Next Article in Journal
Volatile Compounds as Markers of Terroir and Winemaking Practices in Fetească Albă Wines of Romania
Previous Article in Journal
Synergistic Bioactive Potential of Combined Fermented Kombucha and Water Kefir
Previous Article in Special Issue
A Freshly Prepared Guava and Mamey Beverage Induces Subjective Satiety in Healthy Adults, Similar to a Commercial Control
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Olive Leaf Extracts as a Medicinal Beverage: Origin, Physico-Chemical Properties, and Bio-Functionality

by
Andreas Alexandros Panou
and
Ioannis Konstantinos Karabagias
*
Department of Food Science & Technology, School of Agricultural Sciences, University of Patras, 30100 Agrinio, Greece
*
Author to whom correspondence should be addressed.
Beverages 2025, 11(3), 66; https://doi.org/10.3390/beverages11030066
Submission received: 11 March 2025 / Revised: 29 April 2025 / Accepted: 30 April 2025 / Published: 6 May 2025
(This article belongs to the Special Issue Opportunities and Challenges for Functional and Medicinal Beverages)

Abstract

:
Medicinal beverages are herbal drinks that are consumed by people in numerous countries including China, India, Brazil, Greece, Turkey, and others. These herbal drinks possess many beneficial properties for human health, such as antioxidant, antimicrobial, anti-inflammatory, anticancer, antiaging, anti-fertility, antitumor properties, and anti-diabetic effects. Flavonoids, phenols, carotenoids, sterols, glucosinolates, alkaloids, polyacetylenes, coumarins, saponins, and terpenoids are the main constituents which offer benefits to human health. In this context, this review aimed to highlight medicinal beverages’ origin, physico-chemical properties, and bio-functionality, focusing mainly on olive leaf extracts and their bioactive components that have not been extensively studied.

1. Introduction

Progress in the functional foods and food plants sector has led to the reduction in synthetic antibiotics use and the alleviation of physical disorders [1,2]. Phenols, tannins, flavonoids, stilbenes, and anthocyanins are the main constituents of plants to which the anticancer, anti-diabetic and antiaging properties are attributed [2,3,4,5]. Some representative species are rosemary, sage, and peppermint, which in addition to their contribution to the improvement and enforcement of flavors and aromas, possess anticancer, antimicrobial, and anti-diabetic properties due to their essential oils [3,6,7,8]. The high demand for the plant species is due to the easy access, lower cost in comparison to convectional drugs and demand for natural alternatives choices [9]. An important substance which is contained both in sage and rosemary is rosmarinic acid, which possesses anti-inflammatory properties [3,6,7]. The positive effects of sage in hypolipidemia, irritation and memory enhancement have also been demonstrated [7]. Other herbs and vegetables that are used for the improvement of aroma and flavor and possess antioxidants and antimicrobial properties are dill leaves, bay leaves, basil, thyme, garlic, onion, tarragon, marjoram, celery, and shallot [10]. These herbs and vegetables can also find applications in medicine. The incorporation of these herbs into meat products and several other products such as sauces, vinegar, and mustards can contribute to the improvement of product appearance and increase in nutritional and medicinal value [11,12].
All herbal drinks have physical and chemical properties. Physical properties include temperature turbidity color, solid content, odor, and taste. Mineral content is included in chemical properties [13]. The chemical composition of herbal drinks consists of phytochemical compounds such as phenols, flavonoids, sterols, carotenoids, alkaloids, glucosinolates, saponins, coumarins, terpenoids, polyacetylenes, and other sulfur-containing compounds. Flowers, roots, and leaves present proportionally different phytochemical properties compared to herbal drinks. The compounds above can inhibit product oxidation and decrease disease risk conditions. Also, these phytochemical compounds are responsible for the pharmacological action of herbal drinks, such as antibacterial, antioxidant, anticancer, anti-inflammatory, anti-fertility, and antitumor properties, and anti-diabetic effects. The pharmacological properties may differ from type to type of herbal drink. The pharmacological action of herbal drinks is in accordance with the content of bioactive constituents. The most popular herbal drinks are many species of tea and olive leaf extract. The olive leaf extract possesses a distinctive position in herbal drink category on account of its high antioxidant, anti-inflammatory, anticancer, anticholesterolemic, antihypertensive, antimicrobial and antiviral properties, which are mentioned more extensively in Section 2. Table 1 presents the chemical composition of some selected herb species and herbal drinks containing broccoli by-products, among others, given their high content of polyphenols.

Consumption of Herbal Drinks in Several Countries and Their Positive Effect on Human Health

Herbal drinks are used in Africa and India for medicinal purposes and health improvement [17]. Herbal drinks can be found in the market either in isolated form (roots, flowers, leaves, and bark) or as complex herbal mixtures [18]. According to the World Health Organization, more than 80% of people in Africa and Asia use herbal medicines. Herbal drinks can be prepared by several means such as ‘akar kayu’, ‘jamu’, drink, herbal tea, herbal decoction, and herbal infusion [19]. Herbal tea has gained the interest of many countries such as Turkey, China, Brazil, India and others. Herbal teas are used in traditional medicine and pharmacy in Greece and the Eastern Mediterranean countries [20]. The overall consumption of herbal teas in 152 countries increased by 25.1% from 2003 to 2013. Countries that possess the highest percent of tea consumption are China, India, and Brazil, followed by Japan, Iran, USA, Russia, Argentina, and Turkey and others. The highest number of tea drinkers was noted in Ireland, Turkey, and the United Kingdom [21]. A rapid increase in tea consumption was recorded in Japan from 1986 to 2007, and the next year, it decreased [22]. This increase was attributed to the health benefits offered by catechins. Leung cha is a tea that is consumed in China and is utilized for the cure of fever [23].
In 2016, the population of overweight and obese people increased to over 1.9 billion adults [24]. The medicinal herbs that were used for the therapy of obesity by the Brazilian Health Regulatory Agency were green tea, green tea–Camellia sinensis (L.) Kuntze; carqueja–Baccharis trimera (L.) DC; hibiscus-Hibiscus sabdariffa L. and soursop Annona muricata L. [25]. Green tea is one of the most popular consumed plant species worldwide, either isolated or intercropped. The world production of green tea is going to be increased by 7.5% per year [26]. Generally, the combination of various plant species is effective in the cure of some types of cancer and obesity [27]. As far as the effectiveness and safety of herbal mixtures are concerned, there is a need for more research. In 2013, concerns related to the need for formal recognition in the countries of origin about the commerce and consumption of products were expressed at an event in China. The recent years the health benefits of herbal teas and spices consumption, has led to a rapid expansion of the sector, concerning both organic and conventional spices and herbs, integral to herbal tea production, that is rapidly growing and is expected to reach approximately $31.95 billion by 2028 [28]. Considering the above, the main scope of the review article was to highlight the medicinal beverages’ origin, physico-chemical properties, and bio-functionality, focusing mainly on olive leaf extracts and their bioactive components that have not been extensively studied in the literature.

2. Olive Leaf Extracts

Olive leaves are the leaves of the olive tree (Olea europaea L.), which has been cultivated in Mediterranean countries for centuries [29]. For example, Egypt produces approximately 332,321 tons of olives annually and has a total cultivation area equal to 1% of the world’s total (100,000 ha) [30]. In Algeria and Tunisia, approximately 8 million hectares are cultivated with olive trees [31]. Olive leaves are a by-product that represent more than 10% of the weight of harvested olives [32]. Olive leaf extracts have been used for the cure of fever and malaria [33] and in various pharmacological applications [34]. Olive leaves contain organic matter (76.4–92.7 g/100 g dry matter), low quantities of crude protein (6.31–10.9 g/100 g dry matter), amino acids (89.9 g/100 g total nitrogen), nitrogen attached to the cell walls (49.2 and 35.4 g/100 g total nitrogen), crude fat (2.28–9.57 g/100 g dry matter), mannitol and glucose, bioactive components, and inorganic matter in different proportions, depending on: (i) terroir, (ii) cultivar, (iii) geographical position, (iv) methods of drying and extraction, (v) period of harvest, and (vi) standardized methods of phenolic compounds’ concentration, among others [35,36,37]. Bioactive components include phenols, such as flavonoids (luteolin, apigenin, luteolin-7-O-glucoside, etc.), tyrosol, hydroxytyrosol, ferulic acids and caffeic, and secoiridoids (oleuropein, dimethyloleuropein, ligstroside) [16,38,39].
The major polyphenolic compounds of olive leaves are oleuropein (24.54%), hydroxytyrosol (1.46%), tyrosol (0.71%), luteolin-7-O-glucoside (1.38%), apigenin-7-O-glucoside (1.37%), and verbascoside (1.11%) [40]. Caffeic acid, vanillic acid, and luteolin are present in lower amounts. Several factors, such as hydric deficiency, salinity, fertilization, geographical zone, sampling time, light exposition, frost stress, fungi, bacteria, genotypes, leaves age, and alternate bearing patterns of olive can change the synthesis and the number of phenols [41]. The above-mentioned polyphenols and other flavonoids, phenolic acids, phenolic alcohols, and secoiridoids are found also in different proportions in various parts of the olive tree, including the tree (branches or twigs), fruits, leaves, and virgin olive oil [12], and have antioxidant, antimicrobial, antiviral, antihypertensive, anticholesterolemic, anti-inflammatory, hypoglycemic, and anticancer activities [42,43,44,45,46,47]. Also, they decrease the risk of cardiovascular diseases and regulate blood pressure and cholesterol levels in animals [48,49]. All of the activities above are attributed mainly to the predominant phenolic compound oleuropein. Oleuropein has a diverse concentration (mg/kg) in olive leaves ranging from 5526 mg/kg to 26,471 mg/kg, whereas much lower amounts (≤2 mg/kg) are found in olive oil [49]. However, before going any further, it is mandatory to elucidate the biochemical pathway of oleuropein in olive fruits and leaves. Mostly, enzymes, including β-glucosidase, esterases and oxidoreductases, control the hydrolysis and the oxidation of oleuropein. In particular, β-glucosidase releases glucose, forming thus, the aldehydic form of oleuropein aglycon and the dialdehydic form of decarboxymethyl oleuropein aglycon, while esterases hydrolyze the ester bonds of oleuropein, producing glucosyl derivates, hydroxytyrosol and elenolic acid. In the gastrointestinal tract, oleuropein is hydrolysed by the action of enzyme β-glucosidase as a result the production of oleuropein aglycone [50]. Figure 1 shows the typical structures of oleuropein and its hydrolyzed derivatives.

2.1. Antioxidant Activity

Olive leaves exhibited the highest antioxidant activity against free radicals and reduced the oxidative stress in mice [43,44]. Oleuropein is found in the highest amounts in olive leaves and possesses antioxidant, antihypertensive, anti-inflammatory, hypoglycemic, and antiarrhythmic activities [44,51,52,53]. In a study, the intake of olive leaf extract and oleuropein by mice led to a significant reduction in the oxidative action of cyclophosphamide [54]. Oleuropein can contribute to the prevention of low-density lipoprotein oxidation (LDL). This prevention depends on its concentration [55,56]. Also, oleuropein can scavenge hypochlorous acid, which is produced in vivo by neutrophil myeloperoxidase at the site of inflammation and can oxidize the proteins and enzymes [55]. Oleuropein has also a scavenging effect against nitric oxide and increases the inducible nitric oxide synthase (iNOS) expression [57]. An experiment conducted on rabbits showed that oleuropein increases the oxidative resistance of LDL and simultaneously induces a decrease in the levels of total, free, and esterified cholesterol in plasma [58].
Manna et al. [59] investigated the protective effect of oleuropein in an isolated rat heart. Their results show that oleuropein induces a reduction in the release of a significant susceptible indicator of the exposure of the heart to oxidative stress called oxidized glutathione. A prevention was also observed in membrane lipid peroxidation. Puerta et al. [60] reported that an oleuropein concentration equal to 80 μM inhibited the leukotriene B4 generation peritoneal leukocytes of intact rats after their stimulation with calcium ionophore. Visioli et al. [61] researched the effect of oleuropein aglycone on human volunteers. The results of this study revealed that aglycone of oleuropein induced a decrease in an indicator of oxidative stress urinary production of 8-iso-PGF2α. There was no dependence between this reduction and concentration of oleuropein aglycone. According to studies, oleuropein can activate the transcription of nuclear factor erythroid 2-related factor 2, which leads to the stimulation of the intracellular antioxidant enzymes’ expression and increase in concentration of glutathione, vitamin C, β-carotene, and α-tocopherol [62,63,64,65]. In another study, oleuropein at concentrations of 50 and 100 µM caused a reduction in cell death caused by H2O2. The percentages of cell death were 6.4% for 50 µM and 9.2% for 100 µM [66]. Also, it was supported that the antioxidant effects of oleuropein have a relationship with the decrease in radical oxygen species in HaCaT cells. IL-1β and thioredoxin also take part in the protective action of oleuropein in HaCaT cells. Oleuropein decreases the concentration of IL-1β and inhibits the activity of caspase-1/IL-1-converting enzymes.

2.2. Anti-Inflammatory Activity

Inflammation is a crucial and defensive response that appears after an infection or damaged tissue and is a responsible factor for the promotion of many diseases such as atherosclerosis, diabetes mellitus, metabolic syndrome, cancer, and diseases of the kidney and nervous system [67]. In a previous study, oleuropein at concentrations 12 μΜ and 200 µM induced a decrease in the level of proinflammatory cytokine interleukin (IL)-6 [64]. This reduction was equal to 38.8% for 12 μM and 45.5% for 200 μM. Castejon et al. [68] found that oleuropein caused an inhibition in murine lupus nephritis NLRP3 inflammasome-related signaling pathways. Acute and chronic inflammation are the two main categories of inflammation. In chronic inflammation, the reactive oxygen (ROS) species take part and are responsible for oxidative degradation and the depletion of antioxidants [69]. IL-1, IL-6, TNF-a, and IFN-γ are the main mediators of the inflammatory response [70,71,72,73]. A damaged tissue, in addition to inflammatory cytokines or mediators, also releases monocyte chemoattractant proteins (MCP-1), cyclooxygenase (COX), inducible form of nitric oxide synthase (iNOS), metalloproteinases (MMP), and adhesion molecules. Furthermore, a significant role in the production of countless pro-inflammatory mediators plays nuclear factor Kappa β (NF-kβ) [42].
Studies have shown the reduction in serum C-reactive protein (CRP), IL-6, endothelial and monocyte adherence molecules (ICAM-1 and VCAM-1), and chemokines using the Mediterranean Diet (MD), which included extra virgin olive oil [74]. A study revealed that oleuropein inhibited the secretion of IL-1β [75]. Ryu et al. [76] reported that oleuropein downregulates the key markers iNOS, COX-2, NF-Kβ, and JNK, and of the two pro-inflammatory interleukins, IL-6 and IL-1β. In mice, the symptoms of colitis, which is caused by dextran sulfate sodium (DSS), were receded by the intake of oleuropein [77]. Oleuropein induced a decrease in COX-2, iNOS, and MMP-9, and suppressed p38 MAPK phosphorylation, which may be due to the upregulation of annexin A1 [78]. The production of TNF-α and IL-1β was attenuated by oleuropein in a mouse model of carrageenan-induced pleurisy, in a rat model of post-traumatic stress disorder, and in rats with spinal cord trauma [79,80,81]. In another study, oleuropein had a beneficial effect on ovariectomy/inflammation experimental model of bone loss in rats [82]. Larussa et al. [83] claimed that the levels of COX-2 and IL-17 and the inflammation of the colon tissue can be reduced by the provision of oleuropein on colon biopsies taken from ulcerative colitis subjects.

2.3. Hypoglycemic Activity

The symptom of diabetic patients (both type 1 and 2), chronic hyperglycemia, is responsible for damages of organs such as heart and blood vessels, nerves, kidneys and eyes [84]. Oxidative stress is responsible factor for diabetes complications and the increase in insulin resistance and diabetes mellitus [85,86,87,88,89]. Flavonoids and polyphenols can contribute to the reduction in negative effects of oxidative stress and free radicals in diabetic patients [90]. The high concentration of phenols in olive leaves is very significant for human health [91]. The most predominant phenolic compounds that possess many beneficial properties for human health are oleuropein and its hydrolysis products, elenolic acid and hydroxytyrosol [92,93]. All these beneficial properties are probably attributed to the antioxidant properties of oleuropein and its hydrolyzed products [91].
The antihyperglycemic properties of oleuropein have been supported by Gonzalez et al. [92] and Romani et al. [93]. Also, according to Al-Azzawie et al. [94], oleuropein may cause inhibition in hyperglycemia and oxidative stress caused by diabetes, and the supply of oleuropein may suppress diabetic complications that come from oxidative stress. In a study, the supply of oleuropein in diabetic type 1 rats induced a significant inhibition in the increase in glucose compared to the untreated rats with oleuropein rats [95]. The provision, for 4 weeks, of oleuropein (8 mg/kg body weight) and hydroxytyrosol (16 mg/kg body weight) rich extracts caused a significant reduction in the serum glucose levels [63]. Oleuropein on physiological concentrations activated the adenosine monophosphate-activated protein kinase on cultured mouse byoplast, which has been connected with the increased cellular internalization of glucose [96]. Oleuropein caused a decrease in glucose transporter GLUT-2 (glucose transporter-2) of cultured cells [64]. GLUT-2 regulates the absorption of intestinal glucose and insulin secretion.
According to Wu et al. [97], the secretion of insulin by pancreatic β-cells is facilitated by oleuropein, which is connected with the activation of the MAPK pathway. Also, the cytotoxicity of β-cell-caused amyloids is prevented by oleuropein. A recent study showed that the pancreatic β-cell cytotoxicity induced by human islet amyloid polypeptide aggregates was inhibited by oleuropein aglycone at a concentration of 10 μΜ, as a result the protection of the plasma membrane from permeabilization and the prevention of cell death [98]. A reduction was achieved in the concentration of blood glucose, plasma insulin, and hepatic glycogen of mice after the intake of oleuropein at concentrations of 5–10 ppm per day [99]. Also, another study showed a significant decline in blood glucose levels, an increase in glucose tolerance, a reduction in the homeostasis model assessment insulin-resistance index, and a promotion of protein kinase B activation after the intake of oleuropein at concentrations of 200 ppm for 15 days in diabetic mice [100]. All the histopathological changes in mice are restored by the oleuropein. Also, important changes were recorded in the composition of the gut microflora of diabetic mice after oleuropein consumption. A study demonstrated that polyphenols of extra virgin olive oil (EVOO) had important beneficial effects on intestinal bacteria [101]. Generally, the dysregulation of gut microbiota can induce a modification in intestinal permeability and affect several pathophysiological mechanisms that can have a negative effect on glucose metabolism. Oleuropein participates beneficially in glucose metabolism and can cause changes in the glucose transport and intracellular metabolism, increase in insulin susceptibility and glucose-stimulated insulin production by pancreatic β-cells.
The consumption of polyphenol-rich EVOO (25 mL per day, 577 mg of phenolic compounds per kg of body weight) for 4 weeks induced an important decrease in body weight, fasting plasma glucose, and glycated hemoglobin [102]. These changes were connected with an important mitigation in serum visfatin, a pro-inflammatory adipocytokine. Another study showed that the intake of EVOO decreased the risk of developing type 2 diabetes by 16% [103]. Also, the levels of plasma glucose and glycated hemoglobin in type 2 diabetics were significantly reduced by the supplementation of EVOO, showing that the consumption of EVOO could be useful for the treatment of type 2 diabetes. The intake of oleuropein in capsules (51 mg/day) or a placebo for 12 weeks significantly improved the insulin sensitivity and pancreatic β-cells secretory capacity of 46 middle-aged, overweight men [104]. In a study, the intake of oleuropein (35 to 200 mg/day) reduced the post-prandial blood glucose response after the intake of sucrose but did not affect post-prandial glucose after the consumption of bread or glucose [105]. Oleuropein inhibited the sucrase and GLUT-2-mediated transport, but α-amylase was not affected significantly. A study showed a fast reduction in blood glucose after the oral supplementation of two capsules of Tensiofytol per day (100 mg of oleuropein and 20 mg of hydroxytyrosol) by hypertensive patients suffering from obesity and/or diabetes [106].
Blood glucose and dipeptidyl-peptidase 4 (DPP-4) protein concentration and activity were lower after the supplementation of 20 mg oleuropein in human patients subjected to the Mediterranean diet compared to those people who were subjected to the Mediterranean diet without the addition of oleuropein [98]. Also, higher levels of serum insulin and glucagon-like peptide-1 (GLP1) were monitored. The supplementation of oleuropein at 20 mg before the lunch on blood glucose and DPP-4 was reported, and an important decline in markers of oxidative stress, such as soluble NADPH oxidase-derived peptide activity and 8-iso-prostaglandin-2α was reported [107]. Mediterranean diet meals containing the amount of oleuropein that is contained in 10 g of EVOO reduced blood glucose levels and DPP-4 activity and increased insulin and GLP-1 levels in comparison to a conventional Mediterranean diet meal without oleuropein [108]. In another study, blood glucose levels exhibited a lower increase in type 2 diabetic patients after the consumption of 40 g of oleuropein-enriched chocolate in comparison to the intake of conventional chocolate [109]. These findings prove that oleuropein might induce a decline in post-prandial blood glucose by its involvement in a mechanism that counteracts oxidative stress-mediated incretin downregulation.

2.4. Anticholesterolemic Activity

The supply of oleuropein to diabetic rats inhibited significantly (24.80%) the increase in cholesterol compared to the no-fed-oleuropein diabetic rats (Table 2) [95]. The provision of oleuropein and hydroxytyrosol-rich extracts for 28 days significantly decreased the cholesterol levels of rats [63].
A study conducted in mice treated with oleuropein (100 mg/kg/day) for 42 days showed that oleuropein acts as a ligand of the peroxisome proliferator-activated-receptor alpha (PPARα), which is responsible for the activation of gene encoding enzymes that tale part in fatty acid cell uptake, mitochondrial β-oxidation, microsomal ω-oxidation, genes that encode many apolipoproteins and a family of genes that critically participate in many metabolic pathways [110]. Consequently, PPARα controls the circulating fatty acids and biochemical processes associated with insulin sensitivity and glucose metabolism. The oral intake of oleuropein at a concentration of 50 mg/kg by cholesterol-fed rats induced mitigation in body weight and adipose tissue mass [111]. This reduction in body weight is related to the significant inhibition of peroxisome proliferator-activated-receptor gamma (PPARγ) and an increase in serum adiponectin levels. The hypocholesterolemic activity of oleuropein was reported by Romani et al. [93].

2.5. Antihypertensive Activity

The hypotensive effect of olive leaves has been well studied [112,113,114]. The most popular phenolic constituents that are contained in olive leaves are oleuropein and oleacein; however, the latter is active only in good quality extra virgin olive oil [115]. According to Petkov and Manolov [116] oleuropein had a hypotensive, coronary dilating, and antiarrhythmic action. Another study demonstrated that hydroxytyrosol exhibited a calcium-antagonistic action [117]. Hansen et al. [118] reported that oleacein can inhibit the distinct angiotensin-converting enzyme (ACE). Bioactive peptides that act as inhibitors of ACE have been obtained from various sources of animal and vegetable origin. The majority of ACE inhibitory peptides are isolated by the action of specific proteases on different sources of dietary proteins. These include dairy and fermented milks, eggs, soybeans, chickpeas, peanuts, tuna, sardines, shrimp, chicken, and squid, among others. A representative example of ACE inhibitor peptides is found in hydrolysates of milk proteins.
The development of severe hypertension and atherosclerosis in insulin-resistant rats was prevented after the supply of isolated triterpenoids from African wild olive leaves, Greek olive leaves, and olive leaves from Cape Town [119]. The isolate of the African wild olive leaves contained 0.27% iso-molecular mixture of oleanolic acid and ursolic acid. The isolate of Greek olive leaves consisted of 0.71% oleanolic acid, and the Cape Town olive leaves contained 2.47% oleanolic acid. The provision of Arbequina table olives (3.85 g/kg) in rats that suffered from hypertension caused a transient reduction in blood pressure, which was observed on the second week after the provision and lasted until the seventh week [120]. Generally, some studies have demonstrated the antihypertensive effect of olive oil polyphenols in rats that suffered from hypertension [121], people who suffered from high cholesterol levels [122], pre-hypertensive [123,124] and people who suffered from hypertension [125], and coronary heart disease patients [126]. A study has shown that the consumption of olive oils with at least 150 mg/L (ppm) of polyphenols may have positive effects on systolic blood pressure [127]. A significant blood pressure reduction has been observed by the consumption of triterpenes for many years [128,129]. Recently, a study revealed that olive oils containing high amounts of triterpenes (86–389 ppm) did not affect blood pressure [130]. A study showed that OLE exhibited a protective action against the rise in blood pressure caused by nitro-L-arginine methyl ester at concentrations of 50 and 100 ppm [131]. A decrease in systolic left ventricular pressure and heart rate of rabbit hearts was achieved after the intake of oleuropein [132]. The possible mechanisms of the cardioprotective effects of olive leaf extract are the blockade of beta receptors [133], angiotensin-converting enzyme (ACE) inhibition, Ca2 + antagonism [134], and vasodilatory NO pathways [93].
In a study conducted in 20 monozygotic adult twin pairs suffering from mild hypertension in Germany, the intake of olive leaf extract at a dose of 0.5 or 1.0 g daily for 56 days led to a significant decline in systolic and diastolic blood pressure [135]. The consumption of olive (Olea europaea L.) leaf extract (EFLA®943) by patients at the dose of 0.5 g twice daily in a flat-dose manner for 56 days caused a mitigation in systolic and diastolic blood pressures of patients who were taking Captopril at a dose of 12.5–25 mg twice daily [136]. An important decrease in blood pressure in patients was observed after the intake of oleuropein in a dose of 1.0 g four times daily for 84 days [137]. A reduction in the digital volume pulse stiffness index was also found after the consumption of 51 mg of oleuropein once by 18 male and female volunteers [135]. Consuming olive leaf extract containing high amounts of polyphenols (136 mg) for 42 days reduced systolic and diastolic blood pressure throughout the day, and the 24 h measure in comparison to the control group [137]. Another study demonstrated that the consumption of olive leaf extract tablets containing 16% oleuropein and at least 0.62 mg of luteolin induced a decrease in inflammatory factors associated with hypertension, while no changes were exhibited in renal and liver functions [138].
The provision of olive leaf extract containing oleuropein at concentrations of 20–60 ppm on type 2 diabetic animals accompanied by renal hypertension, caused a decline in systolic blood pressure and a rise in nitric oxide concentration, which is connected with beneficial vasodilator effect [126]. Researchers have attributed the protective action of oleuropein against the cardiovascular system in type 2 diabetes to the inhibition of the angiotensin-converting enzyme action, the stenosis of calcium channels, the swelling of blood vessels, the restoration of endothelium function, and the scavenging of oxygen free radicals [64,139,140]. Oxidative stress plays an important role in the pathogenesis of cardiovascular disorders, and the increase in reactive oxygen species in human carotid arteries of older people, which results in the excessive accumulation of superoxide ions by NADPH oxidase [141]. The increase in oxygen-reacted species in the vascular tissue of hypertensive rats has been connected with the generation of hypertension [142]. A significant beneficial effects of different polyphenol-rich plant products on hypertension-induced alterations of the carotid have been reported [143,144]. This beneficial effect leads to a significant reduction in blood pressure and systemic oxidative stress.

2.6. Anticancer Activity

Generally, cancer is treated by surgery in the early stage or by specific chemotherapies and immunotherapies when it has spread. The disadvantage of chemotherapies and immunotherapies is the resistance to drugs and the generation of host side effects. One way for the reduction in drug resistance and increase in treatment efficacy is the combination of conventional treatment with biological agents (complementary therapy).
Bouallagui et al. [145] observed an inhibition of accumulation of luminal breast cancer cells (MCF-7 cells) by the supply of extract rich in oleuropein and its derivative hydroxytyrosol. Similar results were reported by Han et al. [146]. Oleuropein can also negatively affect the NF-kB expression, which is responsible for the control of many genes driving cancer development and progression [147,148]. Oleuropein exhibited a pro-oxidant and anti-proliferative effect in androgen-insensitive DU145 prostate cancer cells [146]. Oleuropein also prevents azoxymethane, which causes pre-neoplastic lesions of the colon and decreases dysplasia and DNA damage [147]. A previous study reported that HIF-1a was inhibited significantly by oleuropein and hydroxytyrosol, and the expression of p53 in HT-29 human colon adenocarcinoma cells was promoted. The latter led to apoptosis [148]. The oleuropein inhibitory effect on human colon carcinoma cells may be facilitated by long-chain fatty acids and their esters, such as hydroxytyrosol oleate [149]. A high anti-proliferative activity on pancreatic cancer cells (MiaPaCa-2 cells) by oleuropein has also been reported for Corregiola leaf extracts.
In a previous study, 51 analogs of oleuropein were studied on several human cancer cells and it was reported that analog 24 expressed the highest inhibitory effect in vitro (human colon cancer cells HCT-116, human cervical carcinoma cells HeLa, MCF-7 cells) as well as in vivo (B16-F10 mouse melanoma cells) [150]. It was found that oleuropein promotes apoptosis in cancer cells, like HeLa cells [151], HepG2 human hepatoma cells [152], SH-SY5Y human neuroblastoma cells [153], and HCT116 cells [154]. Also, oleuropein exhibited pro-apoptotic activity against HL60 human promyelocytic leukemia cells [155]. Studies have proven the effect of oleuropein on the most aggressive skin cancer, melanoma [156]. A study revealed that oleuropein altered the dynamics of intracellular Ca2+ in mesothelioma cancer cells and changed the T-type Ca2+ channels [157]. Another study showed that oleuropein can affect the modulation of onco-miRNAs (miRNA-21 and miR-155) genes, which suppress tumor development [158]. Tezcan et al. [159] claimed that oleuropein may affect the expression of miRNAs, miR-137, -145, and -153, in glioblastoma multiforme (GBM) cancer stem cells. A significant role in cancer treatment, play the histone deacetylases (HDAC2 and HDAC3) inhibitors. In a previous study, it was supported that oleuropein may inhibit histone deacetylases inhibitors (HDAC2 and HDAC3) [160].
Obesity is a reason for the development of many cancers and metastatic dissemination. In mice melanoma cells, oleuropein inhibited tumor growth and lymph node metastasis and abrogated angiogenesis and lymphangiogenesis through the decrease in the peroxisome proliferator-activated receptor γ and the infiltration of M2 macrophages [161]. Anter et al. [155] reported that oleuropein displayed anticancer/pro-apoptotic activity on leukemic cells, like HL60 human promyelocytic leukemia cells. Oleuropein showed anticancer activity in glioma cells by reducing the ability of MMP-2 and -9 to invade extracellular tissues [162]. The promotion of a decline in the metastatic ability of breast cancer cells by oleuropein was demonstrated by Hassan et al. [163]. This reduction was attributed to MMP-2 and MMP -9 decrease, strengthened by the promotion of tissue inhibitors of MMP (TIMP) 1,3,4. The authors also reported that the cancer cell invasion and propagation were regulated by the balance of MMP and TIMP.

3. Additional Anticancer, Antiviral, and Antimicrobial Properties of Oleuropein and Olive Leaf Extracts

Several studies have been carried out on the use of oleuropein in complementary cancer therapy on account of its anticancer properties, anti-proliferative, and pro-apoptosis activity. The side effects and toxicity in tumor-bearing patients caused by anticancer drugs could be treated by oleuropein. The cell accumulation of PC3 prostate cancer cells was affected by the application of co-treatment with very low amounts of doxorubicin and Ole [164]. This co-treatment could cause a significant promotion of autophagy. The relationship of cancer cells by autophagy reveals a new therapeutic treatment for Ole. The protection of cardiotoxicity of doxorubicin by oleuropein could be a good reason for the application of oleuropein in cancer co-treatments [165]. Oleuropein synergistically increases bevacizumab’s effect, which is used in the treatment of the most malignant human cancer GMB by preventing VEGFA, MMP-2, and -9 activities [166]. A study has shown the participation of PI3K/Akt/mTOR in the promotion of protein synthesis and suppression of autophagy [167].
The mTOR inhibition and autophagy induction caused by oleuropein could be used in the development of new therapeutic methods against GBM. Oleuropein can affect cell proliferation in BRAF melanoma cells by downregulation of the pAKT/pS6 pathway [168]. Also, oleuropein enhances the cytotoxic effect of dacarbazine against BRAF melanoma cells. The exposure of melanoma cells to oleuropein leads to reverse trastuzumab resistance in HER2-overexpressing breast cancer cells [169]. Oleuropein at a concentration of 200 μΜ can enhance the decrease in MMP-7 gene expression in HepG2 cells caused by cisplatin [170]. This was attributed to the reduction in the cancer-promoting ability of nerve growth factor (NGF) on HepG2 cells. The mature NGF is produced from its precursor form, pro-NGF, through MMP-7 proteolytic cleavage and exhibits a pro-survival effect on HCC cells. The stimulation of oleuropein with cisplatin simultaneously potentiates caspase-3 gene expression, which leads to the enhancement of apoptotic rate. A previous study supported the idea that oleuropein potentiates the sensitivity of nasopharyngeal carcinoma cells to radiation [171]. Generally, oleuropein can discriminate the cancer from normal cells, prevent accumulation, and promote apoptosis in several tumors such as mesothelioma, GBM, and melanoma. These abilities above have gained the researchers’ interest in the discovery of new cancer methods of treatment.

3.1. Antiviral Activity

The antiviral activities of O. europea against viral hemorrhagic septicemia virus (VHSV), herpes virus, hepatitis virus, rotavirus, bovine rhinovirus, canine parvovirus, and feline leukemia virus have been proven [172,173]. In Vero cells, olive leaf hydroalcoholic extract displayed antiviral activity against the herpes simplex virus type at concentrations higher of 1 mg/mL [174]. Leaf extracts from Olea europea L. var. sativa (OESA) and Olea europea var. sylvestris (OESY) showed significant antiviral activity against the herpes simplex type 1 (HSV-1) were assessed on Vero cells at a concentration of 200 ppm and 820 ppm, respectively [175]. Also, Lee-Huang et al. discovered that olive leaf extracts-displayed antiviral action against HIV-1 infection and replication at a concentration of 200 ppb (μg/L) [176]. The replication of HSV-1 in HeLa cancer cells was inhibited by the half-maximal effective concentration of oleuropein 0.241 mg/mL [177]. A study showed that oleuropein extracted from Jasminum officinale L. var. grandiflorum inhibited HBV action in HepG2 2.2.15 cells test in vitro and DHBV activity in ducklings test in vivo [178]. Oleuropein effectively blocks HBsAg production in HepG2 2.2.15 cells. The effectiveness of the block production of HBsAg was analogous to the oleuropein dose with IC50 equal to 23.2 μg/mL. The reduction in DHBV replication by oleuropein in infected ducklings by DHBV in vivo was achieved at a concentration of 80 mg/kg of oleuropein.
According to a recent study, olive leaf phytoconstituents induced a significant inhibition in SARS-CoV-2 [179]. The standardized olive leaf extract (SOLE) containing 20% oleuropein exhibited moderate antiviral activity against SARS-CoV-2 with an IC50 of 118.3 μg/mL. Also, olive leaves can significantly inhibit respiratory viruses, such as the syncytial virus and type 3 parainfluenza virus [180]. Oleuropein and other phytochemical constituents of olive leaves (3,4-hydroxyphenylethanol, 3β-Hydroxyolean-12-en-28-oic acid, 2α,3β-Dihydroxyolean-12-en-28-oic acid, 3′,4′,5,7-Tetrahydroxyflavone, 7-O-beta-D-Glucosyl-5,7,3′,4′-tetrahydroxyflavone, verbascoside, apigenin-7-O-glucoside and 3,4′,5,7-Tetrahydroxyflavone) have shown in silico antiviral activities against SARS-CoV-2 viral proteases (Mpro/3CLpro, PLpro), TLRs, ACE2, RBD, NSP15, HSPA5, SBDβ, TMPRSS2, S protein and Furin [181,182,183,184,185,186,187,188]. Hydroxytyrosol, luteolin, and kaempferol have shown antiviral action against SARS-CoV-2 in vitro [181,188,189,190,191]. Non-structural protein 12 (nsp12), 3C-like protease (3CLpro), and papain-like protease (PLPro) are essential keys for the preparation of targeted antiviral drugs (DAAs) against COVID-19. The 3C-like protease (3CLpro) and papain-like protease (PLPro) take part in the SARS-CoV-2 replication cycle by controlling the polyprotein produced during transcription into functional subunits [192,193,194].

3.2. Antimicrobial Activity

Oleuropein and other phytochemicals found in olive leaf extracts can inhibit or delay the growth rate of a plethora of bacteria and fungi. Olive extract’s oleuropein has shown high antimicrobial activity on a wide range of Gram-positive and Gram-negative bacteria and fungi, such as Bacillus cereus, Bacillus subtilis, Staphylococcus aureus, Pseudomonas aeruginosa, Escherichia coli, Klebsiella pneumoniae, Candida albicans, and Cryptococcus neoformans in vitro [195,196]. Oleuropein can suppress the growth of both ATTC bacterial strains and clinical bacterial strains (e.g., Haemophilus influenzae, Moraxella catarrhalis, Salmonella typhi, Vibrio parahaemolyticus, Staphylococcus aureus) [197,198]. The dialdehydic form of decarboxymethyl oleuropein and ligstroside aglycons, hydroxytyrosol, and tyrosol exhibited bactericidal activity against harmful bacteria of the microflora of gut Clostridium perfringens and Escherichia coli and against beneficial bacteria such as Lactobacillus acidophilus and Bifidobacterium bifidum [198]. Foodborne pathogens such as Listeria monocytogenes, Staphylococcus aureus, Salmonella enterica, Yersinia sp., and Shigella sonnei are killed after contact with the phenols above for 1 h. In a study, the growth of Staphylococcus aureus and Bacillus subtilis were inhibited at minimum inhibitory concentrations 0.6 to 1.6 mg/mL and 1.2 to 1.8 mg/mL, respectively [199]. According to Bisignano et al. [200], Staphylococcus aureus exhibited minimum inhibitory concentration for oleuropein from 0.0625 to 0.5 mg/mL for certified strains of the American Type Culture Collection (ATCC) and from 0.0312 to 0.250 mg/mL for clinical isolates. In a study, it was found that oleuropein displayed high inhibitory action against Campylobacter jejuni, Helicobacter pylori, and Staphylococcus aureus with minimum inhibitory concentration as low as 0.31% v/v, 0.62% v/v, and 0.78% v/v, respectively [200]. Previous studies have demonstrated that Campylobacter jejuni isolates were more sensitive to various natural compounds compared to other microorganisms such as Escherichia coli, Salmonella, and Listeria [201].
A previous study reported that Staphylococcus aureus exhibited the highest sensitivity to oleuropein extracts, while Escherichia coli O1577:H7 displayed the highest resistance [202]. Leaf extracts of Olea europea L. var. sativa (OESA) and Olea europea var. sylvestris (OESY) exhibited antimicrobial action against Gram-positive bacteria at minimum inhibitory concentrations of 7.81 and 15.61 μg/mL and Staphylococcus aureus ATCC 6538 at minimum inhibitory concentrations of 15.61 and 31.25 μg/mL [174]. Moreover, oleuropein may enhance the bactericidal action of peracetic acid against L. monocytogenes biofilms [203]. The combination of oleuropein with commercial sanitizers increased their bactericidal action, especially for Chlorhexidine digluconate, which resulted in an approximately 60-fold decrease in the minimum inhibitory concentrations values for Staphylococcus aureus and Listeria monocytogenes (2.5 mg/mL) [204]. Oleuropein can interact with the phosphate group and modify the phospholipid/water interface properties [205]. In another study, it was reported that oleuropein interacted with phosphatidylglycerol and phosphatidylethanolamine of Staphylococcus aureus and Escherichia coli, and the antimicrobial activity was exhibited only at the cell surface because oleuropein is not able to form stable mixed monolayers with the lipids in the interface [206]. This phenomenon may be attributed to the prevention of the glycosidic group of oleuropein diffusion through the cell membrane [197]. Juven et al. [207] reported that oleuropein affected a significant leakage of glutamate, potassium, and inorganic phosphate from Lactobacillus plantarum. Oleuropein did not change the rate of glycolysis when incorporated into resting cells of L. plantarum, but it decreased the ATP content of the cells, whereas other studies [208,209,210,211] have reported a considerable antimicrobial activity and sanitizing effects of oleuropein.

4. Conclusions

Olive leaf extracts give a great and useful opportunity for the discovery of new treatment methods due to their high antioxidant, anti-inflammatory, anticancer, anticholesterolemic, antihypertensive, antiviral, and antimicrobial activities. These properties above are mainly associated with the phenolic constituents of olives, olive oil, and herein in olive leaf extracts, such as oleuropein and its hydrolyzed derivatives elenolic acid, and hydroxytyrosol. Therefore, the food and beverage industry should consider olive leaf extracts as an excellent ingredient in the development of new fortified beverages that strengthen the immune system and longevity. In this context, more extensive research studies are required in the near future to optimize the extraction methods of oleuropein and its hydrolyzed derivatives and convey more exhaustive clinical trials to fix and standardize the effective concentration of these polyphenols against chronic or other pathophysiological disorders.

Author Contributions

Conceptualization, I.K.K. and A.A.P.; methodology, I.K.K. and A.A.P.; software, I.K.K. and A.A.P.; validation, I.K.K.; formal analysis, A.A.P. and I.K.K.; investigation, A.A.P. and I.K.K.; resources, I.K.K.; data curation, A.A.P. and I.K.K.; writing—original draft preparation, A.A.P.; writing—review and editing, I.K.K.; visualization, A.A.P. and I.K.K.; supervision, I.K.K.; project administration, I.K.K.; funding acquisition, I.K.K. and A.A.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The original contributions presented in this study are included in this article. Further inquiries can be directed to the corresponding author.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

DPP-4Dipeptidyl peptidase-4 inhibitor
EVOOExtra virgin olive oil
OLEOlive leaf extract
iNOSNitric oxide synthase
LDLLow-density lipoprotein
8-iso-PGF2α8-iso Prostaglandin F2α
NrF2Nuclear factor erythroid 2-related factor 2
HaCaT cellsaneuploid immortal keratinocyte cell line
IL-1βInterleukin-1 beta (IL-1β)
NLRP3NLR family pyrin containing NACHT—NAIP neuronal apoptosis inhibitor protein, LRR—“leucine-rich repeat” and PYD—“PYRIN domain”
ROSReactive oxygen species
IL-1Interleukin-1
IL-6Interleukin 6
IL-17Interleukin 17
TNF-aTumor necrosis factor-a
IFN-γInterferon gamma
MCP-1Monocyte chemoattractant proteins
COXCyclooxygenase
MMPMetalloproteinases
NF-kβnuclear factor Kappa β
CRPC-reactive protein endothelial and monocyte adhesion molecules
ICAM-1Intercellular adhesion molecule 1
VCAM-1Vascular cell adhesion protein 1
JNKc-Jun N-terminal kinases
MMP-9Matrix metalloproteinase-9
p38 MAPKp38 mitogen-activated protein kinases
GLUT2Glucose transporter 2
MAPKMitogen-activated protein kinase
DPP-4Dipeptidyl-peptidase 4
GLP1Glucagon-like peptide-1
NADPH oxidaseNicotinamide adenine dinucleotide phosphate oxidase
PPARαProliferator-activated-receptor alpha
PPARγProliferator-activated-receptor gamma
MCF-7Michigan Cancer Foundation-7
HIF1AHypoxia-inducible factor 1-alpha
MIA PaCa-2Pancreatic cancer cell line
HCT116Human colon cancer cell line
HL-60Human leukemia cell line
GBMGlioblastoma multiforme
HDAC2 and HDAC3Histone deacetylase 2 and Histone deacetylase 3
TIMPMetallopeptidase inhibitor 1
PC3Human prostate cancer cell line
GBMGlioblastoma multiforme
VEGF-AVascular endothelial growth factor A
PI3KsPhosphoinositide 3-kinases
AktProtein kinase B (PKB)
mTORMammalian target of rapamycin
HER2Human epidermal growth factor receptor 2
NGFNerve growth factor
HCCHepatocellular carcinoma
VHSVViral hemorrhagic septicemia virus
HSV-1Herpes simplex type 1
HIVHuman immunodeficiency viruses
HBVHepatitis B virus
DHBVDuck hepatitis B virus
IC50Half maximal inhibitory concentration
MICMinimum inhibitory concentration
SARS-CoV-2Severe acute respiratory syndrome coronavirus 2
SOLEStandardized olive leaf extract
TMPRSS2Transmembrane protease, serine 2
BiPSBinding immunoglobulin protein
ACE2Angiotensin-converting enzyme 2
TLRsToll-like receptors
PLProPapain-like protease protein
3CLpro3C-like protease
Nsp12Non-structural protein
ATCCAmerican Type Culture Collection
OESAOlea europea L. var. sativa
OESYOlea europea var. sylvestris
ACEAngiotensin-converting enzyme

References

  1. Li, Q.; Tu, Y.; Zhu, C.; Luo, W.; Huang, W.; Liu, W.; Li, Y. Cholinesterase, β-amyloid aggregation inhibitory and antioxidant capacities of Chinese medicinal plants. Ind. Crop. Prod. 2017, 108, 512–519. [Google Scholar] [CrossRef]
  2. Nollet, L.M.; Gutierrez-Uribe, J.A. Phenolic Compounds in Food: Characterization and Analysis, 1st ed.; CRC Press: Boca Raton, FL, USA, 2018; p. 167. [Google Scholar]
  3. Uritu, C.M.; Mihai, C.T.; Stanciu, G.D.; Dodi, G.; Alexa-Stratulat, T.; Luca, A.; Leon-Constantin, M.M.; Stefanescu, R.; Bild, V.; Melnic, S.; et al. Medicinal plants of the family Lamiaceae in pain therapy: A review. Pain Res. Manag. 2018, 2018, 7801543. [Google Scholar] [CrossRef] [PubMed]
  4. Etkin, N.L. Plants and Indigenous Medicine and Diet: Biobehavioral Approaches eBook; Taylor & Francis: Abingdon, UK, 2019; 336p. [Google Scholar]
  5. Watson, R.R.; Preedy, V.R. Fruits, Vegetables, and Herbs: Bioactive Foods in Health Promotion, 1st ed.; Elsevier Academic Press: Cambridge, MA, USA, 2016. [Google Scholar]
  6. Alavi, M.S.; Fanoudi, S.; Ghasemzadeh Rahbardar, M.; Mehri, S.; Hosseinzadeh, H. An updated review of protective effects of rosemary and its active constituents against natural and chemical toxicities. Phytother. Res. 2021, 35, 1313–1328. [Google Scholar] [CrossRef] [PubMed]
  7. Ghorbani, A.; Esmaeilizadeh, M. Pharmacological properties of Salvia ofcinalis and its components. J. Tradit. Complement. Med. 2017, 7, 433–440. [Google Scholar] [CrossRef] [PubMed]
  8. Mahendran, G.; Rahman, L.-U. Ethnomedicinal, phytochemical and pharmacological updates on Peppermint (Mentha × piperita L.)—A review. Phytother. Res. 2020, 34, 2088–2139. [Google Scholar] [CrossRef]
  9. Teschke, R.; Eickhoff, A. Herbal hepatotoxicity in traditional and modern medicine: Actual key issues and new encouraging steps. Front. Pharmacol. 2015, 6, 72. [Google Scholar] [CrossRef]
  10. Adeeyo, A.O.; Ndou, T.M.; Alabi, M.A.; Mkoyi, H.D.; Enitan, E.M.; Beswa, D.; Makungo, R.; Odiyo, J.O. Structure: Activity and Emerging Applications of Spices and Herbs. Herbs and spices—New Processing Technologies; IntechOpen: London, UK, 2021. [Google Scholar] [CrossRef]
  11. El-Sayed, S.M.; Youssef, A.M. Potential application of herbs and spices and their effects in functional dairy products. Heliyon 2019, 5, e01989. [Google Scholar] [CrossRef]
  12. Asmaey, M.A.; Elsoghiar, A.A.M.; Shaaban, M.; Moharram, A.M.; El-Gaby, M.S.A. Phenolics and Other Structural Compounds from Leaves of Olea europaea L.: Extraction Techniques and Pharmacological Activities. Chem. Afr. 2024, 7, 5123–5148. [Google Scholar] [CrossRef]
  13. Rahmanian, N.; Ali, S.H.B.; Homayoonfard, M.; Ali, N.J.; Rehan, M.M.; Sadef, Y.; Nizami, A.S. Analysis of physiochemical parameters to evaluate the drinking water quality in the state of Perak, Malaysia. J. Chem. 2015, 2015, 716125. [Google Scholar] [CrossRef]
  14. Carbonara, T.; Pascale, R.; Argentieri, M.P.; Papadia, P.; Fanizzi, F.P.; Villanova, L.; Avato, P. Phytochemical analysis of A herbal tea from Artemisia annua L. J. Pharm. Biomed. Anal. 2012, 62, 79–86. [Google Scholar] [CrossRef]
  15. Cahyawati, P.N.; Lestari, A.; Subrata, T.; Dewi, N.W.E.S.; Wiadnyana, I.G.P. Phytochemical test on herbal drinks loloh cemcem at Penglipuran Village, Bali. J. Phys. Conf. Ser. 2019, 1402, 055030. [Google Scholar] [CrossRef]
  16. Quirantes-Piné, R.; Lozano-Sánchez, J.; Herrero, M.; Ibáñez, E.; Segura-Carretero, A.; Fernández Gutiérrez, A. HPLC-ESI-QTOF-MS as a powerful analytical tool for characterising phenolic compounds in olive-leaf extracts. Phytochem. Anal. 2013, 24, 213–223. [Google Scholar] [CrossRef] [PubMed]
  17. Saad, J.M.; Prochaska, J.O. A philosophy of health: Life as reality, health as a universal value. Palgrave Commun. 2020, 6, 45. [Google Scholar] [CrossRef] [PubMed]
  18. Jamshidi-Kia, F.; Lorigooini, Z.; Amini-Khoei, H. Medicinal plants: Past history and future perspective. J. Herbmed Pharmacol. 2018, 7, 1–7. [Google Scholar] [CrossRef]
  19. Rashid, N.; Gbedomon, R.C.; Ahmad, M.; Salako, V.K.; Zafar, M.; Malik, K. Traditional knowledge on herbal drinks among indigenous communities in Azad Jammu and Kashmir, Pakistan. J. Ethnobiol. Ethnomed. 2018, 14, 16. [Google Scholar] [CrossRef]
  20. Ob’on, C.; Rivera, D.; Fonoll’a, E.; Alcaraz, F.; Attieh, L. A comparison study on traditional mixtures of herbal teas used in eastern mediterranean area. Front. Pharmacol. 2021, 12, 632692. [Google Scholar] [CrossRef]
  21. Martin. Top 10 Tea Drinking Countries in the World. 2019. Available online: https://www.storiesabouttea.com/top-10-tea-drinking-countries-in-the-world/ (accessed on 11 May 2022).
  22. Hara, Y. Tea catechins and their applications as supplements and pharmaceutics. Pharmacol. Res. 2011, 64, 100–104. [Google Scholar] [CrossRef]
  23. Long, T.; Hu, R.; Cheng, Z.; Xu, C.; Hu, Q.; Liu, Q.; Gu, R.; Huang, Y.; Long, C. Ethnobotanical study on herbal tea drinks in Guangxi, China. J. Ethnobiol. Ethnomed. 2023, 19, 10. [Google Scholar] [CrossRef]
  24. World Health Organization. Obesity and Overweight. 2021. Available online: https://www.who.int/news-room/fact-sheets/detail/obesity-and-overweight (accessed on 1 November 2024).
  25. Cercato, L.M.; White, P.A.S.; Nampo, F.K.; Santos, M.R.V.; Camargo, E.A. A systematic review of medicinal plants used for weight loss in Brazil: Is there potential for obesity treatment? J. Ethnopharmacol. 2015, 176, 286–296. [Google Scholar] [CrossRef]
  26. Food and Agriculture Organization of the United Nations (FAO/UN). Current Market Situation and Medium-Term Outlook. In Proceedings of the Twenty-Third Session of the Intergovernmental Group on Tea, Hangzhou, China, 17–20 May 2018; pp. 13–16. Available online: http://www.fao.org/3/a-i4480e.pdf (accessed on 1 September 2024).
  27. Yang, Y.; Zhang, Z.; Li, S.; Ye, X.; Li, X.; He, K. Synergy effects of herb extracts: Pharmacokinetics and pharmacodynamic basis. Fitoterapia 2014, 92, 133–147. [Google Scholar] [CrossRef]
  28. Carbone, R.; Caracciolo, F.; Di Vita, G.; D’Amico, M.; Spina, D. Consumer Trends in the Herbal Tea Market: A Systematic Literature Review. Food Rev. Int. 2025, 1–19. [Google Scholar] [CrossRef]
  29. Cavalheiro, C.V.; Picoloto, R.S.; Cichoski, A.J.; Wagner, R.; de Menezes, C.R.; Zepka, L.Q.; Da Croce, D.M.; Barin, J.S. Olive leaves offer more than phenolic compounds—Fatty acids and mineral composition of varieties from Southern Brazil. Ind. Crop. Prod. 2015, 71, 122–127. [Google Scholar] [CrossRef]
  30. Kabbash, E.M.; Ayoub, I.M.; Gad, H.A.; Abdel-Shakour, Z.T.; El-Ahmady, S.H. Quality assessment of leaf extracts of 12 olive cultivars and impact of seasonal variation based on UV spectroscopy and phytochemcial content using multivariate analyses. Phytochem. Anal. 2021, 32, 932–941. [Google Scholar] [CrossRef] [PubMed]
  31. Tarchoune, I.; Sgherri, C.; Eddouzi, J.; Zinnai, A.; Quartacci, M.F.; Zarrouk, M. Olive leaf addition increases olive oil nutraceutical properties. Molecules 2019, 24, 545. [Google Scholar] [CrossRef]
  32. Irakli, M.; Chatzopoulou, P.; Ekateriniadou, L. Optimization of ultrasound-assisted extraction of phenolic compounds: Oleuropein, phenolic acids, phenolic alcohols and flavonoids from olive leaves and evaluation of its antioxidant activities. Ind. Crop. Prod. 2018, 124, 382–388. [Google Scholar] [CrossRef]
  33. Sifaoui, I.; López-Arencibia, A.; Martín-Navarro, C.M.; Chammem, N.; Reyes-Batlle, M.; Mejri, M.; Lorenzo-Morales, J.; Abderabba, M.; Piñero, J.E. Activity of olive leaf extracts against the promastigote stage of Leishmania species and their correlation with the antioxidant activity. Exp. Parasitol. 2014, 141, 106–111. [Google Scholar] [CrossRef]
  34. Salem, M.; Affes, H.; Ksouda, K.; Sahnoun, Z.; Zeghal, K.; Hammami, S. Pharmacological activities of Olea europaea leaves. J. Food Process. Preserv. 2014, 39, 3128–3136. [Google Scholar] [CrossRef]
  35. Rahmanian, N.; Jafari, S.; Wani, T. A comprehensive review on the bioactive profile, dehydration, extraction and application of the bioactive components of olive leaves. Trends Food Sci. Technol. 2015, 42, 150–172. [Google Scholar] [CrossRef]
  36. Boeing, J.S.; Barizão, É.O.; E Silva, B.C.; Montanher, P.F.; de Cinque Almeida, V.; Visentainer, J.V. Evaluation of solvent effect on the extraction of phenolic compounds and antioxidant capacities from the berries: Application of principal component analysis. Chem. Cent. J. 2014, 8, 48. [Google Scholar] [CrossRef]
  37. Ferreira, D.M.; de Oliveira, N.M.; Chéu, M.H.; Meireles, D.; Lopes, L.; Oliveira, M.B.; Machado, J. Updated Organic Composition and Potential Therapeutic Properties of Different Varieties of Olive Leaves from Olea europaea. Plants 2023, 12, 688. [Google Scholar] [CrossRef]
  38. Bouaziz, M.; Sayadi, S. Isolation and evaluation of antioxidants from leaves of a Tunisian cultivar olive tree. Eur. J. Lipid Sci. Technol. 2005, 107, 497–504. [Google Scholar] [CrossRef]
  39. Japón-Luján, R.; Luque de Castro, M.D. Static–dynamic superheated liquid extraction of hydroxytyrosol and other biophenols from alperujo (a semisolid residue of the olive oil industry). J. Agric. Food Chem. 2007, 55, 3629–3634. [Google Scholar] [CrossRef] [PubMed]
  40. Žugčić, T.; Abdelkebir, R.; Baena, C.; Collado, M.C.; García-Pérez, J.; Meléndez-Martínez, A.J.; Režek Jambrak, A.; Lorenzo, J.M.; Barba, F.J. From extraction of valuable compounds to health promoting benefits of olive leaves through bioaccessibility, bioavailability and impact on gut microbiota. Trends Food Sci. Technol. 2018, 83, 63–77. [Google Scholar] [CrossRef]
  41. Talhaoui, N.; Taamalli, A.; Gómez-Caravaca, A.M.; Fernández-Gutiérrez, A.; Segura-Carretero, A. Phenolic compounds in olive leaves: Analytical determination, biotic and abiotic influence, and health benefits. Food Res. Int. 2015, 77, 92–108. [Google Scholar] [CrossRef]
  42. Hassen, I.; Casabianca, H.; Hosni, K. Biological activities of the natural antioxidant oleuropein: Exceeding the expectation—A mini-review. J. Funct. Foods 2015, 18, 926–940. [Google Scholar] [CrossRef]
  43. Benavente-Garcıa, O.; Castillo, J.; Lorente, J.; Ortuño, A.; Del Rio, J. Antioxidant activity of phenolics extracted from Olea europaea L. leaves. Food Chem. 2000, 68, 457–462. [Google Scholar] [CrossRef]
  44. Poudyal, H.; Campbell, F.; Brown, L. Olive leaf extract attenuates cardiac, hepatic, and metabolic changes in high carbohydrate–, high fat–fed rats. J. Nutr. 2010, 140, 946–953. [Google Scholar] [CrossRef]
  45. Samet, I.; Villareal, M.O.; Motojima, H.; Han, J.; Sayadi, S.; Isoda, H. Olive leaf components apigenin 7-glucoside and luteolin 7-glucoside direct human hematopoietic stem cell differentiation towards erythroid lineage. Differentiation 2015, 89, 146–155. [Google Scholar] [CrossRef]
  46. Rostamzadeh, A.; Amini-Khoei, H.; Mardani Korani, M.J.; Rahimi-Madiseh, M. Comparison effects of olive leaf extract and oleuropein compounds on male reproductive function in cyclophosphamide exposed mice. Heliyon 2020, 6, e03785. [Google Scholar] [CrossRef]
  47. Nunes, M.; Pimentel, F.; Costa, A.; Alves, R.; Oliveira, M. Olive by-products for functional and food applications: Challenging opportunities to face environmental constraints. Innov. Food Sci. Emerg. Technol. 2016, 35, 139–148. [Google Scholar] [CrossRef]
  48. El, S.N.; Karakaya, S. Olive tree (Olea europaea) leaves: Potential beneficial effects on human health. Nutr. Rev. 2009, 67, 632–638. [Google Scholar] [CrossRef] [PubMed]
  49. Lockyer, S.; Yaqoob, P.; Spencer, J.; Rowland, I. Olive leaf phenolics and cardiovascular risk reduction: Physiological effects and mechanisms of action. Nutr. Aging 2012, 1, 125–140. [Google Scholar] [CrossRef]
  50. De Leonardis, A.; Macciola, V.; Cuomo, F.; Lopez, F. Evidence of oleuropein degradation by olive leaf protein extract. Food Chem. 2015, 175, 568–574. [Google Scholar] [CrossRef]
  51. Guinda Garín, M.Á.; Castellano, J.M.; Sántos-Lozano, J.M.; Delgado Hervás, T.; Gutiérrez-Adánez, P.; Rada, M. Determination of Major Bioactive Compounds from Olive Leaf. J. Food Sci. Technol. 2015, 64, 431–438. [Google Scholar] [CrossRef]
  52. Karakaya, S. Studies of olive tree leaf extract indicate seveal potential health benefits. Nutr. Rev. 2009, 67, 632–639. [Google Scholar]
  53. Dekanski, D.; Ristic, S.; Mitrovic, D. Antioxidant effect of dry olive (Olea europaea L.) leaf extract on ethanol-induced gastric lesions in rats. Mediterr. J. Nutr. Metab. 2009, 2, 205–211. [Google Scholar] [CrossRef]
  54. Carito, V.; Venditti, A.; Bianco, A.; Ceccanti, M.; Serrilli, A.M.; Chaldakov, G.; Tarani, L.; De Nicolò, S.; Fiore, M. Effects of olive leaf polyphenols on male mouse brain NGF, BDNF and their receptors TrkA, TrkB and p75. Nat. Prod. Res. 2014, 28, 1970–1984. [Google Scholar] [CrossRef]
  55. Visioli, F.; Galli, C.; Galli, G.; Caruso, D. Biological activities and metabolic fate of olive oil phenols. Eur. J. Lipid Sci. Technol. 2002, 104, 677–684. [Google Scholar] [CrossRef]
  56. Visioli, F.; Bogani, P.; Galli, C. Healthful properties of olive oil minor components. In Olive Oil, Chemistry and Technology; Boskou, D., Ed.; AOCS Press: Champaign, IL, USA, 2006; pp. 173–190. [Google Scholar] [CrossRef]
  57. De la Puerta, R.; Dominguez, M.E.M.; Ruiz-Guttierrez, V.; Flavill, J.A.; Hoult, J.R.S. Effects of olive oil phenolics on scavenging of reactive nitrogen species and upon nitrergic neurotransmission. Life Sci. 2001, 69, 1213–1222. [Google Scholar] [CrossRef]
  58. Coni, E.; Benedetto, R.; Pasquale, M.; Masella, R.; Modesti, D.; Mattei, R.; Carline, E.A. Protective effect of oleuropein, an olive oil biophenol, on low density lipoprotein oxidizability in rabbits. Lipids 2000, 35, 45–54. [Google Scholar] [CrossRef]
  59. Manna, C.; Migliardi, V.; Golino, P.; Scognamiglio, A.; Galletti, P.; Chiariello, M.; Zappia, V. Oleuropein prevents oxidative myocardial injury induced by ischemia and reperfusion. J. Nutr. Biochem. 2004, 15, 461–466. [Google Scholar] [CrossRef]
  60. De la Puerta, R.; Ruiz Gutierrez, V.; Hoult, J.R. Inhibition of leukocyte 5-lipoxygenase by phenolics from virgin olive oil. Biochem. Pharmacol. 1999, 57, 445–449. [Google Scholar] [CrossRef]
  61. Visioli, F.; Caruso, D.; Galli, C.; Viappiani, S.; Galli, G.; Sala, A. Olive Oils Rich in Natural Catecholic Phenols Decrease Isoprostane Excretion in Humans. Biochem. Biophys. Res. Commun. 2000, 278, 797–799. [Google Scholar] [CrossRef]
  62. Parzonko, A.; Czerwińska, M.E.; Kiss, A.K.; Naruszewicz, M. Oleuropein and oleacein may restore biological functions of endothelial progenitor cells impaired by angiotensin II via activation of Nrf2/heme oxygenase-1 pathway. Phytomedicine 2013, 20, 1088–1094. [Google Scholar] [CrossRef]
  63. Al-Azzawie, H.F.; Alhamdani, M.-S.S. Hypoglycemic and antioxidant effect of oleuropein in alloxan-diabetic rabbits. Life Sci. 2006, 78, 1371–1377. [Google Scholar] [CrossRef]
  64. Jemai, H.; El Feki, A.; Sayadi, S. Antidiabetic and Antioxidant Effects of Hydroxytyrosol and Oleuropein from Olive Leaves in Alloxan-Diabetic Rats. J. Agric. Food Chem. 2009, 57, 8798–8804. [Google Scholar] [CrossRef]
  65. Kotyzová, D.; Hodková, A.; Eybl, V. The effect of olive oil phenolics—Hydroxytyrosol and oleuropein on antioxidant defence status in acute arsenic exposed rats. Toxicol. Lett. 2011, 205, S222. [Google Scholar] [CrossRef]
  66. Li, H.; Deng, N.; Yang, J.; Zhao, Y.; Jin, X.; Cai, A.; Seeram, N.P.; Ma, H.; Li, D.; Yang, H.; et al. Anti-inflammatory and antioxidant properties of oleuropein in human keratinocytes characterized by bottom-up proteomics. Front. Pharmacol. 2025, 15, 1496078. [Google Scholar] [CrossRef]
  67. Dessì, M.; Noce, A.; Agnoli, A.; De Angelis, S.; Fuiano, L.; Tozzo, C.; Taccone-Gallucci, M.; Fuiano, G.; Federici, G. The usefulness of the prognostic inflammatory and nutritional index (PINI) in a haemodialysis population. Nutr. Metab. Cardiovasc. Dis. 2009, 19, 811–815. [Google Scholar] [CrossRef]
  68. Castejon, M.L.; Sánchez-Hidalgo, M.; Aparicio-Soto, M.; Montoya, T.; Martín-LaCave, I.; Fernández-Bolaños, J.G.; Alarcón-de-la-Lastra, C. Dietary oleuropein and its new acyl-derivate attenuate murine lupus nephritis through HO-1/Nrf2 activation and suppressing JAK/STAT, NF-κB, MAPK and NLRP3 inflammasome signaling pathways. J. Nutr. Biochem. 2019, 74, 108229. [Google Scholar] [CrossRef]
  69. Jacob, K.D.; Noren Hooten, N.; Trzeciak, A.R.; Evans, M.K. Markers of oxidant stress that are clinically relevant in aging and age-related disease. Mech. Ageing Dev. 2013, 134, 139–157. [Google Scholar] [CrossRef] [PubMed]
  70. Bandeen-Roche, K.; Walston, J.D.; Huang, Y.; Semba, R.D.; Ferrucci, L. Measuring Systemic Inflammatory Regulation in Older Adults: Evidence and Utility. Rejuvenat. Res. 2009, 12, 403–410. [Google Scholar] [CrossRef] [PubMed]
  71. Barzilay, J.I.; Blaum, C.; Moore, T.; Xue, Q.L.; Hirsch, C.H.; Walston, J.D.; Fried, L.P. Insulin Resistance and Inflammation as Precursors of Frailty: The Cardiovascular Health Study. Arch. Intern. Med. 2007, 167, 635–641. [Google Scholar] [CrossRef] [PubMed]
  72. Collerton, J.; Martin-Ruiz, C.; Davies, K.; Hilkens, C.M.; Isaacs, J.; Kolenda, C.; Parker, C.; Dunn, M.; Catt, M.; Jagger, C.; et al. Frailty and the role of inflammation, immunosenescence and cellular ageing in the very old: Cross-sectional findings from the Newcastle 85+ Study. Mech. Ageing Dev. 2012, 133, 456–466. [Google Scholar] [CrossRef]
  73. Fried, L.P.; Xue, Q.-L.; Cappola, A.R.; Ferrucci, L.; Chaves, P.; Varadhan, R.; Guralnik, J.M.; Leng, S.X.; Semba, R.D.; Walston, J.D.; et al. Nonlinear Multisystem Physiological Dysregulation Associated With Frailty in Older Women: Implications for Etiology and Treatment. J. Gerontol. Ser. A Biol. Sci. Med. Sci. 2009, 64, 1049–1057. [Google Scholar] [CrossRef]
  74. Estruch, R.; Martínez-González, M.A.; Corella, D.; Salas-Salvadó, J.; Ruiz-Gutiérrez, V.; Covas, M.I.; Fiol, M.; Gómez-Gracia, E.; López-Sabater, M.C.; Vinyoles, E.; et al. Effects of a Mediterranean-Style Diet on Cardiovascular Risk Factors: A Randomized Trial. Ann. Intern. Med. 2006, 145, 1–11. [Google Scholar] [CrossRef]
  75. Miles, E.A.; Zoubouli, P.; Calder, P.C. Differential anti-inflammatory effects of phenolic compounds from extra virgin olive oil identified in human whole blood cultures. Nutrition 2005, 21, 389–394. [Google Scholar] [CrossRef]
  76. Ryu, S.J.; Choi, H.S.; Yoon, K.Y.; Lee, O.H.; Kim, K.J.; Lee, B.Y. Oleuropein Suppresses LPS-Induced Inflammatory Responses in RAW 264.7 Cell and Zebrafish. J. Agric. Food Chem. 2015, 63, 2098–2105. [Google Scholar] [CrossRef]
  77. Giner, E.; Recio, M.C.; Ríos, J.L.; Giner, R.M. Oleuropein Protects against Dextran Sodium Sulfate-Induced Chronic Colitis in Mice. J. Nat. Prod. 2013, 76, 1113–1120. [Google Scholar] [CrossRef]
  78. Visioli, F.; Bellomo, G.; Galli, C. Free Radical-Scavenging Properties of Olive Oil Polyphenols. Biochem. Biophys. Res. Commun. 1998, 247, 60–64. [Google Scholar] [CrossRef]
  79. Impellizzeri, D.; Esposito, E.; Mazzon, E.; Paterniti, I.; Di Paola, R.; Bramanti, P.; Morittu, V.M.; Procopio, A.; Britti, D.; Cuzzocrea, S. The effects of oleuropein aglycone, an olive oil compound, in a mouse model of carrageenan-induced pleurisy. Clin. Nutr. 2011, 30, 533–540. [Google Scholar] [CrossRef] [PubMed]
  80. Lee, B.; Shim, I.; Lee, H.; Hahm, D.-H. Effect of oleuropein on cognitive deficits and changes in hippocampal brain-derived neurotrophic factor and cytokine expression in a rat model of post-traumatic stress disorder. J. Nat. Med. 2018, 72, 44–56. [Google Scholar] [CrossRef] [PubMed]
  81. Khalatbary, A.; Zarrinjoei, G. Anti-Inflammatory Effect of Oleuropein in Experimental Rat Spinal Cord Trauma. Iran. Red. Crescent Med. J. 2012, 14, 229–234. [Google Scholar] [PubMed]
  82. 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. 2006, 25, 859–868. [Google Scholar] [CrossRef]
  83. 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]
  84. WHO. Classification of Diabetes Mellitus 2019; WHO: Geneva, Switzerland, 2019.
  85. Yaribeygi, H.; Farrokhi, F.R.; Butler, A.E.; Sahebkar, A. Insulin resistance: Review of the underlying molecular mechanisms. J. Cell. Physiol. 2019, 234, 8152–8161. [Google Scholar] [CrossRef]
  86. Yaribeygi, H.; Butler, A.E.; Barreto, G.E.; Sahebkar, A. Antioxidative potential of antidiabetic agents: A possible protective mechanism against vascular complications in diabetic patients. J. Cell. Physiol. 2019, 234, 2436–2446. [Google Scholar] [CrossRef]
  87. Yaribeygi, H.; Mohammadi, M.T.; Sahebkar, A. Crocin potentiates antioxidant defense system and improves oxidative damage in liver tissue in diabetic rats. Biomed. Pharmacother. 2018, 98, 333–337. [Google Scholar] [CrossRef]
  88. Yaribeygi, H.; Mohammadi, M.T.; Sahebkar, A. PPAR-α agonist improves hyperglycemia-induced oxidative stress in pancreatic cells by potentiating antioxidant defense system. Drug Res. 2018, 68, 355–360. [Google Scholar] [CrossRef]
  89. Hurrle, S.; Hsu, W.H. The etiology of oxidative stress in insulin resistance. Biomed. J. 2017, 40, 257–262. [Google Scholar] [CrossRef]
  90. Asgary, S.; Naderi, G.A.; Sarraf Zadegan, N.; Vakili, R. The inhibitory effects of pure flavonoids on in vitro protein glycosylation. J. Herb. Pharmacother. 2002, 2, 47–55. [Google Scholar] [CrossRef] [PubMed]
  91. Visioli, F.; Poli, A.; Galli, C. Antioxidant and other biological activities of phenols from olives and olive oil. Med. Res. Rev. 2002, 22, 65–75. [Google Scholar] [CrossRef] [PubMed]
  92. Diaz, A.M.; Abad, M.J.; Fernandez, L.; Recuero, C.; Villaescusa, L.; Silvan, A.M.; Bermejo, P. In vitro anti-inflammatory activity of iridoids and triterpenoid compounds isolated from Phillyrea latifolia L. Biol. Pharm. Bull. 2000, 23, 1307–1313. [Google Scholar] [CrossRef] [PubMed]
  93. Somova, L.I.; Shode, F.O.; Mipando, M. Cardiotonic and antidysrhythmic effects of oleanolic and ursolic acids, methyl maslinate and uvaol. Phytomedicine 2004, 11, 121–129. [Google Scholar] [CrossRef]
  94. Gonzalez, M.; Zarzuelo, A.; Gamez, M.J.; Utrilla, M.P.; Jimenez, J.; Osuna, I. Hypoglycemic activity of olive leaf. Planta Med. 1992, 58, 513–515. [Google Scholar] [CrossRef]
  95. Romani, A.; Mulinacci, N.; Pinelli, P.; Vincieri, F.; Cimato, A. Polyphenolic content in five Tuscany cultivars of Olea europaea L. J. Agric. Food Chem. 1999, 47, 964–967. [Google Scholar] [CrossRef]
  96. Ahmadvand, H.; Noori, A.; Dehnoo, M.G.; Bagheri, S.; Cheraghi, R.A. Hypoglycemic, hypolipidemic and antiatherogenic effects of oleuropein in alloxan-induced Type 1 diabetic rats. Asian Pac. J. Trop. Dis. 2014, 4, S421–S425. [Google Scholar] [CrossRef]
  97. Fujiwara, Y.; Tsukahara, C.; Ikeda, N.; Sone, Y.; Ishikawa, T.; Ichi, I.; Koike, T.; Aoki, Y. Oleuropein improves insulin resistance in skeletal muscle by promoting the translocation of GLUT4. J. Clin. Biochem. Nutr. 2017, 61, 196–202. [Google Scholar] [CrossRef]
  98. 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]
  99. Wu, L.; Velander, P.; Liu, D.; Xu, B. Olive Component Oleuropein Promotes β-Cell Insulin Secretion and Protects β-Cells from Amylin Amyloid-Induced Cytotoxicity. Biochemistry 2017, 56, 5035–5039. [Google Scholar] [CrossRef]
  100. Chaari, A. Inhibition of human islet amyloid polypeptide aggregation an cellular toxicity by oleuropein and derivatives from olive oil. Int. J. Biol. Macromol. 2020, 162, 284–300. [Google Scholar] [CrossRef] [PubMed]
  101. Zhang, Z.; Zhao, H.; Wang, A. Oleuropein alleviated gestational diabetes mellitus by activating AMPK signalling. Endocr. Connect. 2021, 10, 45–53. [Google Scholar] [CrossRef] [PubMed]
  102. Zheng, S.; Wang, Y.; Fang, J.; Geng, R.; Li, M.; Zhao, Y.; Kang, S.G.; Huang, K.; Tong, T. Oleuropein ameliorates advanced stage of type 2 diabetes in db/db mice by regulating gut microbiota. Nutrients 2021, 13, 2131. [Google Scholar] [CrossRef]
  103. Marcelino, G.; Hiane, P.A.; de Cassia Freitas, K.; Figueiredo Santana, L.; Pott, A.; Rodrigues Donadon, J.; de Cassia Avellaneda Guimares, R. Effects of olive oil and its minor components on cardiovascular diseases, inflammation, and gut microbiota. Nutrients 2019, 11, 1826. [Google Scholar] [CrossRef] [PubMed]
  104. Santangelo, C.; Filesi, C.; Vari, R.; Scazzocchio, B.; Filardi, T.; Fogliano, V.; D’Archivio, M.; Giovannini, C.; Lenzi, A.; Morano, S.; et al. Consumption of extra-virgin oil rich in phenolic compounds improves metabolic control in patients with type 2 diabetes mellitus: A possible involvement of reduced levels of circulating visfatin. J. Endocrinol. Invest. 2016, 39, 1295–1301. [Google Scholar] [CrossRef]
  105. Schwingshackl, L.; Lampousi, A.M.; Portillo, M.P.; Romaguera, D.; Hoffmann, G.; Boeing, H. Olive oil in the prevention and management of type 2 diabetes mellitus: A systematic review and meta-analysis of cohort studies and intervention trials. Nutr. Diabetes 2017, 7, e262. [Google Scholar] [CrossRef]
  106. De Bock, M.; Derraik, J.G.; 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]
  107. Hermans, M.P.; Lempereur, P.; Salembier, J.P.; Maes, N.; Albert, A.; Jansen, O.; Pincemail, J. Supplementation effect of a combination of olive (Olea europea L.) leaf and fruit extracts in the clinical management of hypertension and metabolic syndrome. Antioxidants 2020, 9, 872. [Google Scholar] [CrossRef]
  108. Violi, F.; Loffredo, L.; Pignatelli, P.; Angelico, F.; Bartimoccia, S.; Nocella, C.; Cangemi, R.; Petruccioli, A.; Monticolo, R.; Pastori, D.; et al. Extra virgin olive oil use is associated with improved post-prandial blood glucose and LDL cholesterol in healthy subjects. Nutr. Diabetes 2015, 5, e172. [Google Scholar] [CrossRef]
  109. 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. Pharmacol. 2018, 84, 1566–1574. [Google Scholar] [CrossRef]
  110. Carnevale, R.; Loffredo, L.; Del Ben, M.; Angelico, F.; Nocella, C.; Petruccioli, A.; Bartimoccia, S.; Monticolo, R.; Cava, E.; Violi, F. Extra virgin olive oil improves post-prandial glycemic and lipid profile in patients with impaired fasting glucose. Clin. Nutr. 2017, 36, 782–787. [Google Scholar] [CrossRef] [PubMed]
  111. Del Ben, M.; Nocella, C.; Loffredo, L.; Bartimoccia, S.; Cammisotto, V.; Mancinella, M.; Angelico, F.; Valenti, V.; Cavarretta, E.; Carnevale, R.; et al. Oleuropein-enriched chocolate by extra virgin olive oil blunts hyperglycaemia in diabetic patients: Results from a one-time 2-hour post-prandial cross over study. Clin. Nutr. 2020, 39, 2187–2191. [Google Scholar] [CrossRef]
  112. Malliou, F.; Andreadou, I.; Gonzalez, F.J.; Lazou, A.; Xepapadaki, E.; Vallianou, I.; Lambrinidis, G.; Mikros, E.; Marselos, M.; Skaltsounmis, A.L.; et al. The olive constituent oleuropein, as PPARα agonist, markedly reduces serum triglycerides. J. Nutr. Biochem. 2018, 59, 17–28. [Google Scholar] [CrossRef] [PubMed]
  113. Hadrich, F.; Mahmoudi, A.; Bouallagui, Z.; Feki, I.; Isoda, H.; Feve, B.; Sayadi, S. Evaluation of hypocholesterolemic effect of oleuropein in cholesterol-fed rats. Chem. Biol. Interact. 2016, 252, 54–60. [Google Scholar] [CrossRef] [PubMed]
  114. Manceau, P.; Netien, G.; Jardon, P. Hypoglycemic action of extracts of olive leaves. Comptes Rendues Soc. Biol. 1942, 136, 810–811. [Google Scholar]
  115. Capretti, G.; Bonaconza, E. Effects of infusions or decoctions of olive leaves, (O. europaea) on some physical constants of blood and components of metabolism. G. Clin. Med. 1949, 30, 630–642. [Google Scholar]
  116. Ribeiro, R.A.; Fiuza de Melo, M.M.; De Barros, F.; Gomes, C.; Trolin, G. Acute antihypertensive effect in conscious rats produced by some medicinal plants used in the state of Sao Paulo. J. Ethnopharmacol. 1986, 15, 261–269. [Google Scholar] [CrossRef]
  117. Zarzuelo, A.; Duarte, J.; Jimenez, M.; Utrilla, P. Vasodilator effect of olive leaf. Planta Med. 1991, 57, 417–419. [Google Scholar] [CrossRef]
  118. Fehri, B.; Aiache, J.M.; Memmi, A.; Korbi, S.; Yacoubi, M.T.; Mrad, S.; Lamaison, J.L. Hypotension, hypoglycemia and hypouricemia recorded after repeated administration of aqueous leaf extract of O. europaea L. J. Pharm. Belg. 1994, 49, 101–108. [Google Scholar]
  119. Huang, Y.; Guan, Q.; Zhang, Z.; Wang, P.; Li, C. Oleacein: A comprehensive review of its extraction, purification, absorption, metabolism, and health effects. Food Chem. 2024, 433, 137334. [Google Scholar] [CrossRef]
  120. Cherif, S.; Rahal, N.; Haouala, M.; Hizaoui, B.; Dargouth, F.; Gueddiche, M.; Kallel, Z.; Balansard, G.; Boukef, K. A clinical trial of a titrated Olea extract in the treatment of essential arterial hypertension. J. Pharm. Belg. 1996, 51, 69–71. [Google Scholar] [PubMed]
  121. Somova, L.I.; Shode, F.O.; Ramnanan, P.; Nadar, A. Antihypertensive, antiatherosclerotic and antioxidant activity of triterpenoids isolated from Olea europaea, subspecies africana leaves. J. Ethnopharmacol. 2003, 84, 299–305. [Google Scholar] [CrossRef] [PubMed]
  122. Franco-Ávila, T.; Moreno-González, R.; Juan, M.E.; Planas, J.M. Table olive elicits antihypertensive activity in spontaneously hypertensive rats. J. Sci. Food Agric. 2023, 103, 64–72. [Google Scholar] [CrossRef] [PubMed]
  123. Romero, M.; Toral, M.; Gómez-Guzmán, M.; Jiménez, R.; Galindo, P.; Sánchez, M.; Olivares, M.; Gálvez, J.; Duarte, J. Antihypertensive effects of oleuropein-enriched olive leaf extract in spontaneously hypertensive rats. Food Funct. 2016, 7, 584–593. [Google Scholar] [CrossRef]
  124. Ruano, J.; Lopez-Miranda, J.; Fuentes, F.; Moreno, J.A.; Bellido, C.; Perez-Martinez, P.; Lozano, A.; Gómez, P.; Jiménez, Y.; Pérez Jiménez, F. Phenolic content of virgin olive oil improves ischemic reactive hyperemia in hypercholesterolemic patients. J. Am. Coll. Cardiol. 2005, 46, 1864–1868. [Google Scholar] [CrossRef]
  125. Valls, R.M.; Farràs, M.; Suárez, M.; Fernández-Castillejo, S.; Fitó, M.; Konstantinidou, V.; Fuentes, F.; López-Miranda, J.; Giralt, M.; Covas, M.I.; et al. Effects of functional olive oil enriched with its own phenolic compounds on endothelial function in hypertensive patients. A randomised controlled trial. Food Chem. 2015, 167, 30–35. [Google Scholar] [CrossRef]
  126. 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]
  127. Moreno-Luna, R.; Muñoz-Hernandez, R.; Miranda, M.L.; Costa, A.F.; Jimenez-Jimenez, L.; Vallejo-Vaz, A.J.; Muriana, F.J.; 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]
  128. Fitó, M.; Cladellas, M.; De la Torre, R.; Marti, J.; Alcantara, M.; Pujadas-Bastardes, M.; Marrugat, J.; Bruguera, J.; López-Sabater, M.C.; Vila, J.; et al. The members of the SOLOS Investigators. 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]
  129. Hohmann, C.D.; Cramer, H.; Michalsen, A.; Kessler, C.; Steckhan, N.; Choi, K.; Dobos, G. Effects of high phenolic olive oil on cardiovascular risk factors: A systematic review and meta-analysis. Phytomedicine 2015, 22, 631–640. [Google Scholar] [CrossRef]
  130. Somova, L.O.; Nadar, A.; Rammanan, P.; Shode, F.O. Cardiovascular, antihyperlipidemic and antioxidant effects of oleanolic and ursolic acids in experimental hypertension. Phytomedicine 2003, 10, 115–121. [Google Scholar] [CrossRef] [PubMed]
  131. Valero-Muñoz, M.; Martín-Fernández, B.; Ballesteros, S.; de la Fuente, E.; Quintela, J.C.; Lahera, V.; de las Heras, N. Protective effect of a pomace olive oil concentrated in triterpenic acids in alterations related to hypertension in rats: Mechanisms involved. Mol. Nutr. Food Res. 2014, 58, 376–383. [Google Scholar] [CrossRef] [PubMed]
  132. Sanchez-Rodriguez, E.; Lima-Cabello, E.; Biel-Glesson, S.; Fernandez-Navarro, J.R.; Calleja, M.A.; Roca, M.; Espejo-Calvo, J.A.; Gil-Extremera, B.; Soria-Florido, M.; de la Torre, R.; et al. Effects of Virgin Olive Oils Differing in Their Bioactive Compound Contents on Metabolic Syndrome and Endothelial Functional Risk Biomarkers in Healthy Adults: A Randomized Double-Blind Controlled Trial. Nutrients 2018, 10, 626. [Google Scholar] [CrossRef] [PubMed]
  133. Khayyal, M.T.; El-Ghazaly, M.A.; Abdallah, D.M.; Nassar, N.N.; Okpanyi, S.N.; Kreuter, M.H. Blood pressure lowering effect of an olive leaf extract (Olea europaea) in l-name induced hypertension in rats. Arzneim. Forsch./Drug Res. 2002, 52, 797–802. [Google Scholar] [CrossRef]
  134. Scheffler, A.; Rauwald, H.W.; Kampa, B.; Mann, U.; Mohr, F.W.; Dhein, S. Olea europaea leaf extract exerts L-type Ca2+ channel antagonistic effects. J. Ethnopharmacol. 2008, 120, 233–240. [Google Scholar] [CrossRef]
  135. Susalit, E.; Agus, N.; Effendi, I.; Tjandrawinata, R.R.; Nofiarny, D.; Perrinjaquet-Moccetti, T.; Verbruggen, M. Olive (Olea europaea) leaf extract effective in patients with stage-1 hypertension: Comparison with Captopril. Phytomedicine 2011, 18, 251–258. [Google Scholar] [CrossRef]
  136. Miloradović, Z.; Gvozdenov, M.; Jovović, Đ.; Mihailović-Stanojević, N.; Ivanov, M.; Vajić, U.; Karanović, D.; Milanović, S.D.; Grujić-Milanović, J. Uticaj ekstrakta lista masline (Olea europea L.) na hemodinamski status i nivo lipidne peroksidacije kod pacova sa urođenom hipertenzijom/Effect of Olea europea L. leaf extract on haemodynamic status and lipid peroxidation in spontaneously hypertensive rats. Vet. Glas. 2013, 67, 303–315. [Google Scholar] [CrossRef]
  137. Perrinjaquet-Moccetti, T.; Busjahn, A.; Schmidlin, C.; Schmidt, A.; Bradl, B.; Aydogan, C. Food supplementation with an olive (Olea europaea L.) leaf extract reduces blood pressure in borderline hypertensive monozygotic twins. Phytother. Res. 2008, 22, 1239–1242. [Google Scholar] [CrossRef]
  138. 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]
  139. Javadi, H.; Yaghoobzadeh, H.; Esfahani, Z.; Memarzadeh, M.R.; Mirhashemi, S.M. Effects of olive leaf extract on metabolic response, liver and kidney functions and inflammatory biomarkers in hypertensive patients. Pak. J. Biol. Sci. 2019, 22, 342–348. [Google Scholar] [CrossRef]
  140. Nekooeian, A.; Khalili, A.; Khosravi, M. Oleuropein offers cardioprotection in rats with simultaneous type 2 diabetes and renal hypertension. Indian J. Pharmacol. 2014, 46, 398–403. [Google Scholar] [CrossRef] [PubMed]
  141. Rodriguez-Rodriguez, R.; Herrera, M.D.; De Sotomayor, M.A.; Ruiz-Gutierrez, V. Effects of pomace olive oil-enriched diets on endothelial function of small mesenteric arteries from spontaneously hypertensive rats. Br. J. Nutr. 2009, 102, 1435–1444. [Google Scholar] [CrossRef] [PubMed]
  142. Nekooeian, A.A.; Khalili, A.; Khosravi, M.B. Effects of oleuropein in rats with simultaneous type 2 diabetes and renal hypertension: A study of antihypertensive mechanisms. J. Asian Nat. Prod. Res. 2014, 16, 953–962. [Google Scholar] [CrossRef] [PubMed]
  143. Lucas, M.L.; Carraro, C.C.; Belló-Klein, A.; Kalil, A.N.; Aerts, N. Oxidative stress in carotid arteries of patients submitted to carotid endarterectomy. The role of aging process. Acta Cir. Bras. 2016, 31, 564–568. [Google Scholar] [CrossRef]
  144. Vaziri, N.D.; Ni, Z.; Oveisi, F.; Liang, K.; Pandian, R. Enhanced nitric oxide inactivation and protein nitration by reactive oxygen species in renal insufficiency. Hypertension 2002, 39, 135–141. [Google Scholar] [CrossRef]
  145. Mihailovic-Stanojevic, N.; Miloradovic, Z.; Ivanov, M.; Bugarski, B.; Jovovic, D.; Karanovic, D.; Vajić, U.J.; Komes, D.; Grujić-Milanović, J. Upregulation of heme oxygenase-1 in response to wild thyme treatment protects against hypertension and oxidative stress. Oxid. Med. Cell Longev. 2016, 2016, 1458793. [Google Scholar] [CrossRef]
  146. Mihailovic-Stanojevic, N.; Savikin, K.; Zivkovic, J.; Zdunic, G.; Miloradovic, Z.; Ivanov, M.; Karanovic, D.; Vajic, U.J.; Jovovic, D.; Grujic-Milanovic, J. Moderate consumption of alcohol-free red wine provide more beneficial effects on systemic haemodynamics, lipid profile and oxidative stress in spontaneously hypertensive rats than red wine. J. Funct. Foods 2016, 26, 719–730. [Google Scholar] [CrossRef]
  147. Bouallagui, Z.M.; Han, J.; Isoda, H.; Sayadi, S. Hydroxytyrosol rich extract from olive leaves modulates cell cycle progression in MCF-7 human breast cancer cells. Food Chem. Toxicol. 2011, 49, 179–184. [Google Scholar] [CrossRef]
  148. Han, J.; Talorete, T.P.N.; Yamada, P.; Isoda, H. Anti-proliferative and apoptotic effects of oleuropein and hydroxytyrosol on human breast cancer MCF-7 cells. Cytotechnology 2009, 59, 45–53. [Google Scholar] [CrossRef]
  149. Elamin, M.H.; Daghestani, M.H.; Omer, S.A.; Elobeid, M.A.; Virk, P.; Al-Olayan, E.M.; Hassan, Z.K.; Mohammed, O.B.; Aboussekhra, A. Olive oil oleuropein has anti-breast cancer properties with higher efficiency on ER-negative cells. Food Chem. Toxicol. 2013, 53, 310–316. [Google Scholar] [CrossRef]
  150. Liu, L.; Ahn, K.S.; Shanmugam, M.K.; Wang, H.; Shen, H.; Arfuso, F.; Chinnathambi, A.; Alharbi, S.A.; Chang, Y.; Sethi, G.; et al. Oleuropein induces apoptosis via abrogating NF-κB activation cascade in estrogen receptor–negative breast cancer cells. J. Cell. Biochem. 2019, 120, 4504–4513. [Google Scholar] [CrossRef] [PubMed]
  151. Acquaviva, R.; Di Giacomo, C.; Sorrenti, V.; Galvano, F.; Santangelo, R.; Cardile, V.; Gangia, S.; D’Orazio, N.; Abraham, N.G.; Vanella, L. Antiproliferative effect of oleuropein in prostate cell lines. Int. J. Oncol. 2012, 41, 31–38. [Google Scholar] [CrossRef] [PubMed]
  152. Sepporta, M.V.; Fuccelli, R.; Rosignoli, P.; Ricci, G.; Servili, M.; Fabiani, R. Oleuropein Prevents Azoxymethane-Induced Colon Crypt Dysplasia and Leukocytes DNA Damage in A/J Mice. J. Med. Food 2016, 19, 983–989. [Google Scholar] [CrossRef] [PubMed]
  153. Cárdeno, A.; Sánchez-Hidalgo, M.; Rosillo, M.A.; de la Lastra, C.A. Oleuropein, a Secoiridoid Derived from Olive Tree, Inhibits the Proliferation of Human Colorectal Cancer Cell Through Downregulation of HIF-1α. Nutr. Cancer 2013, 65, 147–156. [Google Scholar] [CrossRef]
  154. Bernini, R.; Carastro, I.; Palmini, G.; Tanini, A.; Zonefrati, R.; Pinelli, P.; Brandi, M.L.; Romani, A. Lipophilization of Hydroxytyrosol-Enriched Fractions from Olea europaea L. Byproducts and Evaluation of the in Vitro Effects on a Model of Colorectal Cancer Cells. J. Agric. Food Chem. 2017, 65, 6506–6512. [Google Scholar] [CrossRef]
  155. Samara, P.; Christoforidou, N.; Lemus, C.; Argyropoulou, A.; Ioannou, K.; Vougogiannopoulou, K.; Aligiannis, N.; Paronis, E.; Gaboriaud-Kolar, N.; Tsitsilonis, O.; et al. New semi-synthetic analogs of oleuropein show improved anticancer activity in vitro and in vivo. Eur. J. Med. Chem. 2017, 137, 11–29. [Google Scholar] [CrossRef]
  156. Yao, J.; Wu, J.; Yang, X.; Yang, J.; Zhang, Y.; Du, L. Oleuropein induced apoptosis in HeLa cells via a mitochondrial apoptotic cascade associated with activation of the c-Jun NH2-terminal kinase. J. Pharmacol. Sci. 2014, 125, 300–311. [Google Scholar] [CrossRef]
  157. Yan, C.M.; Chai, E.Q.; Cai, H.Y.; Miao, G.Y.; Ma, W. Oleuropein induces apoptosis via activation of caspases and suppression of phosphatidylinositol 3-kinase/protein kinase B pathway in HepG2 human hepatoma cell line. Mol. Med. Rep. 2015, 11, 4617–4624. [Google Scholar] [CrossRef]
  158. Seçme, M.; Eroğlu, C.; Dodurga, Y.; Bağcı, G. Investigation of anticancer mechanism of oleuropein via cell cycle and apoptotic pathways in SH-SY5Y neuroblastoma cells. Gene 2016, 585, 93–99. [Google Scholar] [CrossRef]
  159. Zeriouh, W.; Nani, A.; Belarbi, M.; Dumont, A.; de Rosny, C.; Aboura, I.; Ghanemi, F.Z.; Murtaza, B.; Patoli, D.; Thomas, C.; et al. Correction: Phenolic extract from oleaster (Olea europaea var. Sylvestris) leaves reduces colon cancer growth and induces caspase-dependent apoptosis in colon cancer cells via the mitochondrial apoptotic pathway. PLoS ONE 2017, 12, e0176574. [Google Scholar] [CrossRef]
  160. Anter, J.; Fernández-Bedmar, Z.; Villatoro-Pulido, M.; Demyda-Peyras, S.; Moreno-Millán, M.; Alonso-Moraga, Á.; Muñoz-Serrano, A.; Luque de Castro, M.D. A pilot study on the DNA-protective, cytotoxic, and apoptosis-inducing properties of olive-leaf extracts. Mutat. Res. Genet. Toxicol. Environ. Mutagen. 2011, 723, 165–170. [Google Scholar] [CrossRef] [PubMed]
  161. Momtaz, S.; Niaz, K.; Maqbool, F.; Abdollahi, M.; Rastrelli, L.; Nabavi, S.M. STAT3 targeting by polyphenols: Novel therapeutic strategy for melanoma: STAT3 targeting by polyphenols. BioFactors 2017, 43, 347–370. [Google Scholar] [CrossRef] [PubMed]
  162. Marchetti, C.; Clericuzio, M.; Borghesi, B.; Cornara, L.; Ribulla, S.; Gosetti, F.; Marengo, E.; Burlando, B. Oleuropein-Enriched Olive Leaf Extract Affects Calcium Dynamics and Impairs Viability of Malignant Mesothelioma Cells. Evid.-Based Complement. Altern. Med. 2015, 2015, 908493. [Google Scholar] [CrossRef] [PubMed]
  163. Abtin, M.; Alivand, M.R.; Khaniani, M.S.; Bastami, M.; Zaeifizadeh, M.; Derakhshan, S.M. Simultaneous downregulation of miR-21 and miR-155 through oleuropein for breast cancer prevention and therapy. J. Cell. Biochem. 2018, 119, 7151–7165. [Google Scholar] [CrossRef]
  164. Tezcan, G.; Tunca, B.; Bekar, A.; Budak, F.; Sahin, S.; Cecener, G.; Egeli, U.; Taskapılıoglu, M.O.; Kocaeli, H.; Tolunay, S.; et al. Olea europaea leaf extract improves the treatment response of GBM stem cells by modulating miRNA expression. Am. J. Cancer Res. 2014, 4, 572–590. [Google Scholar]
  165. Bayat, S.; Shekari Khaniani, M.; Choupani, J.; Alivand, M.R.; Mansoori Derakhshan, S. HDACis (class I), cancer stem cell, and phytochemicals: Cancer therapy and prevention implications. Biomed. Pharmacother. 2018, 97, 1445–1453. [Google Scholar] [CrossRef]
  166. Song, H.; Lim, D.Y.; Jung, J.I.; Cho, H.J.; Park, S.Y.; Kwon, G.T.; Kang, Y.H.; Lee, K.W.; Choi, M.S.; Park, J.H.Y. Dietary oleuropein inhibits tumor angiogenesis and lymphangiogenesis in the B16F10 melanoma allograft model: A mechanism for the suppression of high-fat diet-induced solid tumor growth and lymph node metastasis. Oncotarget 2017, 8, 32027–32042. [Google Scholar] [CrossRef]
  167. Liu, M.; Wang, J.; Huang, B.; Chen, A.; Li, X. Oleuropein inhibits the proliferation and invasion of glioma cells via suppression of the AKT signaling pathway. Oncol. Rep. 2016, 36, 2009–2016. [Google Scholar] [CrossRef]
  168. Hassan, Z.K.; Elamin, M.H.; Daghestani, M.H.; Omer, S.A.; Al-Olayan, E.M.; Elobeid, M.A.; Virk, P.; Mohammed, O.B. Oleuropein induces anti-metastatic effects in breast cancer. Asian Pac. J. Cancer Prev. 2012, 13, 4555–4559. [Google Scholar] [CrossRef]
  169. Papachristodoulou, A.; Tsoukala, M.; Benaki, D.; Kostidis, S.; Gioti, K.; Aligiannis, N.; Pratsinis, H.; Kletsas, D.; Skaltsounis, A.-L.; Mikros, E.; et al. Oleuropein is a Powerful Sensitizer of Doxorubicin-mediated Killing of Prostate Cancer Cells and Exerts Its Action via Induction of Autophagy. J. Cancer Res. Treat. 2018, 4, 61–68. [Google Scholar] [CrossRef]
  170. 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] [PubMed]
  171. Tezcan, G.; Taskapilioglu, M.O.; Tunca, B.; Bekar, A.; Demirci, H.; Kocaeli, H.; Aksoy, S.A.; Egeli, U.; Cecener, G.; Tolunay, S. Olea europaea leaf extract and bevacizumab synergistically exhibit beneficial efficacy upon human glioblastoma cancer stem cells through reducing angiogenesis and invasion in vitro. Biomed. Pharmacother. 2017, 90, 713–723. [Google Scholar] [CrossRef] [PubMed]
  172. Ryskalin, L.; Gaglione, A.; Limanaqi, F.; Biagioni, F.; Familiari, P.; Frati, A.; Esposito, V.; Fornai, F. The Autophagy Status of Cancer Stem Cells in Gliobastoma Multiforme: From Cancer Promotion to Therapeutic Strategies. IJMS 2019, 20, 3824. [Google Scholar] [CrossRef] [PubMed]
  173. Ruzzolini, J.; Peppicelli, S.; Andreucci, E.; Bianchini, F.; Scardigli, A.; Romani, A.; la Marca, G.; Nediani, C.; Calorini, L. Oleuropein, the Main Polyphenol of Olea europaea Leaf Extract, has an Anti-Cancer Effect on Human BRAF Melanoma Cells and Potentiates the Cytotoxicity of Current Chemotherapies. Nutrients 2018, 10, 1950. [Google Scholar] [CrossRef]
  174. Menendez, J.A.; Vazquez-Martin, A.; Colomer, R.; Brunet, J.; Carrasco-Pancorbo, A.; Garcia-Villalba, R.; Fernandez-Gutierrez, A.; Segura-Carretero, A. Olive oil’s bitter principle reverses acquired autoresistance to trastuzumab (HerceptinTM) in HER2-overexpressing breast cancer cells. BMC Cancer 2007, 7, 80. [Google Scholar] [CrossRef]
  175. Sherif, I.O.; Al-Gayyar, M.M.H. Oleuropein potentiates anti-tumor activity of cisplatin against HepG2 through affecting proNGF/NGF balance. Life Sci. 2018, 198, 87–93. [Google Scholar] [CrossRef]
  176. Xu, T.; Xiao, D. Oleuropein enhances radiation sensitivity of nasopharyngeal carcinoma by downregulating PDRG1 through HIF1α-repressed microRNA-519d. J. Exp. Clin. Cancer Res. 2017, 36, 3. [Google Scholar] [CrossRef]
  177. Fredrickson, W.R. Method and Composition for Antiviral Therapy with Olive Leaves. U.S. Patent No. 6,117,844, 2000. [Google Scholar]
  178. Micol, V.; Caturla, N.; Pe’rez-Fons, L.; Ma’s, V.; Pe’rez, L.; Estepa, A. The olive leaf extract exhibits antiviral activity against viral haemorrhagic septicaemia rhabdovirus (VHSV). Antivir. Res. 2005, 66, 129–136. [Google Scholar] [CrossRef]
  179. Motamedifar, M.; Nekooeian, A.A.; Moatari, A. The Effect of Hydroalcoholic Extract of Olive Leaves against Herpes Simplex Virus Type 1. Iran. J. Med. Sci. 2007, 32, 222–227. [Google Scholar]
  180. Ben-Amor, I.; Musarra-Pizzo, M.; Smeriglio, A.; D’Arrigo, M.; Pennisi, R.; Attia, H.; Gargouri, B.; Trombetta, D.; Mandalari, G.; Sciortino, M.T. Phytochemical Characterization of Olea europea Leaf Extracts and Assessment of Their Anti-Microbial and Anti-HSV-1 Activity. Viruses 2021, 13, 1085. [Google Scholar] [CrossRef]
  181. Lee-Huang, S.; Zhang, L.; Huang, P.L.; Chang, Y.T.; Huang, P.L. Anti-HIV activity of olive leaf extract (OLE) and modulation of host cell gene expression by HIV-1 infection and OLE treatment. Biochem. Biophys. Res. Commun. 2003, 307, 1029–1037. [Google Scholar] [CrossRef] [PubMed]
  182. Pennisi, R.; Ben Amor, I.; Gargouri, B.; Attia, H.; Zaabi, R.; Chira, A.B.; Saoudi, M.; Piperno, A.; Trischitta, P.; Tamburello, M.P.; et al. Analysis of Antioxidant and Antiviral Effects of Olive (Olea europaea L.) Leaf Extracts and Pure Compound Using Cancer Cell Model. Biomolecules 2023, 13, 238. [Google Scholar] [CrossRef] [PubMed]
  183. Zhao, G.; Yin, Z.; Dong, J. Antiviral efficacy against hepatitis B virus replication of oleuropein isolated from Jasminum officinale L. var. grandiflorum. J. Ethnopharmacol. 2009, 125, 265–268. [Google Scholar] [CrossRef] [PubMed]
  184. Majrashi, T.A.; El Hassab, M.A.; Mahmoud, S.H.; Mostafa, A.; Wahsh, E.A.; Elkaeed, E.B.; Hassan, F.E.; EldehnaID, W.M.; Abdelgawad, S.M. In vitro biological evaluation and in silico insights into the antiviral activity of standardized olive leaves extract against SARS-CoV-2. PLoS ONE 2024, 19, e0301086. [Google Scholar] [CrossRef]
  185. Ma, S.C.; He, Z.D.; Deng, X.L.; But, P.P.H.; Ooi, V.E.C.; Xu, H.X.; Lee, S.H.S.; Lee, S.F. In vitro evaluation of secoiridoid glucosides from the fruits of Ligustrum lucidum as antiviral agents. Chem. Pharm. Bull. 2001, 49, 1471–1473. [Google Scholar] [CrossRef]
  186. Abdelgawad, S.M.; El Hassab, M.A.; Abourehab, M.A.; Elkaeed, E.B.; Eldehna, W.M. Olive Leaves as a Potential Phytotherapy in the Treatment of COVID-19 Disease; A Mini-Review. Front. Pharmacol. 2022, 13, 879118. [Google Scholar] [CrossRef]
  187. Khaerunnisa, S.; Kurniawan, H.; Awaluddin, R.; Suhartati, S.; Soetjipto, S. Potential inhibitor of COVID-19 main protease (Mpro) from several medicinal plant compounds by molecular docking study. PPR 2020, 2020030226. [Google Scholar]
  188. Vijayan, R.; Gourinath, S. Structure-based inhibitor screening of natural products against NSP15 of SARS-CoV-2 revealed Thymopentin and Oleuropein as potent inhibitors. Protein J. 2021, 12, 71–80. [Google Scholar] [CrossRef]
  189. Sampangi-Ramaiah, M.H.; Vishwakarma, R.; Shaanker, R.U. Molecular docking analysis of selected natural products from plants for inhibition of SARS-CoV-2 main protease. Curr. Sci. 2020, 118, 1087–1092. [Google Scholar] [CrossRef]
  190. Vardhan, S.; Sahoo, S.K. In silico ADMET and molecular docking study on searching potential inhibitors from limonoids and triterpenoids for COVID-19. Comput. Biol. Med. 2020, 124, 103936. [Google Scholar] [CrossRef]
  191. Yu, R.; Chen, L.; Lan, R.; Shen, R.; Li, P. Computational screening of antagonists against the SARS-CoV-2 (COVID-19) coronavirus by molecular docking. Int. J. Antimicrob. Agents 2020, 56, 106012. [Google Scholar] [CrossRef] [PubMed]
  192. Shawky, E.; Nada, A.A.; Ibrahim, R.S. Potential role of medicinal plants and their constituents in the mitigation of SARS-CoV-2: Identifying related therapeutic targets using network pharmacology and molecular docking analyses. RSC Adv. 2020, 10, 27961–27983. [Google Scholar] [CrossRef] [PubMed]
  193. Khan, A.; Heng, W.; Wang, Y.; Qiu, J.; Wei, X.; Peng, S.; Saleem, S.; Khan, M.; Ali, S.S.; Wei, D.Q. In silico and in vitro evaluation of kaempferol as a potential inhibitor of the SARS-CoV-2 main protease (3CLpro). Phytother. Res. 2021, 35, 2841–2845. [Google Scholar] [CrossRef] [PubMed]
  194. Topuz, S.; Bayram, M. Oleuropein extraction from leaves of three olive varieties (Olea europaea L.): Antioxidant and antimicrobial properties of purified oleuropein and oleuropein extracts. J. Food Process. Preserv. 2021, 46, e15697. [Google Scholar] [CrossRef]
  195. Takeda, Y.; Jamsransuren, D.; Matsuda, S.; Crea, R.; Ogawa, H. The SARS-CoV-2-Inactivating Activity of Hydroxytyrosol-Rich Aqueous Olive Pulp Extract (HIDROX®) and its Use as a Virucidal Cream for Topical Application. Viruses 2021, 13, 232. [Google Scholar] [CrossRef]
  196. Yi, L.; Li, Z.; Yuan, K.; Qu, X.; Chen, J.; Wang, G.; Zhang, H.; Luo, H.; Zhu, L.; Jiang, P.; et al. Small molecules blocking the entry of severe acute respiratory syndrome coronavirus into host cells. Virol. J. 2004, 78, 11334–11339. [Google Scholar] [CrossRef]
  197. Borgio, J.F.; Alsuwat, H.S.; Al Otaibi, W.M.; Ibrahim, A.M.; Almandil, N.; Al Asoom, L.I.; Salahuddin, M.; Kamaraj, B.; AbdulAzeez, S. State-of-the-art tools unveil potent drug targets amongst clinically approved drugs to inhibit helicase in SARS-CoV-2. Arch. Med. Sci. 2020, 16, 508–518. [Google Scholar] [CrossRef]
  198. Li, G.; De Clercq, E. Therapeutic options for the 2019 novel coronavirus (2019-nCoV). Nature reviews. Drug Discov. 2020, 19, 149–150. [Google Scholar] [CrossRef]
  199. Jeong, G.U.; Song, H.; Yoon, G.Y.; Kim, D.; Kwon, Y.C. Therapeutic strategies against COVID-19 and structural characterization of SARS-CoV-2: A review. Front. Microbiol. 2020, 11, 1723. [Google Scholar] [CrossRef]
  200. Pereira, A.P.; Ferreira, I.C.; Marcelino, F.; Valentão, P.; Andrade, P.B.; Seabra, R.; Estevinho, L.; Bento, A.; Pereira, J.A. Phenolic compounds and antimicrobial activity of olive (Olea europaea L. Cv. Cobrançosa) leaves. Molecules 2007, 12, 1153–1162. [Google Scholar] [CrossRef]
  201. Tuck, K.L.; Hayball, P.J. Major phenolic compounds in olive oil: Metabolism and health effects. J. Nutr. Biochem. 2002, 13, 636–644. [Google Scholar] [CrossRef]
  202. Medina, E.; de Castro, A.; Roero, C.; Brenes, M. Comparison of the concentrations of phenolic compounds in olive oils and other plant oils: Correlation with antimicrobial activity. J. Agric. Food Chem. 2006, 54, 4954–4961. [Google Scholar] [CrossRef] [PubMed]
  203. Laincer, F.; Laribi, R.; Tamendjari, A.; Arrar, L.; Rovellini, P.; Venturini, S. Olive oils from Algeria: Phenolic compounds, antioxidant and antibacterial activities. Grasas Aceites 2014, 65, e001. [Google Scholar] [CrossRef]
  204. Bisignano, G.; Tomaino, A.; Cascio, R.L.; Crisafi, G.; Uccella, N.; Saija, A. On the in vitro antimicrobial activity of oleuropein and hydroxytyrosol. J. Pharm. Pharmacol. 1999, 51, 971–974. [Google Scholar] [CrossRef] [PubMed]
  205. 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. Antimicrob. Agents 2009, 33, 461–463. [Google Scholar] [CrossRef]
  206. Friedman, M.; Henika, P.; Mandrell, R. Bactericidal activities of plant essential oils and some of their isolated constituents against Campylobacter jejuni, Escherichia coli, Listeria monocytogenes, and Salmonella enterica. J. Food Prot. 2002, 65, 1545–1560. [Google Scholar] [CrossRef]
  207. Russo, M.; Moccia, S.; Spagnuolo, C.; Tedesco, I.; Russo, G.L. Roles of flavonoids against coronavirus infection. Chem. Biol. Interact. 2020, 328, 109211. [Google Scholar] [CrossRef]
  208. Dominciano, L.C.C.; Lee, S.H.I.; Corassin, C.H.; Martinis, E.C.P.; Oliveira, C.A.F. Effects of oleuropein and peracetic acid as sanitizing agents for inactivation of biofilms. Open Conf. Proc. J. 2016, 7, 1–6. [Google Scholar] [CrossRef]
  209. Dominciano, L.C.C.; Oliveira, C.A.F.; Lee, S.H.; Corassin, C.H. Individual and Combined Antimicrobial Activity of Oleuropein and Chemical Sanitizers. J. Food Chem. Nanotechnol. 2016, 2, 124–127. [Google Scholar] [CrossRef]
  210. Caturla, N.; Pérez-Fons, L.; Estepa, A.; Micol, V. Differential effects of oleuropein, a biophenol from Olea europaea, on anionic and zwiterionic phospholipid model membranes. Chem. Phys. Lipids 2005, 137, 2–17. [Google Scholar] [CrossRef]
  211. Casas-Sanchez, J.; Alsina, A.M.; Herrlein, M.K.; Mestres, C. Interaction between the antibacterial compound, oleuropein, and model membranes. Colloid Polym. Sci. 2007, 285, 1351–1360. [Google Scholar] [CrossRef]
Figure 1. Structure of oleuropein and its hydrolyzed derivatives (Source: PubChem).
Figure 1. Structure of oleuropein and its hydrolyzed derivatives (Source: PubChem).
Beverages 11 00066 g001
Table 1. Chemical composition of selected herb species and herbal drinks.
Table 1. Chemical composition of selected herb species and herbal drinks.
Herb Species and Herbal DrinksChemical CompositionReferences
Green teaFlavonoids, terpenoids, cardiac glycosides saponins and tannins[14]
Green tea infusions containing broccoli by-productsCatechins, hydroxycinnamic acids, flavonols and glucosinolates[14]
Tea from Artemisia annua L.Coumarins, terpenes, flavonoids, acetylenes and phenols[14]
Loloh cemcem, Bali traditional drinkTannins, terpenoids, flavonoids, alkaloids, and phenols[15]
Olive leaf extractsFlavonoids (luteolin, apigenin, luteolin-7-O-glucoside, etc.), ferulic acid, caffeic acid, tyrosol and hydroxytyrosol and secoiridoids (ligstroside, oleuropein dimethyloleuropein),[16]
Table 2. Effect of oleuropein on triglyceride (TG), total cholesterol (TC), low-density lipoprotein (LDL), high-density lipoprotein (HDL), and very low-density lipoprotein (VLDL) in diabetic rats [95].
Table 2. Effect of oleuropein on triglyceride (TG), total cholesterol (TC), low-density lipoprotein (LDL), high-density lipoprotein (HDL), and very low-density lipoprotein (VLDL) in diabetic rats [95].
ParameterControlDiabeticDiabetic + Oleuropein
TG (mg/dL)82.33 ± 14.75126.57 ± 18.5997.69 ± 13.91
TC (mg/dL)72.01 ± 16.35110.88 ± 28.4883.38 ± 20.75
HDL (mg/dL)39.00 ± 13.2925.01 ± 9.1938.13 ± 10.67
LDL (mg/dL)26.52 ± 21.5860.57 ± 28.1825.73 ± 21.06
VLDL (mg/dL)16.47 ± 2.4925.30 ± 3.4419.55 ± 2.78
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Panou, A.A.; Karabagias, I.K. Olive Leaf Extracts as a Medicinal Beverage: Origin, Physico-Chemical Properties, and Bio-Functionality. Beverages 2025, 11, 66. https://doi.org/10.3390/beverages11030066

AMA Style

Panou AA, Karabagias IK. Olive Leaf Extracts as a Medicinal Beverage: Origin, Physico-Chemical Properties, and Bio-Functionality. Beverages. 2025; 11(3):66. https://doi.org/10.3390/beverages11030066

Chicago/Turabian Style

Panou, Andreas Alexandros, and Ioannis Konstantinos Karabagias. 2025. "Olive Leaf Extracts as a Medicinal Beverage: Origin, Physico-Chemical Properties, and Bio-Functionality" Beverages 11, no. 3: 66. https://doi.org/10.3390/beverages11030066

APA Style

Panou, A. A., & Karabagias, I. K. (2025). Olive Leaf Extracts as a Medicinal Beverage: Origin, Physico-Chemical Properties, and Bio-Functionality. Beverages, 11(3), 66. https://doi.org/10.3390/beverages11030066

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