Next Article in Journal
Diallyl Trisulfide Induces Apoptosis in Breast Ductal Carcinoma In Situ Derived and Minimally Invasive Breast Cancer Cells
Next Article in Special Issue
Molecular Mechanisms Underlying the Effects of Olive Oil Triterpenic Acids in Obesity and Related Diseases
Previous Article in Journal
Amino Acid-Related Metabolic Signature in Obese Children and Adolescents
Previous Article in Special Issue
Protective Effects and Benefits of Olive Oil and Its Extracts on Women’s Health
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Therapeutic Properties and Use of Extra Virgin Olive Oil in Clinical Nutrition: A Narrative Review and Literature Update

by
Andrés Jiménez-Sánchez
1,2,*,
Antonio Jesús Martínez-Ortega
2,3,
Pablo Jesús Remón-Ruiz
1,2,
Ana Piñar-Gutiérrez
1,2,
José Luis Pereira-Cunill
1,2 and
Pedro Pablo García-Luna
1,2,*
1
Unidad de Gestión Clínica de Endocrinología y Nutrición, Hospital Universitario Virgen del Rocío, Avda. Manuel Siurot s/n, 41013 Seville, Spain
2
Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Avda. Manuel Siurot s/n, 41013 Seville, Spain
3
Unidad de Gestión Clínica de Endocrinología y Nutrición, Hospital Torrecárdenas, C. Hermandad de Donantes de Sangre, s/n, 04009 Almería, Spain
*
Authors to whom correspondence should be addressed.
Nutrients 2022, 14(7), 1440; https://doi.org/10.3390/nu14071440
Submission received: 17 February 2022 / Revised: 24 March 2022 / Accepted: 25 March 2022 / Published: 31 March 2022
(This article belongs to the Special Issue Virgin Olive Oil and Health)

Abstract

:
Extra virgin olive oil (EVOO) is a cornerstone of the Mediterranean diet (MedD). In this narrative review, we synthesize and illustrate the various characteristics and clinical applications of EVOO and its components—such as oleic acid, hydroxytyrosol, and oleuropein—in the field of clinical nutrition and dietetics. The evidence is split into diet therapy, oleic acid-based enteral nutrition formulations and oral supplementation formulations, oleic acid-based parenteral nutrition, and nutraceutical supplementation of minor components of EVOO. EVOO has diverse beneficial health properties, and current evidence supports the use of whole EVOO in diet therapy and the supplementation of its minor components to improve cardiovascular health, lipoprotein metabolism, and diabetes mellitus in clinical nutrition. Nevertheless, more intervention studies in humans are needed to chisel specific recommendations for its therapeutic use through different formulations in other specific diseases and clinical populations.

1. Introduction

Olive oil (OO) is the juice of the Olea europaea fruit. When it is obtained by physical processes and without any further treatment other than washing, decantation, centrifugation, and filtration, it is considered virgin olive oil. When VOO reaches the highest standards set by the EU Commission Regulation No 2568/91 [1] and the Trade Standard of the International Olive Council (IOC) [2], it can be classified as extra virgin olive oil (EVOO). Both regulations share the following requirements in chemical composition to define an OO as EVOO: a free acidity (expressed as percentage of free oleic acid) ≤0.8°%, a peroxide value ≤20 mEq O2/kg, UV spectrometry values that reflect minimal oxidation (K270 ≤ 0.22; K232 ≤ 2.50; ∆K ≤ 0.01), a favorable organoleptic assessment (median of 0 defects and median of “fruity” flavor >0), and a percentage of fatty acid ethyl esters (FAEEs) ≤30 mg/kg of EVOO (in crops harvested since 2015). The IOC also has a higher FAEE threshold (≤35 mg/kg of EVOO) and includes further qualifying criteria, such as ≤0.20% of moisture and volatile matter in the oil, ≤0.10% insoluble impurities in light petroleum, and low levels of trace metals (≤3.0 for iron and ≤0.1 for copper). If an OO complies with all the quality criteria to be defined as EVOO, then purity criteria must be met to maintain this certification, such as a ≤0.05 stigmadiene content (mg/kg of EVOO), small amounts of trans-isomers of unsaturated fatty acids (≤0.05 for the sum of C18:1 and ≤0.05 for the sum of C18:2 and C18:3), fatty acid content under specific thresholds (for myristic, linolenic, arachidic, eicosenoic, behenic, and lignoceric acids), ∆ECN42 (the difference between the theoretical and measured ECN42 by HPLC) ≤|0.2|, levels of 2-glyceryl-monopalmitate (depending on the palmitic acid percentage), certain requirements regarding sterols (a total sterol content ≥1000 mg/kg of EVOO, fixed maximum percentages for cholesterol, brassicasterol, campesterol, stigmasterol, ∆-7-stigmasterol, and the apparent sum of β-sitosterol, as well as a sum of erythrodiol and uvaol ≤4.5%), and total waxes (C42 + C44 + C46) ≤150 mg/kg of EVOO. The IOC has extra criteria, such as fixed ranges of other free fatty acids (palmitic, palmitoleic, heptadecanoic, heptadecenoic, stearic, and linoleic) and a higher threshold for eicosenoic acid (≤0.50 instead of ≤0.40) and total unsaponifiable matter (≤15 g/kg of EVOO).
EVOO has been a cornerstone in the civilizations that have developed over millennia in the Mediterranean Basin. Since antiquity, peoples of the Mare Nostrum have been aware of its beneficial properties for health and have used this product in their gastronomy and pharmacopoeia [3,4,5]. The aim of this narrative review is to synthesize and illustrate the various characteristics, clinical applications, and mechanisms of action of EVOO and its components in the field of clinical nutrition and dietetics. More specifically, the following minor topics are discussed: diet therapy, enteral nutrition, oral nutritional supplementation, parenteral nutrition, and nutraceutical supplements. Our focus is on intervention studies in humans. When not available, evidence from other type of studies and/or animal models is reviewed.

2. Materials and Methods

Each minor topic was individually explored in a comprehensive narrative literature review during October–November 2021 using PubMed and Web of Science search engines. MeSH terms were used when applicable, and different search strategies were undertaken combining the following keywords: Mediterranean diet”, “extra virgin olive oil”, “olive oil”, “enteral nutrition”, “parenteral nutrition”, “oleic acid”, “tyrosol”, hydroxytyrosol”, “phenolic”, “oleuropein”, “oleocanthal”, “olive leaf”, “olive mill wastewater”, “antioxidant”, “inflammat*”, “glucose”, “cholesterol”, “metabolic syndrome”, “diabetes”, “obesity”, “lipoprotein”, “cancer”, “vascular”, “heart”, “osteoporosis”, “bone”, “prostat*”, “intestinal”, “neuro*”, “Alzheimer”, “fatty liver”, “elder*”, “microbio*”, and “arthritis”. Our primary search was conducted in humans and intervention studies. If no evidence was available, then a secondary search was conducted selecting other type of studies and/or animal models. Finally, information was pooled and synthesized.

3. Results

3.1. Characteristics of EVOO

3.1.1. Chemical Composition

The chemical composition of EVOO depends, among others, on the place of production, cultivar, and degree of ripeness of the olives, which can vary significantly [6,7,8,9,10,11]. From a chemical perspective, EVOO can be divided into three main groups of substances:
1.
Saponifiable fraction. Accounts for 98.5–99.5% in total weight [6,7,8,9,12,13,14]. It is mainly made up of the following:
(1)
Free fatty acids. The most frequent are palmitic (C16:0), palmitoleic (C16:1), stearic (C18:0), oleic (C18:1), linoleic (C18:2), and linolenic (C18:3) acids. Myristic (C14:0), heptadecanoic, and eicosanoic acids are also present in small amounts [6,7,8,9,12,13,14]. See Table S1 for further information on the percentages of fatty acids present in EVOO from the main cultivars in the Mediterranean Area [6,15,16,17,18,19,20,21,22,23,24,25,26].
(2)
Triglycerides (TAGs) account for 99% of the saponifiable fraction. The most prevalent (43.5%) is triolein, composed of three chains of acid. Other esterification possibilities are one palmitic acid in position sn-3 and two oleic acids in sn-1 and sn-2 positions (accounting for 18.4% of TAGs), as well as one linoleic acid in sn-2 position bordered by two oleic acids (6.8%) [6,7,8,9,12,13,14].
(3)
Partial acylglycerols (diacylglycerols, DAGs, or monoacylglycerols, MAGs). Their presence in EVOO is due to incomplete triacylglycerol biosynthesis or hydrolytic reactions. A good-quality oil should have no more than 1–2.8% DAGs, with higher values being considered an indication of poor processing or storage. The 1,2-DAGs—typical of freshly produced oil—gradually transform into the more stable 1,3-DAGs. Therefore, the 1,3-DAG/1,2-DAG ratio is considered a reliable parameter to determine the age of EVOO. MAGs are much less prevalent (less than 0.25%) and vary widely according to the production area: Spanish, Portuguese, Italian, or Greek OOs are richer in oleic-based MAGs, while Tunisian OOs are richer in palmitic- and linoleic-based MAGs [6,7,8,9,12,13,14].
2.
Unsaponifiable fraction. Constitutes 0.5–1.5% of EVOO. It can be grouped into the following:
(1)
Tocopherols. The most abundant is α-tocopherol (150 to 250 mg / kg of EVOO). The optimal ratio of vitamin E to polyunsaturated fatty acids of 1.5–2.0. α-Tocopherol is able to neutralize free radicals and prevent peroxidation of membrane lipids and low-density lipoproteins (LDLs). β-tocopherol (15–20%) and γ-tocopherol (7–23%) are present in smaller proportions [6,7,8,9,12,13,14].
(2)
Phytosterols. The amount varies from 100 to 250 mg/100 g of EVOO. Sterols are categorized into 4,4-dimethylsterols or triterpenic alcohols (two methyl groups at the C-4 position), 4-monomethylsterols or methylsterols (one methyl group at the C-4 position), and 4-desmethylsterols or phytosterols (no methyl group), the latter being the main sterols in EVOO [6]. EVOO has many phytosterols, such as β-sitosterol, campesterol, ∆5-avenasterol, stigmasterol, cholesterol, cholesterol, brassicasterol, sitostanol, ergosterol, campestanol, ∆7-cholestenol, ∆7-avenasterol, ∆7-stigmasterol, ∆7-campesterol, ∆5,24-stigmastadienol, ∆5,23-stigmastadienol, ∆7,24-ergostadienol, ∆7,22-ergostadienol, 22,23-dihydrobrassicasterol, and 24-methylene-cholesterol [27]. The main one is β-sitosterol, accounting for 75–90% of total phytosterols in EVOO [28]. Different vegetable oils have different proportions of phytosterols, such as higher campesterol for seed oil, and Δ7-stigmastenol for sunflower and saffron oil [29]. The IOC and EU Commission define the sterol “fingerprint” of EVOO as that with >93.0% of apparent β-sitosterol, which is the sum of the percentages of β-sitosterol plus Δ5,23-stigmastadienol, sitostanol, Δ5-avenasterol, Δ5,24-stigmastadienol, and chlerosterol [1,2]. For example, EVOOs from Picholine and Sinopolese cultivars in Italy have shown high and steady amounts of β-sitosterol through different seasons [30]. On the other hand, these regulations also limit the relative proportion of erythrodiol and uvaol as 4.5% [1,2], as excessive amounts of these triterpenes may be a sign of adulteration with olive pomace oil or grape seed oil. For example, EVOOs from Manzanilla Cacereña in Spain are well under this limit [31].
3.
Other minor components are the following:
(1)
Phenolic compounds. They are divided into phenolic acids (p-coumaric, gallic, vanillic, and caffeic acid, as well as others), flavonoids (luteolin, apigenin, and their derivatives), lignans (pinoresinol and 1-acetoxypinoresinol), isochromans (1-phenyl-6,7-dihydroxy-isochroman, and 1-(30-methoxy-40-hydroxy)phenyl-6,7-dihydroxy-isochroman), secoiridoids (demethyloleuropein, oleuropein, ligstroside, and their aglycone derivatives), and phenolic alcohols (tyrosol, hydroxytyrosol, and homovanillyl alcohol) [27]. Tyrosol, 5-hydroxytyrosol, and its elenoic acid ester—oleuropein—have potent antioxidant activity in vitro and in vivo, along with anti-inflammatory activity. Combining an in vivo and in vitro approach, it was demonstrated that oleocanthal (a secoiridoid derivative) is a potent activator of the transient receptor potential ankyrin 1 (TRPA1) ion channel expressed in trigeminal sensory neurons. When activated, TRPA1 creates a Ca2+-mediated cell depolarization interpreted as a pungent sensation that triggers a defensive cough response, to prevent the entrance of potentially harmful compounds to the airway. This effect takes place mainly in the pharynx—where the density of TRPA1 receptors doubles that of the tongue—and independently of the lipophilicity of the food matrix [32]. Smaller amounts of simpler phenolic compounds—such as caffeic, vanillic, and ferulic acids—have the role of protecting and enhancing the bioavailability of α-tocopherol [6,7,8,9,12,13,14,33,34]. See Table S2 for further information regarding antioxidants in the unsaponifiable fraction of EVOO from the main cultivars in the Mediterranean Area [17,19,20,21,22,23,25,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51].
(2)
Chlorophyllic and carotenoid pigments. Present in small amounts, they color EVOO. Of note are β-carotene and lycopene. [6,7,8,9,12,13,14].
(3)
Volatile compounds. More than 100 volatile compounds are responsible for the characteristic odor of EVOO. These are small molecules (<300 Da) that vaporize at room temperature [52] and are present in EVOO’s headspace in higher concentrations than lower-quality OOs [53,54]. The major volatile compounds in EVOO are C6 and C5 alcohols and aldehydes formed from polyunsaturated fatty acids (PUFAs) through the lipoxygenase pathway. Although the former are present in higher quantities, the latter have lower detection thresholds in olfactory receptors [55]. 2(E)-Hexenal is the main C6 aldehyde in EVOO, giving “green” and “fruity” notes to EVOO, while C5 aldehydes and alcohols give pungent sensations [56] that may be appreciated by consumers [57]. The concentration of volatile compounds in EVOO depends on the cultivar and geographical area where the olive tree lives [58]; hence, EVOOs with different origins can be sorted by their characteristic profiles [59]. Using this approach, a study clustered 18 Italian cultivars into eight groups on the basis of their distinct levels of ethanol, 2-methyl propanol, pentanol, cis-2-penten-1-ol, cis-3-hexenol, and octanol [60]. Unripe fruit is associated with hexanal, trans-3-hexexnol, cis-3-hexen-1-ol, and cis-2-hexenol, associated with compelling sensory attributes such as “fruity”, “grassy”, and “pungent” [61]. Higher oxygenation [62] and temperatures around 20 °C during malaxation [63], as well as an adequate storage temperature [64] and minimization of storage time [62], maximize the amount and quality of EVOO’s volatiles.
(4)
Aliphatic alcohols or fatty alcohols (FAL). They are useful to classify different categories of OO in relation to the wax content of the product. Even-chain FALs (ECFALS) are more frequent than odd-chain FALs (OCFALs). Policosanol is the mixture of the most common ECFALs: docosanol (C22), tetracosanol (C24), hexacosanol (C26), and octacosanol (C28). Policosanol content in EVOO is regulated by the IOC and EU Commission [1,2]. According to previous studies, hexacosanol seems to be the most abundant FAL in Croatian [65], Greek [66], Italian [67], Spanish [68], and Tunisian [69] cultivars. The FAL content of EVOO depends mainly on the cultivar [67]. Both OCFAL and ECFAL quantities decrease with the increased ripening of the fruit, but there are differences between studies regarding the FAL most affected by ripening. It was hexacosanol in Italian cultivars [70] and pentacosanol in Croatian cultivars [65].
(5)
Alkanes and alkenes. These linear hydrocarbons occur naturally in EVOO as precursors during wax biosynthesis. Alkanes and alkenes are mostly odd-numbered, as they come from the loss of a carboxyl group in very-long-chain acids (VLCA), which are initially formed by the fatty-acid elongation complex in the endoplasmic reticulum (using palmitic and stearic acids as substrates) [71]. The alkane content and profile in EVOO depend on its cultivar [72], harvest time [73], and geographical region: the most frequent n-alkanes are n-C25 in continental Europe, n-C23 in north Africa and southern Italy, and n-C29 in the Iberian Peninsula [73,74]. This different geographical location may be related to meteorological conditions, as the intensity of precipitation is associated with larger alkanes and alkenes [75]. In addition to their usefulness in tracing the geographical origin and cultivar of EVOO, alkanes and alkenes are also useful for detecting counterfeits, because vegetable oils, as well as olive oils of different qualities, have distinctive fingerprints regarding these compounds. Alkanes are absorbed and then metabolized to FALs in the small intestine [72].

3.1.2. Production of EVOO and Its Minor Components

The profile and concentration of compounds in the unsaponifiable fraction of EVOO depend on the cultivar [76], irrigation [77], mode of cultivation [47], and ripening of the fruit [78]. Cultivar, time, and growing conditions can modify the expression of the β-glucosidase, a main enzyme in the synthesis of secoiridoid derivatives in Olea europaea [79]. Malaxation temperature [46] and storage temperature [80,81] may also affect the phenolic content of EVOO. During storage, there is time-dependent conversion of their secoiridoids and derivatives (mainly oleacein and oleocanthal) into byproducts such as hydroxytyrosol and oleocanthalic acid. During this process, different cultivars and geographical areas may present different transformation patterns [82]. There is no actual consensus on the definition of EVOO with high phenolic content. Some authors have proposed EVOOs with >500 mg of polyphenols per kg of product [76]. Polyphenols and other minor components can be extracted from olive pomace, olive mill wastewater, and olive leaves, creating a circular agriculture system. The phenolic profile of these non-oil sources of polyphenols is distinctive. Amongst them, olive tree leaves have the highest yield of phenols due to their greater content of secoiridoids (mainly oleuropein) [83,84]. The major determinant of the phenolic yield of olive tree leaves is the type of cultivar [85]. Other factors are the color [86] and dryness of the leaves [87], as well as the geographical location of the olive tree and leaf collection time [88].

3.1.3. Cooking with EVOO

Crude EVOO is the ideal food matrix for absorption of its hydroxytyrosol in the intestines, ensuring maximum concentrations in blood [89,90]. Among heat-based cooking methods, frying and steaming may preserve or enhance EVOO’s antioxidant capacity, while sautéing, pressure cooking, boiling, and microwaving may lead to significant reductions in its total phenolic content [91,92]. When boiling vegetables in a pressure cooker, phenolic compounds are transferred to cooking water from both EVOO and vegetables, with a maximum transfer in the case of eggplant, tomato, and pumpkin [92]. Therefore, incorporating this cooking water into the final recipe—instead of discarding it—may be of interest to the consumer. During boiling, secoiridoids are transformed into tyrosol, which is increased, while tocopherol content is lowered. Interestingly, hydroxytyrosol content may increase for the first hours when using EVOO of the Picual cultivar, while the intensity of loss of total phenolic content may be cultivar-dependent [93].
During frying, hydroxytyrosol and tyrosol may have the lowest thermal resistance, and deep frying and pan frying may affect the type of phenolic compounds lost during heating [94]. When frying with EVOO, temperature seems a much more important factor than time; approximately, its phenolic content can be decreased by 40% at 120 °C and 75% at 170 °C with 15–60 min of cooking [95], while its scavenging activity may be halved at approximately 180 °C after just 15 min [96]. During intensive heating (180–220 °C for 30–120 min), EVOO can degrade more than other vegetable oils such as pomace, soybean, and palm oil, developing higher levels of peroxides and free fatty acids, as well as undergoing the highest relative loss in phenolic content [97]. Nevertheless, EVOO is more resistant to oxidation [98] and may produce higher yields of tocopherols at 180 °C than other monounsaturated fatty acid (MUFA)-rich cooking oils [99]. In this regard, using OO to fry blue fish (another important component of theMedD) may transfer significant amounts of squalene to the food matrix and protect its ω-3 content better than sunflower oil [100]. In this process, the loss of hydroxytyrosol and its secoiridoids seems to be greater than the loss of tyrosol and its derivatives [101]. Interestingly, the loss of total phenolic content under frying seems to take place early during cooking and stabilize from 60 min of cooking [102]. Under these conditions, 2(E)-hexenal content is drastically reduced in a time- and temperature-dependent manner, while new and undesirable volatiles appear due to oleic acid oxidation, thus reducing its sensory properties [103].
In a head-to-head study, microwaving for 5 min at 500 W did not generate significant losses in total phenolic content, while baking at 180 °C for 45 was associated with an 11% loss. In any case, both methods were less detrimental to the phenolic content than boiling at 100 °C for 40–80 min or frying at 180 °C for 1–5 h [1]. Nevertheless, microwaving EVOO at 500 W for longer periods of time is not recommended, as it may generate quantitatively greater oxidation and loss of phenolic content [104], as well as faster flavor deterioration through the formation of acrolein. Nevertheless, these results are limited by certain experimental conditions (microwave and oven samples did not have the same volume) [105]. Although lipid oxidation takes place during baking, the substitution of margarine for EVOO in bakery products can prevent triglyceride oxidation and increase its shelf life [106] without substantially altering the sensory and physical properties of the food [107]. This may be due to a greater presence of antioxidant molecules in the final product. In this regard, oleuropein levels may be higher in starch matrices that have been baked instead of boiled, while having greater bio-accessibility than lipid-based carriers [90]. A study on Caco cells suggested there may not be a close correlation between the total phenolic content of a certain polyphenol-enriched bakery product and its ability to exert anti-inflammatory properties, as it depends on the different bio-accessibility and bioavailability of each of these compounds. This may depend on the source of phenolic compounds, the type of molecule, the characteristics of the fermentation process, and the selected time and temperature during baking [108].
Consuming EVOO with egg or whey protein [109] and fortifying foods with polyphenols or secoiridoids may be of interest to people with an aversion to the bitter taste of these compounds [110]. Liquid preparations with oleuropein in comparison with capsules have shown a higher plasma peak of oleuropein and a faster absorption of hydroxytyrosol metabolites [111], which could explain the greater therapeutic efficacy associated with this pharmaceutical preparation [112]. Polyphenol absorption occurs in proximal sections of the small intestine in humans [113]. Drugs can interfere with the absorption of β-sitosterol. Ezetimibe is an approved treatment for phytosterolemia [114] that inhibits β-sitosterol absorption either as monotherapy [115] or in combination with statins [116]. β-Sitosterol absorption increases with atorvastatin [117].

3.1.4. Absorption and Metabolization of EVOO and Its Minor Components

Secoiridoid derivatives and phenolic acids are hydroxylated in the mouth, stomach, and duodenum, releasing phenolic alcohols (tyrosol and hydroxytyrosol) [45] This transformation can be enhanced in vivo in mice with the addition of the probiotic Lactobacillus plantarum [118]. Interestingly, the bio-accessibility (fraction of bioactive compounds freed from the food matrix and available for their intestinal absorption) of phenolic compounds may be not only dose-dependent, but also contingent upon the different stability of these compounds with regard to digestive tract conditions and the characteristics of the food matrix. In a study with in vitro enzymatic digestion of EVOOs from five different cultivars, Sevillana had the lowest total phenolic content (571 ± 1.6 nmol/g of EVOO) and the second lowest hydroxytyrosol content (6.11 ± 0.0 nmol / g of EVOO), yet the highest bio-accessibility index for total phenolic compounds (36%) and hydroxytyrosol (2452%) [45]. Regarding the absorption of olive leaf extracts, a study found a total phenolic content of 94 ± 2 mg/g of dry extract, with oleuropein as its major constituent (76.1 ± 0.8 mg/g of dry extract). In this case, oleuropein also presented the highest bio-accessibility index (109.86%) after 240 min of enzymatic digestion [119]. Further studies focusing on the bio-accessibility of polyphenols after enzymatic digestion of different Olea europaea products have been extensively reviewed elsewhere [120]. Oleuropein and hydroxytyrosol antioxidant activity seemed to be similar in an ex vivo study in humans [121], while both seemed to be more potent than homovanillic alcohol in vitro [122]. Hydroxytyrosol plasma concentrations diminish within approximately 1 h after EVOO ingestion, as they are transformed into glucuronidated forms, which have similar antioxidant potency to tyrosol [123]. In humans, the absorbed dietary tyrosol is metabolized to hydroxytyrosol and homovanillic alcohol in the liver by the CYP2A6 and CYP2D6 enzymes [124].
Although phytosterol and phytostanol absorption in humans is poor, ∆5-phytosterols (sitosterol and campesterol) may have higher absorption and lower turnover rates than 5α-phytostanols (sitostanol and campestanol) [125]. This absorption takes place using the Niemann-Pick 1C-like 1 transporter (NPC1L1), the same route used by cholesterol. Therefore, the inhibition of cholesterol absorption is competitive and dose-dependent. Afterward, phytosterols are then esterified within enterocytes and incorporated in chylomicrons to reach the liver. Finally, hepatocytes secrete these phytosterols into the bile through ABCG5/G8 transporters [126]. Phytosterol composition, the characteristics of its food matrix, clinical conditions that affect cholesterol metabolism such as type 2 diabetes mellitus (DM2), and single-nucleotide polymorphisms (SNPs) in NPC1L1 and apolipoprotein E (ApoE) may modify both their bioavailability and their efficacy [126,127].

3.1.5. Relevant Effects of EVOO on Pathophysiological Processes

Although EVOO has a high concentration of MUFA (oleic acid) compared to other fatty acids, EVOO’s minor components appear to be responsible for the greatest number of its beneficial effects. There is abundant evidence about the mechanisms of action and the therapeutic potential or capacity of these minor components, mainly in animal models or ex vivo studies in humans. The following is a very brief summary of the most clinically relevant evidence on the effects of EVOO:
  • Anti-inflammatory. Consumption of EVOO in humans has shown significant reductions in inflammatory and cell adhesion mediators such as, but not limited to, vascular cell adhesion molecule 1 (VCAM-1), intercellular adhesion molecule 1 (ICAM-1), interleukin 6 (IL-6), and C-reactive protein (CRP), both pre-prandially and postprandially. After consuming a meal rich in EVOO, reduced levels of thromboxane B2, leukotriene B 4, tumor necrosis factor-α (TNF-α) mRNA, arachidonic acid synthesis, and natural killer (NK) cell activity have been observed in ex vivo human studies. These effects could be mediated—among other mechanisms—by direct activation of peroxisome proliferator-activated receptors- α/γ (PPAR-α/γ), repression of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), inhibition of cyclooxygenases 1 and 2 (COX1-COX2), lipoxygenases, MAP kinases (MAPK), and Janus kinases / signal transducer and activator of transcription proteins (JAK/STAT) pathways [12,14,128,129,130].
  • Coagulation. EVOO appears to inhibit platelet adhesion by decreasing the von Willebrand factor expression, and regular consumption of EVOO may also reduce factor VII activation and circulating plasminogen activator inhibitor-1 (PAI-1) levels. [12,13,128].
  • Vasodilation. In murine models, triterpenes generate aortic vasodilation in a NO-dependent and COX-independent manner [131] that is endothelium-mediated [132]. EVOO’s minor components may also negatively regulate ACE and NR1H2 genes [130].
  • Improved endothelial health and lipoprotein composition. The combination of the previously described properties of EVOO allows it to improve endothelial function [12,14] and reduce the risk of atheromatous plaque development or complication [12,13,14,128,130]. Regular consumption of EVOO increases the high-density lipoprotein (HDL) fraction and the phenolic content of LDLs, improving their resistance to oxidation and, thus, reducing their atherogenic capacity. Although mixed, some evidence indicates that EVOO may also reduce abdominal adiposity [130].
  • Control of cell proliferation. Phenolic compounds, such as oleocanthal, are able to act on the expression of genes that control the proliferation, apoptosis, and differentiation of cancer cells [13,33,34,133]. There is in vitro evidence showing that polyphenols in EVOO alter microRNAs in cancer cells, while hydroxytyrosol appears to exert strong antiproliferative effects in human colon adenocarcinoma cells [14].
  • Microbiological properties. Oleuropein has in vitro antimicrobial activity against multiple Gram-positive and Gram-negative bacteria, mycoplasma, and viruses [134]. Some of these microorganisms are of particular interest, such as methicillin-resistant Staphylococcus aureus (MRSA) [135]. Oleuropein caused improved survival in an animal model of multidrug-resistant Pseudomonas aeruginosa sepsis, explained by ex vivo stimulation of phagocytosis and inhibition of proinflammatory cytokines [136]. Hydroxytyrosol has shown ex vivo and in vivo anti-HIV-1 activity in rabbits, with no associated toxicity [137]. EVOO consumption has been linked to increased proportions of Clostridium cluster XIVa and Bifidobacterium, Parascardovia, Bacteroides fragilis, and Lactobacillus johnsonii, as well as a reduction of the presence of Lactobacillus animalis, Lactobacillus taiwanensis, Lactococcus spp., Proteobacteria, Deferribacteres, and Rikenella [130].
  • Bone homeostasis. In an animal model, both hydroxytyrosol and tyrosol appeared to have a positive effect on bone health [128]. In vitro studies in the human osteoblast cell line MG63 have shown differential effects of caffeic, ferulic and coumaric acid, apigenin and luteolin on gene expression [138,139]. The addition of different varieties of EVOO—with unique compositions in minor components—had divergent effects on the differentiation, antigenic expression, and antigenic capacity of this cell line [140].
  • Cardiac function. In animal models, oleuropein protected in a dose-dependent fashion against loss of ejection fraction and cardiac output due to myocardial infarction at 24 h [141] and 5 weeks [142]. Oleuropein also protected in a redox-dependent manner against reperfusion injury in similar studies [143,144]. In animal models, oleuropein prevented myocardial remodeling in heart failure—by inhibiting the angiotensin-converting enzyme [145], in doxorubicin cardiomyopathy—by modulating nitric oxide synthases and other mechanisms [146], and in autoimmune myocarditis [147] and sepsis-induced myocardial injury—by NF-κB inhibition [148]. Oleuropein and hydroxytyrosol exert antiapoptotic effects on cardiomyocytes by reducing endoplasmic reticulum stress [149]. In an animal model of obesity, oleuropein and caffeic acid improved myocardiocyte energy yield without improving redox markers [150].
  • Amyloid production. Oleuropein offered protection against amyloid deposition in a human cell line that mimics familial systemic amyloidosis [151].

3.2. Use of EVOO in Diet Therapy

The Mediterranean diet (MedD) is a dietary pattern typical of countries characterized by their abundance of olive trees and the extensive use of OO in their diets, being their main source of fat. Interest in the MedD began in the 1960s with the Seven Countries population-based study, which observed up to three times lower mortality due to heart disease in Mediterranean countries compared to those of northern Europe and the United States [152]. The composition of EVOO and its effects on multiple pathophysiological processes may synergize with other factors in this dietary pattern—which seem to be key to inducing the beneficial effects of MedD—such as an active lifestyle with regular physical exercise, high fiber intake, consumption of foods naturally rich in ω-3 fatty acids, and compliance with nutritional recommendations on micro and macronutrient composition, among others. The consumption of EVOO has shown beneficial effects in many different pathologies, all of which are related to its antioxidant, immunomodulatory, and inflammatory response-regulating effects. Below, we summarize the evidence for high-impact pathologies in the general population.
  • Cardiovascular function and health. The MUFA composition of EVOO has been associated with better peripheral insulin sensitivity and glucose tolerance, reduced exogenous insulin requirements, improved endothelial function, and antithrombotic properties [153]. Clinical trials have shown that the phenolic content of EVOO is responsible for its antihypertensive properties [154,155] and its improvement of fasting and postprandial lipid profile [153] in a dose-dependent manner [156] in different clinical populations, including DM2 [157]. Clinical trials with polyphenol-rich EVOO have also shown its ability to modify levels of antioxidants, proinflammatory proteins, biomarkers of endothelial dysfunction, and DNA and lipid oxidation in a dose-dependent manner, in various clinical populations [155,158,159,160,161,162,163]. In this regard, a meta-analysis of clinical trials—with a majority of healthy adults—confirmed that high phenolic EVOO (defined as ≥200 mg/kg of phenolic content) has the greatest impact in the reduction of oxidized LDL-cholesterol (HR: −0.68; 95% CI: −1.31, −0.04) [164]. The PREDIMED study has been a milestone in this topic. It was an unblinded clinical trial, designed to test the efficacy of MedD in the primary prevention of cardiovascular major events (a composite of myocardial infarction, stroke, and death from cardiovascular causes) in a high-risk ambulatory population. The treatment arm exposed to dietary training to increase adhesion to MedD, and four tablespoons (approximately 52 g) of EVOO per day had a 31% risk reduction (HR: 0.69; 95% CI: 0.53, 0.91) of cardiovascular major events [165]. An observational post-trial follow-up in the PREDIMED study found a 10% risk reduction (HR: 0.93; 95% CI: 085, 0.95) in cardiovascular major events for each 10 g of EVOO that participants consumed at baseline, independent of their compliance to the MedD and other confounding factors at that moment [166]. A meta-analysis of cohort studies attributed a reduction in cardiovascular mortality of 21% to MedD [167]. Participants in the NHANES and UPFS cohorts that consumed more than 7 g of OO per day had a 19% risk reduction (HR: 0.81; 95% CI: 0.75, 0.87) in comparison with those that never or seldom did [168]. The substitution of an equivalent dose of margarine or butter for a tablespoon of OO showed a 6% risk reduction (HR: 0.94; 95% CI: 0.90, 0.97) in the NIH-AARP Diet and Health Study cohort [169]. Population studies also associate EVOO consumption with reduced vascular stiffness [170] and attribute the reduction in cardiovascular mortality from MedD to hydroxytyrosol and its metabolites [171]. A clinical trial in healthy volunteers found that EVOO improved proteomic markers associated with coronary artery disease, while no differences were found for those associated with chronic kidney disease or diabetes [172].
  • Lipoprotein metabolism. In 2011, the European Food Safety Authority (EFSA) issued a Scientific Opinion accepting the benefits of the daily intake of 5 mg of hydroxytyrosol and its derivatives from EVOO to prevent oxidation of LDL particles in the general population [173]. In randomized, blinded, and crossover clinical trials, the addition of polyphenol-rich EVOO was shown to reduce the expression of genes related to inflammation, atherogenesis, oxidation, metabolic syndrome, dyslipidemia, and DM2 in different clinical populations [158,174,175,176], as well as increasing the expression of genes involved in cholesterol efflux to HDL particles in people with hypertension [177]. Similarly, polyphenol-rich EVOO has the highest capacity to improve the number, proportions, and morphology of lipoproteins [178,179], due to the incorporation of ingested polyphenols into these particles [180]. Results from an animal model suggest that polyphenol supplementation and exercise may synergize to achieve a greater reduction of oxidized LDL particles [181]. On the other hand, an isocaloric high-MUFA (sourced from sunflower oil and margarine), high-fiber, and cholesterol-restricted diet was able to maintain apolipoprotein AI levels while providing the same decrease in apolipoprotein B100 as an isocaloric low-MUFA, high-fiber, and cholesterol-restricted diet in a clinical trial with people with dyslipidemia [182].
  • Alterations in carbohydrate metabolism. MedD has been associated with weight loss and protection against metabolic syndrome in humans [156]. The consumption of EVOO in the general population, at the expense of its phenolic content, has been associated with a 40% relative risk reduction in the incidence of DM2 [183]. Metabolic control in people with DM2 may also benefit from the use of EVOO, by delaying the need for initiation of antidiabetic treatment [184] and reducing exogenous insulin requirements [153]. In pregnant women, consumption of EVOO has been associated with a 27% relative risk reduction of newly diagnosed gestational diabetes [185]. In people with type 1 diabetes mellitus (DM1), EVOO has been associated with an improvement in weight, waist circumference [186], lipid profile [187], and quality of life [188]. Lastly, polyphenol-rich EVOO has maintained insulin secretion and insulin sensitivity during a high-fat diet in animal models [189].
  • Neurodegenerative diseases. Many neurodegenerative diseases share oxidative stress and neuroinflammation in their genesis and progression. Oleuropein, hydroxytyrosol, and oleocanthal are components of EVOO specifically related to the reduction in oxidative stress. Despite a suitable theoretical framework, there are not many studies that allow us to elucidate the effects of EVOO consumption on neurodegenerative diseases in humans [190]. In this regard, a long-term follow-up (mean of 6.5 years) of the PREDIMED clinical trial showed—in comparison to a low-fat diet—cognitive improvements in the EVOO supplementation and MedD dietary training group, which suggested a neuroprotective effect [191]. These findings are congruent with the results obtained in the NHANES and HPFS cohorts. Participants that consumed more than 7 g of OO per day showed a 29% lower risk of neurodegenerative disease mortality (HR: 0.71; 95% CI: 0.64–0.78), in comparison with those that never or seldom consumed OO [168].
  • Cancer. An ex vivo study in humans showed how EVOO modifies the expression of oncogenesis-related genes [192]. In observational and experimental studies in humans, EVOO consumption was associated with an overall 31% risk reduction in the incidence of any type of malignancy, both within and outside the MedD setting. The highest risk reduction appeared in pancreatic, esophageal, and urinary cancer (54%), followed by breast cancer (33%) and gastrointestinal cancer (23%). This protection may be mediated by decreased expression of receptor tyrosine-protein kinase erbB-2 (HER-2), upregulation of repressor of HER-2 expression (PEA3), increased expression of cannabinoid receptor type 1 (CNR-1), inhibition of extracellular signal-regulated kinases 1/2 (ERK1/2) phosphorylation, or reduced expression of cyclin D1 [193].
  • Gut microbiota. A 3 year sub-analysis of CORDIOPREV—a large clinical trial in people with metabolic syndrome and stable coronary heart disease—showed that the modifications of gut microbiota by EVOO supplementation (up to 138 g of EVOO per day, with >300 mg of phenolic compounds per kg of EVOO) and MedD differed according to sex [194]. In the MaPLE study—a crossover, randomized controlled clinical trial—a polyphenol-rich diet based on green tea, blood orange, Renetta apples, blueberries, and pomegranate juice (summing up to 724 mg of phenolic compounds per day) was able to improve the intestinal permeability and profile in the elderly [195]. In a small-sampled clinical trial, polyphenol-rich EVOO (500 mg of phenolic compounds per kg of EVOO) increased the proportion of IgA-coated bacteria—suggesting an increase in intestinal immunity [196]—while increasing the excretion of phenolic metabolites in feces without significantly changing the microbiome profile [197].

3.3. Oleic Acid-Based Formulations in Enteral Nutrition and Oral Supplementation

In this section, we summarize the evidence present in human studies on the use of lipids naturally present in olive oil or its major fatty component, oleic acid, in commercial enteral nutrition or oral nutritional supplementation formulations. Most of the available studies were conducted in the following clinical populations:
  • Diabetes mellitus. Diabetes-specific enteral formulations (FEED) contain fructose, low-glycemic-index carbohydrates, and a higher proportion of fat than standard formulations (40–50%), mainly MUFAs in the form of oleic acid [198]. A large cohort found an initially neutral and subsequently protective effect of dietary oleic acid against the development of DM2 [199]. Most of the commercially available enteral nutrition and oral supplementation formulations source their oleic acid from sunflower and canola oil, as it is more cost-efficient than obtaining it from OO. A clinical trial in critically ill patients without diabetes compared two formulations with the same chemical composition—one sourcing its oleic acid from sunflower oil and the other from a mixture of sunflower oil and OO—finding no differences regarding the protective effect for this intervention on the development of stress hyperglycemia [200]. We did not find further specific data regarding OO-based enteral nutrition formulations in humans; thus, we focus on oleic-acid based FEEDs irrespective of their source of oleic acid. Meta-analyses and clinical trials have shown that FEED in critically ill and noncritically ill patients with DM2—whether administered orally or with an enteral access—can improve long-term glycemic control, decrease insulin requirements, and lower postprandial glycaemia [201,202]. Table 1 summarizes the main and latest clinical trials regarding oleic acid-based FEEDs in DM2 or stress hyperglycemia in different clinical settings. The improvement in glycemic control associated with FEEDs can be attributed to several mechanisms. On one hand, the high fat content delays gastric emptying, slowing glucose absorption [203]. This may lead to nausea, especially in patients with predisposing conditions [204]. On the other hand, oleic acid increases glucagon-like peptide 1 (GLP-1) and other neuropeptides via its metabolite 2-oleoyl glycerol [205]. An ex vivo study in humans associated the level of oleic acid in spinal fluid with improved respiratory quotient and glycemic response after oral glucose overload [206]. Ethnic origins may be taken into consideration when assessing the possible benefits of an oleic acid-based enteral formulation, as a clinical trial suggested that populations of African origin may have a lower insulin secretory response following an oleic acid-based high-fat diet [207].
2.
Cardiovascular risk. The administration of FEEDs in clinical trials increased HDL-cholesterol and reduced LDL-cholesterol in noncritically ill patients with DM2 [203,226]. A meta-analysis found a significant improvement in HDL-cholesterol levels in people treated with FEED, although significance was lost in the sensitivity analysis [227]. Oleic acid was shown in a crossover clinical trial in people with DM2 to increase the concentration of the antioxidant enzyme paraoxonase 1 (PON1) in both chylomicrons and VLDL-cholesterol particles [228]. In a crossover clinical trial in healthy adults, a 3 week oral diet rich in oleic acid (comprising 28% of daily energy requirements) increased fasting respiratory quotient, reduced plasma LDL-cholesterol levels, tended to reduce fatty-acid oxidation, and significantly increased the expression of insulin-induced gene-1 (INSIG-1), a molecule that modulates the activity of the sterol regulatory element-binding protein-2/1c (SREBP-2/1c) in women [229]. Of particular interest in DM2 and other clinical populations with high cardiovascular risk is to assess the long-term cardiovascular safety of an oleic acid-based formulation, although no specific studies are available regarding this topic. In a meta-analysis of cohort studies, MUFA intake did not present a lower risk for the development of cardiovascular events than saturated fat intake, but the authors stated that the mainly animal origin of MUFAs in the selected studies may have acted as a confounding factor [230]. MUFA intake has been associated with lower diastolic blood pressure [231]. Nevertheless, MUFA intake did not change surrogate biochemical markers of cardiovascular risk [232] or hemostasis parameters [233] in clinical trials, yet the oleic acid content of the erythrocyte membrane was inversely associated with biomarkers of inflammation [234].
3.
Obesity. Of particular interest in this clinical population are the possible effects of oleic acid on satiety and oral tolerance to oleic acid-based formulas. In clinical trials with healthy volunteers, oleic acid ingestion was shown to induce greater postprandial satiety [132] by increasing levels of the endocannabinoid oleylethanolamide, which was associated with a reduced caloric intake at the next meal [133]. However, this satiating capacity may be lost in overweight or obese individuals, due to a reduced hormonal and motor response to oleic acid intake because of their higher BMI and habitual dietary fat intake [134]. The oral taste detection threshold for oleic acid may be higher than for other fatty acids [135]. Adiponectin levels may be higher in TNF-α-stimulated adipocytes after a combination of hydroxytyrosol (1–10 μM) and oleic acid (10 μM) than in TNF-α-naïve adipocytes, as suggested in an in vitro study [235]. Therefore, creating formulas based on whole-EVOO instead of oleic acid might be of interest to potentiate this anti-obesogenic effect.
4.
Inflammatory bowel disease (IBD). Studies evaluating the role of oleic acid or MUFAs in the enteral nutrition received by patients with Crohn’s disease are scarce. In clinical trials, formulations rich in oleic acid and other MUFAs were less effective than those based on linoleic acid and PUFAs [236,237]. This is consistent with previous case–control studies that showed higher dietary intake of oleic acid and plasma levels of oleic acid in people with ulcerative colitis [238,239].
5.
Elder population. In a randomized clinical trial, the administration of an oral oleic acid-based formulation during medication rounds in elderly patients admitted to a geriatric ward was safe and well tolerated, and it increased appetite and daily caloric intake, while modestly improving lipid profile [240]. A similar study that orally supplemented an oleic acid-based formulation in elderly people with DM2 found a dose-dependent improvement in glycosylated hemoglobin [241]. To our knowledge, there are no published studies to date assessing the impact of an oleic acid-based enteral nutrition or MUFA-based enteral nutrition formula on sarcopenia in the elderly. In this respect, clinical trials in other clinical populations showed discordant results regarding the role of oleic acid-based formulations in the development of myosteatosis [242,243]. In our review of the available literature, we did not find intervention studies in humans that considered the role of oleic acid or MUFA-based enteral nutrition formulae on cognition in the elderly, although an oleic acid-based high-fat diet intervention was neutral for parameters of brain inflammation or impairment of memory tests in a crossover clinical trial in young people [244]. It is of interest to note that the anti-inflammatory and antiaging effect of MUFA consumption can be modulated by polymorphisms in the telomerase RNA component (TERC) in adults [245].

3.4. Oleic Acid-Based Formulations in Parenteral Nutrition

Intravenous lipid emulsions (ILEs) are a source of essential fatty acids and nonprotein calories used in the formulation of parenteral nutrition. Traditionally, soybean oil (SO) was used in their manufacturing. In the third generation of ILEs, OO was introduced in 80:20 ratio with SO, generating a formulation composed of 15% saturated fatty acids (SFAs), 65% MUFAs (especially oleic acid, which is an ω-9 fatty acid), and 20% essential PUFAs (linoleic acid and alpha-linoleic acid, which are ω-6 fatty acids) [246]. In the fourth generation of ILEs, the SMOFlipid® 20% formulation (Fresenius Kabi Limited, Cheshire, UK) was introduced, which includes 25% OO in its composition, as well as 30% SO, 30% MCT, and 15% fish oil.
Evidence supports that OO-based formulations are associated with lower lipid peroxidation [129,247]. This results from the higher proportion of MUFAs than PUFAs in OO, which also results in these formulations not altering (or only slightly altering) inflammatory parameters when compared to traditional SO or SO emulsified with medium chain triglycerides (MCTs) [248,249]. In addition, MUFAs are associated with health benefits in the general population [250,251,252]. Another factor that may contribute to lower lipid peroxidation is the high α-tocopherol content of OO compared to SO, which contains higher concentrations of γ-tocopherol [253].
Third-generation and fourth-generation ILEs have lower ω-6:ω-3 fatty acid ratios than traditional ILEs [246]. Potential health benefits may derive from this, as ω-3 fatty acids have been associated with greater anti-inflammatory activity and vice versa [254]. ILEs containing OO could preserve the immune function, mainly through innate immunity [255,256,257]. In a study involving patients with advanced head and neck squamous carcinoma, the use of OO-based ILEs demonstrated an increase in natural and adaptive immunity, postulating that this could favorably influence their response to chemotherapy [258]. There are scarce data regarding infection in patients on parenteral nutrition as a function of the ILEs used; however, in the largest clinical trial to date, OO-based ILEs were associated with fewer infections when compared with soybean oil (SO)-based ILEs [259].
Another concern regarding the use of parenteral nutrition is its possible deleterious effect on the lipid profile of patients. OO-based ILEs may have beneficial effects on total cholesterol levels [129,260], and, although their relationship with blood TAGs is less clear, it has been shown in both children and adults that these formulas are safe and have little effect on the lipid profile when used long-term [248,261]. Regarding glucose metabolism, although little data exist, no differences have been found between OO-based and SO-based ILEs in adults [260].
With regard to liver function, there is no evidence that these OO-based ILEs alter liver function when used long-term [129] When comparing OO-based ILEs with other oil-based formulations, there are mixed results and differences in bilirubin, γ-glutamyl transferase (GGT), and alanine and aspartate aminotransferase (ALT, AST) levels that—while statistically significant—are not necessarily clinically significant, since these levels were within the normal ranges or within 1.5× upper limit of normal (ULN). Therefore, more studies are needed in this area [129].
Current results regarding clinical outcomes such as mortality and hospital or ICU stay did not differ between different types of ILEs in most studies, although two of them demonstrated a shorter duration of mechanical ventilation in patients receiving OO-based ILEs [262,263].
The maximum dose for OO-based ILEs is 2 g/kg of weight/day and should be reduced or discontinued if blood TAGs exceed 4.5 mmol/L [264] Regarding contraindications to the use of OO-based ILEs, they should not be used in patients with hypersensitivity to egg, OO, soy, peanuts, or any component thereof. In addition, OO-based ILEs formulations should be administered with caution in patients requiring anticoagulant therapy, as their lipids contain only 10–50 µg/L of vitamin K [250].

3.5. Supplementation of Nutraceuticals from EVOO

In this section, we summarize the studies that supplemented minor components of EVOO, either in isolation or within food matrices such as EVOO itself. Most of the available studies were conducted in the following clinical populations:
  • Diabetes. Human clinical trials have demonstrated that oleuropein can improve postprandial glycemic response in healthy individuals [112], in overweight people without impaired carbohydrate metabolism [265], and in those with carbohydrate intolerance [266] and DM2 at the expense of increased GLP1 and insulin, by reducing oxidative stress through inhibition of NADPH oxidase 2 (Nox2) [267]. In a randomized, blinded clinical trial, olive leaf extract was also shown to improve postprandial glycemic response and metabolic control in people with DM2 [268]. In a prospective study in people with hydrocarbon intolerance, plasma β-sitosterol level acted as a protective factor against the development of DM2 [269]. Ex vivo studies in humans have indicated that oleuropein and hydroxytyrosol can inhibit α-amylase, α-glucosidase, sucrase, lipase, and glucose transporter 2 (GLUT-2) [112,270,271,272]. In animal models, oleanolic acid and luteolin have shown a superior ability to olive leaf extract in reducing postprandial blood glucose, all being significantly superior to placebo [266]. A meta-analysis of clinical trials found that phytosterols reduce insulin resistance, but not enough to generate clinically significant improvements in glucose metabolism [273]. This is backed by in vitro studies that demonstrated increased expression levels and activation of different components of the insulin receptor signaling pathway by phytosterols [274,275].
  • Cardiovascular risk. In randomized, controlled, double-blind, and crossover clinical trials, administration of oleuropein and hydroxytyrosol [276]—as well as an olive leaf extract [277]—significantly reduced in vivo vascular stiffness and interleukin-8 production in prehypertensive individuals and healthy volunteers, respectively. Polyphenol administration in people with pre-diabetes or metabolic syndrome [278], as well as in overweight or obese people [279], improved biomarkers of endothelial dysfunction. This improvement in endothelial function was not associated with stimulation of angiogenesis [124]. In a double-blind, controlled, crossover clinical trial, hydroxytyrosol 7.5 mg b.i.d. increased antioxidant enzyme expression and reduced levels of oxidative stress markers [280]. A similar study—in which hydroxytyrosol 5 mg q.d and 25 mg q.d was administrated for 1 week—found no difference in levels of biomarkers of inflammatory and redox status, angiogenesis, and liver health, suggesting that this intervention requires a longer exposure time [281]. In vitro, both hydroxytyrosol and its metabolites lowered E-selectin, P-selectin, VCAM-1, ICAM-1, and MCP-1 levels in TNF-α-stimulated human endothelial cells, yet hydroxytyrosol metabolites interestingly exerted a greater effect than hydroxytyrosol at low concentrations (1 μM) [282]. A double-blind, controlled clinical trial in people with metabolic syndrome showed how supplementation of two minor components (chlorogenic acid and luteolin) improved intima-media thickness and flow-mediated dilation [283]. Regarding FALs, policosanol is a multifaceted drug with antiaggregant properties through direct inhibition of thromboxane A2 synthesis [284] and a lipid-lowering effect through the inhibition of cholesteryl ester transfer protein (CETP) activity, offering protection of LDL particles from oxidation [285,286]. In a double-blind, placebo-controlled, parallel clinical trial with ischemic heart disease patients, policosanol 5 mg b.i.d. with or without aspirin 125 mg q.d. reduced cardiac events and enhanced exercise tolerance [287]. Clinical trials have shown that policosanol 20 mg q.d. may have less adverse effects and better efficacy than low-intensity statins and low-dose aspirin in adults with dyslipidemia [288,289]. A meta-analysis calculated an LDL-cholesterol reduction of −23.7% for each 12 mg of policosanol per day [290], although other clinical trials found no beneficial effects in mild hypercholesterolemia [291]. Policosanol reduced LDL-cholesterol and triglyceride levels, increased HDL-cholesterol levels, and reduced systolic blood pressure in a double-blind, placebo-controlled, and parallel clinical trial with women with prehypertension [285]. In comparison to a high-fat diet, healthy C57BL/6 mice that were fed a high-fat diet with 0.5% policosanol for 18 weeks developed a better lipid profile, a reduction in brown and white inguinal adipose tissue, a distinctive microbiota profile, and enhanced lipolysis and thermogenesis [292], while, in a similar study, 60 mg/kg of either octacosanol or policosanol protected against visceral fat hyperplasia and liver steatosis [293]. Beneficial effects of phytosterols on LDL-cholesterol are well established, with a mean reduction of 8–10% [294] and significant reductions in ApoCII and ApoE [295]. An extensive review of clinical trials with phytosterols in cardiovascular risk reduction was published elsewhere [126]. There is no definitive evidence on the impact of dividing the daily phytosterol intake on cholesterol metabolism [296,297]; therefore, further studies are needed to make recommendations. The FDA has supported the claim that plant sterols and stanols—as part of a diet low in saturated fat and cholesterol—may reduce the risk of coronary heart disease (CHD), recommending an intake of 1.3 g or more per day of plant sterol esters (at least 0.65 g per serving) or 3.4 g or more per day of plant stanol esters (at least 1.7 g per serving) [298]. The phytosterol content of typical serving sizes of EVOO is well below the 0.65 g threshold. The association between phytosterols and CHD is complex, and new evidence supports that it may be different depending on the food matrix and demographics [299,300], clinical factors, and genetic traits [301,302]. Plant sterol oxidation products (POPs) and their consumption by nontarget population groups are also a concern. The EFSA has calculated that the approved maximum intake of 3 g of plant sterols per person per day may be deemed either safe or unsafe depending on its oxidation rate, which may depend on the cooking method, if the residual fat is discarded, and the characteristics of the food matrix [303]. Nevertheless, studies in animal models have shown that some POPs may have beneficial health properties, such as lipid-lowering properties (via PPAR-α activation, induction of cytochrome P450, and inhibition of enzymes involved in hepatic fatty-acid synthesis) [304].
  • Obesity. There was a randomized, controlled, double-blind, and crossover clinical trial in which the addition of an olive leaf extract did not improve body composition or cardiovascular risk surrogates [265]. Oleuropein and hydroxytyrosol can reduce adipocyte hyperplasia and hypertrophy, by inhibition of adipogenesis-related genes in human cell lines [305,306] and stimulation of mitochondrial biogenesis-related genes in murine adipocytes [307]. When fed high-fat diets, EVOO-supplemented mice underwent lower weight gains [189], and they showed less metabolic, inflammatory, and microbiota disruption [308], as well as more modest subcutaneous and visceral fat development—in relation to a higher basal energy expenditure proportional to their weight—which could be explained by a higher expression of Ucp genes in muscle and visceral fat [309]. Regarding BMI reduction, a meta-analysis of clinical trials calculated statistically significant improvements for food enrichment with phytosterols and phytostanols in dyslipidemic, overweight, and obese subjects [310].
  • Nonalcoholic fatty liver disease (NAFLD). To our knowledge, there have been no intervention studies in humans that supplement minor components in EVOO in patients with NAFLD. In humans, levels of sitosterols in the liver in adults with NAFLD have shown an inverse correlation with the degree of steatosis and lobular inflammation by liver biopsy [311] In animal models, liver damage induced by a high-fat diet was significantly reduced by supplementation with EVOO [312,313], EPA + DHA [314,315,316], DHA + EVOO [317] EPA + hydroxytyrosol [318,319,320,321], or hydroxytyrosol [322], showing beneficial effects on inflammation, lipid content, and hepatocyte gene expression. This evidence is mixed, as other animal models have not demonstrated a positive effect of EVOO and its phenolic compounds on the development of NAFLD [189] or its regression [323]. The beneficial effect of EVOO against NAFLD could be explained by mitochondrial protection—preventing the decrease in nitrated fatty-acid levels caused by a high-fat diet [324]—this ability being directly related to its phenolic content [325]. In addition, EVOO’s lipid profile is low in linoleic acid (ω-6), preventing a proinflammatory state due to an ω-6/ω-3 imbalance [326], with a high content of oleic acid (MUFA), transformed into oleylethanolamide and stimulating PPAR-α, and α-linolenic acid (PUFA), an ω-3 precursor that triggers antisteatotic, antioxidant, and anti-inflammatory responses in the liver via multiple molecular mechanisms [327].
  • Cancer. A nutrigenomic study demonstrated ex vivo that oleuropein leaf extract can modify the expression of genes involved in oncogenesis in healthy humans [328]. There is an extensive review of direct antitumoral effects of oleuropein and hydroxytyrosol in in vitro and ex vivo models [329], which are linked to in vitro evidence of the intratumoral transformation of oleuropein into hydroxytyrosol [330]. Pleiotropic effects in vitro and ex vivo resulting from combinations of oleuropein, hydroxytyrosol, and other minor components with chemotherapeutics and immunomodulators, leading to drug synergism or a reduction in undesirable effects, were thoroughly reviewed elsewhere [33]. An extensive review of the effects of EVOO and its minor components in preclinical models of colon cancer is also available [331]. Oleuropein-rich EVOO stimulated in vivo apoptosis of colon cancer cells and peritoneal macrophages in an animal model [332], and a much more potent nanoencapsulated microemulsion of oleuropein was recently developed and tested in an animal model [333]. While the addition of oleic acid induced DNA synthesis and growth of the human colorectal adenocarcinoma cell line Caco-2, the subsequent addition of EVOO’s minor components prevented this effect, even inducing apoptosis in the case of hydroxytyrosol and maslinic acid [330]. Regarding mechanisms of action, oleuropein was able to decrease cell number and motility in the human melanoma cell line A375—decreasing their glycolytic capacity (Warburg effect)—at the expense of glucose transporter 1 (GLUT-1), pyruvate kinase M2 (PKM2), and monocarboxylate transporter 4 (MCT4) inhibition [334].
  • Neurological diseases. In a randomized clinical trial, an oral palmitoylethanolamide and luteolin extract improved the detection threshold, discrimination ability, and smell identification in people with COVID-19 hyposmia [335]. Nevertheless, oral luteolin supplementation in a quasi-experimental study was not superior to placebo at reducing symptoms of Gulf War Illness, a neuroinflammatory condition [336]. Oleuropein has demonstrated neuroprotective effects in animal models of cerebral ischemia, cerebral reperfusion injury, ageing, Alzheimer’s disease, Huntington’s disease, multiple sclerosis, Parkinson’s disease, peripheral neuropathy, spinal cord injury, and epilepsy [337,338]. With respect to Alzheimer’s disease, oleocanthal was shown to increase neuronal clearance of β-amyloid by increasing P-glycoprotein (P-gp) and LDL receptor-related protein-1 (LRP1) levels in mouse brains [339], while restoring the blood–brain barrier function and reducing neuroinflammation through inhibition of PYD domain-containing protein 3 (NLRP3) inflammasome and inducing autophagy through activation of the AMP-activated protein kinase/Unc-51-like autophagy activating kinase 1 (AMPK/ULK1) pathway [340].
  • IBD. In contrast to its lipid profile, the minority components of EVOO may have a beneficial effect on IBD, although we found no human intervention studies. Ex vivo studies in humans have pointed to the ability of oleuropein to reduce inflammatory infiltrate and inflammatory markers, improving tissue architecture [341,342]. In colitis animal models, oleuropein supplementation decreased mortality and improved histopathology by increasing antioxidant enzyme levels and Bcl2 expression while reducing proinflammatory cytokine and Bax expression [343], while EVOO supplementation improved rectal bleeding [344] and the visceromotor response to mechanical distension [345]. These beneficial effects may be enhanced by delivering oleuropein via lipid nanoparticles orally [346]. To gain further insight into this topic, we point the reader to an excellent review [347].
  • Benign prostatic hypertrophy (BPH). In three randomized, double-blind, placebo-controlled clinical trials, β-sitosterol demonstrated superiority in improving self-reported somatic and sexual symptoms, quality of life, peak urine flow, post-void residual volume, and plasma free testosterone levels [127,348,349]. As possible explanatory mechanisms, this molecule reduced 5α-reductase activity and prostate volume in a hamster model of BPH [350], while unfractionated olive oil reduced prostate volume in a murine model of BPH, at the expense of decreasing prostatic testosterone (T) and dihydrotestosterone (DHT) concentrations [351].
  • Osteoporosis. In a randomized, double-blind, 12 month clinical trial of postmenopausal women with mild osteoporosis and no drugs or diseases affecting phospho-calcium metabolism, the addition of an oral olive leaf extract containing 40% oleuropein to 1000 mg of calcium carbonate q.d maintained bone mineral density levels by DEXA and increased osteocalcin in a safe and well-tolerated manner [352]. An ex vivo study in humans using oleuropein [353] and in vivo studies in murine models of osteoporosis using EVOO and vitamin D3 [354], oleuropein [355,356], and hydroxytyrosol [357] have shown an anti-inflammatory effect—inhibiting osteoclastic activity and reducing trabecular bone loss—while in some cases also stimulating osteoblastic activity. An excellent review of previous studies indicated that EVOO’s bone protection is probably mediated by its anti-inflammatory capacity [358].
  • Rheumatoid arthritis (RA). In a pilot intervention study in newly diagnosed patients, the addition of 35 mg oleuropein to methotrexate generated the maximum reduction in markers of cell damage, oxidative stress, and inflammation at 6 weeks [359]. In a randomized clinical trial with a mean duration of approximately 7 years, the addition of an olive leaf, fig leaf, and fruit extract to methotrexate improved patient-perceived health status without changing disease activity scales by 16 weeks [360]. In a randomized, crossover clinical trial, the addition of EVOO and fish oil to disease-modifying drugs (DMARDs) showed improved patient-perceived health status, increased functionality, and lowered rheumatoid factor levels at 24 weeks compared to the addition of fish oil to DMARDs [218]. All these interventions were safe and well tolerated. In the TOMORROW cohort, MUFA intake was significantly lower in RA patients, while MUFA intake tended to be an independent predictor of disease remission [361]. In a cross-sectional study, the consumption of olive products in the usual diet of people with RA showed a good correlation and explanation of variance in principal component analysis with respect to their calculated amount of dietetical plant-derived PUFAs. However, the amount of PUFAs was most strongly related to the consumption of walnuts and linoleic and alpha-linolenic acids, acting as possible confounding factors. In this study, consumption of PUFAs was associated with a lower degree of disease activity according to Disease Activity Score on 28 joints (DAS28), independently of antibody positivity and with higher protection in more severe or chronic forms of the disease [362].

4. Discussion

EVOO has a pivotal role in the MedD. With its well-balanced mixture of oleic acid and minor components, the culinary use of EVOO in the general population may contribute to the daily requirements of essential fatty acids and antioxidant body supply. Likewise, whole EVOO, its lipids, or minor components can be supplemented—in a safe, effective, and well-tolerated manner—in fortified foods, oral, enteral, and intravenous pharmacological formulations in clinical nutrition. Furthermore, diet therapy with EVOO and the nutraceutical supplementation of its minor components currently have a very scientifically sound rationale after showing multiple beneficial effects—in both intervention and mechanistic studies—in important conditions for public health, such as cardiovascular health, lipoprotein metabolism, and diabetes mellitus. Currently, promising therapeutic evidence—albeit in different stages of development, and too early to recommend specific EVOO-based dietary or nutraceutical interventions—is available for conditions such as obesity, NAFLD, neurodegenerative diseases, osteoporosis, cancer, and inflammatory diseases, amongst others. Regarding EVOO-based formulations in clinical nutrition, oleic acid-based ILEs already are well established parenteral formulations with widespread use in the clinical setting. To our knowledge, both oral and enteral oleic acid-based formulas may facilitate metabolic control, although adequately blinded, crossover clinical trials with a high number of participants and an intention-to-treat analysis are needed to further evaluate their safety and efficacy in specific situations.
Considering the aforementioned and possible future lines of research, it would be of high interest to develop intervention trials to further evaluate the effect of EVOO and its minor components on hard endpoints in high-impact clinical conditions such as heart disease, NAFLD, IBD, bacterial infections, or HIV. It would be of particular interest to assess the long-term cardiovascular safety and effects on hemostatic and inflammatory parameters of oleic acid-based formulations for enteral nutrition or oral supplementation—especially with minor components from EVOO—in groups with high cardiovascular risk. Clinical nutrition could also benefit from further studies comparing liver function under different OO-based ILEs in a randomized fashion in different clinical populations. Lastly, sarcopenia and cognition in the elderly are ever-developing fields that could benefit from well-planned studies assessing the impact of oleic acid-based oral and enteral nutrition formulations, as well as clinical trials designed to further test the efficacy and tolerability of EVOO’s minor components in age-related conditions such as osteoporosis, cancer, and dementia.

5. Conclusions

EVOO has diverse beneficial health properties and a well-established role in clinical nutrition. More specifically, the current evidence supports the use of whole EVOO in diet therapy and the supplementation of EVOO’s minor components to improve cardiovascular health, lipoprotein metabolism, and diabetes mellitus, as well as enough evidence for the use—when available—of oleic acid-based ILEs in parenteral nutrition. EVOO-based nutritional interventions have thus far shown very promising results in other high-impact clinical conditions, but more intervention studies in humans are needed in this case to develop specific recommendations and grant new therapeutic uses of EVOO through different formulations and in specific clinical populations.

Supplementary Materials

The following supporting information can be downloaded at https://www.mdpi.com/article/10.3390/nu14071440/s1: Table S1. Composition of fatty acids in EVOO produced from different Mediterranean cultivars; Table S2. Composition of some of the antioxidants in EVOO produced from different Mediterranean cultivars.

Author Contributions

Conceptualization and methodology, P.P.G.-L. and J.L.P.-C.; investigation, A.J.-S., A.J.M.-O. P.J.R.-R. and A.P.-G.; writing—original draft preparation, A.J.-S., A.J.M.-O., P.J.R.-R. and A.P.-G..; writing—review and editing, A.J.-S.; supervision and project administration, P.P.G.-L. All authors have read and agreed to the published version of the manuscript.

Funding

This article is part of the R+D+i project PID2019-107837RB-I00, funded by MCIN/AEI/10.13039/501100011033/.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Commission Regulation (EEC) No 2568/91 of 11 July 1991 on the characteristics of olive oil and olive-residue oil and on the relevant methods of analysis. Consolidated Text. 01991R2568—EN—04.12.2016—031.005—1. Off. J. Eur. Union 1991, 248, 12–23. [Google Scholar]
  2. International Olive Council. Trade Standard Applying to Olive Oils and Olive Pomace Oils; COI/T.15/NC No 3/Rev. 13; International Olive Council: Madrid, Spain, 2019; English version, Original: French version. [Google Scholar]
  3. Hippocrates. Tratados Hipocráticos: Tratados Quirúrgicos; María Dolores, L.N., Translator; International Olive Council, Gredos: Madrid, Spain, 1993; Volume VII, ISBN 978-84-249-1610-7. [Google Scholar]
  4. Dioscorides, P.; García Valdés, M. Plantas y Remedios Medicinales: De Materia Medica; Libros I–III; Biblioteca Clásica Gredos: Madrid, Spain, 1998; ISBN 978-84-249-1958-0. [Google Scholar]
  5. Plinius Secundus, G. Historia Natural; Letras Universales; Cantó, M.J., Ed.; Cátedra: Madrid, Spain, 2002; ISBN 978-84-376-1958-3. [Google Scholar]
  6. Aparicio, R.; Harwood, J.L. Handbook of Olive Oil: Analysis and Properties, 2nd ed.; Springer: New York, NY, USA, 2013; ISBN 978-1-4614-7776-1. [Google Scholar]
  7. Preedy, V.R.; Watson, R.R. Olives and Olive Oil in Health and Disease Prevention, 1st ed.; Elsevier: Amsterdam, The Netherlands, 2010; ISBN 978-0-12-374420-3. [Google Scholar]
  8. Miloš, J. (Ed.) Handbook of Olive Oil: Phenolic Compounds, Production and Health Benefits; Food Science and Technology; Nova Science Publishers: New York, NY, USA, 2017; ISBN 978-1-5361-2356-2. [Google Scholar]
  9. Boskou, D. Olives and Olive Oil Bioactive Constituents; AOCS Press: Urbana, IL, USA, 2015; ISBN 978-1-63067-041-2. [Google Scholar]
  10. Boskou, D. Olive Oil. World Rev. Nutr. Diet 2000, 87, 56–77. [Google Scholar] [CrossRef] [PubMed]
  11. Aparicio-Ruíz, R. SEXIA © (Expert System for Olive Oil Characterization) Databases; Instituto de La Grasa-CSIC: Seville, Spain, 2013. [Google Scholar]
  12. Pérez Martínez, P.; López-Miranda, J.; Delgado-Lista, J.; López-Segura, F.; Pérez Jiménez, F. Aceite de oliva y prevención cardiovascular: Más que una grasa. Clínica Investig. Arterioscler. 2006, 18, 195–206. [Google Scholar] [CrossRef] [Green Version]
  13. Gouvinhas, I.; Machado, N.; Sobreira, C.; Domínguez-Perles, R.; Gomes, S.; Rosa, E.; Barros, A.I. Critical review on the significance of olive phytochemicals in plant physiology and human health. Molecules 2017, 22, 1986. [Google Scholar] [CrossRef] [Green Version]
  14. Visioli, F.; Davalos, A.; López de Las Hazas, M.-C.; Crespo, M.C.; Tomé-Carneiro, J. An overview of the pharmacology of olive oil and its active ingredients. Br. J. Pharm. 2020, 177, 1316–1330. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Jukić Špika, M.; Perica, S.; Žanetić, M.; Škevin, D. Virgin olive oil phenols, fatty acid composition and sensory profile: Can cultivar overpower environmental and ripening effect? Antioxidants 2021, 10, 689. [Google Scholar] [CrossRef]
  16. Mastralexi, A.; Tsimidou, M.Z. The potential of virgin olive oil from cv. Chondrolia chalkidikis and Chalkidiki (Greece) to bear health claims according to the European legislation. Molecules 2021, 26, 3184. [Google Scholar] [CrossRef] [PubMed]
  17. Vekiari, S.A.; Oreopoulou, V.; Kourkoutas, Y.; Kamoun, N.; Msallem, M.; Psimouli, V.; Arapoglou, D. Characterization and seasonal variation of the quality of virgin olive oil of the Throumbolia and Koroneiki varieties from Southern Greece. Grasas Aceites 2010, 61, 221–231. [Google Scholar] [CrossRef] [Green Version]
  18. Katsoyannos, E.; Batrinou, A.; Chatzilazarou, A.; Bratakos, S.M.; Stamatopoulos, K.; Sinanoglou, V.J. Quality parameters of olive oil from stoned and nonstoned Koroneiki and Megaritiki Greek olive varieties at different maturity levels. Grasas Aceites 2015, 66, e067. [Google Scholar] [CrossRef] [Green Version]
  19. Squeo, G.; Silletti, R.; Mangini, G.; Summo, C.; Caponio, F. The potential of apulian olive biodiversity: The case of Oliva rossa virgin olive oil. Foods 2021, 10, 369. [Google Scholar] [CrossRef]
  20. Di Vaio, C.; Graziani, G.; Gaspari, A.; De Luca, L.; Aiello, A.; Cirillo, A.; Bruno, A.; Romano, R.; Ritieni, A. Drupe characters, fatty acids, polyphenolic and aromatic profile of olive oil obtained from “Oliva bianca”, minor autochthonous cultivar of campania. Plants 2021, 10, 1119. [Google Scholar] [CrossRef] [PubMed]
  21. El Riachy, M.; Hamade, A.; Ayoub, R.; Dandachi, F.; Chalak, L. Oil content, fatty acid and phenolic profiles of some olive varieties growing in Lebanon. Front. Nutr. 2019, 6, 94. [Google Scholar] [CrossRef] [PubMed]
  22. Ramos-Escudero, F.; Morales, M.T.; Asuero, A.G. Characterization of bioactive compounds from monovarietal virgin olive oils: Relationship between phenolic compounds-antioxidant capacities. Int. J. Food Prop. 2015, 18, 348–358. [Google Scholar] [CrossRef]
  23. Montaño, A.; Hernández, M.; Garrido, I.; Llerena, J.; Espinosa, F. Fatty acid and phenolic compound concentrations in eight different monovarietal virgin olive oils from Extremadura and the relationship with oxidative stability. Int. J. Mol. Sci. 2016, 17, 11960. [Google Scholar] [CrossRef] [Green Version]
  24. Youssef, O.; Guido, F.; Daoud, D.; Mokhtar, Z. Effect of cultivar on minor components in Tunisia olive fruits cultivated in microclimate. Afr. J. Wood Sci. For. 2020, 8, 001–008. [Google Scholar]
  25. Gargouri, B.; Ammar, S.; Zribi, A.; Mansour, A.B.; Bouaziz, M. Effect of growing region on quality characteristics and phenolic compounds of Chemlali extra-virgin olive oils. Acta Physiol. Plant. 2013, 35, 2801–2812. [Google Scholar] [CrossRef]
  26. Chtourou, F.; Valli, E.; Ben Mansour, A.; Bendini, A.; Gallina Toschi, T.; Bouaziz, M. Characterization of virgin olive oils obtained from minor Tunisian varieties for their valorization. Food Meas. 2021, 15, 5060–5070. [Google Scholar] [CrossRef]
  27. Jimenez-Lopez, C.; Carpena, M.; Lourenço-Lopes, C.; Gallardo-Gomez, M.; Lorenzo, J.M.; Barba, F.J.; Prieto, M.A.; Simal-Gandara, J. Bioactive compounds and quality of extra virgin olive oil. Foods 2020, 9, 1014. [Google Scholar] [CrossRef] [PubMed]
  28. Kyçyk, O.; Aguilera, M.P.; Gaforio, J.J.; Jiménez, A.; Beltrán, G. Sterol composition of virgin olive oil of forty-three olive cultivars from the world collection olive germplasm bank of Cordoba. J. Sci. Food Agric. 2016, 96, 4143–4150. [Google Scholar] [CrossRef] [PubMed]
  29. Lukić, M.; Lukić, I.; Moslavac, T. Sterols and triterpene diols in virgin olive oil: A comprehensive review on their properties and significance, with a special emphasis on the influence of variety and ripening degree. Horticulturae 2021, 7, 493. [Google Scholar] [CrossRef]
  30. Giuffrè, A.M.; Louadj, L. Influence of crop season and cultivar on sterol composition of monovarietal olive oils in Reggio Calabria (Italy). Czech. J. Food Sci. 2013, 31, 256–263. [Google Scholar] [CrossRef] [Green Version]
  31. Cano, M.M.; de Miguel Gordillo, C.; Mendoza, M.F.; Vertedor, D.M.; Casas, J.S. The sterol and erythrodiol + uvaol content of virgin olive oils produced in five olive-growing zones of Extremadura (Spain). J. Am. Oil Chem. Soc. 2016, 93, 227–235. [Google Scholar] [CrossRef]
  32. Peyrot des Gachons, C.; Uchida, K.; Bryant, B.; Shima, A.; Sperry, J.B.; Dankulich-Nagrudny, L.; Tominaga, M.; Smith, A.B.; Beauchamp, G.K.; Breslin, P.A.S. Unusual pungency from extra-virgin olive oil is attributable to restricted spatial expression of the receptor of oleocanthal. J. Neurosci. 2011, 31, 999–1009. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Torić, J.; Marković, A.K.; Brala, C.J.; Barbarić, M. Anticancer effects of olive oil polyphenols and their combinations with anticancer drugs. Acta Pharm. 2019, 69, 461–482. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Rahmani, A.H.; Albutti, A.S.; Aly, S.M. Therapeutics role of olive fruits/oil in the prevention of diseases via modulation of anti-oxidant, anti-tumour and genetic activity. Int. J. Clin. Exp. Med. 2014, 7, 799–808. [Google Scholar] [PubMed]
  35. Douzane, M.; Tamendjari, A.; Abdi, A.K.; Daas, M.S.; Mehdid, F.; Bellal, M.M. Phenolic compounds in mono-cultivar extra virgin olive oils from Algeria. Grasas Aceites 2013, 64, 285–294. [Google Scholar] [CrossRef]
  36. Torić, J.; Barbarić, M.; Uršić, S.; Jakobušić Brala, C.; Karković Marković, A.; Zebić Avdičević, M.; Benčić, Đ. Antique traditional practice: Phenolic profile of virgin olive oil obtained from fruits stored in seawater. Foods 2020, 9, 1347. [Google Scholar] [CrossRef]
  37. Diamantakos, P.; Giannara, T.; Skarkou, M.; Melliou, E.; Magiatis, P. Influence of harvest time and malaxation conditions on the concentration of individual phenols in extra virgin olive oil related to its healthy properties. Molecules 2020, 25, 2449. [Google Scholar] [CrossRef] [PubMed]
  38. Negro, C.; Aprile, A.; Luvisi, A.; Nicolì, F.; Nutricati, E.; Vergine, M.; Miceli, A.; Blando, F.; Sabella, E.; De Bellis, L. Phenolic profile and antioxidant activity of Italian monovarietal extra virgin olive oils. Antioxidants 2019, 8, 161. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Różańska, A.; Russo, M.; Cacciola, F.; Salafia, F.; Polkowska, Ż.; Dugo, P.; Mondello, L. Concentration of potentially bioactive compounds in Italian extra virgin olive oils from various sources by using LC-MS and multivariate data analysis. Foods 2020, 9, 1120. [Google Scholar] [CrossRef] [PubMed]
  40. Fanali, C.; Della Posta, S.; Vilmercati, A.; Dugo, L.; Russo, M.; Petitti, T.; Mondello, L.; de Gara, L. Extraction, analysis, and antioxidant activity evaluation of phenolic compounds in different Italian extra-virgin olive oils. Molecules 2018, 23, 3249. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Barbalace, M.C.; Zallocco, L.; Beghelli, D.; Ronci, M.; Scortichini, S.; Digiacomo, M.; Macchia, M.; Mazzoni, M.R.; Fiorini, D.; Lucacchini, A.; et al. Antioxidant and neuroprotective activity of extra virgin olive oil extracts obtained from Quercetano cultivar trees grown in different areas of the Tuscany region (Italy). Antioxidants 2021, 10, 421. [Google Scholar] [CrossRef] [PubMed]
  42. El Hilali, H.; El Hilali, F.; Porter, S.E.G.; Ghali, S.A.; Meyls, H.M.; Ouazzani, N.; Laziri, F.; Barber, A. Olive oil varieties cultivated in Morocco reduce reactive oxygen species and cell viability of human cervical cancer cells. Mediterr. J. Nutr. Metab. 2020, 13, 89–100. [Google Scholar] [CrossRef]
  43. Bouymajane, A.; Oulad El Majdoub, Y.; Cacciola, F.; Russo, M.; Salafia, F.; Trozzi, A.; Rhazi Filali, F.; Dugo, P.; Mondello, L. Characterization of phenolic compounds, vitamin e and fatty acids from monovarietal virgin olive oils of “Picholine marocaine” cultivar. Molecules 2020, 25, 5428. [Google Scholar] [CrossRef] [PubMed]
  44. Becerra-Herrera, M.; Vélez-Martín, A.; Ramos-Merchante, A.; Richter, P.; Beltrán, R.; Sayago, A. Characterization and evaluation of phenolic profiles and color as potential discriminating features among spanish extra virgin olive oils with protected designation of origin. Food Chem. 2018, 241, 328–337. [Google Scholar] [CrossRef]
  45. Quintero-Flórez, A.; Pereira-Caro, G.; Sánchez-Quezada, C.; Moreno-Rojas, J.M.; Gaforio, J.J.; Jimenez, A.; Beltrán, G. Effect of olive cultivar on bioaccessibility and antioxidant activity of phenolic fraction of virgin olive oil. Eur. J. Nutr. 2018, 57, 1925–1946. [Google Scholar] [CrossRef] [PubMed]
  46. López-Yerena, A.; Ninot, A.; Jiménez-Ruiz, N.; Lozano-Castellón, J.; Pérez, M.; Escribano-Ferrer, E.; Romero-Aroca, A.; Lamuela-Raventós, R.M.; Vallverdú-Queralt, A. Influence of the ripening stage and extraction conditions on the phenolic fingerprint of ‘Corbella’ extra-virgin olive oil. Antioxidants 2021, 10, 877. [Google Scholar] [CrossRef] [PubMed]
  47. López-Yerena, A.; Lozano-Castellón, J.; Olmo-Cunillera, A.; Tresserra-Rimbau, A.; Quifer-Rada, P.; Jiménez, B.; Pérez, M.; Vallverdú-Queralt, A. Effects of organic and conventional growing systems on the phenolic profile of extra-virgin olive oil. Molecules 2019, 24, 1986. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Bayram, B.; Esatbeyoglu, T.; Schulze, N.; Ozcelik, B.; Frank, J.; Rimbach, G. Comprehensive analysis of polyphenols in 55 extra virgin olive oils by HPLC-ECD and their correlation with antioxidant activities. Plant Foods Hum. Nutr. 2012, 67, 326–336. [Google Scholar] [CrossRef]
  49. Loubiri, A.; Taamalli, A.; Talhaoui, N.; Mohamed, S.N.; Carretero, A.S.; Zarrouk, M. Usefulness of phenolic profile in the classification of extra virgin olive oils from autochthonous and introduced cultivars in Tunisia. Eur. Food Res. Technol. 2017, 243, 467–479. [Google Scholar] [CrossRef]
  50. Ouni, Y.; Taamalli, A.; Gómez-Caravaca, A.M.; Segura-Carretero, A.; Fernández-Gutiérrez, A.; Zarrouk, M. Characterisation and quantification of phenolic compounds of extra-virgin olive oils according to their geographical origin by a rapid and resolutive LC–ESI-TOF MS Method. Food Chem. 2011, 127, 1263–1267. [Google Scholar] [CrossRef] [PubMed]
  51. Alkan, D.; Tokatli, F.; Ozen, B. Phenolic characterization and geographical classification of commercial extra virgin olive oils produced in Turkey. J. Am. Oil Chem. Soc. 2012, 89, 261–268. [Google Scholar] [CrossRef]
  52. Garcia-Oliveira, P.; Jimenez-Lopez, C.; Lourenço-Lopes, C.; Chamorro, F.; Pereira, A.G.; Carrera-Casais, A.; Fraga-Corral, M.; Carpena, M.; Simal-Gandara, J.; Prieto, M.A. Evolution of flavors in extra virgin olive oil shelf-life. Antioxidants 2021, 10, 368. [Google Scholar] [CrossRef] [PubMed]
  53. Yan, J.; Alewijn, M.; van Ruth, S.M. From extra virgin olive oil to refined products: Intensity and balance shifts of the volatile compounds versus odor. Molecules 2020, 25, 2469. [Google Scholar] [CrossRef] [PubMed]
  54. Volpe, M.G.; De Cunzo, F.; Siano, F.; Paolucci, M.; Barbarisi, C.; Cammarota, G. Influence of extraction techniques on physical-chemical characteristics and volatile compounds of extra virgin olive oil. J. Oleo Sci. 2014, 63, 875–883. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Angerosa, F.; Servili, M.; Selvaggini, R.; Taticchi, A.; Esposto, S.; Montedoro, G. Volatile compounds in virgin olive oil: Occurrence and their relationship with the quality. J. Chromatogr. A 2004, 1054, 17–31. [Google Scholar] [CrossRef]
  56. Kalua, C.M.; Allen, M.S.; Bedgood, D.R.; Bishop, A.G.; Prenzler, P.D.; Robards, K. Olive oil volatile compounds, flavour development and quality: A critical review. Food Chem. 2007, 100, 273–286. [Google Scholar] [CrossRef]
  57. Zamuz, S.; Purriños, L.; Tomasevic, I.; Domínguez, R.; Brnčić, M.; Barba, F.J.; Lorenzo, J.M. Consumer acceptance and quality parameters of the commercial olive oils manufactured with cultivars grown in Galicia (NW Spain). Foods 2020, 9, 427. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Romero, N.; Saavedra, J.; Tapia, F.; Sepúlveda, B.; Aparicio, R. Influence of agroclimatic parameters on phenolic and volatile compounds of Chilean virgin olive oils and characterization based on geographical origin, cultivar and ripening stage. J. Sci. Food Agric. 2016, 96, 583–592. [Google Scholar] [CrossRef] [PubMed]
  59. Zhou, Q.; Liu, S.; Liu, Y.; Song, H. Comparison of flavour fingerprint, electronic nose and multivariate analysis for discrimination of extra virgin olive oils. R. Soc. Open Sci. 2019, 6, 190002. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Tura, D.; Failla, O.; Bassi, D.; Pedò, S.; Serraiocco, A. Cultivar influence on virgin olive (Olea europaea) oil flavor based on aromatic compounds and sensorial profile. Sci. Hortic. 2008, 118, 139–148. [Google Scholar] [CrossRef]
  61. Caporaso, N. Virgin olive oils: Environmental conditions, agronomical factors and processing technology affecting the chemistry of flavor profile. J. Food Chem. Nanotechnol. 2016, 2, 21–31. [Google Scholar] [CrossRef]
  62. Raffo, A.; Bucci, R.; D’Aloise, A.; Pastore, G. Combined effects of reduced malaxation oxygen levels and storage time on extra-virgin olive oil volatiles investigated by a novel chemometric approach. Food Chem. 2015, 182, 257–267. [Google Scholar] [CrossRef]
  63. Vidal, A.M.; Alcalá, S.; De Torres, A.; Moya, M.; Espínola, J.M.; Espínola, F. Fresh and aromatic virgin olive oil obtained from Arbequina, Koroneiki, and Arbosana cultivars. Molecules 2019, 24, 3587. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Hachicha Hbaieb, R.; Kotti, F.; Gargouri, M.; Msallem, M.; Vichi, S. Ripening and storage conditions of chétoui and arbequina olives: Part I. Effect on olive oils volatiles profile. Food Chem. 2016, 203, 548–558. [Google Scholar] [CrossRef] [PubMed]
  65. Lukić, M.; Lukić, I.; Sladonja, B.; Piližota, V. Policosanol variation in olive oil as a result of variety, ripening, and storage. Eur. J. Lipid Sci. Technol. 2015, 117, 1248–1260. [Google Scholar] [CrossRef]
  66. Aparicio, R.; Luna, G. Characterisation of monovarietal virgin olive oils. Eur. J. Lipid Sci. Technol. 2002, 104, 614–627. [Google Scholar] [CrossRef]
  67. Giuffrè, A.M. The Effects of cultivar and harvest year on the fatty alcohol composition of olive oils from Southwest Calabria (Italy). Grasas Aceites 2014, 65, e011. [Google Scholar] [CrossRef] [Green Version]
  68. Orozco-Solano, M.; Ruiz-Jiménez, J.; Luque de Castro, M.D. Ultrasound-assisted extraction and derivatization of sterols and fatty alcohols from olive leaves and drupes prior to determination by gas chromatography-tandem mass spectrometry. J. Chromatogr. A 2010, 1217, 1227–1235. [Google Scholar] [CrossRef] [PubMed]
  69. Krichène, D.; Allalout, A.; Salvador, M.D.; Fregapane, G.; Zarrouk, M. Fatty acids, volatiles, sterols and triterpenic alcohols of six monovarietal Tunisian virgin olive oils. Eur. J. Lipid Sci. Technol. 2010, 112, 400–409. [Google Scholar] [CrossRef]
  70. Giuffrè, A.M. Evolution of fatty alcohols in olive oils produced in Calabria (Southern Italy) during fruit ripening. J. Oleo Sci. 2014, 63, 485–496. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Gonzales-Vigil, E.; Hefer, C.A.; von Loessl, M.E.; La Mantia, J.; Mansfield, S.D. Exploiting natural variation to uncover an alkene biosynthetic enzyme in poplar. Plant Cell 2017, 29, 2000–2015. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Giuffrè, A.M. The Effect of cultivar and harvest season on the n-alkane and the n-alkene composition of virgin olive oil. Eur. Food Res. Technol. 2021, 247, 25–36. [Google Scholar] [CrossRef]
  73. Giuffrè, A.M. N-Alkanes and n-alkenes in virgin olive oil from calabria (south Italy): The effects of cultivar and harvest date. Foods 2021, 10, 290. [Google Scholar] [CrossRef]
  74. Srbinovska, A.; Conchione, C.; Menegoz Ursol, L.; Lucci, P.; Moret, S. Occurrence of n-alkanes in vegetable oils and their analytical determination. Foods 2020, 9, 1546. [Google Scholar] [CrossRef]
  75. Mihailova, A.; Abbado, D.; Kelly, S.D.; Pedentchouk, N. The impact of environmental factors on molecular and stable isotope compositions of n-alkanes in Mediterranean extra virgin olive oils. Food Chem. 2015, 173, 114–121. [Google Scholar] [CrossRef]
  76. Diamantakos, P.; Ioannidis, K.; Papanikolaou, C.; Tsolakou, A.; Rigakou, A.; Melliou, E.; Magiatis, P. A New definition of the term “high-phenolic olive oil” based on large scale statistical data of Greek olive oils analyzed by QNMR. Molecules 2021, 26, 1115. [Google Scholar] [CrossRef]
  77. Faghim, J.; Mohamed, M.B.; Bagues, M.; Guasmi, F.; Triki, T.; Nagaz, K. Irrigation effects on phenolic profile and extra virgin olive oil quality of “‘chemlali’” variety grown in South Tunisia. S. Afr. J. Bot. 2021, 141, 322–329. [Google Scholar] [CrossRef]
  78. Gelmini, F.; Ruscica, M.; Macchi, C.; Bianchi, V.; Maffei Facino, R.; Beretta, G.; Magni, P. Unsaponifiable fraction of unripe fruits of Olea europaea: An interesting source of anti-inflammatory constituents. Planta Med. 2016, 82, 273–278. [Google Scholar] [CrossRef] [Green Version]
  79. Velázquez-Palmero, D.; Romero-Segura, C.; García-Rodríguez, R.; Hernández, M.L.; Vaistij, F.E.; Graham, I.A.; Pérez, A.G.; Martínez-Rivas, J.M. An oleuropein β-glucosidase from olive fruit is involved in determining the phenolic composition of virgin olive oil. Front. Plant Sci. 2017, 8, 1902. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Caipo, L.; Sandoval, A.; Sepúlveda, B.; Fuentes, E.; Valenzuela, R.; Metherel, A.H.; Romero, N. Effect of storage conditions on the quality of arbequina extra virgin olive oil and the impact on the composition of flavor-related compounds (phenols and volatiles). Foods 2021, 10, 2161. [Google Scholar] [CrossRef]
  81. Krichene, D.; Salvador, M.D.; Fregapane, G. Stability of virgin olive oil phenolic compounds during long-term storage (18 months) at temperatures of 5–50 °C. J. Agric. Food Chem. 2015, 63, 6779–6786. [Google Scholar] [CrossRef]
  82. Castillo-Luna, A.; Criado-Navarro, I.; Ledesma-Escobar, C.A.; López-Bascón, M.A.; Priego-Capote, F. The decrease in the health benefits of extra virgin olive oil during storage is conditioned by the initial phenolic profile. Food Chem. 2021, 336, 127730. [Google Scholar] [CrossRef] [PubMed]
  83. Abbattista, R.; Ventura, G.; Calvano, C.D.; Cataldi, T.R.I.; Losito, I. Bioactive compounds in waste by-products from olive oil production: Applications and structural characterization by mass spectrometry techniques. Foods 2021, 10, 1236. [Google Scholar] [CrossRef]
  84. Centrone, M.; D’Agostino, M.; Difonzo, G.; De Bruno, A.; Di Mise, A.; Ranieri, M.; Montemurro, C.; Valenti, G.; Poiana, M.; Caponio, F.; et al. Antioxidant efficacy of olive by-product extracts in human colon HCT8 cells. Foods 2021, 10, 11. [Google Scholar] [CrossRef]
  85. Mallamaci, R.; Budriesi, R.; Clodoveo, M.L.; Biotti, G.; Micucci, M.; Ragusa, A.; Curci, F.; Muraglia, M.; Corbo, F.; Franchini, C. Olive tree in circular economy as a source of secondary metabolites active for human and animal health beyond oxidative stress and inflammation. Molecules 2021, 26, 1072. [Google Scholar] [CrossRef] [PubMed]
  86. Ranalli, A.; Contento, S.; Lucera, L.; Di Febo, M.; Marchegiani, D.; Di Fonzo, V. Factors affecting the contents of iridoid oleuropein in olive leaves (Olea europaea L.). J. Agric. Food Chem. 2006, 54, 434–440. [Google Scholar] [CrossRef]
  87. Ghomari, O.; Sounni, F.; Massaoudi, Y.; Ghanam, J.; Drissi Kaitouni, L.B.; Merzouki, M.; Benlemlih, M. Phenolic profile (HPLC-UV) of olive leaves according to extraction procedure and assessment of antibacterial activity. Biotechnol. Rep. 2019, 23, e00347. [Google Scholar] [CrossRef] [PubMed]
  88. Lukić, I.; Pasković, I.; Žurga, P.; Majetić Germek, V.; Brkljača, M.; Marcelić, Š.; Ban, D.; Grozić, K.; Lukić, M.; Užila, Z.; et al. Determination of the variability of biophenols and mineral nutrients in olive leaves with respect to cultivar, collection period and geographical location for their targeted and well-timed exploitation. Plants 2020, 9, 1667. [Google Scholar] [CrossRef] [PubMed]
  89. Alemán-Jiménez, C.; Domínguez-Perles, R.; Medina, S.; Prgomet, I.; López-González, I.; Simonelli-Muñoz, A.; Campillo-Cano, M.; Auñón, D.; Ferreres, F.; Gil-Izquierdo, Á. Pharmacokinetics and bioavailability of hydroxytyrosol are dependent on the food matrix in humans. Eur. J. Nutr. 2021, 60, 905–915. [Google Scholar] [CrossRef]
  90. Pacheco, C.; Gonzalez, E.; Robert, P.; Parada, J. Retention and pre-colon bioaccessibility of oleuropein in starchy food matrices, and the effect of microencapsulation by using inulin. J. Funct. Foods 2018, 41, 112–117. [Google Scholar] [CrossRef]
  91. Murador, D.; Braga, A.R.; Da Cunha, D.; De Rosso, V. Alterations in phenolic compound levels and antioxidant activity in response to cooking technique effects: A meta-analytic investigation. Crit. Rev. Food Sci. Nutr. 2018, 58, 169–177. [Google Scholar] [CrossRef] [PubMed]
  92. Del Pilar Ramírez-Anaya, J.; Castañeda-Saucedo, M.C.; Olalla-Herrera, M.; Villalón-Mir, M.; López-García de la Serrana, H.; Samaniego-Sánchez, C. Changes in the antioxidant properties of extra virgin olive oil after cooking typical Mediterranean vegetables. Antioxidants 2019, 8, 246. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Brenes, M.; García, A.; Dobarganes, M.C.; Velasco, J.; Romero, C. Influence of thermal treatments simulating cooking processes on the polyphenol content in virgin olive oil. J. Agric. Food Chem. 2002, 50, 5962–5967. [Google Scholar] [CrossRef]
  94. Andrikopoulos, N.K.; Dedoussis, G.V.Z.; Falirea, A.; Kalogeropoulos, N.; Hatzinikola, H.S. Deterioration of natural antioxidant species of vegetable edible oils during the domestic deep-frying and pan-frying of potatoes. Int. J. Food Sci. Nutr. 2002, 53, 351–363. [Google Scholar] [CrossRef] [PubMed]
  95. Lozano-Castellón, J.; Vallverdú-Queralt, A.; Rinaldi de Alvarenga, J.F.; Illán, M.; Torrado-Prat, X.; Lamuela-Raventós, R.M. Domestic sautéing with EVOO: Change in the phenolic profile. Antioxidants 2020, 9, 77. [Google Scholar] [CrossRef] [Green Version]
  96. Abril, D.; Mirabal-Gallardo, Y.; González, A.; Marican, A.; Durán-Lara, E.F.; Silva Santos, L.; Valdés, O. Comparison of the oxidative stability and antioxidant activity of extra-virgin olive oil and oils extracted from seeds of colliguaya integerrima and cynara cardunculus under normal conditions and after thermal treatment. Antioxidants 2019, 8, 470. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Giuffrè, A.M.; Caracciolo, M.; Aappia, C.; Capocasale, M.; Poiana, M. Effect of heating on chemical parameters of extra virgin olive oil, pomace olive oil, soybean oil and palm oil. Ital. J. Food Sci. 2018, 30. [Google Scholar] [CrossRef]
  98. Santos, C.S.P.; García, L.M.; Cruz, R.; Cunha, S.C.; Fernandes, J.O.; Casal, S. Impact of potatoes deep-frying on common monounsaturated-rich vegetable oils: A comparative study. J. Food Sci. Technol. 2019, 56, 290–301. [Google Scholar] [CrossRef] [PubMed]
  99. Marmesat, S.; Morales, A.; Velasco, J.; Dobarganes, M.C. Action and fate of natural and synthetic antioxidants during frying. Grasas Aceites 2010, 61, 333–340. [Google Scholar] [CrossRef] [Green Version]
  100. Zotos, A.; Kotaras, A.; Mikras, E. Effect of baking of sardine (Sardina pilchardus) and frying of anchovy (engraulis encrasicholus) in olive and sunflower oil on their quality. Food Sci. Technol. Int. 2013, 19, 11–23. [Google Scholar] [CrossRef] [PubMed]
  101. Gómez-Alonso, S.; Fregapane, G.; Salvador, M.D.; Gordon, M.H. Changes in phenolic composition and antioxidant activity of virgin olive oil during frying. J. Agric. Food Chem. 2003, 51, 667–672. [Google Scholar] [CrossRef] [PubMed]
  102. Ambra, R.; Lucchetti, S.; Pastore, G. A Review of the effects of olive oil-cooking on phenolic compounds. Molecules 2022, 27, 661. [Google Scholar] [CrossRef]
  103. Giuffrè, A.M.; Capocasale, M.; Macrì, R.; Caracciolo, M.; Zappia, C.; Poiana, M. Volatile profiles of extra virgin olive oil, olive pomace oil, soybean oil and palm oil in different heating conditions. LWT 2020, 117, 108631. [Google Scholar] [CrossRef]
  104. Caponio, F.; Pasqualone, A.; Gomes, T. Effects of conventional and microwave heating on the degradation of olive oil. Eur. Food Res. Technol. 2002, 215, 114–117. [Google Scholar] [CrossRef]
  105. Kishimoto, N. Microwave heating induces oxidative degradation of extra virgin olive oil. Food Sci. Technol. Res. 2019, 25, 75–79. [Google Scholar] [CrossRef]
  106. Caponio, F.; Giarnetti, M.; Paradiso, V.M.; Summo, C.; Gomes, T. Potential use of extra virgin olive oil in bakery products rich in fats: A comparative study with refined oils. Int. J. Food Sci. Technol. 2013, 48, 82–88. [Google Scholar] [CrossRef]
  107. Giuffrè, A.M.; Caracciolo, M.; Capocasale, M.; Zappia, C.; Poiana, M. Effects of shortening replacement with extra virgin olive oil on the physical–chemical–sensory properties of Italian cantuccini biscuits. Foods 2022, 11, 299. [Google Scholar] [CrossRef] [PubMed]
  108. Di Nunzio, M.; Picone, G.; Pasini, F.; Chiarello, E.; Caboni, M.F.; Capozzi, F.; Gianotti, A.; Bordoni, A. Olive oil by-product as functional ingredient in bakery products. influence of processing and evaluation of biological effects. Food Res. Int. 2020, 131, 108940. [Google Scholar] [CrossRef] [Green Version]
  109. Peyrot des Gachons, C.; O’Keefe, A.J.; Slade, L.; Beauchamp, G.K. Protein suppresses both bitterness and oleocanthal-elicited pungency of extra virgin olive oil. Sci. Rep. 2021, 11, 11851. [Google Scholar] [CrossRef] [PubMed]
  110. Spinelli, S.; Prescott, J.; Pierguidi, L.; Dinnella, C.; Arena, E.; Braghieri, A.; Di Monaco, R.; Toschi, T.G.; Endrizzi, I.; Proserpio, C.; et al. Phenol-rich food acceptability: The influence of variations in sweetness optima and sensory-liking patterns. Nutrients 2021, 13, 866. [Google Scholar] [CrossRef] [PubMed]
  111. De Bock, M.; Thorstensen, E.B.; Derraik, J.G.B.; Henderson, H.V.; Hofman, P.L.; Cutfield, W.S. Human absorption and metabolism of oleuropein and hydroxytyrosol ingested as olive (Olea europaea L.) leaf extract. Mol. Nutr. Food Res. 2013, 57, 2079–2085. [Google Scholar] [CrossRef] [PubMed]
  112. Kerimi, A.; Nyambe-Silavwe, H.; Pyner, A.; Oladele, E.; Gauer, J.S.; Stevens, Y.; Williamson, G. Nutritional implications of olives and sugar: Attenuation of post-prandial glucose spikes in healthy volunteers by inhibition of sucrose hydrolysis and glucose transport by oleuropein. Eur. J. Nutr. 2019, 58, 1315–1330. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Vissers, M.N.; Zock, P.L.; Roodenburg, A.J.C.; Leenen, R.; Katan, M.B. Olive oil phenols are absorbed in humans. J. Nutr. 2002, 132, 409–417. [Google Scholar] [CrossRef]
  114. Sizar, O.; Nassereddin, A.; Talati, R. Ezetimibe. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2022. [Google Scholar]
  115. Ballantyne, C.M.; Hoogeveen, R.C.; Raya, J.L.; Cain, V.A.; Palmer, M.K.; Karlson, B.W. GRAVITY study investigators efficacy, safety and effect on biomarkers related to cholesterol and lipoprotein metabolism of rosuvastatin 10 or 20 mg plus ezetimibe 10 mg vs. simvastatin 40 or 80 mg plus ezetimibe 10 mg in high-risk patients: Results of the GRAVITY randomized study. Atherosclerosis 2014, 232, 86–93. [Google Scholar] [CrossRef] [PubMed]
  116. Thongtang, N.; Lin, J.; Schaefer, E.J.; Lowe, R.S.; Tomassini, J.E.; Shah, A.K.; Tershakovec, A.M. Effects of ezetimibe added to statin therapy on markers of cholesterol absorption and synthesis and LDL-c lowering in hyperlipidemic patients. Atherosclerosis 2012, 225, 388–396. [Google Scholar] [CrossRef] [Green Version]
  117. Malina, D.M.T.; Fonseca, F.A.; Barbosa, S.A.; Kasmas, S.H.; Machado, V.A.; França, C.N.; Borges, N.C.; Moreno, R.A.; Izar, M.C. Additive effects of plant sterols supplementation in addition to different lipid-lowering regimens. J. Clin. Lipidol. 2015, 9, 542–552. [Google Scholar] [CrossRef]
  118. Aponte, M.; Ungaro, F.; d’Angelo, I.; De Caro, C.; Russo, R.; Blaiotta, G.; Dal Piaz, F.; Calignano, A.; Miro, A. Improving in vivo conversion of oleuropein into hydroxytyrosol by oral granules containing probiotic Lactobacillus plantarum 299v and an Olea europaea standardized extract. Int. J. Pharm. 2018, 543, 73–82. [Google Scholar] [CrossRef]
  119. Duque-Soto, C.; Quirantes-Piné, R.; Borrás-Linares, I.; Segura-Carretero, A.; Lozano-Sánchez, J. Characterization and influence of static in vitro digestion on bioaccessibility of bioactive polyphenols from an olive leaf extract. Foods 2022, 11, 743. [Google Scholar] [CrossRef] [PubMed]
  120. Reboredo-Rodríguez, P.; González-Barreiro, C.; Martínez-Carballo, E.; Cambeiro-Pérez, N.; Rial-Otero, R.; Figueiredo-González, M.; Cancho-Grande, B. Applicability of an in-vitro digestion model to assess the bioaccessibility of phenolic compounds from olive-related products. Molecules 2021, 26, 6667. [Google Scholar] [CrossRef] [PubMed]
  121. Zorić, N.; Kopjar, N.; Rodriguez, J.V.; Tomić, S.; Kosalec, I. Protective effects of olive oil phenolics oleuropein and hydroxytyrosol against hydrogen peroxide-induced DNA damage in human peripheral lymphocytes. Acta Pharm. 2021, 71, 131–141. [Google Scholar] [CrossRef] [PubMed]
  122. Umeno, A.; Takashima, M.; Murotomi, K.; Nakajima, Y.; Koike, T.; Matsuo, T.; Yoshida, Y. Radical-scavenging activity and antioxidative effects of olive leaf components oleuropein and hydroxytyrosol in comparison with homovanillic alcohol. J. Oleo Sci. 2015, 64, 793–800. [Google Scholar] [CrossRef] [Green Version]
  123. Khymenets, O.; Fitó, M.; Touriño, S.; Muñoz-Aguayo, D.; Pujadas, M.; Torres, J.L.; Joglar, J.; Farré, M.; Covas, M.-I.; de la Torre, R. Antioxidant activities of hydroxytyrosol main metabolites do not contribute to beneficial health effects after olive oil ingestion. Drug Metab. Dispos. 2010, 38, 1417–1421. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Boronat, A.; Mateus, J.; Soldevila-Domenech, N.; Guerra, M.; Rodríguez-Morató, J.; Varon, C.; Muñoz, D.; Barbosa, F.; Morales, J.C.; Gaedigk, A.; et al. Cardiovascular benefits of tyrosol and its endogenous conversion into hydroxytyrosol in humans. A Randomized, controlled trial. Free Radic. Biol. Med. 2019, 143, 471–481. [Google Scholar] [CrossRef] [PubMed]
  125. Ostlund, R.E.; McGill, J.B.; Zeng, C.-M.; Covey, D.F.; Stearns, J.; Stenson, W.F.; Spilburg, C.A. Gastrointestinal absorption and plasma kinetics of soy delta(5)-phytosterols and phytostanols in humans. Am. J. Physiol. Endocrinol. Metab. 2002, 282, E911–E916. [Google Scholar] [CrossRef]
  126. Poli, A.; Marangoni, F.; Corsini, A.; Manzato, E.; Marrocco, W.; Martini, D.; Medea, G.; Visioli, F. Phytosterols, cholesterol control, and cardiovascular disease. Nutrients 2021, 13, 2810. [Google Scholar] [CrossRef] [PubMed]
  127. Salehi, B.; Quispe, C.; Sharifi-Rad, J.; Cruz-Martins, N.; Nigam, M.; Mishra, A.P.; Konovalov, D.A.; Orobinskaya, V.; Abu-Reidah, I.M.; Zam, W.; et al. Phytosterols: From preclinical evidence to potential clinical applications. Front. Pharm. 2020, 11, 599959. [Google Scholar] [CrossRef] [PubMed]
  128. Cicerale, S.; Lucas, L.; Keast, R. Biological activities of phenolic compounds present in virgin olive oil. Int. J. Mol. Sci. 2010, 11, 458–479. [Google Scholar] [CrossRef] [Green Version]
  129. Cai, W.; Calder, P.C.; Cury-Boaventura, M.F.; De Waele, E.; Jakubowski, J.; Zaloga, G. Biological and clinical aspects of an olive oil-based lipid emulsion-a review. Nutrients 2018, 10, 776. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  130. Marcelino, G.; Hiane, P.A.; de Cássia Freitas, K.; Santana, L.F.; Pott, A.; Donadon, J.R.; de Cássia Avellaneda Guimarães, R. Effects of olive oil and its minor components on cardiovascular diseases, inflammation, and gut microbiota. Nutrients 2019, 11, 1826. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  131. Rodríguez-Rodríguez, R.; Herrera, M.D.; Perona, J.S.; Ruiz-Gutiérrez, V. Potential vasorelaxant effects of oleanolic acid and erythrodiol, two triterpenoids contained in “orujo” olive oil, on rat aorta. Br. J. Nutr. 2004, 92, 635–642. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Luna-Vázquez, F.J.; Ibarra-Alvarado, C.; Rojas-Molina, A.; Romo-Mancillas, A.; López-Vallejo, F.H.; Solís-Gutiérrez, M.; Rojas-Molina, J.I.; Rivero-Cruz, F. Role of nitric oxide and hydrogen sulfide in the vasodilator effect of ursolic acid and uvaol from black cherry prunus serotina fruits. Molecules 2016, 21, 78. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. El Haouari, M.; Quintero, J.E.; Rosado, J.A. Anticancer molecular mechanisms of oleocanthal. Phytother. Res. 2020, 34, 2820–2834. [Google Scholar] [CrossRef] [PubMed]
  134. Omar, S.H. Oleuropein in olive and its pharmacological effects. Sci. Pharm. 2010, 78, 133–154. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Sudjana, A.N.; D’Orazio, C.; Ryan, V.; Rasool, N.; Ng, J.; Islam, N.; Riley, T.V.; Hammer, K.A. Antimicrobial activity of commercial Olea europaea (olive) leaf extract. Int. J. Antimicrobal Agents 2009, 33, 461–463. [Google Scholar] [CrossRef] [PubMed]
  136. Giamarellos-Bourboulis, E.J.; Geladopoulos, T.; Chrisofos, M.; Koutoukas, P.; Vassiliadis, J.; Alexandrou, I.; Tsaganos, T.; Sabracos, L.; Karagianni, V.; Pelekanou, E.; et al. Oleuropein: A novel immunomodulator conferring prolonged survival in experimental sepsis by: Pseudomonas aeruginosa. Shock 2006, 26, 410–416. [Google Scholar] [CrossRef] [PubMed]
  137. Bedoya, L.M.; Beltran, M.; Obregon-Calderon, P.; Garcia-Perez, J.; de la Torre, H.E.; Gonzalez, N.; Perez-Olmeda, M.; Aunon, D.; Capa, L.; Gomez-Acebo, E.; et al. Hydroxytyrosol: A new class of microbicide displaying broad anti-HIV-1 activity. Aids 2016, 30, 2767–2776. [Google Scholar] [CrossRef] [Green Version]
  138. Melguizo-Rodríguez, L.; Manzano-Moreno, F.J.; Illescas-Montes, R.; Ramos-Torrecillas, J.; de Luna-Bertos, E.; Ruiz, C.; García-Martínez, O. Bone protective effect of extra-virgin olive oil phenolic compounds by modulating osteoblast gene expression. Nutrients 2019, 11, 1722. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Melguizo-Rodríguez, L.; Manzano-Moreno, F.J.; De Luna-Bertos, E.; Rivas, A.; Ramos-Torrecillas, J.; Ruiz, C.; García-Martínez, O. Effect of olive oil phenolic compounds on osteoblast differentiation. Eur. J. Clin. Investig. 2018, 48, e12904. [Google Scholar] [CrossRef] [PubMed]
  140. Melguizo-Rodríguez, L.; Ramos-Torrecillas, J.; Manzano-Moreno, F.J.; Illescas-Montes, R.; Rivas, A.; Ruiz, C.; De Luna-Bertos, E.; García-Martínez, O. Effect of Phenolic extracts from different extra-virgin olive oil varieties on osteoblast-like cells. PLoS ONE 2018, 13, e0196530. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  141. Janahmadi, Z.; Nekooeian, A.A.; Moaref, A.R.; Emamghoreishi, M. Oleuropein offers cardioprotection in rats with acute myocardial infarction. Cardiovasc. Toxicol. 2015, 15, 61–68. [Google Scholar] [CrossRef] [PubMed]
  142. Janahmadi, Z.; Nekooeian, A.A.; Moaref, A.R.; Emamghoreishi, M. Oleuropein attenuates the progression of heart failure in rats by antioxidant and antiinflammatory effects. Naunyn-Schmiedebergs Arch. Pharmacol. 2017, 390, 245–252. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Tsoumani, M.; Georgoulis, A.; Nikolaou, P.-E.; Kostopoulos, I.V.; Dermintzoglou, T.; Papatheodorou, I.; Zoga, A.; Efentakis, P.; Konstantinou, M.; Gikas, E.; et al. Acute administration of the olive constituent, oleuropein, combined with ischemic postconditioning increases myocardial protection by modulating oxidative defense. Free Radic. Biol. Med. 2021, 166, 18–32. [Google Scholar] [CrossRef] [PubMed]
  144. Jin, H.-X.; Zhang, Y.-H.; Guo, R.-N.; Zhao, S.-N. Inhibition of MEK/ERK/STAT3 signaling in oleuropein treatment inhibits myocardial ischemia/reperfusion. Int. J. Mol. Med. 2018, 42, 1034–1043. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Mnafgui, K.; Khlif, I.; Hajji, R.; Derbali, F.; Kraiem, F.; Ellefi, H.; Michel, T.; Halabalaki, M.; Skaltsounis, A.L.; Elfeki, A.; et al. Preventive effects of oleuropein against cardiac remodeling after myocardial infarction in wistar rat through inhibiting angiotensin-converting enzyme activity. Toxicol. Mech. Methods 2015, 25, 538–546. [Google Scholar] [CrossRef] [PubMed]
  146. Andreadou, I.; Mikros, E.; Ioannidis, K.; Sigala, F.; Naka, K.; Kostidis, S.; Farmakis, D.; Tenta, R.; Kavantzas, N.; Bibli, S.-I.; et al. Oleuropein prevents doxorubicin-induced cardiomyopathy interfering with signaling molecules and cardiomyocyte metabolism. J. Mol. Cell. Cardiol. 2014, 69, 4–16. [Google Scholar] [CrossRef]
  147. Zhang, J.-Y.; Yang, Z.; Fang, K.; Shi, Z.-L.; Ren, D.-H.; Sun, J. Oleuropein prevents the development of experimental autoimmune myocarditis in rats. Int. Immunopharmacol. 2017, 48, 187–195. [Google Scholar] [CrossRef]
  148. Xing, C.; Xu, L.; Yao, Y. Beneficial role of oleuropein in sepsis-induced myocardial injury. Possible involvement of GSK-3 beta/NF-KB pathway. Acta Cirúrgica Bras. 2021, 36, e360107. [Google Scholar] [CrossRef] [PubMed]
  149. Xu, Y.; Wu, L.; Chen, A.; Xu, C.; Feng, Q. Protective effects of olive leaf extract on acrolein-exacerbated myocardial infarction via an endoplasmic reticulum stress pathway. Int. J. Mol. Sci. 2018, 19, 20493. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  150. Ebaid, G.M.X.; Seiva, F.R.F.; Rocha, K.K.H.R.; Souza, G.A.; Novelli, E.L.B. Effects of olive oil and its minor phenolic constituents on obesity-induced cardiac metabolic changes. Nutr. J. 2010, 9, 46. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  151. Leri, M.; Oropesa-Nunez, R.; Canale, C.; Raimondi, S.; Giorgetti, S.; Bruzzone, E.; Bellotti, V.; Stefani, M.; Bucciantini, M. Oleuropein aglycone: A polyphenol with different targets against amyloid toxicity. Biochim. Biophys. Acta—Gen. Subj. 2018, 1862, 1432–1442. [Google Scholar] [CrossRef] [PubMed]
  152. Keys, A.; Menotti, A.; Karvonen, M.J.; Aravanis, C.; Blackburn, H.; Buzina, R.; Djordjevic, B.S.; Dontas, A.S.; Fidanza, F.; Keys, M.H. The diet and 15-year death rate in the Seven Countries study. Am. J. Epidemiol. 1986, 124, 903–915. [Google Scholar] [CrossRef] [PubMed]
  153. Mangas-Cruz, M.A.; Martínez-Brocca, M.; Ortiz-Leyba, C.; Garnacho-Montero, J.; Cunill, J.L.P.; García-Luna, P.P. Olive oil in clinical nutrition. Grasas Aceites 2004, 55, 76–83. [Google Scholar] [CrossRef]
  154. Moreno-Luna, R.; Muñoz-Hernandez, R.; Miranda, M.L.; Costa, A.F.; Jimenez-Jimenez, L.; Vallejo-Vaz, A.J.; Muriana, F.J.G.; Villar, J.; Stiefel, P. Olive oil polyphenols decrease blood pressure and improve endothelial function in young women with mild hypertension. Am. J. Hypertens. 2012, 25, 1299–1304. [Google Scholar] [CrossRef] [Green Version]
  155. Fitó, M.; Cladellas, M.; de la Torre, R.; Martí, J.; Alcántara, M.; Pujadas-Bastardes, M.; Marrugat, J.; Bruguera, J.; López-Sabater, M.C.; Vila, J.; et al. Antioxidant effect of virgin olive oil in patients with stable coronary heart disease: A randomized, crossover, controlled, clinical trial. Atherosclerosis 2005, 181, 149–158. [Google Scholar] [CrossRef] [PubMed]
  156. Ditano-Vázquez, P.; Torres-Peña, J.D.; Galeano-Valle, F.; Pérez-Caballero, A.I.; Demelo-Rodríguez, P.; Lopez-Miranda, J.; Katsiki, N.; Delgado-Lista, J.; Alvarez-Sala-Walther, L.A. The fluid aspect of the Mediterranean diet in the prevention and management of cardiovascular disease and diabetes: The role of polyphenol content in moderate consumption of wine and olive oil. Nutrients 2019, 11, 2833. [Google Scholar] [CrossRef] [Green Version]
  157. Gomez-Marin, B.; Gomez-Delgado, F.; Lopez-Moreno, J.; Alcala-Diaz, J.F.; Jimenez-Lucena, R.; Torres-Peña, J.D.; Garcia-Rios, A.; Ortiz-Morales, A.M.; Yubero-Serrano, E.M.; Del Mar Malagon, M.; et al. Long-term consumption of a Mediterranean diet improves postprandial lipemia in patients with type 2 diabetes: The CORDIOPREV randomized trial. Am. J. Clin. Nutr. 2018, 108, 963–970. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Camargo, A.; Ruano, J.; Fernandez, J.M.; Parnell, L.D.; Jimenez, A.; Santos-Gonzalez, M.; Marin, C.; Perez-Martinez, P.; Uceda, M.; Lopez-Miranda, J.; et al. Gene expression changes in mononuclear cells in patients with metabolic syndrome after acute intake of phenol-rich virgin olive oil. BMC Genom. 2010, 11, 253. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  159. Castañer, O.; Covas, M.-I.; Khymenets, O.; Nyyssonen, K.; Konstantinidou, V.; Zunft, H.-F.; de la Torre, R.; Muñoz-Aguayo, D.; Vila, J.; Fitó, M. Protection of LDL from oxidation by olive oil polyphenols is associated with a downregulation of CD40-ligand expression and its downstream products in vivo in humans. Am. J. Clin. Nutr. 2012, 95, 1238–1244. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  160. Weinbrenner, T.; Fitó, M.; de la Torre, R.; Saez, G.T.; Rijken, P.; Tormos, C.; Coolen, S.; Albaladejo, M.F.; Abanades, S.; Schroder, H.; et al. Olive oils high in phenolic compounds modulate oxidative/antioxidative status in men. J. Nutr. 2004, 134, 2314–2321. [Google Scholar] [CrossRef] [Green Version]
  161. Gimeno, E.; Fitó, M.; Lamuela-Raventós, R.M.; Castellote, A.I.; Covas, M.; Farré, M.; de La Torre-Boronat, M.C.; López-Sabater, M.C. Effect of ingestion of virgin olive oil on human low-density lipoprotein composition. Eur. J. Clin. Nutr. 2002, 56, 114–120. [Google Scholar] [CrossRef] [Green Version]
  162. Luomala, M.; Laaksonen, R.; Janatuinen, T.; Vesalainen, R.; Nuutila, P.; Saarela, M.; Mattila, K.; Kalijärvi, M.; Solakivi, T.; Knuuti, J.; et al. High plasma levels of CD40 are associated with low coenzyme Q and vitamin E content of low-density lipoprotein in healthy men. Scand. J. Clin. Lab. Investig. 2007, 67, 115–122. [Google Scholar] [CrossRef]
  163. Sarapis, K.; George, E.S.; Marx, W.; Mayr, H.L.; Willcox, J.; Esmaili, T.; Powell, K.L.; Folasire, O.S.; Lohning, A.E.; Garg, M.; et al. Extra virgin olive oil high in polyphenols improves antioxidant status in adults: A double-blind, randomized, controlled, cross-over study (OLIVAUS). Eur. J. Nutr. 2022, 61, 1073–1086. [Google Scholar] [CrossRef] [PubMed]
  164. Schwingshackl, L.; Krause, M.; Schmucker, C.; Hoffmann, G.; Rücker, G.; Meerpohl, J.J. Impact of different types of olive oil on cardiovascular risk factors: A systematic review and network meta-analysis. Nutr. Metab. Cardiovasc. Dis. 2019, 29, 1030–1039. [Google Scholar] [CrossRef] [PubMed]
  165. Estruch, R.; Ros, E.; Salas-Salvadó, J.; Covas, M.-I.; Corella, D.; Arós, F.; Gómez-Gracia, E.; Ruiz-Gutiérrez, V.; Fiol, M.; Lapetra, J.; et al. Primary prevention of cardiovascular disease with a Mediterranean diet supplemented with extra-virgin olive oil or nuts. N. Engl. J. Med. 2018, 378, e34. [Google Scholar] [CrossRef] [PubMed]
  166. Guasch-Ferré, M.; Hu, F.B.; Martínez-González, M.A.; Fitó, M.; Bulló, M.; Estruch, R.; Ros, E.; Corella, D.; Recondo, J.; Gómez-Gracia, E.; et al. Olive oil intake and risk of cardiovascular disease and mortality in the PREDIMED study. BMC Med. 2014, 12, 78. [Google Scholar] [CrossRef] [Green Version]
  167. Becerra-Tomás, N.; Blanco Mejía, S.; Viguiliouk, E.; Khan, T.; Kendall, C.W.C.; Kahleova, H.; Rahelić, D.; Sievenpiper, J.L.; Salas-Salvadó, J. Mediterranean diet, cardiovascular disease and mortality in diabetes: A systematic review and meta-analysis of prospective cohort studies and randomized clinical trials. Crit. Rev. Food Sci. Nutr. 2020, 60, 1207–1227. [Google Scholar] [CrossRef]
  168. Guasch-Ferré, M.; Li, Y.; Willett, W.C.; Sun, Q.; Sampson, L.; Salas-Salvadó, J.; Martínez-González, M.A.; Stampfer, M.J.; Hu, F.B. Consumption of olive oil and risk of total and cause-specific mortality among U.S. adults. J. Am. Coll. Cardiol. 2022, 79, 101–112. [Google Scholar] [CrossRef]
  169. Zhang, Y.; Zhuang, P.; Wu, F.; He, W.; Mao, L.; Jia, W.; Zhang, Y.; Chen, X.; Jiao, J. Cooking oil/fat consumption and deaths from cardiometabolic diseases and other causes: Prospective analysis of 521,120 individuals. BMC Med. 2021, 19, 92. [Google Scholar] [CrossRef]
  170. Cicero, A.F.G.; Fogacci, F.; Grandi, E.; Rizzoli, E.; Bove, M.; D’Addato, S.; Borghi, C. Prevalent seasoning and cooking fats, arterial stiffness and blood lipid pattern in a rural population sample: Data from the Brisighella Heart Study. Nutrients 2020, 12, 3063. [Google Scholar] [CrossRef] [PubMed]
  171. De la Torre, R.; Corella, D.; Castañer, O.; Martínez-González, M.A.; Salas-Salvado, J.; Vila, J.; Estruch, R.; Sorli, J.V.; Arós, F.; Fiol, M.; et al. Protective effect of homovanillyl alcohol on cardiovascular disease and total mortality: Virgin olive oil, wine, and catechol-methylation. Am. J. Clin. Nutr. 2017, 105, 1297–1304. [Google Scholar] [CrossRef]
  172. Silva, S.; Bronze, M.R.; Figueira, M.E.; Siwy, J.; Mischak, H.; Combet, E.; Mullen, W. 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. 2015, 101, 44–54. [Google Scholar] [CrossRef] [PubMed]
  173. EFSA Panel on Dietetic Products, Nutrition and Allergies (NDA). 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 1924/2006. EFSA J. 2011, 9, 2033. [Google Scholar] [CrossRef]
  174. Farràs, M.; Canyelles, M.; Fitó, M.; Escolà-Gil, J.C. Effects of virgin olive oil and phenol-enriched virgin olive oils on lipoprotein atherogenicity. Nutrients 2020, 12, 601. [Google Scholar] [CrossRef] [Green Version]
  175. Konstantinidou, V.; Khymenets, O.; Covas, M.-I.; de la Torre, R.; Muñoz-Aguayo, D.; Anglada, R.; Farré, M.; Fito, M. Time course of changes in the expression of insulin sensitivity-related genes after an acute load of virgin olive oil. OMICS J. Integr. Biol. 2009, 13, 431–438. [Google Scholar] [CrossRef] [PubMed]
  176. Konstantinidou, V.; Covas, M.-I.; Muñoz-Aguayo, D.; Khymenets, O.; de la Torre, R.; Saez, G.; del Carmen Tormos, M.; Toledo, E.; Marti, A.; Ruiz-Gutiérrez, V.; et al. In vivo nutrigenomic effects of virgin olive oil polyphenols within the frame of the mediterranean diet: A randomized controlled trial. FASEB J. 2010, 24, 2546–2557. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  177. Farras, M.; Valls, R.M.; Fernandez-Castillejo, S.; Giralt, M.; Sola, R.; Subirana, I.; Motilva, M.-J.; Konstantinidou, V.; Covas, M.-I.; Fito, M. Olive oil polyphenols enhance the expression of cholesterol efflux related genes in vivo in humans. a randomized controlled trial. J. Nutr. Biochem. 2013, 24, 1334–1339. [Google Scholar] [CrossRef] [PubMed]
  178. Hernáez, Á.; Remaley, A.T.; Farràs, M.; Fernández-Castillejo, S.; Subirana, I.; Schröder, H.; Fernández-Mampel, M.; Muñoz-Aguayo, D.; Sampson, M.; Solà, R.; et al. Olive oil polyphenols decrease LDL concentrations and LDL atherogenicity in men in a randomized controlled trial. J. Nutr. 2015, 145, 1692–1697. [Google Scholar] [CrossRef]
  179. Fernández-Castillejo, S.; Valls, R.-M.; Castañer, O.; Rubió, L.; Catalán, Ú.; Pedret, A.; Macià, A.; Sampson, M.L.; Covas, M.-I.; Fitó, M.; et al. Polyphenol rich olive oils improve lipoprotein particle atherogenic ratios and subclasses profile: A randomized, crossover, controlled trial. Mol. Nutr. Food Res. 2016, 60, 1544–1554. [Google Scholar] [CrossRef] [Green Version]
  180. Gimeno, E.; de la Torre-Carbot, K.; Lamuela-Raventós, R.M.; Castellote, A.I.; Fitó, M.; de la Torre, R.; Covas, M.-I.; López-Sabater, M.C. Changes in the phenolic content of low density lipoprotein after olive oil consumption in men. a randomized crossover controlled trial. Br. J. Nutr. 2007, 98, 1243–1250. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  181. Dupuit, M.; Chavanelle, V.; Chassaing, B.; Perriere, F.; Etienne, M.; Plissonneau, C.; Boscaro, A.; Barnich, N.; Pialoux, V.; Maugard, T.; et al. The TOTUM-63 supplement and high-intensity interval training combination limits weight gain, improves glycemic control, and influences the composition of gut mucosa-associated bacteria in rats on a high fat diet. Nutrients 2021, 13, 1569. [Google Scholar] [CrossRef] [PubMed]
  182. Jenkins, D.J.A.; Chiavaroli, L.; Wong, J.M.W.; Kendall, C.; Lewis, G.F.; Vidgen, E.; Connelly, P.W.; Leiter, L.A.; Josse, R.G.; Lamarche, B. Adding monounsaturated fatty acids to a dietary portfolio of cholesterol-lowering foods in hypercholesterolemia. CMAJ 2010, 182, 1961–1967. [Google Scholar] [CrossRef] [Green Version]
  183. Salas-Salvadó, J.; Bulló, M.; Estruch, R.; Ros, E.; Covas, M.-I.; Ibarrola-Jurado, N.; Corella, D.; Arós, F.; Gómez-Gracia, E.; Ruiz-Gutiérrez, V.; et al. Prevention of diabetes with Mediterranean diets: A subgroup analysis of a randomized trial. Ann. Intern. Med. 2014, 160, 1–11. [Google Scholar] [CrossRef] [Green Version]
  184. Basterra-Gortari, F.J.; Ruiz-Canela, M.; Martínez-González, M.A.; Babio, N.; Sorlí, J.V.; Fito, M.; Ros, E.; Gómez-Gracia, E.; Fiol, M.; Lapetra, J.; et al. Effects of a Mediterranean eating plan on the need for glucose-lowering medications in participants with type 2 diabetes: A subgroup analysis of the PREDIMED trial. Diabetes Care 2019, 42, 1390–1397. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Assaf-Balut, C.; García de la Torre, N.; Durán, A.; Fuentes, M.; Bordiú, E.; Del Valle, L.; Familiar, C.; Ortolá, A.; Jiménez, I.; Herraiz, M.A.; et al. A Mediterranean diet with additional extra virgin olive oil and pistachios reduces the incidence of gestational diabetes mellitus (GDM): A randomized controlled trial: The St. Carlos GDM prevention study. PLoS ONE 2017, 12, e0185873. [Google Scholar] [CrossRef] [PubMed]
  186. Fortin, A.; Rabasa-Lhoret, R.; Lemieux, S.; Labonté, M.-E.; Gingras, V. Comparison of a Mediterranean to a low-fat diet intervention in adults with type 1 diabetes and metabolic syndrome: A 6-month randomized trial. Nutr. Metab. Cardiovasc. Dis. 2018, 28, 1275–1284. [Google Scholar] [CrossRef]
  187. Cadario, F.; Prodam, F.; Pasqualicchio, S.; Bellone, S.; Bonsignori, I.; Demarchi, I.; Monzani, A.; Bona, G. Lipid Profile and nutritional intake in children and adolescents with type 1 diabetes improve after a structured dietician training to a Mediterranean-style diet. J. Endocrinol. Investig. 2012, 35, 160–168. [Google Scholar] [CrossRef]
  188. Granado-Casas, M.; Martin, M.; Martínez-Alonso, M.; Alcubierre, N.; Hernández, M.; Alonso, N.; Castelblanco, E.; Mauricio, D. The Mediterranean diet is associated with an improved quality of life in adults with type 1 diabetes. Nutrients 2020, 12, 131. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  189. Álvarez-Amor, L.; Sierra, A.L.; Cárdenas, A.; López-Bermudo, L.; López-Beas, J.; Andújar, E.; Pérez-Alegre, M.; Gallego-Durán, R.; Varela, L.M.; Martin-Montalvo, A.; et al. Extra virgin olive oil improved body weight and insulin sensitivity in high fat diet-induced obese LDLr−/−.Leiden mice without attenuation of steatohepatitis. Sci. Rep. 2021, 11, 8250. [Google Scholar] [CrossRef]
  190. Angeloni, C.; Malaguti, M.; Barbalace, M.C.; Hrelia, S. Bioactivity of olive oil phenols in neuroprotection. Int. J. Mol. Sci. 2017, 18, 12230. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  191. Martínez-Lapiscina, E.H.; Clavero, P.; Toledo, E.; Estruch, R.; Salas-Salvadó, J.; San Julián, B.; Sanchez-Tainta, A.; Ros, E.; Valls-Pedret, C.; Martinez-Gonzalez, M.Á. Mediterranean diet improves cognition: The PREDIMED-NAVARRA randomised trial. J. Neurol. Neurosurg. Psychiatry 2013, 84, 1318–1325. [Google Scholar] [CrossRef] [Green Version]
  192. Konstantinidou, V.; Khymenets, O.; Fito, M.; De la Torre, R.; Anglada, R.; Dopazo, A.; Covas, M.I. Characterization of human gene expression changes after olive oil ingestion: An exploratory approach. Folia Biol. 2009, 55, 85–91. [Google Scholar]
  193. Markellos, C.; Ourailidou, M.-E.; Gavriatopoulou, M.; Halvatsiotis, P.; Sergentanis, T.N.; Psaltopoulou, T. Olive oil intake and cancer risk: A systematic review and meta-analysis. PLoS ONE 2022, 17, e0261649. [Google Scholar] [CrossRef] [PubMed]
  194. Santos-Marcos, J.A.; Haro, C.; Vega-Rojas, A.; Alcala-Diaz, J.F.; Molina-Abril, H.; Leon-Acuña, A.; Lopez-Moreno, J.; Landa, B.B.; Tena-Sempere, M.; Perez-Martinez, P.; et al. Sex differences in the gut microbiota as potential determinants of gender predisposition to disease. Mol. Nutr. Food Res. 2019, 63, e1800870. [Google Scholar] [CrossRef] [PubMed]
  195. Del Bo’, C.; Bernardi, S.; Cherubini, A.; Porrini, M.; Gargari, G.; Hidalgo-Liberona, N.; González-Domínguez, R.; Zamora-Ros, R.; Peron, G.; Marino, M.; et al. A polyphenol-rich dietary pattern improves intestinal permeability, evaluated as serum zonulin levels, in older subjects: The MaPLE randomised controlled trial. Clin. Nutr. 2021, 40, 3006–3018. [Google Scholar] [CrossRef]
  196. Martín-Peláez, S.; Castañer, O.; Solà, R.; Motilva, M.J.; Castell, M.; Pérez-Cano, F.J.; Fitó, M. Influence of phenol-enriched olive oils on human intestinal immune function. Nutrients 2016, 8, 213. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  197. Martín-Peláez, S.; Mosele, J.I.; Pizarro, N.; Farràs, M.; de la Torre, R.; Subirana, I.; Pérez-Cano, F.J.; Castañer, O.; Solà, R.; Fernandez-Castillejo, S.; et al. Effect of virgin olive oil and thyme phenolic compounds on blood lipid profile: Implications of human gut microbiota. Eur. J. Nutr. 2017, 56, 119–131. [Google Scholar] [CrossRef] [PubMed]
  198. Buranapin, S.; Siangruangsang, S.; Chantapanich, V.; Hengjeerajarus, N. The comparative study of diabetic specific formula and standard formula on postprandial plasma glucose control in type 2 DM patients. J. Med. Assoc. Thail./Chotmaihet Thangphaet 2014, 97, 582–588. [Google Scholar]
  199. Ma, W.; Wu, J.H.Y.; Wang, Q.; Lemaitre, R.N.; Mukamal, K.J.; Djoussé, L.; King, I.B.; Song, X.; Biggs, M.L.; Delaney, J.A.; et al. Prospective association of fatty acids in the de novo lipogenesis pathway with risk of type 2 diabetes: The cardiovascular health study. Am. J. Clin. Nutr. 2015, 101, 153–163. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  200. Nourmohammadi, M.; Moghadam, O.M.; Lahiji, M.N.; Hatamian, S.; Shariatpanahi, Z.V. Effect of fat-based versus carbohydrate-based enteral feeding on glycemic control in critically ill patients: A randomized clinical trial. Indian J. Crit. Care Med. 2017, 21, 500–505. [Google Scholar] [CrossRef] [PubMed]
  201. Sanz-París, A.; Matía-Martín, P.; Martín-Palmero, Á.; Gómez-Candela, C.; Camprubi Robles, M. Diabetes-specific formulas high in monounsaturated fatty acids and metabolic outcomes in patients with diabetes or hyperglycaemia. a systematic review and meta-analysis. Clin. Nutr. 2020, 39, 3273–3282. [Google Scholar] [CrossRef]
  202. Ojo, O.; Weldon, S.M.; Thompson, T.; Crockett, R.; Wang, X.-H. The effect of diabetes-specific enteral nutrition formula on cardiometabolic parameters in patients with type 2 diabetes: A systematic review and meta–analysis of randomised controlled trials. Nutrients 2019, 11, 1905. [Google Scholar] [CrossRef] [Green Version]
  203. Gulati, S.; Misra, A.; Nanda, K.; Pandey, R.M.; Garg, V.; Ganguly, S.; Cheung, L. Efficacy and tolerance of a diabetes specific formula in patients with type 2 diabetes mellitus: An open label, randomized, crossover study. Diabetes Metab. Syndr. Clin. Res. Rev. 2015, 9, 252–257. [Google Scholar] [CrossRef]
  204. León-Sanz, M.; García-Luna, P.P.; Sanz-París, A.; Gómez-Candela, C.; Casimiro, C.; Chamorro, J.; Pereira-Cunill, J.L.; Martin-Palmero, A.; Trallero, R.; Martínez, J.; et al. Glycemic and lipid control in hospitalized type 2 diabetic patients: Evaluation of 2 enteral nutrition formulas (low carbohydrate-high monounsaturated fat vs high carbohydrate). JPEN J. Parenter. Enter. Nutr. 2005, 29, 21–29. [Google Scholar] [CrossRef]
  205. Mandoe, M.J.; Hansen, K.B.; Hartmann, B.; Rehfeld, J.F.; Holst, J.J.; Hansen, H.S. The 2-monoacylglycerol moiety of dietary fat appears to be responsible for the fat-induced release of GLP-1 in humans. Am. J. Clin. Nutr. 2015, 102, 548–555. [Google Scholar] [CrossRef] [PubMed]
  206. Jumpertz, R.; Guijarro, A.; Pratley, R.E.; Mason, C.C.; Piomelli, D.; Krakoff, J. Associations of fatty acids in cerebrospinal fluid with peripheral glucose concentrations and energy metabolism. PLoS ONE 2012, 7, e41503. [Google Scholar] [CrossRef] [PubMed]
  207. Goree, L.L.; Chandler-Laney, P.; Ellis, A.C.; Casazza, K.; Granger, W.M.; Gower, B.A. Dietary macronutrient composition affects beta cell responsiveness but not insulin sensitivity. Am. J. Clin. Nutr. 2011, 94, 120–127. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  208. Eckert, I.; Kumbier, M.C.C.; Silva, F.M.; Franzosi, O.S.; de Almeida, J.C. Association of specialized enteral nutrition with glycemic control and clinical outcomes in critically ill patients: A meta-analysis of randomized controlled trials. Clin. Nutr. 2021, 40, 3940–3949. [Google Scholar] [CrossRef] [PubMed]
  209. Lansink, M.; Hofman, Z.; Genovese, S.; Rouws, C.H.F.C.; Ceriello, A. Improved glucose profile in patients with type 2 diabetes with a new, high-protein, diabetes-specific tube feed during 4 hours of continuous feeding. JPEN J. Parenter. Enter. Nutr. 2017, 41, 968–975. [Google Scholar] [CrossRef]
  210. Alish, C.J.; Garvey, W.T.; Maki, K.C.; Sacks, G.S.; Hustead, D.S.; Hegazi, R.A.; Mustad, V.A. A Diabetes-specific enteral formula improves glycemic variability in patients with type 2 diabetes. Diabetes Technol. Ther. 2010, 12, 419–425. [Google Scholar] [CrossRef] [PubMed]
  211. Pohl, M.; Mayr, P.; Mertl-Roetzer, M.; Lauster, F.; Haslbeck, M.; Hipper, B.; Steube, D.; Tietjen, M.; Eriksen, J.; Rahlfs, V.W. Glycemic control in patients with type 2 diabetes mellitus with a disease-specific enteral formula: Stage II of a randomized, controlled multicenter trial. JPEN J. Parenter. Enter. Nutr. 2009, 33, 37–49. [Google Scholar] [CrossRef]
  212. Vaisman, N.; Lansink, M.; Rouws, C.H.; van Laere, K.M.; Segal, R.; Niv, E.; Bowling, T.E.; Waitzberg, D.L.; Morley, J.E. Tube Feeding with a diabetes-specific feed for 12 weeks improves glycaemic control in type 2 diabetes patients. Clin. Nutr. 2009, 28, 549–555. [Google Scholar] [CrossRef]
  213. Vanschoonbeek, K.; Lansink, M.; van Laere, K.M.J.; Senden, J.M.G.; Verdijk, L.B.; van Loon, L.J.C. Slowly digestible carbohydrate sources can be used to attenuate the postprandial glycemic response to the ingestion of diabetes-specific enteral formulas. Diabetes Educ. 2009, 35, 631–640. [Google Scholar] [CrossRef] [PubMed]
  214. Magnoni, D.; Rouws, C.H.F.C.; Lansink, M.; van Laere, K.M.J.; Campos, A.C. Long-term use of a diabetes-specific oral nutritional supplement results in a low-postprandial glucose response in diabetes patients. Diabetes Res. Clin. Pract. 2008, 80, 75–82. [Google Scholar] [CrossRef] [PubMed]
  215. Voss, A.C.; Maki, K.C.; Garvey, W.T.; Hustead, D.S.; Alish, C.; Fix, B.; Mustad, V.A. Effect of two carbohydrate-modified tube-feeding formulas on metabolic responses in patients with type 2 diabetes. Nutrition 2008, 24, 990–997. [Google Scholar] [CrossRef] [PubMed]
  216. Yokoyama, J.; Someya, Y.; Yoshihara, R.; Ishii, H. Effects of high-monounsaturated fatty acid enteral formula versus high-carbohydrate enteral formula on plasma glucose concentration and insulin secretion in healthy individuals and diabetic patients. J. Int. Med. Res. 2008, 36, 137–146. [Google Scholar] [CrossRef] [Green Version]
  217. Hofman, Z.; Lansink, M.; Rouws, C.; van Drunen, J.D.E.; Kuipers, H. Diabetes specific tube feed results in improved glycaemic and triglyceridaemic control during 6h continuous feeding in diabetes patients. e-SPEN Eur. e-J. Clin. Nutr. Metab. 2007, 2, 44–50. [Google Scholar] [CrossRef] [Green Version]
  218. Pohl, M.; Mayr, P.; Mertl-Roetzer, M.; Lauster, F.; Lerch, M.; Eriksen, J.; Haslbeck, M.; Rahlfs, V.W. Glycaemic control in type ii diabetic tube-fed patients with a new enteral formula low in carbohydrates and high in monounsaturated fatty acids: A randomised controlled trial. Eur. J. Clin. Nutr. 2005, 59, 1221–1232. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  219. Doola, R.; Deane, A.M.; Tolcher, D.M.; Presneill, J.J.; Barrett, H.L.; Forbes, J.M.; Todd, A.S.; Okano, S.; Sturgess, D.J. The effect of a low carbohydrate formula on glycaemia in critically ill enterally-fed adult patients with hyperglycaemia: A blinded randomised feasibility trial. Clin. Nutr. ESPEN 2019, 31, 80–87. [Google Scholar] [CrossRef] [PubMed]
  220. Vahabzadeh, D.; Valizadeh Hasanloei, M.A.; Vahdat Shariatpanahi, Z. Effect of high-fat, low-carbohydrate enteral formula versus standard enteral formula in hyperglycemic critically ill patients: A randomized clinical trial. Int. J. Diabetes Dev. Ctries. 2019, 39, 173–180. [Google Scholar] [CrossRef]
  221. Van Steen, S.C.; Rijkenberg, S.; Sechterberger, M.K.; DeVries, J.H.; van der Voort, P.H.J. Glycemic effects of a low-carbohydrate enteral formula compared with an enteral formula of standard composition in critically ill patients: An open-label randomized controlled clinical trial. JPEN J. Parenter. Enter. Nutr. 2018, 42, 1035–1045. [Google Scholar] [CrossRef] [PubMed]
  222. Wewalka, M.; Drolz, A.; Seeland, B.; Schneeweiss, M.; Schmid, M.; Schneeweiss, B.; Zauner, C. Different enteral nutrition formulas have no effect on glucose homeostasis but on diet-induced thermogenesis in critically ill medical patients: A randomized controlled trial. Eur. J. Clin. Nutr. 2018, 72, 496–503. [Google Scholar] [CrossRef]
  223. Mesejo, A.; Montejo-González, J.C.; Vaquerizo-Alonso, C.; Lobo-Tamer, G.; Zabarte-Martinez, M.; Herrero-Meseguer, J.I.; Acosta-Escribano, J.; Blesa-Malpica, A.; Martinez-Lozano, F. Diabetes-specific enteral nutrition formula in hyperglycemic, mechanically ventilated, critically ill patients: A prospective, open-label, blind-randomized, multicenter study. Crit. Care 2015, 19, 390. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  224. Egi, M.; Toda, Y.; Katayama, H.; Yokoyama, M.; Morita, K.; Arai, H.; Yamatsuji, T.; Bailey, M.; Naomoto, Y. Safer glycemic control using isomaltulose-based enteral formula: A pilot randomized crossover trial. J. Crit. Care 2010, 25, 90–96. [Google Scholar] [CrossRef] [PubMed]
  225. Mesejo, A.; Acosta, J.A.; Ortega, C.; Vila, J.; Fernández, M.; Ferreres, J.; Sanchis, J.C.; López, F. Comparison of a high-protein disease-specific enteral formula with a high-protein enteral formula in hyperglycemic critically ill patients. Clin. Nutr. 2003, 22, 295–305. [Google Scholar] [CrossRef]
  226. Ceriello, A.; Lansink, M.; Rouws, C.H.F.C.; van Laere, K.M.J.; Frost, G.S. Administration of a new diabetes-specific enteral formula results in an improved 24h glucose profile in type 2 diabetic patients. Diabetes Res. Clin. Pract. 2009, 84, 259–266. [Google Scholar] [CrossRef]
  227. Imamura, F.; Micha, R.; Wu, J.H.Y.; de Oliviera Otto, M.C.; Otite, F.O.; Abioye, A.I.; Mozaffarian, D. Effects of saturated fat, polyunsaturated fat, monounsaturated fat, and carbohydrate on glucose-insulin homeostasis: A systematic review and meta-analysis of randomised controlled feeding trials. PLoS Med. 2016, 13, e1002087. [Google Scholar] [CrossRef] [Green Version]
  228. Manning, P.J.; de Jong, S.A.; Ryalls, A.R.; Sutherland, W.H.F. Paraoxonase 1 activity in chylomicrons and VLDL: The effect of type 2 diabetes and meals rich in saturated fat and oleic acid. Lipids 2012, 47, 259–267. [Google Scholar] [CrossRef] [PubMed]
  229. Kien, C.L.; Bunn, J.Y.; Stevens, R.; Bain, J.; Ikayeva, O.; Crain, K.; Koves, T.R.; Muoio, D.M. Dietary intake of palmitate and oleate has broad impact on systemic and tissue lipid profiles in humans. Am. J. Clin. Nutr. 2014, 99, 436–445. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  230. Jakobsen, M.U.; O’Reilly, E.J.; Heitmann, B.L.; Pereira, M.A.; Bälter, K.; Fraser, G.E.; Goldbourt, U.; Hallmans, G.; Knekt, P.; Liu, S.; et al. Major types of dietary fat and risk of coronary heart disease: A pooled analysis of 11 cohort studies. Am. J. Clin. Nutr. 2009, 89, 1425–1432. [Google Scholar] [CrossRef] [PubMed]
  231. Miura, K.; Stamler, J.; Brown, I.J.; Ueshima, H.; Nakagawa, H.; Sakurai, M.; Chan, Q.; Appel, L.J.; Okayama, A.; Okuda, N.; et al. Relationship of dietary monounsaturated fatty acids to blood pressure: The international study of macro/micronutrients and blood pressure. J. Hypertens. 2013, 31, 1144–1150. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  232. Voon, P.T.; Ng, T.K.W.; Lee, V.K.M.; Nesaretnam, K. Diets high in palmitic acid (16:0), lauric and myristic acids (12:0+14:0), or oleic acid (18:1) do not alter postprandial or fasting plasma homocysteine and inflammatory markers in healthy malaysian adults. Am. J. Clin. Nutr. 2011, 94, 1451–1457. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  233. Gebauer, S.K.; Tracy, R.P.; Baer, D.J. Impact of stearic acid and oleic acid on hemostatic factors in the context of controlled diets consumed by healthy men. Eur. J. Clin. Nutr. 2014, 68, 1072–1074. [Google Scholar] [CrossRef] [Green Version]
  234. Belury, M.A.; Cole, R.M.; Bailey, B.E.; Ke, J.-Y.; Andridge, R.R.; Kiecolt-Glaser, J.K. Erythrocyte linoleic acid, but not oleic acid, is associated with improvements in body composition in men and women. Mol. Nutr. Food Res. 2016, 60, 1206–1212. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  235. Scoditti, E.; Massaro, M.; Carluccio, M.A.; Pellegrino, M.; Wabitsch, M.; Calabriso, N.; Storelli, C.; De Caterina, R. Additive regulation of adiponectin expression by the Mediterranean diet olive oil components oleic acid and hydroxytyrosol in human adipocytes. PLoS ONE 2015, 10, e0128218. [Google Scholar] [CrossRef]
  236. Gassull, M.A.; Fernández-Bañares, F.; Cabré, E.; Papo, M.; Giaffer, M.H.; Sánchez-Lombraña, J.L.; Richart, C.; Malchow, H.; González-Huix, F.; Esteve, M.; et al. Fat composition may be a clue to explain the primary therapeutic effect of enteral nutrition in Crohn’s disease: Results of a double blind randomised multicentre European trial. Gut 2002, 51, 164–168. [Google Scholar] [CrossRef]
  237. Ajabnoor, S.M.; Forbes, A. Effect of fat composition in enteral nutrition for Crohn’s disease in adults: A systematic review. Clin. Nutr. 2019, 38, 90–99. [Google Scholar] [CrossRef] [PubMed]
  238. Wiese, D.M.; Horst, S.N.; Brown, C.T.; Allaman, M.M.; Hodges, M.E.; Slaughter, J.C.; Druce, J.P.; Beaulieu, D.B.; Schwartz, D.A.; Wilson, K.T.; et al. Serum fatty acids are correlated with inflammatory cytokines in ulcerative colitis. PLoS ONE 2016, 11, e0156387. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  239. Scoville, E.A.; Allaman, M.M.; Adams, D.W.; Motley, A.K.; Peyton, S.C.; Ferguson, S.L.; Horst, S.N.; Williams, C.S.; Beaulieu, D.B.; Schwartz, D.A.; et al. Serum polyunsaturated fatty acids correlate with serum cytokines and clinical disease activity in Crohn’s disease. Sci. Rep. 2019, 9, 2882. [Google Scholar] [CrossRef] [PubMed]
  240. Faxen-Irving, G.; Cederholm, T. Energy dense oleic acid rich formula to newly admitted geriatric patients—Feasibility and effects on energy intake. Clin. Nutr. 2011, 30, 202–208. [Google Scholar] [CrossRef]
  241. De Luis, D.A.; Izaola, O.; de la Fuente, B.; Terroba, M.C.; Cuellar, L.; Cabezas, G. A Randomized clinical trial with two doses of an enteral diabetes-specific suplements in elderly patients with diabetes mellitus type 2. Eur. Rev. Med. Pharm. Sci. 2013, 17, 1626–1630. [Google Scholar]
  242. Kien, C.L.; Everingham, K.I.; Stevens, R.D.; Fukagawa, N.K.; Muoio, D.M. Short-term effects of dietary fatty acids on muscle lipid composition and serum acylcarnitine profile in human subjects. Obesity 2011, 19, 305–311. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  243. Kien, C.L.; Bunn, J.Y.; Poynter, M.E.; Stevens, R.; Bain, J.; Ikayeva, O.; Fukagawa, N.K.; Champagne, C.M.; Crain, K.I.; Koves, T.R.; et al. A lipidomics analysis of the relationship between dietary fatty acid composition and insulin sensitivity in young adults. Diabetes 2013, 62, 1054–1063. [Google Scholar] [CrossRef] [Green Version]
  244. Dumas, J.A.; Bunn, J.Y.; Nickerson, J.; Crain, K.I.; Ebenstein, D.B.; Tarleton, E.K.; Makarewicz, J.; Poynter, M.E.; Kien, C.L. Dietary saturated fat and monounsaturated fat have reversible effects on brain function and the secretion of pro-inflammatory cytokines in young women. Metabolism 2016, 65, 1582–1588. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  245. Gomez-Delgado, F.; Delgado-Lista, J.; Lopez-Moreno, J.; Rangel-Zuñiga, O.A.; Alcala-Diaz, J.F.; Leon-Acuña, A.; Corina, A.; Yubero-Serrano, E.; Torres-Peña, J.D.; Camargo, A.; et al. Telomerase RNA Component genetic variants interact with the mediterranean diet modifying the inflammatory status and its relationship with aging: CORDIOPREV study. J. Gerontol. Ser. A 2018, 73, 327–332. [Google Scholar] [CrossRef] [Green Version]
  246. Biesboer, A.N.; Stoehr, N.A. A product review of alternative oil-based intravenous fat emulsions. Nutr. Clin. Pract. 2016, 31, 610–618. [Google Scholar] [CrossRef]
  247. Xu, Z.; Harvey, K.A.; Pavlina, T.M.; Zaloga, G.P.; Siddiqui, R.A. Distribution of tocopherols and tocotrienols in guinea pig tissues following parenteral lipid emulsion infusion. JPEN J. Parenter. Enter. Nutr. 2016, 40, 672–681. [Google Scholar] [CrossRef]
  248. Reimund, J.-M.; Rahmi, G.; Escalin, G.; Pinna, G.; Finck, G.; Muller, C.D.; Duclos, B.; Baumann, R. Efficacy and safety of an olive oil-based intravenous fat emulsion in adult patients on home parenteral nutrition. Aliment. Pharm. Ther. 2005, 21, 445–454. [Google Scholar] [CrossRef] [PubMed]
  249. Eritsland, J. Safety considerations of polyunsaturated Fatty Acids. Am. J. Clin. Nutr. 2000, 71, 197S–201S. [Google Scholar] [CrossRef]
  250. Pontes-Arruda, A. Biological benefits of an oleic acid–rich lipid emulsion for parenteral nutrition. Clin. Nutr. Suppl. 2009, 4, 19–23. [Google Scholar] [CrossRef]
  251. Abdullah, M.M.H.; Jew, S.; Jones, P.J.H. Health benefits and evaluation of healthcare cost savings if oils rich in monounsaturated fatty acids were substituted for conventional dietary oils in the united states. Nutr. Rev. 2017, 75, 163–174. [Google Scholar] [CrossRef] [PubMed]
  252. Carrillo Pérez, C.; Cavia Camarero, M.D.M.; Alonso de la Torre, S. Role of oleic acid in immune system; mechanism of action: A review. Nutr. Hosp. 2012, 27, 978–990. [Google Scholar] [CrossRef] [Green Version]
  253. Sadu Singh, B.K.; Narayanan, S.S.; Khor, B.H.; Sahathevan, S.; Abdul Gafor, A.H.; Fiaccadori, E.; Sundram, K.; Karupaiah, T. Composition and functionality of lipid emulsions in parenteral nutrition: Examining evidence in clinical applications. Front. Pharm. 2020, 11, 506. [Google Scholar] [CrossRef] [PubMed]
  254. Raman, M.; Almutairdi, A.; Mulesa, L.; Alberda, C.; Beattie, C.; Gramlich, L. Parenteral nutrition and lipids. Nutrients 2017, 9, 388. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  255. Yaqoob, P. Monounsaturated fatty acids in parenteral nutrition; evaluation of risks and benefits. Br. J. Nutr. 2005, 94, 867–868. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  256. Buschmann, K.; Poeschl, J.; Braach, N.; Hudalla, H.; Kuss, N.; Frommhold, D. The olive oil-based lipid clinoleic blocks leukocyte recruitment and improves survival during systemic inflammation: A comparative in vivo study of different parenteral lipid emulsions. Mediat. Inflamm. 2015, 2015, 757059. [Google Scholar] [CrossRef] [Green Version]
  257. Garnacho-Montero, J.; Ortiz-Leyba, C.; Garnacho-Montero, M.C.; Garcia-Garmendia, J.L.; Pérez-Paredes, C.; Moyano-Del Estad, M.R.; Barrero-Almodóvar, A.; Jiménez-Jiménez, F.J. Effects of three intravenous lipid emulsions on the survival and mononuclear phagocyte function of septic rats. Nutrition 2002, 18, 751–754. [Google Scholar] [CrossRef]
  258. Candiloro, F.; Borioli, V.; Borsellino, G.; Picozza, M.; Pellini, R.; Cereda, E.; Gargano, F.; Caraccia, M.; Nardi, M.T.; Bellu, L.; et al. Influence of different lipid emulsions on specific immune cell functions in head and neck cancer patients receiving supplemental parenteral nutrition: An exploratory analysis. Nutrition 2021, 86, 111178. [Google Scholar] [CrossRef]
  259. Jia, Z.-Y.; Yang, J.; Xia, Y.; Tong, D.-N.; Zaloga, G.P.; Qin, H.-L. Oliclinomel N4 study group safety and efficacy of an olive oil-based triple-chamber bag for parenteral nutrition: A prospective, randomized, multi-center clinical trial in China. Nutr. J. 2015, 14, 119. [Google Scholar] [CrossRef] [Green Version]
  260. Siqueira, J.; Smiley, D.; Newton, C.; Le, N.-A.; Gosmanov, A.R.; Spiegelman, R.; Peng, L.; Osteen, S.J.; Jones, D.P.; Quyyumi, A.A.; et al. Substitution of standard soybean oil with olive oil-based lipid emulsion in parenteral nutrition: Comparison of vascular, metabolic, and inflammatory effects. J. Clin. Endocrinol. Metab. 2011, 96, 3207–3216. [Google Scholar] [CrossRef]
  261. Goulet, O.; de Potter, S.; Antébi, H.; Driss, F.; Colomb, V.; Béréziat, G.; Alcindor, L.G.; Corriol, O.; Le Brun, A.; Dutot, G.; et al. Long-term efficacy and safety of a new olive oil-based intravenous fat emulsion in pediatric patients: A double-blind randomized study. Am. J. Clin. Nutr. 1999, 70, 338–345. [Google Scholar] [CrossRef]
  262. Huschak, G.; Zur Nieden, K.; Hoell, T.; Riemann, D.; Mast, H.; Stuttmann, R. Olive oil based nutrition in multiple trauma patients: A pilot study. Intensive Care Med. 2005, 31, 1202–1208. [Google Scholar] [CrossRef] [PubMed]
  263. Manzanares, W.; Dhaliwal, R.; Jurewitsch, B.; Stapleton, R.D.; Jeejeebhoy, K.N.; Heyland, D.K. Alternative lipid emulsions in the critically ill: A systematic review of the evidence. Intensive Care Med. 2013, 39, 1683–1694. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  264. Gramlich, L.; Atkins, M. Alberta Health Services Cutrition Practice Guidelines-Intravenous Lipid Emulsions. 2017; unpublished work. [Google Scholar]
  265. De Bock, M.; Derraik, J.G.B.; Brennan, C.M.; Biggs, J.B.; Morgan, P.E.; Hodgkinson, S.C.; Hofman, P.L.; Cutfield, W.S. Olive (Olea europaea L.) leaf polyphenols improve insulin sensitivity in middle-aged overweight men: A randomized, placebo-controlled, crossover trial. PLoS ONE 2013, 8, e57622. [Google Scholar] [CrossRef] [PubMed]
  266. Komaki, E.; Yamaguchi, S.; Maru, I.; Kinoshita, M.; Kakehi, K.; Ohta, Y.; Tsukada, Y. Identification of anti-α-amylase components from olive leaf extracts. Food Sci. Technol. Res. 2003, 9, 35–39. [Google Scholar] [CrossRef] [Green Version]
  267. Carnevale, R.; Silvestri, R.; Loffredo, L.; Novo, M.; Cammisotto, V.; Castellani, V.; Bartimoccia, S.; Nocella, C.; Violi, F. Oleuropein, a component of extra virgin olive oil, lowers postprandial glycaemia in healthy subjects. Br. J. Clin. Pharm. 2018, 84, 1566–1574. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  268. Wainstein, J.; Ganz, T.; Boaz, M.; Bar Dayan, Y.; Dolev, E.; Kerem, Z.; Madar, Z. Olive leaf extract as a hypoglycemic agent in both human diabetic subjects and in rats. J. Med. Food 2012, 15, 605–610. [Google Scholar] [CrossRef] [PubMed]
  269. De Mello, V.D.F.; Lindström, J.; Eriksson, J.G.; Ilanne-Parikka, P.; Keinänen-Kiukaanniemi, S.; Pihlajamäki, J.; Tuomilehto, J.; Uusitupa, M. Markers of cholesterol metabolism as biomarkers in predicting diabetes in the finnish diabetes prevention study. Nutr. Metab. Cardiovasc. Dis. 2015, 25, 635–642. [Google Scholar] [CrossRef]
  270. Hadrich, F.; Bouallagui, Z.; Junkyu, H.; Isoda, H.; Sayadi, S. The α-glucosidase and α-amylase enzyme inhibitory of hydroxytyrosol and oleuropein. J. Oleo Sci. 2015, 64, 835–843. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  271. Tundis, R.; Conidi, C.; Loizzo, M.R.; Sicari, V.; Romeo, R.; Cassano, A. Concentration of bioactive phenolic compounds in olive mill wastewater by direct contact membrane distillation. Molecules 2021, 26, 1808. [Google Scholar] [CrossRef] [PubMed]
  272. Sultana, N.; Ata, A. Oleanolic acid and related derivatives as medicinally important compounds. J. Enzym. Inhib. Med. Chem. 2008, 23, 739–756. [Google Scholar] [CrossRef] [PubMed]
  273. Salehi-Sahlabadi, A.; Varkaneh, H.K.; Shahdadian, F.; Ghaedi, E.; Nouri, M.; Singh, A.; Farhadnejad, H.; Găman, M.-A.; Hekmatdoost, A.; Mirmiran, P. Effects of phytosterols supplementation on blood glucose, glycosylated hemoglobin (HbA1c) and insulin levels in humans: A systematic review and meta-analysis of randomized controlled trials. J. Diabetes Metab. Disord. 2020, 19, 625–632. [Google Scholar] [CrossRef] [PubMed]
  274. Pandey, J.; Dev, K.; Chattopadhyay, S.; Kadan, S.; Sharma, T.; Maurya, R.; Sanyal, S.; Siddiqi, M.I.; Zaid, H.; Tamrakar, A.K. β-Sitosterol-D-Glucopyranoside mimics estrogenic properties and stimulates glucose utilization in skeletal muscle cells. Molecules 2021, 26, 3129. [Google Scholar] [CrossRef]
  275. Prasad, M.; Jayaraman, S.; Eladl, M.A.; El-Sherbiny, M.; Abdelrahman, M.A.E.; Veeraraghavan, V.P.; Vengadassalapathy, S.; Umapathy, V.R.; Jaffer Hussain, S.F.; Krishnamoorthy, K.; et al. A Comprehensive review on therapeutic perspectives of phytosterols in insulin resistance: A mechanistic approach. Molecules 2022, 27, 1595. [Google Scholar] [CrossRef] [PubMed]
  276. Lockyer, S.; Rowland, I.; Spencer, J.P.E.; Yaqoob, P.; Stonehouse, W. Impact of phenolic-rich olive leaf extract on blood pressure, plasma lipids and inflammatory markers: A randomised controlled trial. Eur. J. Nutr. 2017, 56, 1421–1432. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  277. Lockyer, S.; Corona, G.; Yaqoob, P.; Spencer, J.P.E.; Rowland, I. Secoiridoids delivered as olive leaf extract induce acute improvements in human vascular function and reduction of an inflammatory cytokine: A randomised, double-blind, placebo-controlled, cross-over trial. Br. J. Nutr. 2015, 114, 75–83. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  278. Njike, V.Y.; Ayettey, R.; Treu, J.A.; Doughty, K.N.; Katz, D.L. Post-prandial effects of high-polyphenolic extra virgin olive oil on endothelial function in adults at risk for type 2 diabetes: A randomized controlled crossover trial. Int. J. Cardiol. 2021, 330, 171–176. [Google Scholar] [CrossRef] [PubMed]
  279. Detopoulou, M.; Ntzouvani, A.; Petsini, F.; Gavriil, L.; Fragopoulou, E.; Antonopoulou, S. Consumption of enriched yogurt with paf inhibitors from olive pomace affects the major enzymes of PAF metabolism: A randomized, double blind, three arm trial. Biomolecules 2021, 11, 801. [Google Scholar] [CrossRef] [PubMed]
  280. Colica, C.; Di Renzo, L.; Trombetta, D.; Smeriglio, A.; Bernardini, S.; Cioccoloni, G.; Costa de Miranda, R.; Gualtieri, P.; Sinibaldi Salimei, P.; De Lorenzo, A. Antioxidant effects of a hydroxytyrosol-based pharmaceutical formulation on body composition, metabolic state, and gene expression: A randomized double-blinded, placebo-controlled crossover trial. Oxid. Med. Cell Longev. 2017, 2017, 2473495. [Google Scholar] [CrossRef]
  281. Crespo, M.C.; Tomé-Carneiro, J.; Burgos-Ramos, E.; Loria Kohen, V.; Espinosa, M.I.; Herranz, J.; Visioli, F. One-week administration of hydroxytyrosol to humans does not activate phase II enzymes. Pharm. Res. 2015, 95–96, 132–137. [Google Scholar] [CrossRef] [PubMed]
  282. Catalán, Ú.; López de Las Hazas, M.-C.; Rubió, L.; Fernández-Castillejo, S.; Pedret, A.; de la Torre, R.; Motilva, M.-J.; Solà, R. Protective effect of hydroxytyrosol and its predominant plasmatic human metabolites against endothelial dysfunction in human aortic endothelial cells. Mol. Nutr. Food Res. 2015, 59, 2523–2536. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  283. Castellino, G.; Nikolic, D.; Magán-Fernández, A.; Malfa, G.A.; Chianetta, R.; Patti, A.M.; Amato, A.; Montalto, G.; Toth, P.P.; Banach, M.; et al. Altilix® supplement containing chlorogenic acid and luteolin improved hepatic and cardiometabolic parameters in subjects with metabolic syndrome: A 6 month randomized, double-blind, placebo-controlled study. Nutrients 2019, 11, 2580. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  284. Li, C.; Ding, Y.; Si, Q.; Li, K.; Xu, K. Multiple functions of policosanol in elderly patients with dyslipidemia. J. Int. Med. Res. 2020, 48, 300060520936082. [Google Scholar] [CrossRef]
  285. Cho, K.-H.; Kim, S.-J.; Yadav, D.; Kim, J.-Y.; Kim, J.-R. Consumption of Cuban policosanol improves blood pressure and lipid profile via enhancement of HDL functionality in healthy women subjects: Randomized, double-blinded, and placebo-controlled study. Oxid. Med. Cell Longev. 2018, 2018, 4809525. [Google Scholar] [CrossRef] [Green Version]
  286. Cho, K.-H.; Bae, M.-A.; Kim, J.-R. Cuban sugar cane wax acid and policosanol showed similar atheroprotective effects with inhibition of LDL oxidation and cholesteryl ester transfer via enhancement of high-density lipoproteins functionality. Cardiovasc. Ther. 2019, 2019, 8496409. [Google Scholar] [CrossRef] [PubMed]
  287. Stüsser, R.; Batista, J.; Padrón, R.; Sosa, F.; Pereztol, O. Long-term therapy with policosanol improves treadmill exercise-ECG Testing performance of coronary heart disease patients. Int. J. Clin. Pharm. Ther. 1998, 36, 469–473. [Google Scholar]
  288. Arruzazabala, M.L.; Molina, V.; Mas, R.; Fernandez, L.; Carbajal, D.; Valdes, S.; Castano, G. Antiplatelet Effects of policosanol (20 and 40 mg/day) in healthy volunteers and dyslipidaemic patients. Clin. Exp. Pharm. Physiol. 2002, 29, 891–897. [Google Scholar] [CrossRef] [PubMed]
  289. Taylor, J.C.; Rapport, L.; Lockwood, G.B. Octacosanol in human health. Nutrition 2003, 19, 192–195. [Google Scholar] [CrossRef]
  290. Chen, J.T.; Wesley, R.; Shamburek, R.D.; Pucino, F.; Csako, G. Meta-analysis of natural therapies for hyperlipidemia: Plant sterols and stanols versus policosanol. Pharmacotherapy 2005, 25, 171–183. [Google Scholar] [CrossRef] [PubMed]
  291. Lin, Y.; Rudrum, M.; van der Wielen, R.P.J.; Trautwein, E.A.; McNeill, G.; Sierksma, A.; Meijer, G.W. Wheat germ policosanol failed to lower plasma cholesterol in subjects with normal to mildly elevated cholesterol concentrations. Metabolism 2004, 53, 1309–1314. [Google Scholar] [CrossRef]
  292. Zhai, Z.; Liu, J.; Niu, K.-M.; Lin, C.; Tu, Y.; Liu, Y.; Cai, L.; Liu, H.; Ouyang, K. Integrated Metagenomics and Metabolomics to reveal the effects of policosanol on modulating the gut microbiota and lipid metabolism in hyperlipidemic C57BL/6 mice. Front. Endocrinol. 2021, 12, 722055. [Google Scholar] [CrossRef]
  293. Sharma, R.; Matsuzaka, T.; Kaushik, M.K.; Sugasawa, T.; Ohno, H.; Wang, Y.; Motomura, K.; Shimura, T.; Okajima, Y.; Mizunoe, Y.; et al. Octacosanol and policosanol prevent high-fat diet-induced obesity and metabolic disorders by activating brown adipose tissue and improving liver metabolism. Sci. Rep. 2019, 9, 5169. [Google Scholar] [CrossRef] [PubMed]
  294. Kaur, R.; Myrie, S.B. Association of dietary phytosterols with cardiovascular disease biomarkers in humans. Lipids 2020, 55, 569–584. [Google Scholar] [CrossRef] [PubMed]
  295. Ghaedi, E.; Kord-Varkaneh, H.; Mohammadi, H.; Askarpour, M.; Miraghajani, M. Phytosterol supplementation could improve atherogenic and anti-atherogenic apolipoproteins: A systematic review and dose-response meta-analysis of randomized controlled trials. J. Am. Coll. Nutr. 2020, 39, 82–92. [Google Scholar] [CrossRef]
  296. AbuMweis, S.S.; Vanstone, C.A.; Lichtenstein, A.H.; Jones, P.J.H. Plant sterol consumption frequency affects plasma lipid levels and cholesterol kinetics in humans. Eur. J. Clin. Nutr. 2009, 63, 747–755. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  297. Plat, J.; van Onselen, E.N.; van Heugten, M.M.; Mensink, R.P. Effects on serum lipids, lipoproteins and fat soluble antioxidant concentrations of consumption frequency of margarines and shortenings enriched with plant stanol esters. Eur. J. Clin. Nutr. 2000, 54, 671–677. [Google Scholar] [CrossRef] [Green Version]
  298. U.S. Food and Drug Administration. Food Labeling: Health Claims; Plant Sterol/Stanol Esters and Coronary Heart Disease; Interim Final Rule. Fed. Regist. 2000, 65, 54685–54739. [Google Scholar]
  299. Gibney, E.R.; Milenkovic, D.; Combet, E.; Ruskovska, T.; Greyling, A.; González-Sarrías, A.; de Roos, B.; Tomás-Barberán, F.; Morand, C.; Rodriguez-Mateos, A. Factors influencing the cardiometabolic response to (poly)phenols and phytosterols: A review of the COST action POSITIVe activities. Eur. J. Nutr. 2019, 58, 37–47. [Google Scholar] [CrossRef] [Green Version]
  300. Harcombe, Z.; Baker, J.S. Plant sterols lower cholesterol, but increase risk for coronary heart disease. Online J. Biol. Sci. 2014, 14, 167–169. [Google Scholar] [CrossRef] [Green Version]
  301. Scholz, M.; Horn, K.; Pott, J.; Gross, A.; Kleber, M.E.; Delgado, G.E.; Mishra, P.P.; Kirsten, H.; Gieger, C.; Müller-Nurasyid, M.; et al. Genome-wide meta-analysis of phytosterols reveals five novel loci and a detrimental effect on coronary atherosclerosis. Nat. Commun. 2022, 13, 143. [Google Scholar] [CrossRef] [PubMed]
  302. Helgadottir, A.; Thorleifsson, G.; Stefansson, K. Increased absorption of phytosterols is the simplest and most plausible explanation for coronary artery disease risk not accounted for by non-HDL cholesterol in high cholesterol absorbers. Eur. Heart J. 2020, 42, 283–284. [Google Scholar] [CrossRef] [PubMed]
  303. EFSA Panel on Nutrition, Novel Foods and Food Allergens (NDA); Turck, D.; Castenmiller, J.; De Henauw, S.; Hirsch-Ernst, K.I.; Kearney, J.; Maciuk, A.; Mangelsdorf, I.; McArdle, H.J.; Naska, A.; et al. Safety of the extension of use of plant sterol esters as a novel food pursuant to regulation (EU) 2015/2283. EFSA J. 2020, 18, e06135. [Google Scholar] [CrossRef]
  304. Hovenkamp, E.; Demonty, I.; Plat, J.; Lütjohann, D.; Mensink, R.P.; Trautwein, E.A. Biological effects of oxidized phytosterols: A review of the current knowledge. Prog. Lipid Res. 2008, 47, 37–49. [Google Scholar] [CrossRef] [PubMed]
  305. Warnke, I.; Goralczyk, R.; Fuhrer, E.; Schwager, J. Dietary constituents reduce lipid accumulation in murine c3h10 t1/2 adipocytes: A novel fluorescent method to quantify fat droplets. Nutr. Metab. 2011, 8, 30. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  306. Drira, R.; Chen, S.; Sakamoto, K. Oleuropein and hydroxytyrosol inhibit adipocyte differentiation in 3 T3-L1 cells. Life Sci. 2011, 89, 708–716. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  307. Hao, J.; Shen, W.; Yu, G.; Jia, H.; Li, X.; Feng, Z.; Wang, Y.; Weber, P.; Wertz, K.; Sharman, E.; et al. Hydroxytyrosol promotes mitochondrial biogenesis and mitochondrial function in 3T3-L1 adipocytes. J. Nutr. Biochem. 2010, 21, 634–644. [Google Scholar] [CrossRef] [PubMed]
  308. Vezza, T.; Rodríguez-Nogales, A.; Algieri, F.; Garrido-Mesa, J.; Romero, M.; Sánchez, M.; Toral, M.; Martín-García, B.; Gómez-Caravaca, A.M.; Arráez-Román, D.; et al. The metabolic and vascular protective effects of olive (Olea europaea L.) leaf extract in diet-induced obesity in mice are related to the amelioration of gut microbiota dysbiosis and to its immunomodulatory properties. Pharm. Res. 2019, 150, 104487. [Google Scholar] [CrossRef] [PubMed]
  309. Shin, S.; Ajuwon, K.M. Effects of diets differing in composition of 18-C fatty acids on adipose tissue thermogenic gene expression in mice fed high-fat diets. Nutrients 2018, 10, 256. [Google Scholar] [CrossRef] [Green Version]
  310. Ghaedi, E.; Varkaneh, H.K.; Rahmani, J.; Mousavi, S.M.; Mohammadi, H.; Fatahi, S.; Pantovic, A.; Darooghegi Mofrad, M.; Zhang, Y. Possible anti-obesity effects of phytosterols and phytostanols supplementation in humans: A systematic review and dose-response meta-analysis of randomized controlled trials. Phytother. Res. 2019, 33, 1246–1257. [Google Scholar] [CrossRef]
  311. Tauriainen, M.-M.; Männistö, V.; Kaminska, D.; Vaittinen, M.; Kärjä, V.; Käkelä, P.; Venesmaa, S.; Gylling, H.; Pihlajamäki, J. Serum, liver and bile sitosterol and sitostanol in obese patients with and without NAFLD. Biosci. Rep. 2018, 38, BSR20171274. [Google Scholar] [CrossRef] [Green Version]
  312. Sales, R.C.; Medeiros, P.C.; Spreafico, F.; de Velasco, P.C.; Gonçalves, F.K.A.; Martín-Hernández, R.; Mantilla-Escalante, D.C.; Gil-Zamorano, J.; Peres, W.A.F.; de Souza, S.A.L.; et al. Olive oil, palm oil, and hybrid palm oil distinctly modulate liver transcriptome and induce NAFLD in mice fed a high-fat diet. Int. J. Mol. Sci. 2018, 20, 10008. [Google Scholar] [CrossRef] [Green Version]
  313. Jurado-Ruiz, E.; Varela, L.M.; Luque, A.; Berná, G.; Cahuana, G.; Martinez-Force, E.; Gallego-Durán, R.; Soria, B.; de Roos, B.; Romero Gómez, M.; et al. An extra virgin olive oil rich diet intervention ameliorates the nonalcoholic steatohepatitis induced by a high-fat “Western-type” diet in mice. Mol. Nutr. Food Res. 2017, 61, 1600549. [Google Scholar] [CrossRef]
  314. Tapia, G.; Valenzuela, R.; Espinosa, A.; Romanque, P.; Dossi, C.; Gonzalez-Mañán, D.; Videla, L.A.; D’Espessailles, A. N-3 long-chain PUFA supplementation prevents high fat diet induced mouse liver steatosis and inflammation in relation to PPAR-α upregulation and NF-ΚB DNA binding abrogation. Mol. Nutr. Food Res. 2014, 58, 1333–1341. [Google Scholar] [CrossRef] [PubMed]
  315. Dossi, C.G.; Tapia, G.S.; Espinosa, A.; Videla, L.A.; D’Espessailles, A. Reversal of high-fat diet-induced hepatic steatosis by n-3 LCPUFA: Role of PPAR-α and SREBP-1c. J. Nutr. Biochem. 2014, 25, 977–984. [Google Scholar] [CrossRef] [PubMed]
  316. Soni, N.K.; Nookaew, I.; Sandberg, A.-S.; Gabrielsson, B.G. Eicosapentaenoic and docosahexaenoic acid-enriched high fat diet delays the development of fatty liver in mice. Lipids Health Dis. 2015, 14, 74. [Google Scholar] [CrossRef] [Green Version]
  317. Hernández-Rodas, M.C.; Valenzuela, R.; Echeverría, F.; Rincón-Cervera, M.Á.; Espinosa, A.; Illesca, P.; Muñoz, P.; Corbari, A.; Romero, N.; Gonzalez-Mañan, D.; et al. Supplementation with docosahexaenoic acid and extra virgin olive oil prevents liver steatosis induced by a high-fat diet in mice through PPAR-α and Nrf2 upregulation with concomitant SREBP-1c and NF-KB downregulation. Mol. Nutr. Food Res. 2017, 61, 1700479. [Google Scholar] [CrossRef]
  318. Echeverría, F.; Valenzuela, R.; Bustamante, A.; Álvarez, D.; Ortiz, M.; Soto-Alarcon, S.A.; Muñoz, P.; Corbari, A.; Videla, L.A. Attenuation of high-fat diet-induced rat liver oxidative stress and steatosis by combined hydroxytyrosol-(HT-) eicosapentaenoic acid supplementation mainly relies on HT. Oxid. Med. Cell Longev. 2018, 2018, 5109503. [Google Scholar] [CrossRef] [PubMed]
  319. Echeverría, F.; Valenzuela, R.; Espinosa, A.; Bustamante, A.; Álvarez, D.; Gonzalez-Mañan, D.; Ortiz, M.; Soto-Alarcon, S.A.; Videla, L.A. Reduction of high-fat diet-induced liver proinflammatory state by eicosapentaenoic acid plus hydroxytyrosol supplementation: Involvement of resolvins RvE1/2 and RvD1/2. J. Nutr. Biochem. 2019, 63, 35–43. [Google Scholar] [CrossRef] [PubMed]
  320. Echeverría, F.; Valenzuela, R.; Bustamante, A.; Álvarez, D.; Ortiz, M.; Espinosa, A.; Illesca, P.; Gonzalez-Mañan, D.; Videla, L.A. High-fat diet induces mouse liver steatosis with a concomitant decline in energy metabolism: Attenuation by eicosapentaenoic acid (EPA) or hydroxytyrosol (HT) supplementation and the additive effects upon EPA and HT co-administration. Food Funct. 2019, 10, 6170–6183. [Google Scholar] [CrossRef]
  321. Soto-Alarcón, S.A.; Ortiz, M.; Orellana, P.; Echeverría, F.; Bustamante, A.; Espinosa, A.; Illesca, P.; Gonzalez-Mañán, D.; Valenzuela, R.; Videla, L.A. Docosahexaenoic acid and hydroxytyrosol co-administration fully prevents liver steatosis and related parameters in mice subjected to high-fat diet: A molecular approach. Biofactors 2019, 45, 930–943. [Google Scholar] [CrossRef] [PubMed]
  322. Pirozzi, C.; Lama, A.; Simeoli, R.; Paciello, O.; Pagano, T.B.; Mollica, M.P.; Di Guida, F.; Russo, R.; Magliocca, S.; Canani, R.B.; et al. Hydroxytyrosol prevents metabolic impairment reducing hepatic inflammation and restoring duodenal integrity in a rat model of NAFLD. J. Nutr. Biochem. 2016, 30, 108–115. [Google Scholar] [CrossRef] [PubMed]
  323. Rajcic, D.; Brandt, A.; Jin, C.J.; Sánchez, V.; Engstler, A.J.; Jung, F.; Nier, A.; Baumann, A.; Bergheim, I. Exchanging dietary fat source with extra virgin olive oil does not prevent progression of diet-induced non-alcoholic fatty liver disease and insulin resistance. PLoS ONE 2020, 15, e0237946. [Google Scholar] [CrossRef] [PubMed]
  324. Sánchez-Calvo, B.; Cassina, A.; Mastrogiovanni, M.; Santos, M.; Trias, E.; Kelley, E.E.; Rubbo, H.; Trostchansky, A. Olive oil-derived nitro-fatty acids: Protection of mitochondrial function in non-alcoholic fatty liver disease. J. Nutr. Biochem. 2021, 94, 108646. [Google Scholar] [CrossRef] [PubMed]
  325. Lama, A.; Pirozzi, C.; Mollica, M.P.; Trinchese, G.; Di Guida, F.; Cavaliere, G.; Calignano, A.; Mattace Raso, G.; Berni Canani, R.; Meli, R. Polyphenol-rich virgin olive oil reduces insulin resistance and liver inflammation and improves mitochondrial dysfunction in high-fat diet fed rats. Mol. Nutr. Food Res. 2017, 61, 1600418. [Google Scholar] [CrossRef]
  326. Valenzuela, R.; Videla, L.A. The importance of the long-chain polyunsaturated fatty acid n-6/n-3 ratio in development of non-alcoholic fatty liver associated with obesity. Food Funct. 2011, 2, 644–648. [Google Scholar] [CrossRef]
  327. Valenzuela, R.; Videla, L.A. Impact of the co-administration of n-3 fatty acids and olive oil components in preclinical nonalcoholic fatty liver disease models: A mechanistic view. Nutrients 2020, 12, 499. [Google Scholar] [CrossRef] [Green Version]
  328. Boss, A.; Kao, C.H.-J.; Murray, P.M.; Marlow, G.; Barnett, M.P.G.; Ferguson, L.R. Human intervention study to assess the effects of supplementation with olive leaf extract on peripheral blood mononuclear cell gene expression. Int. J. Mol. Sci. 2016, 17, 22019. [Google Scholar] [CrossRef] [Green Version]
  329. Imran, M.; Nadeem, M.; Gilani, S.A.; Khan, S.; Sajid, M.W.; Amir, R.M. Antitumor perspectives of oleuropein and its metabolite hydroxytyrosol: Recent updates. J. Food Sci. 2018, 83, 1781–1791. [Google Scholar] [CrossRef] [Green Version]
  330. Storniolo, C.E.; Martinez-Hovelman, N.; Martinez-Huelamo, M.; Lamuela-Raventos, R.M.; Moreno, J.J. Extra virgin olive oil minor compounds modulate mitogenic action of oleic acid on colon cancer cell line. J. Agric. Food Chem. 2019, 67, 11420–11427. [Google Scholar] [CrossRef]
  331. Borzì, A.M.; Biondi, A.; Basile, F.; Luca, S.; Vicari, E.S.D.; Vacante, M. Olive oil effects on colorectal cancer. Nutrients 2018, 11, 32. [Google Scholar] [CrossRef] [Green Version]
  332. Ruzzolini, J.; Chioccioli, S.; Monaco, N.; Peppicelli, S.; Andreucci, E.; Urciuoli, S.; Romani, A.; Luceri, C.; Tortora, K.; Calorini, L.; et al. Oleuropein-rich leaf extract as a broad inhibitor of tumour and macrophage INOS in an Apc mutant rat model. Antioxidants 2021, 10, 1577. [Google Scholar] [CrossRef] [PubMed]
  333. Al-Karald, R.; Awadallah, A.; Tawfeek, H.M.; Nasr, M. Preparation, Characterization and cytotoxic activity of new oleuropein microemulsion against HCT-116 colon cancer cells. Pharm. Chem. J. 2020, 53, 1118–1121. [Google Scholar] [CrossRef]
  334. Ruzzolini, J.; Peppicelli, S.; Bianchini, F.; Andreucci, E.; Urciuoli, S.; Romani, A.; Tortora, K.; Caderni, G.; Nediani, C.; Calorini, L. Cancer glycolytic dependence as a new target of olive leaf extract. Cancers 2020, 12, 317. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  335. D’Ascanio, L.; Vitelli, F.; Cingolani, C.; Maranzano, M.; Brenner, M.J.; Di Stadio, A. Randomized clinical trial “olfactory dysfunction after COVID-19: Olfactory rehabilitation therapy vs. intervention treatment with palmitoylethanolamide and luteolin”: Preliminary results. Eur. Rev. Med. Pharm. Sci. 2021, 25, 4156–4162. [Google Scholar] [CrossRef]
  336. Hodgin, K.S.; Donovan, E.K.; Kekes-Szabo, S.; Lin, J.C.; Feick, J.; Massey, R.L.; Ness, T.J.; Younger, J.W. A placebo-controlled, pseudo-randomized, crossover trial of botanical agents for gulf war illness: Resveratrol (Polygonum cuspidatum), luteolin, and fisetin (Rhus succedanea). Int. J. Environ. Res. Public Health 2021, 18, 52483. [Google Scholar] [CrossRef]
  337. Butt, M.S.; Tariq, U.; Iahtisham-Ul-Haq; Naz, A.; Rizwan, M. Neuroprotective Effects of Oleuropein: Recent Developments and Contemporary Research. J. Food Biochem. 2021, 45, e13967. [Google Scholar] [CrossRef] [PubMed]
  338. Khalatbary, A.R. Natural polyphenols and spinal cord injury. Iran. Biomed. J. 2014, 18, 120–129. [Google Scholar] [CrossRef]
  339. Abuznait, A.H.; Qosa, H.; Busnena, B.A.; El Sayed, K.A.; Kaddoumi, A. Olive-oil-derived oleocanthal enhances β-amyloid clearance as a potential neuroprotective mechanism against Alzheimer’s disease: In vitro and in vivo studies. ACS Chem. Neurosci. 2013, 4, 973–982. [Google Scholar] [CrossRef] [Green Version]
  340. Al Rihani, S.B.; Darakjian, L.I.; Kaddoumi, A. Oleocanthal-rich extra-virgin olive oil restores the blood–brain barrier function through NLRP3 inflammasome inhibition simultaneously with autophagy induction in TgSwDI mice. ACS Chem. Neurosci. 2019, 10, 3543–3554. [Google Scholar] [CrossRef] [PubMed]
  341. Larussa, T.; Oliverio, M.; Suraci, E.; Greco, M.; Placida, R.; Gervasi, S.; Marasco, R.; Imeneo, M.; Paolino, D.; Tucci, L.; et al. Oleuropein decreases cyclooxygenase-2 and interleukin-17 expression and attenuates inflammatory damage in colonic samples from ulcerative colitis patients. Nutrients 2017, 9, 391. [Google Scholar] [CrossRef] [Green Version]
  342. Vezza, T.; Algieri, F.; Rodríguez-Nogales, A.; Garrido-Mesa, J.; Utrilla, M.P.; Talhaoui, N.; Gómez-Caravaca, A.M.; Segura-Carretero, A.; Rodríguez-Cabezas, M.E.; Monteleone, G.; et al. Immunomodulatory properties of Olea europaea leaf extract in intestinal inflammation. Mol. Nutr. Food Res. 2017, 61, 1601066. [Google Scholar] [CrossRef]
  343. Motawea, M.H.; Abd Elmaksoud, H.A.; Elharrif, M.G.; Desoky, A.A.E.; Ibrahimi, A. Evaluation of anti-inflammatory and antioxidant profile of oleuropein in experimentally induced ulcerative colitis. Int. J. Mol. Cell. Med. 2020, 9, 224–233. [Google Scholar] [CrossRef] [PubMed]
  344. Cariello, M.; Contursi, A.; Gadaleta, R.M.; Piccinin, E.; De Santis, S.; Piglionica, M.; Spaziante, A.F.; Sabbà, C.; Villani, G.; Moschetta, A. Extra-virgin olive oil from Apulian cultivars and intestinal inflammation. Nutrients 2020, 12, 1084. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  345. Parisio, C.; Lucarini, E.; Micheli, L.; Toti, A.; Bellumori, M.; Cecchi, L.; Calosi, L.; Bani, D.; Di Cesare Mannelli, L.; Mulinacci, N.; et al. Extra virgin olive oil and related by-products (Olea europaea L.) as natural sources of phenolic compounds for abdominal pain relief in gastrointestinal disorders in rats. Food Funct. 2020, 11, 10423–10435. [Google Scholar] [CrossRef] [PubMed]
  346. Huguet-Casquero, A.; Xu, Y.; Gainza, E.; Luis Pedraz, J.; Beloqui, A. Oral delivery of oleuropein-loaded lipid nanocarriers alleviates inflammation and oxidative stress in acute colitis. Int. J. Pharm. 2020, 586, 119515. [Google Scholar] [CrossRef]
  347. Larussa, T.; Imeneo, M.; Luzza, F. Olive Tree biophenols in inflammatory bowel disease: When bitter is better. Int. J. Mol. Sci. 2019, 20, 61390. [Google Scholar] [CrossRef] [Green Version]
  348. Sudeep, H.V.; Thomas, J.V.; Shyamprasad, K. A double blind, placebo-controlled randomized comparative study on the efficacy of phytosterol-enriched and conventional saw palmetto oil in mitigating benign prostate hyperplasia and androgen deficiency. BMC Urol. 2020, 20, 86. [Google Scholar] [CrossRef] [PubMed]
  349. Klippel, K.F.; Hiltl, D.M.; Schipp, B. A multicentric, placebo-controlled, double-blind clinical trial of beta-sitosterol (phytosterol) for the treatment of benign prostatic hyperplasia. German BPH-phyto study group. Br. J. Urol. 1997, 80, 427–432. [Google Scholar] [CrossRef] [PubMed]
  350. Cabeza, M.; Bratoeff, E.; Heuze, I.; Ramírez, E.; Sánchez, M.; Flores, E. Effect of beta-sitosterol as inhibitor of 5 alpha-reductase in hamster prostate. Proc. West. Pharm. Soc. 2003, 46, 153–155. [Google Scholar]
  351. Oyelowo, O.; Okafor, C.; Ajibare, A.; Ayomide, B.; Dada, K.; Adelakun, R.; Ahmed, B. Fatty acids in some cooking oils as agents of hormonal manipulation in a rat model of benign prostate cancer. Niger. J. Physiol. Sci. 2019, 34, 69–75. [Google Scholar]
  352. Filip, R.; Possemiers, S.; Heyerick, A.; Pinheiro, I.; Raszewski, G.; Davicco, M.-J.; Coxam, V. Twelve-Month Consumption of a polyphenol extract from olive (Olea europaea) in a double blind, randomized trial increases serum total osteocalcin levels and improves serum lipid profiles in postmenopausal women with osteopenia. J. Nutr. Health Aging 2015, 19, 77–86. [Google Scholar] [CrossRef]
  353. Angeles Rosillo, M.; Montserrat-de-la-Paz, S.; Abia, R.; Luisa Castejon, M.; Millan-Linares, M.C.; Alarcon-de-la-Lastra, C.; Fernandez-Bolaños, J.G.; Muriana, F.J.G. Oleuropein and its peracetylated derivative negatively regulate osteoclastogenesis by controlling the expression of genes involved in osteoclast differentiation and function. Food Funct. 2020, 11, 4038–4048. [Google Scholar] [CrossRef]
  354. Tagliaferri, C.; Davicco, M.-J.; Lebecque, P.; Georgé, S.; Amiot, M.-J.; Mercier, S.; Dhaussy, A.; Huertas, A.; Walrand, S.; Wittrant, Y.; et al. Olive oil and vitamin D synergistically prevent bone loss in mice. PLoS ONE 2014, 9, e115817. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  355. Taskan, M.M.; Balci Yuce, H.; Karatas, O.; Gevrek, F.; Toker, H. Evaluation of the effect of oleuropein on alveolar bone loss, inflammation, and apoptosis in experimental periodontitis. J. Periodontal Res. 2019, 54, 624–632. [Google Scholar] [CrossRef] [PubMed]
  356. Puel, C.; Mathey, J.; Agalias, A.; Kati-coulibaly, S.; Mardon, J.; Obled, C.; Davicco, M.-J.; Lebecque, P.; Horcajada, M.-N.; Skaltsounis, A.L.; et al. Dose–response study of effect of oleuropein, an olive oil polyphenol, in an ovariectomy/inflammation experimental model of bone loss in the rat. Clin. Nutr. ESPEN 2006, 25, 859–868. [Google Scholar] [CrossRef] [PubMed]
  357. Hagiwara, K.; Goto, T.; Araki, M.; Miyazaki, H.; Hagiwara, H. Olive polyphenol hydroxytyrosol prevents bone loss. Eur. J. Pharm. 2011, 662, 78–84. [Google Scholar] [CrossRef]
  358. Sacco, S.M.; Horcajada, M.-N.; Offord, E. Phytonutrients for bone health during ageing. Br. J. Clin. Pharm. 2013, 75, 697–707. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  359. Čabarkapa, A.; Živković, L.; Borozan, S.; Zlatković-Švenda, M.; Dekanski, D.; Jančić, I.; Radak-Perović, M.; Bajić, V.; Spremo-Potparević, B. Dry olive leaf extract in combination with methotrexate reduces cell damage in early rheumatoid arthritis patients-a pilot study. Phytother. Res. 2016, 30, 1615–1623. [Google Scholar] [CrossRef]
  360. Bahadori, S.; Salamzadeh, J.; Kamalinejad, M.; Shams Ardekani, M.R.; Keshavarz, M.; Ahmadzadeh, A. Study of the effect of an oral formulation of fig and olive on rheumatoid arthritis (RA) remission indicators: A randomized clinical trial. Iran. J. Pharm. Res. 2016, 15, 537–545. [Google Scholar]
  361. Matsumoto, Y.; Sugioka, Y.; Tada, M.; Okano, T.; Mamoto, K.; Inui, K.; Habu, D.; Koike, T. Monounsaturated fatty acids might be key factors in the mediterranean diet that suppress rheumatoid arthritis disease activity: The TOMORROW study. Clin. Nutr. 2018, 37, 675–680. [Google Scholar] [CrossRef]
  362. Edefonti, V.; Parpinel, M.; Ferraroni, M.; Boracchi, P.; Schioppo, T.; Scotti, I.; Ubiali, T.; Currenti, W.; De Lucia, O.; Cutolo, M.; et al. A posteriori dietary patterns and rheumatoid arthritis disease activity: A beneficial role of vegetable and animal unsaturated fatty acids. Nutrients 2020, 12, 3856. [Google Scholar] [CrossRef] [PubMed]
Table 1. Characteristics of the latest clinical trials that used MUFA-based FEEDs. Modified from [201,202,208].
Table 1. Characteristics of the latest clinical trials that used MUFA-based FEEDs. Modified from [201,202,208].
AuthorDesignITTSettingGroup CharacteristicsAdministrationTime (Max)I-FormulaI-Fat (%)I-MUFA (%)C-FormulaC-Fat (%)C-MUFA (%)Improvements
Lansink (2017) [209]Double-blind, crossover YesAmbulatoryIntervention and control groups DM2 (n = 24): 64.6 ± 10.7 yearsNasogastric tube, continuous feeding4 hNutrison Advanced Diason Energy HP46.427.6Nutrison Energy Multi Fibre34.419.4Postprandial IS
Postprandial BG
Gulati (2015) [203]Open-label, crossoverNoAmbulatoryIntervention (n = 22), DM2, 45.1 ± 6.9 years;
Control (n = 18), DM2, 47.7 ± 6.8 years
Oral4 hNutren
Diabetes
36.827.0Isocaloric dietNRNRPostprandial IS
Postprandial BG
Alish (2010) [210]Double-blind, crossoverNoAmbulatoryIntervention and control groups (n = 18): HbA1c 6.7% ± 0.2%, 63.1 ± 1.9 yearsOral4 hGlucerna 1.245.027.7“Standard formula”29.010.4Postprandial BG
Pohl (2009) [211]Double-blind, parallel YesNeurological rehab Dept., nursing home, medical wardIntervention (n = 48): DM2, HbA1c ≥ 7.0%, 74 (44–91) years;
Control (n = 49): healthy, 69 (53–86) years
Nasogastric tube, continuous feeding84 daysDiben45.032.2Isoenergetic, isonitrogenous enteral formula30.017.0Insulin requirements
Fasting BG
Vaisman (2009) [212]Double-blind, parallel NoAmbulatoryIntervention (n = 12): HbA1c 7.0% ± 0.7%, 73.0 ± 14.7 years;
Control (n = 13): HbA1c 7.3% ± 0.8%, 79.2 ± 10.4 years
Nasogastric tube, bolus feeding12 weeksNutrison Advanced Diason38.026.0“Standard formula”30.014.0Postprandial BG HbA1c
HDL-cholesterol
Vanschoonbeek (2009) [213]Double-blind, crossover NoAmbulatory3 intervention and control groups (n = 15): DM2, 63.0 ± 1.0 years, HbA1c 7.3% ± 0.2%Oral4 hGlucerna/Nutricia formula 1/Nutricia formula 250.034.8Isosource Fiber30.012.6Postprandial IS
Postprandial BG
Magnoni (2008) [214]Double-blind, parallel YesAmbulatoryIntervention and control groups (n = 39): DM2, HbA1c 6.5–8.5%, 57.5 ± 1.5 yearsOral84 daysDiasip49.034.2“Standard formula”30.017.1Postprandial BG
Voss (2008) [215]Double-blind, crossover NoAmbulatory2 intervention and control groups DM2 (n= 48): 56 ± 1.4 yearsOral4 hDiabetes-specific formula/Slowly digested carbohydrate formula49.0/49.032.0/32.2“Standard formula”29.015.0Postprandial IS
Postprandial BG
Yokoyama (2008) [216]Open-label, crossoverNoAmbulatoryIntervention: DM2 (n = 10), HbA1c 6.6% ± 0.7%, 58.6 ± 7.7 years;
Control: healthy (n = 12), 20.8 ± 1.2 years
Oral7 daysGlucerna49.334.3Enrich-SF (Meiji Milk Co.)30.88.2Lower AUC insulin and plasma glucose at 180 min
Hofman (2007) [217]Double-blind, crossover NoAmbulatory2 intervention and control groups (n = 11): DM2, HbA1c 7.0% ± 1.2%, 63 ± 9.4 yearsNasogastric tube (continuous feeding)6 hDiason/“Test product”38.0 /49.026.1/34.2Nutrison Standard Multifiber35.021.0Postprandial IS
Postprandial BG
Leon-Sanz (2005) [204]Open-label, parallelNoMedical wardIntervention (n = 36) DM2, 7.7% ± 1.7 %, 73.9 ± 9.6 years;
Control (n = 27), 7.8% ± 1.9%, 70.6 ± 8.7 years
Nasogastric tube (continuous feeding)13 daysGlucerna 1.050.034.2Precitene31.09.4Diarrhea, nausea
Pohl (2005) [218]Double-blind, parallel YesNeurological rehab Dept., nursing homeIntervention (n = 35): DM2, HbA1c ≥ 7.0%, 71 (42–86) years;
Control (n = 37): Healthy, 72.0 (51–87) years
Nasogastric tube and gastrostomy, continuous feeding84 daysDiben45.032.2Isoenergetic, isonitrogenous enteral formula30.017.0Insulin requirements
Fasting BG
Doola (2019) [219]Double-blind, parallelNoICUIntervention (n = 21): 62.4 ± 9.9 years;
Control (n = 20): 62.2 ± 11.3 years
Nasogastric or orogastric tube (continuous feeding)2 daysGlucerna Select48.6NRNutrison Protein Plus Multifiber35.3NRInsulin requirements
Mean BG and GV
Vahabzadek (2019) [220]Double-blind, parallelNoICUIntervention (n = 44), stress hyperglycemia, 61 (53–64) years;
Control (n = 44), stress hyperglycemia, 62 (57–64) years
Enteral feeding (nasogastric tube, orogastric tube or gastrostomy), continuous feeding with night rest15 days“High-fat formula”45.0NR“Standard formula”30.0NRInsulin requirements
Capillary glucose
TAG (at end of study)
Van Steen (2018) [221]Open-label, parallelYesICUIntervention (n = 52), DM2, 66.1 ± 13.2 years;
Control, (n = 49), DM2, 67.1 ± 10.0 years
Nasogastric tube3 daysGlucerna 1.545.029.4Fresubin Energy Fiber34.822.2Capillary and interstitial glucose (flash monitoring) mean absolute change
Wewalka (2018) [222]Open-label, parallelNoICUIntervention (n = 30), 60 ± 12 years;
Control (n = 30), 58 ± 16 years
Nasogastric tube (continuous feeding)7 daysDiben45.0NRFresubin Original Fiber30.0NRFasting BG
Diarrhea
Nourmohammadi (2017) [200]Double-blind, parallelNoICUOO + sunflower oil-based formula (n = 16), 58.50 ± 22.25 years;
Sunflower oil-based formula (n = 16), 57.56 ± 23.96 years;
Control (n = 16), 56.81 ± 24.17 years
Nasogastric tube (continuous feeding)10 daysOO + sunflower oil-based formula/sunflower oil-based formula45.0 /45.0NR/NR“Carbohydrate-based formula”30.0NRHDL-cholesterol (only for OO-based formula)
Diarrhea
Mesejo (2015) [223]Open-label, parallel YesICUGlucerna Select (n = 53):60 (45–71) years;
Diaba HP (n = 52): 57 (43–70) years;
Isosource Protein Fiber (n = 52):58 (46–68) years
Nasogastric tube28 daysGlucerna Select/Diaba HP49.0 /40.032.2/20.0Isosource Protein Fiber30.012.9Insulin requirements
Fasting BG
Hypoglycemia
GV
Egi (2010) [224]Open-label, crossover NoICUIntervention and control groups: stress hyperglycemia after esophagectomy (n = 8)Jejunostomy, continuous feeding16 h“Intervention formula”29.721.5“Standard formula”25.212.6Interstitial glucose (CGM)
HGI
Mesejo (2003) [225]Single-blind, parallel YesICUDM2 or stress hyperglycemia (n = 50);
Intervention (n = 26): 65.27 ± 14.95 years;
Control (n = 24): 64.67 ± 9.63 years
Nasgastric tube, continuous feeding14 daysNovasource Diabet Plus40.023.2Isosource Protein29.011.4Insulin requirements
Capillary and BG
ITT: intention to treat; I-formula: intervention formula; I-Fat: fat (as %) in the intervention formula; I-MUFA: MUFAs (as %) in the intervention formula; C-Formula: control formula; C-Fat: fat (as %) in the control formula; C-MUFA: MUFA (as %) in the control formula; OO: olive oil; BG: blood glucose; GV: glucose variability, HGI: high glucose index; CGM: continuous glucose monitoring; HDL: high-density lipoprotein; AUC: area under curve, NR: not reported; HbA1c: glycated hemoglobin; TAG: triglycerides.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Jiménez-Sánchez, A.; Martínez-Ortega, A.J.; Remón-Ruiz, P.J.; Piñar-Gutiérrez, A.; Pereira-Cunill, J.L.; García-Luna, P.P. Therapeutic Properties and Use of Extra Virgin Olive Oil in Clinical Nutrition: A Narrative Review and Literature Update. Nutrients 2022, 14, 1440. https://doi.org/10.3390/nu14071440

AMA Style

Jiménez-Sánchez A, Martínez-Ortega AJ, Remón-Ruiz PJ, Piñar-Gutiérrez A, Pereira-Cunill JL, García-Luna PP. Therapeutic Properties and Use of Extra Virgin Olive Oil in Clinical Nutrition: A Narrative Review and Literature Update. Nutrients. 2022; 14(7):1440. https://doi.org/10.3390/nu14071440

Chicago/Turabian Style

Jiménez-Sánchez, Andrés, Antonio Jesús Martínez-Ortega, Pablo Jesús Remón-Ruiz, Ana Piñar-Gutiérrez, José Luis Pereira-Cunill, and Pedro Pablo García-Luna. 2022. "Therapeutic Properties and Use of Extra Virgin Olive Oil in Clinical Nutrition: A Narrative Review and Literature Update" Nutrients 14, no. 7: 1440. https://doi.org/10.3390/nu14071440

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