Next Article in Journal
Consumption of Barley, Buckwheat and Quinoa in the United States: Associations with Diet and Metabolic Health
Previous Article in Journal
Vitamin D’s Impact on Cancer Incidence and Mortality: A Systematic Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Extra Virgin Olive Oil (EVOO) Components: Interaction with Pro-Inflammatory Cytokines Focusing on Cancer and Skeletal Muscle Biology

by
Daniela De Stefanis
and
Paola Costelli
*
Department of Clinical and Biological Sciences, University of Turin, 10125 Turin, Italy
*
Author to whom correspondence should be addressed.
Nutrients 2025, 17(14), 2334; https://doi.org/10.3390/nu17142334
Submission received: 17 June 2025 / Revised: 11 July 2025 / Accepted: 14 July 2025 / Published: 16 July 2025
(This article belongs to the Section Phytochemicals and Human Health)

Abstract

The advantages of extra virgin olive oil (EVOO) intake as part of a varied, healthy and balanced diet were demonstrated by many epidemiological studies. In particular, several components present in EVOO, such as tocopherols, carotenoids and phenolic compounds, play an important protective role in mitigating inflammatory diseases, atherosclerosis, neurodegenerative diseases and cancer. The protective effect exerted by EVOO was proposed to be accounted for by its antioxidant, anti-inflammatory or anti-proliferative properties. The present review will focus on the interactions among EVOO’s components and pro-inflammatory cytokines, aiming to reveal the mechanisms potentially involved in the anticancer action of EVOO. Cancer patients very frequently develop a devastating syndrome known as cachexia, which negatively impinges on their outcome. The main features of cachexia include progressive body weight loss, fat and muscle wasting, and dysmetabolism, all of which partially result from the onset of systemic inflammation. In this regard, the possibility that EVOO could be beneficial to cancer patients by mitigating cachexia will be reviewed, focusing on the skeletal muscle.

1. Introduction

Olive oil is one of the most ancient foods. Together with the olive tree, it has been known for thousands of years, and it was always used as a seasoning and cooking medium. In the last forty years, however, nutritional facts about the different types of olive oil were deeply investigated and EVOO is now considered one of the most relevant components of the Mediterranean diet.
The chemical composition of olive oils particularly depends on several factors linked to the specific cultivar, to peculiar environmental features as well as to both production and transformation processes, with the latter considered highly relevant in terms of the quality of the final product. EVOO is obtained from the fruit of the olive tree by mechanical crushing in the absence of solvents or chemical processes, followed by washing, decantation or centrifugation, and filtration [1]. EVOO is particularly enriched in healthy nutrients such as mono-unsaturated fatty acids, oleic acid in particular, essential fatty acids and bioactive compounds such as polyphenols, phytosterols, squalene, etc. While these latter compounds represent about 2% of EVOO’s composition, they are crucial to the definition of some organoleptic properties of EVOO, such as the taste and flavour. In addition, they are mainly responsible for the beneficial action of EVOO, being endowed with both antioxidant and anti-inflammatory activity. In this regard, epidemiologic data showed that regularly consuming EVOO was associated with protection against cardiovascular diseases, neurodegenerative and chronic inflammatory diseases and cancer.

2. Olive Oil Components

2.1. Biological Activity of EVOO

EVOO is mainly composed (98–99%) of a lipophilic fraction characterized by a high content of monounsaturated fatty acids (MUFAs), ranging from 65% to 83%, with oleic acid being the predominant MUFA. Additionally, EVOO contains polyunsaturated fatty acids (PUFAs), such as linoleic acid. The remaining 1–2% is represented by a hydrophilic fraction containing tocopherols, hydrocarbons, polyphenols, secoiridoids, aldehydes, ketones, beta-carotene, esters, alcohol, sterols, lignans, flavonoids, etc., also referred to as EVOO derivatives (Figure 1) [1].
Several studies demonstrated the preventive properties of EVOO against atherosclerosis, cardiometabolic disorders, neurodegenerative diseases, and cancers [2,3,4,5,6]. These beneficial properties are mainly linked to the polyphenols present in EVOO, such as tyrosol, hydroxytyrosol (HT), oleuropein (OLE), and oleocanthal (OC), which were shown to display anticancer activity and to counteract oxidative stress and inflammation (Table 1) [7,8,9].
Olive oil forms a micellar solution, allowing most phenolic compounds in EVOO to pass through the mouth and stomach largely unchanged. These compounds then reach the small intestine and colon [10], where they are directly absorbed, metabolized, and distributed to tissues, or transformed by the gut microbiota [11,12,13]. In the intestinal tract, hydroxytyrosol and tyrosol are assimilated at rates ranging from approximately 40% to 95%, depending on the dose [14]. While secoiridoids are highly stable in the mouth, they experience significant loss in the stomach, duodenum, and colon, with the recovery rates in the duodenum ranging between 7% and 34%. Their absorption involves processes such as glycosylation and the cleavage of glycosidic bonds. Additionally, some secoiridoids, like oleacein, are likely absorbed in the small intestine, primarily through passive diffusion across the intestinal cell membranes [10].
The 2011 PREDIMED study showed that EVOO’s consumption can improve glucose metabolism, preventing the onset of diabetes [15]. A subsequent PREDIMED study (2018) involving people at high risk of cardiovascular events showed that such a risk was lower in patients on a diet supplemented with EVOO than in those assigned to a low-fat diet [16]. A different study showed that metabolic markers, the abdominal fat distribution and the levels of pro-inflammatory cytokines in patients affected by metabolic syndrome were improved by the consumption of EVOO for 60 days [17]. Furthermore, in patients presenting with obesity and prediabetes, EVOO rich in OC and oleacein (OA) was more effective than a common olive oil in promoting body weight loss, improving fasting glucose and redox homeostasis, and decreasing circulating interferon-γ (IFN-γ) [18].
Table 1. Main bioactivities of EVOO’s polyphenols. Symbols: ↓ reduce; ↑ increment.
Table 1. Main bioactivities of EVOO’s polyphenols. Symbols: ↓ reduce; ↑ increment.
Activity
Category
ReferencesPolyphenolsEffectsRange of Concentration
Antioxidant[2,4,5,7,8,9,19]Hydroxytyrosol, Oleuropein,
Tyrosol
Free radical
scavenging
↓ oxidative stress in cells and tissues.
In vitro 10–100 μM
In vivo 10–50 mg/kg/day
Anti-inflammatory[3,4,5,6,7,19]Oleuropein,
Hydroxytyrosol,
Oleocanthal,
↓ Cytokines (TNF-α, IL-6)
↓ NF-κB pathway
↓ inflammation markers
In vitro 10–100 μM
Antitumoral[6,20,21]Oleuropein,
Hydroxytyrosol,
Oleocanthal
↑ apoptosis
↓ proliferation of various cancer cell lines (e.g., colon, breast, hepatoma, melanoma)
Modulates miRNA
expression profiles
In vitro 20–100 μM.
In vivo doses depend on model systems.
Gut Microbiota
Modulation
[2,7,22,23]Hydroxytyrosol, OleuropeinPromotes beneficial bacteria growth (Lactobacillus, Bifidobacterium)
improves gut health
Dietary supplementation (~50 mg/day)
Atherosclerosis and
Cardiometabolic
Disorders
[3,4,8,24,25,26,27,28]Oleuropein,
Hydroxytyrosol
Improves lipid profiles
↓ vascular inflammation ↓ foam cell formation
↓ blood pressure
In vivo 10–50 mg/kg/dye
In human studies by Mediterranean diet
Neurodegenerative
Disease
[4,5,9,29]Hydroxytyrosol, Oleuropein↓ neuroinflammation Potential to improve cognitive functionIn vitro 10–50 μM
In vivo doses vary depending on model
Due to their antioxidant properties, the polyphenols in EVOO reduced LDL oxidation and optimized the circulating cholesterol and triglyceride levels [30,31], counteracting the formation of atherosclerotic plaques [2]. Consistently, Casas et al. [24] demonstrated that adherence to the Mediterranean diet decreased the blood levels of inflammatory biomarkers related to the onset of atheromas in elderly people at high cardiovascular risk. Moreover, studies performed using a murine model of atherosclerosis showed that HT reduced both oxidative stress and inflammation in the vasculature, attenuating the progression of the disease [25]. The antioxidant and anti-inflammatory action of polyphenols and other compounds in EVOO also had beneficial implications for the blood pressure, thanks to a direct effect on the blood vessels (improved elasticity) [26], for the induction of nitric oxide (NO) synthesis by endothelial cells [32] and for the modulation of the expression of genes pertaining to the renin–angiotensin system [27].
Several studies showed the neuroprotective effect of EVOO’s components [29]. Experimental results obtained on a rat brain exposed to an ischemia–reperfusion protocol demonstrated that the antioxidant action of HT included the reduction of reactive nitrogen and oxygen (ROS) species, of lactate dehydrogenase levels and of pro-inflammatory cytokines [33,34]. Such effects were likely accounted for by the increasing hydrogen peroxide and hydroxyl radical scavenging, mainly through activation of the Nrf2 and JNK-p62/SQSTM1 pathways, and by inducing heme-oxygenase-1 [28,35,36,37]. Finally, in animals exposed to brain ischemia, HT improved both the blood flow and the connections among the brain regions, reduced inflammation and was able to positively impinge on the recovery of muscle function in the 15-day post-ischemia time window [38,39].

2.2. Epigenetic Effects of EVOO Derivatives

In the last few years, several pieces of evidence have demonstrated that the bioactivity of EVOO, and of the phenols it contains, can result from epigenetic-related mechanisms [40,41]. The latter may reflect DNA methylation, histone modification (acetylation and methylation), and noncoding RNAs such as microRNAs (miRNAs). Such epigenetic modifications are tissue-specific and stable enough to be inherited for a few generations [42].
The ability of EVOO to impinge on epigenetic regulation could contribute to preventing the development of some chronic diseases, such as cancer (see below) or cardiovascular events [40,41]. Consistently, patients on a Mediterranean diet showed changes in the methylation of the CpG sites of genes related to inflammation, diabetes, intermediary metabolism and a number of signal transduction pathways [43]. Similarly, studies performed using an experimental model of Alzheimer’s disease showed that HT was able to positively modulate the epigenetic dysregulation, resulting in an improvement of the cognitive impulsivity associated with the disease [44].
Many studies suggest that EVOO, or its derivatives, induces epigenetic changes by modulating miRNAs. In this regard, HT was reported to protect human chondrocytes from H2O2-induced cell death and the expression of some markers of osteoarthritis by reducing the miR-9 levels [45,46]. D’Amore et al. [47] compared the effect of high-polyphenol EVOO intake on the expression of the genes and miRNAs of peripheral blood mononuclear cells in healthy subjects and metabolic syndrome patients. In the former, but not in the latter, EVOO was able to downregulate the miRNAs involved in inflammation (miR-181b-5p and miR-23b-3p), insulin resistance (miR-107), and cancer (miR-19a-3p and miR-519b-3p), while resulted in the upregulation of miRNAs associated with anti-inflammatory (miR-23b-3p) and tumour-suppressing activity (miR-519b-3p). Finally, Carpi et al. [48] showed that modulations of NF-κB-related miRs, namely miR-34, miR-155 and let-7c, as induced by TNF-α in Simpson–Golabi–Behmel syndrome adipocytes, could be prevented by pretreatment with OC or OA.

3. EVOO’s Anticancer Activity

Tumour development is a multistep process that includes several events (neoplastic transformation, clonal expansion, angiogenesis and metastasis) that are strictly associated with inflammation [49,50]. Along this line, dietary EVOO was shown to exert a protective action against breast, colon and liver cancer.
Colorectal cancer (CRC) is a widespread tumour with a high mortality rate [51]. Several studies have confirmed an association between EVOO consumption and reduced onset of CRC [22]. A possible protective mechanism was proposed by Rodríguez-García et al. [52]. In their study, mice receiving three different types of high-fat diet (HFD) containing, as a unique fat source, coconut oil or sunflower oil or EVOO were compared. The EVOO-receiving mice showed a preventive effect against CRC development, which was associated with anti-inflammatory changes in the gut microbiota composition, such as decreased abundance of Enterococcus and Pseudomonas and an increased Firmicutes/Bacteroidetes ratio. By contrast, the coconut- and sunflower HFDs produced a dysbiosis that increased the CRC risk.
Consistently, the exposure to an EVOO extract enriched in OC and the ligstroside aglycone of hepatoma cells (HepG2, Huh7, Hep3B) reduced cell proliferation, increased cell death, and induced autophagy [20]. In addition, OC was shown to reduce liver tumour growth induced in nude mice by orthotopic implantation of human HCCLM3 cells, mainly by inhibiting STAT3 activity [53].
Garcia-Guasch et al. [54] showed that, compared to a diet rich in EVOO, a high-PUFA diet promoted the onset of breast tumour with a high degree of histopathological and proliferative alterations. By contrast, the high-EVOO diet, mainly through HT and/or OC, positively modulated the expression of proteins involved in different cell death pathways, favouring apoptosis. Similarly, OC was shown to suppress cell proliferation in two human melanoma cell lines, the highly tumorigenic A375 and the metastatic 501Mel, by downregulating Bcl-2 gene expression and reducing ERK and Akt phosphorylation [21].
Angiogenesis is an important factor for tumour survival, promoting both tumour growth and metastasis by providing nutrients and oxygen. The phenolic compounds from EVOO were demonstrated to reduce angiogenesis by modulating, in endothelial cells, the expression of proteins involved in proliferation, migration and invasion, adhesion, and survival [55,56]. Marrero et al. [57] demonstrated in vitro that OC and OA reduced the activity of matrix metalloproteinase (MMP)-2, thus inhibiting the ability of endothelial cells to invade matrigel and to form tubes, eventually resulting in endothelial cell death. More recently, treatment of HUVEC cells with an EVOO phenolic fraction was shown to reduce the expression levels of proteins associated with angiogenesis, inhibiting the ability of endothelial cells to migrate, adhere and form tubes. Such an effect was associated with reduced extracellular matrix degradation and enhanced endothelial cell apoptosis [58].
The chemopreventive role of EVOO in carcinogenesis could also be accounted for by modulations of the methylation status of genes involved in breast [59,60] and colon [61] cancer progression. Similarly, in rats hosting colon cancer, EVOO administration increased the methylation of the promoter region of the genes coding for NF-κB, VEGF, and MMP-9, downregulating their expression and counteracting tumour progression. Moreover, EVOO induced the hypomethylation of the promoter region of the genes encoding miR-143 and miR-145, upregulating their expression and eventually counteracting the NF-kB pathway. Finally, EVOO-induced hypomethylation was also observed in the promoter region of the genes coding for caspase-3 and caspase-9, increasing their expression and restoring apoptosis [62].
EVOO and its derivatives were shown to also inhibit breast cancer by modulating the expression of miRNAs, both in vitro and in vivo. In MDA-MB-231 and MCF-7 cells, OLE increased the expression of tumour suppressor miRNAs (miR-125b, miR-16, miR-34a) and pro-apoptotic genes (p53, p21 and TNFRS10B), while it decreased the expression of onco-miRNAs (miR-221, miR-29a, miR-21 and miR-155) and anti-apoptotic genes (bcl-2, mcl1) [63,64]. In addition, in MDA-MB-231 cells, OLE resulted in decreased levels of miR-194-5p and of its target PD-L1, one of the most relevant immune-escape-associated factors [65].
The antioxidant action of EVOO was shown to counteract the early stages of tumour development [7,8,9]; however, several studies reported that OLE, by inhibiting mitochondrial function, caused excessive ROS accumulation and apoptosis in different cancer cell lines (HepG2, A549, MCF-7, and MDA-MB-231) [66,67,68,69]. Similarly, the exposure of cancer cells to OC was reported to inhibit cell proliferation and colony formation, and to induce apoptosis, mainly by stimulating mitochondrial depolarization and intracellular ROS production [70].
Finally, EVOO-derived compounds appeared to also exert protective effects against chemotherapy-associated toxicity, one of the most relevant complications in the management of cancer patients. Indeed, both in vitro and in vivo experiments showed that secoiridoids such as OC, OLE, or tyrosol combined with chemotherapeutic drugs synergistically to reduce tumour cell proliferation [71,72,73,74,75], without affecting healthy cells [70,76,77].

EVOO Interaction with Pro-Inflammatory Cytokines

The onset and progression of most solid and hematopoietic tumours is associated with chronic inflammation that affects all stages of tumour growth (Figure 2) [49]. As an example, the low-grade inflammation occurring in patients with obesity and/or consuming an HFD supports gastrointestinal cancer development [78]. Similarly, pathogens, such as viruses and bacteria, frequently activate the inflammatory response, promoting the onset of cancer, as occurs in gastric cancer associated with Helicobacter pylori or in hepatocellular carcinoma favoured by hepatitis B/C viruses [79].
EVOO-derived polyphenols were reported to exert a protective effect against the onset of inflammatory bowel disease [80], a chronic disease of the gastrointestinal tract that can evolve into ulcerative colitis (UC) and Crohn’s disease (CD) [81]. Chicco et al. [82] analysed patients with IBD (84 with CD and 58 with UC) who were fed a Mediterranean diet for 6 months, showing that most of them experienced decreased liver steatosis and levels of inflammatory biomarkers (C-reactive protein and faecal calprotectin), improved quality of life and reduced disease activity. Moreover, daily consumption of EVOO decreased the inflammatory markers and improved the gastrointestinal symptoms in UC patients [83]. Finally, in vitro and in vivo experiments demonstrated that EVOO and its derivatives preserved the homeostasis of the intestinal epithelium by counteracting both oxidative stress and inflammation, maintaining or improving the gut microbiota and the immune response [23].
The exposure of differentiated colon cancer Caco-2 cells to oxysterols increased the production of pro-inflammatory cytokines and ROS; such an effect was inhibited by pre-treatment with a phenolic extract of EVOO, resulting in reduced IL-8, IL-6, and iNOS release in the culture medium [84]. Furthermore, in Caco-2 cells exposed to bacterial lipopolysaccharide (LPS), pretreatment with HT or tyrosol, or with their sulphate and glucuronide metabolites, inhibited iNOS expression and NF-κB activity [85].
In addition to favouring cancer onset, inflammation also plays crucial roles during cancer progression. Indeed, inflammatory cells infiltrate the tumour stroma and release mediators such as ROS, pro- and anti-inflammatory cytokines, chemokines, and growth factors. Moreover, cytokines can also be produced by the tumour itself, generating a tumour-promoting inflammatory loop that boosts its own development [49]. This whole scenario is regulated by multiple signalling pathways and transcription factors, with the dominant role being played by the axis impinging on IKK-NF-κB, JAK-signal transducer, STAT3, and MAPK-AP1, which can be influenced, directly or indirectly, by EVOO and its derivatives [86,87,88,89,90]. This was clearly shown by a study in which mice suffering from colitis induced by dextran sodium sulphate (DSS) were exposed to a standard diet, an EVOO-containing diet or an HT-enriched EVOO-containing diet. The EVOO-containing diet reduced by 50% the DSS-induced mortality observed in the standard diet group. Such an effect was associated with decreased levels of TNF-α and iNOS, reduced p38 MAPK activation and increased expression of the anti-inflammatory cytokine IL-10. This pattern was further improved in mice receiving the HT-enriched EVOO-containing diet [91].
Huguet-Casquero et al. [92] explored the effects of OLE alone or loaded on nanostructured lipid carriers in mice affected by acute colitis, showing that both treatments significantly decreased the myeloperoxidase activity, TNF-α, and IL-6 concentration in the colon mucosa, confirming the anti-inflammatory properties of EVOO derivatives, which likely reduce the risk of developing CRC. In the liver, inflammation and progressive tissue damage (e.g., steatosis, NASH, cirrhosis) result in preneoplastic lesions, which eventually evolve into hepatocellular carcinomas [93].
A number of studies showed that EVOO and its derivatives exert protective effects against liver damage, mainly by activating the Nrf2-dependent antioxidant response, counteracting inflammation through NF-κB inactivation, and inhibiting PERK, thus affording protection against endoplasmic reticulum stress, excess autophagy, and lipogenesis [19]. Several experiments showed that oleic acid and HT prevented lipid peroxidation in the rat liver [94], resulting in inhibition of hepatic fibrogenesis [95,96], liver steatosis [97,98,99], and hepatocyte ballooning [19]. Furthermore, HT downregulated the TNF-α and IL-6 mRNA and COX-2 expression in the liver of young male rats exposed to HFD, decreasing both steatosis and inflammation [19]. In addition, tyrosol administration to mice affected by NASH was shown to modulate the hepatic immune milieu, reducing inflammation and improving steatosis and fibrosis [100]. Finally, in HFD-fed mice, OLE lowered the LPS, TNF-α, and IFN-γ in the serum, and downregulated the intestinal and liver TLR4+ macrophages, reducing liver inflammation and steatosis [101]. Such effects were also proposed to be achieved by the OC-induced inhibition of the STAT3-dependent pathway, which was shown to result in reduced proliferation, epithelial–mesenchymal transition, migration and invasiveness and enhanced apoptosis in hepatocellular carcinoma cells as well as in an orthotopically implanted experimental HCC [53].

4. EVOO and the Skeletal Muscle

4.1. Skeletal Muscle Homeostasis

The skeletal muscle contains most of the body’s protein and represents about 40% of the human body weight [102]. Myogenesis occurs during embryonal, foetal and neonatal life. However, it can be reactivated during adult life in the case of muscle injury [103]. During muscle development, individual cells, the myoblasts, which derive from a population of multipotent mesodermal progenitors, fuse to form muscle fibres. The survival of these progenitor cells is guaranteed by the transcription factors Pax3 and Pax7, and their subsequent progression towards myoblasts is operationalized by the myogenic regulatory factors (MRFs: MyoD, Myf5, Mrf4, myogenin) [103,104]. During embryonic development, Myf5 is the first MRF to be expressed by the progenitor cells, which can subsequently differentiate into mononuclear myocytes that form the early muscle tissue of the embryo under the control of Mrf4 and myogenin. As the differentiation process progresses, stem cells expressing Pax3/Pax7, Myf5, and MyoD are recruited and contribute to the development of muscle tissue. At this point, Mrf4 is no longer expressed, while MyoD and myogenin drive the formation of primary myofibers through the fusion of myoblasts, in a process called primary myogenesis. Secondary myogenesis completes muscle formation: myogenin and Mrf4 drive the fusion of further proliferating myocytes into primary myofibers, which will then follow the specification into the oxidative (slow) or glycolytic (fast) phenotype [103,105]. In adults, myogenesis occurs following tissue damage through the recruitment of undifferentiated progenitor cells in a process that closely resembles the embryonal one [106].
The skeletal muscle performs several functions, such as force generation, movement, metabolic and endocrine regulation [103]. During fasting, the muscle can provide amino acids as precursors for gluconeogenesis, and its mass is directly related to bone density and mineralization [107]. Maintaining skeletal muscle mass, metabolism and function allows the body to cope with stressful conditions and helps prevent the onset of some chronic diseases [108].
Muscle mass depends on the balance between catabolic and anabolic processes, which are influenced by nutritional status, mechanical stress, age and concurrent pathologies. Muscle homeostasis is controlled and regulated through the pathways dependent on insulin/IGF1–Akt–mTOR (anabolic) and TGF-β/myostatin/BMP (catabolic), among others [109]. In particular, while protein catabolism is controlled by the Akt/FOXO pathway, protein synthesis is regulated by the Akt/mTOR axis. The interaction of these pathways is regulated, partially at least, by myostatin and insulin/IGF-1: overproduction of myostatin downregulates the Akt/FOXO axis, activating protein degradation and resulting in decreased muscle mass, while insulin/IGF-1 reduces muscle atrophy by switching off FOXO via Akt stimulation [110,111].
The complex interaction among Akt, FOXO and mTOR was demonstrated in mice characterized by skeletal-muscle-specific Akt deficiency, which showed a loss of muscle mass and function, associated with reduced oxidative metabolism. Such a pattern mainly resulted from decreased protein synthesis without significant changes in protein breakdown. In the same mouse model, mTORC1 activation or FOXO1 inhibition alone was not sufficient to rescue the loss of muscle mass, which is counteracted when both pathways are convergently (FOXO1 inhibited and mTORC1 activated) and simultaneously modulated [112].
As for the TGF-β/myostatin/BMP pathway, myostatin binding to its receptor activates ALK4/5/7, leading to phosphorylation of Smad2/3, which will interact with Smad4, forming a complex able to translocate into the nucleus. On the other side, when BMP ligands engage the receptor, ALK2/3/6 is recruited to activate Smad 1/5/8. The latter binds Smad4 as well, thus competing with Smad2/3 and counteracting the drift towards muscle atrophy [109].

4.2. Relevance of Pro-Inflammatory Cytokines

The pro-inflammatory cytokines can influence skeletal muscle homeostasis positively or negatively, depending on the level of cytokine expression, the type of producing cells and the type of signalling pathway(s) activated. Generally speaking, if cytokines are produced by the muscle itself in an autocrine manner, their levels are generally low and positively impinge on myogenic differentiation. If cytokines are released by infiltrating immune cells at skeletal muscle injury sites, muscle repair and regeneration are facilitated [113]. As an example, TNFα, the main mediator of the initial inflammatory response during skeletal muscle regeneration, was shown to promote C2C12 myoblast proliferation by modulating Myf5, and by upregulating myostatin mRNA, to reduce myoblast differentiation, suppressing premature differentiation after muscle injury [113]. Furthermore, TNF-α increased the expression of chemokines and cytokines, which influence immune cell recruitment, inflammation, and tissue regeneration [114].
IL-6 also, at low concentrations, exerts a pro-myogenic function. During load-induced compensatory hypertrophy, the IL-6 expression increased in both myogenic progenitors and myofibers [115,116], while the myogenesis-associated skeletal muscle hypertrophy was impaired in mice genetically lacking IL-6 [115]. Consistently, both IL-6 and the closely related cytokine LIF were expressed during skeletal muscle regeneration after acute injury [117,118,119,120]. Similarly to TNFα and IL-6, IFN-γ was reported to exert a positive function in regenerating muscles upon injury. Indeed, administration of an IFN-γ receptor-blocking antibody or deletion of IFN-γ in KO mice impaired muscle regeneration [121].
When cytokines are produced by activated immune cells or by damaged muscle, as in the case of dystrophy, their concentrations in the bloodstream are usually high, resulting in potent inhibition of myogenic differentiation. Along this line, the regenerative potential of satellite cells in dystrophic muscles was inhibited by elevated levels of TNF-α and by over-activation of NF-κB [122]. IL-6 is over-expressed in the muscle of patients affected by Duchenne muscular dystrophy and in mdx dystrophic mice. In the latter, treatment with antibodies against the IL-6 receptor was shown to enhance muscle regeneration, improving the dystrophic phenotype [123]. Furthermore, a reduction in the IL-6 levels was associated with attenuated muscle damage in adult mdx mice [124]. Inhibition or rescue of muscle regeneration was also reported in mice exposed to exogenous IFN-γ or to anti-IFN-γ antibodies, respectively [125]. Consistently, C2C12 myoblast proliferation and fusion were inhibited by exposure to an antibody against the IFN-γ receptor, thus favouring myogenic differentiation [121], which was inhibited by IFN-γ in both murine and human myoblasts cultures [126,127]. In the skeletal muscle, pro-inflammatory cytokines can modulate the balance between protein synthesis and degradation. Several pathologies, such as cancer, chronic heart failure, diabetes mellitus and sepsis, are associated with increased levels of pro-inflammatory cytokines, which could activate protein catabolism [128,129]. The imbalance towards protein degradation causes a loss of muscle mass, which a frequent occurrence of chronic diseases, including cancer. In this regard, cancer-induced muscle wasting is one of the most relevant features of cachexia, a complex syndrome that frequently complicates patient management.

4.3. Cancer Cachexia

Cancer cachexia is a multifactorial syndrome characterized by progressive body weight loss, reduction of muscle mass and adipose tissue, and metabolic alterations, which negatively impinges on patient prognosis, quality of life, tolerance to chemotherapy and survival [130,131]. In patients with cancer, cachexia develops differently, depending on the type of tumour [132,133,134]. Lung, liver and gastrointestinal tumours have a very high incidence of cachexia (between 70 and 90%) compared to leukaemia, breast cancer and favourable non-Hodgkin’s lymphoma (about 30%) [131,133]. The occurrence of cachexia in cancer patients increases with age, advanced Classification of Malignant Tumours (TNM) stage and sex, with males being more affected than females [133]. At present, cachexia is an unmet medical need and there are no effective treatments available. Along this line, investigations aimed at defining new suitable therapeutic approaches based on molecular evidences are warranted.
Skeletal muscle wasting featuring cancer cachexia was demonstrated to result from enhanced rates of protein degradation, mainly operationalized by the ubiquitin–proteasome and autophagic–lysosomal proteolytic systems, which is frequently associated with altered rates of protein [135]. The modulations of protein turnover were proposed to result, partially at least, from impaired energy metabolism. Indeed, the presence of a tumour, which competes with the host organism for energy and substrates to sustain its own metabolism, increases the patient’s energy expenditure [136,137]. In addition, it is now well accepted that cancer-induced muscle wasting is also characterized by altered mitochondrial function, resulting in reduced energy production and increased ROS levels [138].
An important contribution to the development of cachexia is made by pro-inflammatory cytokines such as TNF-α, IL-6, LIF and IFN-γ, produced by both the host and the tumour [139]. Indeed, cytokines are well known to promote muscle atrophy by NF-κB activation and by suppressing the Akt/mTOR pathway, resulting in enhanced protein breakdown, frequently associated with reduced protein synthesis rates [136]. Furthermore, cytokines act at the central level as well, promoting anorexia and the release of corticosteroids, which are well known to induce both protein and fat breakdown [136,140].

4.4. Impact of EVOO on Skeletal Muscle Homeostasis

Few in vitro and in vivo studies have analysed the mechanisms by which EVOO’s phenolic derivatives impinge on skeletal muscle homeostasis. For example, OLE counteracted the ROS increase caused in C2C12 myotubes by H2O2 and reduced the mitochondrial ROS generation in chicken muscle cells modulating the expression of Sirt1 and PGC1-α, with the latter being the master regulator of mitochondrial biogenesis [141,142]. HT administration to obese mice or to rats practising strenuous exercise was shown to improve the muscle mass and function by acting as an ROS scavenger, enhancing the activity of the endogenous antioxidant systems and stimulating mitochondrial biogenesis [143,144]. More specifically, the induction of mitochondrial biogenesis by EVOO was associated with enhanced activation of the signalling pathway dependent on AMP-activated protein kinase (AMPK), which triggers the overexpression of genes such as PGC1-α, NRF-1 and TFAM [145]. Along the same line, EVOO supplementation in animals exposed to a high-fat diet resulted in increased levels of the markers of autophagy, associated with a reduced pFOXO3/FOXO ratio [146]. Another study showed that both FOXO1 upregulation and the reduction of mTOR pathway activity were counteracted in diabetic pregnant rats consuming an EVOO-enriched diet [147].
Most studies have demonstrated the effectiveness of EVOO consumption in counteracting sarcopenia in the elderly. In particular, EVOO intake was proposed to activate anabolic pathways and to prevent mitochondrial damage and inflammatory processes responsible for sarcopenia [148,149].
Despite the observations reported above, there are still very few studies aimed at understanding if EVOO and its derivatives can counteract muscle atrophy. In this regard, experiments performed on C2C12 cells cultured in high glucose medium, which mimics the persistent hyperglucose environment of diabetes, showed that exposure to tyrosol protected myoblasts by reducing ROS production, resulting in increased cell proliferation, suppressed apoptosis, and restored ability to release angiogenic factors into the culture medium. Consistently, hindlimb ischemia in diabetic mice could be improved by recovering blood perfusion through tyrosol injection into the gastrocnemius muscle [150].
Few pieces of evidence are actually available concerning the possibility that the EVOO-enriched Mediterranean diet or EVOO components, known to exert a protective action against cancer both in vitro and in vivo, could also positively impinge on cancer-induced skeletal muscle wasting (Figure 3). Until now, only three clinical studies have analysed the correlation between the Mediterranean diet and cancer cachexia. In a study conducted in prostate cancer patients exposed to androgen deprivation therapy, in which obesity was a treatment-induced negative side effect, Baguley et al. [151] showed that those adhering to the Mediterranean diet experienced improvements in their quality of life and fatigue, as well as a reduction in their total body mass, fat mass and IL-8 levels, compared to patients on the standard diet.
Another study analysed the effects of a Mediterranean diet in patients affected by lung cancer, showing that it was able to reduce both the inflammation index and the C-reactive protein blood concentrations [152]. Finally, Bagheri et al. [153] compared two groups of CRC patients with cachexia, one given the Mediterranean diet and one fed their normal diet (controls). The loss of body weight, adipose tissue, lean body mass and muscle function were significantly improved in patients fed the Mediterranean diet compared to the control group. The circulating levels of TNF-α, C-reactive protein, and IL-6 were considerably reduced in the Mediterranean diet group, with an improvement in the general health status and exercise performance [153]. Recently, OC was reported to protect C2C12 myotubes against the reduction in size deriving from exposure to TNF-α or to the medium conditioned by cultured C26 cells (CM-C26), a tumour well known to induce cachexia in the host mice. Indeed, when added to TNF-α- or CM-C26-treated C2C12 cultures, OC restored the myotube morphology and size, also normalizing the expression of the accepted markers of protein degradation atrogin-1 and MuRF1. Furthermore, OC positively modulated the expression of Pax7, myogenin, and MyHC, three molecules involved in myogenesis [154]. Consistently, in a previous study, tyrosol was shown to counteract the negative effects exerted by dexamethasone in C2C12 myotubes. Indeed, morpho-functional analyses revealed that dexamethasone reduced the myotube size and induced an immature syncytia phenotype compared to the control cultures. Furthermore, the presence of dysfunctional mitochondria and the accumulation of autophagic vacuoles contributed to myotube degeneration and death. Administering tyrosol prior to glucocorticoid treatment mitigated the myotube damage and restored both mitochondrial and lysosomal function [155].

5. Conclusions and Future Perspectives

Numerous epidemiological studies have highlighted the health benefits of EVOO. Due to its antioxidant and anti-inflammatory properties, EVOO plays a crucial role in protecting against inflammatory diseases, atherosclerosis, neurodegenerative disorders, and cancer. In the present review, the potential interactions among EVOO, pro-inflammatory cytokines, cancer, and cachexia are highlighted. In this context, EVOO can help in reducing inflammation and oxidative stress, thereby acting on one side on cancer onset and progression, while on the other side, it could protect the skeletal muscle tissue, preserving its mass and function.
However, most of the available data come from in vitro reports, with very few studies performed on preclinical models or human beings. This is particularly relevant when taking into account issues such as EVOO’s polyphenol concentration, absorption, bioavailability and metabolism. In this regard, it must be considered that many of the bioactivities exerted in vitro by EVOO derivatives are not confirmed by in vivo experimental and clinical studies. Several factors could account for such a discrepancy: (i) the concentrations used in vitro are generally higher than those occurring in the human diet; and (ii) the amount of the bioactive compound that can reach the circulation can be markedly lower than expected. As an example, the concentration of non-metabolized hydroxytyrosol reaching the bloodstream is less than 1% of the amount introduced with the diet [156]. On the other side, the concentrations of metabolites derived from hydroxytyrosol and tyrosol can become markedly high, and they can share the biological effects with the original compounds, being able to modulate the intracellular signalling pathways that account for the effects of EVOO derivatives [157]. Just as another example, a high polyphenol dosage could result in pro-oxidant rather than antioxidant action, leading to altered mitochondrial function and enhanced mutation rates [158]. Finally, gender differences in terms of metabolism, the metabolic response to EVOO derivatives and the extent of the biological effects, as well as polymorphisms in the drug-metabolizing systems, are additional issues to be considered when approaching the use of EVOO derivatives in vivo. An additional note is that the effects of chronic assumption of EVOO polyphenols should be investigated.
Keeping in mind the potential limitations above, the existing knowledge suggests that EVOO might be a nutraceutical intervention useful for contributing to cancer prevention and to the management of the associated cachexia. Indeed, by safeguarding muscle, EVOO may lower the risk of cancer cachexia and assist patients in better tolerating anti-tumour therapies, ultimately enhancing their quality of life. Such a hypothesis is supported by studies showing that the combination of supplements such as essential amino acids, antioxidants, or other agents that promote muscle protein synthesis and reduce inflammation may be more effective than conventional treatments (chemotherapy and dietary interventions). For example, an association treatment for cancer cachexia, which included a polyphenol-enriched diet, antioxidants, medroxyprogesterone acetate, and celecoxib, was shown to be effective and safe in an early phase II study [159].
In recent decades, nanotechnology has become increasingly vital in improving the solubility, bioavailability, permeability, and stability of natural compounds and extracts [160]. Polymeric micelles have been developed to improve the intestinal permeability of OLE [161]. Encapsulating in polymeric micelles both hydrophobic and hydrophilic natural compounds prolongs the circulation time in the bloodstream and protects against chemical and enzymatic breakdown in the gastrointestinal tract, ultimately increasing their ability to cross the intestinal barrier, and reducing oral doses and potential toxicity.
On the whole, despite the still unclear issues above, the integration of EVOO into the diet, or the fortification of foods with EVOO polyphenols, could reveal a useful strategy for improving the management of cancer patients. Along this line, both preclinical and clinical studies are warranted.

Author Contributions

Writing—original draft preparation, D.D.S.; writing—review and editing, D.D.S. and P.C.; project administration, P.C.; funding acquisition, P.C. All authors have read and agreed to the published version of the manuscript.

Funding

This publication is part of the project NODES, which has received funding from the MUR–M4C2 1.5 of PNRR (grant agreement no. ECS00000036). It was also supported by the University of Torino, Italy (local research grants to P.C.) and by the Italian Ministry of University and Research, PRIN project (to P.C.).

Conflicts of Interest

On behalf of all the authors, the corresponding author states that there are no conflicts of interest. The authors declare that this study received funding from MUR–M4C2 1.5 of PNRR. The funder was not involved in the study design, collection, analysis, interpretation of data, the writing of this article or the decision to submit it for publication.

References

  1. Jimenez-Lopez, C.; Carpena, M.; Lourenço-Lopes, C.; Gallardo-Gomez, M.; Lorenzo, J.M.; Barba, F.J.; Prieto, M.A.; Simal-Gandara, J. Bioactive Compounds and Quality of Extra Virgin Olive Oil. Foods 2020, 9, 1014. [Google Scholar] [CrossRef] [PubMed]
  2. Wongwarawipat, T.; Papageorgiou, N.; Bertsias, D.; Siasos, G.; Tousoulis, D. Olive Oil-related Anti-inflammatory Effects on Atherosclerosis: Potential Clinical Implications. Endocr. Metab. Immune Disord. Drug Targets 2018, 18, 51–62. [Google Scholar] [CrossRef] [PubMed]
  3. Widmer, R.J.; Flammer, A.J.; Lerman, L.O.; Lerman, A. The Mediterranean diet, its components, and cardiovascular disease. Am. J. Med. 2015, 128, 229–238. [Google Scholar] [CrossRef] [PubMed]
  4. McEvoy, C.T.; Guyer, H.; Langa, K.M.; Yaffe, K. Neuroprotective diets are associated with better cognitive function: The Health and Retirement Study. J. Am. Geriatr. Soc. 2017, 65, 1857–1862. [Google Scholar] [CrossRef] [PubMed]
  5. Franco, G.A.; Interdonato, L.; Cordaro, M.; Cuzzocrea, S.; Di Paola, R. Bioactive compounds of the Mediterranean diet as nutritional support to fight neurodegenerative disease. Int. J. Mol. Sci. 2023, 24, 7318. [Google Scholar] [CrossRef] [PubMed]
  6. Di Daniele, N.; Noce, A.; Vidiri, M.F.; Moriconi, E.; Marrone, G.; Annicchiarico-Petruzzelli, M.; D’Urso, G.; Tesauro, M.; Rovella, V.; De Lorenzo, A. Impact of Mediterranean diet on metabolic syndrome, cancer and longevity. Oncotarget 2017, 8, 8947–8979. [Google Scholar] [CrossRef] [PubMed]
  7. González-Rodríguez, M.; Ait Edjoudi, D.; Cordero-Barreal, A.; Farrag, M.; Varela-García, M.; Torrijos-Pulpón, C.; Ruiz-Fernández, C.; Capuozzo, M.; Ottaiano, A.; Lago, F.; et al. Oleocanthal, an Antioxidant Phenolic Compound in Extra Virgin Olive Oil (EVOO): A Comprehensive Systematic Review of Its Potential in Inflammation and Cancer. Antioxidants 2023, 14, 2112. [Google Scholar] [CrossRef] [PubMed]
  8. Velotti, F.; Bernini, R. Expand Hydroxytyrosol Interference with Inflammaging via Modulation of Inflammation and Autophagy. Nutrients 2023, 5, 1774. [Google Scholar] [CrossRef] [PubMed]
  9. Rufino-Palomares, E.E.; Pérez-Jiménez, A.; García-Salguero, L.; Mokhtari, K.; Reyes-Zurita, F.J.; Peragón-Sánchez, J.; Lupiáñez, J.A. Nutraceutical Role of Polyphenols and Triterpenes Present in the Extracts of Fruits and Leaves of Olea europaea as Antioxidants, Anti-Infectives and Anticancer Agents on Healthy Growth. Molecules 2022, 27, 2341. [Google Scholar] [CrossRef] [PubMed]
  10. Del Rio, D.; Rodriguez-Mateos, A.; Spencer, J.P.; Tognolini, M.; Borges, G.; Crozier, A. Dietary (Poly)phenolics in Human Health: Structures, Bioavailability, and Evidence of Protective Effects Against Chronic Diseases. Antioxid. Redox Signal. 2013, 18, 1818–1832. [Google Scholar] [CrossRef] [PubMed]
  11. de Bock, M.; Thorstensen, E.B.; Derraik, J.G.; Henderson, H.V.; Hofman, P.L.; Cutfield, W.S. Human Absorption and Metabolism of Oleuropein and Hydroxytyrosol Ingested as Olive (Olea europaea L.) Leaf Extract. Mol. Nutr. Food Res. 2013, 57, 2079–2085. [Google Scholar] [CrossRef] [PubMed]
  12. Pereira-Caro, G.; Sarriá, B.; Madrona, A.; Espartero, J.L.; Escuderos, M.E.; Bravo, L.; Mateos, R. Digestive Stability of Hydroxytyrosol, Hydroxytyrosyl Acetate and Alkyl Hydroxytyrosyl Ethers. Int. J. Food Sci. Nutr. 2012, 63, 703–707. [Google Scholar] [CrossRef] [PubMed]
  13. Pinto, J.; Paiva-Martins, F.; Corona, G.; Debnam, E.S.; Jose Oruna-Concha, M.; Vauzour, D.; Gordon, M.H.; Spencer, J.P. Absorption and metabolism of olive oil secoiridoids in the small intestine. Br. J. Nutr. 2011, 105, 1607–1618. [Google Scholar] [CrossRef] [PubMed]
  14. Cicerale, S.; Lucas, L.; Keast, R. Biological activities of phenolic compounds present in virgin olive oil. Int. J. Mol. Sci. 2010, 11, 458–479. [Google Scholar] [CrossRef] [PubMed]
  15. Salas-Salvadó, J.; Bulló, M.; Babio, N.; Martínez-González, M.Á.; Ibarrola-Jurado, N.; Basora, J.; Estruch, R.; Covas, M.I.; Corella, D.; Arós, F.; et al. Reduction in the incidence of type 2 diabetes with the Mediterranean diet: Results of the PREDIMED-Reus nutrition intervention randomized trial. Diabetes Care 2011, 34, 14–19. [Google Scholar] [CrossRef] [PubMed]
  16. Estruch, R.; Ros, E.; Salas-Salvadó, J.; Covas, M.I.; Corella, D.; Arós, F.; Gómez-Gracia, E.; Ruiz-Gutiérrez, V.; Fiol, M.; Lapetra, J.; et al. Primary Prevention of Cardiovascular Disease with a Mediterranean Diet Supplemented with Extra-Virgin Olive Oil or Nuts. N. Engl. J. Med. 2018, 378, e34. [Google Scholar] [CrossRef] [PubMed]
  17. Patti, A.M.; Carruba, G.; Cicero, A.F.G.; Banach, M.; Nikolic, D.; Giglio, R.V.; Terranova, A.; Soresi, M.; Giannitrapani, L.; Montalto, G.; et al. Daily Use of Extra Virgin Olive Oil with High Oleocanthal Concentration Reduced Body Weight, Waist Circumference, Alanine Transaminase, Inflammatory Cytokines and Hepatic Steatosis in Subjects with the Metabolic Syndrome: A 2-Month Intervention Study. Metabolites 2020, 10, 392. [Google Scholar] [CrossRef] [PubMed]
  18. Ruiz-García, I.; Ortíz-Flores, R.; Badía, R.; García-Borrego, A.; García-Fernández, M.; Lara, E.; Martín-Montañez, E.; García-Serrano, S.; Valdés, S.; Gonzalo, M.; et al. Rich oleocanthal and oleacein extra virgin olive oil and inflammatory and antioxidant status in people with obesity and pre-diabetes. The APRIL study: A randomised, controlled crossover study. Clin. Nutr. 2023, 42, 1389–1398. [Google Scholar] [CrossRef] [PubMed]
  19. Soto-Alarcon, S.A.; Valenzuela, R.; Valenzuela, A.; Videla, L.A. Liver protective effects of extra virgin olive oil: Interaction between its chemical composition and the cell-signaling pathways involved in protection. Endocr. Metab. Immune Disord. Drug Targets 2018, 18, 75–84. [Google Scholar] [CrossRef] [PubMed]
  20. De Stefanis, D.; Scimè, S.; Accomazzo, S.; Catti, A.; Occhipinti, A.; Bertea, C.M.; Costelli, P. Anti-Proliferative Effects of an Extra-Virgin Olive Oil Extract Enriched in Ligstroside Aglycone and Oleocanthal on Human Liver Cancer Cell Lines. Cancers 2019, 11, 1640. [Google Scholar] [CrossRef] [PubMed]
  21. Fogli, S.; Arena, C.; Carpi, S.; Polini, B.; Bertini, S.; Digiacomo, M.; Gado, F.; Saba, A.; Saccomanni, G.; Breschi, M.C.; et al. Cytotoxic Activity of Oleocanthal Isolated from Virgin Olive Oil on Human Melanoma Cells. Nutr. Cancer 2016, 68, 873–877. [Google Scholar] [CrossRef] [PubMed]
  22. Memmola, R.; Petrillo, A.; Di Lorenzo, S.; Altuna, S.C.; Habeeb, B.S.; Soggiu, A.; Bonizzi, L.; Garrone, O.; Ghidini, M. Correlation between Olive Oil Intake and Gut Microbiota in Colorectal Cancer Prevention. Nutrients 2022, 14, 3749. [Google Scholar] [CrossRef] [PubMed]
  23. Deiana, M.; Serra, G.; Corona, G. Modulation of intestinal epithelium homeostasis by extra virgin olive oil phenolic compounds. Food Funct. 2018, 9, 4085–4099. [Google Scholar] [CrossRef] [PubMed]
  24. Casas, R.; Urpi-Sardà, M.; Sacanella, E.; Arranz, S.; Corella, D.; Castañer, O.; Lamuela-Raventós, R.M.; Salas-Salvadó, J.; Lapetra, J.; Portillo, M.P.; et al. Anti-inflammatory effects of the Mediterranean diet in the early and late stages of atheroma plaque development. Mediat. Inflamm. 2017, 2017, 3674390. [Google Scholar] [CrossRef] [PubMed]
  25. Hara, T.; Fukuda, D.; Ganbaatar, B.; Pham, P.T.; Aini, K.; Rahadian, A.; Suto, K.; Yagi, S.; Kusunose, K.; Yamada, H.; et al. Olive mill wastewater and hydroxytyrosol inhibit atherogenesis in apolipoprotein E-deficient mice. Heart Vessel. 2023, 38, 1386–1394. [Google Scholar] [CrossRef] [PubMed]
  26. Terés, S.; Barceló-Coblijn, G.; Benet, M.; Alvarez, R.; Bressani, R.; Halver, J.E.; Escribá, P.V. Oleic acid content is responsible for the reduction in blood pressure induced by olive oil. Proc. Natl. Acad. Sci. USA 2008, 105, 13811–13816. [Google Scholar] [CrossRef] [PubMed]
  27. Martín-Peláez, S.; Castañer, O.; Konstantinidou, V.; Subirana, I.; Muñoz-Aguayo, D.; Blanchart, G.; Gaixas, S.; de la Torre, R.; Farré, M.; Sáez, G.T.; et al. Effect of olive oil phenolic compounds on the expression of blood pressure-related genes in healthy individuals. Eur. J. Nutr. 2017, 56, 663–670. [Google Scholar] [CrossRef] [PubMed]
  28. Zou, X.; Feng, Z.; Li, Y.; Wang, Y.; Wertz, K.; Weber, P.; Fu, Y.; Liu, J. Stimulation of GSH synthesis to prevent oxidative stress-induced apoptosis by hydroxytyrosol in human retinal pigment epithelial cells: Activation of Nrf2 and JNK-p62/SQSTM1 pathways. J. Nutr. Biochem. 2011, 23, 994–1006. [Google Scholar] [CrossRef] [PubMed]
  29. Gonçalves, M.; Vale, N.; Silva, P. Neuroprotective effects of olive oil: A comprehensive review of antioxidant properties. Antioxidants 2024, 13, 762. [Google Scholar] [CrossRef] [PubMed]
  30. de la Torre-Carbot, K.; Chávez-Servín, J.L.; Jaúregui, O.; Castellote, A.I.; Lamuela-Raventós, R.M.; Nurmi, T.; Poulsen, H.E.; Gaddi, A.V.; Kaikkonen, J.; Zunft, H.F.; et al. Elevated circulating LDL phenol levels in men who consumed virgin rather than refined olive oil are associated with less oxidation of plasma LDL. J. Nutr. 2010, 140, 501–508. [Google Scholar] [CrossRef] [PubMed]
  31. Covas, M.I.; Nyyssönen, K.; Poulsen, H.E.; Kaikkonen, J.; Zunft, H.J.; Kiesewetter, H.; Gaddi, A.; de la Torre, R.; Mursu, J.; Bäumler, H.; et al. The effect of polyphenols in olive oil on heart disease risk factors: A randomized trial. Ann. Intern. Med. 2006, 145, 333–341. [Google Scholar] [CrossRef] [PubMed]
  32. Yubero-Serrano, E.M.; Lopez-Moreno, J.; Gomez-Delgado, F.; Lopez-Miranda, J. Extra virgin olive oil: More than a healthy fat. Eur. J. Clin. Nutr. 2019, 72, 8–17. [Google Scholar] [CrossRef] [PubMed]
  33. Cabrerizo, S.; De La Cruz, J.P.; López-Villodres, J.A.; Muñoz-Marín, J.; Guerrero, A.; Reyes, J.J.; Labajos, M.T.; González-Correa, J.A. Role of the inhibition of oxidative stress and inflammatory mediators in the neuroprotective effects of hydroxytyrosol in rat brain slices subjected to hypoxia reoxygenation. J. Nutr. Biochem. 2013, 24, 2152–2157. [Google Scholar] [CrossRef] [PubMed]
  34. De La Cruz, J.P.; Ruiz-Moreno, M.I.; Guerrero, A.; Reyes, J.J.; Benitez-Guerrero, A.; Espartero, J.L.; González-Correa, J.A. Differences in the neuroprotective effect of orally administered virgin olive oil (Olea europaea) polyphenols tyrosol and hydroxytyrosol in rats. J. Agric. Food Chem. 2015, 63, 5957–5963. [Google Scholar] [CrossRef] [PubMed]
  35. Fabiani, R.; Sepporta, M.V.; Rosignoli, P.; De Bartolomeo, A.; Crescimanno, M.; Morozzi, G. Anti-proliferative and pro-apoptotic activities of hydroxytyrosol on different tumour cells: The role of extracellular production of hydrogen peroxide. Eur. J. Nutr. 2012, 51, 455–464. [Google Scholar] [CrossRef] [PubMed]
  36. O’Dowd, Y.; Driss, F.; Dang, P.M.; Elbim, C.; Gougerot-Pocidalo, M.A.; Pasquier, C.; El-Benna, J. Antioxidant effect of hydroxytyrosol, a polyphenol from olive oil: Scavenging of hydrogen peroxide but not superoxide anion produced by human neutrophils. Biochem. Pharmacol. 2004, 68, 2003–2008. [Google Scholar] [CrossRef] [PubMed]
  37. Zrelli, H.; Matsuoka, M.; Kitazaki, S.; Araki, M.; Kusunoki, M.; Zarrouk, M.; Miyazaki, H. Hydroxytyrosol induces proliferation and cytoprotection against oxidative injury in vascular endothelial cells: Role of Nrf2 activation and HO-1 induction. J. Agric. Food Chem. 2011, 59, 4473–4482. [Google Scholar] [CrossRef] [PubMed]
  38. Calahorra, J.; Shenk, J.; Wielenga, V.H.; Verweij, V.; Geenen, B.; Dederen, P.J.; Peinado, M.A.; Siles, E.; Wiesmann, M.; Kiliaan, A.J. Hydroxytyrosol, the Major Phenolic Compound of Olive Oil, as an Acute Therapeutic Strategy after Ischemic Stroke. Nutrients 2019, 11, 2430. [Google Scholar] [CrossRef] [PubMed]
  39. Barca, C.; Wiesmann, M.; Calahorra, J.; Wachsmuth, L.; Doring, C.; Foray, C.; Heiradi, A.; Hermann, S.; Peinado, M.A.; Siles, E.; et al. Impact of hydroxytyrosol on stroke: Tracking therapy response on neuroinflammation and cerebrovascular parameters using PET-MR imaging and on functional outcomes. Theranostics 2021, 11, 4030–4049. [Google Scholar] [CrossRef] [PubMed]
  40. Fabiani, R.; Vella, N.; Rosignoli, P. Epigenetic modifications induced by olive oil and its phenolic compounds: A systematic review. Molecules 2021, 26, 273. [Google Scholar] [CrossRef] [PubMed]
  41. Del Saz-Lara, A.; López de Las Hazas, M.C.; Visioli, F.; Dávalos, A. Nutri-epigenetic effects of phenolic compounds from extra virgin olive oil: A systematic review. Adv. Nutr. 2022, 13, 2039–2060. [Google Scholar] [CrossRef] [PubMed]
  42. Casadesús, J.; Noyer-Weidner, M. Epigenetics. Brenner’s Encyclopedia of Genetics, 2nd ed.; Academic Press: San Diego, CA, USA, 2013. [Google Scholar]
  43. Arpón, A.; Riezu-Boj, J.I.; Milagro, F.I.; Marti, A.; Razquin, C.; Martínez-González, M.A.; Corella, D.; Estruch, R.; Casas, R.; Fitó, M.; et al. Adherence to Mediterranean diet is associated with methylation changes in inflammation-related genes in peripheral blood cells. J. Physiol. Biochem. 2016, 73, 445–455. [Google Scholar] [CrossRef] [PubMed]
  44. ArunSundar, M.; Shanmugarajan, T.S.; Ravichandiran, V. 3, 4-Dihydroxyphenylethanol assuages cognitive impulsivity in Alzheimer’s disease by attuning HPA-axis via differential crosstalk of α7 nAChR with microRNA-124 and HDAC6. ACS Chem. Neurosci. 2018, 9, 2904–2916. [Google Scholar] [CrossRef] [PubMed]
  45. D’Adamo, S.; Cetrullo, S.; Borzì, R.M.; Flamigni, F. Effect of oxidative stress and 3-hydroxytyrosol on DNA methylation levels of miR-9 promoters. J. Cell. Mol. Med. 2019, 23, 7885–7889. [Google Scholar] [CrossRef] [PubMed]
  46. D’Adamo, S.; Cetrullo, S.; Guidotti, S.; Borzì, R.M.; Flamigni, F. Hydroxytyrosol modulates the levels of microRNA-9 and its target sirtuin-1 thereby counteracting oxidative stress-induced chondrocyte death. Osteoarthr. Cartil. 2017, 25, 600–610. [Google Scholar] [CrossRef] [PubMed]
  47. D’Amore, S.; Vacca, M.; Cariello, M.; Graziano, G.; D’Orazio, A.; Salvia, R.; Sasso, R.C.; Sabbà, C.; Palasciano, G.; Moschetta, A. Genes and miRNA expression signatures in peripheral blood mononuclear cells in healthy subjects and patients with metabolic syndrome after acute intake of extra virgin olive oil. Biochim. Biophys. Acta 2016, 1861, 1671–1680. [Google Scholar] [CrossRef] [PubMed]
  48. Carpi, S.; Scoditti, E.; Massaro, M.; Polini, B.; Manera, C.; Digiacomo, M.; Esposito Salsano, J.; Poli, G.; Tuccinardi, T.; Doccini, S.; et al. The extra-virgin olive oil polyphenols oleocanthal and oleacein counteract inflammation-related gene and miRNA expression in adipocytes by attenuating NF-κB activation. Nutrients 2019, 11, 2855. [Google Scholar] [CrossRef] [PubMed]
  49. Hibino, S.; Kawazoe, T.; Kasahara, H.; Itoh, S.; Ishimoto, T.; Sakata-Yanagimoto, M.; Taniguchi, K. Inflammation-induced tumorigenesis and metastasis. Int. J. Mol. Sci. 2021, 22, 5421. [Google Scholar] [CrossRef] [PubMed]
  50. Tripathi, S.; Sharma, Y.; Kumar, D. Unveiling the link between chronic inflammation and cancer. Metabol. Open 2025, 25, 100347. [Google Scholar] [CrossRef] [PubMed]
  51. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef] [PubMed]
  52. Rodríguez-García, C.; Sánchez-Quesada, C.; Algarra, I.; Gaforio, J.J. The High-Fat Diet Based on Extra-Virgin Olive Oil Causes Dysbiosis Linked to Colorectal Cancer Prevention. Nutrients 2020, 12, 1705. [Google Scholar] [CrossRef] [PubMed]
  53. Pei, T.; Meng, Q.; Han, J.; Sun, H.; Li, L.; Song, R.; Sun, B.; Pan, S.; Liang, D.; Liu, L. (-)-Oleocanthal inhibits growth and metastasis by blocking activation of STAT3 in human hepatocellular carcinoma. Oncotarget 2016, 7, 43475–43491. [Google Scholar] [CrossRef] [PubMed]
  54. Garcia-Guasch, M.; Medrano, M.; Costa, I.; Vela, E.; Grau, M.; Escrich, E.; Moral, R. Extra-Virgin Olive Oil and Its Minor Compounds Influence Apoptosis in Experimental Mammary Tumors and Human Breast Cancer Cell Lines. Cancers 2022, 14, 905. [Google Scholar] [CrossRef] [PubMed]
  55. Calabriso, N.; Massaro, M.; Scoditti, E.; D’Amore, S.; Gnoni, A.; Pellegrino, M.; Storelli, C.; De Caterina, R.; Palasciano, G.; Carluccio, M.A. Extra virgin olive oil rich in polyphenols modulates VEGF-induced angiogenic responses by preventing NADPH oxidase activity and expression. J. Nutr. Biochem. 2016, 28, 19–29. [Google Scholar] [CrossRef] [PubMed]
  56. Scoditti, E.; Calabriso, N.; Massaro, M.; Pellegrino, M.; Storelli, C.; Martines, G.; De Caterina, R.; Carluccio, M.A. Mediterranean diet polyphenols reduce inflammatory angiogenesis through MMP-9 and COX-2 inhibition in human vascular endothelial cells: A potentially protective mechanism in atherosclerotic vascular disease and cancer. Arch. Biochem. Biophys. 2012, 527, 81–89. [Google Scholar] [CrossRef] [PubMed]
  57. Marrero, A.D.; Ortega-Vidal, J.; Salido, S.; Castilla, L.; Vidal, I.; Quesada, A.R.; Altarejos, J.; Martínez-Poveda, B.; Medina, M.A. Anti-angiogenic effects of oleacein and oleocanthal: New bioactivities of compounds from extra virgin olive oil. Biomed. Pharmacother. 2023, 165, 115234. [Google Scholar] [CrossRef] [PubMed]
  58. Marrero, A.D.; Cárdenas, C.; Castilla, L.; Ortega-Vidal, J.; Quesada, A.R.; Martínez-Poveda, B.; Medina, M.A. Antiangiogenic potential of an olive oil extract: Insights from a proteomic study. J. Agric. Food Chem. 2024, 72, 13023–13038. [Google Scholar] [CrossRef] [PubMed]
  59. Rodríguez-Miguel, C.; Moral, R.; Escrich, R.; Vela, E.; Solanas, M.; Escrich, E. The role of dietary extra virgin olive oil and corn oil on the alteration of epigenetic patterns in the rat DMBA-induced breast cancer model. PLoS ONE 2015, 10, e0138980. [Google Scholar] [CrossRef] [PubMed]
  60. Govindarajah, V.; Leung, Y.K.; Ying, J.; Gear, R.; Bornschein, R.L.; Medvedovic, M.; Ho, S.M. In utero exposure of rats to high-fat diets perturbs gene expression profiles and cancer susceptibility of prepubertal mammary glands. J. Nutr. Biochem. 2016, 29, 73–82. [Google Scholar] [CrossRef] [PubMed]
  61. Di Francesco, A.; Falconi, A.; Di Germanio, C.; Micioni Di Bonaventura, M.V.; Costa, A.; Caramuta, S.; Del Carlo, M.; Compagnone, D.; Dainese, E.; Cifani, C.; et al. Extra-virgin olive oil up-regulates CB1 tumor suppressor gene in human colon cancer cells and in rat colon via epigenetic mechanisms. J. Nutr. Biochem. 2015, 26, 250–258. [Google Scholar] [CrossRef] [PubMed]
  62. Nanda, N.; Mahmood, S.; Bhatia, A.; Mahmood, A.; Dhawan, D.K. Chemopreventive role of olive oil in colon carcinogenesis by targeting noncoding RNAs and methylation machinery. Int. J. Cancer 2019, 144, 1180–1194. [Google Scholar] [CrossRef] [PubMed]
  63. Asgharzadeh, S.; Sheikhshabani, S.H.; Ghasempour, E.; Heidari, R.; Rahmati, S.; Mohammadi, M.; Jazaeri, A.; Amini-Farsani, Z. The effect of oleuropein on apoptotic pathway regulators in breast cancer cells. Eur. J. Pharmacol. 2020, 886, 173509. [Google Scholar] [CrossRef] [PubMed]
  64. 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] [PubMed]
  65. Hamed, M.M.; Handoussa, H.; Hussein, N.H.; Eissa, R.A.; Abdel-Aal, L.K.; El Tayebi, H.M. Oleuropein controls miR-194/XIST/PD-L1 loop in triple negative breast cancer: New role of nutri-epigenetics in immune-oncology. Life Sci. 2021, 277, 119353. [Google Scholar] [CrossRef] [PubMed]
  66. Cheng, J.S.; Chou, C.T.; Liu, Y.Y.; Sun, W.C.; Shieh, P.; Kuo, D.H.; Kuo, C.C.; Jan, C.R.; Liang, W.Z. The effect of oleuropein from olive leaf (Olea europaea) extract on Ca2+ homeostasis, cytotoxicity, cell cycle distribution and ROS signalling in HepG2 human hepatoma cells. Food Chem. Toxicol. 2016, 9, 151–166. [Google Scholar] [CrossRef] [PubMed]
  67. 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] [PubMed]
  68. Antognelli, C.; Frosini, R.; Santolla, M.F.; Peirce, M.J.; Talesa, V.N. Corrigendum to “Oleuropein-Induced Apoptosis Is Mediated by Mitochondrial Glyoxalase 2 in NSCLC A549 Cells: A Mechanistic Inside and a Possible Novel Nonenzymatic Role for an Ancient Enzyme”. Oxid. Med. Cell. Longev. 2020, 2020, 3045908. [Google Scholar] [CrossRef] [PubMed]
  69. 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]
  70. Cusimano, A.; Balasus, D.; Azzolina, A.; Augello, G.; Emma, M.R.; Di Sano, C.; Gramignoli, R.; Strom, S.C.; McCubrey, J.A.; Montalto, G.; et al. Oleocanthal exerts antitumor effects on human liver and colon cancer cells through ROS generation. Int. J. Oncol. 2017, 51, 533–544. [Google Scholar] [CrossRef] [PubMed]
  71. Ayoub, N.M.; Siddique, A.B.; Ebrahim, H.Y.; Mohyeldin, M.M.; El Sayed, K.A. The olive oil phenolic (-)-oleocanthal modulates estrogen receptor expression in luminal breast cancer in vitro and in vivo and synergizes with tamoxifen treatment. Eur. J. Pharmacol. 2017, 810, 100–111. [Google Scholar] [CrossRef] [PubMed]
  72. Siddique, A.B.; Kilgore, P.C.S.R.; Tajmim, A.; Singh, S.S.; Meyer, S.A.; Jois, S.D.; Cvek, U.; Trutschl, M.; Sayed, K.A.E. (-)-Oleocanthal as a dual c-MET-COX2 inhibitor for the control of lung cancer. Nutrients 2019, 11, 412. [Google Scholar] [CrossRef] [PubMed]
  73. El-Azem, N.; Pulido-Moran, M.; Ramirez-Tortosa, C.L.; Quiles, J.L.; Cara, F.E.; Sanchez-Rovira, P.; Granados-Principal, S.; Ramirez-Tortosa, M. Modulation by hydroxytyrosol of oxidative stress and antitumor activities of paclitaxel in breast cancer. Eur. J. Nutr. 2019, 58, 1203–1211. [Google Scholar] [CrossRef] [PubMed]
  74. 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] [PubMed]
  75. Menendez, J.A.; Vazquez-Martin, A.; Oliveras-Ferraros, C.; Garcia-Villalba, R.; Carrasco-Pancorbo, A.; Fernandez-Gutierrez, A.; Segura-Carretero, A. Effects of extra-virgin olive oil polyphenols on breast cancer-associated fatty acid synthase protein expression using reverse-phase protein microarrays. Int. J. Mol. Med. 2008, 22, 433–439. [Google Scholar] [CrossRef] [PubMed]
  76. Gu, Y.; Wang, J.; Peng, L. (-)-Oleocanthal exerts anti-melanoma activities and inhibits STAT3 signaling pathway. Oncol. Rep. 2016, 37, 483–491. [Google Scholar] [CrossRef] [PubMed]
  77. Zubair, H.; Bhardwaj, A.; Ahmad, A.; Srivastava, S.K.; Khan, M.A.; Patel, G.K.; Singh, S.; Singh, A.P. Hydroxytyrosol induces apoptosis and cell cycle arrest and suppresses multiple oncogenic signaling pathways in prostate cancer cells. Nutr. Cancer 2017, 69, 932–942. [Google Scholar] [CrossRef] [PubMed]
  78. Cani, P.D.; Jordan, B.F. Gut microbiota-mediated inflammation in obesity: A link with gastrointestinal cancer. Nat. Rev. Gastroenterol. Hepatol. 2018, 15, 671–682. [Google Scholar] [CrossRef] [PubMed]
  79. Rositch, A.F. Global burden of cancer attributable to infections: The critical role of implementation science. Lancet Glob. Health. 2020, 8, e153–e154. [Google Scholar] [CrossRef] [PubMed]
  80. Vrdoljak, J.; Kumric, M.; Vilovic, M.; Martinovic, D.; Tomic, I.J.; Krnic, M.; Ticinovic Kurir, T.; Bozic, J. Effects of olive oil and its components on intestinal inflammation and inflammatory bowel disease. Nutrients 2022, 14, 757. [Google Scholar] [CrossRef] [PubMed]
  81. Shen, B. Chapter 1—Introduction and classification of inflammatory bowel diseases. In Atlas of Endoscopy Imaging in Inflammatory Bowel Disease; Shen, B., Ed.; Academic Press: Cambridge, MA, USA, 2020; pp. 1–8. [Google Scholar]
  82. Chicco, F.; Magrì, S.; Cingolani, A.; Paduano, D.; Pesenti, M.; Zara, F.; Tumbarello, F.; Urru, E.; Melis, A.; Casula, L.; et al. Multidimensional impact of Mediterranean diet on IBD patients. Inflamm. Bowel Dis. 2021, 27, 1–9. [Google Scholar] [CrossRef] [PubMed]
  83. Morvaridi, M.; Jafarirad, S.; Seyedian, S.S.; Alavinejad, P.; Cheraghian, B. Effects of extra virgin olive oil and canola oil on inflammatory markers and gastrointestinal symptoms in patients with ulcerative colitis. Eur. J. Clin. Nutr. 2020, 74, 891–899. [Google Scholar] [CrossRef] [PubMed]
  84. Serra, G.; Deiana, M.; Spencer, J.P.E.; Corona, G. Olive oil phenolics prevent oxysterol-induced proinflammatory cytokine secretion and reactive oxygen species production in human peripheral blood mononuclear cells, through modulation of p38 and JNK pathways. Mol. Nutr. Food Res. 2017, 61, 1700283. [Google Scholar] [CrossRef] [PubMed]
  85. Serreli, G.; Melis, M.P.; Corona, G.; Deiana, M. Modulation of LPS-induced nitric oxide production in intestinal cells by hydroxytyrosol and tyrosol metabolites: Insight into the mechanism of action. Food Chem. Toxicol. 2019, 125, 520–527. [Google Scholar] [CrossRef] [PubMed]
  86. Grivennikov, S.I.; Greten, F.R.; Karin, M. Immunity, inflammation, and cancer. Cell 2010, 140, 883–899. [Google Scholar] [CrossRef] [PubMed]
  87. Taniguchi, K.; Karin, M. IL-6 and related cytokines as the critical lynchpins between inflammation and cancer. Semin. Immunol. 2014, 26, 54–74. [Google Scholar] [CrossRef] [PubMed]
  88. Taniguchi, K.; Karin, M. NF-κB, inflammation, immunity and cancer: Coming of age. Nat. Rev. Immunol. 2018, 18, 309–324. [Google Scholar] [CrossRef] [PubMed]
  89. Levy, D.E.; Lee, C.K. What does Stat3 do? J. Clin. Investig. 2002, 109, 1143–1148. [Google Scholar] [CrossRef] [PubMed]
  90. Johnson, D.E.; O’Keefe, R.A.; Grandis, J.R. Targeting the IL-6/JAK/STAT3 signalling axis in cancer. Nat. Rev. Clin. Oncol. 2018, 15, 234–248. [Google Scholar] [CrossRef] [PubMed]
  91. Sánchez-Fidalgo, S.; Sánchez de Ibargüen, L.; Cárdeno, A.; Alarcón de la Lastra, C. Influence of extra virgin olive oil diet enriched with hydroxytyrosol in a chronic DSS colitis model. Eur. J. Nutr. 2011, 51, 497–506. [Google Scholar] [CrossRef] [PubMed]
  92. Huguet-Casquero, A.; Xu, Y.; Gainza, E.; Pedraz, J.L.; Beloqui, A. Oral delivery of oleuropein-loaded lipid nanocarriers alleviates inflammation and oxidative stress in acute colitis. Int. J. Pharm. 2020, 586, 119515. [Google Scholar] [CrossRef] [PubMed]
  93. Zender, L.; Spector, M.S.; Xue, W.; Flemming, P.; Cordon-Cardo, C.; Silke, J.; Fan, S.T.; Luk, J.M.; Wigler, M.; Hannon, G.J.; et al. Identification and validation of oncogenes in liver cancer using an integrative oncogenomic approach. Cell 2006, 125, 1253–1267. [Google Scholar] [CrossRef] [PubMed]
  94. Gutierrez-V, R.; de la Puerta, R.; Catalá, A. The effect of tyrosol, hydroxytyrosol and oleuropein on the non-enzymatic lipid peroxidation of rat liver microsomes. Mol. Cell. Biochem. 2001, 217, 35–41. [Google Scholar] [CrossRef] [PubMed]
  95. Magdaleno, F.; Blajszczak, C.C.; Nieto, N. Key events participating in the pathogenesis of alcoholic liver disease. Biomolecules 2017, 7, 9. [Google Scholar] [CrossRef] [PubMed]
  96. Lee, D.H.; Han, D.H.; Nam, K.T.; Park, J.S.; Kim, S.H.; Lee, M.; Kim, G.; Min, B.S.; Cha, B.S.; Lee, Y.S.; et al. Ezetimibe, an NPC1L1 inhibitor, is a potent Nrf2 activator that protects mice from diet-induced nonalcoholic steatohepatitis. Free Radic. Biol. Med. 2016, 99, 520–532. [Google Scholar] [CrossRef] [PubMed]
  97. Zhao, B.; Ma, Y.; Xu, Z.; Wang, J.; Wang, F.; Wang, D.; Pan, S.; Wu, Y.; Pan, H.; Xu, D.; et al. Hydroxytyrosol from olive oil suppresses growth of human hepatocellular carcinoma cells via inactivating AKT and NF-κB pathways. Cancer Lett. 2014, 347, 79–87. [Google Scholar] [CrossRef] [PubMed]
  98. Martín, M.A.; Ramos, S.; Granado-Serrano, A.B.; Rodríguez-Ramiro, I.; Trujillo, M.; Bravo, L.; Goya, L. Hydroxytyrosol induces antioxidant/detoxificant enzymes and Nrf2 translocation via ERK and PI3K/Akt pathways in HepG2 cells. Mol. Nutr. Food Res. 2010, 54, 956–966. [Google Scholar] [CrossRef] [PubMed]
  99. Pirozzi, C.; Lama, A.; Simeoli, R.; Paciello, O.; Pagano, T.B.; Mollica, M.P.; Di Guida, F.; Russo, R.; Magliocca, S.; Canani, R.B.; et al. Hydroxytyrosol prevents metabolic impairment by reducing hepatic inflammation and restoring duodenal integrity in a rat model of NAFLD. J. Nutr. Biochem. 2016, 30, 108–115. [Google Scholar] [CrossRef] [PubMed]
  100. Gabbia, D.; Sayaf, K.; Zanotto, I.; Colognesi, M.; Frion-Herrera, Y.; Carrara, M.; Russo, F.P.; De Martin, S. Tyrosol attenuates NASH features by reprogramming the hepatic immune milieu. Eur. J. Pharmacol. 2024, 969, 176453. [Google Scholar] [CrossRef] [PubMed]
  101. Schirone, L.; Overi, D.; Carpino, G.; Carnevale, R.; De Falco, E.; Nocella, C.; D’Amico, A.; Bartimoccia, S.; Cammisotto, V.; Castellani, V.; et al. Oleuropein, a component of extra virgin olive oil, improves liver steatosis and lobular inflammation by lipopolysaccharides-TLR4 axis downregulation. Int. J. Mol. Sci. 2024, 25, 5580. [Google Scholar] [CrossRef] [PubMed]
  102. Frontera, W.R.; Ochala, J. Skeletal muscle: A brief review of structure and function. Calcif. Tissue Int. 2015, 96, 183–195. [Google Scholar] [CrossRef] [PubMed]
  103. Feng, L.T.; Chen, Z.N.; Bian, H. Skeletal muscle: Molecular structure, myogenesis, biological functions and diseases. Med. Comm. 2024, 5, e649. [Google Scholar] [CrossRef] [PubMed]
  104. Sambasivan, R.; Tajbakhsh, S. Skeletal muscle stem cell birth and properties. Semin. Cell. Dev. Biol. 2007, 18, 870–882. [Google Scholar] [CrossRef] [PubMed]
  105. Zammit, P.S. Function of the myogenic regulatory factors Myf5, MyoD, Myogenin and MRF4 in skeletal muscle, satellite cells and regenerative myogenesis. Semin. Cell. Dev. Biol. 2017, 72, 19–32. [Google Scholar] [CrossRef] [PubMed]
  106. Bentzinger, C.F.; Wang, Y.X.; Rudnicki, M.A. Building muscle: Molecular regulation of myogenesis. Cold Spring Harb. Perspect. Biol. 2012, 4, a008342. [Google Scholar] [CrossRef] [PubMed]
  107. Felig, P. The glucose-alanine cycle. Metab.-Clin. Exp. 1973, 22, 179–207. [Google Scholar] [CrossRef] [PubMed]
  108. Wolfe, R.R. The underappreciated role of muscle in health and disease. Am. J. Clin. Nutr. 2006, 84, 475–482. [Google Scholar] [CrossRef] [PubMed]
  109. Sartori, R.; Romanello, V.; Sandri, M. Mechanisms of muscle atrophy and hypertrophy: Implications in health and disease. Nat. Commun. 2021, 12, 330. [Google Scholar] [CrossRef] [PubMed]
  110. Ding, S.; Dai, Q.; Huang, H.; Xu, Y.; Zhong, C. An overview of muscle atrophy. Adv. Exp. Med. Biol. 2018, 1088, 3–19. [Google Scholar] [CrossRef] [PubMed]
  111. Rüegg, M.A.; Glass, D.J. Molecular mechanisms and treatment options for muscle wasting diseases. Annu. Rev. Pharmacol. Toxicol. 2011, 51, 373–595. [Google Scholar] [CrossRef] [PubMed]
  112. Jaiswal, N.; Gavin, M.; Loro, E.; Sostre-Colón, J.; Roberson, P.A.; Uehara, K.; Rivera-Fuentes, N.; Neinast, M.; Arany, Z.; Kimball, S.R.; et al. AKT controls protein synthesis and oxidative metabolism via combined mTORC1 and FOXO1 signalling to govern muscle physiology. J. Cachexia Sarcopenia Muscle 2022, 13, 495–514. [Google Scholar] [CrossRef] [PubMed]
  113. Waldemer-Streyer, R.J.; Kim, D.; Chen, J. Muscle cell-derived cytokines in skeletal muscle regeneration. FEBS J. 2022, 289, 6463–6483. [Google Scholar] [CrossRef] [PubMed]
  114. Alvarez, A.M.; Trufen, C.E.M.; Buri, M.V.; de Sousa, M.B.N.; Arruda-Alves, F.I.; Lichtenstein, F.; Castro de Oliveira, U.; Junqueira-de-Azevedo, I.L.M.; Teixeira, C.; Moreira, V. Tumor necrosis factor-alpha modulates expression of genes involved in cytokines and chemokine pathways in proliferative myoblast cells. Cells 2024, 13, 1161. [Google Scholar] [CrossRef] [PubMed]
  115. Serrano, A.L.; Baeza-Raja, B.; Perdiguero, E.; Jardí, M.; Muñoz-Cánoves, P. Interleukin-6 is an essential regulator of satellite cell-mediated skeletal muscle hypertrophy. Cell Metab. 2008, 7, 33–44. [Google Scholar] [CrossRef] [PubMed]
  116. Guerci, A.; Lahoute, C.; Hébrard, S.; Collard, L.; Graindorge, D.; Favier, M.; Cagnard, N.; Batonnet-Pichon, S.; Précigout, G.; Garcia, L.; et al. Srf-dependent paracrine signals produced by myofibers control satellite cell-mediated skeletal muscle hypertrophy. Cell Metab. 2012, 15, 25–37. [Google Scholar] [CrossRef] [PubMed]
  117. Kurek, J.B.; Nouri, S.; Kannourakis, G.; Murphy, M.; Austin, L. Leukemia inhibitory factor and interleukin-6 are produced by diseased and regenerating skeletal muscle. Muscle Nerve 1996, 19, 1291–1301. [Google Scholar] [CrossRef]
  118. Sakuma, K.; Watanabe, K.; Sano, M.; Uramoto, I.; Totsuka, T. Differential adaptation of growth and differentiation factor 8/myostatin, fibroblast growth factor 6 and leukemia inhibitory factor in overloaded, regenerating and denervated rat muscles. Biochim. Biophys. Acta 2000, 1497, 77–88. [Google Scholar] [CrossRef] [PubMed]
  119. Kami, K.; Senba, E. Localization of leukemia inhibitory factor and interleukin-6 messenger ribonucleic acids in regenerating rat skeletal muscle. Muscle Nerve 1998, 21, 819–822. [Google Scholar] [CrossRef]
  120. Barnard, W.; Bower, J.; Brown, M.A.; Murphy, M.; Austin, L. Leukemia inhibitory factor (LIF) infusion stimulates skeletal muscle regeneration after injury: Injured muscle expresses lif mRNA. J. Neurol. Sci. 1994, 123, 108–113. [Google Scholar] [CrossRef] [PubMed]
  121. Cheng, M.; Nguyen, M.H.; Fantuzzi, G.; Koh, T.J. Endogenous interferon-gamma is required for efficient skeletal muscle regeneration. Am. J. Physiol. Cell Physiol. 2008, 294, C1183–C1191. [Google Scholar] [CrossRef] [PubMed]
  122. Acharyya, S.; Sharma, S.M.; Cheng, A.S.; Ladner, K.J.; He, W.; Kline, W.; Wang, H.; Ostrowski, M.C.; Huang, T.H.; Guttridge, D.C. TNF inhibits Notch-1 in skeletal muscle cells by Ezh2 and DNA methylation mediated repression: Implications in duchenne muscular dystrophy. PLoS ONE 2010, 5, e12479. [Google Scholar] [CrossRef] [PubMed]
  123. Pelosi, L.; Berardinelli, M.G.; De Pasquale, L.; Nicoletti, C.; D’Amico, A.; Carvello, F.; Moneta, G.M.; Catizone, A.; Bertini, E.; De Benedetti, F.; et al. Functional and morphological improvement of dystrophic muscle by interleukin 6 receptor blockade. EBioMedicine 2015, 2, 285–293. [Google Scholar] [CrossRef] [PubMed]
  124. Pelosi, L.; Berardinelli, M.G.; Forcina, L.; Spelta, E.; Rizzuto, E.; Nicoletti, C.; Camilli, C.; Testa, E.; Catizone, A.; De Benedetti, F.; et al. Increased levels of interleukin-6 exacerbate the dystrophic phenotype in mdx mice. Hum. Mol. Genet. 2015, 24, 6041–6053. [Google Scholar] [CrossRef] [PubMed]
  125. Zhang, L.; Wang, G.; Chen, X.; Zhang, C.; Jiang, Y.; Zhao, W.; Li, H.; Sun, J.; Li, X.; Xu, H.; et al. Irgm1 knockout indirectly inhibits regeneration after skeletal muscle injury in mice. Int. Immunopharmacol. 2020, 84, 106515. [Google Scholar] [CrossRef] [PubMed]
  126. Grzelkowska-Kowalczyk, K.; Wicik, Z.; Majewska, A.; Tokarska, J.; Grabiec, K.; Kozłowski, M.; Milewska, M.; Błaszczyk, M. Transcriptional regulation of important cellular processes in skeletal myogenesis through interferon-γ. J. Interferon Cytokine Res. 2015, 35, 89–99. [Google Scholar] [CrossRef] [PubMed]
  127. Kalovidouris, A.E.; Plotkin, Z.; Graesser, D. Interferon-gamma inhibits proliferation, differentiation, and creatine kinase activity of cultured human muscle cells. II. A possible role in myositis. J. Rheumatol. 1993, 20, 1718–1723. [Google Scholar] [PubMed]
  128. Ji, Y.; Li, M.; Chang, M.; Liu, R.; Qiu, J.; Wang, K.; Deng, C.; Shen, Y.; Zhu, J.; Wang, W.; et al. Inflammation: Roles in skeletal muscle atrophy. Antioxidants 2022, 11, 1686. [Google Scholar] [CrossRef] [PubMed]
  129. Cohen, S.; Nathan, J.A.; Goldberg, A.L. Muscle wasting in disease: Molecular mechanisms and promising therapies. Nat. Rev. Drug Discov. 2015, 14, 58–74. [Google Scholar] [CrossRef] [PubMed]
  130. Ru, Q.; Chen, L.; Xu, G.; Wu, Y. Exosomes in the pathogenesis and treatment of cancer-related cachexia. J. Transl. Med. 2024, 22, 408. [Google Scholar] [CrossRef] [PubMed]
  131. Fearon, K.; Strasser, F.; Anker, S.D.; Bosaeus, I.; Bruera, E.; Fainsinger, R.L.; Jatoi, A.; Loprinzi, C.; MacDonald, N.; Mantovani, G.; et al. Definition and classification of cancer cachexia: An international consensus. Lancet Oncol. 2011, 12, 489–495. [Google Scholar] [CrossRef] [PubMed]
  132. Dewys, W.D.; Begg, C.; Lavin, P.T.; Band, P.R.; Bennett, J.M.; Bertino, J.R.; Cohen, M.H.; Douglass, H.O., Jr.; Engstrom, P.F.; Ezdinli, E.Z.; et al. Prognostic effect of weight loss prior to chemotherapy in cancer patients. Eastern Cooperative Oncology Group. Am. J. Med. 1980, 69, 491–497. [Google Scholar] [CrossRef] [PubMed]
  133. Li, X.; Hu, C.; Zhang, Q.; Wang, K.; Li, W.; Xu, H.; Fu, Z.; Wang, C.; Guo, Z.; Weng, M.; et al. Cancer cachexia statistics in China. Precis. Nutr. 2022, 1, e00005. [Google Scholar] [CrossRef]
  134. Poisson, J.; Martinez-Tapia, C.; Heitz, D.; Geiss, R.; Albrand, G.; Falandry, C.; Gisselbrecht, M.; Couderc, A.L.; Boulahssass, R.; Liuu, E.; et al. Prevalence and prognostic impact of cachexia among older patients with cancer: A nationwide cross-sectional survey (NutriAgeCancer). J. Cachexia Sarcopenia Muscle 2021, 12, 1477–1488. [Google Scholar] [CrossRef] [PubMed]
  135. Argilés, J.M.; López-Soriano, F.J.; Stemmler, B.; Busquets, S. Cancer-associated cachexia—Understanding the tumour macroenvironment and microenvironment to improve management. Nat. Rev. Clin. Oncol. 2023, 20, 250–264. [Google Scholar] [CrossRef] [PubMed]
  136. Baracos, V.E.; Martin, L.; Korc, M.; Guttridge, D.C.; Fearon, K.C.H. Cancer-associated cachexia. Nat. Rev. Dis. Primers 2018, 4, 17105. [Google Scholar] [CrossRef] [PubMed]
  137. Ni, J.; Zhang, L. Cancer Cachexia: Definition, Staging and Emerging Treatments. Cancer Manag. Res. 2020, 12, 5597–5605. [Google Scholar] [CrossRef] [PubMed]
  138. Penna, F.; Ballarò, R.; Costelli, P. The redox balance: A target for interventions against muscle wasting in cancer cachexia? Antioxid. Redox Signal. 2020, 33, 542–558. [Google Scholar] [CrossRef] [PubMed]
  139. Wu, Q.; Liu, Z.; Li, B.; Liu, Y.E.; Wang, P. Immunoregulation in cancer-associated cachexia. J. Adv. Res. 2024, 58, 45–62. [Google Scholar] [CrossRef] [PubMed]
  140. Wang, Y.; Ding, S. Extracellular vesicles in cancer cachexia: Deciphering pathogenic roles and exploring therapeutic horizons. J. Transl. Med. 2024, 22, 506. [Google Scholar] [CrossRef] [PubMed]
  141. Hadrich, F.; Garcia, M.; Maalej, A.; Moldes, M.; Isoda, H.; Feve, B.; Sayadi, S. Oleuropein activated AMPK and induced insulin sensitivity in C2C12 muscle cells. Life Sci. 2016, 151, 167–173. [Google Scholar] [CrossRef] [PubMed]
  142. Kikusato, M.; Muroi, H.; Uwabe, Y.; Furukawa, K.; Toyomizu, M. Oleuropein induces mitochondrial biogenesis and decreases reactive oxygen species generation in cultured avian muscle cells, possibly via an up-regulation of peroxisome proliferator-activated receptor γ coactivator-1α. Anim. Sci. J. 2016, 87, 1371–1378. [Google Scholar] [CrossRef] [PubMed]
  143. Cao, K.; Xu, J.; Zou, X.; Li, Y.; Chen, C.; Zheng, A.; Li, H.; Li, H.; Szeto, I.M.; Shi, Y.; et al. Hydroxytyrosol prevents diet-induced metabolic syndrome and attenuates mitochondrial abnormalities in obese mice. Free Radic. Biol. Med. 2014, 67, 396–407. [Google Scholar] [CrossRef] [PubMed]
  144. Feng, Z.; Bai, L.; Yan, J.; Li, Y.; Shen, W.; Wang, Y.; Wertz, K.; Weber, P.; Zhang, Y.; Chen, Y.; et al. Mitochondrial dynamic remodeling in strenuous exercise-induced muscle and mitochondrial dysfunction: Regulatory effects of hydroxytyrosol. Free Radic. Biol. Med. 2011, 50, 1437–1446. [Google Scholar] [CrossRef] [PubMed]
  145. Gwal, R.; Reutzel, M.; Dilberger, B.; Hein, H.; Zotzel, J.; Marx, S.; Tretzel, J.; Sarafeddinov, A.; Fuchs, C.; Eckert, G.P. Purified oleocanthal and ligstroside protect against mitochondrial dysfunction in models of early Alzheimer’s disease and brain ageing. Exp. Neurol. 2020, 328, 113248. [Google Scholar] [CrossRef]
  146. Yeo, D.; Zhang, T.; Liu, T.; Zhang, Y.; Kang, C.; Ji, L.L. Protective effects of extra virgin olive oil and exercise training on rat skeletal muscle against high-fat diet feeding. J. Nutr. Biochem. 2022, 100, 108902. [Google Scholar] [CrossRef] [PubMed]
  147. Gatti, C.R.; Schibert, F.; Taylor, V.S.; Capobianco, E.; Montero, V.; Higa, R.; Jawerbaum, A. Maternal dietary olive oil protects diabetic rat offspring from impaired uterine decidualization. Placenta 2024. [Google Scholar] [CrossRef] [PubMed]
  148. Yarla, N.S.; Polito, A.; Peluso, I. Effects of Olive Oil on TNF-α and IL-6 in Humans: Implication in Obesity and Frailty. Endocr. Metab. Immune Disord. Drug Targets 2018, 18, 63–74. [Google Scholar] [CrossRef] [PubMed]
  149. Salucci, S.; Bartoletti-Stella, A.; Bavelloni, A.; Aramini, B.; Blalock, W.L.; Fabbri, F.; Vannini, I.; Sambri, V.; Stella, F.; Faenza, I. Extra Virgin Olive Oil (EVOO), a Mediterranean Diet Component, in the Management of Muscle Mass and Function Preservation. Nutrients 2022, 14, 3567. [Google Scholar] [CrossRef] [PubMed]
  150. Zhang, J.; Nugrahaningrum, D.A.; Marcelina, O.; Ariyanti, A.D.; Wang, G.; Liu, C.; Wu, S.; Kasim, V. Tyrosol Facilitates Neovascularization by Enhancing Skeletal Muscle Cells Viability and Paracrine Function in Diabetic Hindlimb Ischemia Mice. Front. Pharmacol. 2019, 10, 909. [Google Scholar] [CrossRef] [PubMed]
  151. Baguley, B.J.; Skinner, T.L.; Jenkins, D.G.; Wright, O.R.L. Mediterranean-style dietary pattern improves cancer-related fatigue and quality of life in men with prostate cancer treated with androgen deprivation therapy: A pilot randomized control trial. Clin. Nutr. 2021, 40, 245–254. [Google Scholar] [CrossRef] [PubMed]
  152. Gioxari, A.; Tzanos, D.; Kostara, C.; Papandreou, P.; Mountzios, G.; Skouroliakou, M. Mediterranean Diet Implementation to Protect against Advanced Lung Cancer Index (ALI) Rise: Study Design and Preliminary Results of a Randomized Controlled Trial. Int. J. Environ. Res. Public Health 2021, 18, 3700. [Google Scholar] [CrossRef] [PubMed]
  153. Bagheri, A.; Asoudeh, F.; Rezaei, S.; Babaei, M.; Esmaillzadeh, A. The Effect of Mediterranean Diet on Body Composition, Inflammatory Factors and Nutritional Status in Patients with Cachexia Induced by Colorectal Cancer: A Randomized Clinical Trial. Integr. Cancer Ther. 2023, 22, 15347354231195322. [Google Scholar] [CrossRef] [PubMed]
  154. De Stefanis, D.; Balestrini, A.; Costelli, P. Oleocanthal Protects C2C12 Myotubes against the Pro-Catabolic and Anti-Myogenic Action of Stimuli Able to Induce Muscle Wasting In Vivo. Nutrients 2024, 16, 1302. [Google Scholar] [CrossRef] [PubMed]
  155. Salucci, S.; Burattini, S.; Versari, I.; Bavelloni, A.; Bavelloni, F.; Curzi, D.; Battistelli, M.; Gobbi, P.; Faenza, I. Morpho-Functional Analyses Demonstrate That Tyrosol Rescues Dexamethasone-Induced Muscle Atrophy. J. Funct. Morphol. Kinesiol. 2024, 9, 124. [Google Scholar] [CrossRef] [PubMed]
  156. Pastor, A.; Rodríguez-Morató, J.; Olesti, E.; Pujadas, M.; Pérez-Mañá, C.; Khymenets, O.; Fitó, M.; Covas, M.I.; Solá, R.; Motilva, M.J.; et al. Analysis of free hydroxytyrosol in human plasma following the administration of olive oil. J. Chromatogr. A 2016, 1437, 183–190. [Google Scholar] [CrossRef] [PubMed]
  157. Boronat, A.; Rodriguez-Morató, J.; Serreli, G.; Fitó, M.; Tyndale, R.F.; Deiana, M.; de la Torre, R. Contribution of Biotransformations Carried Out by the Microbiota, Drug-Metabolizing Enzymes, and Transport Proteins to the Biological Activities of Phytochemicals Found in the Diet. Adv. Nutr. (Bethesda Md.) 2021, 12, 2172–2189. [Google Scholar] [CrossRef] [PubMed]
  158. Naoi, M.; Wu, Y.; Shamoto-Nagai, M.; Maruyama, W. Mitochondria in Neuroprotection by Phytochemicals: Bioactive Polyphenols Modulate Mitochondrial Apoptosis System, Function and Structure. Int. J. Mol. Sci. 2019, 20, 2451. [Google Scholar] [CrossRef] [PubMed]
  159. Mantovani, G.; Macciò, A.; Madeddu, C.; Gramignano, G.; Lusso, M.R.; Serpe, R.; Massa, E.; Astara, G.; Deiana, L. A phase II study with antioxidants, both in the diet and supplemented, pharmaconutritional support, progestagen, and anti-cyclooxygenase-2 showing efficacy and safety in patients with cancer-related anorexia/cachexia and oxidative stress. Cancer Epidemiol. Biomark. Prev. 2006, 15, 1030–1034. [Google Scholar] [CrossRef] [PubMed]
  160. Yang, B.; Dong, Y.; Wang, F.; Zhang, Y. Nanoformulations to Enhance the Bioavailability and Physiological Functions of Polyphenols. Molecules 2020, 25, 4613. [Google Scholar] [CrossRef] [PubMed]
  161. Bergonzi, M.C.; De Stefani, C.; Vasarri, M.; Ivanova Stojcheva, E.; Ramos-Pineda, A.M.; Baldi, F.; Bilia, A.R.; Degl’Innocenti, D. Encapsulation of Olive Leaf Polyphenol-Rich Extract in Polymeric Micelles to Improve Its Intestinal Permeability. Nanomaterials 2023, 13, 3147. [Google Scholar] [CrossRef] [PubMed]
Figure 1. EVOO’s composition.
Figure 1. EVOO’s composition.
Nutrients 17 02334 g001
Figure 2. Modulation of carcinogenesis by the interaction of EVOO’s components with pro-inflammatory cytokines.
Figure 2. Modulation of carcinogenesis by the interaction of EVOO’s components with pro-inflammatory cytokines.
Nutrients 17 02334 g002
Figure 3. Potential role of EVOO in the prevention of cancer cachexia. Symbols: ↓ reduce; ↑ increment.
Figure 3. Potential role of EVOO in the prevention of cancer cachexia. Symbols: ↓ reduce; ↑ increment.
Nutrients 17 02334 g003
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

De Stefanis, D.; Costelli, P. Extra Virgin Olive Oil (EVOO) Components: Interaction with Pro-Inflammatory Cytokines Focusing on Cancer and Skeletal Muscle Biology. Nutrients 2025, 17, 2334. https://doi.org/10.3390/nu17142334

AMA Style

De Stefanis D, Costelli P. Extra Virgin Olive Oil (EVOO) Components: Interaction with Pro-Inflammatory Cytokines Focusing on Cancer and Skeletal Muscle Biology. Nutrients. 2025; 17(14):2334. https://doi.org/10.3390/nu17142334

Chicago/Turabian Style

De Stefanis, Daniela, and Paola Costelli. 2025. "Extra Virgin Olive Oil (EVOO) Components: Interaction with Pro-Inflammatory Cytokines Focusing on Cancer and Skeletal Muscle Biology" Nutrients 17, no. 14: 2334. https://doi.org/10.3390/nu17142334

APA Style

De Stefanis, D., & Costelli, P. (2025). Extra Virgin Olive Oil (EVOO) Components: Interaction with Pro-Inflammatory Cytokines Focusing on Cancer and Skeletal Muscle Biology. Nutrients, 17(14), 2334. https://doi.org/10.3390/nu17142334

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