Next Article in Journal
Anticancer Activity of Propolis and Its Compounds
Next Article in Special Issue
Machine Learning and In Vitro Chemical Screening of Potential α-Amylase and α-Glucosidase Inhibitors from Thai Indigenous Plants
Previous Article in Journal
Physiological Functions of Threonine in Animals: Beyond Nutrition Metabolism
Previous Article in Special Issue
Systematic Bioinformatic Analyses of Nutrigenomic Modifications by Polyphenols Associated with Cardiometabolic Health in Humans—Evidence from Targeted Nutrigenomic Studies
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Possible Biochemical Processes Underlying the Positive Health Effects of Plant-Based Diets—A Narrative Review

1
Institute of Nutritional Sciences and Dietetics, Faculty of Health Sciences, University of Pecs, 7621 Pecs, Hungary
2
Department of Biochemistry and Medical Chemistry, Medical School, University of Pecs, 7624 Pecs, Hungary
3
Doctoral School of Health Sciences, Faculty of Health Sciences, University of Pecs, 7621 Pecs, Hungary
4
Institute of Bioanalysis, Medical School, University of Pecs, 7624 Pecs, Hungary
5
Szentagothai Research Center, University of Pecs, 7624 Pecs, Hungary
6
Institute for Translational Medicine, Medical School, University of Pecs, 7624 Pecs, Hungary
7
Institute of Health Insurance, Faculty of Health Sciences, University of Pecs, 7621 Pecs, Hungary
8
2nd Department of Internal Medicine and Nephrology Centre, Clinical Centre, University of Pecs, 7624 Pecs, Hungary
*
Author to whom correspondence should be addressed.
Nutrients 2021, 13(8), 2593; https://doi.org/10.3390/nu13082593
Submission received: 31 May 2021 / Revised: 26 July 2021 / Accepted: 27 July 2021 / Published: 28 July 2021

Abstract

:
Plant-based diets are becoming more popular for many reasons, and epidemiological as well as clinical data also suggest that a well-balanced vegan diet can be adopted for the prevention, and in some cases, in the treatment of many diseases. In this narrative review, we provide an overview of the relationships between these diets and various conditions and their potential biochemical background. As whole plant foods are very rich in food-derived antioxidants and other phytochemicals, they have many positive physiological effects on different aspects of health. In the background of the beneficial health effects, several biochemical processes could stand, including the reduced formation of trimethylamine oxide (TMAO) or decreased serum insulin-like growth factor 1 (IGF-1) levels and altered signaling pathways such as mechanistic target of rapamycin (mTOR). In addition, the composition of plant-based diets may play a role in preventing lipotoxicity, avoiding N-glycolylneuraminic acid (Neu5Gc), and reducing foodborne endotoxin intake. In this article, we attempt to draw attention to the growing knowledge about these diets and provide starting points for further research.

1. Introduction

The definition of plant-based diets is widely used, and its main focus is consuming raw or minimally processed vegetables, fruits, whole grains, legumes, nuts and seeds, spices, and herbs. Besides that, these diets often minimize or exclude all animal products [1]. A well-balanced plant-based diet is useful [2,3] for the primary prevention of several health conditions. It can be also used as a complementary treatment in chronic diseases, amongst others in cardiovascular diseases [2,4,5,6,7], obesity [8,9], certain types of cancer [10,11,12], type 2 diabetes mellitus (T2DM) [13,14,15], and stroke [16,17].
Several well-known healthcare institutes such as the Academy of Nutrition and Dietetics [18], the American Diabetes Association [19], the British Dietetic Association [20], the American Dietetic Association and Dietitians of Canada [21], the Directorate-General of Health of Portugal [22], and the German Nutrition Society [23] have given attention to plant-based nutrition, but these recommendations and position papers focus mainly on the macro- and micronutrient content of these diets and their potentially beneficial health roles. Although these diets are acceptable for example in the management of T2DM, with the evidence level B [19] and these recommendations mentioned above list a number of chronic diseases in which a vegetarian, vegan, or other plant-based diet might be beneficial, little is said about the physiological and biochemical mechanisms underlying these positive effects.
Plant-based diets could be an acceptable solution for improving and maintaining health as well as reversing some diseases, but it would also be important to know the underlying processes for recommendations. Therefore, the aim of this review is to provide an overview of some possible biochemical processes behind the positive physiological effects of these diets demonstrated in previous scientific literature.

2. Antioxidants

A typical Western-type diet is rich in refined carbohydrates (sugar), and saturated fats of animal origin, but low in fiber; however, high consumption of the first two ingredients can lead to a pro-oxidative state after each meal due to their active oxidation and simultaneous reactive oxygen species (ROS) formation [24]. Increasing evidence shows that this postprandial pro-oxidative state can induce inflammation after meals and is an important contributing factor besides obesity in several other chronic diseases, such as atherosclerosis, high systolic blood pressure, and insulin resistance [24,25]. This type of low-grade inflammation can lead to endothelial dysfunction; reduced insulin sensitivity and, consequently, reduced nitric oxide (NO) synthesis; and elevated oxidized low-density lipoprotein (LDL). Most of animal-based foods have significant proinflammatory and endothelial-dysfunction-inducing effects, although the exact mechanisms are not well understood [26]. These alterations can be detected even several hours after the consumption of animal-based foods. These factors can play a significant role in the development and progression of many diseases, such as metabolic syndrome, atherosclerosis, T2DM, hypertension, and stroke [24,27].
The acute negative effect of lipemia caused by a single high fat meal seems to be insignificant regard to compensatory mechanisms, which counteract most of these negative effects, but it is not negligible. Eventually, the undesirable effects accumulate and contribute to increased cardiovascular mortality [28]. Thus, consuming a variety of plant foods containing many antioxidants after every meal could be a reasonable way to reduce pro-oxidative state [29] caused by high calorie or high lipid containing meals.
The human metabolism can mitigate the negative consequences of this imbalance in many ways (superoxide dismutase, glutathione peroxidase, catalase, etc.). Numerous studies have shown that the use of exogenous antioxidants derived from plant foods can improve the efficiency of the endogenous enzymatic defense [30,31]. Plant-based foods contain the highest amounts of food antioxidants; by contrast, the antioxidant contents of animal-based foods are negligible, based on data for 3100 investigated foods, beverages, spices, herbs, and supplements used worldwide [32]. The consumption of plant foods containing a high number of antioxidants seems to be protective against the development of certain diseases (cancer, cardiovascular diseases, T2DM, osteoporosis, and neurodegenerative diseases) [33]. By contrast, animal-based foods contain certain catalytic molecules (e.g., iron, myoglobin, and hemoglobin), which, if consumed on a regular basis, could increase lipid peroxidation in the stomach. In different types of in vitro gastric model tests, 100-fold increase in the level of lipid hydroperoxides has been observed [34]. Proteins and lipids oxidized by these catalytic molecules, which can be found in significant quantities in processed meat, butter, and some heated plant-based oils (such as olive oil), may affect the development of Parkinson’s disease [35], inflammatory bowel disease [36], and insulin resistance [37].
Vitamin C, a reducing agent and electron-donor antioxidant, cannot be synthesized endogenously through the human metabolism, so the amount of ascorbic acid in the body depends on the dietary intake [38]. Vitamin C participates in the first line of antioxidant defense and is effective against the superoxide radical ion, hydrogen peroxide, hydroxyl radical, and singlet oxygen, as well as nitrogen oxide species. An in vitro experiment showed that vitamin C could increase the efficiency of vitamin E by decreasing tocopheroxyl radicals [39]. An eight-week-long interventional human study showed that the consumption of red fruit juice rich in anthocyanin and vitamin C increased the activity of superoxide dismutase [40]. Adequate vitamin C intake contributes to cardiovascular health, such as by improving blood pressure [41] and endothelial function [42].
Vitamin E is a plant-derived, potent peroxyl-radical-scavenging antioxidant. It can prevent the propagation of all free radicals in lipoproteins and membranes [43]. Animal and human studies have determined that vitamin E plays a major role in protecting the cerebellum from free radical species [44,45,46]. Four weeks of the daily consumption of roasted almonds caused a significant increase in plasma α-tocopherol concentrations but also a significant decrease in the level of IL-10, as well as lowering the inflammatory markers CAM-1, IL-1ß, and IL-6 in the serum [47].
Carotenoids are fat-soluble pigments that can be found in many plant-based foods. The antioxidant effect of carotenoids has been proven to be linked to their chemical structure. The presence of conjugated double bonds enables these molecules to accept electrons from reactive species and neutralize free radicals [48].
β-carotene is a provitamin that operates as a chain-breaking antioxidant but is less potent in the scavenging of peroxyl radicals. Zeaxanthin can effectively participate in the prevention of lipid peroxidation. A cross-sectional study showed that higher intake of fruits and vegetables rich in vitamin A (orange, peach, collard greens and kale) was associated with lower risk of developing glaucoma among older women. There was also a trend in risk reduction for β-carotene, lutein, and zeaxanthin, but it was not significant [49]. Lutein may play a protective role in reducing oxidative stress and damage of retina and optic nerve [50]. Lycopene is one of the most effective carotenoids in quenching singlet oxygen and participates in scavenging hypochlorous acid [51]. Lycopene is the predominant carotenoid in tomato, and it was shown that its bioavailability from tomato paste is higher than from fresh tomatoes [52]. In a randomized controlled study, the consumption of lycopene-rich tomato paste had protective effects against skin burn caused by UV light. The dietary intervention decreased the formation of erythema and inhibited the expression of matrix metalloproteinase-1 (MMP1) caused by UV radiation, which is a key regulator in the photoaging process [53].
A randomized controlled trial demonstrated that higher vegetable and fruit intake can increase the antioxidant property of HDL (high-density lipoprotein) due to the natural lycopene content of those foods [54]. In addition, it can reduce the risk of cardiovascular disease by decreasing the hydrogen-peroxide-induced oxidative injury of endothelial cells [55].
Due to the high antioxidant capacity of carotenoids, antioxidant and anticarcinogenic effects were hypothesized. To examine these phenomena, in two separate interventional studies, patients were supplemented with β-carotene. The β-Carotene and Retinol Efficacy Trial (CARET) demonstrated that the daily supplementation of β-carotene and vitamin A increased the prevalence of lung cancer in frequent smokers or in patients with substantial occupational exposure to asbestos in the USA [56]. These results were highly consistent with those found for β-carotene and vitamin E supplementation in the Alpha-Tocopherol Beta-Carotene (ATBC) cancer prevention study in male smokers in Finland [57]. The results of the long-term follow-up of the ATBC study confirm that β-carotene supplementation increases the lung cancer risk of smokers, regardless of the nicotine or tar content of cigarettes [58]. The best strategy for decreasing the prevalence of lung cancer among smokers is for them to quit smoking. Besides that, fruit and vegetable consumption have a protective effect in lung cancer’s etiology [59,60,61]. The dietary intake of carotenoids has some additional positive health effects, such as reducing the risk of T2DM [62] and metabolic syndrome [63].
There are plenty of phytochemicals of plant origin known to have potent biological antioxidant effects. The number of polyphenol-type antioxidant molecules alone could be more than 8000, as demonstrated in a previous study [33]. These compounds have significant positive effects (cancer, cardiovascular disease, neurodegeneration) on the human body in many ways [64]; however, their exact mechanisms of action are not yet fully understood. By manipulating many molecular signaling mechanisms such as PI3K, Akt, NF-κB, p53, and many others, these molecules positively affect the body’s oxidoreductive homeostasis [65].
The antioxidant effects of phenol-type compounds of plant origin are diverse. The phenol-type antioxidant activity depends on the compound’s ability to donate electrons or hydrogen, from which its potential for action as an antioxidant can be predicted. For instance, phenol-type molecules from fruits are capable of neutralizing superoxide anions, singlet oxygen, and lipid peroxides (‘scavenger function’) [66]. In addition, some flavonoids can recycle or reduce the body’s own antioxidant molecules and can also form chelates with metallic ions (Fe2+, Fe3+, and Cu2+), meaning that these molecules inhibit the pro-oxidative effect of metallic compounds. These chelates have an additional antioxidant effect similar to that of superoxide dismutase. This can serve as a further explanation for the positive physiological effects of antioxidant compounds [67,68,69].
Flavonoid–lipid and flavonoid–protein interactions are possible even if these compounds are present in small quantities. These interactions can induce numerous biological mechanisms, such as activating the body’s own antioxidant enzyme defense system and suppressing reactive oxygen species (ROS)-producing processes. Flavonoids and other antioxidant molecules of plant origin can contribute to a decreased absorption of lipid hydroxyperoxides. This may provide a further explanation for the positive physiological effects of antioxidant compounds [70,71].
Antioxidant supplementation, especially in the form of isolated components, seems to be ineffective [72,73]. Additionally, no beneficial effect on mortality has been described. In some cases, the use of antioxidant supplements may lead to undesirable health consequences, such as a higher occurrence of cancer and T2DM [74].
Data from clinical trials suggest that supplementation with an isolated component (vitamin E) or mixture of antioxidants (vitamin A and zinc) has unfavorable effects, such as increasing the incidence of hemorrhagic stroke and total mortality [75,76]. According to some researchers, antioxidant mixtures derived from natural sources are better than simple antioxidant formulas that is due to synergism between antioxidants [77].
Therefore, it seems beneficial to consume these antioxidants in their natural form as part of whole-plant foods. Fruits and vegetables are rich sources of polyphenols and, therefore, can have anti-inflammatory and antioxidant effects and could play a key role in the prevention or adjunctive therapy of different chronic diseases. Pomegranates are an excellent source of phytochemicals, such as anthocyanins, ellagic acid, and ellagitannins. In a randomized controlled trial, the consumption of one serving of pomegranate in its natural form decreased the plasma concentration of a potent inflammatory cytokine. This suppression of MCP1 (monocyte chemoattractant protein-1) was not detected after the ingestion of a dietary supplement rich in ellagic acid [78]. As antioxidants, polyphenol-containing fruits (e.g., strawberries) [28] and vegetables (e.g.,: kale) [79] can decrease LDL oxidation and as a result they can decrease the risk of coronary heart disease. Through different biochemical mechanisms (inhibiting regulatory enzymes and transcription factors involved in inflammation as well as scavenging free radicals) a diet high in antioxidants and polyphenols can prevent asthma, decrease the frequency of its exacerbation [80] and might be protective in the development and severity of different food allergies like peanut allergy [81].
Dietary antioxidants also have anti-aging effects due to their antioxidant and free radical scavenging potential. It is a well-known phenomenon that vegans, especially those who consume whole-food plant-based diet seems to look healthier and younger. Although this phenomenon has not been disclosed yet, the consumption of fruits and vegetables is a promising strategy to help maintaining youthful appearing of the skin [82,83]. Aging affects not only our skin, but other tissues also, for example our brain. In a follow-up study, women who had a higher intake of leafy greens or cruciferous vegetables from their diet showed the lowest degrees of cognitive decline [84].
Consuming various types of plant foods together can result in a difference between the expected and the measured antioxidant effects in favor of the latter. This phenomenon is often referred as “synergy”, which marks the additive positive effects of foodborne antioxidant molecules. [85]. Many examples prove the supremacy of consuming whole-plant foods over their isolated constituents [86], which gives us further verification that whole-plant consumption could be more beneficial most of the cases, due to their wide-range known and (probably still) unidentified micronutrient composition. However, to understand these complex relationships, the evaluation of each macro-, micronutrients, and phytochemicals should be examined in an extended way including the above mentioned “synergistic” phenomenon [87,88].
From these few examples, it seems clear that these unique whole-plant food compounds can help to maintain overall state of health and even contribute to the prevention of certain diseases (Figure 1) through various mechanisms, which still have to be confirmed.

3. Lipotoxicity

Lipids have key importance, not only in the mechanism of action of antioxidants. Lipids may play a prominent role in the positive health effects of plant-based diets [89].
The accumulation of ectopic lipid in non-fatty tissues is called lipotoxicity, which is a complex condition caused by increased plasma free fatty acids reaching toxic levels in non-adipose tissues when fat cells’ normal fat-storing capability is compromised [90].
The presence of excess fatty acids leads to the accumulation of intramyocellular lipid (IMCL—lipid deposition within myocytes) species such as diacylglycerol (DAG), ceramide, and long-chain acyl-CoAs [91]. If these metabolites occur in ß-cells, they can disrupt their functions [92,93]. Healthy subjects with normal body weight and without diabetes were examined using nuclear MRI spectroscopy, and it was found that IMCL could be a good predictor of susceptibility to insulin resistance [94]. Other clinical studies suggest that vegans have significantly lower IMCL levels than omnivores [95,96]. Furthermore, plant-based diets have protective effects on ß-cell function by increasing glucose sensitivity, decreasing basal insulin secretion and the mean glucose level [97]. Applying a low-fat, plant-based diet leads to a decrease in free fatty acid levels and better glycemic control compared to a low-carbohydrate omnivorous diet [98].
The further consequences of lipotoxicity can include the induction of proinflammatory processes [99], oxidative stress [100], and mitochondrial dysfunction [101].
Insulin resistance, mitochondrial dysfunction, and the alteration of intracellular signaling pathways lead to liver injury, and this can contribute to non-alcoholic fatty liver disease (NAFLD) [102]. The incidence of NAFLD is now at an endemic level, and it is currently the most common form of chronic liver disease worldwide, affecting about 25% of the general population [102,103]. The typical Western dietary pattern (high intakes of fast food, red meats, processed meats, full-fat dairy products, fried potatoes, high carbohydrate containing refined foods and soft drinks) is closely associated with the development and progression of NAFLD [104]. The connection between NAFLD and lipotoxicity is complex. Among the possible factors playing a role in the pathogenesis of NAFLD, free fatty acids seem to contribute to the development of lipotoxicity inducing lipid accumulation and lipotoxicity in liver cell cultures [105]. The connection between free fatty acid intake and insulin resistance has been reported in several studies [106,107,108,109]. The mitochondrial dysfunction caused by palmitic acid-induced oxidative stress can increase the damaging effects of ROS and disrupt insulin signaling [110]. The high dietary fat intake characteristic for Western-type diets can increase the serum free fatty acid levels, which is an independent risk factor for the development and worsening of NAFLD [111]. The possible consequences of lipotoxicity are summarized in Figure 2.
Adherence to plant-based diets appears to be protective against NAFLD (mainly because enhanced glycemic control, improved insulin sensitivity, and decreased chronic inflammation) [112,113], but further clinical trials are needed in order to clarify this relationship in more detail. A better understanding of lipid irregularity may eventually modify the concept of lipotoxicity as a key pathogenic factor in many diseases [102,114].

4. Trimethylamine N-Oxide

The mechanisms discussed in the previous sections can also have profound effects on the cardiovascular system through a number of mechanisms. More recently, studies have also focused on the trimethylamine N-oxide (TMAO) molecule, which reveals a highly significant relationship between diet and gastrointestinal and cardiovascular health.
TMAO is an amine oxide with the formula (CH3)3NO; it is an oxidized form of trimethylamine (TMA). TMAO is primarily formed from nutritional substrates from the metabolism of carnitine, dimethylglycine, phosphatidylcholine, choline, and betaine by intestinal microflora in the colon [115]. These substrates are mainly derived from products of animal origin but may also be of plant origin. Choline- and carnitine-rich foods include animal-based foods such as eggs, dairy, harslet, red meat, poultry, seafood, and fish [116], but carnitine can be found only in limited amounts in plant foods [117,118]; for example, ground beef contains about 400 times more carnitine than whole wheat bread does [119]. TMA may be present in the diet, but its dietary intake is negligible. After the precursors have been transformed into TMA by bacteria, it is absorbed into the bloodstream. TMA can be transformed into TMAO by hepatic enzymes called flavin monooxygenases (FMO1 and FMO3). Unabsorbed TMA is decomposed into methylamine, dimethylamine (DMA), and ammonia within the colon [120].
The biologically active stereoisomer form of carnitine (L-carnitine) participates in fatty acid metabolism, the maintenance of plant and animal cell homeostasis, and signaling pathways in both plants and animals. L-carnitine, derived from the diet or supplements, is absorbed by active and passive transport through intestinal cell membranes. The liver and the kidneys are the main organs responsible for the biosynthesis of carnitine. Carnitine plays an essential role in the transport of long-chain fatty acids into the mitochondria, which is a rate-limiting step in fatty acid oxidation [121]. L-carnitine is more bioavailable for vegetarians, and their daily loss is also minimal due to their adaption to low-carnitine diets compared to those following omnivorous diets [122].
The precursor for TMA production in vegetarians, vegans, and omnivores is phosphatidylcholine, which is the main dietary source of choline. Soy, cruciferous vegetables such as cauliflower, Brussels sprouts, cabbage, etc., and whole grains are plant-based sources of choline. Although Brussels sprouts are the most abundant source of choline among plant foods, after its consumption instead of an increase, a significant decrease in the urinary TMAO profile has been observed. This can be due to two indole-containing compounds that are potent inhibitors of human FMO3 [123]. This study raises the possibility that plant-based choline sources could have different effect on TMAO production compared to animal-based sources due to their phytochemical composition.

TMAO and Its Clinical Importance

According to prospective observational studies, plasma TMAO levels are related to the incidence of cardiovascular diseases. Both in vivo studies in mice and in vitro studies on human cells have suggested that physiological levels of TMAO stimulate the expression of inflammatory cytokines and adhesion molecules [124]. TMAO can induce the formation of foam cells, and it was also found that the formation of foam cells triggered by oxidized LDL was enhanced by TMAO [125,126]. Moreover, clinical studies in patients with heart failure [127,128] and hemodialysis [129] proposed a preatherogenic role for TMAO in the development of atherosclerosis.
Numerous studies suggest that human gut microbiota may be a double-edged sword. Lifestyle and dietary choices can have a beneficial or detrimental effect on the human health by altering the gut microbiota [130,131]. It plays a role in the development of cardiovascular diseases due to its production of TMAO from carnitine and choline. Red meat consumption has been found to be one of the main risk factors for the development of cardiovascular diseases [132]. Researchers have shown that red meat caused the highest TMAO concentration in the blood compared to the concentrations observed in white meat eaters and non-carnivorous (vegetarian) groups [133]. In 2019, a study was carried out in which omnivores and vegans/vegetarians consumed 450 mg/day of choline for about two months. The results showed that the TMAO levels were elevated from the baseline in both groups, although the vegans/vegetarians had much lower TMAO and platelet aggregation than the omnivores. However, as time passed, the microbiomes of the vegans/vegetarians started to adapt to the choline supplementation, which resulted in higher concentrations of TMAO and platelet aggregation by the end of the study. Despite these differences, at the end of the study’s two-month period, the vegans/vegetarians showed elevated but still much lower levels of TMAO than the omnivorous group [134]. Supplementation of TMAO’s substrates may alter TMAO levels in any dietary groups, even those who primarily had non (or less)-TMAO producing gut flora.
Omnivorous individuals ingest 2–12 μmol/kg of carnitine of body weight/day, which provides 75% of the body’s carnitine sources [117,121]. As carnitine is mainly present in animal-based foods, vegetarians and vegans only consume very small amounts of carnitine in their diets (around 1 μmol/kg/day). Therefore, those following plant-based diets obtain more than 90% of their carnitine through biosynthesis [121].
Previous research has reported the cardioprotective effects of plant-based diets [2,4,5,6,7], which may be partially explained by vegetarians’/vegans’ reduced capacity to produce TMAO [135]. A recent study of healthy subjects concluded that TMAO production from carnitine is higher in omnivores than vegans [136]. In this context, the modification of the gut microbiota composition and diversity with plant-based diets appears to be a useful option in the treatment of diseases related to high TMAO levels [137]. In addition, several studies have proven that the decreased TMAO levels can be partially explained by the remodeling potential of plant compounds, which leads to decreased TMA formation. In an animal study, resveratrol-supplemented chow feed altered the gut microbiome in mice, which might have contributed to the decreased gut microbial TMA production [138]. These results could also be corroborated in a randomized placebo-controlled clinical trial in healthy subjects supplemented with resveratrol-containing grape pomace extract. After four weeks supplementation, the TMAO levels were significantly decreased in the experimental group suggesting microbiota remodeling in the gut [139].
Carnitine supplementation in a regular diet can increase TMAO production [140].
In a short-term randomized controlled trial, fecal microbial transplantation from donors following a vegan diet changed the composition of the gut microbiota but did not affect the TMAO production in obese, omnivorous, atherosclerotic patients. Nonetheless, the study had the limitation that the participants insisted on following their own omnivorous diets after this intervention [141].
The level of TMAO production can vary from person to person. Many studies have mentioned that individuals with different phenotypes presumably produce different amounts of TMAO in the gut [136]. The clinical relevance of this claim has not yet been proven, so further research is needed to find out more about the mechanisms of action involved.
Therefore, the findings of these studies show that the gut microbiota may play an important role in plant-based dietary interventions aiming to decrease the risk of developing cardiovascular diseases. Further clinical studies are needed to establish the beneficial effects of plant-based diets, especially for decreasing the level of TMAO production in the gut.

5. Insulin-like Growth Factor-1

In the previous sections, we examined components consumed through the diet or metabolized in the body from dietary components, but we now want to discuss different regulatory mechanisms that are highly influenced by dietary factors.
Human insulin-like growth factor-1 (IGF-1), or somatomedin C, is an anabolic hormone produced by hepatocytes that consists of 70 amino acids in a single polypeptide chain, with intramolecular disulfide bridges. The insulin-like growth factor-1 system includes IGF-1, IGF-1 receptor (IGF-1R), and IGF-binding proteins (IGFBPs) [142].
IGF-1 regulates many functions of cell metabolism, mediates the growth effect of human growth hormone (hGH), promotes the proliferation of many cells, and inhibits apoptosis; as a consequence, it could promote the survival of malignant cells [143,144].
IGF-1’s serum levels progressively decline with age [145] and are influenced by nutrition [146]. The effects of certain nutrients on serum IGF-1 and its expression at the mRNA and protein levels seem to be decisive; in different tissues such as the liver and intestines, the levels of IGF-1 tend to decrease with fasting and are restored with refeeding [147]. Beyond energy intake from different macronutrients, a higher intake of protein seems to regulate the level of IGF-1, which may contribute to a higher body mass in early childhood [148,149,150].
Significant energy and protein restriction (especially restricting the essential amino acids [151]) can reduce IGF-1 plasma levels in both animals and humans in general [152]. The growth hormones IGF-1 and IGFBPs play a role in the growth of children by stimulating the longitudinal growth of the bones [148]. In vegan children, a low and limited intake of essential amino acids and proteins can affect their growth velocity [153]. Recent data suggest that there are no additional nutritional risks among vegan children compared to omnivores [154].
In theory, following high-calorie, low nutrient-dense diets (such as the typical Western-type diet) early in life adversely program the principal components of metabolic syndrome and other conditions by promoting growth acceleration [155]. In contrast, in some earlier studies, relative undernutrition and slower growth (compared to Western-type diet) in early life may result in a lower risk of developing cardiovascular diseases later [156]. The early life environment as well as early nutrition (both undernutrition [157] and overnutrition [158]) can play a key role in the later health and development of different diseases (better known as “Developmental Origins of Health and Disease” (DoHaD) theory [159], or early programming theory [160]), but the complex biochemical processes behind this phenomenon (and the potential role of IGF-1) needs to be clarified. Therefore, optimal early nutrition, with lower calorie and higher nutrient-content compared to Western-type intake, can play a key role in the prevention of developing chronic diseases (e.g., obesity, cardiovascular diseases) in adulthood.

IGF-1 and Cancer

Cancer is one of the leading causes of death globally, and the incidence of cases increased by 33% between 2007 and 2017 [161,162,163].
Data on molecular mechanisms suggest that the activation of different genes combined with IGF-1 signaling can perturb the normal homeostasis of the cell, and these irregularities can be found in different carcinogenic processes [164,165,166,167]. High levels of circulating IGF-1 and IGFBP-3 in the blood are related to a higher risk of certain cancers, such as colorectal, prostate, and breast cancers [168,169,170].
Higher levels of energy and protein intake [171], as well as milk [172,173,174] and meat [175] consumption, are associated with elevated IGF-1 levels, which may increase the risk of developing prostate cancer in men [176,177,178] and breast cancer in women [171,179,180]. By contrast, the consumption of plant-based diets (especially vegan diets with a low consumption of soy milk) can reduce IGF-1 levels [181]. Moreover, both epidemiological studies [179,180] and clinical data [182] have shown that following a plant-based diet (the regular consumption of fruit, vegetables, legumes, or whole grains) may reduce the risk of developing certain cancerous diseases, such as colorectal, prostate, and breast cancer [11,18,183,184,185]. Based on data from the Adventist Health Study 2, which is the study with the largest cohort of vegans and vegetarians to date, the tumor risk was significantly lower in vegans/vegetarians than in non-vegetarians. In both sexes, vegan diets showed a remarkably higher protection against cancerous diseases. The authors explained these differences as being due to, among other factors, the differences in IGF-1 levels [12].
It is not well understood what mediates the positive effect of plant-based diets on the IGF-1 system (plant-based protein, high fruit and vegetable consumption, a high fiber intake, and low fat consumption); further evidence and clinical research are needed. IGF-1 should be targeted for cancer prevention, and the regulation of its level could be a potential therapeutic point in general and lifestyle medicine [186].

6. Mechanistic Target of Rapamycin

In close association with IGF-1, there is another potent regulatory mechanism that is highly influenced by dietary factors: the mechanistic target of rapamycin (mTOR).
mTOR is a serine/threonine protein kinase from the phosphatidylinositol-3-kinase (PI3K) superfamily. It is present in all eukaryotes and plays a key role in regulating cell growth and proliferation, cellular energy levels, oxygen levels, and mitogenic signals [187]. mTOR has two functional units (mTOR complex 1 (mTORC1) and mTOR complex 2 (mTORC2)), which act as central connectors of nutrient signaling pathways and are involved in the regulation of the cell cycle [188]. It was discovered in the 1970s as a result of the search for the target of rapamycin, a macrolide unit produced by Streptomyces hygroscopicus bacteria. Rapamycin is a selective inhibitor of mTOR. Rapamycin inhibits the transcriptional activity of cytokines by suspending their production, but it also has antifungal and antitumor effects, and it is used as an immunosuppressant [189]. Rapamycin appears to have life-extending properties [190,191], but its widespread recreational use has been inhibited by its side-effect profile (hyperlipidemia, high blood sugar, anemia, and inflammation of the oral mucosa) [192]. mTOR could be a key factor in the development and progression of many conditions (such as metabolic diseases, obesity, cancer, and ageing). The manipulation of mTOR mechanisms through dietary interventions seems to be suitable for the prevention and treatment of these conditions.

6.1. mTOR Complex 1

mTORC1 controls major regulatory processes (as a nutrient sensor, responding to dynamic changes in amino acid levels, ATP, and growth factor signaling), apoptosis, and stress responses [191,193,194]. These inputs are capable of synergizing and antagonizing each other, enabling the cell to fine-tune the action of mTORC1. Therefore, the deregulation of mTORC1 activation is associated with many diseases (including cancer and T2DM).
Amino acids such as leucine, arginine, and glutamine are important signals for mTORC1 activation [193,195,196,197]. Moreover, leucine-mediated mTORC1–ribosomal protein S6 kinase beta-1 (S6K1) signaling induces insulin resistance by the phosphorylation of insulin receptor substrate 1 (IRS-1). Leucine-mediated mTORC1–S6K1 signaling also plays an essential role in adipogenesis, thus increasing the risk of obesity-related insulin resistance [194].
In normal conditions, mTORC1 regulates lipid accumulation in fat cells, primarily through storage as white adipose tissue (WAT). The adipocyte-specific deletion of raptor (which is an mTOR binding partner that is necessary for mTOR signal transduction, binding to mTORC1, and the phosphorylation of mTOR-catalyzed substrates) reduces the amount of WAT in adipose-specific raptor knockout mice and increases the oxidation of fatty acids [198]. Accordingly, long-term chronic mTORC1 hyperactivity causes increased lipogenesis in the liver and WAT, which can lead to obesity and insulin resistance [199].

6.2. mTOR Complex 2

mTORC2 plays a key role in cell survival and the regulation of anabolic processes [200,201]; the endogenous activity of mTORC2 is localized to the plasma membrane and mitochondrial and endosomal spaces with distinct sensitivities to phosphoinositide 3-kinase (PI3K) and growth factor signaling, promoting Akt phosphorylation by encouraging Akt to localize to the plasma membrane. The stimulation of cells by insulin promotes the S473 phosphorylation of Akt by mTORC2. In addition, mTORC2 phosphorylates the AGC kinase family members and activates Akt, serum- and glucocorticoid-induced protein kinase (SGK), and protein kinase C (PKC), which regulates cell survival, cell cycle progression, and anabolism [199,202]
mTORC1 and mTORC2 cooperate closely, regulating many different processes (autophagy, apoptosis, neurodevelopment, cell migration, dendritic arborization, and adipocyte formation). Although, mTOR pathways are very diverse in the eukaryotic cells most details of their regulated processes are still unclear [202]. Whether plant-based diets can act via mTOR signaling pathway leading better health and decreasing the risk of chronic diseases needs to be further investigated in human trials.

6.2.1. PI3K–mTOR Pathway in Cancer

The PI3K–Akt–mTOR pathway plays an important role in the regulation of multiple cellular functions, affecting both anabolism and catabolism [194]. The dysregulation of the PI3K–Akt–mTOR pathway is involved in the development of numerous human diseases, such as cancer [203]. mTOR overactivity is mainly caused by the activation of the PI3K–Akt signaling pathway and occurs in virtually every type of tumor cell, including breast cancer cells, colorectal cancer cells, and gastric cancer cells [204]. In the case of breast cancer, the overactivation of this pathway leads to protein synthesis, which, in turn, contributes to the increase in tumor cell proliferation and cell growth. Moreover, increased mTOR signaling can stimulate angiogenesis and may confer resistance to estrogen endocrine therapy [205].
The PI3K–Akt–mTOR pathway is also a strong regulator of autophagy [206] and is involved in the development and promotion of pathological disorders such as cancer. Activated mTORC1 inhibits autophagy through the inhibitory phosphorylation of Unc-51, such as by autophagy activating kinase (ULK1) [193]. Thus, hampering PI3K-Akt-mTOR-mediated autophagy may be an important therapeutic strategy in the treatment of various tumors. The activation of 5′ AMP-activated protein kinase (AMPK) can downregulate ULK1 and, therefore, inhibiting both early and late phase of autophagy [207].

6.2.2. Caloric Restriction, Fasting, and mTOR

The cell senses nutrient supply through different signal transductional pathways (AMPK, recombination-activating gene (Rag), PI3K/Akt, etc.). Different states of nutrition alter the cell’s energetic homeostasis through various pathways (anabolism and catabolism). Amino acids, especially leucine, are capable of inhibiting AMPK, which leads to mTORC1 activation [194]. For this reason, restricting protein and calories can be an option for suppressing mTOR [208]. Short and longer time fasting upregulated in both in vitro cell and in vivo animal study the expression of farnesyl-diphosphate farnesyltransferase 1 (FDFT1), which acts as a critical tumor suppressor in colorectal cancer by negatively regulating Akt–mTOR–hypoxia inducible factor-1α (HIF1α) signaling and, therefore, resulted in slower tumor growth [209].
Animal models and clinical studies showed a wide range of beneficial effect of longer-time caloric restriction for several health conditions (e.g., obesity, T2DM, cardiovascular disease, cancers, neurologic disorder), which appears to be safe if it is conducted appropriately (e.g., intermittent fasting) [210].

6.3. Plant-Based Diets and mTOR

Energy restriction has been recognized as a life-extending factor [211,212,213]. In many cases, the application of energy restriction is impossible or difficult to achieve. The effect of restricting protein consumption on longevity, can be similar to that of energy restriction [214]. In this case, the inhibition of both the IGF-1 and mTOR signaling pathways, as described earlier, may be responsible for an increased life expectancy [215]. Otherwise, a fully plant-based (“vegan”) diet has the possibility to enhance longevity through protein and amino acid restriction and other key mechanisms (fibroblast growth factor 21 (FGF21) induction, gut microbiome diversity), but it has to be confirmed [216].
The quantity and quality of protein consumption can alter mTOR’s activity. As discussed earlier, amino acids (of which leucine has the strongest effect) are important signals for mTORC1 activation [217]. Therefore, a significant reduction in leucine intake reduces the activity of the mTORC1 signaling pathway as much as a reduction in total amino acid intake [218,219] that can be an effective tool for the prevention of several chronic diseases like T2DM, obesity, and cancer [197]. As animal-based foods (meat, eggs, milk, and dairy products) contain the highest levels of leucine, the only way to reduce leucine intake is to eliminate or at least strictly reduce the consumption of these food sources and increase the consumption of plant-derived foods that are low in leucine [194].
Based on different in vitro cell culture data whole-plant food components have inhibitory effect on mTORC1 activation [220]. Furthermore, polyphenols, flavonoids, and curcumin are regarded as natural inhibitors of mTORC1 and exert antidiabetic and anti-obesity effects [221,222,223,224,225]. There is evidence that polyphenol supplementation can effectively reduce fasting blood glucose levels in both T2DM patients or individuals who are at risk of developing diabetes [226]. Anti-obesity effect of different phytochemicals seems promising based on in vitro and animal studies, but evidence for clinical relevance is very low because of the limited number of clinical trials [227].The synergistic effects of these phytonutrients could be much greater than the effects of consuming each component alone or only the active ingredients of whole foods in isolation [228], but more clinical trials are needed that can corroborate this theory.
Despite the fact that there are very limited clinical data available demonstrating the exact relationship between plant-based diets and mTOR, it seems plausible that further studies would confirm the theoretical link between mTOR complexes and dietary factors.

7. Other Factors

7.1. N-Glycolylneuraminic Acid

By consuming animal flesh and meat products, certain molecules that can be found in farm animals but cannot be produced by the human body itself may trigger immune responses and chronic inflammation. This molecular mechanism could be another explanatory factor in the relationship between the consumption of animal-based foods and certain pathological processes.
Sialic acids are well-known molecules that were discussed by numerous research groups in the 1940 and the 1950s [229,230]. Sialic acid and its derivatives are believed to play an important role in immunological processes [231]. The two most common forms of sialic acid in mammals are N-acetylneuraminic acid (Neu5Ac) and N-glycolylneuraminic acid (Neu5Gc) [232]. Human cells cannot synthesize Neu5Gc because of the irreversible mutation of the cytidine monophospho-N-acetylneuraminic acid hydroxylase (CMAH) gene [233]; however, small amounts can be found in different human cells due to dietary intake [234]. Since plants, fungi, and the microorganisms in our bodies are unable to produce Neu5Gc, the primary source of the Neu5Gc molecule in the human body is animal-based foods [233,235] with the highest concentrations in caviar (446–531 μg/g), beef (134–231 μg/g), and lamb (19–57 μg/g) [236].
Neu5Gc exposure causes the production of anti-Neu5Gc antibodies, which can participate in the propagation of autoimmune processes. In addition, most cancerous cells exhibit a high affinity for the accumulation of the Neu5Gc molecule, resulting in the production of anti-Neu5Gc antibodies. Subsequently, the already-mentioned “low-grade inflammation” condition develops, contributing to the survival, further proliferation, and angiogenesis of malignant cells [237]. These mechanisms may also contribute to the association of red meat consumption with the prevalence of T2DM [236].
The Neu5Gc molecule is also highly accumulated in epithelial cells, especially at atherosclerotic sites [233]. Thus, it is suggested that anti-Neu5Gc antibodies can contribute to the progression of atherosclerosis [238]. The potential role of Neu5Gc in the development of anti-Neu5Gc antibody-related diseases is summarized in Figure 3.
Intriguingly, SARS-CoV-2 may use host Neu5Gc molecules, similar to MERS-CoV. A current hypothesis suggests that SARS-CoV-2 has a sialylated glycan shield, which may lend support to the idea that Neu5Gc and its derivatives are possible virus receptors [239].
Although the data described here may be of interest, anti-Neu5Gc-antibody-related disorders need to be confirmed by well-designed human clinical trials [240].

7.2. Endotoxemia

Endotoxins (Lipopolysaccharides (LPS)) are structural components of Gram-negative bacteria and therefore the main targets of antibody production [241] and possible contributors of inflammation [242]. When the LPS level in the blood is elevated, the production of toll-like receptor-2 (TLR2) and toll-like receptor-4 (TLR4) can be stimulated [226]. As TLR4 is one of the activators of the NF-κB protein complex, its inadequate regulation may be related to inflammatory processes when NF-κB stimulates the transcription of proinflammatory genes [227].
In the Bruneck study, LPS was found in healthy individuals in low concentrations (median 14.3 pg/mL), but at levels of 50 pg/mL or above, they increased the risk of atherosclerosis, that was more pronounced in ex-smokers and current smokers [109,243]. Dietary factors, as high saturated fat diet can also increase postprandial plasma levels of LPS leading to inflammatory response [244].
Based on human epidemiological studies, consumption of several prebiotics (e.g.,: insoluble dietary fiber, resistant dextrin, galacto-oligosaccharides, oligofructose, inulin) that is typical in plant-based diets can decrease the level of LPS [245], while many foodstuffs typical for Western-diet (pork, turkey, soft cheese, ice cream, and chocolate) contained TLR2 stimulants in an in vitro study, and, therefore, can induce endotoxemic state [246].
As we mentioned in relation to TMAO, the gut flora have the ability to significantly modify these processes also [247]. The mechanisms underlying the connection between nutrition, microbiome, health, and certain diseases go far beyond the scope of this narrative review, but the use of plant-based diet in this context also seems beneficial [248,249].
Contradictory data are available concerning the relation of on the one hand certain diets and on the other hand endotoxemia and postprandial inflammation [250,251], but it seems that consuming foods rich in saturated fats as well as several food typical for Western-type diet can increase the levels of LPS for a short time, even in healthy subjects [252], while different dietary factors typical for plant-based diets can decrease LPS levels in a few weeks on average [245]. To confirm the relative usefulness of plant-based diets in this context need to be confirmed with more well-designed clinical trials.

8. Summary

The adoption of plant-based diets is becoming popular in the Western world, so it is necessary for practicing healthcare professionals to be well informed about these diets. Support for the implementation of plant-based diets by dietitians will be more effective if they better understand the positive physiological consequences of these diets. While this review is not comprehensive, we have tried to describe some of the huge amount of scientific evidence confirming the efficacy and usefulness of plant-based diets. Thus, explanatory mechanisms could become partly recognizable too. Nevertheless, the exact mechanisms involved in a wide range of positive health effects remain unclear. This review draws attention to the need for further evidence-based, high-quality studies.

Author Contributions

Conceptualization, Z.S.; software, T.M.; writing—original draft preparation, Z.S., V.K., K.F., A.E., E.P. and E.F.; writing—review and editing, T.M., E.S. and Z.V.; supervision, M.F. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by NKFIH a, grant number K-120193 b. This research was supported within the framework of the 2020-4.1.1-TKP2020 3rd thematic program of the University of Pécs. M.T. is supported by the UNKP-20-3-I-PTE-648 New National Excellence Program of the Ministry for Innovation and Technology from the Source of the National Research, Development and Innovation Found. The research was financed and supported by the Thematic Excellence Program 2020—Institutional Excellence Sub-programme/National Excellence Sub-program of the Ministry for Innovation and Technology in Hungary, within the framework of the 2nd thematic programme of the University of Pécs (2020-4.1.1-TEP2020).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Acknowledgments

The authors would like to thank Gábor Varga, an IT specialist at Faculty of Health Sciences, University of Pecs, Pecs, Hungary, for the preparation of the graphical abstract.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Ostfeld, R.J. Definition of a plant-based diet and overview of this special issue. J. Geriatr. Cardiol. 2017, 14, 315. [Google Scholar] [CrossRef]
  2. Hu, F.B. Plant-based foods and prevention of cardiovascular disease: An overview. Am. J. Clin. Nutr. 2003, 78, 544S–551S. [Google Scholar] [CrossRef] [Green Version]
  3. Dinu, M.; Abbate, R.; Gensini, G.F.; Casini, A.; Sofi, F. Vegetarian, vegan diets and multiple health outcomes: A systematic review with meta-analysis of observational studies. Crit. Rev. Food. Sci. Nutr. 2017, 57, 3640–3649. [Google Scholar] [CrossRef] [PubMed]
  4. Orlich, M.J.; Singh, P.N.; Sabate, J.; Jaceldo-Siegl, K.; Fan, J.; Knutsen, S.; Beeson, W.L.; Fraser, G.E. Vegetarian dietary patterns and mortality in Adventist Health Study 2. JAMA Intern. Med. 2013, 173, 1230–1238. [Google Scholar] [CrossRef]
  5. Ornish, D.; Scherwitz, L.W.; Billings, J.H.; Brown, S.E.; Gould, K.L.; Merritt, T.A.; Sparler, S.; Armstrong, W.T.; Ports, T.A.; Kirkeeide, R.L.; et al. Intensive lifestyle changes for reversal of coronary heart disease. JAMA 1998, 280, 2001–2007. [Google Scholar] [CrossRef] [PubMed]
  6. Kahleova, H.; Levin, S.; Barnard, N. Cardio-Metabolic Benefits of Plant-Based Diets. Nutrients 2017, 9, 848. [Google Scholar] [CrossRef] [PubMed]
  7. Kim, H.; Caulfield, L.E.; Garcia-Larsen, V.; Steffen, L.M.; Coresh, J.; Rebholz, C.M. Plant-Based Diets Are Associated With a Lower Risk of Incident Cardiovascular Disease, Cardiovascular Disease Mortality, and All-Cause Mortality in a General Population of Middle-Aged Adults. J. Am. Heart Assoc. 2019, 8, e012865. [Google Scholar] [CrossRef] [PubMed]
  8. Tran, E.; Dale, H.F.; Jensen, C.; Lied, G.A. Effects of Plant-Based Diets on Weight Status: A Systematic Review. Diabetes Metab. Syndr. Obes. Targets Ther. 2020, 13, 3433–3448. [Google Scholar] [CrossRef] [PubMed]
  9. Huang, R.Y.; Huang, C.C.; Hu, F.B.; Chavarro, J.E. Vegetarian Diets and Weight Reduction: A Meta-Analysis of Randomized Controlled Trials. J. Gen. Intern. Med. 2016, 31, 109–116. [Google Scholar] [CrossRef] [Green Version]
  10. Anand, P.; Kunnumakkara, A.B.; Sundaram, C.; Harikumar, K.B.; Tharakan, S.T.; Lai, O.S.; Sung, B.; Aggarwal, B.B. Cancer is a preventable disease that requires major lifestyle changes. Pharm. Res. 2008, 25, 2097–2116. [Google Scholar] [CrossRef]
  11. Huang, T.; Yang, B.; Zheng, J.; Li, G.; Wahlqvist, M.L.; Li, D. Cardiovascular disease mortality and cancer incidence in vegetarians: A meta-analysis and systematic review. Ann. Nutr. Metab. 2012, 60, 233–240. [Google Scholar] [CrossRef] [PubMed]
  12. Tantamango-Bartley, Y.; Jaceldo-Siegl, K.; Fan, J.; Fraser, G. Vegetarian Diets and the Incidence of Cancer in a Low-risk Population. Cancer Epidemiol. Biomark. 2013, 22, 286–294. [Google Scholar] [CrossRef] [Green Version]
  13. Kahleova, H.; Matoulek, M.; Malinska, H.; Oliyarnik, O.; Kazdova, L.; Neskudla, T.; Skoch, A.; Hajek, M.; Hill, M.; Kahle, M.; et al. Vegetarian diet improves insulin resistance and oxidative stress markers more than conventional diet in subjects with Type 2 diabetes. Diabet. Med. 2011, 28, 549–559. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Tonstad, S.; Butler, T.; Yan, R.; Fraser, G.E. Type of vegetarian diet, body weight, and prevalence of type 2 diabetes. Diabetes Care 2009, 32, 791–796. [Google Scholar] [CrossRef] [Green Version]
  15. Qian, F.; Liu, G.; Hu, F.B.; Bhupathiraju, S.N.; Sun, Q. Association between Plant-Based Dietary Patterns and Risk of Type 2 Diabetes: A Systematic Review and Meta-analysis. JAMA Intern. Med. 2019, 179, 1335–1344. [Google Scholar] [CrossRef]
  16. Campbell, T. A plant-based diet and stroke. J. Geriatr. Cardiol. 2017, 14, 321–326. [Google Scholar] [CrossRef]
  17. Baden, M.Y.; Shan, Z.; Wang, F.; Li, Y.; Manson, J.E.; Rimm, E.B.; Willett, W.C.; Hu, F.B.; Rexrode, K.M. Quality of Plant-based Diet and Risk of Total, Ischemic, and Hemorrhagic Stroke. Neurology 2021, 96, e1940–e1953. [Google Scholar] [CrossRef]
  18. Melina, V.; Craig, W.; Levin, S. Position of the Academy of Nutrition and Dietetics: Vegetarian Diets. J. Acad. Nutr. Diet. 2016, 116, 1970–1980. [Google Scholar] [CrossRef]
  19. American Diabetes, A. 4. Lifestyle Management. Diabetes Care 2017, 40, S33–S43. [Google Scholar] [CrossRef] [Green Version]
  20. Garton, L.; Hood, S. Food Fact Sheet Plant-Based Diet; The British Dietetic Association (BDA): Birmingham, UK. Available online: https://www.bda.uk.com/uploads/assets/3f9e2928-ca7a-4c1e-95b87c839d2ee8a1/Plant-based-diet-food-fact-sheet.pdf (accessed on 27 July 2021).
  21. American Dietetic, A.; Dietitians of, C. Position of the American Dietetic Association and Dietitians of Canada: Vegetarian diets. J. Am. Diet. Assoc. 2003, 103, 748–765. [Google Scholar] [CrossRef]
  22. Gomes Silva, C.S.; Pinho, J.P.; Borges, C.; Teixeira Santos, C.; Santos, A.; Graca, P. Guidelines for a Healthy Vegetarian Diet; Direção-Geral da Saúde: Lisbon, Portugal, 2015; pp. 1–45. [Google Scholar]
  23. Richter, M.; Boeing, H.; Grünewald-Funk, D.; Heseker, H.; Kroke, A.; Leschik-Bonnet, E.; Oberritter, H.; Strohm, D.; Watzl, B. Vegan Diet Position of the German Nutrition Society (DGE). Ernaehrungs Umsch. Int. 2016, 63, 11. [Google Scholar] [CrossRef]
  24. Burton-Freeman, B. Postprandial metabolic events and fruit-derived phenolics: A review of the science. Br. J. Nutr. 2010, 104 (Suppl. S3), S1–S14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Emerson, S.R.; Kurti, S.P.; Harms, C.A.; Haub, M.D.; Melgarejo, T.; Logan, C.; Rosenkranz, S.K. Magnitude and Timing of the Postprandial Inflammatory Response to a High-Fat Meal in Healthy Adults: A Systematic Review. Adv. Nutr. 2017, 8, 213–225. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Tan, B.L.; Norhaizan, M.E.; Liew, W.P. Nutrients and Oxidative Stress: Friend or Foe? Oxid. Med. Cell Longev. 2018, 2018, 9719584. [Google Scholar] [CrossRef]
  27. Vogel, R.A.; Corretti, M.C.; Plotnick, G.D. Effect of a single high-fat meal on endothelial function in healthy subjects. Am. J. Cardiol. 1997, 79, 350–354. [Google Scholar] [CrossRef]
  28. Burton-Freeman, B.; Linares, A.; Hyson, D.; Kappagoda, T. Strawberry modulates LDL oxidation and postprandial lipemia in response to high-fat meal in overweight hyperlipidemic men and women. J. Am. Coll. Nutr. 2010, 29, 46–54. [Google Scholar] [CrossRef]
  29. Prior, R.L.; Go, L.W.; Wu, X.L.; Jacob, R.A.; Sotoudeh, G.; Kader, A.A.; Cook, R.A. Plasma antioxidant capacity changes following a meal as a measure of the ability of a food to alter in vivo antioxidant status. J. Am. Coll. Nutr. 2007, 26, 170–181. [Google Scholar] [CrossRef] [PubMed]
  30. Peng, C.; Wang, X.; Chen, J.; Jiao, R.; Wang, L.; Li, Y.M.; Zuo, Y.; Liu, Y.; Lei, L.; Ma, K.Y.; et al. Biology of ageing and role of dietary antioxidants. Biomed Res. Int. 2014, 2014, 831841. [Google Scholar] [CrossRef] [Green Version]
  31. Winiarska-Mieczan, A.; Baranowska-Wojcik, E.; Kwiecien, M.; Grela, E.R.; Szwajgier, D.; Kwiatkowska, K.; Kiczorowska, B. The Role of Dietary Antioxidants in the Pathogenesis of Neurodegenerative Diseases and Their Impact on Cerebral Oxidoreductive Balance. Nutrients 2020, 12, 435. [Google Scholar] [CrossRef] [Green Version]
  32. Carlsen, M.H.; Halvorsen, B.L.; Holte, K.; Bohn, S.K.; Dragland, S.; Sampson, L.; Willey, C.; Senoo, H.; Umezono, Y.; Sanada, C.; et al. The total antioxidant content of more than 3100 foods, beverages, spices, herbs and supplements used worldwide. Nutr. J. 2010, 9, 3. [Google Scholar] [CrossRef]
  33. Pandey, K.B.; Rizvi, S.I. Plant polyphenols as dietary antioxidants in human health and disease. Oxid. Med. Cell. Longev. 2009, 2, 270–278. [Google Scholar] [CrossRef] [Green Version]
  34. Kanner, J.; Lapidot, T. The stomach as a bioreactor: Dietary lipid peroxidation in the gastric fluid and the effects of plant-derived antioxidants. Free Radic. Biol. Med. 2001, 31, 1388–1395. [Google Scholar] [CrossRef]
  35. Miranda-Diaz, A.G.; Garcia-Sanchez, A.; Cardona-Munoz, E.G. Foods with Potential Prooxidant and Antioxidant Effects Involved in Parkinson’s Disease. Oxid. Med. Cell. Longev. 2020, 2020, 6281454. [Google Scholar] [CrossRef] [PubMed]
  36. Keshavarzian, A.; Banan, A.; Farhadi, A.; Komanduri, S.; Mutlu, E.; Zhang, Y.; Fields, J.Z. Increases in free radicals and cytoskeletal protein oxidation and nitration in the colon of patients with inflammatory bowel disease. Gut 2003, 52, 720–728. [Google Scholar] [CrossRef] [Green Version]
  37. Ding, Y.Y.; Li, Z.Q.; Cheng, X.R.; Ran, Y.M.; Wu, S.J.; Shi, Y.; Le, G. Dityrosine administration induces dysfunction of insulin secretion accompanied by diminished thyroid hormones T3 function in pancreas of mice. Amino Acids 2017, 49, 1401–1414. [Google Scholar] [CrossRef] [PubMed]
  38. Rouhier, N.; Lemaire, S.D.; Jacquot, J.P. The role of glutathione in photosynthetic organisms: Emerging functions for glutaredoxins and glutathionylation. Annu. Rev. Plant Biol. 2008, 59, 143–166. [Google Scholar] [CrossRef]
  39. Sato, K.; Niki, E.; Shimasaki, H. Free radical-mediated chain oxidation of low density lipoprotein and its synergistic inhibition by vitamin E and vitamin C. Arch. Biochem. Biophys. 1990, 279, 402–405. [Google Scholar] [CrossRef]
  40. Bakuradze, T.; Tausend, A.; Galan, J.; Groh, I.A.M.; Berry, D.; Tur, J.A.; Marko, D.; Richling, E. Antioxidative activity and health benefits of anthocyanin-rich fruit juice in healthy volunteers. Free Radic. Res. 2019, 53, 1045–1055. [Google Scholar] [CrossRef] [PubMed]
  41. Juraschek, S.P.; Guallar, E.; Appel, L.J.; Miller, E.R., III. Effects of vitamin C supplementation on blood pressure: A meta-analysis of randomized controlled trials. Am. J. Clin. Nutr. 2012, 95, 1079–1088. [Google Scholar] [CrossRef]
  42. Ashor, A.W.; Lara, J.; Mathers, J.C.; Siervo, M. Effect of vitamin C on endothelial function in health and disease: A systematic review and meta-analysis of randomised controlled trials. Atherosclerosis 2014, 235, 9–20. [Google Scholar] [CrossRef] [PubMed]
  43. Traber, M.G.; Stevens, J.F. Vitamins C and E: Beneficial effects from a mechanistic perspective. Free Radic. Biol. Med. 2011, 51, 1000–1013. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Saito, Y.; Nishio, K.; Akazawa, Y.O.; Yamanaka, K.; Miyama, A.; Yoshida, Y.; Noguchi, N.; Niki, E. Cytoprotective effects of vitamin E homologues against glutamate-induced cell death in immature primary cortical neuron cultures: Tocopherols and tocotrienols exert similar effects by antioxidant function. Free Radic. Biol. Med. 2010, 49, 1542–1549. [Google Scholar] [CrossRef]
  45. Ulatowski, L.M.; Manor, D. Vitamin E and neurodegeneration. Neurobiol. Dis. 2015, 84, 78–83. [Google Scholar] [CrossRef] [PubMed]
  46. Lebold, K.M.; Lohr, C.V.; Barton, C.L.; Miller, G.W.; Labut, E.M.; Tanguay, R.L.; Traber, M.G. Chronic vitamin E deficiency promotes vitamin C deficiency in zebrafish leading to degenerative myopathy and impaired swimming behavior. Comp. Biochem. Physiol. Toxicol. Pharmacol. 2013, 157, 382–389. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Jung, H.; Chen, C.O.; Blumberg, J.B.; Kwak, H.K. The effect of almonds on vitamin E status and cardiovascular risk factors in Korean adults: A randomized clinical trial. Eur. J. Nutr. 2018, 57, 2069–2079. [Google Scholar] [CrossRef] [Green Version]
  48. Milani, A.; Basirnejad, M.; Shahbazi, S.; Bolhassani, A. Carotenoids: Biochemistry, pharmacology and treatment. Br. J. Pharmacol. 2017, 174, 1290–1324. [Google Scholar] [CrossRef] [Green Version]
  49. Giaconi, J.A.; Yu, F.; Stone, K.L.; Pedula, K.L.; Ensrud, K.E.; Cauley, J.A.; Hochberg, M.C.; Coleman, A.L. The Association of Consumption of Fruits/Vegetables With Decreased Risk of Glaucoma Among Older African-American Women in the Study of Osteoporotic Fractures. Am. J. Ophthalmol. 2012, 154, 635–644. [Google Scholar] [CrossRef] [Green Version]
  50. Ozawa, Y.; Sasaki, M.; Takahashi, N.; Kamoshita, M.; Miyake, S.; Tsubota, K. Neuroprotective Effects of Lutein in the Retina. Curr. Pharm. Des. 2012, 18, 51–56. [Google Scholar] [CrossRef] [Green Version]
  51. Müller, L.; Caris-Veyrat, C.; Lowe, G.; Böhm, V. Lycopene and Its Antioxidant Role in the Prevention of Cardiovascular Diseases—A Critical Review. Crit. Rev. Food Sci. Nutr. 2015, 56, 1868–1879. [Google Scholar] [CrossRef]
  52. Gärtner, C.; Stahl, W.; Sies, H. Lycopene is more bioavailable from tomato paste than from fresh tomatoes. Am. J. Clin. Nutr. 1997, 66, 116–122. [Google Scholar] [CrossRef]
  53. Rizwan, M.; Rodriguez-Blanco, I.; Harbottle, A.; Birch-Machin, M.A.; Watson, R.E.B.; Rhodes, L.E. Tomato paste rich in lycopene protects against cutaneous photodamage in humans in vivo: A randomized controlled trial. Br. J. Dermatol. 2011, 164, 154–162. [Google Scholar] [CrossRef]
  54. Daniels, J.A.; Mulligan, C.; McCance, D.; Woodside, J.V.; Patterson, C.; Young, I.S.; McEneny, J. A randomised controlled trial of increasing fruit and vegetable intake and how this influences the carotenoid concentration and activities of PON-1 and LCAT in HDL from subjects with type 2 diabetes. Cardiovasc. Diabetol. 2014, 13, 16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Tang, X.; Yang, X.; Peng, Y.; Lin, J. Protective effects of lycopene against H2O2-induced oxidative injury and apoptosis in human endothelial cells. Cardiovasc. Drugs. Ther. 2009, 23, 439–448. [Google Scholar] [CrossRef]
  56. Omenn, G.S.; Goodman, G.E.; Thornquist, M.D.; Balmes, J.; Cullen, M.R.; Glass, A.; Keogh, J.P.; Meyskens, F.L., Jr.; Valanis, B.; Williams, J.H., Jr.; et al. Risk factors for lung cancer and for intervention effects in CARET, the Beta-Carotene and Retinol Efficacy Trial. J. Natl. Cancer Inst. 1996, 88, 1550–1559. [Google Scholar] [CrossRef]
  57. Alpha-Tocopherol Beta Carotene Cancer Prevention Study Group. The effect of vitamin E and beta carotene on the incidence of lung cancer and other cancers in male smokers. N. Engl. J. Med. 1994, 330, 1029–1035. [Google Scholar] [CrossRef]
  58. Middha, P.; Weinstein, S.J.; Mannisto, S.; Albanes, D.; Mondul, A.M. β-Carotene Supplementation and Lung Cancer Incidence in the Alpha-Tocopherol, Beta-Carotene Cancer Prevention Study: The Role of Tar and Nicotine. Nicotine Tob. Res. Off. J. Soc. Res. Nicotine Tob. 2019, 21, 1045–1050. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Bradbury, K.E.; Appleby, P.N.; Key, T.J. Fruit, vegetable, and fiber intake in relation to cancer risk: Findings from the European Prospective Investigation into Cancer and Nutrition (EPIC). Am. J. Clin. Nutr. 2014, 100, 394S–398S. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Gnagnarella, P.; Maisonneuve, P.; Bellomi, M.; Rampinelli, C.; Bertolotti, R.; Spaggiari, L.; Palli, D.; Veronesi, G. Nutrient intake and nutrient patterns and risk of lung cancer among heavy smokers: Results from the COSMOS screening study with annual low-dose CT. Eur. J. Epidemiol. 2013, 28, 503–511. [Google Scholar] [CrossRef] [PubMed]
  61. Takata, Y.; Xiang, Y.-B.; Yang, G.; Li, H.; Gao, J.; Cai, H.; Gao, Y.-T.; Zheng, W.; Shu, X.-O. Intakes of Fruits, Vegetables, and Related Vitamins and Lung Cancer Risk: Results from the Shanghai Men’s Health Study (2002–2009). Nutr. Cancer 2013, 65, 51–61. [Google Scholar] [CrossRef] [PubMed]
  62. Sluijs, I.; Cadier, E.; Beulens, J.W.; van der, A.D.; Spijkerman, A.M.; van der Schouw, Y.T. Dietary intake of carotenoids and risk of type 2 diabetes. Nutr. Metab. Cardiovasc. Dis. 2015, 25, 376–381. [Google Scholar] [CrossRef]
  63. Beydoun, M.A.; Chen, X.; Jha, K.; Beydoun, H.A.; Zonderman, A.B.; Canas, J.A. Carotenoids, vitamin A, and their association with the metabolic syndrome: A systematic review and meta-analysis. Nutr. Rev. 2019, 77, 32–45. [Google Scholar] [CrossRef] [PubMed]
  64. Del Rio, D.; Rodriguez-Mateos, A.; Spencer, J.P.E.; 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, 1818–1892. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Anantharaju, P.G.; Gowda, P.C.; Vimalambike, M.G.; Madhunapantula, S.V. An overview on the role of dietary phenolics for the treatment of cancers. Nutr. J. 2016, 15, 99. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Losada-Barreiro, S.; Bravo-Diaz, C. Free radicals and polyphenols: The redox chemistry of neurodegenerative diseases. Eur. J. Med. Chem. 2017, 133, 379–402. [Google Scholar] [CrossRef]
  67. Kostyuk, V.A.; Potapovich, A.I.; Strigunova, E.N.; Kostyuk, T.V.; Afanas’ev, I.B. Experimental evidence that flavonoid metal complexes may act as mimics of superoxide dismutase. Arch. Biochem. Biophys. 2004, 428, 204–208. [Google Scholar] [CrossRef] [PubMed]
  68. Olszowy, M. What is responsible for antioxidant properties of polyphenolic compounds from plants? Plant Physiol. Biochem. 2019, 144, 135–143. [Google Scholar] [CrossRef]
  69. Hussain, T.; Tan, B.; Yin, Y.; Blachier, F.; Tossou, M.C.; Rahu, N. Oxidative Stress and Inflammation: What Polyphenols Can Do for Us? Oxid. Med. Cell. Longev. 2016, 2016, 7432797. [Google Scholar] [CrossRef] [Green Version]
  70. Urquiaga, I.; Avila, F.; Echeverria, G.; Perez, D.; Trejo, S.; Leighton, F. A Chilean Berry Concentrate Protects against Postprandial Oxidative Stress and Increases Plasma Antioxidant Activity in Healthy Humans. Oxid. Med. Cell. Longev. 2017, 2017, 8361493. [Google Scholar] [CrossRef] [Green Version]
  71. Li, Z.; Wong, A.; Henning, S.M.; Zhang, Y.; Jones, A.; Zerlin, A.; Thames, G.; Bowerman, S.; Tseng, C.H.; Heber, D. Hass avocado modulates postprandial vascular reactivity and postprandial inflammatory responses to a hamburger meal in healthy volunteers. Food Funct. 2013, 4, 384–391. [Google Scholar] [CrossRef] [Green Version]
  72. Tongtako, W.; Klaewsongkram, J.; Mickleborough, T.D.; Suksom, S. Effects of aerobic exercise and vitamin C supplementation on rhinitis symptoms in allergic rhinitis patients. Asian Pac. J. Allergy Immunol. 2018, 36, 222–231. [Google Scholar] [CrossRef] [PubMed]
  73. Pearson, P.J.K. Vitamin E supplements in asthma: A parallel group randomised placebo controlled trial. Thorax 2004, 59, 652–656. [Google Scholar] [CrossRef] [Green Version]
  74. Bjelakovic, G.; Nikolova, D.; Gluud, C. Antioxidant supplements and mortality. Curr. Opin. Clin. Nutr. Metab. Care 2014, 17, 40–44. [Google Scholar] [CrossRef]
  75. Sesso, H.D.; Buring, J.E.; Christen, W.G.; Kurth, T.; Belanger, C.; MacFadyen, J.; Bubes, V.; Manson, J.E.; Glynn, R.J.; Gaziano, J.M. Vitamins E and C in the prevention of cardiovascular disease in men: The Physicians’ Health Study II randomized controlled trial. JAMA 2008, 300, 2123–2133. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Qiao, Y.L.; Dawsey, S.M.; Kamangar, F.; Fan, J.H.; Abnet, C.C.; Sun, X.D.; Johnson, L.L.; Gail, M.H.; Dong, Z.W.; Yu, B.; et al. Total and cancer mortality after supplementation with vitamins and minerals: Follow-up of the Linxian General Population Nutrition Intervention Trial. J. Natl. Cancer Inst. 2009, 101, 507–518. [Google Scholar] [CrossRef]
  77. Sadowska-Bartosz, I.; Bartosz, G. Effect of Antioxidants Supplementation on Aging and Longevity. BioMed Res. Int. 2014, 2014, 1–17. [Google Scholar] [CrossRef] [PubMed]
  78. Long, J.; Guo, Y.; Yang, J.; Henning, S.M.; Lee, R.P.; Rasmussen, A.; Zhang, L.; Lu, Q.Y.; Heber, D.; Li, Z. Bioavailability and bioactivity of free ellagic acid compared to pomegranate juice. Food Funct. 2019, 10, 6582–6588. [Google Scholar] [CrossRef]
  79. Samec, D.; Urlic, B.; Salopek-Sondi, B. Kale (Brassica oleracea var. acephala) as a superfood: Review of the scientific evidence behind the statement. Crit. Rev. Food. Sci. Nutr. 2019, 59, 2411–2422. [Google Scholar] [CrossRef]
  80. Wood, L.G.; Garg, M.L.; Smart, J.M.; Scott, H.A.; Barker, D.; Gibson, P.G. Manipulating antioxidant intake in asthma: A randomized controlled trial. Am. J. Clin. Nutr. 2012, 96, 534–543. [Google Scholar] [CrossRef] [Green Version]
  81. Maleki, S.J.; Crespo, J.F.; Cabanillas, B. Anti-inflammatory effects of flavonoids. Food Chem. 2019, 299, 125124. [Google Scholar] [CrossRef]
  82. Cao, C.; Xiao, Z.; Wu, Y.; Ge, C. Diet and Skin Aging—From the Perspective of Food Nutrition. Nutrients 2020, 12, 870. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Schagen, S.K.; Zampeli, V.A.; Makrantonaki, E.; Zouboulis, C.C. Discovering the link between nutrition and skin aging. Dermato-Endocrinology 2014, 4, 298–307. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Kang, J.H.; Ascherio, A.; Grodstein, F. Fruit and vegetable consumption and cognitive decline in aging women. Ann. Neurol. 2005, 57, 713–720. [Google Scholar] [CrossRef] [PubMed]
  85. Wang, S.; Meckling, K.A.; Marcone, M.F.; Kakuda, Y.; Tsao, R. Synergistic, additive, and antagonistic effects of food mixtures on total antioxidant capacities. J. Agric. Food Chem. 2011, 59, 960–968. [Google Scholar] [CrossRef]
  86. Jacobs, D.R., Jr.; Gross, M.D.; Tapsell, L.C. Food synergy: An operational concept for understanding nutrition. Am. J. Clin. Nutr. 2009, 89, 1543S–1548S. [Google Scholar] [CrossRef] [PubMed]
  87. Stonehouse, W.; Gammon, C.S.; Beck, K.L.; Conlon, C.A.; von Hurst, P.R.; Kruger, R. Kiwifruit: Our daily prescription for health. Can. J. Physiol. Pharmacol. 2013, 91, 442–447. [Google Scholar] [CrossRef] [PubMed]
  88. Carr, A.; Bozonet, S.; Pullar, J.; Simcock, J.; Vissers, M. A Randomized Steady-State Bioavailability Study of Synthetic versus Natural (Kiwifruit-Derived) Vitamin C. Nutrients 2013, 5, 3684–3695. [Google Scholar] [CrossRef] [Green Version]
  89. Trautwein, E.A.; McKay, S. The Role of Specific Components of a Plant-Based Diet in Management of Dyslipidemia and the Impact on Cardiovascular Risk. Nutrients 2020, 12, 2671. [Google Scholar] [CrossRef]
  90. Engin, A.B. What Is Lipotoxicity? In Obesity and Lipotoxicity; Engin, A.B., Engin, A., Eds.; Springer International Publishing: Cham, Switzerland, 2017; pp. 197–220. [Google Scholar] [CrossRef]
  91. Kraegen, E.W.; Cooney, G.J. Free fatty acids and skeletal muscle insulin resistance. Curr. Opin. Lipidol. 2008, 19, 235–241. [Google Scholar] [CrossRef]
  92. Sharma, R.B.; Alonso, L.C. Lipotoxicity in the Pancreatic Beta Cell: Not Just Survival and Function, but Proliferation as Well? Curr. Diabetes Rep. 2014, 14, 492. [Google Scholar] [CrossRef] [Green Version]
  93. Sokolowska, E.; Blachnio-Zabielska, A. The Role of Ceramides in Insulin Resistance. Front. Endocrinol. 2019, 10, 577. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Krssak, M.; Falk Petersen, K.; Dresner, A.; DiPietro, L.; Vogel, S.M.; Rothman, D.L.; Roden, M.; Shulman, G.I. Intramyocellular lipid concentrations are correlated with insulin sensitivity in humans: A 1H NMR spectroscopy study. Diabetologia 1999, 42, 113–116. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Goff, L.M.; Bell, J.D.; So, P.W.; Dornhorst, A.; Frost, G.S. Veganism and its relationship with insulin resistance and intramyocellular lipid. Eur. J. Clin. Nutr. 2005, 59, 291–298. [Google Scholar] [CrossRef] [Green Version]
  96. Gojda , J.; Patkova , J.; Jaček, M.; Potočková, J.; Trnka, J.; Kraml, P.; Anděl, M. Higher insulin sensitivity in vegans is not associated with higher mitochondrial density. Eur. J. Clin. Nutr. 2013, 67, 1310–1315. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Kahleova, H.; Tura, A.; Hill, M.; Holubkov, R.; Barnard, N.D. A Plant-Based Dietary Intervention Improves Beta-Cell Function and Insulin Resistance in Overweight Adults: A 16-Week Randomized Clinical Trial. Nutrients 2018, 10, 189. [Google Scholar] [CrossRef] [Green Version]
  98. Hall, K.D.; Guo, J.; Courville, A.B.; Boring, J.; Brychta, R.; Chen, K.Y.; Darcey, V.; Forde, C.G.; Gharib, A.M.; Gallagher, I.; et al. Effect of a plant-based, low-fat diet versus an animal-based, ketogenic diet on ad libitum energy intake. Nat. Med. 2021, 27, 344–353. [Google Scholar] [CrossRef]
  99. Alvarez-Garcia, O.; Rogers, N.H.; Smith, R.G.; Lotz, M.K. Palmitate Has Proapoptotic and Proinflammatory Effects on Articular Cartilage and Synergizes With Interleukin-1. Arthritis Rheumatol. 2014, 66, 1779–1788. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  100. Nishi, H.; Higashihara, T.; Inagi, R. Lipotoxicity in Kidney, Heart, and Skeletal Muscle Dysfunction. Nutrients 2019, 11, 1664. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Ge, M.; Fontanesi, F.; Merscher, S.; Fornoni, A. The Vicious Cycle of Renal Lipotoxicity and Mitochondrial Dysfunction. Front. Physiol. 2020, 11, 732. [Google Scholar] [CrossRef]
  102. Svegliati-Baroni, G.; Pierantonelli, I.; Torquato, P.; Marinelli, R.; Ferreri, C.; Chatgilialoglu, C.; Bartolini, D.; Galli, F. Lipidomic biomarkers and mechanisms of lipotoxicity in non-alcoholic fatty liver disease. Free Radic. Biol. Med. 2019, 144, 293–309. [Google Scholar] [CrossRef] [PubMed]
  103. Godoy-Matos, A.F.; Silva Junior, W.S.; Valerio, C.M. NAFLD as a continuum: From obesity to metabolic syndrome and diabetes. Diabetol. Metab. Syndr. 2020, 12, 60. [Google Scholar] [CrossRef]
  104. Oddy, W.H.; Herbison, C.E.; Jacoby, P.; Ambrosini, G.L.; O’Sullivan, T.A.; Ayonrinde, O.T.; Olynyk, J.K.; Black, L.J.; Beilin, L.J.; Mori, T.A.; et al. The Western Dietary Pattern Is Prospectively Associated With Nonalcoholic Fatty Liver Disease in Adolescence. Am. J. Gastroenterol. 2013, 108, 778–785. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Yao, H.R.; Liu, J.; Plumeri, D.; Cao, Y.B.; He, T.; Lin, L.; Li, Y.; Jiang, Y.Y.; Li, J.; Shang, J. Lipotoxicity in HepG2 cells triggered by free fatty acids. Am. J. Transl. Res. 2011, 3, 284–291. [Google Scholar]
  106. Ye, J. Role of insulin in the pathogenesis of free fatty acid-induced insulin resistance in skeletal muscle. Endocr. Metab. Immune Disord. Drug Targets 2007, 7, 65–74. [Google Scholar] [CrossRef]
  107. Frohnert, B.I.; Jacobs, D.R.; Steinberger, J.; Moran, A.; Steffen, L.M.; Sinaiko, A.R. Relation Between Serum Free Fatty Acids and Adiposity, Insulin Resistance, and Cardiovascular Risk Factors From Adolescence to Adulthood. Diabetes 2013, 62, 3163–3169. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Capurso, C.; Capurso, A. From excess adiposity to insulin resistance: The role of free fatty acids. Vasc. Pharm. 2012, 57, 91–97. [Google Scholar] [CrossRef]
  109. Estadella, D.; da Penha Oller do Nascimento, C.; Oyama, L.M.; Ribeiro, E.B.; Damaso, A.R.; de Piano, A. Lipotoxicity: Effects of dietary saturated and transfatty acids. Mediat. Inflamm. 2013, 2013, 137579. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Makinen, S.; Nguyen, Y.H.; Skrobuk, P.; Koistinen, H.A. Palmitate and oleate exert differential effects on insulin signalling and glucose uptake in human skeletal muscle cells. Endocr. Connect. 2017, 6, 331–339. [Google Scholar] [CrossRef] [Green Version]
  111. McCarthy, E.M.; Rinella, M.E. The role of diet and nutrient composition in nonalcoholic Fatty liver disease. J. Acad. Nutr. Diet. 2012, 112, 401–409. [Google Scholar] [CrossRef]
  112. Mazidi, M.; Kengne, A.P. Higher adherence to plant-based diets are associated with lower likelihood of fatty liver. Clin. Nutr. 2019, 38, 1672–1677. [Google Scholar] [CrossRef]
  113. Alferink, L.J.M.; Erler, N.S.; de Knegt, R.J.; Janssen, H.L.A.; Metselaar, H.J.; Darwish Murad, S.; Kiefte-de Jong, J.C. Adherence to a plant-based, high-fibre dietary pattern is related to regression of non-alcoholic fatty liver disease in an elderly population. Eur. J. Epidemiol. 2020, 35, 1069–1085. [Google Scholar] [CrossRef] [Green Version]
  114. Geng, Y.; Faber, K.N.; de Meijer, V.E.; Blokzijl, H.; Moshage, H. How does hepatic lipid accumulation lead to lipotoxicity in non-alcoholic fatty liver disease? Hepatol. Int. 2021, 15, 21–35. [Google Scholar] [CrossRef]
  115. Wang, Z.; Tang, W.H.W.; Buffa, J.A.; Fu, X.; Britt, E.B.; Koeth, R.A.; Levison, B.S.; Fan, Y.; Wu, Y.; Hazen, S.L. Prognostic value of choline and betaine depends on intestinal microbiota-generated metabolite trimethylamine-N-oxide. Eur. Heart J. 2014, 35, 904–910. [Google Scholar] [CrossRef] [PubMed]
  116. Wang, Z.; Klipfell, E.; Bennett, B.J.; Koeth, R.; Levison, B.S.; Dugar, B.; Feldstein, A.E.; Britt, E.B.; Fu, X.; Chung, Y.M.; et al. Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature 2011, 472, 57–63. [Google Scholar] [CrossRef] [Green Version]
  117. Flanagan, J.L.; Simmons, P.A.; Vehige, J.; Willcox, M.D.; Garrett, Q. Role of carnitine in disease. Nutr. Metab. 2010, 7, 30. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Jacques, F.; Rippa, S.; Perrin, Y. Physiology of L-carnitine in plants in light of the knowledge in animals and microorganisms. Plant Sci. Int. J. Exp. Plant Biol. 2018, 274, 432–440. [Google Scholar] [CrossRef] [PubMed]
  119. Carnitine Fact Sheet for Health Professionals. Office of Dietary Supplements, National Institutes of Health, US Department of Health & Human Services, Ed. Available online: https://meatscience.org/docs/default-source/publications-resources/Hot-Topics/carnitine-health-professional-fact-sheet.pdf?sfvrsn=0 (accessed on 28 July 2021).
  120. Gatarek, P.; Kaluzna-Czaplinska, J. Trimethylamine N-oxide (TMAO) in human health. EXCLI J. 2021, 20, 19. [Google Scholar] [CrossRef]
  121. El-Hattab, A.W.; Scaglia, F. Disorders of carnitine biosynthesis and transport. Mol. Genet. Metab. 2015, 116, 107–112. [Google Scholar] [CrossRef]
  122. Pekala, J.; Patkowska-Sokola, B.; Bodkowski, R.; Jamroz, D.; Nowakowski, P.; Lochynski, S.; Librowski, T. L-carnitine--metabolic functions and meaning in humans life. Curr. Drug. Metab. 2011, 12, 667–678. [Google Scholar] [CrossRef] [PubMed]
  123. Cashman, J.R.; Xiong, Y.; Lin, J.; Verhagen, H.; van Poppel, G.; van Bladeren, P.J.; Larsen-Su, S.; Williams, D.E. In vitro and in vivo inhibition of human flavin-containing monooxygenase form 3 (FMO3) in the presence of dietary indoles. Biochem. Pharmacol. 1999, 58, 1047–1055. [Google Scholar] [CrossRef]
  124. Velasquez, M.T.; Ramezani, A.; Manal, A.; Raj, D.S. Trimethylamine N-Oxide: The Good, the Bad and the Unknown. Toxins 2016, 8, 326. [Google Scholar] [CrossRef] [Green Version]
  125. Qi, J.; You, T.; Li, J.; Pan, T.; Xiang, L.; Han, Y.; Zhu, L. Circulating trimethylamine N-oxide and the risk of cardiovascular diseases: A systematic review and meta-analysis of 11 prospective cohort studies. J. Cell. Mol. Med. 2018, 22, 185–194. [Google Scholar] [CrossRef] [PubMed]
  126. Geng, J.; Yang, C.; Wang, B.; Zhang, X.; Hu, T.; Gu, Y.; Li, J. Trimethylamine N-oxide promotes atherosclerosis via CD36-dependent MAPK/JNK pathway. Biomed. Pharmacother. Biomed. Pharmacother. 2018, 97, 941–947. [Google Scholar] [CrossRef]
  127. Suzuki, T.; Heaney, L.M.; Bhandari, S.S.; Jones, D.J.; Ng, L.L. Trimethylamine N-oxide and prognosis in acute heart failure. Heart 2016, 102, 841–848. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Troseid, M.; Ueland, T.; Hov, J.R.; Svardal, A.; Gregersen, I.; Dahl, C.P.; Aakhus, S.; Gude, E.; Bjorndal, B.; Halvorsen, B.; et al. Microbiota-dependent metabolite trimethylamine-N-oxide is associated with disease severity and survival of patients with chronic heart failure. J. Intern. Med. 2015, 277, 717–726. [Google Scholar] [CrossRef] [PubMed]
  129. Shafi, T.; Powe, N.R.; Meyer, T.W.; Hwang, S.; Hai, X.; Melamed, M.L.; Banerjee, T.; Coresh, J.; Hostetter, T.H. Trimethylamine N-Oxide and Cardiovascular Events in Hemodialysis Patients. J. Am. Soc. Nephrol. 2017, 28, 321–331. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  130. Daïen, C.I.; Pinget, G.V.; Tan, J.K.; Macia, L. Detrimental Impact of Microbiota-Accessible Carbohydrate-Deprived Diet on Gut and Immune Homeostasis: An Overview. Front. Immunol. 2017, 8, 548. [Google Scholar] [CrossRef] [Green Version]
  131. Singh, R.K.; Chang, H.-W.; Yan, D.; Lee, K.M.; Ucmak, D.; Wong, K.; Abrouk, M.; Farahnik, B.; Nakamura, M.; Zhu, T.H.; et al. Influence of diet on the gut microbiome and implications for human health. J. Transl. Med. 2017, 15, 73. [Google Scholar] [CrossRef] [Green Version]
  132. Micha, R.; Michas, G.; Mozaffarian, D. Unprocessed red and processed meats and risk of coronary artery disease and type 2 diabetes--an updated review of the evidence. Curr. Atheroscler. Rep. 2012, 14, 515–524. [Google Scholar] [CrossRef] [Green Version]
  133. Wang, Z.; Bergeron, N.; Levison, B.S.; Li, X.S.; Chiu, S.; Jia, X.; Koeth, R.A.; Li, L.; Wu, Y.; Tang, W.H.W.; et al. Impact of chronic dietary red meat, white meat, or non-meat protein on trimethylamine N-oxide metabolism and renal excretion in healthy men and women. Eur. Heart J. 2019, 40, 583–594. [Google Scholar] [CrossRef]
  134. Zhu, W.; Wang, Z.; Tang, W.H.W.; Hazen, S.L. Gut Microbe-Generated Trimethylamine N-Oxide From Dietary Choline Is Prothrombotic in Subjects. Circulation 2017, 135, 1671–1673. [Google Scholar] [CrossRef] [Green Version]
  135. Heianza, Y.; Ma, W.; DiDonato, J.A.; Sun, Q.; Rimm, E.B.; Hu, F.B.; Rexrode, K.M.; Manson, J.E.; Qi, L. Long-Term Changes in Gut Microbial Metabolite Trimethylamine N-Oxide and Coronary Heart Disease Risk. J. Am. Coll. Cardiol. 2020, 75, 763–772. [Google Scholar] [CrossRef]
  136. Wu, W.K.; Chen, C.C.; Liu, P.Y.; Panyod, S.; Liao, B.Y.; Chen, P.C.; Kao, H.L.; Kuo, H.C.; Kuo, C.H.; Chiu, T.H.T.; et al. Identification of TMAO-producer phenotype and host-diet-gut dysbiosis by carnitine challenge test in human and germ-free mice. Gut 2019, 68, 1439–1449. [Google Scholar] [CrossRef] [Green Version]
  137. Lyu, M.; Wang, Y.F.; Fan, G.W.; Wang, X.Y.; Xu, S.Y.; Zhu, Y. Balancing Herbal Medicine and Functional Food for Prevention and Treatment of Cardiometabolic Diseases through Modulating Gut Microbiota. Front. Microbiol. 2017, 8, 2146. [Google Scholar] [CrossRef] [Green Version]
  138. Chen, M.L.; Yi, L.; Zhang, Y.; Zhou, X.; Ran, L.; Yang, J.; Zhu, J.D.; Zhang, Q.Y.; Mi, M.T. Resveratrol Attenuates Trimethylamine-N-Oxide (TMAO)-Induced Atherosclerosis by Regulating TMAO Synthesis and Bile Acid Metabolism via Remodeling of the Gut Microbiota. mBio 2016, 7, e02210-15. [Google Scholar] [CrossRef] [Green Version]
  139. Annunziata, G.; Maisto, M.; Schisano, C.; Ciampaglia, R.; Narciso, V.; Tenore, G.C.; Novellino, E. Effects of Grape Pomace Polyphenolic Extract (Taurisolo®) in Reducing TMAO Serum Levels in Humans: Preliminary Results from a Randomized, Placebo-Controlled, Cross-Over Study. Nutrients 2019, 11, 139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  140. Sawicka, A.K.; Renzi, G.; Olek, R.A. The bright and the dark sides of L-carnitine supplementation: A systematic review. J. Int. Soc. Sports Nutr. 2020, 17, 49. [Google Scholar] [CrossRef]
  141. Smits, L.P.; Kootte, R.S.; Levin, E.; Prodan, A.; Fuentes, S.; Zoetendal, E.G.; Wang, Z.; Levison, B.S.; Cleophas, M.C.P.; Kemper, E.M.; et al. Effect of Vegan Fecal Microbiota Transplantation on Carnitine- and Choline-Derived Trimethylamine-N-Oxide Production and Vascular Inflammation in Patients With Metabolic Syndrome. J. Am. Heart Assoc. 2018, 7, e008342. [Google Scholar] [CrossRef] [PubMed]
  142. Pang, A.L.Y.; Martin, M.M.; Martin, A.L.; Chan, W.Y. Molecular basis of diseases of the endocrine system. In Essential Concepts in Molecular Pathology, 2nd ed.; William Coleman, G.T., Ed.; Academic Press: Cambridge, MA, USA, 2020; p. 28. [Google Scholar] [CrossRef]
  143. Werner, H.; Sarfstein, R.; LeRoith, D.; Bruchim, I. Insulin-like Growth Factor 1 Signaling Axis Meets p53 Genome Protection Pathways. Front. Oncol. 2016, 6, 159. [Google Scholar] [CrossRef]
  144. Ziegler, A.N.; Levison, S.W.; Wood, T.L. Insulin and IGF receptor signalling in neural-stem-cell homeostasis. Nat. Rev. Endocrinol. 2015, 11, 161–170. [Google Scholar] [CrossRef] [Green Version]
  145. Bartke, A.; Chandrashekar, V.; Dominici, F.; Turyn, D.; Kinney, B.; Steger, R.; Kopchick, J.J. Insulin-like growth factor 1 (IGF-1) and aging: Controversies and new insights. Biogerontology 2003, 4, 1–8. [Google Scholar] [CrossRef] [PubMed]
  146. Brabant, G.; Wallaschofski, H. Normal levels of serum IGF-I: Determinants and validity of current reference ranges. Pituitary 2007, 10, 129–133. [Google Scholar] [CrossRef]
  147. Estivariz, C.F.; Ziegler, T.R. Nutrition and the insulin-like growth factor system. Endocrine 1997, 7, 65–71. [Google Scholar] [CrossRef]
  148. Switkowski, K.M.; Jacques, P.F.; Must, A.; Fleisch, A.; Oken, E. Associations of protein intake in early childhood with body composition, height, and insulin-like growth factor I in mid-childhood and early adolescence. Am. J. Clin. Nutr. 2019, 109, 1154–1163. [Google Scholar] [CrossRef]
  149. Xu, S.; Xue, Y. Protein intake and obesity in young adolescents. Exp. Ther. Med. 2016, 11, 1545–1549. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  150. Sonntag, W.E.; Carter, C.S.; Ikeno, Y.; Ekenstedt, K.; Carlson, C.S.; Loeser, R.F.; Chakrabarty, S.; Lee, S.; Bennett, C.; Ingram, R.; et al. Adult-onset growth hormone and insulin-like growth factor I deficiency reduces neoplastic disease, modifies age-related pathology, and increases life span. Endocrinology 2005, 146, 2920–2932. [Google Scholar] [CrossRef] [Green Version]
  151. Miura, Y.; Kato, H.; Noguchi, T. Effect of dietary proteins on insulin-like growth factor-1 (IGF-1) messenger ribonucleic acid content in rat liver. Br. J. Nutr. 1992, 67, 257–265. [Google Scholar] [CrossRef] [Green Version]
  152. Thissen, J.P.; Ketelslegers, J.M.; Underwood, L.E. Nutritional regulation of the insulin-like growth factors. Endocr. Rev. 1994, 15, 80–101. [Google Scholar] [CrossRef]
  153. Semba, R.D.; Shardell, M.; Sakr Ashour, F.A.; Moaddel, R.; Trehan, I.; Maleta, K.M.; Ordiz, M.I.; Kraemer, K.; Khadeer, M.A.; Ferrucci, L.; et al. Child Stunting is Associated with Low Circulating Essential Amino Acids. EBioMedicine 2016, 6, 246–252. [Google Scholar] [CrossRef] [Green Version]
  154. Alexy, U.; Fischer, M.; Weder, S.; Längler, A.; Michalsen, A.; Sputtek, A.; Keller, M. Nutrient Intake and Status of German Children and Adolescents Consuming Vegetarian, Vegan or Omnivore Diets: Results of the VeChi Youth Study. Nutrients 2021, 13, 1707. [Google Scholar] [CrossRef]
  155. Kopp, W. How Western Diet And Lifestyle Drive The Pandemic Of Obesity And Civilization Diseases. Diabetes Metab. Syndr. Obes. Targets Ther. 2019, 12, 2221–2236. [Google Scholar] [CrossRef] [Green Version]
  156. Singhal, A.; Lucas, A. Early origins of cardiovascular disease: Is there a unifying hypothesis? Lancet 2004, 363, 1642–1645. [Google Scholar] [CrossRef]
  157. Hoffman, D.J.; Reynolds, R.M.; Hardy, D.B. Developmental origins of health and disease: Current knowledge and potential mechanisms. Nutr. Rev. 2017, 75, 951–970. [Google Scholar] [CrossRef] [Green Version]
  158. Ong, Z.Y.; Gugusheff, J.R.; Muhlhausler, B.S. Perinatal overnutrition and the programming of food preferences: Pathways and mechanisms. J. Dev. Orig. Health Dis. 2012, 3, 299–308. [Google Scholar] [CrossRef] [Green Version]
  159. Fukuoka, H. DOHaD (Developmental Origins of Health and Disease) and Birth Cohort Research. J. Nutr. Sci. Vitaminol. 2015, 61, S2–S4. [Google Scholar] [CrossRef] [Green Version]
  160. English, S.; Uller, T. Does early-life diet affect longevity? A meta-analysis across experimental studies. Biol. Lett. 2016, 12, 20160291. [Google Scholar] [CrossRef] [PubMed]
  161. Lin, L.; Yan, L.; Liu, Y.; Yuan, F.; Li, H.; Ni, J. Incidence and death in 29 cancer groups in 2017 and trend analysis from 1990 to 2017 from the Global Burden of Disease Study. J. Hematol. Oncol. 2019, 12, 96. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  163. Global Burden of Disease Cancer Collaboration; Fitzmaurice, C.; Abate, D.; Abbasi, N.; Abbastabar, H.; Abd-Allah, F.; Abdel-Rahman, O.; Abdelalim, A.; Abdoli, A.; Abdollahpour, I.; et al. Global, Regional, and National Cancer Incidence, Mortality, Years of Life Lost, Years Lived With Disability, and Disability-Adjusted Life-Years for 29 Cancer Groups, 1990 to 2017: A Systematic Analysis for the Global Burden of Disease Study. JAMA Oncol. 2019, 5, 1749–1768. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Tracz, A.F.; Szczylik, C.; Porta, C.; Czarnecka, A.M. Insulin-like growth factor-1 signaling in renal cell carcinoma. BMC Cancer 2016, 16, 453. [Google Scholar] [CrossRef] [Green Version]
  165. Werner, H. Tumor suppressors govern insulin-like growth factor signaling pathways: Implications in metabolism and cancer. Oncogene 2012, 31, 2703–2714. [Google Scholar] [CrossRef]
  166. Weroha, S.J.; Haluska, P. The insulin-like growth factor system in cancer. Endocrinol. Metab. Clin. N. Am. 2012, 41, 335–350. [Google Scholar] [CrossRef] [Green Version]
  167. Grimberg, A. Mechanisms by which IGF-I may promote cancer. Cancer Biol. Ther. 2003, 2, 630–635. [Google Scholar] [CrossRef] [Green Version]
  168. Rinaldi, S.; Cleveland, R.; Norat, T.; Biessy, C.; Rohrmann, S.; Linseisen, J.; Boeing, H.; Pischon, T.; Panico, S.; Agnoli, C.; et al. Serum levels of IGF-I, IGFBP-3 and colorectal cancer risk: Results from the EPIC cohort, plus a meta-analysis of prospective studies. Int. J. Cancer 2010, 126, 1702–1715. [Google Scholar] [CrossRef]
  169. Roddam, A.W.; Allen, N.E.; Appleby, P.; Key, T.J.; Ferrucci, L.; Carter, H.B.; Metter, E.J.; Chen, C.; Weiss, N.S.; Fitzpatrick, A.; et al. Insulin-like growth factors, their binding proteins, and prostate cancer risk: Analysis of individual patient data from 12 prospective studies. Ann. Intern. Med. 2008, 149, 461–471. [Google Scholar] [CrossRef] [Green Version]
  170. Key, T.J.; Appleby, P.N.; Reeves, G.K.; Roddam, A.W.; Breast, T.E.H. Insulin-like growth factor 1 (IGF1), IGF binding protein 3 (IGFBP3), and breast cancer risk: Pooled individual data analysis of 17 prospective studies. Lancet Oncol. 2010, 11, 530–542. [Google Scholar] [CrossRef] [Green Version]
  171. Gunnell, D.; Oliver, S.E.; Peters, T.J.; Donovan, J.L.; Persad, R.; Maynard, M.; Gillatt, D.; Pearce, A.; Hamdy, F.C.; Neal, D.E.; et al. Are diet-prostate cancer associations mediated by the IGF axis? A cross-sectional analysis of diet, IGF-I and IGFBP-3 in healthy middle-aged men. Br. J. Cancer 2003, 88, 1682–1686. [Google Scholar] [CrossRef] [Green Version]
  172. Epstein, S.S. Re: Role of the insulin-like growth factors in cancer development and progression. J. Natl. Cancer Inst. 2001, 93, 238. [Google Scholar] [CrossRef] [Green Version]
  173. Key, T.J. Diet, insulin-like growth factor-1 and cancer risk. Proc. Nutr. Soc. 2011, 70, 385–388. [Google Scholar] [CrossRef] [Green Version]
  174. Melnik, B.C.; John, S.M.; Schmitz, G. Over-stimulation of insulin/IGF-1 signaling by western diet may promote diseases of civilization: Lessons learnt from laron syndrome. Nutr. Metab. 2011, 8, 41. [Google Scholar] [CrossRef] [Green Version]
  175. Dawson-Hughes, B.; Harris, S.S.; Rasmussen, H.; Song, L.; Dallal, G.E. Effect of dietary protein supplements on calcium excretion in healthy older men and women. J. Clin. Endocrinol. Metab. 2004, 89, 1169–1173. [Google Scholar] [CrossRef] [Green Version]
  176. Chan, J.M.; Stampfer, M.J.; Giovannucci, E.; Gann, P.H.; Ma, J.; Wilkinson, P.; Hennekens, C.H.; Pollak, M. Plasma insulin-like growth factor-I and prostate cancer risk: A prospective study. Science 1998, 279, 563–566. [Google Scholar] [CrossRef]
  177. Qin, L.Q.; Xu, J.Y.; Wang, P.Y.; Kaneko, T.; Hoshi, K.; Sato, A. Milk consumption is a risk factor for prostate cancer: Meta-analysis of case-control studies. Nutr. Cancer 2004, 48, 22–27. [Google Scholar] [CrossRef] [PubMed]
  178. Torfadottir, J.E.; Steingrimsdottir, L.; Mucci, L.; Aspelund, T.; Kasperzyk, J.L.; Olafsson, O.; Fall, K.; Tryggvadottir, L.; Harris, T.B.; Launer, L.; et al. Milk Intake in Early Life and Risk of Advanced Prostate Cancer. Am. J. Epidemiol. 2012, 175, 144–153. [Google Scholar] [CrossRef] [Green Version]
  179. Levine, M.E.; Suarez, J.A.; Brandhorst, S.; Balasubramanian, P.; Cheng, C.W.; Madia, F.; Fontana, L.; Mirisola, M.G.; Guevara-Aguirre, J.; Wan, J.; et al. Low protein intake is associated with a major reduction in IGF-1, cancer, and overall mortality in the 65 and younger but not older population. Cell Metab. 2014, 19, 407–417. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  180. Key, T.J.; Appleby, P.N.; Spencer, E.A.; Travis, R.C.; Roddam, A.W.; Allen, N.E. Cancer incidence in vegetarians: Results from the European Prospective Investigation into Cancer and Nutrition (EPIC-Oxford). Am. J. Clin. Nutr. 2009, 89, S1620–S1626. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  181. Allen, N.E.; Appleby, P.N.; Davey, G.K.; Kaaks, R.; Rinaldi, S.; Key, T.J. The associations of diet with serum insulin-like growth factor I and its main binding proteins in 292 women meat-eaters, vegetarians, and vegans. Cancer Epidemiol. Biomark. 2002, 11, 1441–1448. [Google Scholar]
  182. Saxe, G.A.; Major, J.M.; Nguyen, J.Y.; Freeman, K.M.; Downs, T.M.; Salem, C.E. Potential attenuation of disease progression in recurrent prostate cancer with plant-based diet and stress reduction. Integr. Cancer Ther. 2006, 5, 206–213. [Google Scholar] [CrossRef] [PubMed]
  183. Harvie, M.; Howell, A.; Evans, D.G. Can diet and lifestyle prevent breast cancer: What is the evidence? Am. Soc. Clin. Oncol. Educ. Book 2015, 35, e66–e73. [Google Scholar] [CrossRef] [PubMed]
  184. Berkow, S.E.; Barnard, N.D.; Saxe, G.A.; Ankerberg-Nobis, T. Diet and survival after prostate cancer diagnosis. Nutr. Rev. 2007, 65, 391–403. [Google Scholar] [CrossRef] [PubMed]
  185. Jasek, K.; Kubatka, P.; Samec, M.; Liskova, A.; Smejkal, K.; Vybohova, D.; Bugos, O.; Biskupska-Bodova, K.; Bielik, T.; Zubor, P.; et al. DNA Methylation Status in Cancer Disease: Modulations by Plant-Derived Natural Compounds and Dietary Interventions. Biomolecules 2019, 9, 289. [Google Scholar] [CrossRef] [Green Version]
  186. Qian, F.; Huo, D. Circulating Insulin-Like Growth Factor-1 and Risk of Total and 19 Site-Specific Cancers: Cohort Study Analyses from the UK Biobank. Cancer Epidemiol. Prev. Biomark. 2020, 29, 2332–2342. [Google Scholar] [CrossRef]
  187. Liu, P.; Cheng, H.; Roberts, T.M.; Zhao, J.J. Targeting the phosphoinositide 3-kinase pathway in cancer. Nat. Rev. Drug Discov. 2009, 8, 627–644. [Google Scholar] [CrossRef] [Green Version]
  188. Crino, P.B. The mTOR signalling cascade: Paving new roads to cure neurological disease. Nat. Rev. Neurol. 2016, 12, 379–392. [Google Scholar] [CrossRef]
  189. Compound Summary for CID 5284616. National Center for Biotechnology Information: PubChem Compound Database. 2021. Available online: https://pubchem.ncbi.nlm.nih.gov/compound/Sirolimus (accessed on 27 July 2021).
  190. Ehninger, D.; Neff, F.; Xie, K. Longevity, aging and rapamycin. Cell. Mol. Life Sci. 2014, 71, 4325–4346. [Google Scholar] [CrossRef] [Green Version]
  191. Lamming, D.W. Inhibition of the Mechanistic Target of Rapamycin (mTOR)-Rapamycin and Beyond. Cold Spring Harb. Perspect. Med. 2016, 6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  192. Lee, S.H.; Min, K.J. Caloric restriction and its mimetics. BMB Rep. 2013, 46, 181–187. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Kim, J.; Guan, K.L. mTOR as a central hub of nutrient signalling and cell growth. Nat. Cell Biol. 2019, 21, 63–71. [Google Scholar] [CrossRef]
  194. Melnik, B.C. Leucine signaling in the pathogenesis of type 2 diabetes and obesity. World J. Diabetes 2012, 3, 38–53. [Google Scholar] [CrossRef] [PubMed]
  195. Ma, X.; Han, M.; Li, D.; Hu, S.; Gilbreath, K.R.; Bazer, F.W.; Wu, G. L-Arginine promotes protein synthesis and cell growth in brown adipocyte precursor cells via the mTOR signal pathway. Amino Acids 2017, 49, 957–964. [Google Scholar] [CrossRef] [PubMed]
  196. Mossmann, D.; Park, S.; Hall, M.N. mTOR signalling and cellular metabolism are mutual determinants in cancer. Nat. Rev. Cancer 2018, 18, 744–757. [Google Scholar] [CrossRef]
  197. Wang, R.; Jiao, H.; Zhao, J.; Wang, X.; Lin, H. L-Arginine Enhances Protein Synthesis by Phosphorylating mTOR (Thr 2446) in a Nitric Oxide-Dependent Manner in C2C12 Cells. Oxid. Med. Cell Longev. 2018, 2018, 7569127. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  198. Polak, P.; Cybulski, N.; Feige, J.N.; Auwerx, J.; Ruegg, M.A.; Hall, M.N. Adipose-specific knockout of raptor results in lean mice with enhanced mitochondrial respiration. Cell Metab. 2008, 8, 399–410. [Google Scholar] [CrossRef]
  199. Zoncu, R.; Efeyan, A.; Sabatini, D.M. mTOR: From growth signal integration to cancer, diabetes and ageing. Nat. Rev. Mol. Cell Biol. 2011, 12, 21–35. [Google Scholar] [CrossRef] [Green Version]
  200. Facchinetti, V.; Ouyang, W.; Wei, H.; Soto, N.; Lazorchak, A.; Gould, C.; Lowry, C.; Newton, A.C.; Mao, Y.; Miao, R.Q.; et al. The mammalian target of rapamycin complex 2 controls folding and stability of Akt and protein kinase C. EMBO J. 2008, 27, 1932–1943. [Google Scholar] [CrossRef] [PubMed]
  201. Ikenoue, T.; Inoki, K.; Yang, Q.; Zhou, X.; Guan, K.L. Essential function of TORC2 in PKC and Akt turn motif phosphorylation, maturation and signalling. EMBO J. 2008, 27, 1919–1931. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  202. Liu, G.Y.; Sabatini, D.M. mTOR at the nexus of nutrition, growth, ageing and disease. Nat. Rev. Mol. Cell. Biol. 2020, 21, 183–203. [Google Scholar] [CrossRef] [PubMed]
  203. Xu, F.; Na, L.; Li, Y.; Chen, L. Roles of the PI3K/AKT/mTOR signalling pathways in neurodegenerative diseases and tumours. Cell Biosci. 2020, 10, 54. [Google Scholar] [CrossRef] [Green Version]
  204. Ghosh, J.C.; Siegelin, M.D.; Vaira, V.; Faversani, A.; Tavecchio, M.; Chae, Y.C.; Lisanti, S.; Rampini, P.; Giroda, M.; Caino, M.C.; et al. Adaptive mitochondrial reprogramming and resistance to PI3K therapy. J. Natl. Cancer Inst. 2015, 107. [Google Scholar] [CrossRef] [Green Version]
  205. Fontana, L.; Adelaiye, R.M.; Rastelli, A.L.; Miles, K.M.; Ciamporcero, E.; Longo, V.D.; Nguyen, H.; Vessella, R.; Pili, R. Dietary protein restriction inhibits tumor growth in human xenograft models. Oncotarget 2013, 4, 2451–2461. [Google Scholar] [CrossRef]
  206. Xu, Z.; Han, X.; Ou, D.; Liu, T.; Li, Z.; Jiang, G.; Liu, J.; Zhang, J. Targeting PI3K/AKT/mTOR-mediated autophagy for tumor therapy. Appl. Microbiol. Biotechnol. 2020, 104, 575–587. [Google Scholar] [CrossRef]
  207. Nwadike, C.; Williamson, L.E.; Gallagher, L.E.; Guan, J.L.; Chan, E.Y.W. AMPK Inhibits ULK1-Dependent Autophagosome Formation and Lysosomal Acidification via Distinct Mechanisms. Mol. Cell Biol. 2018, 38, e00023-18. [Google Scholar] [CrossRef] [Green Version]
  208. Yin, J.; Ren, W.; Huang, X.; Li, T.; Yin, Y. Protein restriction and cancer. Biochim. Biophys. Acta Rev. Cancer 2018, 1869, 256–262. [Google Scholar] [CrossRef]
  209. Weng, M.L.; Chen, W.K.; Chen, X.Y.; Lu, H.; Sun, Z.R.; Yu, Q.; Sun, P.F.; Xu, Y.J.; Zhu, M.M.; Jiang, N.; et al. Fasting inhibits aerobic glycolysis and proliferation in colorectal cancer via the Fdft1-mediated AKT/mTOR/HIF1alpha pathway suppression. Nat. Commun. 2020, 11, 1869. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  210. De Cabo, R.; Longo, D.L.; Mattson, M.P. Effects of Intermittent Fasting on Health, Aging, and Disease. N. Engl. J. Med. 2019, 381, 2541–2551. [Google Scholar] [CrossRef] [PubMed]
  211. Mccay, C.M.; Crowell, M.F.; Maynard, L.A. Nutrition Metabolism Classic—The Effect of Retarded Growth Upon the Length of Life-Span and Upon the Ultimate Body Size. Nutrition 1989, 5, 155–171. [Google Scholar] [PubMed]
  212. Heilbronn, L.K.; Ravussin, E. Calorie restriction and aging: Review of the literature and implications for studies in humans. Am. J. Clin. Nutr. 2003, 78, 361–369. [Google Scholar] [CrossRef] [PubMed]
  213. Wolf, G. Calorie restriction increases life span: A molecular mechanism. Nutr. Rev. 2006, 64, 89–92. [Google Scholar] [CrossRef]
  214. Nakagawa, S.; Lagisz, M.; Hector, K.L.; Spencer, H.G. Comparative and meta-analytic insights into life extension via dietary restriction. Aging Cell 2012, 11, 401–409. [Google Scholar] [CrossRef]
  215. Fontana, L.; Partridge, L.; Longo, V.D. Extending Healthy Life Span-From Yeast to Humans. Science 2010, 328, 321–326. [Google Scholar] [CrossRef] [Green Version]
  216. Norman, K.; Klaus, S. Veganism, aging and longevity: New insight into old concepts. Curr. Opin. Clin. Nutr. Metab. Care 2020, 23, 145–150. [Google Scholar] [CrossRef]
  217. Yan, L.J.; Lamb, R.F. Amino acid sensing and regulation of mTORC1. Semin. Cell Dev. Biol. 2012, 23, 621–625. [Google Scholar] [CrossRef]
  218. Wang, X.; Proud, C.G. Nutrient control of TORC1, a cell-cycle regulator. Trends Cell Biol. 2009, 19, 260–267. [Google Scholar] [CrossRef]
  219. McCarty, M.F. mTORC1 activity as a determinant of cancer risk--rationalizing the cancer-preventive effects of adiponectin, metformin, rapamycin, and low-protein vegan diets. Med. Hypotheses 2011, 77, 642–648. [Google Scholar] [CrossRef]
  220. Tong, X.; Pelling, J.C. Targeting the PI3K/Akt/mTOR axis by apigenin for cancer prevention. Anti-Cancer Agents Med. Chem. 2013, 13, 971–978. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  221. Beevers, C.S.; Chen, L.; Liu, L.; Luo, Y.; Webster, N.J.; Huang, S. Curcumin disrupts the Mammalian target of rapamycin-raptor complex. Cancer Res. 2009, 69, 1000–1008. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  222. Van Aller, G.S.; Carson, J.D.; Tang, W.; Peng, H.; Zhao, L.; Copeland, R.A.; Tummino, P.J.; Luo, L. Epigallocatechin gallate (EGCG), a major component of green tea, is a dual phosphoinositide-3-kinase/mTOR inhibitor. Biochem. Biophys. Res. Commun. 2011, 406, 194–199. [Google Scholar] [CrossRef] [PubMed]
  223. Lin, J.N.; Lin, V.C.; Rau, K.M.; Shieh, P.C.; Kuo, D.H.; Shieh, J.C.; Chen, W.J.; Tsai, S.C.; Way, T.D. Resveratrol modulates tumor cell proliferation and protein translation via SIRT1-dependent AMPK activation. J. Agric. Food Chem. 2010, 58, 1584–1592. [Google Scholar] [CrossRef]
  224. Adhami, V.M.; Syed, D.N.; Khan, N.; Mukhtar, H. Dietary flavonoid fisetin: A novel dual inhibitor of PI3K/Akt and mTOR for prostate cancer management. Biochem. Pharmacol. 2012, 84, 1277–1281. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  225. Hosseinzade, A.; Sadeghi, O.; Naghdipour Biregani, A.; Soukhtehzari, S.; Brandt, G.S.; Esmaillzadeh, A. Immunomodulatory Effects of Flavonoids: Possible Induction of T CD4+ Regulatory Cells Through Suppression of mTOR Pathway Signaling Activity. Front. Immunol. 2019, 10, 51. [Google Scholar] [CrossRef] [Green Version]
  226. Raimundo, A.F.; Félix, F.; Andrade, R.; García-Conesa, M.-T.; González-Sarrías, A.; Gilsa-Lopes, J.; do Ó, D.; Raimundo, A.; Ribeiro, R.; Rodriguez-Mateos, A.; et al. Combined effect of interventions with pure or enriched mixtures of (poly)phenols and anti-diabetic medication in type 2 diabetes management: A meta-analysis of randomized controlled human trials. Eur. J. Nutr. 2020, 59, 1329–1343. [Google Scholar] [CrossRef]
  227. Mopuri, R.; Islam, M.S. Medicinal plants and phytochemicals with anti-obesogenic potentials: A review. Biomed. Pharmacother. 2017, 89, 1442–1452. [Google Scholar] [CrossRef]
  228. Melnik, B.C. Western Diet-Mediated mTORC1-Signaling in Acne, Psoriasis, Atopic Dermatitis, and Related Diseases of Civilization: Therapeutic Role of Plant-Derived Natural mTORC1 Inhibitors. In Bioactive Dietary Factors and Plant Extracts in Dermatology; Watson, R.S.Z., Ed.; Humana Press: Totowa, NJ, USA, 2013. [Google Scholar] [CrossRef]
  229. Lundblad, A. Gunnar Blix and his discovery of sialic acids. Fascinating molecules in glycobiology. Upsala J. Med. Sci. 2015, 120, 104–112. [Google Scholar] [CrossRef]
  230. Gottschalk, A. Structural Relationship between Sialic Acid, Neuraminic Acid and 2-Carboxy-Pyrrole. Nature 1955, 176, 2. [Google Scholar] [CrossRef]
  231. Takahashi, T.; Takano, M.; Kurebayashi, Y.; Masuda, M.; Kawagishi, S.; Takaguchi, M.; Yamanaka, T.; Minami, A.; Otsubo, T.; Ikeda, K.; et al. N-Glycolylneuraminic Acid on Human Epithelial Cells Prevents Entry of Influenza A Viruses That Possess N-Glycolylneuraminic Acid Binding Ability. J. Virol. 2014, 88, 8445–8456. [Google Scholar] [CrossRef] [Green Version]
  232. Okerblom, J.; Varki, A. Biochemical, Cellular, Physiological, and Pathological Consequences of Human Loss of N-Glycolylneuraminic Acid. ChemBioChem 2017, 18, 1155–1171. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  233. Varki, A. Colloquium paper: Uniquely human evolution of sialic acid genetics and biology. Proc. Natl. Acad. Sci. USA 2010, 107 (Suppl. 2), 8939–8946. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  234. Bardor, M.; Nguyen, D.H.; Diaz, S.; Varki, A. Mechanism of uptake and incorporation of the non-human sialic acid N-glycolylneuraminic acid into human cells. J. Biol. Chem. 2005, 280, 4228–4237. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  235. Bergfeld, A.K.; Varki, A. Cytidine Monophospho-NAcetylneuraminic Acid Hydroxylase (CMAH). In Handbook of Glycosyl-transferases and Related Genes, 2nd ed.; Taniguchi, N., Honke, K., Fukuda, M., Narimatsu, H., Yamaguchi, Y., Angata, T., Eds.; Springer: Berlin/Heidelberg, Germany, 2014; pp. 1559–1576. [Google Scholar] [CrossRef]
  236. Samraj, A.N.; Pearce, O.M.; Laubli, H.; Crittenden, A.N.; Bergfeld, A.K.; Banda, K.; Gregg, C.J.; Bingman, A.E.; Secrest, P.; Diaz, S.L.; et al. A red meat-derived glycan promotes inflammation and cancer progression. Proc. Natl. Acad. Sci. USA 2015, 112, 542–547. [Google Scholar] [CrossRef] [Green Version]
  237. Hedlund, M.; Padler-Karavani, V.; Varki, N.M.; Varki, A. Evidence for a human-specific mechanism for diet and antibody-mediated inflammation in carcinoma progression. Proc. Natl. Acad. Sci. USA 2008, 105, 18936–18941. [Google Scholar] [CrossRef] [Green Version]
  238. Pham, T.; Gregg, C.J.; Karp, F.; Chow, R.; Padler-Karavani, V.; Cao, H.; Chen, X.; Witztum, J.L.; Varki, N.M.; Varki, A. Evidence for a novel human-specific xeno-auto-antibody response against vascular endothelium. Blood 2009, 114, 5225–5235. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  239. Pruimboom, L. SARS-CoV 2; Possible alternative virus receptors and pathophysiological determinants. Med. Hypotheses 2021, 146, 110368. [Google Scholar] [CrossRef]
  240. Soulillou, J.P.; Cozzi, E.; Bach, J.M. Challenging the Role of Diet-Induced Anti-Neu5Gc Antibodies in Human Pathologies. Front. Immunol. 2020, 11, 834. [Google Scholar] [CrossRef]
  241. Raetz, C.R.H.; Whitfield, C. Lipopolysaccharide Endotoxins. Annu. Rev. Biochem. 2002, 71, 635–700. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  242. Kell, D.B.; Pretorius, E. On the translocation of bacteria and their lipopolysaccharides between blood and peripheral locations in chronic, inflammatory diseases: The central roles of LPS and LPS-induced cell death. Integr. Biol. 2015, 7, 1339–1377. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  243. Stoll, L.L.; Denning, G.M.; Weintraub, N.L. Potential role of endotoxin as a proinflammatory mediator of atherosclerosis. Arterioscler. Thromb. Vasc. Biol. 2004, 24, 2227–2236. [Google Scholar] [CrossRef]
  244. López-Moreno, J.; García-Carpintero, S.; Jimenez-Lucena, R.; Haro, C.; Rangel-Zúñiga, O.A.; Blanco-Rojo, R.; Yubero-Serrano, E.M.; Tinahones, F.J.; Delgado-Lista, J.; Pérez-Martínez, P.; et al. Effect of Dietary Lipids on Endotoxemia Influences Postprandial Inflammatory Response. J. Agric. Food Chem. 2017, 65, 7756–7763. [Google Scholar] [CrossRef]
  245. Fuke, N.; Nagata, N.; Suganuma, H.; Ota, T. Regulation of Gut Microbiota and Metabolic Endotoxemia with Dietary Factors. Nutrients 2019, 11, 2277. [Google Scholar] [CrossRef] [Green Version]
  246. Erridge, C. The capacity of foodstuffs to induce innate immune activation of human monocytes in vitro is dependent on food content of stimulants of Toll-like receptors 2 and 4. Br. J. Nutr. 2011, 105, 15–23. [Google Scholar] [CrossRef] [Green Version]
  247. Saad, M.J.A.; Santos, A.; Prada, P.O. Linking Gut Microbiota and Inflammation to Obesity and Insulin Resistance. Physiology 2016, 31, 283–293. [Google Scholar] [CrossRef]
  248. David, L.A.; Maurice, C.F.; Carmody, R.N.; Gootenberg, D.B.; Button, J.E.; Wolfe, B.E.; Ling, A.V.; Devlin, A.S.; Varma, Y.; Fischbach, M.A.; et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature 2013, 505, 559–563. [Google Scholar] [CrossRef] [Green Version]
  249. Medawar, E.; Huhn, S.; Villringer, A.; Veronica Witte, A. The effects of plant-based diets on the body and the brain: A systematic review. Transl. Psychiatry 2019, 9, 226. [Google Scholar] [CrossRef]
  250. Ahola, A.J.; Lassenius, M.I.; Forsblom, C.; Harjutsalo, V.; Lehto, M.; Groop, P.-H. Dietary patterns reflecting healthy food choices are associated with lower serum LPS activity. Sci. Rep. 2017, 7, 6511. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  251. Hwang, D.H.; Rutledge, J.C.; Burnett, D.J.; Huang, S.; Mo, Z. Endotoxin May Not Be the Major Cause of Postprandial Inflammation in Adults Who Consume a Single High-Fat or Moderately High-Fat Meal. J. Nutr. 2020, 150, 1303–1312. [Google Scholar] [CrossRef]
  252. Erridge, C.; Attina, T.; Spickett, C.M.; Webb, D.J. A high-fat meal induces low-grade endotoxemia: Evidence of a novel mechanism of postprandial inflammation. Am. J. Clin. Nutr. 2007, 86, 1286–1292. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Influence of natural-source antioxidants on health.
Figure 1. Influence of natural-source antioxidants on health.
Nutrients 13 02593 g001
Figure 2. The potential role and consequence of plant-based diets in preventing lipotoxicity. IMCL: intramyocellular lipid.
Figure 2. The potential role and consequence of plant-based diets in preventing lipotoxicity. IMCL: intramyocellular lipid.
Nutrients 13 02593 g002
Figure 3. The potential role of animal food-derived Neu5Gc in the risk of certain diseases. Neu5Gc: N-glycolylneuraminic acid.
Figure 3. The potential role of animal food-derived Neu5Gc in the risk of certain diseases. Neu5Gc: N-glycolylneuraminic acid.
Nutrients 13 02593 g003
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Szabo, Z.; Koczka, V.; Marosvolgyi, T.; Szabo, E.; Frank, E.; Polyak, E.; Fekete, K.; Erdelyi, A.; Verzar, Z.; Figler, M. Possible Biochemical Processes Underlying the Positive Health Effects of Plant-Based Diets—A Narrative Review. Nutrients 2021, 13, 2593. https://doi.org/10.3390/nu13082593

AMA Style

Szabo Z, Koczka V, Marosvolgyi T, Szabo E, Frank E, Polyak E, Fekete K, Erdelyi A, Verzar Z, Figler M. Possible Biochemical Processes Underlying the Positive Health Effects of Plant-Based Diets—A Narrative Review. Nutrients. 2021; 13(8):2593. https://doi.org/10.3390/nu13082593

Chicago/Turabian Style

Szabo, Zoltan, Viktor Koczka, Tamas Marosvolgyi, Eva Szabo, Eszter Frank, Eva Polyak, Kata Fekete, Attila Erdelyi, Zsofia Verzar, and Maria Figler. 2021. "Possible Biochemical Processes Underlying the Positive Health Effects of Plant-Based Diets—A Narrative Review" Nutrients 13, no. 8: 2593. https://doi.org/10.3390/nu13082593

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