Next Article in Journal
The Ultra-Processed Food Content of School Meals and Packed Lunches in the United Kingdom
Previous Article in Journal
Ischemic Stroke and Dietary Vitamin B12 Deficiency in Old-Aged Females: Impaired Motor Function, Increased Ischemic Damage Size, and Changed Metabolite Profiles in Brain and Cecum Tissue
Previous Article in Special Issue
Modulation of Inflammation by Plant-Derived Nutraceuticals in Tendinitis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Hesperidin Functions as an Ergogenic Aid by Increasing Endothelial Function and Decreasing Exercise-Induced Oxidative Stress and Inflammation, Thereby Contributing to Improved Exercise Performance

by
Maria Imperatrice
1,*,†,
Iris Cuijpers
2,*,†,
Freddy J. Troost
2 and
Mireille M. J. P. E. Sthijns
2
1
BioActor BV, Gaetano Martinolaan 50, 6229 GS Maastricht, The Netherlands
2
Food Innovation and Health, Department of Human Biology, Maastricht University, 6200 MD Maastricht, The Netherlands
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Nutrients 2022, 14(14), 2955; https://doi.org/10.3390/nu14142955
Submission received: 20 June 2022 / Revised: 12 July 2022 / Accepted: 15 July 2022 / Published: 19 July 2022
(This article belongs to the Special Issue Polyphenols for Health and Sport)

Abstract

:
The regulation of blood flow to peripheral muscles is crucial for proper skeletal muscle functioning and exercise performance. During exercise, increased mitochondrial oxidative phosphorylation leads to increased electron leakage and consequently induces an increase in ROS formation, contributing to DNA, lipid, and protein damage. Moreover, exercise may increase blood- and intramuscular inflammatory factors leading to a deterioration in endurance performance. The aim of this review is to investigate the potential mechanisms through which the polyphenol hesperidin could lead to enhanced exercise performance, namely improved endothelial function, reduced exercise-induced oxidative stress, and inflammation. We selected in vivo RCTs, animal studies, and in vitro studies in which hesperidin, its aglycone form hesperetin, hesperetin-metabolites, or orange juice are supplemented at any dosage and where the parameters related to endothelial function, oxidative stress, and/or inflammation have been measured. The results collected in this review show that hesperidin improves endothelial function (via increased NO availability), inhibits ROS production, decreases production and plasma levels of pro-inflammatory markers, and improves anaerobic exercise outcomes (e.g., power, speed, energy). For elite and recreational athletes, hesperidin could be used as an ergogenic aid to enhance muscle recovery between training sessions, optimize oxygen and nutrient supplies to the muscles, and improve anaerobic performance.

1. Introduction

During exercise, skeletal muscle cells convert biological fuel (e.g., lipids, carbohydrates) into mechanical force to allow muscle contraction and therefore movement. The energy required for this motion is largely provided by the breakdown of adenosine triphosphate (ATP). Intramuscular stores of ATP can sustain only a short period of muscle activity. Therefore, ATP needs to be generated by anaerobic glycolysis and oxidative phosphorylation [1]. During exercise bouts lasting several minutes to hours, mitochondrial oxidative phosphorylation is responsible for almost all the ATP generated for the contracting skeletal muscles. This process is critically dependent on the respiratory and cardiovascular systems to ensure an adequate oxygen supply [2].
Blood flow is the main regulator of the skeletal muscles’ oxygen supply. Skeletal muscle contains a dense capillary network that serves to deliver oxygen and nutrients and remove waste products from the skeletal muscle cells [3]. To ensure adequate muscle oxygenation, blood flow increases during exercise capacity (i.e., capillaries’ numbers and diameters) [4,5].
The endothelium plays a major role in the regulation of blood flow to peripheral muscles and is crucial for muscle perfusion [6]. The vascular wall is composed of a monolayer of specialized cells, the endothelial cells, which form the interface between the underlying smooth muscle cells and the vascular lumen [7]. Endothelial cells regulate vascular permeability and maintain vascular tone [8]. Normal arterial function requires a balance between vasodilation and vasoconstriction, which is important for regulating blood flow and vascular tone during rest and exercise [9,10]. Nitric oxide (NO) is a strong vasodilatory and anti-inflammatory signalling molecule, that regulates vascular tone [7]. The release of NO by endothelial cells causes the dilation of an artery, which leads to an increase in blood flow. On the other hand, vasoconstriction is induced by the release of endothelin-1 (ET-1). Endothelial dysfunction (ED) can lead to a reduced NO availability or an increased ET-1 synthesis, release, or activity [7,11]. In addition, hydrogen peroxide (H2O2) might be an important factor in the regulation of vascular tone by functioning as an endothelium-derived hyperpolarizing factor (EDHF) leading to vasoconstriction [12]. The main mechanisms underlying the pathophysiology of ED are increased reactive oxygen species (ROS), inflammation and diminished NO production, and bioavailability [13].
Investigating the link between an individual’s endothelial function and skeletal muscle function is of great interest in the field of exercise physiology [14]. Oxygen delivery and the related mitochondrial capacity of the muscles are regarded as the primary limiting factors for endurance performance [15]. When muscle mass is overperfused during exercise, it has an extremely high capacity for consuming oxygen [15]. Therefore, improved muscle perfusion during exercise via vascular endothelial function can positively impact endurance exercise performance.

1.1. Excessive Production of ROS Results in Decreased Force Output and Decreased NO Availability

Skeletal muscle tissue contraction, which induces a higher oxygen demand, could induce an increased formation of ROS as a result of the increased mitochondrial activity. This can lead to incomplete oxidative phosphorylation during exercise. Short-term increased ROS formation during physical activity, if not excessive, has shown to be important for exercise-induced adaptations including enhanced mitochondrial biogenesis, cardiovascular adaptations, as well as the regulation of contractile force [16,17]. ROS induces redox-sensitive signalling pathways involving redox-sensitive kinases, phosphatases, and the transcription factor nuclear factor-κB leading to an induced skeletal muscle adaptation [16]. The harmful effects of excessive ROS formation can be counteracted by the endogenous antioxidant system comprising superoxide dismutase (SOD), catalase (CAT), reduced glutathione (GSH), and glutathione peroxidase (GPx). However, during long-term, high-intensity endurance exercise, the continuous ROS production may exceed the capacity of the cellular defence system leading to damage to DNA, lipids (lipid peroxidation), or protein in the muscles [18]. Furthermore, ROS generated during exercise modulates muscle contraction signalling pathways; low levels of ROS stimulate force output, whereas high levels attenuate this [19,20].
A possible trigger of ROS production in vascular cells could be the increased blood flow during exercise, thereby increasing the shear stress [21]. There is accumulating evidence suggesting that in ageing and certain disease states such as hypertension, atherosclerosis, and heart failure, there might be an excessive formation of ROS in response to exercise resulting in decreased NO availability through reaction of NO with superoxides [22,23,24]. Via a similar mechanism, the overproduction of ROS during high-intensity exercise (in healthy individuals) leads to a decline in NO availability, whereas supplementation with antioxidants (e.g., hesperidin) can reverse these adverse effects [25]. In this way, the overproduction of ROS is linked to impaired vascular homeostasis and ED.

1.2. Detrimental Effects of Post-Exercise Inflammation on Endurance Performance and Endothelial Function

The immune system plays a key role not only in protecting our bodies from invading microorganisms and disease prevention but also in wound-healing processes [26] and tissue-remodelling mechanisms [27]. Macrophages are specialized cells affecting inflammation and the healing response to acute injury [28]. When it comes to exercise immunity, macrophages play a key role in skeletal muscle regeneration [29]. Despite the fact that the exact macrophage-mediated signalling of inflammation and muscle regeneration is not yet fully understood, several cytokines, including tumour necrosis factor-alpha (TNFα), interferon-gamma (IFNγ), interleukin 6 (IL-6), and interleukin 10 (IL-10), appear to play key roles in the muscle regeneration process [30,31,32].
Although the exercise-induced inflammatory response is important to stimulate muscle adaptations [33,34,35], the post-exercise recovery period is equally critical in providing sufficient time for metabolic and structural adaptations to occur within skeletal muscle, e.g., skeletal muscle hypertrophy [30] and exercise-induced angiogenesis [36]. Unaccustomed exercise (in type, intensity, and duration of training), especially if it requires eccentric (muscle-lengthening) contractions, frequently leads to exercise-induced muscle damage (EIMD) [26]. EIMD is linked to an increase in inflammatory markers both within the injured muscle and the blood, the increased appearance of muscle proteins in the blood, and the delayed onset of muscle soreness (DOMS) [26,37,38]. The acute inflammatory response following EIMD is characterized by increased levels of circulatory and intramuscular inflammatory markers such as C-reactive protein (CRP) and cytokines (e.g., TNF-α and IL-6) [39,40,41].
Without adequate post-exercise/competition recovery periods, an excessive inflammatory response could lead to impaired muscle contractions and force production [30,42]. Moreover, post-exercise inflammation can inhibit the recovery of muscle function, thereby negatively impacting short-term recovery [43,44]. Therefore, if the highly demanding training schedule of professional athletes is not tempered with periods of rest and recovery, a short-term performance decrement can be experienced. This phenomenon, known as overtraining syndrome (OTS) [45], has been associated with a deterioration in endurance performance [46].
The vascular system is important for the inflammatory response because of the transport of systemic immune cells to the site of inflammation. In vessels, acute and chronic inflammation could damage the arterial wall and lead to ED. The generation of ROS released by immune cells plays a central role in limiting the bioavailability of NO and increasing the formation of peroxynitrite (ONOO), which is a highly unstable ROS involved in vascular inflammation, hypertrophy, fibrosis, and ED [47].

1.3. Hesperidin Supplementation: A Potential Ergogenic Aid

The use of ergogenic aids as a strategy to improve exercise performance is widespread among elite as well as recreational athletes [48,49,50,51]. The term “ergogenic aid” includes any training method, mechanic device, or nutritional and pharmacological approach that can improve exercise performance capacity and/or enhance training adaptation [52].
Polyphenols, among other nutritional supplements, have been investigated as ergogenic aids. As their antioxidant and anti-inflammatory role is well-known, polyphenol supplementation could provide an efficient strategy to counteract exercise-related inflammation and prevent cell damage due to an excess of reactive oxygen species (ROS) [53]. Moreover, polyphenols showed the ability to attenuate the delayed onset of muscle soreness (DOMS) [54,55,56,57], a symptom of exercise-induced muscle damage [58]. Other beneficial effects of polyphenols are their capacity to improve physical performance [59] and increase time to exhaustion [60], their anti-fatigue effect [61], and their ability to increase markers of mitochondrial biogenesis (e.g., PGC-1α, SIRT1, mtDNA, and cytochrome c) that are associated with maximal endurance capacity [62]. Taken together, polyphenol supplementation can be used as an ergogenic aid to positively impact exercise performance capacity.
Hesperidin (C28H34O15) is a flavanone belonging to the class of flavonoids, one of the most common and widely distributed groups of plant phenolics, which is abundantly present in citrus fruits [63,64]. Orally administered hesperidin (hesperetin-7-O-rutinoside) is converted to the active aglycone hesperetin by an enzyme that is expressed by intestinal microbiota and subsequently absorbed by the gastrointestinal tract [65]. Human studies showed that after the consumption of orange juice, the maximal plasma concentration of hesperidin (0.1–2.2 micromol/L) is reached between 5 and 7 h after ingestion and is still detected in plasma after 10 h [66,67,68,69]. There are indications that hesperidin supplementation has anti-inflammatory [70,71,72], lipid-lowering [73,74,75], neuro-protective [76,77], and insulin-sensitizing properties [78]. Interestingly, hesperidin has also been investigated for its effects on exercise performance. It is essential to measure the level of endogenous antioxidants, endothelial function, and muscle oxygen supply of a person to determine the right dosage of hesperidin supplementation [79].
This review aims to provide an overview of the existing research evidence on hesperidin supplementation as a potential ergogenic aid. The growing interest in the effects of hesperidin on improved human performance is translated into an increasing number of randomized controlled trials (RCTs) performed on athletes. The exact molecular mechanisms through which hesperidin could lead to enhanced exercise performance are not yet clear. Therefore, in this paper, we investigate the potential molecular mechanisms that could provide sufficient scientific evidence regarding its efficacy, namely improved endothelial function and reduced exercise-induced oxidative stress and inflammation. We selected in vivo RCTs, animal studies, and in vitro studies in which hesperidin, its aglycone form hesperetin, hesperetin-metabolites, or orange juice (in which the hesperidin content is known) are supplemented at any dosage. Studies using combined supplements have been excluded from this narrative review.

2. Hesperidin Increases Endothelial Function

2.1. Hesperidin and Hesperetin Increase NO Production and Decrease Monocyte Adhesion in Endothelial Cells

The effects of hesperidin and hesperetin or their metabolites on endothelial function have been shown in vitro (Table 1). Rizza et al. found that the treatment of bovine aortic endothelial cells (BAECs) with 1μM and 10 μM of hesperetin for 10 min acutely increased cellular levels of phosphorylated (p) 5′AMP-activated protein kinase (AMPK) and protein kinase B (Akt) [71]. Both kinases regulate the activity of endothelial nitric oxide synthase (eNOS), resulting in the increased production of NO. Accordingly, hesperetin treatment of BAECs increased the levels of p-eNOS with a corresponding increase in the NO production [71]. Other studies confirmed the stimulatory effects of hesperidin, hesperetin, or their metabolites with exposure times ranging from 30 min to 24 h on NO production in human umbilical vein endothelial cells (HUVECs) [80,81,82]. The effects of hesperidin on NO production seem to be dose-dependent. Chiou et al. also found decreased levels of strain-induced ET-1 after treatment with hesperidin [82].
In addition to these effects on vasoactive factors, Rizza et al. showed that pre-treatment with hesperetin (10 µM, for 1 h) reduces the TNF-α-stimulated expression of vascular cell adhesion molecule 1 (VCAM-1) as well as TNF-α-stimulated monocyte adhesion [71]. This is in line with other studies showing decreased VCAM-1 levels and decreased monocyte adhesion in TNF-α-stimulated HUVECs pre-treated with hesperetin and its metabolites [83,84]. However, no significant effect was found on the intracellular adhesion molecule 1 (ICAM-1) protein expression [84]. VCAM-1 and ICAM-1 are endothelial adhesion molecules that promote monocyte accumulation in the arterial intima. Increased expression of VCAM-1 was shown to play a major role in the initiation of atherosclerosis [85].

2.2. Hesperidin and Hesperetin Decrease Blood Pressure and Increase Endothelium-Dependent Vasodilation in Hypertensive Rats

The effects of hesperidin, hesperetin, and their metabolites on blood pressure and the vasodilatory response were examined in hypertensive rat models (Table 2). Male Sprague–Dawley rats with hypertension showed decreased systolic blood pressure (SBP) and diastolic blood pressure (DBP) when treated with 15 mg/kg and 30 mg/kg of hesperidin for 5 weeks. Furthermore, increased plasma levels of nitric oxide metabolites (NOx) were found [86]. Administration with hesperetin and its metabolite hesperetin-7-O-glucuronide (HPT7G) (but not Hesperetin-3′-O-glucuronide (HPT3′G)) for 3 min resulted in decreased SBP in hypertensive rats, whereas DBP did not change [87]. In the same study, thoracic aortic rings were isolated from spontaneously hypertensive rats (SHRs) and exposed to 100 µM of HPT7G and HPT3′G. HPT7G but not HPT3′G treatment significantly enhanced endothelium-dependent vasodilation but did not alter endothelium-independent vasodilation. In aortic rings from normotensive control rats (Wistar Kyoto rats), the hesperetin metabolites did not change endothelium-dependent and endothelium-independent vasodilation [87].

2.3. Hesperidin Increases Flow-Mediated Vasodilation and Decreases sVCAM-1 and sICAM-1 in Humans

RCT studies investigating the effects of hesperidin supplementation on endothelial function are collected in Table 3. ED is characterized by reduced vasodilation, which is non-invasively evaluated in vivo via ultrasound flow-mediated vasodilation (FMD) of the peripheral artery [88]. Since FMD responds rapidly to new drug and bioactive substance therapies, it is considered a good marker to assess endothelial function in interventional trials [88]. ED is also characterized by a pro-inflammatory state, which creates favourable conditions for cytokine secretion by immune cells and an increased expression of adhesion molecules on the endothelial cells of the damaged arterial wall [89]. Through the mechanisms of proteolytic cleavage or alternative splicing, adhesion proteins are released in a circulatory form that can be measured in the plasma [90,91]. The released adhesion molecules are an indicator of ED and the pro-inflammatory state. Hence, the studies collected in this review also evaluated the effects of hesperidin supplementation on endothelial function through the increased serum levels of adhesion molecules such as soluble VCAM-1, soluble ICAM-1, and soluble P-selectin (sP-selectin) [92,93]. Hypertension, also known as high blood pressure, is an important risk factor for ED [93]. Therefore, in the following studies, alterations in SBP and DBP were also assessed. When discussing the results of these studies, a distinction has been made between acute and chronic hesperidin supplementation.

2.4. Acute Supplementation

Two studies indicate that blood flow parameters improve 6h after hesperidin supplementation [94,95]. In healthy subjects, the acute administration of 292 mg of hesperidin was able to improve microvascular reactivity measured using combined laser-Doppler flowmetry and iontophoresis [94]. The acute administration of 600 mg of hesperidin significantly improved ischaemic reactive hyperaemia (IRH), a measure of endothelial-dependent vasomotor function, in hypertensive subjects [95]. IRH was measured using a laser-Doppler linear flowmeter taking into account blood perfusion, whereas distal ischaemia was induced by inflating a blood-pressure cuff placed above the elbow to supra-systolic pressure.
Furthermore, supplementation with water-dispersible hesperetin was able to positively impact blood flow in women with cold sensitivity within 70 min after intake [80]. Both concentrations of 17 mg and 170 mg significantly suppressed the drop in blood flow in the air-conditioned room at 22 °C. Schar et al., on the other hand, did not observe any statistically significant changes in multiple vascular function parameters (P-selectin expression, blood pressure, and baroreflex sensitivity) when 320 mg of hesperidin was ingested 5 h before testing in men at moderate risk of cardiovascular disease (CVD) [96]. As noted by the authors, this could be explained by the fact that the plasma concentrations of total flavanone metabolites are only increased until 5 h after hesperidin ingestion.

2.5. Chronic Supplementation

FMD significantly improved in three studies evaluating the chronic supplementation of hesperidin: with an oral dosage of 159.5 mg/day of hesperidin for 7 days in adult subjects with increased cardiovascular risk [97] and 500 mg/d for 3 weeks in individuals with metabolic syndrome [71]. Salden et al. induced acute, reversible ED using a high-fat meal in subjects with a baseline FMD ≥3% [98]. In this study, hesperidin supplementation (450 mg/day for 6 weeks) significantly protected against postprandial FMD impairment compared to the placebo. Yari et al. recorded a BP-lowering effect after hesperidin intake; SBP significantly decreased in subjects with metabolic syndrome after hesperidin intake (1 g/day for 12 weeks) compared to the placebo [75]. In the study by Morand et al., DBP was significantly decreased after 4 weeks of hesperidin supplementation (292 mg/day) [94]. The endothelial-dependent vasomotor function marker IRH improved after oral intake of 600 mg/day of hesperidin during an intervention of 12 weeks [95]. In one study, the chronic effect of hesperidin supplementation (292 mg/day for 4 weeks) was evaluated on NO production. Despite no significant change compared to the placebo, an increasing trend in NOx was recorded in the intervention group [94].
A significant decrease in sVCAM-1 and sICAM-1 was observed after 6 weeks of hesperidin supplementation (450 mg/day) compared to the placebo [98]. No significant changes were recorded in the same biomarkers in the studies of Rizza et al. and Morand et al. where the hesperidin supplementation lasted 3 weeks (500 mg/day) and 4 weeks (292 mg/day), respectively [71,94]. Those findings may suggest that a longer supplementation with the flavanone hesperidin is required to significantly affect the serum levels of the abovementioned cellular adhesion molecules.

3. Hesperidin Reduces Exercise-Induced Oxidative Stress

3.1. Hesperidin and Hesperetin Function as an Antioxidant In Vitro

The results showed that the ROS scavenging activity (with the exception of ·NO scavenging) of hesperidin/hesperetin was comparable to the mentioned standards (Table 4) [99,100]. Furthermore, hesperetin decreased cellular ROS formation induced by tert-butylhydroperoxide (t-BHP) and lipopolysaccharides (LPS) in in vitro models using multiple cell types (including endothelial cells, hepatic cells, macrophage cells, and fibroblasts) [82,100,101,102]. Additionally, Kaplana et al. and Chen et al. showed that hesperidin treatment reduced the by-products of lipid peroxidation in the human erythrocyte membrane, measured as thiobarbituric acid-reactive substances (TBARS) and malondialdehyde (MDA), respectively [99,101]. Hesperidin and other polyphenols also showed the potential to affect the endogenous antioxidant status by increasing nuclear factor erythroid 2-related factor 2 (Nrf2) nuclear translocation. In human hepatocytes, increased Nrf2 translocation leads to increased mRNA and protein levels of endogenous antioxidants (e.g., SOD1, GST, thioredoxin, and HO-1) and enhances their activities [101,103].

3.2. Hesperidin Decreases ROS and Increases Antioxidant Markers in Rats

Rats were supplemented with hesperidin for a duration ranging from 10 days to 5 weeks (Table 5). In the study of Estruel-Amades et al., Wister rats were trained for five weeks (five days per week) including two exhaustion tests and three trainings per week [104]. The oxidative status was determined before and immediately after an additional exhaustion test. Hesperidin prevented the increase in ROS production by peritoneal macrophages induced by the exhaustion test. Moreover, supplementation with hesperidin avoided the decrease in SOD activity in the thymus and the decrease in CAT activity in the spleen and liver induced by the exhaustion test. Sedentary animals supplemented with hesperidin showed decreased activity of SOD, CAT, and GPx in the mentioned tissue sections. The same applies to the trained animals in which hesperidin supplementation led to either a decrease or no change in antioxidant activity compared to the controls (Table 5) [104]. In the study of El-Sayed et al., the neurotoxin acrylonitrile was used to induce ROS formation in rat brain tissue [105]. Supplementation with hesperidin (200 mg/kg/day) ameliorated the acrylonitrile-induced alterations in brain lipid peroxidation and increased the acrylonitrile-induced reduction in GSH, SOD, CAT, GPx, and glutathione-s-transferase (GST) levels in the brain. Furthermore, increased SOD and GPx levels and decreased CAT levels were found in hesperidin-supplemented rats compared to control rats without any treatment with acrylonitrile. According to this, Sahu et al. showed that hesperidin supplementation with the same dosage for 10 days leads to decreased cisplatin (a cancer treatment known to induce nephrotoxicity)-induced levels of ROS and TBARS and increased activity of antioxidants (including SOD, GSH, CAT, GPx, GST, and glutathione reductase (GR)) in rat kidneys [106]. Without stimulating ROS production, no significant differences in oxidative status were found between hesperidin-treated and control animals [106]. Moreover, a study in hypertensive rats showed decreased vascular superoxide production and decreased plasma levels of MDA after a 5-week administration with hesperidin [86].

3.3. Hesperidin Supplementation Increases CAT and Decreases MDA after Strenuous Exercise Performance in Humans

Acute supplementation of hesperidin (500 mg) increased the endogenous antioxidant enzyme catalase (CAT) in venous blood samples after a strenuous exercise performance measured by a Wingate test on a cycle ergometer in male amateur cyclists (Table 6) [107]. On the other hand, the concentration of other endogenous antioxidant markers, such as superoxide dismutase (SOD) and glutathione (GSH), and lipid oxidation markers, such as thiobarbituric acid-reactive substances (TBARS), did not show any significant difference between the intervention and control groups, despite a decreasing trend observed for SOD in the intervention group.
Acute supplementation with 217 mg of hesperidin in healthy soccer players decreased the lipid peroxidation marker malondialdehyde (MDA) post-exercise in plasma [108]. Plasma total antioxidant status (TAS) significantly increased after exercise in both the intervention and placebo groups; however, there was no significant difference present between the control and hesperidin intervention groups.

4. Hesperidin Reduces Inflammatory Markers

4.1. Hesperidin and Hesperetin Decrease Pro-Inflammatory Responses in LPS-Stimulated Macrophages

The effects of hesperidin/hesperetin on inflammatory responses were investigated in Macrophage RAW264.7 cells (Table 7). Treatment with HPT7G for 24 h showed decreased LPS-induced inflammatory responses measured by a decrease in the mRNA expression of IL-6, IL-1β, TNF-α (only at a concentration of 50μM), and COX-2, and a decreased production of NO, IL-6, and IL-1β. No effects of the flavanone were found on the LPS-induced production of TNF-α [109]. Other studies of the same cell type found that hesperidin and hesperetin exposure with a duration ranging from 30 min to 24 h resulted in a decreased LPS-induced production of PGE2, NO, and NO2, and decreased protein levels of COX-2 and iNOS [102,110,111]. Furthermore, conflicting results were found concerning the effects of hesperidin and hesperetin on the activation of NF-κB. Although one study found decreased NF-κB activity, the study of Kazlowska et al. showed no effects of the flavanone on NF-κB and iNOS promotor activity [102,111].

4.2. Hesperidin Decreases Renal and Plasma Levels of TNF-α in Rat and Mouse Models

The effects of short-term (3 h) and long-term (10 days and 5 weeks) supplementation with hesperidin on TNF-α levels were investigated in animal studies (Table 8). Treatment with 0.3 mg, 1 mg, and 3 mg hesperidin three hours before LPS stimulation led to decreased plasma levels of TNF-α in female mice [112]. The same was observed in male Wistar rats in which 10 days of supplementation with 200 mg/kg/day of hesperidin led to a decrease in the cisplatin (a cancer treatment known to induce nephrotoxicity)-induced increase in renal TNF-α. In the same study, a reduction in cisplatin-induced neutrophil infiltration was observed after supplementation with hesperidin, assessed by the measurement of renal myeloperoxidase (MPO) activity [106]. Moreover, in hypertensive rats, 5 weeks of hesperidin supplementation resulted in decreased plasma values of TNF-α [86].

4.3. Hesperidin Decreases CRP, TNF-α, and IL-6 in Humans

In RCTs with hesperidin supplementation for a period of 1–12 weeks performed in healthy adults as well as individuals with medical conditions, such as rheumatoid arthritis, metabolic syndrome (MetS), or increased cardiovascular risk, decreased levels of CRP tumour necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6) were found (Table 9). A significant decrease in CRP, TNF-α, and IL-6 concentrations was measured after 7 days of hesperidin supplementation (159.5 mg/day) in subjects with increased cardiovascular risk [97]. An amount of 1 g of hesperidin per day for 12 weeks decreased TNF-α but not in CRP in subjects with MetS [75]. Kometani et al. recorded a significant decrease in CRP concentration in subjects with arthritis after 12 weeks of supplementation with 3 g of hesperidin per day compared to the placebo [113]. When tested in healthy men, 292 mg hesperidin per day for 4 weeks did not show an effect on IL-6 and CRP concentrations in the intervention group compared to the placebo [94].
In summary, the available human studies indicate that the CRP concentration in serum can be decreased by hesperetin supplementation. Two of these studies also decreased TNF-α levels in serum after hesperidin supplementation compared to controls, whereas the effects of the supplementation on IL-6 levels were inconclusive.

5. Hesperidin Improves Exercise Performance

5.1. Hesperidin Supplementation Increases Maximum Running Performance in Rats

For a period of five weeks, female rats performed a maximum distance run until exhaustion two times per week and were supplemented with 200 mg/kg of hesperidin or a placebo three times per week. Non-supplemented animals achieved the highest performance in week two, in which they ran about 134% of the maximum distance compared to the first exhaustion test. Animals supplemented with hesperidin showed a significantly better performance compared to the control group, reaching their peak performance in week three, running 158% of the maximum distance compared to the first test (Table 10) [104].

5.2. Hesperidin Improves Anaerobic Exercise Performance Outcomes in Human

Ingesting 500 mg of hesperidin 5h before a repeated sprints test (Wingate test) was able to improve anaerobic performance outcomes (average power (W); maximal speed (rpm); and total energy (J)) in the intervention group compared to the placebo [107] (Table 11). Ingesting 217 mg of hesperidin 2.5 h before a Yo-Yo intermittent recovery test (YYIRT) did not result in a significant improvement in the ratings of perceived exertion (RPE) and maximal oxygen uptake (VO2max) [108]. However, an increasing trend in VO2max was recorded in the intervention group compared to the placebo. VO2max is defined as the maximum rate of oxygen consumption measured during severe exercise [114]. In exercise physiology, VO2max is used to assess endurance performance and it is limited by the ability of the cardiorespiratory system to deliver oxygen to the exercising muscles [15]. No significant improvement in the estimated VO2max was recorded during a 10 min time trial on a cycle ergometer after 4 weeks of hesperidin (450 mg/day) supplementation [115]. In the same study, performance outcomes such as power (W) and VO2/power ratio significantly improved in the intervention group compared to the placebo, resulting in a higher amount of power produced per unit of oxygen consumed (VO2/power ratio).
Martínez-Noguera et al. and Van Iersel et al. both tested the chronic effects of hesperidin supplementation on sport performance outcomes after a Wingate test [116,117]. Supplementation with 500 mg/day of hesperidin for 8 weeks significantly increased absolute peak power (W) and relative peak power (W) in male amateur cyclists [116]. The oral ingestion of 360 mg and 450 mg of hesperidin/day for 4 weeks significantly improved average power (W) and 5 s peak power (W) recorded during a Wingate anaerobic test performed in trained healthy subjects [117]. Average power (W) was still significantly improved after 8 weeks of hesperidin supplementation (360 mg/day). Moreover, Martínez- Noguera et al. also evaluated the effects of a 500 mg/day hesperidin supplementation for 8 weeks after an incremental test until exhaustion and found a significant improvement in maximum power (W) and estimated functional threshold power (FTP) (W) [116].

6. Discussion and Conclusions

The studies collected in this review show the potential of hesperidin, hesperetin, and their metabolites to enhance exercise performance by (i) improving endothelial function (via increased NO availability; Figure 1), (ii) reducing oxidative stress (by acting as an antioxidant, e.g., as a ROS scavenger or enhancer of endogenous antioxidant capacity; Figure 2), and (iii) inhibiting the production of pro-inflammatory cytokines to prevent excessive post-exercise inflammation (Figure 3).
In vitro studies investigating the effects of hesperidin, hesperetin, and their metabolites in endothelial cells highlight the potential of the flavanone to enhance the production of NO in the vascular endothelium. There is growing evidence showing that increased NO availability can improve exercise-related performance through enhanced tissue oxygenation (due to blood vessel vasodilation) combined with increased metabolic efficiency in active skeletal muscle [118]. Increased NO availability can enhance skeletal muscle metabolic efficiency by increasing contractile function through alterations in calcium availability and sensitivity in the sarcoplasmic reticulum, resulting in the reduced ATP cost of the muscle force production [119]. Skeletal muscle contraction requires ATP both for the interaction between actin and myosin (actomyosin-ATPase) and for the calcium (Ca2+) pumping in the sarcoplasmic reticulum (Ca2+-ATPase) [119]. NO, being able to reduce Ca2+ release from the sarcoplasmic reticulum [120] and inhibit Ca2+-ATPase activity [121], can decrease the energetic cost of muscle force production. This allows high-intensity exercise to be tolerated for a greater period of time. The combination of improved oxygen delivery to the muscle and the related mitochondrial capacity is very important as too much oxygen could induce oxidative stress by overloading the mitochondrial respiration system. Exhaustive aerobic exercise has recently been shown to attenuate maximal skeletal muscle mitochondrial respiratory capacity through the inhibition of oxidative phosphorylation [122]. When it comes to athletes, this likely transient, mitochondrial defect could amplify the exercise-induced development of fatigue [123]. Therefore, investigating the effects of hesperidin on mitochondrial capacity could be an important area for future research.
The studies collected in this review showed enhanced vasodilator responses after supplementation with hesperidin in both healthy and unhealthy individuals/animals. Although in subjects with hypertension, there is a different regulation in blood vessel vasomotor responses compared to healthy people and, therefore, athletes [93,124]. Future studies performed on healthy, trained subjects are needed to assess the efficacy of hesperidin supplementation on vasomotor responses and endothelial function and to eventually translate those effects into improvements in exercise performance.
The included studies were consistent in the ability of hesperidin and hesperetin to inhibit ROS production in a variety of cell types and tissues. Despite the broad amount of literature supporting the role of hesperidin in antioxidant cellular defences, there is still a lack of studies focusing specifically on its effects on skeletal muscles. More RCTs should be conducted to ascertain the effects of hesperidin on oxidative status after exercise [125]. Furthermore, future investigations should assess the baseline levels of endogenous antioxidants in the muscles and endothelium of trained/untrained and healthy/unhealthy subjects. As there could be differences in the baseline antioxidants between individuals, this knowledge could be used to determine the most effective and personalized dose of hesperidin supplementation. Moreover, it is important to highlight the fact that hesperidin works as an exogenous antioxidant and if reacted with ROS, it cannot be converted to its reduced form again by endogenous antioxidant enzymes. Therefore, it is recommended to supplement hesperidin multiple times per day depending on individual needs to ensure the sufficient availability of the reduced form of hesperidin or enhance the endogenous antioxidant network to channel the reactivity of radicals into the antioxidant network [126]. Finally, it would be interesting to investigate whether hesperidin can decrease ROS formation in vessels surrounding the contracting muscles to see if this can be linked to improvements in NO availability and muscle perfusion during exercise.
Hesperidin and hesperetin showed good anti-inflammatory properties by decreasing the production and plasma levels of pro-inflammatory markers. Despite the evidence from studies performed on untrained and unhealthy subjects, we do not have enough data to support the role of hesperidin in restraining systemic inflammation in overtrained subjects. More research is needed to validate our hypothesis that the anti-inflammatory properties of hesperidin can lead to a reduction in intramuscular inflammation and muscle damage, and in this way result in increased exercise performance. Future studies should not only investigate the effects of hesperidin supplementation on systemic post-exercise inflammation markers but also evaluate the changes in intramuscular inflammation markers via skeletal muscle biopsies.
Finally, the effects of hesperidin supplementation on improved exercise performance have been investigated. In rats, supplementation with hesperidin led to increased performance in maximal running distance. In trained athletes, both acute and chronic hesperidin intake was able to improve multiple anaerobic exercise outcomes (e.g., power, speed, energy). Further studies are needed to assess the effects of hesperidin supplementation on endurance exercise in humans.
In conclusion, the ergogenic effects that hesperidin can bring to the spectrum of improved exercise performance are promising and should be investigated further. For elite and recreational athletes, hesperidin could be a promising food supplement to optimize the oxygen and nutrient supplies of the muscles, stimulate muscle contraction, and enhance muscle recovery between training sessions. During exercise, hesperidin supplementation can increase endothelial function, thereby contributing to increased skeletal muscle perfusion and increasing oxygen (O2) efflux to the muscle, which is associated with increased endurance performance. Moreover, hesperidin can decrease ROS-mediated damage in muscle cells, which enhances muscle function. Finally, hesperidin can decrease post-exercise-induced inflammation, which potentially speeds up the recovery process and can thereby improve exercise performance. In this way, personalized supplementation with hesperidin seems to increase anaerobic exercise performance, although further research is necessary to draw conclusions regarding the efficiency of hesperidin supplementation for endurance athletes.

Author Contributions

M.I. and I.C. contributed equally to this paper. Conceptualization, M.I., I.C., and M.M.J.P.E.S.; investigation, M.I. and I.C.; writing—original draft preparation, M.I. and I.C.; writing—review and editing, M.M.J.P.E.S. and F.J.T.; visualization, M.I. and I.C.; supervision, M.M.J.P.E.S. and F.J.T. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

M.I. is an employee of BioActor BV. All other authors declare no conflicts of interest.

References

  1. Barclay, C.J. Energy demand and supply in human skeletal muscle. J. Muscle Res. Cell Motil. 2017, 38, 143–155. [Google Scholar] [CrossRef] [PubMed]
  2. Hawley, J.A.; Hargreaves, M.; Joyner, M.J.; Zierath, J.R. Integrative biology of exercise. Cell 2014, 159, 738–749. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Poole, D.C.; Copp, S.W.; Hirai, D.M.; Musch, T.I. Dynamics of muscle microcirculatory and blood-myocyte O2 flux during contractions. Acta Physiol. 2011, 202, 293–310. [Google Scholar] [CrossRef] [PubMed]
  4. Segal, S.S.; Kurjiaka, D.T. Coordination of blood flow control in the resistance vasculature of skeletal muscle. Med. Sci. Sports Exerc. 1995, 27, 1158–1164. [Google Scholar] [CrossRef]
  5. Brodal, P.; Ingjer, F.; Hermansen, L. Capillary supply of skeletal muscle fibers in untrained and endurance-trained men. Am. J. Physiol. 1977, 232, H705–H712. [Google Scholar] [CrossRef]
  6. Kabbach, E.Z.; Heubel, A.D.; da Luz Goulart, C.; Di Lorenzo, V.A.P.; Phillips, S.A.; Borghi-Silva, A.; Mendes, R.G. Association of exercise capacity and endothelial function in patients with severe exacerbations of chronic obstructive pulmonary disease. Sci. Rep. 2021, 11, 461. [Google Scholar] [CrossRef]
  7. Cyr, A.R.; Huckaby, L.V.; Shiva, S.S.; Zuckerbraun, B.S. Nitric Oxide and Endothelial Dysfunction. Crit. Care Clin. 2020, 36, 307–321. [Google Scholar] [CrossRef]
  8. Sturtzel, C. Endothelial Cells. In The Immunology of Cardiovascular Homeostasis and Pathology; Sattler, S., Kennedy-Lydon, T., Eds.; Springer International Publishing: Cham, Switzerland, 2017; pp. 71–91. [Google Scholar] [CrossRef]
  9. Lambiase, M.J.; Dorn, J.; Thurston, R.C.; Roemmich, J.N. Flow-mediated dilation and exercise blood pressure in healthy adolescents. J. Sci. Med. Sport 2014, 17, 425–429. [Google Scholar] [CrossRef] [Green Version]
  10. Duncker, D.J.; Bache, R.J. Regulation of coronary blood flow during exercise. Physiol. Rev. 2008, 88, 1009–1086. [Google Scholar] [CrossRef] [Green Version]
  11. Marasciulo, F.L.; Montagnani, M.; Potenza, M.A. Endothelin-1: The yin and yang on vascular function. Curr. Med. Chem. 2006, 13, 1655–1665. [Google Scholar] [CrossRef]
  12. Garry, A.; Edwards, D.H.; Fallis, I.F.; Jenkins, R.L.; Griffith, T.M. Ascorbic acid and tetrahydrobiopterin potentiate the EDHF phenomenon by generating hydrogen peroxide. Cardiovasc. Res. 2009, 84, 218–226. [Google Scholar] [CrossRef] [Green Version]
  13. Vanhoutte, P.M.; Shimokawa, H.; Tang, E.H.; Feletou, M. Endothelial dysfunction and vascular disease. Acta Physiol. 2009, 196, 193–222. [Google Scholar] [CrossRef] [Green Version]
  14. Hendrickse, P.; Degens, H. The role of the microcirculation in muscle function and plasticity. J. Muscle Res. Cell Motil. 2019, 40, 127–140. [Google Scholar] [CrossRef] [Green Version]
  15. Bassett, D.R., Jr.; Howley, E.T. Limiting factors for maximum oxygen uptake and determinants of endurance performance. Med. Sci. Sports Exerc. 2000, 32, 70–84. [Google Scholar] [CrossRef]
  16. Powers, S.K.; Duarte, J.; Kavazis, A.N.; Talbert, E.E. Reactive oxygen species are signalling molecules for skeletal muscle adaptation. Exp. Physiol. 2010, 95, 1–9. [Google Scholar] [CrossRef]
  17. Westerblad, H.; Allen, D.G. Emerging roles of ROS/RNS in muscle function and fatigue. Antioxid. Redox Signal. 2011, 15, 2487–2499. [Google Scholar] [CrossRef]
  18. Mastaloudis, A.; Leonard, S.W.; Traber, M.G. Oxidative stress in athletes during extreme endurance exercise. Free Radic. Biol. Med. 2001, 31, 911–922. [Google Scholar] [CrossRef]
  19. Radak, Z.; Zhao, Z.; Koltai, E.; Ohno, H.; Atalay, M. Oxygen consumption and usage during physical exercise: The balance between oxidative stress and ROS-dependent adaptive signaling. Antioxid. Redox Signal. 2013, 18, 1208–1246. [Google Scholar] [CrossRef] [Green Version]
  20. Radak, Z.; Chung, H.Y.; Koltai, E.; Taylor, A.W.; Goto, S. Exercise, oxidative stress and hormesis. Ageing Res. Rev. 2008, 7, 34–42. [Google Scholar] [CrossRef]
  21. Hsieh, H.-J.; Liu, C.-A.; Huang, B.; Tseng, A.H.H.; Wang, D.L. Shear-induced endothelial mechanotransduction: The interplay between reactive oxygen species (ROS) and nitric oxide (NO) and the pathophysiological implications. J. Biomed. Sci. 2014, 21, 3. [Google Scholar] [CrossRef] [Green Version]
  22. Minuz, P.; Patrignani, P.; Gaino, S.; Degan, M.; Menapace, L.; Tommasoli, R.; Seta, F.; Capone, M.L.; Tacconelli, S.; Palatresi, S.; et al. Increased oxidative stress and platelet activation in patients with hypertension and renovascular disease. Circulation 2002, 106, 2800–2805. [Google Scholar] [CrossRef] [Green Version]
  23. Hajjar, D.P.; Gotto, A.M., Jr. Biological relevance of inflammation and oxidative stress in the pathogenesis of arterial diseases. Am. J. Pathol. 2013, 182, 1474–1481. [Google Scholar] [CrossRef] [Green Version]
  24. Heymes, C.; Bendall, J.K.; Ratajczak, P.; Cave, A.C.; Samuel, J.L.; Hasenfuss, G.; Shah, A.M. Increased myocardial NADPH oxidase activity in human heart failure. J. Am. Coll. Cardiol. 2003, 41, 2164–2171. [Google Scholar] [CrossRef] [Green Version]
  25. Wang, Y.X.; Liu, H.B.; Li, P.S.; Yuan, W.X.; Liu, B.; Liu, S.T.; Qin, K.R. ROS and NO Dynamics in Endothelial Cells Exposed to Exercise-Induced Wall Shear Stress. Cell. Mol. Bioeng. 2019, 12, 107–120. [Google Scholar] [CrossRef]
  26. Clarkson, P.M.; Hubal, M.J. Exercise-induced muscle damage in humans. Am. J. Phys. Med. Rehabil. 2002, 81, S52–S69. [Google Scholar] [CrossRef]
  27. Kratofil, R.M.; Kubes, P.; Deniset, J.F. Monocyte Conversion During Inflammation and Injury. Arter. Thromb. Vasc. Biol. 2017, 37, 35–42. [Google Scholar] [CrossRef] [Green Version]
  28. Oishi, Y.; Manabe, I. Macrophages in inflammation, repair and regeneration. Int. Immunol. 2018, 30, 511–528. [Google Scholar] [CrossRef]
  29. Chazaud, B. Inflammation and Skeletal Muscle Regeneration: Leave It to the Macrophages! Trends Immunol. 2020, 41, 481–492. [Google Scholar] [CrossRef]
  30. Cheng, A.J.; Jude, B.; Lanner, J.T. Intramuscular mechanisms of overtraining. Redox Biol. 2020, 35, 101480. [Google Scholar] [CrossRef]
  31. Paulsen, G.; Crameri, R.; Benestad, H.B.; Fjeld, J.G.; Mørkrid, L.; Hallén, J.; Raastad, T. Time course of leukocyte accumulation in human muscle after eccentric exercise. Med. Sci. Sports Exerc. 2010, 42, 75–85. [Google Scholar] [CrossRef]
  32. Arango Duque, G.; Descoteaux, A. Macrophage cytokines: Involvement in immunity and infectious diseases. Front. Immunol. 2014, 5, 491. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Paulsen, G.; Ramer Mikkelsen, U.; Raastad, T.; Peake, J.M. Leucocytes, cytokines and satellite cells: What role do they play in muscle damage and regeneration following eccentric exercise? Exerc. Immunol. Immunol. Rev. 2012, 18, 42–79. [Google Scholar]
  34. Tidball, J.G. Regulation of muscle growth and regeneration by the immune system. Nat. Rev. Immunol. 2017, 17, 165–178. [Google Scholar] [CrossRef] [PubMed]
  35. Tidball, J.G.; Villalta, S.A. Regulatory interactions between muscle and the immune system during muscle regeneration. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2010, 298, R1173–R1187. [Google Scholar] [CrossRef] [Green Version]
  36. Gorski, T.; De Bock, K. Metabolic regulation of exercise-induced angiogenesis. Vasc. Biol. 2019, 1, H1–H8. [Google Scholar] [CrossRef] [Green Version]
  37. Hyldahl, R.D.; Hubal, M.J. Lengthening our perspective: Morphological, cellular, and molecular responses to eccentric exercise. Muscle Nerve 2014, 49, 155–170. [Google Scholar] [CrossRef]
  38. McFarlin, B.K.; Venable, A.S.; Henning, A.L.; Sampson, J.N.B.; Pennel, K.; Vingren, J.L.; Hill, D.W. Reduced inflammatory and muscle damage biomarkers following oral supplementation with bioavailable curcumin. BBA Clin. 2016, 5, 72–78. [Google Scholar] [CrossRef] [Green Version]
  39. Bell, P.G.; Walshe, I.H.; Davison, G.W.; Stevenson, E.; Howatson, G. Montmorency cherries reduce the oxidative stress and inflammatory responses to repeated days high-intensity stochastic cycling. Nutrients 2014, 6, 829–843. [Google Scholar] [CrossRef] [Green Version]
  40. Buford, T.W.; Cooke, M.B.; Willoughby, D.S. Resistance exercise-induced changes of inflammatory gene expression within human skeletal muscle. Eur. J. Appl. Physiol. 2009, 107, 463–471. [Google Scholar] [CrossRef]
  41. Vella, L.; Markworth, J.F.; Peake, J.M.; Snow, R.J.; Cameron-Smith, D.; Russell, A.P. Ibuprofen supplementation and its effects on NF-κB activation in skeletal muscle following resistance exercise. Physiol. Rep. 2014, 2, e12172. [Google Scholar] [CrossRef]
  42. Place, N.; Ivarsson, N.; Venckunas, T.; Neyroud, D.; Brazaitis, M.; Cheng, A.J.; Ochala, J.; Kamandulis, S.; Girard, S.; Volungevičius, G.; et al. Ryanodine receptor fragmentation and sarcoplasmic reticulum Ca2+ leak after one session of high-intensity interval exercise. Proc. Natl. Acad. Sci. USA 2015, 112, 15492–15497. [Google Scholar] [CrossRef] [Green Version]
  43. Peake, J.M.; Markworth, J.F.; Nosaka, K.; Raastad, T.; Wadley, G.D.; Coffey, V.G. Modulating exercise-induced hormesis: Does less equal more? J. Appl. Physiol. 2015, 119, 172–189. [Google Scholar] [CrossRef] [Green Version]
  44. Pizza, F.X.; Peterson, J.M.; Baas, J.H.; Koh, T.J. Neutrophils contribute to muscle injury and impair its resolution after lengthening contractions in mice. J. Physiol. 2005, 562, 899–913. [Google Scholar] [CrossRef]
  45. Meeusen, R.; Duclos, M.; Foster, C.; Fry, A.; Gleeson, M.; Nieman, D.; Raglin, J.; Rietjens, G.; Steinacker, J.; Urhausen, A. Prevention, diagnosis, and treatment of the overtraining syndrome: Joint consensus statement of the European College of Sport Science and the American College of Sports Medicine. Med. Sci. Sports Exerc. 2013, 45, 186–205. [Google Scholar] [CrossRef] [Green Version]
  46. Lehmann, M.; Foster, C.; Keul, J. Overtraining in endurance athletes: A brief review. Med. Sci. Sports Exerc. 1993, 25, 854–862. [Google Scholar] [CrossRef]
  47. Zanoli, L.; Briet, M.; Empana, J.P.; Cunha, P.G.; Mäki-Petäjä, K.M.; Protogerou, A.D.; Tedgui, A.; Touyz, R.M.; Schiffrin, E.L.; Spronck, B.; et al. Vascular consequences of inflammation: A position statement from the ESH Working Group on Vascular Structure and Function and the ARTERY Society. J. Hypertens. 2020, 38, 1682–1698. [Google Scholar] [CrossRef]
  48. Froiland, K.; Koszewski, W.; Hingst, J.; Kopecky, L. Nutritional supplement use among college athletes and their sources of information. Int. J. Sport Nutr. Exerc. Metab. 2004, 14, 104–120. [Google Scholar] [CrossRef]
  49. Maughan, R.J.; Greenhaff, P.L.; Hespel, P. Dietary supplements for athletes: Emerging trends and recurring themes. J. Sports Sci. 2011, 29 (Suppl. 1), S57–S66. [Google Scholar] [CrossRef]
  50. Jenkinson, D.M.; Harbert, A.J. Supplements and sports. Am. Fam. Physician 2008, 78, 1039–1046. [Google Scholar]
  51. McGuine, T.A.; Sullivan, J.C.; Bernhardt, D.T. Creatine supplementation in high school football players. Clin. J. Sport Med 2001, 11, 247–253. [Google Scholar] [CrossRef]
  52. Kreider, R.B.; Wilborn, C.D.; Taylor, L.; Campbell, B.; Almada, A.L.; Collins, R.; Cooke, M.; Earnest, C.P.; Greenwood, M.; Kalman, D.S.; et al. ISSN exercise & sport nutrition review: Research & recommendations. J. Int. Soc. Sports Nutr. 2010, 7, 7. [Google Scholar] [CrossRef] [Green Version]
  53. Sastre, J.; Asensi, M.; Gascó, E.; Pallardó, F.V.; Ferrero, J.A.; Furukawa, T.; Viña, J. Exhaustive physical exercise causes oxidation of glutathione status in blood: Prevention by antioxidant administration. Am. J. Physiol. 1992, 263, R992–R995. [Google Scholar] [CrossRef]
  54. Romain, C.; Freitas, T.T.; Martínez-Noguera, F.J.; Laurent, C.; Gaillet, S.; Chung, L.H.; Alcaraz, P.E.; Cases, J. Supplementation with a Polyphenol-Rich Extract, TensLess®, Attenuates Delayed Onset Muscle Soreness and Improves Muscle Recovery from Damages After Eccentric Exercise. Phytotherapy Res. 2017, 31, 1739–1746. [Google Scholar] [CrossRef]
  55. Nicol, L.M.; Rowlands, D.S.; Fazakerly, R.; Kellett, J. Curcumin supplementation likely attenuates delayed onset muscle soreness (DOMS). Eur. J. Appl. Physiol. 2015, 115, 1769–1777. [Google Scholar] [CrossRef]
  56. Trombold, J.R.; Barnes, J.N.; Critchley, L.; Coyle, E.F. Ellagitannin consumption improves strength recovery 2-3 d after eccentric exercise. Med. Sci. Sports Exerc. 2010, 42, 493–498. [Google Scholar] [CrossRef] [Green Version]
  57. Ammar, A.; Turki, M.; Chtourou, H.; Hammouda, O.; Trabelsi, K.; Kallel, C.; Abdelkarim, O.; Hoekelmann, A.; Bouaziz, M.; Ayadi, F.; et al. Pomegranate Supplementation Accelerates Recovery of Muscle Damage and Soreness and Inflammatory Markers after a Weightlifting Training Session. PLoS ONE 2016, 11, e0160305. [Google Scholar] [CrossRef] [Green Version]
  58. Gulick, D.T.; Kimura, I.F. Delayed onset muscle soreness: What is it and how do we treat it? J. Sport Rehabil. 1996, 5, 234–243. [Google Scholar] [CrossRef]
  59. Murase, T.; Haramizu, S.; Shimotoyodome, A.; Nagasawa, A.; Tokimitsu, I. Green tea extract improves endurance capacity and increases muscle lipid oxidation in mice. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2005, 288, R708–R715. [Google Scholar] [CrossRef] [Green Version]
  60. Malaguti, M.; Angeloni, C.; Hrelia, S. Polyphenols in exercise performance and prevention of exercise-induced muscle damage. Oxid. Med. Cell. Longev. 2013, 2013, 825928. [Google Scholar] [CrossRef] [Green Version]
  61. Swamy, M.; Naveen, S.; Singsit, D.; Naika, M.; Khanum, F. Anti-fatigue effects of polyphenols extracted from pomegranate peel. Int. J. Integr. Biol. 2011, 11, 69–72. [Google Scholar]
  62. Davis, J.M.; Murphy, E.A.; Carmichael, M.D.; Davis, B. Quercetin increases brain and muscle mitochondrial biogenesis and exercise tolerance. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2009, 296, R1071–R1077. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Bravo, L. Polyphenols: Chemistry, dietary sources, metabolism, and nutritional significance. Nutr. Rev. 1998, 56, 317–333. [Google Scholar] [CrossRef] [PubMed]
  64. Garg, A.; Garg, S.; Zaneveld, L.J.; Singla, A.K. Chemistry and pharmacology of the Citrus bioflavonoid hesperidin. Phytotherapy Res. 2001, 15, 655–669. [Google Scholar] [CrossRef] [PubMed]
  65. Jin, M.J.; Kim, U.; Kim, I.S.; Kim, Y.; Kim, D.-H.; Han, S.B.; Kim, D.-H.; Kwon, O.-S.; Yoo, H.H. Effects of Gut Microflora on Pharmacokinetics of Hesperidin: A Study on Non-Antibiotic and Pseudo-Germ-Free Rats. J. Toxicol. Environ. Health Part A 2010, 73, 1441–1450. [Google Scholar] [CrossRef]
  66. Brett, G.M.; Hollands, W.; Needs, P.W.; Teucher, B.; Dainty, J.R.; Davis, B.D.; Brodbelt, J.S.; Kroon, P.A. Absorption, metabolism and excretion of flavanones from single portions of orange fruit and juice and effects of anthropometric variables and contraceptive pill use on flavanone excretion. Br. J. Nutr. 2009, 101, 664–675. [Google Scholar] [CrossRef] [Green Version]
  67. Nielsen, I.L.; Chee, W.S.; Poulsen, L.; Offord-Cavin, E.; Rasmussen, S.E.; Frederiksen, H.; Enslen, M.; Barron, D.; Horcajada, M.N.; Williamson, G. Bioavailability is improved by enzymatic modification of the citrus flavonoid hesperidin in humans: A randomized, double-blind, crossover trial. J. Nutr. 2006, 136, 404–408. [Google Scholar] [CrossRef] [Green Version]
  68. Erlund, I.; Meririnne, E.; Alfthan, G.; Aro, A. Plasma Kinetics and Urinary Excretion of the Flavanones Naringenin and Hesperetin in Humans after Ingestion of Orange Juice and Grapefruit Juice. J. Nutr. 2001, 131, 235–241. [Google Scholar] [CrossRef]
  69. Manach, C.; Morand, C.; Gil-Izquierdo, A.; Bouteloup-Demange, C.; Rémésy, C. Bioavailability in humans of the flavanones hesperidin and narirutin after the ingestion of two doses of orange juice. Eur. J. Clin. Nutr. 2003, 57, 235–242. [Google Scholar] [CrossRef] [Green Version]
  70. Haidari, F.; Heybar, H.; Jalali, M.T.; Ahmadi Engali, K.; Helli, B.; Shirbeigi, E. Hesperidin supplementation modulates inflammatory responses following myocardial infarction. J. Am. Coll. Nutr. 2015, 34, 205–211. [Google Scholar] [CrossRef]
  71. Rizza, S.; Muniyappa, R.; Iantorno, M.; Kim, J.A.; Chen, H.; Pullikotil, P.; Senese, N.; Tesauro, M.; Lauro, D.; Cardillo, C.; et al. Citrus polyphenol hesperidin stimulates production of nitric oxide in endothelial cells while improving endothelial function and reducing inflammatory markers in patients with metabolic syndrome. J. Clin. Endocrinol. Metab. 2011, 96, E782–E792. [Google Scholar] [CrossRef]
  72. Milenkovic, D.; Deval, C.; Dubray, C.; Mazur, A.; Morand, C. Hesperidin displays relevant role in the nutrigenomic effect of orange juice on blood leukocytes in human volunteers: A randomized controlled cross-over study. PLoS ONE 2011, 6, e26669. [Google Scholar] [CrossRef] [Green Version]
  73. Miwa, Y.; Yamada, M.; Sunayama, T.; Mitsuzumi, H.; Tsuzaki, Y.; Chaen, H.; Mishima, Y.; Kibata, M. Effects of glucosyl hesperidin on serum lipids in hyperlipidemic subjects: Preferential reduction in elevated serum triglyceride level. J. Nutr. Sci. Vitaminol. 2004, 50, 211–218. [Google Scholar] [CrossRef] [Green Version]
  74. Miwa, Y.; Mitsuzumi, H.; Sunayama, T.; Yamada, M.; Okada, K.; Kubota, M.; Chaen, H.; Mishima, Y.; Kibata, M. Glucosyl hesperidin lowers serum triglyceride level in hypertriglyceridemic subjects through the improvement of very low-density lipoprotein metabolic abnormality. J. Nutr. Sci. Vitaminol. 2005, 51, 460–470. [Google Scholar] [CrossRef] [Green Version]
  75. Yari, Z.; Movahedian, M.; Imani, H.; Alavian, S.M.; Hedayati, M.; Hekmatdoost, A. The effect of hesperidin supplementation on metabolic profiles in patients with metabolic syndrome: A randomized, double-blind, placebo-controlled clinical trial. Eur. J. Nutr. 2020, 59, 2569–2577. [Google Scholar] [CrossRef]
  76. Hong, Y.; An, Z. Hesperidin attenuates learning and memory deficits in APP/PS1 mice through activation of Akt/Nrf2 signaling and inhibition of RAGE/NF-κB signaling. Arch. Pharmacal. Res. 2018, 41, 655–663. [Google Scholar] [CrossRef]
  77. Youdim, K.A.; Dobbie, M.S.; Kuhnle, G.; Proteggente, A.R.; Abbott, N.J.; Rice-Evans, C. Interaction between flavonoids and the blood-brain barrier: In vitro studies. J. Neurochem. 2003, 85, 180–192. [Google Scholar] [CrossRef]
  78. Papandreou, D.; Magriplis, E.; Abboud, M.; Taha, Z.; Karavolia, E.; Karavolias, C.; Zampelas, A. Consumption of Raw Orange, 100% Fresh Orange Juice, and Nectar- Sweetened Orange Juice-Effects on Blood Glucose and Insulin Levels on Healthy Subjects. Nutrients 2019, 11, 2171. [Google Scholar] [CrossRef] [Green Version]
  79. Sthijns, M.; van Blitterswijk, C.A.; LaPointe, V.L.S. Redox regulation in regenerative medicine and tissue engineering: The paradox of oxygen. J. Tissue Eng. Regen. Med. 2018, 12, 2013–2020. [Google Scholar] [CrossRef] [Green Version]
  80. Takumi, H.; Nakamura, H.; Simizu, T.; Harada, R.; Kometani, T.; Nadamoto, T.; Mukai, R.; Murota, K.; Kawai, Y.; Terao, J. Bioavailability of orally administered water-dispersible hesperetin and its effect on peripheral vasodilatation in human subjects: Implication of endothelial functions of plasma conjugated metabolites. Food Funct. 2012, 3, 389–398. [Google Scholar] [CrossRef]
  81. Liu, L.; Xu, D.-m.; Cheng, Y.-y. Distinct Effects of Naringenin and Hesperetin on Nitric Oxide Production from Endothelial Cells. J. Agric. Food Chem. 2008, 56, 824–829. [Google Scholar] [CrossRef]
  82. Chiou, C.-S.; Lin, J.-W.; Kao, P.-F.; Liu, J.-C.; Cheng, T.-H.; Chan, P. Effects of hesperidin on cyclic strain-induced endothelin-1 release in human umbilical vein endothelial cells. Clin. Exp. Pharmacol. Physiol. 2008, 35, 938–943. [Google Scholar] [CrossRef]
  83. Chanet, A.; Milenkovic, D.; Claude, S.; Maier, J.A.; Kamran Khan, M.; Rakotomanomana, N.; Shinkaruk, S.; Bérard, A.M.; Bennetau-Pelissero, C.; Mazur, A.; et al. Flavanone metabolites decrease monocyte adhesion to TNF-α-activated endothelial cells by modulating expression of atherosclerosis-related genes. Br. J. Nutr. 2013, 110, 587–598. [Google Scholar] [CrossRef] [Green Version]
  84. Nizamutdinova, I.T.; Jeong, J.J.; Xu, G.H.; Lee, S.H.; Kang, S.S.; Kim, Y.S.; Chang, K.C.; Kim, H.J. Hesperidin, hesperidin methyl chalone and phellopterin from Poncirus trifoliata (Rutaceae) differentially regulate the expression of adhesion molecules in tumor necrosis factor-alpha-stimulated human umbilical vein endothelial cells. Int. Immunopharmacol. 2008, 8, 670–678. [Google Scholar] [CrossRef]
  85. Cybulsky, M.I.; Iiyama, K.; Li, H.; Zhu, S.; Chen, M.; Iiyama, M.; Davis, V.; Gutierrez-Ramos, J.C.; Connelly, P.W.; Milstone, D.S. A major role for VCAM-1, but not ICAM-1, in early atherosclerosis. J. Clin. Investig. 2001, 107, 1255–1262. [Google Scholar] [CrossRef] [Green Version]
  86. Maneesai, P.; Bunbupha, S.; Potue, P.; Berkban, T.; Kukongviriyapan, U.; Kukongviriyapan, V.; Prachaney, P.; Pakdeechote, P. Hesperidin Prevents Nitric Oxide Deficiency-Induced Cardiovascular Remodeling in Rats via Suppressing TGF-β1 and MMPs Protein Expression. Nutrients 2018, 10, 1549. [Google Scholar] [CrossRef] [Green Version]
  87. Yamamoto, M.; Jokura, H.; Hashizume, K.; Ominami, H.; Shibuya, Y.; Suzuki, A.; Hase, T.; Shimotoyodome, A. Hesperidin metabolite hesperetin-7-O-glucuronide, but not hesperetin-3’-O-glucuronide, exerts hypotensive, vasodilatory, and anti-inflammatory activities. Food Funct. 2013, 4, 1346–1351. [Google Scholar] [CrossRef]
  88. Thijssen, D.H.J.; Bruno, R.M.; van Mil, A.; Holder, S.M.; Faita, F.; Greyling, A.; Zock, P.L.; Taddei, S.; Deanfield, J.E.; Luscher, T.; et al. Expert consensus and evidence-based recommendations for the assessment of flow-mediated dilation in humans. Eur. Heart J. 2019, 40, 2534–2547. [Google Scholar] [CrossRef]
  89. Ras, R.T.; Streppel, M.T.; Draijer, R.; Zock, P.L. Flow-mediated dilation and cardiovascular risk prediction: A systematic review with meta-analysis. Int. J. Cardiol. 2013, 168, 344–351. [Google Scholar] [CrossRef]
  90. Sultan, S.; Gosling, M.; Nagase, H.; Powell, J.T. Shear stress-induced shedding of soluble intercellular adhesion molecule-1 from saphenous vein endothelium. FEBS Lett. 2004, 564, 161–165. [Google Scholar] [CrossRef] [Green Version]
  91. Videm, V.; Albrigtsen, M. Soluble ICAM-1 and VCAM-1 as markers of endothelial activation. Scand. J. Immunol. 2008, 67, 523–531. [Google Scholar] [CrossRef]
  92. Jublanc, C.; Beaudeux, J.L.; Aubart, F.; Raphael, M.; Chadarevian, R.; Chapman, M.J.; Bonnefont-Rousselot, D.; Bruckert, E. Serum levels of adhesion molecules ICAM-1 and VCAM-1 and tissue inhibitor of metalloproteinases, TIMP-1, are elevated in patients with autoimmune thyroid disorders: Relevance to vascular inflammation. Nutr. Metab. Cardiovasc. Dis. 2011, 21, 817–822. [Google Scholar] [CrossRef] [PubMed]
  93. Konukoglu, D.; Uzun, H. Endothelial Dysfunction and Hypertension. Adv. Exp. Med. Biol. 2017, 956, 511–540. [Google Scholar] [CrossRef] [PubMed]
  94. Morand, C.; Dubray, C.; Milenkovic, D.; Lioger, D.; Martin, J.F.; Scalbert, A.; Mazur, A. Hesperidin contributes to the vascular protective effects of orange juice: A randomized crossover study in healthy volunteers. Am. J. Clin. Nutr. 2011, 93, 73–80. [Google Scholar] [CrossRef] [PubMed]
  95. Valls, R.M.; Pedret, A.; Calderón-Pérez, L.; Llauradó, E.; Pla-Pagà, L.; Companys, J.; Moragas, A.; Martín-Luján, F.; Ortega, Y.; Giralt, M.; et al. Hesperidin in orange juice improves human endothelial function in subjects with elevated blood pressure and stage 1 hypertension: A randomized, controlled trial (Citrus study). J. Funct. Foods 2021, 85, 104646. [Google Scholar] [CrossRef]
  96. Schär, M.Y.; Curtis, P.J.; Hazim, S.; Ostertag, L.M.; Kay, C.D.; Potter, J.F.; Cassidy, A. Orange juice–derived flavanone and phenolic metabolites do not acutely affect cardiovascular risk biomarkers: A randomized, placebo-controlled, crossover trial in men at moderate risk of cardiovascular disease. Am. J. Clin. Nutr. 2015, 101, 931–938. [Google Scholar] [CrossRef] [Green Version]
  97. Buscemi, S.; Rosafio, G.; Arcoleo, G.; Mattina, A.; Canino, B.; Montana, M.; Verga, S.; Rini, G. Effects of red orange juice intake on endothelial function and inflammatory markers in adult subjects with increased cardiovascular risk. Am. J. Clin. Nutr. 2012, 95, 1089–1095. [Google Scholar] [CrossRef]
  98. Salden, B.N.; Troost, F.J.; de Groot, E.; Stevens, Y.R.; Garcés-Rimón, M.; Possemiers, S.; Winkens, B.; Masclee, A.A. Randomized clinical trial on the efficacy of hesperidin 2S on validated cardiovascular biomarkers in healthy overweight individuals. Am. J. Clin. Nutr. 2016, 104, 1523–1533. [Google Scholar] [CrossRef] [Green Version]
  99. Kalpana, K.B.; Srinivasan, M.; Menon, V.P. Evaluation of antioxidant activity of hesperidin and its protective effect on H2O2 induced oxidative damage on pBR322 DNA and RBC cellular membrane. Mol. Cell. Biochem. 2009, 323, 21–29. [Google Scholar] [CrossRef]
  100. Kim, J.Y.; Jung, K.J.; Choi, J.S.; Chung, H.Y. Hesperetin: A Potent Antioxidant Against Peroxynitrite. Free Radic. Res. 2004, 38, 761–769. [Google Scholar] [CrossRef]
  101. Chen, M.; Gu, H.; Ye, Y.; Lin, B.; Sun, L.; Deng, W.; Zhang, J.; Liu, J. Protective effects of hesperidin against oxidative stress of tert-butyl hydroperoxide in human hepatocytes. Food Chem. Toxicol. 2010, 48, 2980–2987. [Google Scholar] [CrossRef]
  102. Yang, H.L.; Chen, S.C.; Senthil Kumar, K.J.; Yu, K.N.; Lee Chao, P.D.; Tsai, S.Y.; Hou, Y.C.; Hseu, Y.C. Antioxidant and anti-inflammatory potential of hesperetin metabolites obtained from hesperetin-administered rat serum: An ex vivo approach. J. Agric. Food Chem. 2012, 60, 522–532. [Google Scholar] [CrossRef]
  103. Sthijns, M.; Schiffers, P.M.; Janssen, G.M.; Lemmens, K.J.A.; Ides, B.; Vangrieken, P.; Bouwman, F.G.; Mariman, E.C.; Pader, I.; Arnér, E.S.J.; et al. Rutin protects against H2O2-triggered impaired relaxation of placental arterioles and induces Nrf2-mediated adaptation in Human Umbilical Vein Endothelial Cells exposed to oxidative stress. Biochim. Biophys. Acta Gen. Subj. 2017, 1861, 1177–1189. [Google Scholar] [CrossRef]
  104. Estruel-Amades, S.; Massot-Cladera, M.; Garcia-Cerdà, P.; Pérez-Cano, F.J.; Franch, À.; Castell, M.; Camps-Bossacoma, M. Protective Effect of Hesperidin on the Oxidative Stress Induced by an Exhausting Exercise in Intensively Trained Rats. Nutrients 2019, 11, 783. [Google Scholar] [CrossRef] [Green Version]
  105. El-Sayed, E.-S.M.; Abo-Salem, O.M.; Abd-Ellah, M.F.; Abd-Alla, G.M. Hesperidin, an antioxidant flavonoid, prevents acrylonitrile-induced oxidative stress in rat brain. J. Biochem. Mol. Toxicol. 2008, 22, 268–273. [Google Scholar] [CrossRef]
  106. Sahu, B.D.; Kuncha, M.; Sindhura, G.J.; Sistla, R. Hesperidin attenuates cisplatin-induced acute renal injury by decreasing oxidative stress, inflammation and DNA damage. Phytomedicine 2013, 20, 453–460. [Google Scholar] [CrossRef]
  107. Martínez-Noguera, F.J.; Marín-Pagán, C.; Carlos-Vivas, J.; Rubio-Arias, J.A.; Alcaraz, P.E. Acute Effects of Hesperidin in Oxidant/Antioxidant State Markers and Performance in Amateur Cyclists. Nutrients 2019, 11, 1898. [Google Scholar] [CrossRef] [Green Version]
  108. Boussetta, N.; Abedelmalek, S.; Khouloud, A.; Ben Anes, A.; Souissi, N. Does red orange juice supplementation has a protective effect on performance, cardiovascular parameters, muscle damage and oxidative stress markers following the Yo-Yo Intermittent Recovery Test Level-1 under polluted air? Int. J. Environ. Health Res. 2020, 30, 630–642. [Google Scholar] [CrossRef]
  109. Shen, C.-Y.; Lin, J.-J.; Jiang, J.-G.; Wang, T.-X.; Zhu, W. Potential roles of dietary flavonoids from Citrus aurantium L. var. amara Engl. in atherosclerosis development. Food Funct. 2020, 11, 561–571. [Google Scholar] [CrossRef]
  110. Sakata, K.; Hirose, Y.; Qiao, Z.; Tanaka, T.; Mori, H. Inhibition of inducible isoforms of cyclooxygenase and nitric oxide synthase by flavonoid hesperidin in mouse macrophage cell line. Cancer Lett. 2003, 199, 139–145. [Google Scholar] [CrossRef]
  111. Kazłowska, K.; Hsu, T.; Hou, C.-C.; Yang, W.-C.; Tsai, G.-J. Anti-inflammatory properties of phenolic compounds and crude extract from Porphyra dentata. J. Ethnopharmacol. 2010, 128, 123–130. [Google Scholar] [CrossRef]
  112. Kawaguchi, K.; Kikuchi, S.-i.; Hasunuma, R.; Maruyama, H.; Yoshikawa, T.; Kumazawa, Y. A Citrus Flavonoid Hesperidin Suppresses Infection-Induced Endotoxin Shock in Mice. Biol. Pharm. Bull. 2004, 27, 679–683. [Google Scholar] [CrossRef] [Green Version]
  113. Kometani, T.; Fukuda, T.; Kakuma, T.; Kawaguchi, K.; Tamura, W.; Kumazawa, Y.; Nagata, K. Effects of alpha-glucosylhesperidin, a bioactive food material, on collagen-induced arthritis in mice and rheumatoid arthritis in humans. Immunopharmacol. Immunotoxicol. 2008, 30, 117–134. [Google Scholar] [CrossRef]
  114. Hill, A.V.; Lupton, H. Muscular Exercise, Lactic Acid, and the Supply and Utilization of Oxygen. QJM Int. J. Med. 1923, os-16, 135–171. [Google Scholar] [CrossRef]
  115. Overdevest, E.; Wouters, J.A.; Wolfs, K.H.M.; van Leeuwen, J.J.M.; Possemiers, S. Citrus Flavonoid Supplementation Improves Exercise Performance in Trained Athletes. J. Sports Sci. Med. 2018, 17, 24–30. [Google Scholar]
  116. Martínez-Noguera, F.J.; Marín-Pagán, C.; Carlos-Vivas, J.; Alcaraz, P.E. Effects of 8 Weeks of 2S-Hesperidin Supplementation on Performance in Amateur Cyclists. Nutrients 2020, 12, 3911. [Google Scholar] [CrossRef]
  117. Van Iersel, L.E.; Stevens, Y.R.; Conchillo, J.M.; Troost, F.J. The effect of citrus flavonoid extract supplementation on anaerobic capacity in moderately trained athletes: A randomized controlled trial. J. Int. Soc. Sports Nutr. 2021, 18, 2. [Google Scholar] [CrossRef]
  118. Baranauskas, M.N.; Coggan, A.R.; Gruber, A.H.; Altherr, C.A.; Raglin, J.S.; Carter, S.J. Dietary Nitrate Supplementation and Exercise-Related Performance. Nutr. Today 2020, 55, 211–217. [Google Scholar] [CrossRef]
  119. Bailey, S.J.; Fulford, J.; Vanhatalo, A.; Winyard, P.G.; Blackwell, J.R.; DiMenna, F.J.; Wilkerson, D.P.; Benjamin, N.; Jones, A.M. Dietary nitrate supplementation enhances muscle contractile efficiency during knee-extensor exercise in humans. J. Appl. Physiol. 2010, 109, 135–148. [Google Scholar] [CrossRef] [Green Version]
  120. Hart, J.D.; Dulhunty, A.F. Nitric oxide activates or inhibits skeletal muscle ryanodine receptors depending on its concentration, membrane potential and ligand binding. J. Membr. Biol. 2000, 173, 227–236. [Google Scholar] [CrossRef]
  121. Viner, R.I.; Williams, T.D.; Schöneich, C. Nitric oxide-dependent modification of the sarcoplasmic reticulum Ca-ATPase: Localization of cysteine target sites. Free Radic. Biol. Med. 2000, 29, 489–496. [Google Scholar] [CrossRef]
  122. Layec, G.; Blain, G.M.; Rossman, M.J.; Park, S.Y.; Hart, C.R.; Trinity, J.D.; Gifford, J.R.; Sidhu, S.K.; Weavil, J.C.; Hureau, T.J.; et al. Acute High-Intensity Exercise Impairs Skeletal Muscle Respiratory Capacity. Med. Sci. Sports Exerc. 2018, 50, 2409–2417. [Google Scholar] [CrossRef] [PubMed]
  123. Lewis, M.T.; Blain, G.M.; Hart, C.R.; Layec, G.; Rossman, M.J.; Park, S.Y.; Trinity, J.D.; Gifford, J.R.; Sidhu, S.K.; Weavil, J.C.; et al. Acute high-intensity exercise and skeletal muscle mitochondrial respiratory function: Role of metabolic perturbation. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2021, 321, R687–R698. [Google Scholar] [CrossRef] [PubMed]
  124. Versari, D.; Daghini, E.; Virdis, A.; Ghiadoni, L.; Taddei, S. Endothelium-dependent contractions and endothelial dysfunction in human hypertension. Br. J. Pharm. 2009, 157, 527–536. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Parhiz, H.; Roohbakhsh, A.; Soltani, F.; Rezaee, R.; Iranshahi, M. Antioxidant and anti-inflammatory properties of the citrus flavonoids hesperidin and hesperetin: An updated review of their molecular mechanisms and experimental models. Phytotherapy Res. 2015, 29, 323–331. [Google Scholar] [CrossRef]
  126. Jacobs, H.; Moalin, M.; Bast, A.; van der Vijgh, W.J.; Haenen, G.R. An essential difference between the flavonoids monoHER and quercetin in their interplay with the endogenous antioxidant network. PLoS ONE 2010, 5, e13880. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Schematic summary of the potential mechanism of action for the hesperidin effect on endothelial function during exercise. (A) During exercise, the release of nitric oxide (NO) by endothelial cells causes the relaxation of the smooth muscle cells, which leads to the dilation of an artery and an increase in blood flow. (B) Hesperidin increases the endothelial cells’ NO production. This process leads to higher artery dilation, which further improves blood flow. During exercise, improved skeletal muscle perfusion and the consequent increase in oxygen (O2) efflux to the muscle can improve endurance performance. The figure was created with BioRender.com. Abbreviations: HES = hesperidin; NO = nitric oxide; O2 = oxygen; SKM = skeletal muscle; Increased: ↑ < ↑↑; Decreased ↓ < ↓↓.
Figure 1. Schematic summary of the potential mechanism of action for the hesperidin effect on endothelial function during exercise. (A) During exercise, the release of nitric oxide (NO) by endothelial cells causes the relaxation of the smooth muscle cells, which leads to the dilation of an artery and an increase in blood flow. (B) Hesperidin increases the endothelial cells’ NO production. This process leads to higher artery dilation, which further improves blood flow. During exercise, improved skeletal muscle perfusion and the consequent increase in oxygen (O2) efflux to the muscle can improve endurance performance. The figure was created with BioRender.com. Abbreviations: HES = hesperidin; NO = nitric oxide; O2 = oxygen; SKM = skeletal muscle; Increased: ↑ < ↑↑; Decreased ↓ < ↓↓.
Nutrients 14 02955 g001
Figure 2. Schematic summary of potential mechanism of action for the hesperidin effect on exercise-induced oxidative stress. (A) Contractile activity of skeletal muscle tissue leading to a higher oxygen demand could induce an increased formation of ROS as a result of the excessive mitochondrial activity leading to incomplete oxidative phosphorylation during exercise. In athletes performing extreme endurance exercise, the constant rise in ROS production could lead to damage to DNA, lipids (lipid peroxidation), or protein and attenuation in muscle contraction. (B) Increased blood flow (and thereby increased shear stress) during exercise leads to increased endothelial ROS production, which reacts with NO. Increased ROS production by the endothelium leads to decreased NO availability. (C) Hesperidin, acting as an antioxidant, helps to prevent the side effects of excessive ROS formation in the muscle cells. Moreover, hesperidin increases endogenous antioxidant enzymes. These two mechanisms combined help prevent cell damage and the decline in muscle contraction signalling pathways leading to stimulation in force production. (D) When supplemented with hesperidin, endothelial ROS production will be decreased, preventing the decrease in NO production caused by shear stress. The figure was created with BioRender.com. Abbreviations: HES = hesperidin; NO = nitric oxide; NOX = NADPH oxidase; O2 = oxygen; ROS = reactive oxygen species; Increased: ↑; Decreased: ↓.
Figure 2. Schematic summary of potential mechanism of action for the hesperidin effect on exercise-induced oxidative stress. (A) Contractile activity of skeletal muscle tissue leading to a higher oxygen demand could induce an increased formation of ROS as a result of the excessive mitochondrial activity leading to incomplete oxidative phosphorylation during exercise. In athletes performing extreme endurance exercise, the constant rise in ROS production could lead to damage to DNA, lipids (lipid peroxidation), or protein and attenuation in muscle contraction. (B) Increased blood flow (and thereby increased shear stress) during exercise leads to increased endothelial ROS production, which reacts with NO. Increased ROS production by the endothelium leads to decreased NO availability. (C) Hesperidin, acting as an antioxidant, helps to prevent the side effects of excessive ROS formation in the muscle cells. Moreover, hesperidin increases endogenous antioxidant enzymes. These two mechanisms combined help prevent cell damage and the decline in muscle contraction signalling pathways leading to stimulation in force production. (D) When supplemented with hesperidin, endothelial ROS production will be decreased, preventing the decrease in NO production caused by shear stress. The figure was created with BioRender.com. Abbreviations: HES = hesperidin; NO = nitric oxide; NOX = NADPH oxidase; O2 = oxygen; ROS = reactive oxygen species; Increased: ↑; Decreased: ↓.
Nutrients 14 02955 g002
Figure 3. Schematic summary of potential mechanism of action for the hesperidin effect on exercise-induced inflammation. (A) Exhaustive exercise leads to macrophage activation, which activates an acute inflammatory response characterized by increases in circulatory and intramuscular inflammatory markers such as C-reactive protein (CRP), cytokines (tumour necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), and interleukin-1beta (IL-1β). Without adequate post-exercise/competition recovery periods, an excessive inflammatory response could lead to impaired muscle contractions and force generation. (B) Hesperidin shows the potential to inhibit macrophage activation and recruitment and decrease markers of exercise-induced inflammation, potentially speeding up the recovery process and, therefore, improving exercise performance. The figure was created with BioRender.com. Abbreviations: CRP = C-reactive protein; HES = hesperidin; IL-1β = interleukin-1beta; IL-6 = interleukin-6; TNF-α = tumour necrosis factor-alpha; Increased: ↑; Decreased: ↓.
Figure 3. Schematic summary of potential mechanism of action for the hesperidin effect on exercise-induced inflammation. (A) Exhaustive exercise leads to macrophage activation, which activates an acute inflammatory response characterized by increases in circulatory and intramuscular inflammatory markers such as C-reactive protein (CRP), cytokines (tumour necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), and interleukin-1beta (IL-1β). Without adequate post-exercise/competition recovery periods, an excessive inflammatory response could lead to impaired muscle contractions and force generation. (B) Hesperidin shows the potential to inhibit macrophage activation and recruitment and decrease markers of exercise-induced inflammation, potentially speeding up the recovery process and, therefore, improving exercise performance. The figure was created with BioRender.com. Abbreviations: CRP = C-reactive protein; HES = hesperidin; IL-1β = interleukin-1beta; IL-6 = interleukin-6; TNF-α = tumour necrosis factor-alpha; Increased: ↑; Decreased: ↓.
Nutrients 14 02955 g003
Table 1. Studies investigating the effects of hesperidin, hesperetin, and their metabolites on endothelial function markers in studies performed in vitro.
Table 1. Studies investigating the effects of hesperidin, hesperetin, and their metabolites on endothelial function markers in studies performed in vitro.
Author,
Year,
Country
Cell TypeTreatment
Characteristics
Treatment
Duration
Endothelial Function Outcomes
(Hesperidin or Hesperetin vs. Control)
Rizza et al.[71]
2011
Italy
BAECHesperetin
0.01 μM, 0.1 μM, 1 μM, 10 μM
10 min↑pAMPK protein levels (1 μM, 10 μM)
↑pAkt protein levels (1 μM, 10 μM)
↑p-eNOS protein levels (1 μM, 10 μM)
=pAMPK, pAkt and p-eNOS protein levels (0.01 μM, 0.1 μM)
1 h↑NO production (10 μΜ)
=NO production (0.01 μM, 0.1 μM, 1 μM)
↓TNF-α-stimulated VCAM-1 protein levels (10 μM)
↓TNF-α-stimulated monocyte adhesion (10 μM)
Takumi et al.[80]
2012
Japan
HUVECsHesperetin, HPT7G
25 µM, 50 µM
24 h↑Release of NO, in a dose-dependent manner
Liu et al.[81]
2008
China
HUVECsHesperetin
12.5 μM, 25 μM, 50 μM, 100 μM
24 h↑Release of NO in a dose-dependent manner
↑eNOS mRNA expression (50 μM)
↑eNOS protein levels (50 μM)
Chiou et al.[82]
2008
Taiwan
HUVECsHesperidin
1 µM, 10 µM, 100 µM
30 min prior to strain treatment (computer-controlled application of sinusoidal negative pressure)↓strain-induced ET-1 secretion (10 µM, 100 µM)
=strain-induced ET-1 secretion (1 µM)
30 min↑NO production (100 µM)
↑eNOS phosphorylation (100 µM)
↑Akt phosphorylation (100 µM)
60 min↑NO production (10 µM, 100 µM)
=NO production (1 µM)
↑NOS activity (10 µM, 100 µM)
=NOS activity (1 µM)
↑eNOS phosphorylation (100 µM)
=Akt phosphorylation (100 µM)
Chanet et al.[83]
2013
France
HUVECsHesperetin, HPT3′G, HPT3′S, HPT7G
2 μM
24 h↓TNF-α-stimulated monocyte adhesion
Nizamutdinova et al.[84]
2008
Korea
HUVECsHesperidin, hesperidin methyl chalone
1 µM, 5 µM, 10 µM, 50 µM
24 h↓TNF-α-stimulated VCAM-1 protein expression (5 µM, 10 µM, 50 µM)
=TNF-α-stimulated VCAM-1 protein expression (1 µM)
=TNF-α-stimulated ICAM-1 protein expression (1 µM, 5 µM, 10 µM, 50 µM)
↓TNF-α-stimulated monocyte adhesion (5 µM, 10 µM, 50 µM)
↓TNF-α-stimulated monocyte adhesion (1 µM)
↑: statistically significant increase; ↓: statistically significant decrease; = no significant change. Abbreviations: BAEC = bovine aortic endothelial cells; HUVECs = human umbilical vein endothelial cells; (p)AMPK = (phosphorylated) 5′AMP-activated protein kinase; (p)Akt = (phosphorylated) protein kinase B; (p)-Enos = (phosphorylated) endothelial nitric oxide synthase; NO = nitric oxide; TNF-α = tumour necrosis factor-α; VCAM-1 = vascular cell adhesion molecule 1; ICAM-1 = intracellular adhesion molecule 1; HPT7G = hesperetin-7-O-glucuronide; HPTG’3 = hesperetin-3′-O-glucuronide; HPT′3S = hesperetin-3′-O-sulphate; ET-1 = endothelin-1.
Table 2. Studies investigating the effects of hesperidin, hesperetin, or metabolites on endothelial function markers in animal studies.
Table 2. Studies investigating the effects of hesperidin, hesperetin, or metabolites on endothelial function markers in animal studies.
Author,
Year,
Country
Sample
Characteristics
Intervention
Characteristics
Intervention
Duration
Endothelial Function Outcomes
(Hesperidin or Hesperetin vs. Control Groups)
Maneesai et al.[86]
2018
Thailand
Male Sprague–Dawley rats with hypertension
(treated with L-NAME)
Hesperidin,
15 mg/kg/day and 30 mg/kg/day
5 weeks↓SBP, DBP
↑plasma NOx
Yamamoto et al.[87]
2013
Japan
Male SHRsHesperetin, HPT7G, HPT3′G
5 mg/kg
3 min↓SBP (hesperetin, HPT7G)
=SBP (HPT3′G)
=DBP (hesperetin, HPT7G, HPT3′G)
Yamamoto et al.[87]
2013
Japan
Thoracic aortic rings from SHRs and WKY ratsHPT7G
HPT3′G
100 µM
20 minSHRs:
↑ACh-induced endothelium-dependent vasodilation (HPT7G)
=Ach-induced endothelium-dependent vasodilation (HPT3′G)
=SNP-induced endothelium-independent vasodilation (HPT7G, HPT3′G)

WKY:
=ACh-induced endothelium-dependent vasodilation (HPT7G, HPT3′G)
=SNP-induced endothelium-independent vasodilation (HPT7G, HPT3′G)
↑: statistically significant increase; ↓: statistically significant decrease; = no significant change. Abbreviations: L-NAME = Nω-nitro L-arginine methyl ester; SBP = systolic blood pressure; DBP = diastolic blood pressure; Nox = nitric oxide metabolites; SHRs = spontaneously hypertensive rats; HPT7G = hesperetin-7-O-glucuronide; HPTG’3 = hesperetin-3′-O-glucuronide; WKY = Wistar Kyoto; Ach = acetylcholine; SNP = sodium nitroprusside.
Table 3. Studies investigating the effects of hesperidin on endothelial function markers in human studies.
Table 3. Studies investigating the effects of hesperidin on endothelial function markers in human studies.
Author,
Year,
Country
Sample
Characteristics
(Study Design)
Intervention
Characteristics
Intervention
Duration
Endothelial Function Outcomes
(Hesperidin vs. Control Groups)
Morand et al.[94]
2011
France
n = 24 healthy males
Age = 56 (1) y
BMI = 27.4 (0.3) kg/m2
(RCT)
292 mg hesperidin/dayAcute (6 h before test)microvascular reactivity
Chronic (4 weeks)↓DBP
=sICAM-1
=sVCAM-1

=NOx, trend for improvement
Valls et al.[95]
2021
Spain
n = 159 subjects with pre- or stage 1 hypertension
Age = 19–67 y
BMI = 18.5–40.5 kg/m2
(RTC)
600 mg hesperidin/dayAcute (6 h before test)↑IRH
Chronic (12 weeks)↑IRH
Takumi et al.[80]
2011
Japan
n = 10 healthy female subjects
Age = 18–22 y
(RTC)
17 mg or 170 mg hesperidinAcute (test within 70 min after intake)↓drop in blood flow
Comment: while subjects stayed in an air-conditioned room; significant drop in both INT dosages
Schar et al.[96]
2015
UK
n = 16 men at moderate CVD riskAge = 60.6 (8.4) y
BMI = 25.6 (0.8) kg/m2
(RCT)
320 mg hesperidinAcute (5 h before test)=P-selectin expression
= BP
=Cardiac BRS
Buscemi et al.[97]
2012
Italy
n = 21 with increased cardiovascular risk
Age = 19–67 y
BMI = 18.5–40.5 kg/m2
(RCT)
159.5 mg/day hesperidinChronic (7 days)↑FMD
Rizza et al.[71]
2011
Italy
n = 24 with MetS
Age = 52 (2)
BMI = 34.7 (1.5) kg/m2
(RCT)
500 mg/day hesperidinChronic (3 weeks)↑FMD
=VCAM-1
Salden et al.[98]
2016
The Netherlands
n = 48 subjects with baseline FMD ≥3%
Age = 53 (14) y
BMI = 29 (2.6) kg/m2
(RTC)
450 mg/day hesperidinChronic (6 weeks)↑FMD
↓sVCAM-1
↓sICAM-1
Yari et al.[75]
2020
Iran
n = 49 subjects
with MetSAge = 45.1 (11.1) y
BMI = 31.3 (4.9) kg/m2
(RCT)
1 g/day hesperidinChronic (12 weeks)↓SBP
↑: statistically significant increase; ↓: statistically significant decrease; = no significant change; data are presented as mean ± SD or as a range. Abbreviations: Aus = arbitrary units (log); BMI = body max index (kg/m2); BRS = baroreflex sensitivity; BP = blood pressure; CON = control; DBP = diastolic blood pressure; FDM = flow-mediated dilation; INT = intervention; IRH = ischaemic reactive hyperaemia; MetS = metabolic syndrome; Nox = nitric oxide metabolites; RCT = randomized controlled trial; SBP = systolic blood pressure; sICAM-1 = soluble intercellular adhesion molecule 1; sVCAM-1 = soluble vascular cell adhesion molecule 1.
Table 4. Studies investigating the effects of hesperidin, hesperetin and their metabolites on oxidative stress markers in studies performed in vitro.
Table 4. Studies investigating the effects of hesperidin, hesperetin and their metabolites on oxidative stress markers in studies performed in vitro.
Author,
Year,
Country
Cell TypeRadical Scavenging Activity AssayTreatment
Characteristics
Treatment
Duration
Oxidative Stress Outcomes
(Hesperidin or Hesperetin vs. Control)
Kalpana et al. 2009
[99]
India
Human erythrocytes·OH, ·O2, ·NO and ABTS•+ radical scavenging activity assayHesperidin,
0.5 mM, 1 mM, 1.5 mM, 2 mM, 2.5 mM

Hesperidin
0.5 mM, 1 mM, 1.5 mM, 2 mM, 2.5 mM
Assay-dependent=free radical scavenging activity compared to ascorbic acid and trolox, in a dose-dependent manner
30 min↓H2O2-induced TBARS production, in a dose-dependent manner
Kim et al.[100]
2004
South Korea
YPEN-1 prostatic endothelial cellsONOO, ·O2, ·NO scavenging activity assayHesperetin
5 µM, 15 µM, 50 µM, 200 µM
2 h=ONOO and ·O2 scavenging activity compared to penicillamine and Trolox, respectively
↓·NO scavenging activity compared to carboxy-PTIO

t-BHP-induced intracellular ROS generation in a dose-dependent manner
Chiou et al.[82]
2008
Taiwan
HUVECs Hesperidin,
1 µM, 10 µM, 100 µM
1 h exposure in the presence of strain treatment (computer-controlled application of sinusoidal negative pressure)= strain-increased ROS formation (1 µM)
↓strain-increased ROS formation (10 µM, 100 µM)
Chen et al.[101]
2010
China
L02 hepatic cells Hesperidin
20 µM, 40 µM, 80 µM
24 h=t-BHP-induced intracellular ROS levels (20 µM)
t-BHP-induced intracellular ROS levels (40 µM, 80 µM)

=t-BHP-induced MDA production (20 µM)↓t-BHP-induced MDA production (40 µM, 80 µM)
Yang et al.[102]
2012
Taiwan
Macrophage RAW264.7 cells and fibroblast A7r5 cells Hesperetin,
Hesperetin metabolites extracted from rat serum
1 μM, 5 μM, 10 μM
60 min for RAW264.7 cells
5 min for A7r5 cells
↓LPS-induced intracellular ROS level (1 μM, 5 μM, 10 μM)

Hesperetin metabolites showed greater antioxidant potential compared to hesperetin
↓: statistically significant decrease; = no significant change; Abbreviations: HUVECs = human umbilical vein endothelial cells; ROS = reactive oxygen species; ABTS = 2,2′-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid; H2O2 = hydrogen peroxide; TBARS = thiobarbituric acid-reactive substances; ·OH = hydroxyl radical; ONOO = peroxynitrite;·O2 = superoxide anion;·NO = nitric oxide; t-BHP = tert-butylhydroperoxide; MDA = malondialdehyde.
Table 5. Studies investigating the effects of hesperidin on oxidative stress markers in animal studies.
Table 5. Studies investigating the effects of hesperidin on oxidative stress markers in animal studies.
Author,
Year,
Country
Sample
Characteristics
Intervention
Characteristics
Intervention DurationOxidative Stress Outcomes
(Hesperidin vs. Control Groups)
Estruel-Amades et al.[104]
2019
Spain
Groups of Female Wister rats:

Sedentary rats (SED)

5-week-trained rats (T)

5-week-trained rats undergoing an additional exhaustion test (TE)
200 mg/kg of hesperidin three times per week5 weeks↓ROS production by peritoneal macrophages induced by the exhaustion test

In thymus tissue:
=CAT activity in all groupsHesperidin prevented the ↓ in SOD activity induced by the exhaustion test
↓SOD activity in SED group

In spleen tissue:
Hesperidin prevented the ↓ in CAT activity induced by the exhaustion test
↓SOD activity in SED and TE groups
=SOD activity in T group

In liver tissue:
Hesperidin prevented the ↓ in CAT activity induced by the exhaustion test
↓CAT activity in SED group
= CAT activity in T and TE groups
↓SOD activity all groups
↓GPx activity in SED and TE groups
=GPx activity in T group
El-Sayed et al.[105]
2008
Egypt
Brain tissue from male Swiss albino ratsHesperidin 200 mg/kg/day28 days=MDA content
↓Acrylonitrile-induced increase in MDA content

=GSH, GST content
↑SOD, GPx levels
↓CAT levels
↑Acrylonitrile-induced decrease in GSH, SOD, CAT, GPx, GST levels
Sahu et al.[106]
2013
India
Kidney tissue from male Wistar ratsHesperidin
100 mg/kg/day, 200 mg/kg/day
10 days=ROS levels
↓cisplatin-induced increase in ROS (100, 200 mg/kg/day)

=TBARS levels
↓cisplatin-induced increase in TBARS (100, 200 mg/kg/day)

=SOD, GSH, CAT, GPx, GR, GST activity
=cisplatin-induced decrease in GSH, CAT, GPx, GR activity (100 mg/kg/day)
↑cisplatin-induced decrease in SOD, GST activity (100 mg/kg/day)
↑cisplatin-induced decrease in SOD, GSH, CAT, GPx, GR, GST activity (200 mg/kg/day)
Maneesai et al.[86]
2018
Thailand
Male Sprague–Dawley rats with hypertension
(treated with L-NAME)
Hesperidin
15 mg/kg/day, 30 mg/kg/day
5 weeks↓vascular superoxide production (15, 30 mg/kg/day)

↓plasma MDA (15, 30 mg/kg/day)
↑: statistically significant increase; ↓: statistically significant decrease; = no significant change; Abbreviations: L-NAME = Nω-nitro L-arginine methyl ester; MDA = malondialdehyde; ROS = reactive oxygen species; CAT = catalase; SOD = superoxide dismutase; GPx = glutathione peroxidase; GSH = glutathione; GST = glutathione S-transferase; TBARS = thiobarbituric acid-reactive substances; GR = glutathione reductase.
Table 6. Studies investigating the effects of hesperidin on oxidative stress following physical exercise.
Table 6. Studies investigating the effects of hesperidin on oxidative stress following physical exercise.
Author,
Year,
Country
Sample
Characteristics
(Study Design)
Intervention
Characteristics
Intervention
Duration
Exercise TestExercise-Induced Oxidative Stress Outcomes
(Hesperidin vs. Control Groups)
Martínez-Noguera et al.[107]
2019
Spain
n = 15 male amateur cyclists
Age = 18–55 y, BMI = 19–25.5 kg/m2
(RCT)
500 mg hesperidinAcute
(5 h before exercise)
Repeated sprints test (Wingate test)=TBARS
↑CAT
=SOD
=GSH
Boussetta et al.[108]
2019
Tunisia
n = 11 healthy soccer players
Age = 22.4 ± 0.5
BMI = 23.2 ± 0.4 kg/m2
(RCT)
INT: 217 mg hesperidin
CON: placebo
Acute (2.5 h before the test)Yo-Yo Intermittent Recovery Test (YYIRT)=TAS
↓MDA
↑: statistically significant increase; ↓: statistically significant decrease; = no significant change; Abbreviations: BMI = body max index; CAT = catalase; CON = control; GSH = glutathione; INT = intervention; MDA = malondialdehyde; MET = metabolic equivalent; RCT = randomized controlled trial; SOD = Superoxide dismutase; TAS = total antioxidant status; TBARS = thiobarbituric acid-reactive substances; Data are presented as mean ± SD or as a range.
Table 7. Studies investigating the effects of hesperidin and hesperetin on inflammatory markers in studies performed in vitro.
Table 7. Studies investigating the effects of hesperidin and hesperetin on inflammatory markers in studies performed in vitro.
Author,
Year,
Country
Cell TypeTreatment
Characteristics
Treatment DurationInflammatory Outcomes
(Hesperidin or Hesperetin vs. Control)
Shen et al.[109]
2019
China
Macrophage RAW264.7 cellsHPT7G
3.13, 6.25, 12.5, 25, 50, 100 and 200 μg/mL
24 h
12 h (for measurement of mRNA expression)
=LPS-induced NO production (3.13, 6.25 µg/mL)
↓LPS-induced NO production (12.5, 25, 50 µg/mL)

↓LPS-induced IL-6 production (50, 100, 200 µg/mL)
=LPS-induced IL-6 mRNA expression (50 µg/mL)
↓LPS-induced IL-6 mRNA expression (100, 200 µg/mL)

↓LPS-induced IL-1β production (50, 100, 200 µg/mL)
↓LPS-induced IL-1β mRNA expression (50, 100, 200 µg/mL)

=LPS-induced TNF-α production (50, 100, 200 µg/mL)
=LPS-induced TNF-α mRNA expression (100, 200 µg/mL)
↓LPS-induced TNF-α mRNA expression (50 µg/mL)

=LPS-induced COX-2 mRNA expression (50 µg/mL)
↓LPS-induced COX-2 mRNA expression (100, 200 µg/mL)
Yang et al.[102]
2012
Taiwan
Macrophage RAW264.7 cells and fibroblast A7r5 cellsHesperetin,
Hesperetin metabolites extracted from rat serum
1 μM, 5 μM, 10 μM
18 h exposure for RAW264.7 cells
8 h exposure for A7r5 cells
↓LPS-induced PGE2 production (1 μM, 5 μM, 10 μM in both cell types)
↓LPS-induced COX-2 protein levels (1 μM, 5 μM, 10 μM in both cell types)

↓LPS-induced NO production (1 μM, 5 μM, 10 μM in RAW264.7 cells)
=LPS-induced NO production (1 μM, 5 μM, 10 μM in A7r5 cells)
↓iNOS protein levels ((1 μM, 5 μM, 10 μM in both cell types)
↓LPS-induced NF-κB transcriptional activation (1 μM, 5 μM, 10 μM in RAW264.7 cells)

Hesperetin metabolites showed greater anti-inflammatory potential compared to hesperetin
Sakata et al. 2003
[110]
Japan
Macrophage RAW264.7 cellsHesperidin
10 μM, 20 μM, 30 μM
30 min=LPS-induced PGE2 production (10 μM)
↓LPS-induced PGE2 production (20 μM, 30 μM)

=LPS-induced COX-2 protein level ((10 μM, 20 μM, 30 μM))

↓LPS-induced NO2 production (10 μM, 20 μM, 30 μM)
↓LPS-induced iNOS protein level (10 μM, 20 μM, 30 μM)
Kazlowska et al.[111]
2010
Taiwan
Macrophage RAW264.7 cellsHesperidin
5 μg/mL, 15 μg/mL, 80 μg/mL, 125 μg/mL, 150 μg/mL 250 μg/mL
24 h=LPS-induced NO production (5 μg/mL)
↓LPS-induced NO production (15 μg/mL, 125 μg/mL, 250 μg/mL)

=LPS-induced iNOS promoter activity (80 μg/mL, 150 μg/mL, 250 μg/mL)
=LPS-induced NF-κB activity (80 μg/mL, 150 μg/mL, 250 μg/mL)
↓: statistically significant decrease; = no significant change; Abbreviations: HPT7G = hesperetin-7-O-glucopyranoside; LPS = lipopolysaccharides; NO = nitric oxide; IL-6 = interleukin-6; IL-1β = interleukin-1beta; TNF-α = tumour necrosis factor-alpha; COX-2 = cyclo-oxygenase 2; PGE2 = prostaglandin E2; NO2 = nitrogen dioxide; iNOS = nitric oxide synthase; NF-κB = nuclear factor kappa-light-chain-enhancer of activated B cells.
Table 8. Studies investigating the effects of hesperidin on inflammatory markers in animal studies.
Table 8. Studies investigating the effects of hesperidin on inflammatory markers in animal studies.
Author,
Year,
Country
Sample
Characteristics
Intervention
Characteristics
Intervention DurationInflammatory Outcomes
(Hesperidin vs. Control Groups)
Kawaguchi et al.[112]
2004
Japan
Female BALB/c and C57L/6 miceHesperidin,
0.1 mg, 0.3 mg, 1 mg, 3 mg/mouse
3 h before LPS treatment↓LPS-induced increase in plasma TNF-α (0.3 mg, 1 mg, 3 mg/mouse)
=LPS-induced increase in plasma TNF-α (0.1 mg/mouse)
Sahu et al.[106]
2013
India
Male Wistar ratsHesperidin
100 mg/kg/day, 200 mg/kg/day
10 days=renal TNF-α (200 mg/kg/day)
=cisplatin-induced increase in renal TNF-α (100 mg/kg/day)
↓cisplatin-induced increase in renal TNF-α (200 mg/kg/day)

=renal myeloperoxidase (200 mg/kg/day)
↓cisplatin-induced increase in renal myeloperoxidase (100, 200 mg/kg/day)
Maneesai et al.[86]
2018
Thailand
Male Sprague–Dawley rats with hypertension (treated with L-NAME)Hesperidin
15 mg/kg/day and 30 mg/kg/day
5 weeks↓plasma TNF-α (15, 30 mg/kg/day)
↓: statistically significant decrease; = no significant change; Abbreviations: LPS = lipopolysaccharides; TNF-α = tumour necrosis factor-alpha; L-NAME = Nω-nitro L-arginine methyl ester.
Table 9. Studies investigating the effects of hesperidin on inflammatory markers in human studies.
Table 9. Studies investigating the effects of hesperidin on inflammatory markers in human studies.
Author,
Year,
Country
Subject
Characteristics
(Study Design)
Intervention
Characteristics
Intervention
Duration
Inflammatory Outcomes
(Hesperidin vs. Control Groups)
Buscemi et al.[97]
2012
Italy
n = 21 subjects with increased cardiovascular risk
Age = 19–67 y
BMI = 18.5–40.5 kg/m2
(RCT)
159.5 mg/day hesperidin7 days↓hs-CRP
↓IL-6
↓TNF-α
Yari et al.[75]
2020
Iran
n = 49 subjects
with MetS
Age = 45.1 ± 11.1 y
BMI = 31.3 ± 4.9 kg/m2
(RCT)
1 g/day hesperidin12 weeks↓TNF-α
=hs-CRP
Kometani et al.[113]
2008
Japan
n = 19 subjects
with arthritis
Age = 26–49 y
(RCT)
3 g/day hesperidin12 weeks↓CRP
Morand et al.[94]
2011
France
n = 24 healthy males
Age = 56 ± 1 y
BMI = 27.4 ± 0.3 kg/m2
(RCT)
292 mg/day hesperidin4 weeks=CRP
=IL-6
↓: statistically significant decrease; = no significant change; Abbreviations: BMI = body max index (kg/m2); CON = control; CRP = C-reactive protein hs-CRP = high-sensitivity C-reactive protein; IL-6 = interleukin-6; INT = intervention; MetS = metabolic syndrome; RCT = randomized controlled trial; TNF-α = tumour necrosis factor-alpha. Data are presented as mean ± SD or as a range.
Table 10. Studies investigating the effects of hesperidin on exercise performance outcomes in animal studies.
Table 10. Studies investigating the effects of hesperidin on exercise performance outcomes in animal studies.
Author,
Year,
Country
Sample
Characteristics
(Study Design)
Intervention
Characteristics
Intervention
Duration
Exercise TestExercise Performance Outcomes
(Hesperidin vs. Control Groups)
Estruel-Amades et al.[104]
2019
Spain
Female Wistar rats200 mg/kg of hesperidin three times per weekChronic (5 weeks)Maximum distance run until exhaustion test (2 times per week for 5 weeks)↑ maximum distance during all performed tests (week 1–5)
↑: statistically significant increase.
Table 11. Studies investigating the effects of hesperidin on exercise performance outcomes in human studies.
Table 11. Studies investigating the effects of hesperidin on exercise performance outcomes in human studies.
Author,
Year,
Country
Sample Characteristics
(Study Design)
Intervention
Characteristics
Intervention
Duration
Exercise TestExercise Performance Outcomes
(Hesperidin vs. Control Groups)
Martínez-Noguera et al.[107]
2019
Spain
n = 15 male amateur cyclists
Age = 18–55 y, BMI = 19–25.5 kg/m2
(RCT)
500 mg hesperidinAcute
(5 h before exercise)
Repeated sprints test (Wingate test)↑Average power
↑Maximal speed
↑Total energy
Boussetta et al.[108]
2019
Tunisia
n = 11 healthy soccer players
Age = 22.4 ± 0.5 y
BMI = 23.2 ± 0.4 kg/m2
(RCT)
217 mg hesperidinAcute 2.5 h before the test)Yo-Yo intermittent recovery test (YYIRT)=VO2max (increasing trend)
=PRE
Overdevest et al.[115]
2018
The Netherlands
n = 39 trained males Age = 18–25 y
BMI = 22.1 (0.30) kg/m2
(RCT)
500 mg/day citrus fruit extract (450 mg hesperidin/day)Chronic (4 weeks)10 min time-trial on a cycle ergometer↑Δ Power
↓VO2/Power ratio
= Es VO2max
Martínez-Noguera et al.[116]
2020
Spain
n = 40 male amateur cyclists
Age = 18–55 y, BMI = 19–25.5 kg/m2
(RCT)
500 mg/day hesperidinChronic (8 weeks)Repeated sprints test (Wingate test)↑Absolute peak power
↑Relative peak power
Incremental test until exhaustion↑ Maximum power
↑ Estimated FTP
Van Iersel et al.[117]
2021
The Netherlands
n = 92 moderately trained healthy subjects
Age = 24 ± 5 y
BMI = 22.4 ± 2.2 kg/m2
(RCT)
360 mg or 450 mg hesperidinChronic (4 and 8 weeks)Wingate anaerobic test↑Average power (360 mg after 4 weeks)
↑Average power (360 mg after 8 weeks)
↑Average power (450 mg after 4 weeks)
↑5 s Peak power (360 mg after 4 weeks)
↑: statistically significant increase; ↓: statistically significant decrease; = no significant change; Abbreviations: BMI = body max index; CON = control; Es VO2max = Estimated VO2max; FTP = functional threshold power; INT = intervention; MET = metabolic equivalent; RCT = randomized controlled trial; RPE = Ratings of Perceived Exertion; VO2max = maximal oxygen uptake; Data are presented as mean ± SD or as a range.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Imperatrice, M.; Cuijpers, I.; Troost, F.J.; Sthijns, M.M.J.P.E. Hesperidin Functions as an Ergogenic Aid by Increasing Endothelial Function and Decreasing Exercise-Induced Oxidative Stress and Inflammation, Thereby Contributing to Improved Exercise Performance. Nutrients 2022, 14, 2955. https://doi.org/10.3390/nu14142955

AMA Style

Imperatrice M, Cuijpers I, Troost FJ, Sthijns MMJPE. Hesperidin Functions as an Ergogenic Aid by Increasing Endothelial Function and Decreasing Exercise-Induced Oxidative Stress and Inflammation, Thereby Contributing to Improved Exercise Performance. Nutrients. 2022; 14(14):2955. https://doi.org/10.3390/nu14142955

Chicago/Turabian Style

Imperatrice, Maria, Iris Cuijpers, Freddy J. Troost, and Mireille M. J. P. E. Sthijns. 2022. "Hesperidin Functions as an Ergogenic Aid by Increasing Endothelial Function and Decreasing Exercise-Induced Oxidative Stress and Inflammation, Thereby Contributing to Improved Exercise Performance" Nutrients 14, no. 14: 2955. https://doi.org/10.3390/nu14142955

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