoglobin, serum albumin and fibrinogen), and uromodulin were also identified( Reference Coon, Zurbig and Dakna 128 ) in urine which provides additional proof of the suitability of this sample source for proteomic biomarker studies outwith the kidney and urinary tract. Collagens are the most abundant peptides sequenced so far in the CAD biomarker (66 % of all peptides)( Reference Delles, Schiffer and von Zur Muhlen 104 ), with atherosclerosis associated with an increased synthesis of several extracellular matrix components, including collagen types 1 and 3, elastin and several proteoglycans( Reference Lee and Libby 129 ). Changes in the circulating levels of collagenases may mediate these changes in peptides represented in the fingerprint, as reported in coronary atherosclerosis( Reference Zimmerli, Schiffer and Zurbig 100 ) and CKD( Reference Coon, Zurbig and Dakna 128 ).

The progress in urinary proteomics and the use of multiple biomarker classifiers opens the possibility of establishing new tools adapted to different clinical needs( Reference Zurbig, Jerums and Hovind 130 ), enabling direct monitoring of disease overcoming limitations of indirect measurements.

Proteomic in vitro studies on olive oil phenolic compounds

Proteomics has been applied in a number of studies of OO phenolic compounds on cardiovascular health using animal and in vitro studies. The in vitro effects of alperujo extract, an OO production waste product containing phenolic compounds present in olive fruits, were studied on platelet aggregation and changes in the platelet proteome( Reference de Roos, Zhang and Rodriguez Gutierrez 131 ). Nine proteins were differentially regulated by the alperujo extract upon platelet aggregation underlying the anti-platelet effects of the extract. However, like a number of previously mentioned in vitro studies, the effective concentrations (40–500 mg/l) were far above the physiologically concentrations achievable by dietary intake.

The effects of EVOO, with low and high phenolic content, were evaluated in the hepatic proteome in ApoE−/− mice that spontaneously develop atherosclerosis( Reference Arbones-Mainar, Ross and Rucklidge 132 ). For 10 weeks the mice were fed with a high-fat, high-cholesterol diet supplemented with 0·15 % (w/w) cholesterol and either 20 % (w/w) low phenolic EVOO or 20 % (w/w) high phenolic EVOO v. a control group fed with 0·15 % (w/w) cholesterol and 20 % (w/w) palm oil. Within this work a range of hepatic antioxidant enzymes differentially regulated by OO( Reference Arbones-Mainar, Ross and Rucklidge 132 ) were identified. The authors concluded that the upregulation of a large array of antioxidant enzymes might explain anti-atherogenic mechanisms of EVOO( Reference Arbones-Mainar, Ross and Rucklidge 132 ). Again the dose level was above what could be achieved through dietary intake and translation from an animal model to human use has also to be considered.

Urinary proteomics biomarkers, olive oil and CVD

Atherosclerosis is a process of chronic inflammation, characterised by the accumulation of lipids, cells, and fibrous elements in medium and large arteries( Reference Barderas, Vivanco and Alvarez-Llamas 98 ). The extent of inflammation, proteolysis, calcification and neovascularisation influences the development of advanced lesions (atheroma plaques) on the arteries( Reference Barderas, Vivanco and Alvarez-Llamas 98 ).

Classical risk factors in atherosclerosis (hypertension, LDL-cholesterol, C-reactive protein, ageing, smoking, male gender, among others) do not actually measure disease initiation or progression. As such, they cannot be used directly to identify individuals who have developed atherosclerosis and prevent a fatal event( Reference Barderas, Vivanco and Alvarez-Llamas 98 , Reference Ge and Wang 133 ). Other, more recent markers that indicate changes in vascular structure can still only be detected once CVD has progressed to an advanced stage where drug or surgical intervention is required( Reference Sharma, Cosme and Gramolini 134 ).

The analysis of urine samples from diseased and healthy individuals has been used to establish a database of naturally occurring urinary peptides, making a basis for the definition and validation of biomarkers for diagnosis/prognosis/monitoring of a wide range of diseases using proteomic biomarker patterns( Reference Coon, Zurbig and Dakna 128 ), such as CAD( Reference Zimmerli, Schiffer and Zurbig 100 ), emphasising that non-invasive proteomics analysis could become a valuable addition to assess CVD alongside other biomarkers that are indicators of cardiovascular risk.

The first time that urinary proteomics was applied to assess cardiovascular health improvements of OO consumption in human subjects, was in a randomised, parallel, controlled, double-blind study designed to evaluate the impact of a 6-week OO supplementation in healthy adults on urinary proteomic biomarkers of CAD( Reference Silva, Bronze and Figueira 110 ). The impact of the supplementation with OO was also studied on urinary proteomic biomarkers of CKD and diabetes.

The increase or decrease in the concentration of the peptides in the biomarker determines the scoring value of each disease biomarker. The CAD proteomic biomarker developed for clinical diagnosis produces a CAD scoring system from 1 (CAD case) to −1 (healthy artery). A scoring of disease absence, presence and severity is provided, based on the concentration of a group (panel) of urinary peptides measured by capillary electrophoresis coupled with MS, allowing monitoring of progression and/or effect of treatment( Reference Fliser, Novak and Thongboonkerd 135 , Reference Julian, Suzuki and Suzuki 136 ). In the present study, self-reported healthy participants were randomly allocated to supplementation with a daily dose of OO either low or high in phenolic compounds. For 6 weeks, they consumed a daily dose of 20 ml OO (not heated or cooked) as a supplement (no specific time during the day, single intake, equivalent to 6 mg hydroxytyrosol and derivatives for the high phenolic OO), in line with the EFSA and Food and Drug Administration recommendations. The impact of supplementation with OO was evaluated on urinary proteomic biomarkers of CAD with biomarkers being measured at baseline and 3 and 6 weeks. Consumption of both OO significantly improved the proteomic CAD score at endpoint compared with baseline, moving the CAD biomarker pattern in a healthy profile direction (Table 2). No differences were observed for CKD or diabetes proteomic biomarkers, Table 2.

Table 2. Scores of CAD, CKD and diabetes proteomic biomarkers at baseline, middle (3 weeks) and end of intervention (6 weeks)

CAD, coronary artery disease; CKD, chronic kidney disease (adapted from Silva et al. ( Reference Silva, Bronze and Figueira 110 )).

Compared with corresponding baseline value: *P < 0·005, **P < 0·001. There were no significant differences in changes between groups.

A repeated-measures ANOVA test was used with statistical significance at P < 0·05.

In a placebo-controlled intervention, irbesartan (angiotensin II receptor antagonist used for the treatment of hypertension) taken at 300 mg/d over 2 years in hypertensive type 2 diabetes patients, using the CAD 238 biomarker panel, led to a 0·35 point reduction in the CAD score for the drug-controlled group( Reference Delles, Schiffer and von Zur Muhlen 104 ), which saw a significant reduction in incidents of CAD in this group. In the nutritional intervention( Reference Silva, Bronze and Figueira 110 ), the CAD score change in the intervention was significant for both OO tested, using the same CAD 238 biomarker, leading to a similar degree of change as observed for irbersartan over a 6-week period. This evidence highlights the importance of the CAD biomarker as a tool for nutrition and health intervention studies. This type of urinary biomarker enabled the measurement of health effects induced by a change in diet that could not be detected by monitoring the conventional risk markers of CAD such as plasma TAG, oxidised LDL, and LDL-cholesterol. The overall change in CAD score in a short period of time is more likely due to OO major components, such as fatty acids. However, the role of other OO minor components other than phenolic compounds should also be taken into account. Squalene, a polyunsaturated triterpene which makes up 60–75 % of the unsaponifiable fraction of OO( Reference Perona, Cabello-Moruno and Ruiz-Gutierrez 137 ), reduced atherosclerotic lesion size in male mice( Reference Guillen, Acin and Navarro 138 ) and further investigation is needed to clarify its role on CVD.

Our results emphasise further the potential role of nutrition in the prevention or delay of CVD and offer new perspectives on OO applications. These results are highly translatable to guidelines for nutritional recommendations. The biomarkers were originally developed to detect early signs of diseases in clinical setting and to inform clinicians as to the effectiveness of treatment. However, the technology also provides a sensitive tool for the assessment of potential bioactive foods in cardiovascular health, CKD and diabetes, with a range of additional tests under development. Further testing of reportedly bioactive foods can now be carried out which will allow better nutritional health advice to be advanced and could also lead to better food labelling, so that the public can make informed choices on their food purchases.

Exploring olive oil health benefits: perspectives

Although strong evidence from heritability is related with CVD many forms of heart disease are not genome associated( Reference Monte and Vondriska 139 ). The epigenome is a possible link between genetics and environment( Reference Monte and Vondriska 139 ) which includes impact of food components/diet. Omics techniques (such as genomics, transcriptomics, proteomics, epigenomics and metabolomics) have the potential, when integrated, to comprehensively demonstrate the contribution of diet towards the modulation of disease risk( Reference Camargo, Ruano and Fernandez 140 ). Some trials have shown the impact of OO on downregulation of atherosclerosis-related genes( Reference Camargo, Ruano and Fernandez 140 , Reference Konstantinidou, Covas and Munoz-Aguayo 141 ). The effect of Mediterranean diet was studied on urinary metabolome( Reference Vazquez-Fresno, Llorach and Urpi-Sarda 142 ) and related to compounds of the metabolism of carbohydrates, creatine, creatinine, amino acids, lipids and microbial cometabolites.

Phenolic compounds can interact with cellular signalling cascades regulating the activity of transcription factors with impact on gene expression. For instance, phenolic compounds have shown to affect the expression of microRNA( Reference Noratto, Angel-Morales and Talcott 143 ). microRNA are small, non-coding RNA implicated in the regulation of gene expression that control both physiological and pathological processes, influenced by external factors as diet components( Reference Milenkovic, Jude and Morand 144 ). Most of the studies reported in this field are in vitro and more in vivo studies are needed to clarify microRNA targets of dietary phenolic compounds( Reference Milenkovic, Jude and Morand 144 ).

Interactions between genes and the bioactive components present in OO studied by nutrigenomics may help to explain its health benefits( Reference Garcia-Gonzalez and Aparicio 145 ). In this sense, besides their antioxidant and anti-inflammatory capacities, OO phenolic compounds are able to modify gene expression coding in a protective mode for proteins participating in the cellular mechanisms involved in oxidative stress resistance, inflammation or lipid metabolism amongst others( Reference Martin-Pelaez, Covas and Fito 146 ).

Glycation, a non-enzymatic reaction between reducing sugars and proteins, is a proteome wide phenomenon, mainly observed in diabetes due to hyperglycaemia( Reference Dunn 147 ), but also relevant to end organ damage, disease pathogenesis and ageing( Reference Levi and Werman 148 ) and OO phenolic compounds have been reported as potent inhibitors of the formation of advanced glycation end products( Reference Kontogianni, Charisiadis and Margianni 149 ). Our human intervention trial with OO low or high in phenolics did not find a significant impact on the plasma fructosamine levels( Reference Silva, Bronze and Figueira 110 ). A key factor may be the duration of the study (6 weeks) not being sufficient to detect changes in protein modifications such as glycation, and may also be partly related to the quantity and quality of phenolic compounds, which exert differential antioxidant and antiglycative activities depending on structure( Reference Tripoli, Giammanco and Tabacchi 4 , Reference Vlassopoulos, Lean and Combet 150 ). Further studies should proceed in order to clarify anti-glycation properties of OO phenolic compounds, given that glycation is a key driver for tissue damage and is present in all non-communicable disease scenarios.

Conclusion

Results outlined in the present review provide evidence of health benefits related with OO intake. The reported studies may allow the implementation of primary prevention programs of CVD, based on nutritional interventions, useful in non-regular OO consumers groups like the Northern European populations. Interventions in broad populations with highly specific disease biomarkers, as urinary proteomic biomarkers, will offer higher translational value, especially towards development and implementation of new nutritional recommendations.

Human intervention trials focusing on new outcomes related with proteomics and nutrigenomics are needed to better clarify pathways/mechanisms by which oleic acid, phenolic compounds or even other OO components act on CVD risk factors and affect the proteome.

Acknowledgements

The authors would like to acknowledge the support and distinction of Ordem dos Farmacêuticos (Lisbon, Portugal) on the basis of an innovation award attributed to the work ‘Protective effects of hydroxytyrosol-supplemented refined olive oil in animal models of acute inflammation and rheumatoid arthritis'.

Financial Support

QREN project Azeite+ Global no. 12228 and Ordem dos Farmacêuticos (Lisbon, Portugal).

Conflict of Interest

T. K. is employed at Mosaiques Diagnostics, the company that developed the urinary proteomics for capillary electrophoresis coupled with MS technology for clinical application.

Authorship

S. S. conducted the studies described and drafted the manuscript. E. C., M. E. F., W. M. and M. R. B. supervised the studies and contributed to the drafting of the manuscript. All authors were responsible for the critical review of the manuscript.

References

1. Obied, HK, Prenzler, PD, Ryan, D et al. (2008) Biosynthesis and biotransformations of phenol-conjugated oleosidic secoiridoids from Olea europaea L. Nat Prod Rep 25, 11671179.CrossRefGoogle ScholarPubMed
2. European Comission (2011) Official J Eur Union, 27.1.2011, Commission Regulation (EC) No 61/2011 of 24 January 2011. Available at http://eur-lex.europa.eu/legal-content/EN/TXT/?qid=1433842490849&uri=CELEX:32011R0061 Google Scholar
3. European Comission (2008) Official J Eur Union, 5.7.2008, Commission Regulation (EC) No 640/2008 of 4 July 2008. Available at http://eur-lex.europa.eu/legal-content/EN/TXT/?qid=1433776627017&uri=CELEX:32008R0640 Google Scholar
4. Tripoli, E, Giammanco, M, Tabacchi, G et al. (2005) The phenolic compounds of olive oil: structure, biological activity and beneficial effects on human health. Nutr Res Rev 18, 98112.CrossRefGoogle ScholarPubMed
5. Covas, MI (2007) Olive oil and the cardiovascular system. Pharmacol Res 55, 175186.CrossRefGoogle ScholarPubMed
6. Lopez-Miranda, J, Perez-Jimenez, F, Ros, E et al. (2010) Olive oil and health: summary of the II International Conference on olive oil and health consensus report, Jaen and Cordoba (Spain) 2008. Nutr Metab Cardiovasc Dis 20, 284294.Google Scholar
7. Urpi-Sarda, M, Casas, R, Chiva-Blanch, G et al. (2012) Virgin olive oil and nuts as key foods of the Mediterranean diet effects on inflammatory biomarkers related to atherosclerosis. Pharmacol Res 65, 577583.Google Scholar
8. Martinez-Lapiscina, EH, Clavero, P, Toledo, E et al. (2013) Virgin olive oil supplementation and long-term cognition: the PREDIMED-NAVARRA randomized, trial. J Nutr Health Aging 17, 544552.Google Scholar
9. Kastorini, CM, Milionis, HJ, Goudevenos, JA et al. (2010) Mediterranean diet and coronary heart disease: is obesity a link? – A systematic review. Nutr Metab Cardiovasc Dis 20, 536551.Google Scholar
10. Razquin, C, Martinez, JA, Martinez-Gonzalez, MA et al. (2009) A 3 years follow-up of a Mediterranean diet rich in virgin olive oil is associated with high plasma antioxidant capacity and reduced body weight gain. Eur J Clin Nutr 63, 13871393.CrossRefGoogle Scholar
11. Schwingshackl, L & Hoffmann, G (2014) Mediterranean dietary pattern, inflammation and endothelial function: a systematic review and meta-analysis of intervention trials. Nutr Metab Cardiovasc Dis 24, 929939.Google Scholar
12. Schwingshackl, L & Hoffmann, G (2014). Monounsaturated fatty acids, olive oil and health status: a systematic review and meta-analysis of cohort studies. Lipids Health Dis 13, 154.Google Scholar
13. Harwood, J & Aparico, R. (2000) Handbook of Olive oil Analysis and Properties. Gaithersburg, MD: Aspen.Google Scholar
14. Servili, M & Montedoro, G (2002) Contribution of phenolic compounds to virgin olive oil quality. Eur J Lipid Sci Technol 104, 602613.Google Scholar
15. Perez-Jimenez, F, Ruano, J, Perez-Martinez, P et al. (2007) The influence of olive oil on human health: not a question of fat alone. Mol Nutr Food Res 51, 11991208.Google Scholar
16. Di Maio, I, Esposto, S, Taticchi, A et al. (2011) HPLC–ESI–MS investigation of tyrosol and hydroxytyrosol oxidation products in virgin olive oil. Food Chem 125, 2128.Google Scholar
17. Brenes, M, Garcia, A, Garcia, P et al. (2001) Acid hydrolysis of secoiridoid aglycons during storage of virgin olive oil. J Agric food Chem 49, 56095614.Google Scholar
18. Hrncirik, K & Fritsche, S. (2004) Comparability and reliability of different techniques for the determination of phenolic compounds in virgin olive oil. Eur J Lipid Sci Technol 106, 540549.Google Scholar
19. Karkoula, E, Skantzari, A, Melliou, E et al. (2012) Direct measurement of oleocanthal and oleacein levels in olive oil by quantitative (1)H NMR. Establishment of a new index for the characterization of extra virgin olive oils. J Agric Food Chem 60, 1169611703.Google Scholar
20. Corona, G, Tzounis, X, Assunta Dessi, M et al. (2006) The fate of olive oil polyphenols in the gastrointestinal tract: implications of gastric and colonic microflora-dependent biotransformation. Free Radic Res 40, 647658.CrossRefGoogle ScholarPubMed
21. Pinto, J, Paiva-Martins, F, Corona, G et al. (2011) Absorption and metabolism of olive oil secoiridoids in the small intestine. Br J Nutr 105, 16071618.Google Scholar
22. Aranzazu Soler, MPR, Alba, M., Shikha, S et al. (2010) Motilva. Digestion stability and evaluation of the metabolism and transport of olive oil phenols in the human small-intestinal epithelial Caco-2/TC7 cell line. Food Chem 119, 703714.CrossRefGoogle Scholar
23. Vissers, MN, Zock, PL, Roodenburg, AJ et al. (2002) Olive oil phenols are absorbed in humans. J Nutr 132, 409417.Google Scholar
24. Garcia-Villalba, R, Carrasco-Pancorbo, A, Nevedomskaya, E et al. (2010) Exploratory analysis of human urine by LC-ESI-TOF MS after high intake of olive oil: understanding the metabolism of polyphenols. Anal Bioanal Chem 398, 463475.Google Scholar
25. Mateos, R, Goya, L & Bravo, L (2005) Metabolism of the olive oil phenols hydroxytyrosol, tyrosol, and hydroxytyrosyl acetate by human hepatoma HepG2 cells. J Agric Food Chem 53, 98979905.Google Scholar
26. Miro-Casas, E, Covas, MI, Farre, M et al. (2003) Hydroxytyrosol disposition in humans. Clin Chem 49, 945952.Google Scholar
27. Miro Casas, E, Farre Albadalejo, M, Covas Planells, MI et al. (2001) Tyrosol bioavailability in humans after ingestion of virgin olive oil. Clin Chem 47, 341343.Google Scholar
28. Garcia-Villalba, R, Larrosa, M, Possemiers, S et al. (2014) Bioavailability of phenolics from an oleuropein-rich olive (Olea europaea) leaf extract and its acute effect on plasma antioxidant status: comparison between pre- and postmenopausal women. Eur J Nutr 53, 10151027.CrossRefGoogle ScholarPubMed
29. Suarez, M, Valls, RM, Romero, MP et al. (2011) Bioavailability of phenols from a phenol-enriched olive oil. Br J Nutr 106, 16911701.Google Scholar
30. Preedy, VR & Watson, RR (2010). Olives and Olive oil in Health and Disease Prevention, 1st ed. San Diego: Elsevier.Google Scholar
31. Tuck, KL, Freeman, MP, Hayball, PJ et al. (2001). The in vivo fate of hydroxytyrosol and tyrosol, antioxidant phenolic constituents of olive oil, after intravenous and oral dosing of labeled compounds to rats. J Nutr 131, 19931996.CrossRefGoogle ScholarPubMed
32. Gonzalez-Santiago, M, Fonolla, J & Lopez-Huertas, E (2010) Human absorption of a supplement containing purified hydroxytyrosol, a natural antioxidant from olive oil, and evidence for its transient association with low-density lipoproteins. Pharmacol Res 61, 364370.CrossRefGoogle ScholarPubMed
33. Visioli, F, Galli, C, Grande, S et al. (2003) Hydroxytyrosol excretion differs between rats and humans and depends on the vehicle of administration. J Nutr 133, 26122615.Google Scholar
34. Mosele, JI, Martin-Pelaez, S, Macia, A et al. (2014) Faecal microbial metabolism of olive oil phenolic compounds: in vitro and in vivo approaches. Mol Nutr Food Res 58, 18091819.Google Scholar
35. Khymenets, O, Fito, M, Tourino, S et al. (2010) Antioxidant activities of hydroxytyrosol main metabolites do not contribute to beneficial health effects after olive oil ingestion. Drug Metab Dispos: Biol Fate Chem 38, 14171421.Google Scholar
36. Tuck, KL, Hayball, PJ & Stupans, I (2002) Structural characterization of the metabolites of hydroxytyrosol, the principal phenolic component in olive oil, in rats. J Agric Food Chem 50, 24042409.CrossRefGoogle ScholarPubMed
37. Kotronoulas, A, Pizarro, N, Serra, A et al. (2013) Dose-dependent metabolic disposition of hydroxytyrosol and formation of mercapturates in rats. Pharmacol Res 77, 4756.CrossRefGoogle ScholarPubMed
38. Laura Rubió, AS, Alba, M., Carme, P. et al. (2014). In vivo distribution and deconjugation of hydroxytyrosol phase II metabolites in red blood cells: a potential new target for hydroxytyrosol. J Funct Foods 10, 139143.Google Scholar
39. Hamden, K, Allouche, N, Damak, M et al. (2009) Hypoglycemic and antioxidant effects of phenolic extracts and purified hydroxytyrosol from olive mill waste in vitro and in rats. Chem Biol Interact 180, 421432.CrossRefGoogle ScholarPubMed
40. Sanchez de Medina, V, Priego-Capote, F & Luque de Castro, MD (2012) Characterization of refined edible oils enriched with phenolic extracts from olive leaves and pomace. J Agric Food Chem 60, 58665873.Google Scholar
41. Suarez, M, Romero, MP & Motilva, MJ (2010) Development of a phenol-enriched olive oil with phenolic compounds from olive cake. J Agric Food Chem 58, 1039610403.Google Scholar
42. Pashkow, FJ (2011) Oxidative stress and inflammation in heart disease: do antioxidants have a role in treatment and/or prevention? Int J Inflamm 2011, 514623.CrossRefGoogle ScholarPubMed
43. Vlantis, K & Pasparakis, M (2010) Role of TNF in pathologies induced by nuclear factor kappa B deficiency. Curr Dir Autoimmun 11, 8093.Google Scholar
44. Beauchamp, GK, Keast, RS, Morel, D et al. (2005) Phytochemistry: ibuprofen-like activity in extra-virgin olive oil. Nature 437, 4546.CrossRefGoogle ScholarPubMed
45. Beauchamp, GK, Keast, RS, Morel, D et al. (2005) Ibuprofen-like activity in extra-virgin olive oil. Nature 437, 4546.Google Scholar
46. Tulp, M, Bruhn, JG & Bohlin, L (2006) Food for thought. Drug Discov Today 11, 11151121.Google Scholar
47. O'Connor, Á (2014) An overview of the role of diet in the treatment of rheumatoid arthritis. Nutr Bull 39, 7488.Google Scholar
48. Waterman, E & Lockwood, B (2007) Active components and clinical applications of olive oil. Altern Med Rev 12, 331342.Google Scholar
49. Silva, BS, Sepodes, B, Rocha, J et al. (2015) Protective effects of hydroxytyrosol-supplemented refined olive oil in animal models of acute inflammation and rheumatoid arthritis. J Nutr Biochem 26, 360368.Google Scholar
50. European Comission (2012) Official J Eur Union, 25.5.2012, Comission Regulation (EC) No 432/2012 of 16 May 2012. Available at http://eur-lex.europa.eu/legal-content/EN/TXT/?uri=uriserv:OJ.L_.2012.136.01.0001.01.ENG Google Scholar
51. Sanchez-Fidalgo, S, Sanchez de Ibarguen, L, Cardeno, A et al. (2012). Influence of extra virgin olive oil diet enriched with hydroxytyrosol in a chronic DSS colitis model. Eur J Nutr 51, 497506.Google Scholar
52. Rosillo, MA, Alcaraz, MJ, Sanchez-Hidalgo, M et al. (2014) Anti-inflammatory and joint protective effects of extra-virgin olive-oil polyphenol extract in experimental arthritis. J Nutr Biochem 25, 12751281.Google Scholar
53. Impellizzeri, D, Esposito, E, Mazzon, E et al. (2011) Oleuropein aglycone, an olive oil compound, ameliorates development of arthritis caused by injection of collagen type II in mice. J Pharmacol Exp Therap 339, 859869.Google Scholar
54. Martinez-Dominguez, E, de la Puerta, R & Ruiz-Gutierrez, V (2001) Protective effects upon experimental inflammation models of a polyphenol-supplemented virgin olive oil diet. Inflamm Res 50, 102106.Google Scholar
55. Bignotto, L, Rocha, J, Sepodes, B et al. (2009) Anti-inflammatory effect of lycopene on carrageenan-induced paw oedema and hepatic ischaemia-reperfusion in the rat. Br J Nutr 102, 126133.Google Scholar
56. Gong, D, Geng, C, Jiang, L et al. (2009) Effects of hydroxytyrosol-20 on carrageenan-induced acute inflammation and hyperalgesia in rats. Phytother Res 23, 646650.Google Scholar
57. Gimeno, E, de la Torre-Carbot, K, Lamuela-Raventos, RM et al. (2007) Changes in the phenolic content of low density lipoprotein after olive oil consumption in men. A randomized crossover controlled trial. Br J Nutr 98, 12431250.Google Scholar
58. Covas, MI, Nyyssonen, K, Poulsen, HE et al. (2006) The effect of polyphenols in olive oil on heart disease risk factors: a randomized trial. Ann Intern Med 145, 333341.Google Scholar
59. Covas, MI, de la Torre, K, Farre-Albaladejo, M et al. (2006) Postprandial LDL phenolic content and LDL oxidation are modulated by olive oil phenolic compounds in humans. Free Rad Biol Med 40, 608616.Google Scholar
60. Fito, M, Cladellas, M, de la Torre, R et al. (2005) Antioxidant effect of virgin olive oil in patients with stable coronary heart disease: a randomized, crossover, controlled, clinical trial. Atherosclerosis 181, 149158.Google Scholar
61. Weinbrenner, T, Fito, M, de la Torre, R et al. (2004) Olive oils high in phenolic compounds modulate oxidative/antioxidative status in men. J Nutr 134, 23142321.Google Scholar
62. Marrugat, J, Covas, MI, Fito, M et al. (2004) Effects of differing phenolic content in dietary olive oils on lipids and LDL oxidation – a randomized controlled trial. Eur J Nutr 43, 140147.Google Scholar
63. Ruiz-Canela, M & Martinez-Gonzalez, MA (2011) Olive oil in the primary prevention of cardiovascular disease. Maturitas 68, 245250.Google Scholar
64. Katan, MB, Zock, PL & Mensink, RP (1994) Effects of fats and fatty acids on blood lipids in humans: an overview. Am J Clin Nutr 60, 6 Suppl., 1017S1022S.Google Scholar
65. Chrysohoou, C, Panagiotakos, DB, Pitsavos, C et al. (2004) Adherence to the Mediterranean diet attenuates inflammation and coagulation process in healthy adults: the ATTICA Study. J Am Coll Cardiol 44, 152158.Google Scholar
66. Huang, CL & Sumpio, BE (2008) Olive oil, the Mediterranean diet, and cardiovascular health. J Am Coll Surg 207, 407416.Google Scholar
67. Covas, MI, Konstantinidou, V & Fito, M (2009) Olive oil and cardiovascular health. J Cardiovasc Pharmacol 54, 477482.Google Scholar
68. Krauss, RM & Dreon, DM (1995) Low-density-lipoprotein subclasses and response to a low-fat diet in healthy men. Am J Clin Nutr 62, 478S487S.CrossRefGoogle ScholarPubMed
69. Bos, G, Poortvliet, MC, Scheffer, PG et al. (2007) Dietary polyunsaturated fat intake is associated with low-density lipoprotein size, but not with susceptibility to oxidation in subjects with impaired glucose metabolism and type II diabetes: the Hoorn study. Eur J Clin Nutr 61, 205211.CrossRefGoogle Scholar
70. Chait, A, Brazg, RL, Tribble, DL et al. (1993) Susceptibility of small, dense, low-density lipoproteins to oxidative modification in subjects with the atherogenic lipoprotein phenotype, pattern B. Am J Med 94, 350356.Google Scholar
71. Aguilera, CM, Mesa, MD, Ramirez-Tortosa, MC et al. (2004) Sunflower oil does not protect against LDL oxidation as virgin olive oil does in patients with peripheral vascular disease. Clin Nutr 23, 673681.Google Scholar
72. Kratz, M, Cullen, P, Kannenberg, F et al. (2002) Effects of dietary fatty acids on the composition and oxidizability of low-density lipoprotein. Eur J Clin Nutr 56, 7281.Google Scholar
73. FDA (2004) FDA Allows Qualified Health Claim to Decrease Risk of Coronary Heart Disease. http://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/2004/ucm108368.htm Google Scholar
74. Fuentes, F, Lopez-Miranda, J, Perez-Martinez, P et al. (2008) Chronic effects of a high-fat diet enriched with virgin olive oil and a low-fat diet enriched with alpha-linolenic acid on postprandial endothelial function in healthy men. Br J Nutr 100, 159165.CrossRefGoogle Scholar
75. Capurso, C, Massaro, M, Scoditti, E et al. (2014) Vascular effects of the Mediterranean diet Part I: anti-hypertensive and anti-thrombotic effects. Vasc Pharmacol 63, 118126.Google Scholar
76. Khurana, S, Venkataraman, K, Hollingsworth, A et al. (2013) Polyphenols: benefits to the cardiovascular system in health and in aging. Nutrients 5, 37793827.CrossRefGoogle Scholar
77.EFSA Panel on Dietetic Products, Nutrition and Allergies (2011) Scientific Opinion on the substantiation of health claims related to polyphenols in olive and protection of LDL particles from oxidative damage (ID 1333, 1638, 1639, 1696, 2865), maintenance of normal blood HDL-cholesterol concentrations (ID 1639), maintenance of normal blood pressure (ID 3781), “anti-inflammatory properties” (ID 1882), “contributes to the upper respiratory tract health” (ID 3468), “can help to maintain a normal function of gastrointestinal tract” (3779), and “contributes to body defences against external agents” (ID 3467) pursuant to Article 13(1) of Regulation (EC) No 1924/20061. EFSA J 9, 2033.Google Scholar
78.EFSA Panel on Dietetic Products, Nutrition and Allergies (2011) Scientific Opinion on the substantiation of health claims related to olive oil and maintenance of normal blood LDL-cholesterol concentrations (ID 1316, 1332), maintenance of normal (fasting) blood concentrations of triglycerides (ID 1316, 1332), maintenance of normal blood HDL-cholesterol concentrations (ID 1316, 1332) and maintenance of normal blood glucose concentrations (ID 4244) pursuant to Article 13(1) of Regulation (EC) No 1924/2006. EFSA J 9, 2044.Google Scholar
79. Vissers, MN, Zock, PL & Katan, MB (2004) Bioavailability and antioxidant effects of olive oil phenols in humans: a review. Eur J Clin Nutr 58, 955965.Google Scholar
80. Rietjens, SJ, Bast, A & Haenen, GR (2007) New insights into controversies on the antioxidant potential of the olive oil antioxidant hydroxytyrosol. J Agric Food Chem 55, 76097614.Google Scholar
81. Mastralexi, A, Nenadis, N & Tsimidou, MZ (2014) Addressing analytical requirements to support health claims on ‘olive oil polyphenols’ (EC Regulation 432/2012). J Agric Food Chem 62, 24592461.Google Scholar
82. Romero, C & Brenes, M (2012) Analysis of total contents of hydroxytyrosol and tyrosol in olive oils. J Agric Food Chem 60, 90179022.Google Scholar
83. Teres, S, Barcelo-Coblijn, G, Benet, M et al. (2008) Oleic acid content is responsible for the reduction in blood pressure induced by olive oil. Proc Natl Acad Sci USA 105, 1381113816.Google Scholar
84. Yang, Q, Alemany, R, Casas, J et al. (2005) Influence of the membrane lipid structure on signal processing via G protein-coupled receptors. Mol Pharmacol 68, 210217.Google Scholar
85. Lahey, R, Wang, X, Carley, AN et al. (2014) Dietary fat supply to failing hearts determines dynamic lipid signaling for nuclear receptor activation and oxidation of stored triglyceride. Circulation 130, 17901799.Google Scholar
86. Rudolph, V, Rudolph, TK, Schopfer, FJ et al. (2010) Endogenous generation and protective effects of nitro-fatty acids in a murine model of focal cardiac ischaemia and reperfusion. Cardiovasc Res 85, 155166.Google Scholar
87. Coles, B, Bloodsworth, A, Clark, SR et al. (2002) Nitrolinoleate inhibits superoxide generation, degranulation, and integrin expression by human neutrophils: novel antiinflammatory properties of nitric oxide-derived reactive species in vascular cells. Circulat Res 91, 375381.Google Scholar
88. Coles, B, Bloodsworth, A, Eiserich, JP et al. (2002) Nitrolinoleate inhibits platelet activation by attenuating calcium mobilization and inducing phosphorylation of vasodilator-stimulated phosphoprotein through elevation of cAMP. J Biol Chem 277, 58325840.Google Scholar
89. Charles, RL, Rudyk, O, Prysyazhna, O et al. (2014) Protection from hypertension in mice by the Mediterranean diet is mediated by nitro fatty acid inhibition of soluble epoxide hydrolase. Proc Natl Acad Sci USA 111, 81678172.Google Scholar
90. Dhalla, NS, Temsah, RM & Netticadan, T (2000) Role of oxidative stress in cardiovascular diseases. J Hypertens 18, 655673.CrossRefGoogle ScholarPubMed
91. Sugamura, K & Keaney, JF Jr (2011) Reactive oxygen species in cardiovascular disease. Free Radic Biol Med 51, 978992.Google Scholar
92. Raedschelders, K, Ansley, DM & Chen, DD (2012) The cellular and molecular origin of reactive oxygen species generation during myocardial ischemia and reperfusion. Pharmacol Therap 133, 230255.Google Scholar
93. Ross, R (1999) Atherosclerosis – an inflammatory disease. N Engl J Med 340, 115126.Google Scholar
94. Dell'Agli, M, Fagnani, R, Galli, GV et al. (2010) Olive oil phenols modulate the expression of metalloproteinase 9 in THP-1 cells by acting on nuclear factor-kappaB signaling. J Agric Food Chem 58, 22462252.CrossRefGoogle ScholarPubMed
95. Dell'Agli, M, Fagnani, R, Mitro, N et al. (2006) Minor components of olive oil modulate proatherogenic adhesion molecules involved in endothelial activation. J Agric Food Chem 54, 32593264.Google Scholar
96. Gonzalez-Correa, JA, Navas, MD, Munoz-Marin, J et al. (2008) Effects of hydroxytyrosol and hydroxytyrosol acetate administration to rats on platelet function compared to acetylsalicylic acid. J Agric Food Chem 56, 78727876.CrossRefGoogle ScholarPubMed
97. Dollery, CM & Libby, P (2006) Atherosclerosis and proteinase activation. Cardiovasc Res 69, 625635.Google Scholar
98. Barderas, MG, Vivanco, F & Alvarez-Llamas, G (2013) Vascular proteomics. Methods Mol Biol 1000, 120.Google Scholar
99. von Zur Muhlen, C, Schiffer, E, Zuerbig, P et al. (2009) Evaluation of urine proteome pattern analysis for its potential to reflect coronary artery atherosclerosis in symptomatic patients. J Proteome Res 8, 335345.Google Scholar
100. Zimmerli, LU, Schiffer, E, Zurbig, P et al. (2008) Urinary proteomic biomarkers in coronary artery disease. Mol Cell Proteom 7, 290298.Google Scholar
101. Kalela, A, Koivu, TA, Sisto, T et al. (2002) Serum matrix metalloproteinase-9 concentration in angiographically assessed coronary artery disease. Scand J Clin Lab Invest 62, 337342.Google Scholar
102. Scoditti, E, Nestola, A, Massaro, M et al. (2014) Hydroxytyrosol suppresses MMP-9 and COX-2 activity and expression in activated human monocytes via PKCalpha and PKCbeta1 inhibition. Atherosclerosis 232, 1724.Google Scholar
103. Scoditti, E, Calabriso, N, Massaro, M et al. (2012) Mediterranean diet polyphenols reduce inflammatory angiogenesis through MMP-9 and COX-2 inhibition in human vascular endothelial cells: a potentially protective mechanism in atherosclerotic vascular disease and cancer. Arch Biochem Biophys 527, 8189.Google Scholar
104. Delles, C, Schiffer, E, von Zur Muhlen, C et al. (2010) Urinary proteomic diagnosis of coronary artery disease: identification and clinical validation in 623 individuals. J Hypertens 28, 23162322.Google Scholar
105. Monea, S, Lehti, K, Keski-Oja, J et al. (2002) Plasmin activates pro-matrix metalloproteinase-2 with a membrane-type 1 matrix metalloproteinase-dependent mechanism. J Cell Physiol 192, 160170.Google Scholar
106. Lafleur, MA, Hollenberg, MD, Atkinson, SJ et al. (2001) Activation of pro-(matrix metalloproteinase-2) (pro-MMP-2) by thrombin is membrane-type-MMP-dependent in human umbilical vein endothelial cells and generates a distinct 63 kDa active species. Biochem J 357, 107115.Google Scholar
107. Rajagopalan, S, Meng, XP, Ramasamy, S et al. (1996) Reactive oxygen species produced by macrophage-derived foam cells regulate the activity of vascular matrix metalloproteinases in vitro. Implications for atherosclerotic plaque stability. J Clin Invest 98, 25722579.Google Scholar
108. Oak, MH, El Bedoui, J, Anglard, P et al. (2004) Red wine polyphenolic compounds strongly inhibit pro-matrix metalloproteinase-2 expression and its activation in response to thrombin via direct inhibition of membrane type 1-matrix metalloproteinase in vascular smooth muscle cells. Circulation 110, 18611867.Google Scholar
109. El Bedoui, J, Oak, MH, Anglard, P et al. (2005) Catechins prevent vascular smooth muscle cell invasion by inhibiting MT1-MMP activity and MMP-2 expression. Cardiovasc Res 67, 317325.Google Scholar
110. Silva, S, Bronze, MR, Figueira, ME et al. (2015) Impact of a 6-wk olive oil supplementation in healthy adults on urinary proteomic biomarkers of coronary artery disease, chronic kidney disease, and diabetes (types 1 and 2): a randomized, parallel, controlled, double-blind study. Am J Clin Nutr 101, 4454.Google Scholar
111. Wang, M, Lamers, RJ, Korthout, HA et al. (2005) Metabolomics in the context of systems biology: bridging traditional Chinese medicine and molecular pharmacology. Phytother Res 19, 173182.Google Scholar
112. Finley Austin, MJ & Babiss, L (2006) Commentary: where and how could biomarkers be used in 2016? AAPS J 8, E185E189.CrossRefGoogle ScholarPubMed
113. Schanstra, JP & Mischak, H (2015) Proteomic urinary biomarker approach in renal disease: from discovery to implementation. Pediatr Nephrol 30, 713725.Google Scholar
114. Mischak, H, Allmaier, G, Apweiler, R et al. (2010) Recommendations for biomarker identification and qualification in clinical proteomics. Sci Transl Med 2, 46ps42.Google Scholar
115. Stegemann, C, Didangelos, A, Barallobre-Barreiro, J et al. (2013) Proteomic identification of matrix metalloproteinase substrates in the human vasculature. Circ-Cardiovasc Gene 6, 106117.Google Scholar
116. Mischak, H & Rossing, P (2010) Proteomic biomarkers in diabetic nephropathy – reality or future promise? Nephrol Dial Transplant 25, 28432845.Google Scholar
117. Lescuyer, P, Hochstrasser, D & Rabilloud, T (2007) How shall we use the proteomics toolbox for biomarker discovery? J Proteome Res 6, 33713376.Google Scholar
118. Wisniewski, JR, Zougman, A, Nagaraj, N et al. (2009) Universal sample preparation method for proteome analysis. Nat Methods 6, 359362.Google Scholar
119. Husi, H, Van Agtmael, T, Mullen, W et al. (2014) Proteome-based systems biology analysis of the diabetic mouse aorta reveals major changes in fatty acid biosynthesis as potential hallmark in diabetes mellitus-associated vascular disease. Circ-Cardiovasc Gene 7, 161170.Google Scholar
120. Thongboonkerd, V, McLeish, KR, Arthur, JM et al. (2002) Proteomic analysis of normal human urinary proteins isolated by acetone precipitation or ultracentrifugation. Kidney Int 62, 14611469.Google Scholar
121. Thongboonkerd, V & Malasit, P (2005) Renal and urinary proteomics: current applications and challenges. Proteomics 5, 10331042.Google Scholar
122. Mischak, H, Kolch, W, Aivaliotis, M et al. (2010) Comprehensive human urine standards for comparability and standardization in clinical proteome analysis. Proteom Clin Appl 4, 464478.Google Scholar
123. Albalat, A, Franke, J, Gonzalez, J et al. (2013) Urinary proteomics based on capillary electrophoresis coupled to mass spectrometry in kidney disease. Methods Mol Biol 919, 203213.Google Scholar
124. Good, DM, Zurbig, P, Argiles, A et al. (2010) Naturally occurring human urinary peptides for use in diagnosis of chronic kidney disease. Mol Cell Proteom 9, 24242437.Google Scholar
125. Metzger, J, Kirsch, T, Schiffer, E et al. (2010) Urinary excretion of twenty peptides forms an early and accurate diagnostic pattern of acute kidney injury. Kidney Int 78, 12521262.Google Scholar
126. Dawson, J, Walters, M, Delles, C et al. (2012) Urinary proteomics to support diagnosis of stroke. PLoS ONE 7, e35879.Google Scholar
127. Mischak, H & Schanstra, JP (2011) CE-MS in biomarker discovery, validation, and clinical application. Proteom Clin Appl 5, 923.Google Scholar
128. Coon, JJ, Zurbig, P, Dakna, M et al. (2008) CE–MS analysis of the human urinary proteome for biomarker discovery and disease diagnostics. Proteom Clin Appl 2, 964.Google Scholar
129. Lee, RT & Libby, P (1997) The unstable atheroma. Arterioscler Thromb Vasc Biol 17, 18591867.Google Scholar
130. Zurbig, P, Jerums, G, Hovind, P et al. (2012) Urinary proteomics for early diagnosis in diabetic nephropathy. Diabetes 61, 33043313.Google Scholar
131. de Roos, B, Zhang, X, Rodriguez Gutierrez, G et al. (2011) Anti-platelet effects of olive oil extract: in vitro functional and proteomic studies. Eur J Nutr 50, 553562.Google Scholar
132. Arbones-Mainar, JM, Ross, K, Rucklidge, GJ et al. (2007) Extra virgin olive oils increase hepatic fat accumulation and hepatic antioxidant protein levels in APOE-/- mice. J Proteome Res 6, 40414054.Google Scholar
133. Ge, Y & Wang, TJ (2012) Identifying novel biomarkers for cardiovascular disease risk prediction. J Intern Med 272, 430439.Google Scholar
134. Sharma, P, Cosme, J & Gramolini, AO (2013) Recent advances in cardiovascular proteomics. J Proteomics 81, 314.Google Scholar
135. Fliser, D, Novak, J, Thongboonkerd, V et al. (2007) Advances in urinary proteome analysis and biomarker discovery. J Am Soc Nephrol 18, 10571071.Google Scholar
136. Julian, BA, Suzuki, H, Suzuki, Y et al. (2009) Sources of urinary proteins and their analysis by urinary proteomics for the detection of biomarkers of disease. Proteom Clin Appl 3, 10291043.CrossRefGoogle ScholarPubMed
137. Perona, JS, Cabello-Moruno, R & Ruiz-Gutierrez, V (2006) The role of virgin olive oil components in the modulation of endothelial function. J Nutr Biochem 17, 429445.Google Scholar
138. Guillen, N, Acin, S, Navarro, MA et al. (2008) Squalene in a sex-dependent manner modulates atherosclerotic lesion which correlates with hepatic fat content in apoE-knockout male mice. Atherosclerosis 197, 7283.Google Scholar
139. Monte, E & Vondriska, TM (2014) Epigenomes: the missing heritability in human cardiovascular disease? Proteomics Clin Appl 8, 480487.Google Scholar
140. Camargo, A, Ruano, J, Fernandez, JM et al. (2010) Gene expression changes in mononuclear cells in patients with metabolic syndrome after acute intake of phenol-rich virgin olive oil. BMC Genomics 11, 253.Google Scholar
141. Konstantinidou, V, Covas, MI, Munoz-Aguayo, D et al. (2010) In vivo nutrigenomic effects of virgin olive oil polyphenols within the frame of the Mediterranean diet: a randomized controlled trial. FASEB J 24, 25462557.Google Scholar
142. Vazquez-Fresno, R, Llorach, R, Urpi-Sarda, M et al. (2015) Metabolomic pattern analysis after mediterranean diet intervention in a nondiabetic population: A 1- and 3-year follow-up in the PREDIMED study. J Proteome Res 14, 531540.Google Scholar
143. Noratto, GD, Angel-Morales, G, Talcott, ST et al. (2011) Polyphenolics from acai (Euterpe oleracea Mart.) and red muscadine grape (Vitis rotundifolia) protect human umbilical vascular Endothelial cells (HUVEC) from glucose- and lipopolysaccharide (LPS)-induced inflammation and target microRNA-126. J Agric Food Chem 59, 79998012.Google Scholar
144. Milenkovic, D, Jude, B & Morand, C (2013) miRNA as molecular target of polyphenols underlying their biological effects. Free Radic Biol Med 64, 4051.Google Scholar
145. Garcia-Gonzalez, DL & Aparicio, R (2010) Research in olive oil: challenges for the near future. J Agric Food Chem 58, 1256912577.Google Scholar
146. Martin-Pelaez, S, Covas, MI, Fito, M et al. (2013) Health effects of olive oil polyphenols: recent advances and possibilities for the use of health claims. Mol Nutr Food Res 57, 760771.Google Scholar
147. Dunn, MJ (2013) Proteomics clinical applications reviews 2013. Proteomics Clin Appl 7, 47.Google Scholar
148. Levi, B & Werman, MJ (1998) Long-term fructose consumption accelerates glycation and several age-related variables in male rats. J Nutr 128, 14421449.Google Scholar
149. Kontogianni, VG, Charisiadis, P, Margianni, E et al. (2013) Olive leaf extracts are a natural source of advanced glycation end product inhibitors. J Med Food 16, 817822.Google Scholar
150. Vlassopoulos, A, Lean, ME & Combet, E (2014) Protein-phenolic interactions and inhibition of glycation – combining a systematic review and experimental models for enhanced physiological relevance. Food and Funct 5, 26462655.Google Scholar
Figure 0

Table 1. Main classes of phenolic compounds in virgin olive oil

Figure 1

Fig. 1. Chronic inflammation model and impact on rats paw oedema. ANOVA, *P < 0·001 v. positive control – Rheumatoid Arthritis, +P < 0·01 v. Refined Olive Oil; OHTYR, hydroxytyrosol. Adapted from Silva et al.(49).

Figure 2

Table 2. Scores of CAD, CKD and diabetes proteomic biomarkers at baseline, middle (3 weeks) and end of intervention (6 weeks)