Next Article in Journal
Hericium coralloides Ameliorates Alzheimer’s Disease Pathologies and Cognitive Disorders by Activating Nrf2 Signaling and Regulating Gut Microbiota
Previous Article in Journal
The Association of Human Milk Proportion with the Clinical Outcomes of Necrotizing Enterocolitis in Preterm Infants: A Retrospective Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Targeting Metabolic Syndrome in Hidradenitis Suppurativa by Phytochemicals as a Potential Complementary Therapeutic Strategy

1
Psoriasis Research and Treatment Center, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
2
Interdisciplinary Group of Molecular Immunopathology, Dermatology/Medical Immunology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
3
Inflammation and Regeneration of Skin, BIH Center for Regenerative Therapies, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 13353 Berlin, Germany
*
Author to whom correspondence should be addressed.
Nutrients 2023, 15(17), 3797; https://doi.org/10.3390/nu15173797
Submission received: 14 July 2023 / Revised: 9 August 2023 / Accepted: 21 August 2023 / Published: 30 August 2023
(This article belongs to the Section Phytochemicals and Human Health)

Abstract

:
Hidradenitis suppurativa (HS) is a chronic inflammatory disease characterized by the appearance of painful inflamed nodules, abscesses, and pus-draining sinus tracts in the intertriginous skin of the groins, buttocks, and perianal and axillary regions. Despite its high prevalence of ~0.4–1%, therapeutic options for HS are still limited. Over the past 10 years, it has become clear that HS is a systemic disease, associated with various comorbidities, including metabolic syndrome (MetS) and its sequelae. Accordingly, the life expectancy of HS patients is significantly reduced. MetS, in particular, obesity, can support sustained inflammation and thereby exacerbate skin manifestations and the chronification of HS. However, MetS actually lacks necessary attention in HS therapy, underlining the high medical need for novel therapeutic options. This review directs attention towards the relevance of MetS in HS and evaluates the potential of phytomedical drug candidates to alleviate its components. It starts by describing key facts about HS, the specifics of metabolic alterations in HS patients, and mechanisms by which obesity may exacerbate HS skin alterations. Then, the results from the preclinical studies with phytochemicals on MetS parameters are evaluated and the outcomes of respective randomized controlled clinical trials in healthy people and patients without HS are presented.

1. Hidradenitis Suppurativa

Hidradenitis suppurativa (HS) is a chronic inflammatory disease affecting the intertriginous skin, particularly at the axillary, inguinal, gluteal, and perianal sites [1]. Painful inflamed nodules, abscesses, and pus-draining sinus tracts recur in these areas of the skin. In addition, destructive skin remodeling processes in the course of the disease lead to scars that restrict movement (Figure 1). This debilitating disease usually starts in early adulthood and shows an estimated worldwide prevalence of about 0.4–1% [1,2,3,4,5,6].
After the onset of the first symptoms, the diagnosis of HS still takes about 10 years on average, a fact that is important because the disease duration correlates with the number of comorbidities of respective patients [7]. Despite the regional differences, men appear to be as equally affected as women when viewed globally [8,9,10,11,12,13]. Given the great physical and mental burden of the disease, it is not surprising that HS patients have been found to show a considerable reduction in quality of life parameters, and this reduction is even more pronounced compared to other chronic inflammatory skin diseases, including psoriasis or atopic dermatitis [14]. Anxiety, depression, body image impairment, and passive forms of indirect self-destructiveness together with stigmatization and social exclusion or self-isolation are additional aspects frequently associated with HS [15,16,17,18,19,20]. Furthermore, owing to a reduced employment rate and an increased absenteeism and presenteeism, HS leads to a significant loss of national gross value added and, therefore, is of great socio-economic importance [21].
Known predisposing factors for HS include genetic as well as lifestyle factors [22]. Among the lifestyle factors, obesity and smoking, frequently met in HS patients, were linked to the development of skin alterations. However, the mechanism of lesion development is still not fully understood, especially the initial events triggering the disease, which are still unclear. It is assumed that obesity supports a subclinical inflammatory milieu around the hair follicle in apocrine gland-bearing intertriginous skin [22]. In the early stage of HS, epidermal hyperplasia, including acanthosis and hyperkeratosis, leads to infundibular alterations promoting follicular occlusion, whereby secreted inflammatory mediators (e.g., cytokines) from infiltrated mononuclear immune cells may account for this process. Nicotine might contribute to these alterations by promoting epidermal hyperplasia and altering the skin microbiome [23,24,25]. Resulting retention of sebum within the hair follicle then leads to its dilatation, propagation of bacteria, and inflammation [26]. Thus, bacterial components and alarmins released from damaged follicular cells are sensed by local immune cells through pattern recognition receptors, provoking high immune cell infiltration and the formation of inflamed nodules and abscesses [22,26,27]. The continuous cross-talk of cutaneous tissue cells with those activated immune cells, in particular, macrophages, T cells, B/plasma cells, and neutrophilic granulocytes, results in the secretion of further pro-inflammatory cytokines and matrix-degrading enzymes (matrix metalloproteinases), which drive skin destruction and allow for the formation of pus-draining sinus tracts in the chronic stage of HS [27,28,29,30,31,32,33,34,35,36,37]. HS lesions also contain high levels of the anti-inflammatory cytokine interleukin (IL)-10 [26,38]. Interestingly, the long-term effects of bacterial products on monocytes and macrophages are very similar to the effects of IL-10 on these cells [39]. Finally, extensive scarring can develop as the result of the ongoing tissue-remodeling processes. Immune mediators produced locally in HS lesions can enter the circulatory system and act in other organs, promoting the occurrence of comorbidities [27,32,34].

2. Metabolic Alterations in HS

An important clinical aspect associated with HS is the presence of profound metabolic alterations of those affected, including central obesity, hypertriglyceridemia, hypo-high-density lipoprotein (HDL) cholesterolemia, hyperglycemia, and hypertension [40]. When three of these criteria are met, the diagnosis of metabolic syndrome (MetS) can be confirmed. The number of fulfilled MetS criteria typically increases with age, whereas this observation does not apply to HS patients. In fact, already in early life (≤34 years of age), 40% of HS patients are shown to be affected by the MetS compared to 0% in age-matched controls [40]. In addition to MetS, HS is associated with numerous additional comorbidities, including spondyloarthritis, inflammatory bowel disease, non-alcoholic fatty liver disease (NAFLD), and cardiovascular disease [41,42,43,44,45,46,47,48].
Among MetS criteria, central obesity is the most frequent found in ~65% of HS patients (compared to 24% in the healthy controls) and is assumed to play a pathogenetic role in HS [40]. Adipose tissue is able to adapt to times of varying nutrient availability through releasing free fatty acids (FFAs) from stored triglycerides during times of nutrient shortage and through the storing of triglycerides in times of caloric excess [49]. If the physiological storage is at its limit, adipose hyperplasia (increase in cell numbers) occurs. This is associated with decreased blood perfusion, local immune cell activation, apoptosis, and enhanced mechanical stress due to the tightness of cells within the adipose tissue. In contrast to the so-called metabolically healthy obesity (MHO), where the majority of adipose tissue is located in subcutaneous depots, metabolically unhealthy obesity (MUHO) leads to central obesity (visceral adiposity), with triglycerides predominantly deposited in ectopic sites, including visceral adipose tissue or inner organs (e.g., the liver, skeletal muscle, and heart) [49].

3. Proinflammatory Mechanisms of Obesity

Obesity might affect HS in four different ways: at the physical, microbial, immunological, and metabolic levels [22]. First, obesity leads to enlarged skin folds that may support lesion development through continued wetness, maceration, increased mechanical friction, and injury. Second, resulting anaerobic conditions in those skin folds in turn provide the basis for the altered microbiome pattern observed in HS patients. Third, hypertrophic adipose tissue mediates low-grade systemic inflammation through pro-inflammatory mediators (e.g., cytokines and chemokines) secreted by immune cells within the adipose tissue and induces oxidative stress, both of which worsen the skin condition as well as HS comorbidities [50,51]. In comparison to normal-weight individuals, hypertrophic adipose tissue contains increased numbers of neutrophils and macrophages, known to play an important role in the development of low-grade systemic inflammation [52,53]. From the in vivo models, it can be deduced that neutrophils, infiltrated into the hypertrophic adipose tissue in the early stage of obesity, mediate the recruitment and polarization of macrophages through the increased secretion of proinflammatory as well as macrophage recruiting chemotactic mediators (e.g., IL-6, tumor necrosis factor (TNF)-α, CCL2) [54,55]. Furthermore, the tight cross-talk of neutrophils and adipocytes was suggested to mediate NLRP3 inflammasome activation, the inflammation of adipose tissue, and biasing of neutrophils towards a hyperinflammatory state [56,57]. Fourth, adipose tissue is not only an energy reservoir, but also a major endocrine tissue [49,58]. In HS, the pattern of adipokines, peptide hormones derived from adipose tissue that regulate metabolic processes, e.g., insulin sensitivity (regulated by adiponectin) and body weight homeostasis (regulated by leptin) are dysregulated [59,60,61]. In fact, the serum level of anti-inflammatory adiponectin clearly decreases, whereas leptin levels increase in HS compared to healthy donors, indicating the presence of a leptin resistance, which further promotes obesity [59,60]. As immune cells are also directly targeted by adipokines, the altered adipokine pattern might contribute to the development of a pathogenetic immune–metabolic circuit in HS patients [58,62]. Interestingly, the metabolism of blood CD4+ T cells appears to also be altered in HS patients. In fact, it was recently found that the expression of several genes involved in oxidative phosphorylation was downregulated in the blood CD4+ T cells of HS patients and a few transcripts for glycolysis-dependent energy production were increased [63].
It should be noted that obesity is associated with the enhanced systemic level of FFAs [49]. FFAs negatively impact glucose and lipid metabolism, being risk factors for developing insulin resistance and dyslipidemia [49]. Dyslipidemia and hyperglycemia, in turn, are risk factors for developing cardiovascular diseases [49,64,65,66]. Of note, FFAs may also promote inflammation by binding to TLR4 on monocytic immune cells and induce NLRP3-dependent IL-1β production [67,68,69]. As FFAs are released from visceral depots directly into the portal circulation, FFAs also affect liver homeostasis and promote the development of NAFLD [49]. Accordingly, higher prevalences of hyperglycemia, dyslipidemia, cardiovascular alterations, and NAFLD in HS patients compared to the controls was reported [40,41,43,44,46]. In fact, ~26% of HS patients were found to suffer from hyperglycemia compared to 8% in the healthy controls, and the incidence of diabetes increased at least two-fold in HS patients [40,70]. Aspects of dyslipidemia are found in 50% (hypo-HDL cholesterolemia) and 38.8% (hypertriglyceridemia) of HS patients, compared to 18% and 22%, respectively, in the healthy controls [40]. Furthermore, ~70% of HS patients compared to 30% of the controls were also affected by NAFLD [43,46]. The described mitigating impact of bariatric surgery and weight loss on HS severity supports the concept of the significant contribution of adipose tissue to the cutaneous inflammation in HS patients [71,72]. Furthermore, the relevance of inflammation to MetS is supported by the observation that anti-inflammatory therapy targeting TNF-α may improve MetS severity in patients with rheumatoid arthritis [73]. However, the respective data for HS patients are lacking at present. Overall, the strongly increased presence of metabolic alterations and its sequelae in HS patients are serious risk factors contributing to the substantially reduced life expectancy [74]. In fact, HS patients lived an average of 14.7 years less than the controls, with cardiovascular disease being their leading cause of death [74].

4. HS Therapy: Time for a New Perspective?

Therapeutic options for moderate to severe HS include, at present, long-term antibiotic treatment and the surgical excision of skin lesions, whereby these therapies do not result in a sustained improvement of the disease-associated reduced quality of life of patients [14,75]. Furthermore, at present, there are only two approved immune therapies for HS:, the TNF-α-neutralizing antibody adalimumab and the IL-17A-neutralizing antibody secukinumab. In contrast to psoriasis, only a proportion of HS patients benefits from these biologicals. The limited treatment options and consideration that a relevant portion of HS patients refuse single-therapy elements or have relevant contraindications demonstrate the great need for novel and innovative therapeutics for HS treatment [1,76]. The inclusion of MetS comorbidities in therapy concepts, in particular, obesity, as a relevant trigger factor for HS symptoms is also still insufficient. Based on the increased prevalence of mood disorders among HS patients, lifestyle changes are also difficult to realize for those patients. To close this gap, phytotherapy appears to be an appropriate complementary therapeutic approach by targeting MetS elements. In fact, there is a positive perception of alternative therapeutics among both HS patients and dermatologists [77,78]. In this review, we evaluate the potential impact of selected phytomedical drugs on MetS parameters. In this way, we hope to identify candidates that can be tested in future studies on HS patients, applied in daily practice, and complement HS therapy in the long term.

5. Phytotherapy—A Therapeutic Concept with a Long History

The traditional use of herbal plant-based medicine has a very long tradition that dates back several thousand years [79]. The first descriptions of the use of herbal medicine was found on Sumerian clay slabs from Mesopotamia (~3000 BC) and Egyptian papyrus rolls (~1550 BC) [79]. Medicinal plants are also mentioned in the Bible, another old scripture. In the traditional medicine of ancient Greece, the use of locally growing herbs for medical purposes also played an important role and was proposed by Hippocrates (460–377 BC) [79]. The Shen Nong Ben Cao Jing is another early written record (date of origin unclear: 25–220 AD) describing a variety of medical plants and their therapeutic uses according to traditional Chinese medicine [80]. Later, the monastic medicine (i.a. represented by Hildegard von Bingen, 1098–1179) led to the widespread distribution of herb knowledge among the local population. In the 16th century, Paracelsus (1493–1541) laid the foundation for the concept of spagyric medicine, a term derived from the Greek words “spien” (separate) and “agera” (unite) [81]. Two-hundred years later, Carl von Linne finally developed a binary nomenclature for plants that brought the needed system into the plant kingdom [82]. The term phytotherapy was coined by the French physician Henri Leclerc (1870–1955) and comprises the topical application or internal medical administration of plants or herbs. These include their use in the native or processed form as decoctions, extract preparations, or isolated key substances. To date, the European Medicines Agency (EMA) database of the Committee on Herbal Medicinal Products (HMPC) already lists 167 completed monographs for phytomedical plants (https://www.ema.europa.eu/en/medicines, accessed on 7 August 2023).

6. Potential Phytotherapeutic Options for MetS in HS Patients

Targeting metabolic alterations by phytochemicals might be a complementary therapeutic strategy of HS. Especially, their anti-inflammatory, antioxidative, glucose, lipid metabolism regulating, and their described cardio- and hepatoprotective properties are potentially interesting in this regard [83,84]. For this review, phytochemicals were selected within the field of phytotherapy according to the availability of the preclinical and clinical data on a single metabolic syndrome parameter. Accordingly, Olea europea, Withania somnifera, Vitis vinifera, and Camellia sinensis were found to represent appropriate candidates for the indication of MetS (Figure 2). As the effects of phytochemicals on primary and tumor cells are different [85,86], the preclinical data based on primary cells were primarily evaluated in this review. Furthermore, regarding the available human in vivo data, only placebo-controlled, blinded, randomized clinical trials (RCTs) were presented in this review.

6.1. Olea europea

As an important agricultural plant, different parts of the olive tree are used for nutritional and medical purposes, the leaves, olive fruits, and olive oil also being elements of the so-called Mediterranean diet. The main active constituents of olive oil fruits as well as of olive leaf extract (OLE) are the polyphenols oleuropein (secoiridoid) and hydroxytyrosol (phenylethanoid), which is also generated through the metabolization of oleuropein. Several preclinical in vitro and in vivo studies investigating the mode of action of these substances implied the beneficial effect on metabolic dysfunction. In fact, oleuropein and hydroxytyrosol were shown to prevent LDL oxidation and strengthen endogenous antioxidative and arteroprotective mechanisms, reducing endoplasmic reticulum stress and platelet aggregation in vitro [87,88,89,90,91,92,93,94].
In vivo, Olea europea leaf-derived phytochemicals improved dyslipidemia, adipokine profile, glucose homeostasis, and antioxidative capacity in several diabetes, oxidative stress, and obesity animal models [91,95,96,97,98,99,100,101,102,103,104]. In line with these studies, these phytochemicals ameliorated high-fat-diet-induced body weight increase and white adipose tissue hypertrophy in vivo in rodent models [96,100,102,103,104]. The reported enhancing effect of hydroxytyrosol and oleuropein on adipocyte lipolysis in vitro might therefore contribute to their normalizing effect on lipid metabolism in vivo [105,106,107]. The data from Vezza et al. and Wang et al. indicate the normalization of obesity-related dysbiosis and the downregulation of inflammatory cytokines as another mechanism underlying the positive impact of the MetS parameter on high-fed diet-induced murine obesity [100,101]. Moreover, an increase in the systemic adiponectin level and upregulation of MAPK, as well as the suppression of PPARg expression in adipose tissue were suggested by Hadrich et al. and Scoditti et al. to underly the anti-obesity effects of olive leaf phytochemicals [96,108].
Furthermore, an improvement of cardiovascular parameters by hydroxytyrosol using an in vivo diabetes rat model was demonstrated [109]. A cardioprotective role was also suggested for olive leaf phytochemicals using in vivo animal models of high-fat-diet-induced metabolic syndrome, diabetes, arteriosclerosis, and ischemia [104,110,111,112,113,114,115,116]. The underlying mechanism of cardioprotection might involve nitric oxide-mediated vasodilatation and oxidative stress reduction [104,115,117]. Moreover, according to the in vitro data, olive leaf phytochemicals were observed to show anticoagulative properties in healthy and experimentally induced ischemic rodents [87,89,111,118].
For the described hepatoprotective effect of oleuropein and hydroxytyrosol in high-fat-diet-based in vivo rodent models, an attribution to the normalization of hepatic PPARg, Nrf2, and NF-kB pathway activity was suggested [101,102,104,119,120].
The available RCTs on the evaluation of olive leaf extract (OLE), oleuropein, or hydroxytyrosol indicated an attenuating effect on the parameters of MetS, confirming in part the preclinical study data (Table 1). As the individual contribution of containing fatty acids and polyphenols of olive oil to the observed effects in respective RCTs as challenging, only RCTs using OLE, oleuropein, or hydroxytyrosol were considered for the evaluation and were discussed here. RTCs evaluating the potential of OLE on glucose metabolism did not present a consistent picture yet hinted at some beneficial effects contributing to the normalization of glucose homeostasis. In fact, OLEs were found to reduce the postprandial plasma glucose level of healthy, obese, pre-hypertensive, or osteoporosis participants after single or long-term applications in 3 of 4 RCTs evaluating this outcome measure [121,122,123,124]. However, fasting glucose levels were not affected by long-term OLE applications [125,126,127]. A short-term treatment of healthy participants with oleuropein followed by glucose tolerance testing in the absence of oleuropein also did not influence the post-prandial blood glucose level [128]. In contrast, insulin sensitivity and pancreatic β-cell function were improved in obese participants by OLE [122]. However, in obese or hypertensive participants, insulin levels postprandially decreased [122] or remained unchanged [125,127] after long-term OLE treatment; an increase was assessed after a single application in healthy study cohorts [121,124]. Based on the increase of the hormone GLP-1 that supports insulin secretion with a concurrent reduction in its inhibitor DPP-4, an antidiabetic property was suggested for OLE by Carnevale et al. [121]. In contrast to the preclinical studies, the data from respective RCTs regarding the effects of OLE on the lipid profile were not consistent. In two out of four studies evaluating lipid parameters, an improvement of dyslipidemia parameters, including a reduction in total cholesterol (CH), low-density lipoprotein (LDL), and triglyceride (TG) levels after long-term applications, were reported [126,129]. Whether Olea europea phytochemicals might be beneficial in body weight management is not yet clear. The data on the antioxidative capacity of Olea europea phytochemicals from RCTs are sparse. Only one RCT investigated this parameter and found a decrease in postprandial oxidative stress in healthy participants after a single oleuropein application [121]. Regarding the cardiovascular measures, no clear influence of OLE on the blood pressure parameter was found, whereas a slight reduction in systolic and diastolic blood pressure levels was reported by Lockyer et al. after a 6-week OLE application in pre-hypertensive participants; no influences on blood pressure was described by Stevens et al. and de Bock et al. after 8- or 12-week OLE treatments, respectively [122,126,127]. A head-to-head study investigating the effect of OLE and captopril on blood pressure in hypertensive participants revealed a comparable effectivity for both substances [130]. However, the lack of a placebo group and the use of only low-dose captopril were certainly limitations of the study. The impact of OLE treatment on vascular function was also not clear as the short- and long-term studies showed inconsistent data [126,131].

6.2. Withania somnifera

The winter cherry is a common plant predominantly found in Mediterranean regions, with a long history of use in ayurvedic medicine. Among withanolides, secondary phytochemicals present in the root of withania somnifera, withaferin A (steroidal lactone), are the most studied component. Data obtained from the preclinical studies evaluating the effects of withaferin A in vitro and in vivo suggest the antidiabetic, anti-obesity, anti-oxidative, and anti-inflammatory potential of this substance.
In vitro, withaferin A caused an improvement of glucose metabolism, enhanced insulin secretion by pancreatic β-cells, and mediated the protection of pancreatic islet cells against inflammatory cytokine-induced cell death [132,133]. Moreover, the inhibition of adipogenesis by withaferin A was also observed in vitro [134]. Furthermore, in a palmitic acid-induced oxidative stress in vitro model, withaferin A inhibited ROS and inflammatory cytokine production, whereas it restored the impaired insulin signaling and NO production in endothelial cells [135].
In line with the in vitro data, withaferin A was also described to show antidiabetic activity in vivo. In fact, an improvement of insulin resistance, glucose metabolism, and adiponectin level was observed using respective in vivo murine obesity and diabetes models [134,136,137,138]. The suggested underlying mechanisms included the regulation of genes involved in the insulin and PPARγ pathway [134].
Furthermore, withaferin A ameliorated body and adipose tissue weight gain and improved the lipid profile in various murine obesity models [134,136,137,139,140,141]. In line with these observations, withaferin A was identified to act as a leptin sensitizer and inhibit the food restriction-based reduction in basic energy expenditure in obese mice [141]. Furthermore, withaferin A-induced browning of white adipose tissue accompanied by enhanced mitochondrial activity observed in high-fat-diet-fed mice might contribute to its anti-obesity effects [139,140,142]. Accordingly, sympathetic denervation reduced withaferin A-mediated white adipose tissue browning and a decrease in obesity indicated the important role of the sympathetic nerve/adipose axis involving PRDM16 and FATP1 [139].
Furthermore, the data from in vivo rat models of hypertension, ischemia reperfusion injury, and cardiac toxicity suggest the cardioprotective properties of Withania somnifera phytochemicals [143,144,145,146,147]. An enhanced oxidative stress reduction was suggested as one mechanism underlying these findings. The amelioration of hepatic steatosis and normalization of liver enzymes, hepatic inflammatory markers (IL-6, TNF-α, IL-1β, CRP, MCP-1, COX2), endogenous antioxidant system molecules, and regulated enzymes involved in lipid and glucose metabolism in vivo using a high-fat-diet-induced murine obesity model also implied the hepatoprotective potential of withaferin A [134,136,137]. The withaferin A-dependent improvement of steatohepatitis in leptin-signaling-deficient ob/ob mice thereby suggested leptin-independent mechanisms for hepatoprotection [148]. In a further study of murine diet-induced obesity, the hepatoprotective effect of withaferin A was suggested to be related to the direct activation of liver X receptor a/farnesoid X receptor (LXRα/FXR) [137]. Additionally, the data from a murine liver toxicity model reveals that withaferin A is able to reduce liver injury in vivo [149]. This effect was suggested to be attributed to the induction of Nrf2 and genes of the antioxidative glutathion system [149]. The therapeutic hepatoprotective potential of withaferin A was also shown using an in vivo hepatitis model that revealed the effective attenuation of D-galactosamine/LPS-induced liver damage by this phytochemical [150]. The authors suggested the limitation of macrophage NLRP3 activation and IL-1β secretion as possible mechanisms of action of withaferin A in this model [150].
To date, there is limited data on the three available placebo-controlled RCTs on the influence of withania somnifera root extract (WSRE) on the features of metabolic syndrome (Table 2). In chronic, stressed, overweight participants, the application of WSRE provoked a reduction in food craving and perceived stress scores as well as serum cortisol level, whereas the assessed happiness score increased [151]. In line with these data, the body fat percentage of healthy participants undergoing resistance training was more efficiently reduced under WSRE treatment [152]. In healthy athletes, WSRE treatment improved the cardiorespiratory endurance and increased the antioxidative capacity [153]. Moreover, WSRE showed effectivity in improving hypothyreosis. In fact, WSRE treatment led to a significant reduction in TSH and a concomitant increase in triiodothyronine (T3) and thyroxine (T4) levels [154]. The results from three further completed RCTs evaluating the effect of withania somnifera on weight loss and steatohepatitis are expected in the near future (clinicaltrials.gov). Furthermore, as WSRE was tested more extensively for other indications, there were substantial data on pharmacokinetics and safety [151,154,155,156,157,158,159].

6.3. Vitis vinifera

The beneficial properties of wine grapes on human health are not only appreciated in Mediterranean regions, and this is displayed by the extensive research on this topic. Among the range of phytochemicals, resveratrol (phytoalexin) is found in the peel and pulp, whereas the seeds mainly contain polyphenols (proanthocyanidins and flavonoids). Especially for grape seed polyphenols, the potential impact on the metabolic features was described.
In fact, in addition to the anti-inflammatory and antioxidative effects, grape seed extract (GSE) was found to regulate genes involved in metabolic homeostasis in vitro [160,161,162]. Moreover, GSE was reported to inhibit adipogenesis and increases lipolysis via targeting PPARγ in vitro [163,164]. Additionally, using endothelia cells as well as aortic ring cultures, GSE treatment revealed the eNOS-dependent vasodilatative potential in vitro [165,166].
Accordingly, the data from preclinical in vivo studies confirm the glucose and lipid metabolism-regulating as well as hepato- and cardioprotective potential of GSE. Indeed, GSE treatment improved the insulin resistance in in vivo models of obese and fructose-rich-diet rodents and attenuated pancreatic degeneration in a diabetes model [167,168,169,170,171,172,173]. In another study using healthy rats, GSE treatment following glucose intake was found to modulate glucose metabolism by upregulating the incretin GLP-1 and downregulate the GLP-1 inactivating enzyme DPP-4 [160,161,174]. Furthermore, GSE might protect pancreatic b-cell function from lipotoxic stress in vitro and in vivo in Western-diet-fed rats [175].
Moreover, weight gain, fatty liver, adipokine level, and lipid profile were counteracted in vivo in obese or fructose-fed rodents by GSE treatment [167,168,169,176,177,178,179,180,181]. The influence of GSE on weight gain might be related to an increase in portal GLP-1, ghrelin, and decreased cholecystokinin levels, reducing gastric emptying combined with enhanced satiety and reduced food intake [182]. The alleviating effect of GSE on cholesterol levels might be associated with increased bile acid secretion and the upregulation of the cholesterol-metabolizing enzyme CYP7A1 [183]. Metabolic improvements by GSE in obese mice might in part also be related to the upregulation of thermogenesis and adipose tissue browning marker UCP1, BAT and PRDM16 in white adipose tissue, and the improvement of intestinal GLP-1 and DPP-4 expressions [160,161,174,184]. The attenuation of the obesity-induced upregulation of miR-96 and its target mTOR might also contribute to GSE-mediated metabolic improvements in obesity [178]. Furthermore, Pascula-Serrano et al. suggested the GSE-mediated expansion of healthier visceral adipose tissues in obese rats as a mode of action in this model [177]. The normalization of dysbiosis might be a further mechanism of GSE-mediated metabolic improvements and attenuated obesity [173,181,184].
In addition to its protective role in obesity and dyslipidemia, GSE was also assumed to have cardioprotective properties. Indeed, GSE provoked the obesity-related prevention of cardiac siderosis, improvement of ischemia-related cardiac dysfunction and remodeling, attenuation of hypertension-dependent arterial remodeling, as well as protection against toxicity-induced cardiac damage in respective rodent in vivo models [176,185,186,187]. Furthermore, the hepatoprotective role of GSE was suggested based on the results from an in vivo rat NAFLD model, whereby GSE was found to be more effective than metformin [180]. The PPARγ-dependent modulation of hepatic lipid metabolism might be one mechanism underlying the protective effects of GSE on metabolic parameters [188].
To date, a range of RCTs evaluating the clinical potential of GSE on metabolic syndrome features were performed (Table 3). Three RCTs evaluated the impact of GSE on glucose metabolism and overall showed a limited effectivity [189,190,191]. However, in one of these studies, a GSE-mediated improvement of insulin sensitivity (HOMA-IR) was reported; there was no impact on fasting glucose levels but a decreased fructosamine level was reported by another study [189,190]. Furthermore, after long-term GSE treatment, only a tendency for improved fasting glucose and insulin sensitivity (HOMA-IR) was observed by Park et al. [191]. According to the preclinical data, GSE was shown to have a positive impact on the lipid profile parameters of dyslipidemia and overweight participants and heavy smokers [192,193,194,195]. In fact, long-term treatment resulted in reduced total cholesterol [192,193,194,195], LDL [192,193,194] and triglyceride levels [192,195]. Moreover, a GSE-dependent reduction in the artherogenic index of plasma (AIP) was reported by Yousefi et al. [195]. In contrast, no influence on the lipid parameters was observed in two additional studies [191,196]. Results from 4 RCTs suggest the therapeutic use of GSE for body weight management. In one study, GSE treatment for 3 days reduced the 24 h energy intake in the subgroup with an increased basal energy requirement of ≥7.5 MJ/day among the healthy participant cohort [197]. Furthermore, greater reductions in body weight and BMI, waist circumference, and waist to hip ratio of obese participants undergoing a caloric-restriction diet were observed when concomitantly treated with GSE in the long term [198]. Yousefi et al. also found a reduced visceral adiposity index (VAI) in GSE compared to placebo-treated overweight participants on a calorie-restriction diet [195]. Moreover, in postmenopausal women, long-term GSE treatment resulted in significantly heavier muscle mass [199]. An improved endogenous antioxidative capacity [189,194], reduced inflammatory markers (TNF-α, CRP) [198], as well as perceived stress [200], anxiety, and depression scores [199] were described in single RCTs evaluating GSE effects on T2D, healthy smokers, obese, hypertensive, and postmenopausal participants, respectively. Regarding the cardiac parameters, the available data obtained from respective RCTs reveal that the long-term GSE treatment of prehypertensive, mild hypertensive, and postmenopausal participants results in decreased systolic and, in some studies, diastolic blood pressure levels [191,199,200,201]. In two further RCTs, improvements in blood pressure were even measured after a single GSE application in overweight and prehypertensive participants [202,203]. However, no influence of GSE treatment on blood pressure in hypercholesteremia and pre/stage-I hypertensive participants was reported by Ras et al. or Preuss et al. [196,204]. Considering the vascular parameters, no relevant influence on the vasoactive systemic marker level, endothelial function, and flow-mediated dilatation (FMD) was observed after the long-term treatment of pre/stage-I hypertensive and type 2 diabetic participants with GSE [189,191,201,204]. In contrast, an improvement of the vascular health index of heavy smokers after long-term GSE application was described by Weseler et al. and suggested to be associated with the induced increase in endogenous antioxidative potential [194]. Furthermore, the overall cardiac output, assessed by impedance cardiography, was improved after the application of a single GSE dose in obese, but not in healthy, participants [202].

6.4. Camellia sinensis

The tea plant (Camellia sinensis) is found in tropical and subtropical areas with a long history in agricultural use for tea preparation that spans over 1500 years. The main polyphenolic constituents of Camellia sinensis are catechins (flavan-3ols) and their derivatives. Among these, epigallocatechine-3-gallate (EGCG) as well as a whole polyphenol mixture prepared as green tea extract (GTE) are the most studied phytochemicals of Camellia sinensis.
Furthermore, potential antidiabetic properties, including the improvement of blood glucose level and insulin resistance, were suggested for tea catechins on the basis of several preclinical in vivo studies [205,206,207,208,209,210,211]. Accordingly, EGCG was found to provoke a decrease in intestinal glucose absorption [205,212]. Regarding its mode of action, it was suggested that the inhibition of α-amylase α-glucosidase activity as well as the activation of NRF2 signaling and the regulation of glucose transporters might contribute to the antidiabetic effects of EGCG [205,209,213,214]. Whether EGCG influences the tissue uptake of blood glucose is not clear, to date, as there are opposing data on this idea [205,212]. Furthermore, tea catechins might increase blood glucose levels when pre-prandially administered and when already systemically present at the time of glucose-tolerance testing [212].
In high-fat-diet-induced rodent obesity models, EGCG targeted a further metabolic syndrome feature, as it ameliorated dyslipidemia in vivo [208,211,215,216,217]. Furthermore, a decrease in body weight and body fat mass in response to EGCG treatment was observed in vivo [207,208,211,215,218,219,220]. The inhibition of transcriptional activators regulating the expression of proprotein convertase subtilisin/kexin type 9 (PCSK9), thereby increasing hepatic LDL uptake, was discussed as a possible mechanism underlying the LDL lowering effects of EGCG [221]. The upregulation of adipocyte autophagy and regulation of thermogenic and adipogenic genes is a hypothesized mechanism underlying the weight-reduction properties of EGCG [218,219,222,223].
In addition to its anti-obesity and anti-diabetic potential, cardioprotective properties have been postulated for EGCG as well. Using hypoxia-reperfusion injury, diabetes, atherosclerosis, and endothelial dysfunction in vivo rodent models, EGCG was found to ameliorate cardiovascular parameters and endothelial dysfunction [206,224,225,226]. Interestingly, the suppression of eNOS uncoupling, a process that is associated with oxidative stress-induced endothelial dysfunction, by the normalization of BH4 level was identified as a possible underlying mechanism [227,228]. Additionally, the functional inhibition of OMA-1, a metalloendopeptidase that negatively affects mitochondrial function, as well as the inhibition of the mitochondrial apoptosis pathway by EGCG, was suggested to improve cardiomyocyte function [206,229].
Using a bile duct ligation-based liver injury, combined obesity and hypertension, as well as NAFLD in vivo models, the hepatoprotective role of tea catechins was further proposed [217,230,231,232].
To gain further insights into and to evaluate the clinical potential of polyphenols from Camellia sinensis, a variety of RCTs focusing on metabolic syndrome parameters were performed (Table 4). The evaluation of the data obtained from respective RCTs reveal that, in terms of glucose metabolism, tea polyphenols might play a bivalent role. Where some studies showed an improvement in glucose level, insulin sensitivity, and HOMA-IR index in healthy or obese participants [233,234,235,236,237,238], others did not find a positive influence of tea catechins on glucose metabolism [239,240,241,242,243]. In a further RCT, a decrease in fasting insulin after the long-term decaffeinated GTE treatment of obese participants was detected only in the subgroup showing baseline insulin levels ≥ 10 µIU/mL [244]. Of note, the timing of the application might account for the catechin-dependent outcome on the glucose level. In fact, in an open randomized clinical trial, the treatment of healthy participants with green tea catechins one hour before glucose-tolerance testing resulted in higher plasma glucose levels, whereas a reduction in glucose levels was observed when catechins and glucose were concomitantly administered [212]. However, based on the limitation of the available data on the latter issue, a final assessment could not be performed at this point. Regarding the influence of tea catechins on the lipid profile, the available data also do not provide consistent results. In 5 out of 10 evaluated placebo-controlled double-blind RCTs, an improvement of single, but not all, assessed lipid profile parameters, including a decrease in total cholesterol, LDLs, and triglycerides by GTE and EGCG in healthy, obese, and diabetic participants was reported [234,245,246,247,248]. In contrast, no impact of long-term EGCG or GTE treatments on these parameters in obese or postmenopausal participants was found [239,241,242,243,249]. Body weight reduction was observed in only one RTC after the long-term treatment of metabolic syndrome participants [250]. In a further study, a GTE-dependent increased fat oxidation in a healthy study cohort undergoing exercise intervention was observed compared to the exercise intervention group taking a placebo [237]. Moreover, a delayed gastric emptying and increased satiation as well as adiponectin level were found in healthy participants treated with a single dose of EGCG [240]. However, no influence on body weight, BMI, body fat mass, fat oxidation, waist circumference, energy intake, and satiety was reported by GTE and EGCG in obese participants in the majority of the published RCTs [233,235,239,242,249,251,252,253]. Regarding the cardiovascular parameters, reduced arterial stiffness and increased flow mediated dilatation (FMD) after the long-term treatment of coronary artery disease and diabetic participants was reported by Widlansky et al. and Quezada-Fernandez et al. [254,255]. Increased FMD in response to the single application of tea catechins was also observed in a placebo-controlled, but open-label, clinical trial [256]. Furthermore, in obese participants performing physical exercise, a reduction in the resting heart rate was observed [235]. The data from two further RCTs show a reduction in blood pressure parameters in obese participants in response to long-term EGCG treatment [233,239]. In contrast, no clear impact of GTE and EGCG on cardiovascular parameters, including blood pressure and heart rate in hypertensive participants after resistance training, was reported by Arazi et al. [257].

7. General Aspects of the Future Use of Phytochemicals in HS Patients

Metabolic alterations, in particular, obesity, can support sustained inflammation and thereby exacerbate skin manifestations and the chronification of HS. However, they lack the necessary attention in HS therapy. Considering the data from the evaluated preclinical and clinical studies suggest that phytochemicals from Olea europea, Withania somnifera, Camellia sinensis, and Vitis vinifera represent potent candidates for targeting metabolic dysfunction. As the phytochemicals evaluated here have partly overlapping properties, different phytotherapeutic options for the treatment of single metabolic syndrome features central obesity, insulin resistance, triglyceridemia, hypo-high-density lipoprotein (HDL)-cholesterolemia, and hypertension exist. Furthermore, when considering an integrative HS therapy using phytochemicals, the following aspects should be taken into account. First, the priority of MetS parameter(s) that need the relevant improvements should be determined. Second, considering the present concomitant medication of the patient, the relevant potential drug interactions with the phytochemical candidates should be carefully estimated and taken into account for the decision. Third, the decision for the appropriate phytochemical should also depend on the safety profile of the phytochemical of choice, analyzed in regard of the individual clinical condition of the patient. In general, strict medical supervision and monitoring should be prerequisites for performing integrative therapy using phytochemicals. Before and consecutively during therapy with the selected phytochemicals, it is highly recommended to perform a detailed analysis of the relevant physical (e.g., cardiovascular) and laboratory parameters (including indicators of lipid/glucose metabolism, coagulation status, and liver enzymes), as well as HS (e.g., IHS4; [258]) and QoL scoring to enable the careful monitoring of safety, drug interaction, and therapeutic effectivity.

8. Safety and Drug Interaction

Phytochemicals derived from Olea europea, Withania somnifera, and Vitis vinifera showed an overall good tolerability and safety profile during clinical use [122,151,154,155,156,157,159,200,259,260,261,262,263,264,265,266]. Phytochemicals derived from Camellia sinensis were extensively studied in regard of their pharmacokinetics and safety, and for the clinical use of EGCG, an upper safe-dosage limit (338 mg for extracts; 704 mg for beverages) was recommended [267]. This recommendation was based on the described liver toxicity as a possible rare adverse reaction resulting from a high bolus-dose application. In contrast, these safety concerns were not raised for the use of beverages produced from the whole leaves or extract of Camellia sinensis [267]. However, therapy with Camellia sinensis phytochemicals should be avoided for patients with known hepatic dysfunctions.
The main described potential drug interactions of the phytochemicals evaluated here were those related to cytochrome P450-metabolizing/detoxifying enzymes. In fact, for Olea europea-, Camellia sinensis-, and Vitis vinifera-derived phytochemicals, an interaction with cytochrome P450-detoxifying enzymes was reported [268,269,270,271,272]. As this may influence the pharmacokinetics of concomitantly administered P450-metabolized drugs, the efficacy of concomitant medication and, respectively, associated clinical parameters should be monitored during the treatment with these phytochemicals. Whether WSRE from Withania somnifera interacted with cytochrome P450 enzymes was not clarified; however, the precautionary monitoring of the efficacy of concurrent drug medication was also recommended [273,274,275].
Furthermore, phytochemicals from Camellia sinensis were found to be inhibitors of the enzyme catechol-o-methyltransferase (COMT), and might therefore modify the detoxification and metabolization of xenobiotics, catecholamines, and catechol estrogens [276]. For patients carrying the low-activity COMT genotype receiving, e.g., levodopa, apomorphine, isoprenaline, catecholamines, micafungin, or estrogen derivates, or those suffering from estrogen dominance, an awareness for potential drug interactions is needed. For patients with known prediabetes/diabetes, the risk-benefit ratio should also be carefully weighted using this medication based on the possible influence on glucose metabolism [205,212]. Whether the epigenetic modifying potential of EGCG has a clinical relevance for patients remains to be investigated. Of note, Withania somnifera phytochemicals were observed to improve thyroid function, indicated from a decrease in TSH and increase in triiodothyronine (T3) and thyroxine (T4) levels in subclinical hypothyroid patients [154]. The monitoring of thyroid parameters is therefore recommended for hyperthyroid patients as well as patients receiving L-thyroxin supplementation. For Olea europea phytochemicals, the inhibitory property of enzymes that played a role in Alzheimer’s disease progression in vitro was described; however, the clinical relevance of these data remains to be investigated [277]. More detailed information regarding safety and drug interactions are summarized in an previously published review [83].

9. Recommendations for Integrated Phytotherapy Targeting MetS Parameters in HS Patients

For the improvement of glucose metabolism, in principle, OLE (Olea europea) was shown to be eligible (Table 5). A daily dose of 20–160 mg of oleuropein or 250–500 mg of OLE is recommended (Figure 3).
In case of dyslipidemia, GSE (Vitis vinifera) was reported to be eligible (Table 5), doses ranging from 200–300 mg (GSE) daily were recommended (Figure 3). Protective effects regarding the cardiovascular parameters were described for GSE (Vitis vinifera) and EGCG/GTE (Camellia sinensis) (Table 5), whereby more RCTs were available for the latter drug. A daily dosage of 100–400 mg (GSE) or 75–300 mg (EGCG) or 400–1060 mg (EGCG/GTE) were recommended (Figure 3).
The evaluated RCTs reveal that, for weight management, WSRE (Withania somnifera) and GSE (Vitis vinifera) might represent eligible phytochemical drugs (Table 5). There are more data on GSE than for WSRE; however, the data on additional, already completed RCTs evaluating the effect of Withania somnifera on weight loss are awaited in the near future (clinicaltrials.gov). A daily dosage of 600 mg (WSRE) or 100–900 mg (GSE) was recommended (Figure 3).
To date, no RCTs are available evaluating the possible hepatoprotective effects of OLE, WSRE, GSE, and EGCG/GTE (Table 5). Nevertheless, for all these drugs, an improvement of hepatic parameters in various in vivo animal models was reported. However, as described in the above section (Safety and Drug Interaction), EGCG/GTE application is not recommended for patients with hepatic dysfunctions as a safety precaution.

Author Contributions

Conceptualization, K.W. (Katrin Witte), K.W. (Kerstin Wolk) and R.S.; writing—original draft preparation, K.W. (Katrin Witte); writing—review and editing, K.W. (Katrin Witte), K.W. (Kerstin Wolk), E.W.-H., T.K., G.K. and R.S.; visualization, K.W. (Katrin Witte), K.W. (Kerstin Wolk), T.K., G.K. and R.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Photos of skin lesions of HS patients were taken at the Charité-Universitätsmedizin Berlin after receiving the written, informed consent of the patients.

Data Availability Statement

Not applicable.

Acknowledgments

We acknowledge the financial support from the Open Access Publication Fund of Charité–Universitätsmedizin Berlin and the German Research Foundation (DFG). We thank Eva Schmidbauer (Botanischer Garten München-Nymphenburg, head of the outdoor area), Stefanie Kunz, Georgios Kokolakis, and Malte Rozmarynowicz for the botanical pictures.

Conflicts of Interest

K. Wolk received research grants, travel grants, consulting honoraria, and lecturer’s honoraria from AbbVie, Celgene/BMS, Charité Research Organisation, Willmar Schwabe GmbH & Co. KG, Flexopharm, JanssenCilag, Novartis, Pfizer, Sanofi-Aventis, TFS, and UCB. R. Sabat received research grants or honoraria for participation in advisory boards, clinical trials, or as a speaker for one or more of the following: AbbVie, Almirall Hermal, Amgen, Bayer Schering Pharma, Boehringer Ingelheim, Celgene, Charité Research Organisation, CSL Behring, Willmar Schwabe, Flexopharm, ICON PLC, Incyte Corporation, JanssenCilag, La Roche-Posay Laboratoire Dermatologique, MoonLake Immunotherapeutics, Novartis, Parexel, Rheinischen Friedrich-Wilhelms-Universität Bonn, Sanofi–Aventis, TFS, and UCB. The other authors declare no conflict of interest.

References

  1. Sabat, R.; Jemec, G.B.E.; Matusiak, L.; Kimball, A.B.; Prens, E.; Wolk, K. Hidradenitis suppurativa. Nat. Rev. Dis. Prim. 2020, 6, 18. [Google Scholar] [CrossRef] [PubMed]
  2. Alsadhan, H.; Alfawzan, A.I.; Yaqoub, A.; Almoneef, A.; Almohideb, M. Hidradenitis Suppurativa: Estimated Prevalence, Clinical Features, and Risk Factors in Riyadh, Saudi Arabia. Cureus 2022, 14, e23029. [Google Scholar] [CrossRef]
  3. Hagan, P.G.; Bouazzi, D.; Nyarko, G.; Dartey, E.S.; Nunoo-Ghartey, K.B.; Nkum, D.; Ansu, P.; Boakye, J.Y.; Andersen, R.K.; Boer, J.; et al. Prevalence of Hidradenitis Suppurativa in Berekum, Ghana. Br. J. Dermatol. 2022, 187, 586–587. [Google Scholar] [CrossRef] [PubMed]
  4. Fernandez-Avila, D.G.; Anzola, L.C.; Cardona, L.P.G. Prevalence of Hidradenitis Suppurativa in Colombia According to Data from the National Health Registry. Skinmed 2021, 19, 369–373. [Google Scholar] [PubMed]
  5. Han, H.R.; Choi, C.E.E.; Nagad, M.; Patwardhan, K.R.; Boer, J.; Jemec, G.B.E.; Chandran, N.S. Prevalence and perceptions towards hidradenitis suppurativa: A cross-sectional study in a non-dermatological outpatient population. J. Eur. Acad. Dermatol. Venereol. 2022, 36, e392–e394. [Google Scholar] [CrossRef] [PubMed]
  6. Prens, L.M.; Bouwman, K.; Troelstra, L.D.; Prens, E.P.; Alizadeh, B.Z.; Horvath, B. New insights in hidradenitis suppurativa from a population-based Dutch cohort: Prevalence, smoking behaviour, socioeconomic status and comorbidities. Br. J. Dermatol. 2022, 186, 814–822. [Google Scholar] [CrossRef]
  7. Kokolakis, G.; Wolk, K.; Schneider-Burrus, S.; Kalus, S.; Barbus, S.; Gomis-Kleindienst, S.; Sabat, R. Delayed Diagnosis of Hidradenitis Suppurativa and Its Effect on Patients and Healthcare System. Dermatology 2020, 236, 421–430. [Google Scholar] [CrossRef]
  8. Liakou, A.I.; Papadakis, M.; Tsantes, A.G.; Tsante, K.A.; Kontochristopoulos, G.; Marnelakis, I.; Katoulis, A.; Grigoriou, S.; Rigopoulos, D. Perception and Knowledge of Hidradenitis Suppurativa in Greece: A Cross-Sectional Study of 1301 Individuals. Indian J. Dermatol. 2022, 67, 835. [Google Scholar] [CrossRef]
  9. Sabat, R.; Tsaousi, A.; Ghoreschi, K.; Wolk, K.; Schneider-Burrus, S. Sex-disaggregated population analysis in patients with hidradenitis suppurativa. Front. Med. 2022, 9, 1028943. [Google Scholar] [CrossRef]
  10. Schneider-Burrus, S.; Lux, G.; van der Linde, K.; Barbus, S.; Huss-Marp, J.; Tsaousi, A.; Wasem, J.; Wolff, B.; Sabat, R. Hidradenitis suppurativa—Prevalence analyses of German statutory health insurance data. J. Eur. Acad. Dermatol. Venereol. 2021, 35, e32–e35. [Google Scholar] [CrossRef]
  11. Sokumbi, O.; Hodge, D.O.; Ederaine, S.A.; Alavi, A.; Alikhan, A. Comorbid diseases of hidradenitis suppurativa: A 15-year population-based study in Olmsted County, Minnesota, USA. Int. J. Dermatol. 2022, 61, 1372–1379. [Google Scholar] [CrossRef]
  12. Liang, Y.T.; Yeh, C.J.; Huang, J.Y.; Wei, J.C. Epidemiology of hidradenitis suppurativa in Taiwan: A 14-year nationwide population-based study. J. Dermatol. 2021, 48, 613–619. [Google Scholar] [CrossRef] [PubMed]
  13. Lee, J.W.; Heo, Y.W.; Lee, J.H.; Lee, S. Epidemiology and comorbidity of hidradenitis suppurativa in Korea for 17 years: A nationwide population-based cohort study. J. Dermatol. 2023, 50, 778–786. [Google Scholar] [CrossRef] [PubMed]
  14. Schneider-Burrus, S.; Tsaousi, A.; Barbus, S.; Huss-Marp, J.; Witte, K.; Wolk, K.; Fritz, B.; Sabat, R. Features Associated With Quality of Life Impairment in Hidradenitis Suppurativa Patients. Front. Med. 2021, 8, 676241. [Google Scholar] [CrossRef]
  15. Glowaczewska, A.; Reszke, R.; Szepietowski, J.C.; Matusiak, L. Indirect Self-Destructiveness in Hidradenitis Suppurativa Patients. J. Clin. Med. 2021, 10, 4194. [Google Scholar] [CrossRef] [PubMed]
  16. Rymaszewska, J.E.; Krajewski, P.K.; Szczech, J.; Szepietowski, J.C. Depression and anxiety in hidradenitis suppurativa patients: A cross-sectional study among Polish patients. Postep. Dermatol. Alergol. 2023, 40, 35–39. [Google Scholar] [CrossRef]
  17. Schneider-Burrus, S.; Jost, A.; Peters, E.M.J.; Witte-Haendel, E.; Sterry, W.; Sabat, R. Association of Hidradenitis Suppurativa With Body Image. JAMA Dermatol. 2018, 154, 447–451. [Google Scholar] [CrossRef] [PubMed]
  18. Akoglu, G.; Yildiz, I.; Karaismailoglu, E.; Esme, P. Disease severity and poor mental health are the main predictors of stigmatization in patients with hidradenitis suppurativa. Dermatol. Ther. 2021, 34, e14910. [Google Scholar] [CrossRef]
  19. Rymaszewska, J.E.; Krajewski, P.K.; Matusiak, L.; Maj, J.; Szepietowski, J.C. Satisfaction with Life and Coping Strategies among Patients with Hidradenitis Suppurativa: A Cross-Sectional Study. J. Clin. Med. 2023, 12, 2755. [Google Scholar] [CrossRef]
  20. Singh, R.; Kelly, K.A.; Senthilnathan, A.; Feldman, S.R.; Pichardo, R.O. Stigmatization, a social perception which may have a debilitating impact on hidradenitis suppurativa patients: An observational study. Arch. Dermatol. Res. 2023, 315, 1049–1052. [Google Scholar] [CrossRef]
  21. Schneider-Burrus, S.; Kalus, S.; Fritz, B.; Wolk, K.; Gomis-Kleindienst, S.; Sabat, R. The impact of hidradenitis suppurativa on professional life. Br. J. Dermatol. 2023, 188, 122–130. [Google Scholar] [CrossRef]
  22. Wolk, K.; Join-Lambert, O.; Sabat, R. Aetiology and pathogenesis of hidradenitis suppurativa. Br. J. Dermatol. 2020, 183, 999–1010. [Google Scholar] [CrossRef] [PubMed]
  23. Hana, A.; Booken, D.; Henrich, C.; Gratchev, A.; Maas-Szabowski, N.; Goerdt, S.; Kurzen, H. Functional significance of non-neuronal acetylcholine in skin epithelia. Life Sci. 2007, 80, 2214–2220. [Google Scholar] [CrossRef]
  24. Radek, K.A.; Elias, P.M.; Taupenot, L.; Mahata, S.K.; O’Connor, D.T.; Gallo, R.L. Neuroendocrine nicotinic receptor activation increases susceptibility to bacterial infections by suppressing antimicrobial peptide production. Cell Host Microbe 2010, 7, 277–289. [Google Scholar] [CrossRef]
  25. Wu, Y.; Ma, Y.; Xu, T.; Zhang, Q.Z.; Bai, J.; Wang, J.; Zhu, T.; Lou, Q.; Gotz, F.; Qu, D.; et al. Nicotine Enhances Staphylococcus epidermidis Biofilm Formation by Altering the Bacterial Autolysis, Extracellular DNA Releasing, and Polysaccharide Intercellular Adhesin Production. Front. Microbiol. 2018, 9, 2575. [Google Scholar] [CrossRef] [PubMed]
  26. Wolk, K.; Warszawska, K.; Hoeflich, C.; Witte, E.; Schneider-Burrus, S.; Witte, K.; Kunz, S.; Buss, A.; Roewert, H.J.; Krause, M.; et al. Deficiency of IL-22 contributes to a chronic inflammatory disease: Pathogenetic mechanisms in acne inversa. J. Immunol. 2011, 186, 1228–1239. [Google Scholar] [CrossRef]
  27. Witte-Handel, E.; Wolk, K.; Tsaousi, A.; Irmer, M.L.; Mossner, R.; Shomroni, O.; Lingner, T.; Witte, K.; Kunkel, D.; Salinas, G.; et al. The IL-1 Pathway Is Hyperactive in Hidradenitis Suppurativa and Contributes to Skin Infiltration and Destruction. J. Investig. Dermatol. 2019, 139, 1294–1305. [Google Scholar] [CrossRef]
  28. Krajewski, P.K.; Szepietowski, J.C.; Martorell, A. Tunnels in Hidradenitis Suppurativa: Active Inflammatory Entities with Specific Molecular and Genetic Profiles—A Narrative Review. Dermatology 2023, 239, 323–327. [Google Scholar] [CrossRef] [PubMed]
  29. Lima, A.L.; Karl, I.; Giner, T.; Poppe, H.; Schmidt, M.; Presser, D.; Goebeler, M.; Bauer, B. Keratinocytes and neutrophils are important sources of proinflammatory molecules in hidradenitis suppurativa. Br. J. Dermatol. 2016, 174, 514–521. [Google Scholar] [CrossRef]
  30. Moran, B.; Smith, C.M.; Zabarowski, A.; Ryan, M.; Karman, J.; Dunstan, R.W.; Smith, K.M.; Hambly, R.; Musilova, J.; Petrasca, A.; et al. Targeting the NLRP3 inflammasome reduces inflammation in hidradenitis suppurativa skin. Br. J. Dermatol. 2023, ljad184. [Google Scholar] [CrossRef]
  31. Sabat, R.; Simaite, D.; Gudjonsson, J.E.; Brembach, T.C.; Witte, K.; Krause, T.; Kokolakis, G.; Bartnik, E.; Nikolaou, C.; Rill, N.; et al. Neutrophilic granulocyte-derived B-cell activating factor supports B cells in skin lesions in hidradenitis suppurativa. J. Allergy Clin. Immunol. 2023, 151, 1015–1026. [Google Scholar] [CrossRef] [PubMed]
  32. Tsaousi, A.; Witte, E.; Witte, K.; Rowert-Huber, H.J.; Volk, H.D.; Sterry, W.; Wolk, K.; Schneider-Burrus, S.; Sabat, R. MMP8 Is Increased in Lesions and Blood of Acne Inversa Patients: A Potential Link to Skin Destruction and Metabolic Alterations. Mediat. Inflamm. 2016, 2016, 4097574. [Google Scholar] [CrossRef] [PubMed]
  33. Wolk, K.; Brembach, T.C.; Simaite, D.; Bartnik, E.; Cucinotta, S.; Pokrywka, A.; Irmer, M.L.; Triebus, J.; Witte-Handel, E.; Salinas, G.; et al. Activity and components of the granulocyte colony-stimulating factor pathway in hidradenitis suppurativa. Br. J. Dermatol. 2021, 185, 164–176. [Google Scholar] [CrossRef] [PubMed]
  34. Wolk, K.; Wenzel, J.; Tsaousi, A.; Witte-Handel, E.; Babel, N.; Zelenak, C.; Volk, H.D.; Sterry, W.; Schneider-Burrus, S.; Sabat, R. Lipocalin-2 is expressed by activated granulocytes and keratinocytes in affected skin and reflects disease activity in acne inversa/hidradenitis suppurativa. Br. J. Dermatol. 2017, 177, 1385–1393. [Google Scholar] [CrossRef]
  35. Kanni, T.; Zenker, O.; Habel, M.; Riedemann, N.; Giamarellos-Bourboulis, E.J. Complement activation in hidradenitis suppurativa: A new pathway of pathogenesis? Br. J. Dermatol. 2018, 179, 413–419. [Google Scholar] [CrossRef]
  36. Macchiarella, G.; Cornacchione, V.; Cojean, C.; Riker, J.; Wang, Y.; Te, H.; Ceci, M.; Gudjonsson, J.E.; Gaulis, S.; Goetschy, J.F.; et al. Disease Association of Anti–Carboxyethyl Lysine Autoantibodies in Hidradenitis Suppurativa. J. Investig. Dermatol. 2023, 143, 273–283 e212. [Google Scholar] [CrossRef]
  37. Oliveira, C.B.; Byrd, A.S.; Okoye, G.A.; Kaplan, M.J.; Carmona-Rivera, C. Neutralizing Anti–DNase 1 and –DNase 1L3 Antibodies Impair Neutrophil Extracellular Traps Degradation in Hidradenitis Suppurativa. J. Investig. Dermatol. 2023, 143, 57–66. [Google Scholar] [CrossRef]
  38. Sabat, R.; Grutz, G.; Warszawska, K.; Kirsch, S.; Witte, E.; Wolk, K.; Geginat, J. Biology of interleukin-10. Cytokine Growth Factor Rev. 2010, 21, 331–344. [Google Scholar] [CrossRef]
  39. Wolk, K.; Docke, W.; von Baehr, V.; Volk, H.; Sabat, R. Comparison of monocyte functions after LPS- or IL-10-induced reorientation: Importance in clinical immunoparalysis. Pathobiology 1999, 67, 253–256. [Google Scholar] [CrossRef]
  40. Sabat, R.; Chanwangpong, A.; Schneider-Burrus, S.; Metternich, D.; Kokolakis, G.; Kurek, A.; Philipp, S.; Uribe, D.; Wolk, K.; Sterry, W. Increased prevalence of metabolic syndrome in patients with acne inversa. PLoS ONE 2012, 7, e31810. [Google Scholar] [CrossRef]
  41. Damiani, G.; Leone, S.; Fajgenbaum, K.; Bragazzi, N.L.; Pacifico, A.; Conic, R.R.; Pigatto, P.D.; Maiorana, C.; Poli, P.; Berti, E.; et al. Nonalcoholic fatty liver disease prevalence in an Italian cohort of patients with hidradenitis suppurativa: A multi-center retrospective analysis. World J. Hepatol. 2019, 11, 391–401. [Google Scholar] [CrossRef] [PubMed]
  42. Deckers, I.E.; Benhadou, F.; Koldijk, M.J.; Del Marmol, V.; Horvath, B.; Boer, J.; van der Zee, H.H.; Prens, E.P. Inflammatory bowel disease is associated with hidradenitis suppurativa: Results from a multicenter cross-sectional study. J. Am. Acad. Dermatol. 2017, 76, 49–53. [Google Scholar] [CrossRef]
  43. Duran-Vian, C.; Arias-Loste, M.T.; Hernandez, J.L.; Fernandez, V.; Gonzalez, M.; Iruzubieta, P.; Rasines, L.; Gonzalez-Vela, C.; Vaque, J.P.; Blanco, R.; et al. High prevalence of non-alcoholic fatty liver disease among hidradenitis suppurativa patients independent of classic metabolic risk factors. J. Eur. Acad. Dermatol. Venereol. 2019, 33, 2131–2136. [Google Scholar] [CrossRef] [PubMed]
  44. Egeberg, A.; Gislason, G.H.; Hansen, P.R. Risk of Major Adverse Cardiovascular Events and All-Cause Mortality in Patients With Hidradenitis Suppurativa. JAMA Dermatol. 2016, 152, 429–434. [Google Scholar] [CrossRef]
  45. Egeberg, A.; Jemec, G.B.E.; Kimball, A.B.; Bachelez, H.; Gislason, G.H.; Thyssen, J.P.; Mallbris, L. Prevalence and Risk of Inflammatory Bowel Disease in Patients with Hidradenitis Suppurativa. J. Investig. Dermatol. 2017, 137, 1060–1064. [Google Scholar] [CrossRef] [PubMed]
  46. Gonzalez-Villanueva, I.; DeGracia, C.; Planells, M.; Poveda, I.; Alvarez, P.; Schneller-Pavalescu, L.; Betlloch, I.; Jemec, G.B.E.; Ramos, J.M.; Pascual, J.C. Hidradenitis Suppurativa is Associated with Non-alcoholic Fatty Liver Disease: A Cross-sectional Study. Acta Derm. Venereol. 2020, 100, adv00239. [Google Scholar] [CrossRef]
  47. Richette, P.; Molto, A.; Viguier, M.; Dawidowicz, K.; Hayem, G.; Nassif, A.; Wendling, D.; Aubin, F.; Liote, F.; Bachelez, H. Hidradenitis suppurativa associated with spondyloarthritis—Results from a multicenter national prospective study. J. Rheumatol. 2014, 41, 490–494. [Google Scholar] [CrossRef]
  48. Schneider-Burrus, S.; Witte-Haendel, E.; Christou, D.; Rigoni, B.; Sabat, R.; Diederichs, G. High Prevalence of Back Pain and Axial Spondyloarthropathy in Patients with Hidradenitis Suppurativa. Dermatology 2016, 232, 606–612. [Google Scholar] [CrossRef]
  49. Chait, A.; den Hartigh, L.J. Adipose Tissue Distribution, Inflammation and Its Metabolic Consequences, Including Diabetes and Cardiovascular Disease. Front. Cardiovasc. Med. 2020, 7, 22. [Google Scholar] [CrossRef]
  50. Manna, P.; Jain, S.K. Obesity, Oxidative Stress, Adipose Tissue Dysfunction, and the Associated Health Risks: Causes and Therapeutic Strategies. Metab. Syndr. Relat. Disord. 2015, 13, 423–444. [Google Scholar] [CrossRef]
  51. Zatterale, F.; Longo, M.; Naderi, J.; Raciti, G.A.; Desiderio, A.; Miele, C.; Beguinot, F. Chronic Adipose Tissue Inflammation Linking Obesity to Insulin Resistance and Type 2 Diabetes. Front. Physiol. 2019, 10, 1607. [Google Scholar] [CrossRef]
  52. Sanchez-Pino, M.D.; Richardson, W.S.; Zabaleta, J.; Puttalingaiah, R.T.; Chapple, A.G.; Liu, J.; Kim, Y.; Ponder, M.; DeArmitt, R.; Baiamonte, L.B.; et al. Increased inflammatory low-density neutrophils in severe obesity and effect of bariatric surgery: Results from case-control and prospective cohort studies. EBioMedicine 2022, 77, 103910. [Google Scholar] [CrossRef]
  53. Weisberg, S.P.; McCann, D.; Desai, M.; Rosenbaum, M.; Leibel, R.L.; Ferrante, A.W., Jr. Obesity is associated with macrophage accumulation in adipose tissue. J. Clin. Investig. 2003, 112, 1796–1808. [Google Scholar] [CrossRef]
  54. Elgazar-Carmon, V.; Rudich, A.; Hadad, N.; Levy, R. Neutrophils transiently infiltrate intra-abdominal fat early in the course of high-fat feeding. J. Lipid Res. 2008, 49, 1894–1903. [Google Scholar] [CrossRef]
  55. Talukdar, S.; Oh, D.Y.; Bandyopadhyay, G.; Li, D.; Xu, J.; McNelis, J.; Lu, M.; Li, P.; Yan, Q.; Zhu, Y.; et al. Neutrophils mediate insulin resistance in mice fed a high-fat diet through secreted elastase. Nat. Med. 2012, 18, 1407–1412. [Google Scholar] [CrossRef]
  56. Soehnlein, O.; Steffens, S.; Hidalgo, A.; Weber, C. Neutrophils as protagonists and targets in chronic inflammation. Nat. Rev. Immunol. 2017, 17, 248–261. [Google Scholar] [CrossRef] [PubMed]
  57. Watanabe, Y.; Nagai, Y.; Honda, H.; Okamoto, N.; Yanagibashi, T.; Ogasawara, M.; Yamamoto, S.; Imamura, R.; Takasaki, I.; Hara, H.; et al. Bidirectional crosstalk between neutrophils and adipocytes promotes adipose tissue inflammation. FASEB J. 2019, 33, 11821–11835. [Google Scholar] [CrossRef] [PubMed]
  58. Wolk, K.; Sabat, R. Adipokines in psoriasis: An important link between skin inflammation and metabolic alterations. Rev. Endocr. Metab. Disord. 2016, 17, 305–317. [Google Scholar] [CrossRef]
  59. Gonzalez-Lopez, M.A.; Vilanova, I.; Ocejo-Vinals, G.; Arlegui, R.; Navarro, I.; Guiral, S.; Mata, C.; Perez-Paredes, M.G.; Portilla, V.; Corrales, A.; et al. Circulating levels of adiponectin, leptin, resistin and visfatin in non-diabetics patients with hidradenitis suppurativa. Arch. Dermatol. Res. 2020, 312, 595–600. [Google Scholar] [CrossRef] [PubMed]
  60. Malara, A.; Hughes, R.; Jennings, L.; Sweeney, C.M.; Lynch, M.; Awdeh, F.; Timoney, I.; Tobin, A.M.; Lynam-Loane, K.; Tobin, L.; et al. Adipokines are dysregulated in patients with hidradenitis suppurativa. Br. J. Dermatol. 2018, 178, 792–793. [Google Scholar] [CrossRef] [PubMed]
  61. Krajewski, P.K.; Matusiak, L.; Szepietowski, J.C. Adipokines as an important link between hidradenitis suppurativa and obesity: A narrative review. Br. J. Dermatol. 2023, 188, 320–327. [Google Scholar] [CrossRef] [PubMed]
  62. Blaszczak, A.M.; Jalilvand, A.; Hsueh, W.A. Adipocytes, Innate Immunity and Obesity: A Mini-Review. Front. Immunol. 2021, 12, 650768. [Google Scholar] [CrossRef] [PubMed]
  63. Witte, K.; Schneider-Burrus, S.; Salinas, G.; Mossner, R.; Ghoreschi, K.; Wolk, K.; Sabat, R. Blood T Helper Memory Cells: A Tool for Studying Skin Inflammation in HS? Int. J. Mol. Sci. 2023, 24, 8854. [Google Scholar] [CrossRef]
  64. Kadowaki, S.; Okamura, T.; Hozawa, A.; Kadowaki, T.; Kadota, A.; Murakami, Y.; Nakamura, K.; Saitoh, S.; Nakamura, Y.; Hayakawa, T.; et al. Relationship of elevated casual blood glucose level with coronary heart disease, cardiovascular disease and all-cause mortality in a representative sample of the Japanese population. NIPPON DATA80. Diabetologia 2008, 51, 575–582. [Google Scholar] [CrossRef] [PubMed]
  65. Poznyak, A.V.; Litvinova, L.; Poggio, P.; Sukhorukov, V.N.; Orekhov, A.N. Effect of Glucose Levels on Cardiovascular Risk. Cells 2022, 11, 3034. [Google Scholar] [CrossRef] [PubMed]
  66. Riise, H.K.R.; Igland, J.; Sulo, G.; Graue, M.; Haltbakk, J.; Tell, G.S.; Iversen, M.M. Casual blood glucose and subsequent cardiovascular disease and all-cause mortality among 159 731 participants in Cohort of Norway (CONOR). BMJ Open Diabetes Res. Care 2021, 9, e001928. [Google Scholar] [CrossRef]
  67. Shi, H.; Kokoeva, M.V.; Inouye, K.; Tzameli, I.; Yin, H.; Flier, J.S. TLR4 links innate immunity and fatty acid-induced insulin resistance. J. Clin. Investig. 2006, 116, 3015–3025. [Google Scholar] [CrossRef]
  68. Vandanmagsar, B.; Youm, Y.H.; Ravussin, A.; Galgani, J.E.; Stadler, K.; Mynatt, R.L.; Ravussin, E.; Stephens, J.M.; Dixit, V.D. The NLRP3 inflammasome instigates obesity-induced inflammation and insulin resistance. Nat. Med. 2011, 17, 179–188. [Google Scholar] [CrossRef]
  69. Wen, H.; Gris, D.; Lei, Y.; Jha, S.; Zhang, L.; Huang, M.T.; Brickey, W.J.; Ting, J.P. Fatty acid-induced NLRP3-ASC inflammasome activation interferes with insulin signaling. Nat. Immunol. 2011, 12, 408–415. [Google Scholar] [CrossRef]
  70. Miller, I.M.; Ellervik, C.; Vinding, G.R.; Zarchi, K.; Ibler, K.S.; Knudsen, K.M.; Jemec, G.B. Association of metabolic syndrome and hidradenitis suppurativa. JAMA Dermatol. 2014, 150, 1273–1280. [Google Scholar] [CrossRef]
  71. Kromann, C.B.; Ibler, K.S.; Kristiansen, V.B.; Jemec, G.B. The influence of body weight on the prevalence and severity of hidradenitis suppurativa. Acta Derm. Venereol. 2014, 94, 553–557. [Google Scholar] [CrossRef]
  72. Sivanand, A.; Gulliver, W.P.; Josan, C.K.; Alhusayen, R.; Fleming, P.J. Weight Loss and Dietary Interventions for Hidradenitis Suppurativa: A Systematic Review. J. Cutan. Med. Surg. 2020, 24, 64–72. [Google Scholar] [CrossRef] [PubMed]
  73. Stagakis, I.; Bertsias, G.; Karvounaris, S.; Kavousanaki, M.; Virla, D.; Raptopoulou, A.; Kardassis, D.; Boumpas, D.T.; Sidiropoulos, P.I. Anti-tumor necrosis factor therapy improves insulin resistance, beta cell function and insulin signaling in active rheumatoid arthritis patients with high insulin resistance. Arthritis Res. Ther. 2012, 14, R141. [Google Scholar] [CrossRef]
  74. Tiri, H.; Jokelainen, J.; Timonen, M.; Tasanen, K.; Huilaja, L. Substantially reduced life expectancy in patients with hidradenitis suppurativa: A Finnish nationwide registry study. Br. J. Dermatol. 2019, 180, 1543–1544. [Google Scholar] [CrossRef] [PubMed]
  75. Scholl, L.; Schneider-Burrus, S.; Fritz, B.; Sabat, R.; Bechara, F.G. The impact of surgical interventions on the psychosocial well-being of patients with hidradenitis suppurativa. J. Dtsch. Dermatol. Ges. 2023, 21, 131–139. [Google Scholar] [CrossRef]
  76. Ujiie, H.; Rosmarin, D.; Schon, M.P.; Stander, S.; Boch, K.; Metz, M.; Maurer, M.; Thaci, D.; Schmidt, E.; Cole, C.; et al. Unmet Medical Needs in Chronic, Non-communicable Inflammatory Skin Diseases. Front. Med. 2022, 9, 875492. [Google Scholar] [CrossRef]
  77. Price, K.N.; Collier, E.K.; Grogan, T.; Fernandez, J.M.; Alhusayen, R.; Alavi, A.; Hamzavi, I.H.; Lowes, M.A.; Porter, M.J.; Hsiao, J.L.; et al. Physician perspectives on complementary and alternative medicine in hidradenitis suppurativa. Dermatol. Ther. 2021, 34, e14851. [Google Scholar] [CrossRef] [PubMed]
  78. Price, K.N.; Thompson, A.M.; Rizvi, O.; Hendricks, A.J.; Alavi, A.; Hsiao, J.L.; Shi, V.Y. Complementary and Alternative Medicine Use in Patients With Hidradenitis Suppurativa. JAMA Dermatol. 2020, 156, 345–348. [Google Scholar] [CrossRef]
  79. Hassan, H.M. A Short History of the Use of Plants as Medicines from Ancient Times. Chimia 2015, 69, 622–623. [Google Scholar] [CrossRef]
  80. Zhao, Z.; Guo, P.; Brand, E. A concise classification of bencao (materia medica). Chin. Med. 2018, 13, 18. [Google Scholar] [CrossRef]
  81. Petrovska, B.B. Historical review of medicinal plants’ usage. Pharmacogn. Rev. 2012, 6, 1–5. [Google Scholar] [CrossRef] [PubMed]
  82. Calisher, C.H. Taxonomy: What’s in a name? Doesn’t a rose by any other name smell as sweet? Croat. Med. J. 2007, 48, 268–270. [Google Scholar]
  83. Witte, K.; Sabat, R.; Witte-Handel, E.; Ghoreschi, K.; Wolk, K. Phytotherapeuthics Affecting the IL-1/IL-17/G-CSF Axis: A Complementary Treatment Option for Hidradenitis Suppurativa? Int. J. Mol. Sci. 2022, 23, 9057. [Google Scholar] [CrossRef] [PubMed]
  84. Yahfoufi, N.; Alsadi, N.; Jambi, M.; Matar, C. The Immunomodulatory and Anti-Inflammatory Role of Polyphenols. Nutrients 2018, 10, 1618. [Google Scholar] [CrossRef]
  85. Leon-Gonzalez, A.J.; Auger, C.; Schini-Kerth, V.B. Pro-oxidant activity of polyphenols and its implication on cancer chemoprevention and chemotherapy. Biochem. Pharmacol. 2015, 98, 371–380. [Google Scholar] [CrossRef] [PubMed]
  86. Stevens, J.F.; Revel, J.S.; Maier, C.S. Mitochondria-Centric Review of Polyphenol Bioactivity in Cancer Models. Antioxid. Redox Signal. 2018, 29, 1589–1611. [Google Scholar] [CrossRef] [PubMed]
  87. Correa, J.A.; Lopez-Villodres, J.A.; Asensi, R.; Espartero, J.L.; Rodriguez-Gutierez, G.; De La Cruz, J.P. Virgin olive oil polyphenol hydroxytyrosol acetate inhibits in vitro platelet aggregation in human whole blood: Comparison with hydroxytyrosol and acetylsalicylic acid. Br. J. Nutr. 2009, 101, 1157–1164. [Google Scholar] [CrossRef] [PubMed]
  88. Masella, R.; Vari, R.; D’Archivio, M.; Di Benedetto, R.; Matarrese, P.; Malorni, W.; Scazzocchio, B.; Giovannini, C. Extra virgin olive oil biophenols inhibit cell-mediated oxidation of LDL by increasing the mRNA transcription of glutathione-related enzymes. J. Nutr. 2004, 134, 785–791. [Google Scholar] [CrossRef]
  89. Singh, I.; Mok, M.; Christensen, A.M.; Turner, A.H.; Hawley, J.A. The effects of polyphenols in olive leaves on platelet function. Nutr. Metab. Cardiovasc. Dis. 2008, 18, 127–132. [Google Scholar] [CrossRef]
  90. Storniolo, C.E.; Rosello-Catafau, J.; Pinto, X.; Mitjavila, M.T.; Moreno, J.J. Polyphenol fraction of extra virgin olive oil protects against endothelial dysfunction induced by high glucose and free fatty acids through modulation of nitric oxide and endothelin-1. Redox Biol. 2014, 2, 971–977. [Google Scholar] [CrossRef]
  91. Wang, N.; Liu, Y.; Ma, Y.; Wen, D. Hydroxytyrosol ameliorates insulin resistance by modulating endoplasmic reticulum stress and prevents hepatic steatosis in diet-induced obesity mice. J. Nutr. Biochem 2018, 57, 180–188. [Google Scholar] [CrossRef] [PubMed]
  92. Wu, X.; Li, C.; Mariyam, Z.; Jiang, P.; Zhou, M.; Zeb, F.; Haq, I.U.; Chen, A.; Feng, Q. Acrolein-induced atherogenesis by stimulation of hepatic flavin containing monooxygenase 3 and a protection from hydroxytyrosol. J. Cell. Physiol. 2018, 234, 475–485. [Google Scholar] [CrossRef]
  93. Visioli, F.; Poli, A.; Gall, C. Antioxidant and other biological activities of phenols from olives and olive oil. Med. Res. Rev. 2002, 22, 65–75. [Google Scholar] [CrossRef]
  94. Scoditti, E.; Calabriso, N.; Massaro, M.; Pellegrino, M.; Storelli, C.; Martines, G.; De Caterina, R.; Carluccio, M.A. Mediterranean diet polyphenols reduce inflammatory angiogenesis through MMP-9 and COX-2 inhibition in human vascular endothelial cells: A potentially protective mechanism in atherosclerotic vascular disease and cancer. Arch. Biochem. Biophys. 2012, 527, 81–89. [Google Scholar] [CrossRef] [PubMed]
  95. Coni, E.; Di Benedetto, R.; Di Pasquale, M.; Masella, R.; Modesti, D.; Mattei, R.; Carlini, E.A. Protective effect of oleuropein, an olive oil biophenol, on low density lipoprotein oxidizability in rabbits. Lipids 2000, 35, 45–54. [Google Scholar] [CrossRef] [PubMed]
  96. Hadrich, F.; Mahmoudi, A.; Chamkha, M.; Isoda, H.; Sayadi, S. Olive Leaves Extract and Oleuropein Improve Insulin Sensitivity in 3T3-L1 Cells and in High-Fat Diet-Treated Rats via PI3K/AkT Signaling Pathway. Oxid. Med. Cell. Longev. 2023, 2023, 6828230. [Google Scholar] [CrossRef] [PubMed]
  97. Hamden, K.; Allouche, N.; Damak, M.; Elfeki, A. Hypoglycemic and antioxidant effects of phenolic extracts and purified hydroxytyrosol from olive mill waste in vitro and in rats. Chem. Biol. Interact. 2009, 180, 421–432. [Google Scholar] [CrossRef]
  98. Jemai, H.; El Feki, A.; Sayadi, S. Antidiabetic and antioxidant effects of hydroxytyrosol and oleuropein from olive leaves in alloxan-diabetic rats. J. Agric. Food Chem. 2009, 57, 8798–8804. [Google Scholar] [CrossRef]
  99. Khalili, A.; Nekooeian, A.A.; Khosravi, M.B. Oleuropein improves glucose tolerance and lipid profile in rats with simultaneous renovascular hypertension and type 2 diabetes. J. Asian Nat. Prod. Res. 2017, 19, 1011–1021. [Google Scholar] [CrossRef]
  100. Vezza, T.; Rodriguez-Nogales, A.; Algieri, F.; Garrido-Mesa, J.; Romero, M.; Sanchez, M.; Toral, M.; Martin-Garcia, B.; Gomez-Caravaca, A.M.; Arraez-Roman, D.; et al. The metabolic and vascular protective effects of olive (Olea europaea L.) leaf extract in diet-induced obesity in mice are related to the amelioration of gut microbiota dysbiosis and to its immunomodulatory properties. Pharmacol. Res. 2019, 150, 104487. [Google Scholar] [CrossRef]
  101. Wang, N.; Ma, Y.; Liu, Z.; Liu, L.; Yang, K.; Wei, Y.; Liu, Y.; Chen, X.; Sun, X.; Wen, D. Hydroxytyrosol prevents PM2.5-induced adiposity and insulin resistance by restraining oxidative stress related NF-kappaB pathway and modulation of gut microbiota in a murine model. Free Radic. Biol. Med. 2019, 141, 393–407. [Google Scholar] [CrossRef] [PubMed]
  102. Fki, I.; Sayadi, S.; Mahmoudi, A.; Daoued, I.; Marrekchi, R.; Ghorbel, H. Comparative Study on Beneficial Effects of Hydroxytyrosol- and Oleuropein-Rich Olive Leaf Extracts on High-Fat Diet-Induced Lipid Metabolism Disturbance and Liver Injury in Rats. BioMed Res. Int. 2020, 2020, 1315202. [Google Scholar] [CrossRef]
  103. Illesca, P.; Valenzuela, R.; Espinosa, A.; Echeverria, F.; Soto-Alarcon, S.; Ortiz, M.; Videla, L.A. Hydroxytyrosol supplementation ameliorates the metabolic disturbances in white adipose tissue from mice fed a high-fat diet through recovery of transcription factors Nrf2, SREBP-1c, PPAR-gamma and NF-kappaB. Biomed. Pharmacother. 2019, 109, 2472–2481. [Google Scholar] [CrossRef] [PubMed]
  104. Poudyal, H.; Lemonakis, N.; Efentakis, P.; Gikas, E.; Halabalaki, M.; Andreadou, I.; Skaltsounis, L.; Brown, L. Hydroxytyrosol ameliorates metabolic, cardiovascular and liver changes in a rat model of diet-induced metabolic syndrome: Pharmacological and metabolism-based investigation. Pharmacol. Res. 2017, 117, 32–45. [Google Scholar] [CrossRef] [PubMed]
  105. Colitti, M.; Stefanon, B. Different anti-adipogenic effects of bio-compounds on primary visceral pre-adipocytes and adipocytes. EXCLI J. 2016, 15, 362–377. [Google Scholar] [CrossRef]
  106. Drira, R.; Sakamoto, K. Hydroxytyrosol stimulates lipolysis via A-kinase and extracellular signal-regulated kinase activation in 3T3-L1 adipocytes. Eur. J. Nutr. 2014, 53, 743–750. [Google Scholar] [CrossRef]
  107. Stefanon, B.; Colitti, M. Original Research: Hydroxytyrosol, an ingredient of olive oil, reduces triglyceride accumulation and promotes lipolysis in human primary visceral adipocytes during differentiation. Exp. Biol. Med. 2016, 241, 1796–1802. [Google Scholar] [CrossRef]
  108. Scoditti, E.; Massaro, M.; Carluccio, M.A.; Pellegrino, M.; Wabitsch, M.; Calabriso, N.; Storelli, C.; De Caterina, R. Additive regulation of adiponectin expression by the mediterranean diet olive oil components oleic Acid and hydroxytyrosol in human adipocytes. PLoS ONE 2015, 10, e0128218. [Google Scholar] [CrossRef]
  109. Lopez-Villodres, J.A.; Abdel-Karim, M.; De La Cruz, J.P.; Rodriguez-Perez, M.D.; Reyes, J.J.; Guzman-Moscoso, R.; Rodriguez-Gutierrez, G.; Fernandez-Bolanos, J.; Gonzalez-Correa, J.A. Effects of hydroxytyrosol on cardiovascular biomarkers in experimental diabetes mellitus. J. Nutr. Biochem. 2016, 37, 94–100. [Google Scholar] [CrossRef] [PubMed]
  110. Andreadou, I.; Iliodromitis, E.K.; Mikros, E.; Constantinou, M.; Agalias, A.; Magiatis, P.; Skaltsounis, A.L.; Kamber, E.; Tsantili-Kakoulidou, A.; Kremastinos, D.T. The olive constituent oleuropein exhibits anti-ischemic, antioxidative, and hypolipidemic effects in anesthetized rabbits. J. Nutr. 2006, 136, 2213–2219. [Google Scholar] [CrossRef]
  111. Dub, A.M.; Dugani, A.M. Antithrombotic effect of repeated doses of the ethanolic extract of local olive (Olea europaea L.) leaves in rabbits. Libyan J. Med. 2013, 8, 20947. [Google Scholar] [CrossRef] [PubMed]
  112. Nekooeian, A.A.; Khalili, A.; Khosravi, M.B. Oleuropein offers cardioprotection in rats with simultaneous type 2 diabetes and renal hypertension. Indian J. Pharmacol. 2014, 46, 398–403. [Google Scholar] [CrossRef]
  113. Nekooeian, A.A.; Khalili, A.; Khosravi, M.B. Effects of oleuropein in rats with simultaneous type 2 diabetes and renal hypertension: A study of antihypertensive mechanisms. J. Asian Nat. Prod. Res. 2014, 16, 953–962. [Google Scholar] [CrossRef] [PubMed]
  114. Pei, Y.H.; Chen, J.; Xie, L.; Cai, X.M.; Yang, R.H.; Wang, X.; Gong, J.B. Hydroxytyrosol Protects against Myocardial Ischemia/Reperfusion Injury through a PI3K/Akt-Dependent Mechanism. Mediat. Inflamm. 2016, 2016, 1232103. [Google Scholar] [CrossRef] [PubMed]
  115. Romero, M.; Toral, M.; Gomez-Guzman, M.; Jimenez, R.; Galindo, P.; Sanchez, M.; Olivares, M.; Galvez, J.; Duarte, J. Antihypertensive effects of oleuropein-enriched olive leaf extract in spontaneously hypertensive rats. Food Funct. 2016, 7, 584–593. [Google Scholar] [CrossRef]
  116. Zhang, X.; Qin, Y.; Wan, X.; Liu, H.; Iv, C.; Ruan, W.; Lu, L.; He, L.; Guo, X. Hydroxytyrosol Plays Antiatherosclerotic Effects through Regulating Lipid Metabolism via Inhibiting the p38 Signal Pathway. BioMed Res. Int. 2020, 2020, 5036572. [Google Scholar] [CrossRef] [PubMed]
  117. Ilic, S.; Stojiljkovic, N.; Stojanovic, N.; Stoiljkovic, M.; Mitic, K.; Salinger-Martinovic, S.; Randjelovic, P. Effects of oleuropein on rat’s atria and thoracic aorta: A study of antihypertensive mechanisms. Can. J. Physiol. Pharmacol. 2021, 99, 110–114. [Google Scholar] [CrossRef]
  118. Gonzalez-Correa, J.A.; Navas, M.D.; Munoz-Marin, J.; Trujillo, M.; Fernandez-Bolanos, J.; de la Cruz, J.P. Effects of hydroxytyrosol and hydroxytyrosol acetate administration to rats on platelet function compared to acetylsalicylic acid. J. Agric. Food Chem. 2008, 56, 7872–7876. [Google Scholar] [CrossRef]
  119. Pirozzi, C.; Lama, A.; Simeoli, R.; Paciello, O.; Pagano, T.B.; Mollica, M.P.; Di Guida, F.; Russo, R.; Magliocca, S.; Canani, R.B.; et al. Hydroxytyrosol prevents metabolic impairment reducing hepatic inflammation and restoring duodenal integrity in a rat model of NAFLD. J. Nutr. Biochem. 2016, 30, 108–115. [Google Scholar] [CrossRef]
  120. Valenzuela, R.; Echeverria, F.; Ortiz, M.; Rincon-Cervera, M.A.; Espinosa, A.; Hernandez-Rodas, M.C.; Illesca, P.; Valenzuela, A.; Videla, L.A. Hydroxytyrosol prevents reduction in liver activity of Delta-5 and Delta-6 desaturases, oxidative stress, and depletion in long chain polyunsaturated fatty acid content in different tissues of high-fat diet fed mice. Lipids Health Dis. 2017, 16, 64. [Google Scholar] [CrossRef]
  121. Carnevale, R.; Silvestri, R.; Loffredo, L.; Novo, M.; Cammisotto, V.; Castellani, V.; Bartimoccia, S.; Nocella, C.; Violi, F. Oleuropein, a component of extra virgin olive oil, lowers postprandial glycaemia in healthy subjects. Br. J. Clin. Pharmacol. 2018, 84, 1566–1574. [Google Scholar] [CrossRef] [PubMed]
  122. de Bock, M.; Derraik, J.G.; Brennan, C.M.; Biggs, J.B.; Morgan, P.E.; Hodgkinson, S.C.; Hofman, P.L.; Cutfield, W.S. Olive (Olea europaea L.) leaf polyphenols improve insulin sensitivity in middle-aged overweight men: A randomized, placebo-controlled, crossover trial. PLoS ONE 2013, 8, e57622. [Google Scholar] [CrossRef] [PubMed]
  123. Kerimi, A.; Nyambe-Silavwe, H.; Pyner, A.; Oladele, E.; Gauer, J.S.; Stevens, Y.; Williamson, G. Nutritional implications of olives and sugar: Attenuation of post-prandial glucose spikes in healthy volunteers by inhibition of sucrose hydrolysis and glucose transport by oleuropein. Eur. J. Nutr. 2019, 58, 1315–1330. [Google Scholar] [CrossRef] [PubMed]
  124. Tenore, G.C.; Caruso, D.; D’Avino, M.; Buonomo, G.; Caruso, G.; Ciampaglia, R.; Schiano, E.; Maisto, M.; Annunziata, G.; Novellino, E. A Pilot Screening of Agro-Food Waste Products as Sources of Nutraceutical Formulations to Improve Simulated Postprandial Glycaemia and Insulinaemia in Healthy Subjects. Nutrients 2020, 12, 1292. [Google Scholar] [CrossRef]
  125. Javadi, H.; Yaghoobzadeh, H.; Esfahani, Z.; Reza Memarzadeh, M.; Mehdi Mirhashemi, S. Effects of Olive Leaf Extract on Metabolic Response, Liver and Kidney Functions and Inflammatory Biomarkers in Hypertensive Patients. Pak. J. Biol. Sci. 2019, 22, 342–348. [Google Scholar] [CrossRef]
  126. Lockyer, S.; Rowland, I.; Spencer, J.P.E.; Yaqoob, P.; Stonehouse, W. Impact of phenolic-rich olive leaf extract on blood pressure, plasma lipids and inflammatory markers: A randomised controlled trial. Eur. J. Nutr. 2017, 56, 1421–1432. [Google Scholar] [CrossRef] [PubMed]
  127. Stevens, Y.; Winkens, B.; Jonkers, D.; Masclee, A. The effect of olive leaf extract on cardiovascular health markers: A randomized placebo-controlled clinical trial. Eur. J. Nutr. 2021, 60, 2111–2120. [Google Scholar] [CrossRef]
  128. Pyner, A.; Chan, S.Y.; Tumova, S.; Kerimi, A.; Williamson, G. Indirect Chronic Effects of an Oleuropein-Rich Olive Leaf Extract on Sucrase-Isomaltase In Vitro and In Vivo. Nutrients 2019, 11, 1505. [Google Scholar] [CrossRef]
  129. Filip, R.; Possemiers, S.; Heyerick, A.; Pinheiro, I.; Raszewski, G.; Davicco, M.J.; Coxam, V. Twelve-month consumption of a polyphenol extract from olive (Olea europaea) in a double blind, randomized trial increases serum total osteocalcin levels and improves serum lipid profiles in postmenopausal women with osteopenia. J. Nutr. Health Aging 2015, 19, 77–86. [Google Scholar] [CrossRef]
  130. Susalit, E.; Agus, N.; Effendi, I.; Tjandrawinata, R.R.; Nofiarny, D.; Perrinjaquet-Moccetti, T.; Verbruggen, M. Olive (Olea europaea) leaf extract effective in patients with stage-1 hypertension: Comparison with Captopril. Phytomedicine 2011, 18, 251–258. [Google Scholar] [CrossRef]
  131. Lockyer, S.; Corona, G.; Yaqoob, P.; Spencer, J.P.; Rowland, I. Secoiridoids delivered as olive leaf extract induce acute improvements in human vascular function and reduction of an inflammatory cytokine: A randomised, double-blind, placebo-controlled, cross-over trial. Br. J. Nutr. 2015, 114, 75–83. [Google Scholar] [CrossRef]
  132. Gorelick, J.; Rosenberg, R.; Smotrich, A.; Hanus, L.; Bernstein, N. Hypoglycemic activity of withanolides and elicitated Withania somnifera. Phytochemistry 2015, 116, 283–289. [Google Scholar] [CrossRef] [PubMed]
  133. SoRelle, J.A.; Itoh, T.; Peng, H.; Kanak, M.A.; Sugimoto, K.; Matsumoto, S.; Levy, M.F.; Lawrence, M.C.; Naziruddin, B. Withaferin A inhibits pro-inflammatory cytokine-induced damage to islets in culture and following transplantation. Diabetologia 2013, 56, 814–824. [Google Scholar] [CrossRef] [PubMed]
  134. Khalilpourfarshbafi, M.; Devi Murugan, D.; Abdul Sattar, M.Z.; Sucedaram, Y.; Abdullah, N.A. Withaferin A inhibits adipogenesis in 3T3-F442A cell line, improves insulin sensitivity and promotes weight loss in high fat diet-induced obese mice. PLoS ONE 2019, 14, e0218792. [Google Scholar] [CrossRef] [PubMed]
  135. Batumalaie, K.; Amin, M.A.; Murugan, D.D.; Sattar, M.Z.; Abdullah, N.A. Withaferin A protects against palmitic acid-induced endothelial insulin resistance and dysfunction through suppression of oxidative stress and inflammation. Sci. Rep. 2016, 6, 27236. [Google Scholar] [CrossRef]
  136. Abu Bakar, M.H.; Azmi, M.N.; Shariff, K.A.; Tan, J.S. Withaferin A Protects Against High-Fat Diet-Induced Obesity Via Attenuation of Oxidative Stress, Inflammation, and Insulin Resistance. Appl. Biochem. Biotechnol. 2019, 188, 241–259. [Google Scholar] [CrossRef] [PubMed]
  137. Shiragannavar, V.D.; Sannappa Gowda, N.G.; Puttahanumantharayappa, L.D.; Karunakara, S.H.; Bhat, S.; Prasad, S.K.; Kumar, D.P.; Santhekadur, P.K. The ameliorating effect of withaferin A on high-fat diet-induced non-alcoholic fatty liver disease by acting as an LXR/FXR dual receptor activator. Front. Pharmacol. 2023, 14, 1135952. [Google Scholar] [CrossRef]
  138. Tekula, S.; Khurana, A.; Anchi, P.; Godugu, C. Withaferin-A attenuates multiple low doses of Streptozotocin (MLD-STZ) induced type 1 diabetes. Biomed. Pharmacother. 2018, 106, 1428–1440. [Google Scholar] [CrossRef] [PubMed]
  139. Guo, B.; Liu, J.; Wang, B.; Zhang, C.; Su, Z.; Zhao, M.; Qin, L.; Zhang, W.; Zheng, R. Withaferin A Promotes White Adipose Browning and Prevents Obesity Through Sympathetic Nerve-Activated Prdm16-FATP1 Axis. Diabetes 2022, 71, 249–263. [Google Scholar] [CrossRef]
  140. Lee, D.H.; Ahn, J.; Jang, Y.J.; Seo, H.D.; Ha, T.Y.; Kim, M.J.; Huh, Y.H.; Jung, C.H. Withania somnifera Extract Enhances Energy Expenditure via Improving Mitochondrial Function in Adipose Tissue and Skeletal Muscle. Nutrients 2020, 12, 431. [Google Scholar] [CrossRef]
  141. Lee, J.; Liu, J.; Feng, X.; Salazar Hernandez, M.A.; Mucka, P.; Ibi, D.; Choi, J.W.; Ozcan, U. Withaferin A is a leptin sensitizer with strong antidiabetic properties in mice. Nat. Med. 2016, 22, 1023–1032. [Google Scholar] [CrossRef]
  142. Lee, D.H.; Park, S.H.; Lee, E.; Seo, H.D.; Ahn, J.; Jang, Y.J.; Ha, T.Y.; Im, S.S.; Jung, C.H. Withaferin A exerts an anti-obesity effect by increasing energy expenditure through thermogenic gene expression in high-fat diet-fed obese mice. Phytomedicine 2021, 82, 153457. [Google Scholar] [CrossRef]
  143. Gupta, S.K.; Mohanty, I.; Talwar, K.K.; Dinda, A.; Joshi, S.; Bansal, P.; Saxena, A.; Arya, D.S. Cardioprotection from ischemia and reperfusion injury by Withania somnifera: A hemodynamic, biochemical and histopathological assessment. Mol. Cell. Biochem. 2004, 260, 39–47. [Google Scholar] [CrossRef]
  144. Hamza, A.; Amin, A.; Daoud, S. The protective effect of a purified extract of Withania somnifera against doxorubicin-induced cardiac toxicity in rats. Cell Biol. Toxicol. 2008, 24, 63–73. [Google Scholar] [CrossRef] [PubMed]
  145. Kaur, G.; Singh, N.; Samuel, S.S.; Bora, H.K.; Sharma, S.; Pachauri, S.D.; Dwivedi, A.K.; Siddiqui, H.H.; Hanif, K. Withania somnifera shows a protective effect in monocrotaline-induced pulmonary hypertension. Pharm. Biol. 2015, 53, 147–157. [Google Scholar] [CrossRef] [PubMed]
  146. Mohanty, I.R.; Arya, D.S.; Gupta, S.K. Withania somnifera provides cardioprotection and attenuates ischemia-reperfusion induced apoptosis. Clin. Nutr. 2008, 27, 635–642. [Google Scholar] [CrossRef]
  147. Yan, Z.; Guo, R.; Gan, L.; Lau, W.B.; Cao, X.; Zhao, J.; Ma, X.; Christopher, T.A.; Lopez, B.L.; Wang, Y. Withaferin A inhibits apoptosis via activated Akt-mediated inhibition of oxidative stress. Life Sci. 2018, 211, 91–101. [Google Scholar] [CrossRef] [PubMed]
  148. Patel, D.P.; Yan, T.; Kim, D.; Dias, H.B.; Krausz, K.W.; Kimura, S.; Gonzalez, F.J. Withaferin A Improves Nonalcoholic Steatohepatitis in Mice. J. Pharmacol. Exp. Ther. 2019, 371, 360–374. [Google Scholar] [CrossRef]
  149. Jadeja, R.N.; Urrunaga, N.H.; Dash, S.; Khurana, S.; Saxena, N.K. Withaferin-A Reduces Acetaminophen-Induced Liver Injury in Mice. Biochem. Pharmacol. 2015, 97, 122–132. [Google Scholar] [CrossRef]
  150. Xia, Y.; Wang, P.; Yan, N.; Gonzalez, F.J.; Yan, T. Withaferin A alleviates fulminant hepatitis by targeting macrophage and NLRP3. Cell Death Dis. 2021, 12, 174. [Google Scholar] [CrossRef]
  151. Choudhary, D.; Bhattacharyya, S.; Joshi, K. Body Weight Management in Adults Under Chronic Stress Through Treatment With Ashwagandha Root Extract: A Double-Blind, Randomized, Placebo-Controlled Trial. J. Evid. Based Complement. Altern. Med. 2017, 22, 96–106. [Google Scholar] [CrossRef]
  152. Wankhede, S.; Langade, D.; Joshi, K.; Sinha, S.R.; Bhattacharyya, S. Examining the effect of Withania somnifera supplementation on muscle strength and recovery: A randomized controlled trial. J. Int. Soc. Sports Nutr. 2015, 12, 43. [Google Scholar] [CrossRef]
  153. Tiwari, S.; Gupta, S.K.; Pathak, A.K. A double-blind, randomized, placebo-controlled trial on the effect of Ashwagandha (Withania somnifera dunal.) root extract in improving cardiorespiratory endurance and recovery in healthy athletic adults. J. Ethnopharmacol. 2021, 272, 113929. [Google Scholar] [CrossRef]
  154. Sharma, A.K.; Basu, I.; Singh, S. Efficacy and Safety of Ashwagandha Root Extract in Subclinical Hypothyroid Patients: A Double-Blind, Randomized Placebo-Controlled Trial. J. Altern. Complement. Med. 2018, 24, 243–248. [Google Scholar] [CrossRef] [PubMed]
  155. Ambiye, V.R.; Langade, D.; Dongre, S.; Aptikar, P.; Kulkarni, M.; Dongre, A. Clinical Evaluation of the Spermatogenic Activity of the Root Extract of Ashwagandha (Withania somnifera) in Oligospermic Males: A Pilot Study. Evid. Based Complement. Altern. Med. 2013, 2013, 571420. [Google Scholar] [CrossRef] [PubMed]
  156. Chandrasekhar, K.; Kapoor, J.; Anishetty, S. A prospective, randomized double-blind, placebo-controlled study of safety and efficacy of a high-concentration full-spectrum extract of ashwagandha root in reducing stress and anxiety in adults. Indian J. Psychol. Med. 2012, 34, 255–262. [Google Scholar] [CrossRef] [PubMed]
  157. Mahdi, A.A.; Shukla, K.K.; Ahmad, M.K.; Rajender, S.; Shankhwar, S.N.; Singh, V.; Dalela, D. Withania somnifera Improves Semen Quality in Stress-Related Male Fertility. Evid. Based Complement. Altern. Med. 2009, 2011, 576962. [Google Scholar] [CrossRef]
  158. Sandhu, J.S.; Shah, B.; Shenoy, S.; Chauhan, S.; Lavekar, G.S.; Padhi, M.M. Effects of Withania somnifera (Ashwagandha) and Terminalia arjuna (Arjuna) on physical performance and cardiorespiratory endurance in healthy young adults. Int. J. Ayurveda Res. 2010, 1, 144–149. [Google Scholar] [CrossRef] [PubMed]
  159. Verma, N.; Gupta, S.K.; Tiwari, S.; Mishra, A.K. Safety of Ashwagandha Root Extract: A Randomized, Placebo-Controlled, study in Healthy Volunteers. Complement. Ther. Med. 2021, 57, 102642. [Google Scholar] [CrossRef]
  160. Gonzalez-Abuin, N.; Martinez-Micaelo, N.; Blay, M.; Pujadas, G.; Garcia-Vallve, S.; Pinent, M.; Ardevol, A. Grape seed-derived procyanidins decrease dipeptidyl-peptidase 4 activity and expression. J. Agric. Food Chem. 2012, 60, 9055–9061. [Google Scholar] [CrossRef]
  161. Gonzalez-Abuin, N.; Martinez-Micaelo, N.; Margalef, M.; Blay, M.; Arola-Arnal, A.; Muguerza, B.; Ardevol, A.; Pinent, M. A grape seed extract increases active glucagon-like peptide-1 levels after an oral glucose load in rats. Food Funct. 2014, 5, 2357–2364. [Google Scholar] [CrossRef]
  162. Yilmaz, Y.; Toledo, R.T. Major flavonoids in grape seeds and skins: Antioxidant capacity of catechin, epicatechin, and gallic acid. J. Agric. Food Chem. 2004, 52, 255–260. [Google Scholar] [CrossRef]
  163. Wei, S.; Zheng, Y.; Zhang, M.; Zheng, H.; Yan, P. Grape seed procyanidin extract inhibits adipogenesis and stimulates lipolysis of porcine adipocytes in vitro. J. Anim. Sci. 2018, 96, 2753–2762. [Google Scholar] [CrossRef]
  164. Zhang, J.; Huang, Y.; Shao, H.; Bi, Q.; Chen, J.; Ye, Z. Grape seed procyanidin B2 inhibits adipogenesis of 3T3-L1 cells by targeting peroxisome proliferator-activated receptor gamma with miR-483-5p involved mechanism. Biomed. Pharmacother. 2017, 86, 292–296. [Google Scholar] [CrossRef] [PubMed]
  165. Edirisinghe, I.; Burton-Freeman, B.; Tissa Kappagoda, C. Mechanism of the endothelium-dependent relaxation evoked by a grape seed extract. Clin. Sci. 2008, 114, 331–337. [Google Scholar] [CrossRef]
  166. Feng, Z.; Wei, R.B.; Hong, Q.; Cui, S.Y.; Chen, X.M. Grape seed extract enhances eNOS expression and NO production through regulating calcium-mediated AKT phosphorylation in H2O2-treated endothelium. Cell Biol. Int. 2010, 34, 1055–1061. [Google Scholar] [CrossRef]
  167. Decorde, K.; Teissedre, P.L.; Sutra, T.; Ventura, E.; Cristol, J.P.; Rouanet, J.M. Chardonnay grape seed procyanidin extract supplementation prevents high-fat diet-induced obesity in hamsters by improving adipokine imbalance and oxidative stress markers. Mol. Nutr. Food Res. 2009, 53, 659–666. [Google Scholar] [CrossRef]
  168. Meeprom, A.; Sompong, W.; Suwannaphet, W.; Yibchok-anun, S.; Adisakwattana, S. Grape seed extract supplementation prevents high-fructose diet-induced insulin resistance in rats by improving insulin and adiponectin signalling pathways. Br. J. Nutr. 2011, 106, 1173–1181. [Google Scholar] [CrossRef]
  169. Ohyama, K.; Furuta, C.; Nogusa, Y.; Nomura, K.; Miwa, T.; Suzuki, K. Catechin-rich grape seed extract supplementation attenuates diet-induced obesity in C57BL/6J mice. Ann. Nutr. Metab. 2011, 58, 250–258. [Google Scholar] [CrossRef]
  170. Rajasekhar, S.; Subramanyam, M.V.V.; Asha Devi, S. Grape seed proanthocyanidin extract suppresses oxidative stress in the rat pancreas of type-1 diabetes. Arch. Physiol. Biochem. 2021, 1–13. [Google Scholar] [CrossRef]
  171. Suwannaphet, W.; Meeprom, A.; Yibchok-Anun, S.; Adisakwattana, S. Preventive effect of grape seed extract against high-fructose diet-induced insulin resistance and oxidative stress in rats. Food Chem. Toxicol. 2010, 48, 1853–1857. [Google Scholar] [CrossRef] [PubMed]
  172. Wu, Z.; Shen, S.; Jiang, J.; Tan, D.; Jiang, D.; Bai, B.; Sun, X.; Fu, S. Protective effects of grape seed extract fractions with different degrees of polymerisation on blood glucose, lipids and hepatic oxidative stress in diabetic rats. Nat. Prod. Res. 2015, 29, 988–992. [Google Scholar] [CrossRef] [PubMed]
  173. Liu, W.; Zhao, S.; Wang, J.; Shi, J.; Sun, Y.; Wang, W.; Ning, G.; Hong, J.; Liu, R. Grape seed proanthocyanidin extract ameliorates inflammation and adiposity by modulating gut microbiota in high-fat diet mice. Mol. Nutr. Food Res. 2017, 61, 1601082. [Google Scholar] [CrossRef]
  174. Gonzalez-Abuin, N.; Martinez-Micaelo, N.; Blay, M.; Ardevol, A.; Pinent, M. Grape-seed procyanidins prevent the cafeteria-diet-induced decrease of glucagon-like peptide-1 production. J. Agric. Food Chem. 2014, 62, 1066–1072. [Google Scholar] [CrossRef]
  175. Castell-Auvi, A.; Cedo, L.; Pallares, V.; Blay, M.; Pinent, M.; Ardevol, A. Grape seed procyanidins improve beta-cell functionality under lipotoxic conditions due to their lipid-lowering effect. J. Nutr. Biochem. 2013, 24, 948–953. [Google Scholar] [CrossRef] [PubMed]
  176. Charradi, K.; Sebai, H.; Elkahoui, S.; Ben Hassine, F.; Limam, F.; Aouani, E. Grape seed extract alleviates high-fat diet-induced obesity and heart dysfunction by preventing cardiac siderosis. Cardiovasc. Toxicol. 2011, 11, 28–37. [Google Scholar] [CrossRef]
  177. Pascual-Serrano, A.; Blade, C.; Suarez, M.; Arola-Arnal, A. Grape Seed Proanthocyanidins Improve White Adipose Tissue Expansion during Diet-Induced Obesity Development in Rats. Int. J. Mol. Sci. 2018, 19, 2632. [Google Scholar] [CrossRef]
  178. Shi, Y.; Jia, M.; Xu, L.; Fang, Z.; Wu, W.; Zhang, Q.; Chung, P.; Lin, Y.; Wang, S.; Zhang, Y. miR-96 and autophagy are involved in the beneficial effect of grape seed proanthocyanidins against high-fat-diet-induced dyslipidemia in mice. Phytother. Res. 2019, 33, 1222–1232. [Google Scholar] [CrossRef]
  179. Sierra-Cruz, M.; Miguens-Gomez, A.; Grau-Bove, C.; Rodriguez-Gallego, E.; Blay, M.; Pinent, M.; Ardevol, A.; Terra, X.; Beltran-Debon, R. Grape-Seed Proanthocyanidin Extract Reverts Obesity-Related Metabolic Derangements in Aged Female Rats. Nutrients 2021, 13, 2059. [Google Scholar] [CrossRef]
  180. Yogalakshmi, B.; Sreeja, S.; Geetha, R.; Radika, M.K.; Anuradha, C.V. Grape seed proanthocyanidin rescues rats from steatosis: A comparative and combination study with metformin. J. Lipids 2013, 2013, 153897. [Google Scholar] [CrossRef]
  181. Mokrani, M.; Charradi, K.; Limam, F.; Aouani, E.; Urdaci, M.C. Grape seed and skin extract, a potential prebiotic with anti-obesity effect through gut microbiota modulation. Gut Pathog. 2022, 14, 30. [Google Scholar] [CrossRef]
  182. Serrano, J.; Casanova-Marti, A.; Gil-Cardoso, K.; Blay, M.T.; Terra, X.; Pinent, M.; Ardevol, A. Acutely administered grape-seed proanthocyanidin extract acts as a satiating agent. Food Funct. 2016, 7, 483–490. [Google Scholar] [CrossRef] [PubMed]
  183. Jiao, R.; Zhang, Z.; Yu, H.; Huang, Y.; Chen, Z.Y. Hypocholesterolemic activity of grape seed proanthocyanidin is mediated by enhancement of bile acid excretion and up-regulation of CYP7A1. J. Nutr. Biochem. 2010, 21, 1134–1139. [Google Scholar] [CrossRef] [PubMed]
  184. Du, H.; Wang, Q.; Li, T.; Ren, D.; Yang, X. Grape seed proanthocyanidins reduced the overweight of C57BL/6J mice through modulating adipose thermogenesis and gut microbiota. Food Funct. 2021, 12, 8467–8477. [Google Scholar] [CrossRef] [PubMed]
  185. Liang, Y.; Wang, J.; Gao, H.; Wang, Q.; Zhang, J.; Qiu, J. Beneficial effects of grape seed proanthocyanidin extract on arterial remodeling in spontaneously hypertensive rats via protecting against oxidative stress. Mol. Med. Rep. 2016, 14, 3711–3718. [Google Scholar] [CrossRef]
  186. Ruan, Y.; Jin, Q.; Zeng, J.; Ren, F.; Xie, Z.; Ji, K.; Wu, L.; Wu, J.; Li, L. Grape Seed Proanthocyanidin Extract Ameliorates Cardiac Remodelling After Myocardial Infarction Through PI3K/AKT Pathway in Mice. Front. Pharmacol. 2020, 11, 585984. [Google Scholar] [CrossRef] [PubMed]
  187. Sergazy, S.; Shulgau, Z.; Fedotovskikh, G.; Chulenbayeva, L.; Nurgozhina, A.; Nurgaziyev, M.; Krivyh, E.; Kamyshanskiy, Y.; Kushugulova, A.; Gulyayev, A.; et al. Cardioprotective effect of grape polyphenol extract against doxorubicin induced cardiotoxicity. Sci. Rep. 2020, 10, 14720. [Google Scholar] [CrossRef]
  188. Guisantes-Batan, E.; Mazuecos, L.; Rubio, B.; Pereira-Caro, G.; Moreno-Rojas, J.M.; Andres, A.; Gomez-Alonso, S.; Gallardo, N. Grape seed extract supplementation modulates hepatic lipid metabolism in rats. Implication of PPARbeta/delta. Food Funct. 2022, 13, 11353–11368. [Google Scholar] [CrossRef]
  189. Kar, P.; Laight, D.; Rooprai, H.K.; Shaw, K.M.; Cummings, M. Effects of grape seed extract in Type 2 diabetic subjects at high cardiovascular risk: A double blind randomized placebo controlled trial examining metabolic markers, vascular tone, inflammation, oxidative stress and insulin sensitivity. Diabet. Med. 2009, 26, 526–531. [Google Scholar] [CrossRef]
  190. Mohammad, A.; Shahnaz, T.; Sorayya, K. Effect of 8 weeks’ supplementation grape seed extract on insulin resistance in iranian adolescents with metabolic syndrome: A randomized controlled trial. Diabetes Metab. Syndr. 2021, 15, 197–203. [Google Scholar] [CrossRef]
  191. Park, E.; Edirisinghe, I.; Choy, Y.Y.; Waterhouse, A.; Burton-Freeman, B. Effects of grape seed extract beverage on blood pressure and metabolic indices in individuals with pre-hypertension: A randomised, double-blinded, two-arm, parallel, placebo-controlled trial. Br. J. Nutr. 2016, 115, 226–238. [Google Scholar] [CrossRef]
  192. Argani, H.; Ghorbanihaghjo, A.; Vatankhahan, H.; Rashtchizadeh, N.; Raeisi, S.; Ilghami, H. The effect of red grape seed extract on serum paraoxonase activity in patients with mild to moderate hyperlipidemia. Sao Paulo Med. J. 2016, 134, 234–239. [Google Scholar] [CrossRef]
  193. Razavi, S.M.; Gholamin, S.; Eskandari, A.; Mohsenian, N.; Ghorbanihaghjo, A.; Delazar, A.; Rashtchizadeh, N.; Keshtkar-Jahromi, M.; Argani, H. Red grape seed extract improves lipid profiles and decreases oxidized low-density lipoprotein in patients with mild hyperlipidemia. J. Med. Food 2013, 16, 255–258. [Google Scholar] [CrossRef]
  194. Weseler, A.R.; Ruijters, E.J.; Drittij-Reijnders, M.J.; Reesink, K.D.; Haenen, G.R.; Bast, A. Pleiotropic benefit of monomeric and oligomeric flavanols on vascular health—A randomized controlled clinical pilot study. PLoS ONE 2011, 6, e28460. [Google Scholar] [CrossRef]
  195. Yousefi, R.; Parandoosh, M.; Khorsandi, H.; Hosseinzadeh, N.; Madani Tonekaboni, M.; Saidpour, A.; Babaei, H.; Ghorbani, A. Grape seed extract supplementation along with a restricted-calorie diet improves cardiovascular risk factors in obese or overweight adult individuals: A randomized, placebo-controlled trial. Phytother. Res. 2021, 35, 987–995. [Google Scholar] [CrossRef]
  196. Preuss, H.G.; Wallerstedt, D.; Talpur, N.; Tutuncuoglu, S.O.; Echard, B.; Myers, A.; Bui, M.; Bagchi, D. Effects of niacin-bound chromium and grape seed proanthocyanidin extract on the lipid profile of hypercholesterolemic subjects: A pilot study. J. Med. 2000, 31, 227–246. [Google Scholar]
  197. Vogels, N.; Nijs, I.M.; Westerterp-Plantenga, M.S. The effect of grape-seed extract on 24 h energy intake in humans. Eur. J. Clin. Nutr. 2004, 58, 667–673. [Google Scholar] [CrossRef]
  198. Parandoosh, M.; Yousefi, R.; Khorsandi, H.; Nikpayam, O.; Saidpour, A.; Babaei, H. The effects of grape seed extract (Vitis vinifera) supplement on inflammatory markers, neuropeptide Y, anthropometric measures, and appetite in obese or overweight individuals: A randomized clinical trial. Phytother. Res. 2020, 34, 379–387. [Google Scholar] [CrossRef]
  199. Terauchi, M.; Horiguchi, N.; Kajiyama, A.; Akiyoshi, M.; Owa, Y.; Kato, K.; Kubota, T. Effects of grape seed proanthocyanidin extract on menopausal symptoms, body composition, and cardiovascular parameters in middle-aged women: A randomized, double-blind, placebo-controlled pilot study. Menopause 2014, 21, 990–996. [Google Scholar] [CrossRef]
  200. Schon, C.; Allegrini, P.; Engelhart-Jentzsch, K.; Riva, A.; Petrangolini, G. Grape Seed Extract Positively Modulates Blood Pressure and Perceived Stress: A Randomized, Double-Blind, Placebo-Controlled Study in Healthy Volunteers. Nutrients 2021, 13, 654. [Google Scholar] [CrossRef]
  201. Odai, T.; Terauchi, M.; Kato, K.; Hirose, A.; Miyasaka, N. Effects of Grape Seed Proanthocyanidin Extract on Vascular Endothelial Function in Participants with Prehypertension: A Randomized, Double-Blind, Placebo-Controlled Study. Nutrients 2019, 11, 2844. [Google Scholar] [CrossRef] [PubMed]
  202. Dillon, K.N.; Shariffi, B.; Thompson, B.; Steele, R.; Kim, J.K. Effects of Acute Grape Seed Extract Supplementation on Hemodynamics in Normal Body Weight and Obese Males. J. Nutr. Sci. Vitaminol. 2020, 66, 427–431. [Google Scholar] [CrossRef] [PubMed]
  203. Kim, J.K.; Kim, K.A.; Choi, H.M.; Park, S.K.; Stebbins, C.L. Grape Seed Extract Supplementation Attenuates the Blood Pressure Response to Exercise in Prehypertensive Men. J. Med. Food 2018, 21, 445–453. [Google Scholar] [CrossRef] [PubMed]
  204. Ras, R.T.; Zock, P.L.; Zebregs, Y.E.; Johnston, N.R.; Webb, D.J.; Draijer, R. Effect of polyphenol-rich grape seed extract on ambulatory blood pressure in subjects with pre- and stage I hypertension. Br. J. Nutr. 2013, 110, 2234–2241. [Google Scholar] [CrossRef] [PubMed]
  205. Xiao, X.; Erukainure, O.L.; Sanni, O.; Koorbanally, N.A.; Islam, M.S. Phytochemical properties of black tea (Camellia sinensis) and rooibos tea (Aspalathus linearis); and their modulatory effects on key hyperglycaemic processes and oxidative stress. J. Food Sci. Technol. 2020, 57, 4345–4354. [Google Scholar] [CrossRef]
  206. Othman, A.I.; El-Sawi, M.R.; El-Missiry, M.A.; Abukhalil, M.H. Epigallocatechin-3-gallate protects against diabetic cardiomyopathy through modulating the cardiometabolic risk factors, oxidative stress, inflammation, cell death and fibrosis in streptozotocin-nicotinamide-induced diabetic rats. Biomed. Pharmacother. 2017, 94, 362–373. [Google Scholar] [CrossRef]
  207. Li, W.; Zhu, C.; Liu, T.; Zhang, W.; Liu, X.; Li, P.; Zhu, T. Epigallocatechin-3-gallate ameliorates glucolipid metabolism and oxidative stress in type 2 diabetic rats. Diabetes Vasc. Dis. Res. 2020, 17, 1479164120966998. [Google Scholar] [CrossRef]
  208. Ren, Z.; Yang, Z.; Lu, Y.; Zhang, R.; Yang, H. Antiglycolipid disorder effect of epigallocatechin3gallate on highfat diet and STZinduced T2DM in mice. Mol. Med. Rep. 2020, 21, 2475–2483. [Google Scholar] [CrossRef]
  209. Sun, W.; Liu, X.; Zhang, H.; Song, Y.; Li, T.; Liu, X.; Liu, Y.; Guo, L.; Wang, F.; Yang, T.; et al. Epigallocatechin gallate upregulates NRF2 to prevent diabetic nephropathy via disabling KEAP1. Free Radic. Biol. Med. 2017, 108, 840–857. [Google Scholar] [CrossRef]
  210. Zhang, C.; Li, X.; Hu, X.; Xu, Q.; Zhang, Y.; Liu, H.; Diao, Y.; Zhang, X.; Li, L.; Yu, J.; et al. Epigallocatechin-3-gallate prevents inflammation and diabetes -Induced glucose tolerance through inhibition of NLRP3 inflammasome activation. Int. Immunopharmacol. 2021, 93, 107412. [Google Scholar] [CrossRef]
  211. Zhang, J.; Zhao, L.; Cheng, Q.; Ji, B.; Yang, M.; Sanidad, K.Z.; Wang, C.; Zhou, F. Structurally Different Flavonoid Subclasses Attenuate High-Fat and High-Fructose Diet Induced Metabolic Syndrome in Rats. J. Agric. Food Chem. 2018, 66, 12412–12420. [Google Scholar] [CrossRef]
  212. Park, J.H.; Jin, J.Y.; Baek, W.K.; Park, S.H.; Sung, H.Y.; Kim, Y.K.; Lee, J.; Song, D.K. Ambivalent role of gallated catechins in glucose tolerance in humans: A novel insight into non-absorbable gallated catechin-derived inhibitors of glucose absorption. J. Physiol. Pharmacol. 2009, 60, 101–109. [Google Scholar]
  213. Li, X.; Li, S.; Chen, M.; Wang, J.; Xie, B.; Sun, Z. (−)-Epigallocatechin-3-gallate (EGCG) inhibits starch digestion and improves glucose homeostasis through direct or indirect activation of PXR/CAR-mediated phase II metabolism in diabetic mice. Food Funct. 2018, 9, 4651–4663. [Google Scholar] [CrossRef]
  214. Ni, D.; Ai, Z.; Munoz-Sandoval, D.; Suresh, R.; Ellis, P.R.; Yuqiong, C.; Sharp, P.A.; Butterworth, P.J.; Yu, Z.; Corpe, C.P. Inhibition of the facilitative sugar transporters (GLUTs) by tea extracts and catechins. FASEB J. 2020, 34, 9995–10010. [Google Scholar] [CrossRef]
  215. Li, F.; Gao, C.; Yan, P.; Zhang, M.; Wang, Y.; Hu, Y.; Wu, X.; Wang, X.; Sheng, J. EGCG Reduces Obesity and White Adipose Tissue Gain Partly Through AMPK Activation in Mice. Front. Pharmacol. 2018, 9, 1366. [Google Scholar] [CrossRef]
  216. Li, Y.; Wu, S. Epigallocatechin gallate suppresses hepatic cholesterol synthesis by targeting SREBP-2 through SIRT1/FOXO1 signaling pathway. Mol. Cell. Biochem. 2018, 448, 175–185. [Google Scholar] [CrossRef]
  217. Wu, D.; Liu, Z.; Wang, Y.; Zhang, Q.; Li, J.; Zhong, P.; Xie, Z.; Ji, A.; Li, Y. Epigallocatechin-3-Gallate Alleviates High-Fat Diet-Induced Nonalcoholic Fatty Liver Disease via Inhibition of Apoptosis and Promotion of Autophagy through the ROS/MAPK Signaling Pathway. Oxid. Med. Cell. Longev. 2021, 2021, 5599997. [Google Scholar] [CrossRef]
  218. Choi, C.; Song, H.D.; Son, Y.; Cho, Y.K.; Ahn, S.Y.; Jung, Y.S.; Yoon, Y.C.; Kwon, S.W.; Lee, Y.H. Epigallocatechin-3-Gallate Reduces Visceral Adiposity Partly through the Regulation of Beclin1-Dependent Autophagy in White Adipose Tissues. Nutrients 2020, 12, 3072. [Google Scholar] [CrossRef]
  219. Lee, M.S.; Kim, C.T.; Kim, Y. Green tea (−)-epigallocatechin-3-gallate reduces body weight with regulation of multiple genes expression in adipose tissue of diet-induced obese mice. Ann. Nutr. Metab. 2009, 54, 151–157. [Google Scholar] [CrossRef]
  220. Santana, A.; Santamarina, A.; Souza, G.; Mennitti, L.; Okuda, M.; Venancio, D.; Seelaender, M.; do Nascimento, C.O.; Ribeiro, E.; Lira, F.; et al. Decaffeinated green tea extract rich in epigallocatechin-3-gallate improves insulin resistance and metabolic profiles in normolipidic diet—But not high-fat diet-fed mice. J. Nutr. Biochem 2015, 26, 893–902. [Google Scholar] [CrossRef]
  221. Cui, C.J.; Jin, J.L.; Guo, L.N.; Sun, J.; Wu, N.Q.; Guo, Y.L.; Liu, G.; Dong, Q.; Li, J.J. Beneficial impact of epigallocatechingallate on LDL-C through PCSK9/LDLR pathway by blocking HNF1alpha and activating FoxO3a. J. Transl. Med. 2020, 18, 195. [Google Scholar] [CrossRef]
  222. Kim, S.N.; Kwon, H.J.; Akindehin, S.; Jeong, H.W.; Lee, Y.H. Effects of Epigallocatechin-3-Gallate on Autophagic Lipolysis in Adipocytes. Nutrients 2017, 9, 680. [Google Scholar] [CrossRef]
  223. Mi, Y.; Liu, X.; Tian, H.; Liu, H.; Li, J.; Qi, G.; Liu, X. EGCG stimulates the recruitment of brite adipocytes, suppresses adipogenesis and counteracts TNF-alpha-triggered insulin resistance in adipocytes. Food Funct. 2018, 9, 3374–3386. [Google Scholar] [CrossRef]
  224. Yu, J.; Li, W.; Xiao, X.; Huang, Q.; Yu, J.; Yang, Y.; Han, T.; Zhang, D.; Niu, X. (−)-Epicatechin gallate blocks the development of atherosclerosis by regulating oxidative stress in vivo and in vitro. Food Funct. 2021, 12, 8715–8727. [Google Scholar] [CrossRef]
  225. Antonello, M.; Montemurro, D.; Bolognesi, M.; Di Pascoli, M.; Piva, A.; Grego, F.; Sticchi, D.; Giuliani, L.; Garbisa, S.; Rossi, G.P. Prevention of hypertension, cardiovascular damage and endothelial dysfunction with green tea extracts. Am. J. Hypertens. 2007, 20, 1321–1328. [Google Scholar] [CrossRef]
  226. Yin, J.; Huang, F.; Yi, Y.; Yin, L.; Peng, D. EGCG attenuates atherosclerosis through the Jagged-1/Notch pathway. Int. J. Mol. Med. 2016, 37, 398–406. [Google Scholar] [CrossRef]
  227. Faria, A.M.; Papadimitriou, A.; Silva, K.C.; Lopes de Faria, J.M.; Lopes de Faria, J.B. Uncoupling endothelial nitric oxide synthase is ameliorated by green tea in experimental diabetes by re-establishing tetrahydrobiopterin levels. Diabetes 2012, 61, 1838–1847. [Google Scholar] [CrossRef]
  228. Zhang, Z.; Zhang, D. (−)-Epigallocatechin-3-Gallate Inhibits eNOS Uncoupling and Alleviates High Glucose-Induced Dysfunction and Apoptosis of Human Umbilical Vein Endothelial Cells by 186PI3K/AKT/eNOS Pathway. Diabetes Metab. Syndr. Obes. 2020, 13, 2495–2504. [Google Scholar] [CrossRef]
  229. Nan, J.; Nan, C.; Ye, J.; Qian, L.; Geng, Y.; Xing, D.; Rahman, M.S.U.; Huang, M. EGCG protects cardiomyocytes against hypoxia-reperfusion injury through inhibition of OMA1 activation. J. Cell Sci. 2019, 132, jcs220871. [Google Scholar] [CrossRef]
  230. Shen, K.; Feng, X.; Su, R.; Xie, H.; Zhou, L.; Zheng, S. Epigallocatechin 3-gallate ameliorates bile duct ligation induced liver injury in mice by modulation of mitochondrial oxidative stress and inflammation. PLoS ONE 2015, 10, e0126278. [Google Scholar] [CrossRef]
  231. Thitimuta, S.; Pithayanukul, P.; Nithitanakool, S.; Bavovada, R.; Leanpolchareanchai, J.; Saparpakorn, P. Camellia sinensis L. Extract and Its Potential Beneficial Effects in Antioxidant, Anti-Inflammatory, Anti-Hepatotoxic, and Anti-Tyrosinase Activities. Molecules 2017, 22, 401. [Google Scholar] [CrossRef] [PubMed]
  232. Kochi, T.; Shimizu, M.; Terakura, D.; Baba, A.; Ohno, T.; Kubota, M.; Shirakami, Y.; Tsurumi, H.; Tanaka, T.; Moriwaki, H. Non-alcoholic steatohepatitis and preneoplastic lesions develop in the liver of obese and hypertensive rats: Suppressing effects of EGCG on the development of liver lesions. Cancer Lett. 2014, 342, 60–69. [Google Scholar] [CrossRef]
  233. Chatree, S.; Sitticharoon, C.; Maikaew, P.; Pongwattanapakin, K.; Keadkraichaiwat, I.; Churintaraphan, M.; Sripong, C.; Sririwichitchai, R.; Tapechum, S. Epigallocatechin gallate decreases plasma triglyceride, blood pressure, and serum kisspeptin in obese human subjects. Exp. Biol. Med. 2021, 246, 163–176. [Google Scholar] [CrossRef] [PubMed]
  234. de Morais Junior, A.C.; Schincaglia, R.M.; Passarelli, M.; Pimentel, G.D.; Mota, J.F. Acute Epigallocatechin-3-Gallate Supplementation Alters Postprandial Lipids after a Fast-Food Meal in Healthy Young Women: A Randomized, Double-Blind, Placebo-Controlled Crossover Study. Nutrients 2020, 12, 2533. [Google Scholar] [CrossRef] [PubMed]
  235. Hill, A.M.; Coates, A.M.; Buckley, J.D.; Ross, R.; Thielecke, F.; Howe, P.R. Can EGCG reduce abdominal fat in obese subjects? J. Am. Coll. Nutr. 2007, 26, 396S–402S. [Google Scholar] [CrossRef]
  236. Takahashi, M.; Ozaki, M.; Tsubosaka, M.; Kim, H.K.; Sasaki, H.; Matsui, Y.; Hibi, M.; Osaki, N.; Miyashita, M.; Shibata, S. Effects of Timing of Acute and Consecutive Catechin Ingestion on Postprandial Glucose Metabolism in Mice and Humans. Nutrients 2020, 12, 565. [Google Scholar] [CrossRef]
  237. Venables, M.C.; Hulston, C.J.; Cox, H.R.; Jeukendrup, A.E. Green tea extract ingestion, fat oxidation, and glucose tolerance in healthy humans. Am. J. Clin. Nutr. 2008, 87, 778–784. [Google Scholar] [CrossRef]
  238. Zhang, H.; Su, S.; Yu, X.; Li, Y. Dietary epigallocatechin 3-gallate supplement improves maternal and neonatal treatment outcome of gestational diabetes mellitus: A double-blind randomised controlled trial. J. Hum. Nutr. Diet. 2017, 30, 753–758. [Google Scholar] [CrossRef]
  239. Brown, A.L.; Lane, J.; Coverly, J.; Stocks, J.; Jackson, S.; Stephen, A.; Bluck, L.; Coward, A.; Hendrickx, H. Effects of dietary supplementation with the green tea polyphenol epigallocatechin-3-gallate on insulin resistance and associated metabolic risk factors: Randomized controlled trial. Br. J. Nutr. 2009, 101, 886–894. [Google Scholar] [CrossRef]
  240. Fernandes, R.C.; Araujo, V.A.; Giglio, B.M.; Marini, A.C.B.; Mota, J.F.; Teixeira, K.S.; Monteiro, P.A.; Lira, F.S.; Pimentel, G.D. Acute Epigallocatechin 3 Gallate (EGCG) Supplementation Delays Gastric Emptying in Healthy Women: A Randomized, Double-Blind, Placebo-Controlled Crossover Study. Nutrients 2018, 10, 1122. [Google Scholar] [CrossRef]
  241. Hsu, C.H.; Liao, Y.L.; Lin, S.C.; Tsai, T.H.; Huang, C.J.; Chou, P. Does supplementation with green tea extract improve insulin resistance in obese type 2 diabetics? A randomized, double-blind, and placebo-controlled clinical trial. Altern. Med. Rev. 2011, 16, 157–163. [Google Scholar]
  242. Mielgo-Ayuso, J.; Barrenechea, L.; Alcorta, P.; Larrarte, E.; Margareto, J.; Labayen, I. Effects of dietary supplementation with epigallocatechin-3-gallate on weight loss, energy homeostasis, cardiometabolic risk factors and liver function in obese women: Randomised, double-blind, placebo-controlled clinical trial. Br. J. Nutr. 2014, 111, 1263–1271. [Google Scholar] [CrossRef]
  243. Wu, A.H.; Spicer, D.; Stanczyk, F.Z.; Tseng, C.C.; Yang, C.S.; Pike, M.C. Effect of 2-month controlled green tea intervention on lipoprotein cholesterol, glucose, and hormone levels in healthy postmenopausal women. Cancer Prev. Res. 2012, 5, 393–402. [Google Scholar] [CrossRef] [PubMed]
  244. Dostal, A.M.; Samavat, H.; Espejo, L.; Arikawa, A.Y.; Stendell-Hollis, N.R.; Kurzer, M.S. Green Tea Extract and Catechol-O-Methyltransferase Genotype Modify Fasting Serum Insulin and Plasma Adiponectin Concentrations in a Randomized Controlled Trial of Overweight and Obese Postmenopausal Women. J. Nutr. 2016, 146, 38–45. [Google Scholar] [CrossRef]
  245. Bazyar, H.; Hosseini, S.A.; Saradar, S.; Mombaini, D.; Allivand, M.; Labibzadeh, M.; Alipour, M. Effects of epigallocatechin-3-gallate of Camellia sinensis leaves on blood pressure, lipid profile, atherogenic index of plasma and some inflammatory and antioxidant markers in type 2 diabetes mellitus patients: A clinical trial. J. Complement. Integr. Med. 2020, 18, 405–411. [Google Scholar] [CrossRef]
  246. Chen, I.J.; Liu, C.Y.; Chiu, J.P.; Hsu, C.H. Therapeutic effect of high-dose green tea extract on weight reduction: A randomized, double-blind, placebo-controlled clinical trial. Clin. Nutr. 2016, 35, 592–599. [Google Scholar] [CrossRef] [PubMed]
  247. Huang, L.H.; Liu, C.Y.; Wang, L.Y.; Huang, C.J.; Hsu, C.H. Effects of green tea extract on overweight and obese women with high levels of low density-lipoprotein-cholesterol (LDL-C): A randomised, double-blind, and cross-over placebo-controlled clinical trial. BMC Complement. Altern. Med. 2018, 18, 294. [Google Scholar] [CrossRef]
  248. Samavat, H.; Newman, A.R.; Wang, R.; Yuan, J.M.; Wu, A.H.; Kurzer, M.S. Effects of green tea catechin extract on serum lipids in postmenopausal women: A randomized, placebo-controlled clinical trial. Am. J. Clin. Nutr. 2016, 104, 1671–1682. [Google Scholar] [CrossRef] [PubMed]
  249. Hsu, C.H.; Tsai, T.H.; Kao, Y.H.; Hwang, K.C.; Tseng, T.Y.; Chou, P. Effect of green tea extract on obese women: A randomized, double-blind, placebo-controlled clinical trial. Clin. Nutr. 2008, 27, 363–370. [Google Scholar] [CrossRef]
  250. Basu, A.; Sanchez, K.; Leyva, M.J.; Wu, M.; Betts, N.M.; Aston, C.E.; Lyons, T.J. Green tea supplementation affects body weight, lipids, and lipid peroxidation in obese subjects with metabolic syndrome. J. Am. Coll. Nutr. 2010, 29, 31–40. [Google Scholar] [CrossRef]
  251. Brown, A.L.; Lane, J.; Holyoak, C.; Nicol, B.; Mayes, A.E.; Dadd, T. Health effects of green tea catechins in overweight and obese men: A randomised controlled cross-over trial. Br. J. Nutr. 2011, 106, 1880–1889. [Google Scholar] [CrossRef]
  252. Dostal, A.M.; Arikawa, A.; Espejo, L.; Bedell, S.; Kurzer, M.S.; Stendell-Hollis, N.R. Green tea extract and catechol-O-methyltransferase genotype modify the post-prandial serum insulin response in a randomised trial of overweight and obese post-menopausal women. J. Hum. Nutr. Diet. 2017, 30, 166–176. [Google Scholar] [CrossRef]
  253. Thielecke, F.; Rahn, G.; Bohnke, J.; Adams, F.; Birkenfeld, A.L.; Jordan, J.; Boschmann, M. Epigallocatechin-3-gallate and postprandial fat oxidation in overweight/obese male volunteers: A pilot study. Eur. J. Clin. Nutr. 2010, 64, 704–713. [Google Scholar] [CrossRef]
  254. Quezada-Fernandez, P.; Trujillo-Quiros, J.; Pascoe-Gonzalez, S.; Trujillo-Rangel, W.A.; Cardona-Muller, D.; Ramos-Becerra, C.G.; Barocio-Pantoja, M.; Rodriguez-de la Cerda, M.; Nerida Sanchez-Rodriguez, E.; Cardona-Munoz, E.G.; et al. Effect of green tea extract on arterial stiffness, lipid profile and sRAGE in patients with type 2 diabetes mellitus: A randomised, double-blind, placebo-controlled trial. Int. J. Food Sci. Nutr. 2019, 70, 977–985. [Google Scholar] [CrossRef]
  255. Widlansky, M.E.; Hamburg, N.M.; Anter, E.; Holbrook, M.; Kahn, D.F.; Elliott, J.G.; Keaney, J.F., Jr.; Vita, J.A. Acute EGCG supplementation reverses endothelial dysfunction in patients with coronary artery disease. J. Am. Coll. Nutr. 2007, 26, 95–102. [Google Scholar] [CrossRef]
  256. Lorenz, M.; Rauhut, F.; Hofer, C.; Gwosc, S.; Muller, E.; Praeger, D.; Zimmermann, B.F.; Wernecke, K.D.; Baumann, G.; Stangl, K.; et al. Tea-induced improvement of endothelial function in humans: No role for epigallocatechin gallate (EGCG). Sci. Rep. 2017, 7, 2279. [Google Scholar] [CrossRef]
  257. Arazi, H.; Samami, N.; Kheirkhah, J.; Taati, B. The effect of three weeks green tea extract consumption on blood pressure, heart rate responses to a single bout resistance exercise in hypertensive women. High Blood Press. Cardiovasc. Prev. 2014, 21, 213–219. [Google Scholar] [CrossRef]
  258. Zouboulis, C.C.; Tzellos, T.; Kyrgidis, A.; Jemec, G.B.E.; Bechara, F.G.; Giamarellos-Bourboulis, E.J.; Ingram, J.R.; Kanni, T.; Karagiannidis, I.; Martorell, A.; et al. Development and validation of the International Hidradenitis Suppurativa Severity Score System (IHS4), a novel dynamic scoring system to assess HS severity. Br. J. Dermatol. 2017, 177, 1401–1409. [Google Scholar] [CrossRef]
  259. Bentivegna, S.S.; Whitney, K.M. Subchronic 3-month oral toxicity study of grape seed and grape skin extracts. Food Chem. Toxicol. 2002, 40, 1731–1743. [Google Scholar] [CrossRef]
  260. Durg, S.; Bavage, S.; Shivaram, S.B. Withania somnifera (Indian ginseng) in diabetes mellitus: A systematic review and meta-analysis of scientific evidence from experimental research to clinical application. Phytother. Res. 2020, 34, 1041–1059. [Google Scholar] [CrossRef]
  261. Goey, A.K.; Meijerman, I.; Beijnen, J.H.; Schellens, J.H. The effect of grape seed extract on the pharmacokinetics of dextromethorphan in healthy volunteers. Eur. J. Clin. Pharmacol. 2013, 69, 1883–1890. [Google Scholar] [CrossRef]
  262. Kountouri, A.M.; Mylona, A.; Kaliora, A.C.; Andrikopoulos, N.K. Bioavailability of the phenolic compounds of the fruits (drupes) of Olea europaea (olives): Impact on plasma antioxidant status in humans. Phytomedicine 2007, 14, 659–667. [Google Scholar] [CrossRef] [PubMed]
  263. Sano, A. Safety assessment of 4-week oral intake of proanthocyanidin-rich grape seed extract in healthy subjects. Food Chem. Toxicol. 2017, 108, 519–523. [Google Scholar] [CrossRef] [PubMed]
  264. Sano, A.; Uchida, R.; Saito, M.; Shioya, N.; Komori, Y.; Tho, Y.; Hashizume, N. Beneficial effects of grape seed extract on malondialdehyde-modified LDL. J. Nutr. Sci Vitaminol. 2007, 53, 174–182. [Google Scholar] [CrossRef]
  265. Wren, A.F.; Cleary, M.; Frantz, C.; Melton, S.; Norris, L. 90-day oral toxicity study of a grape seed extract (IH636) in rats. J. Agric. Food Chem. 2002, 50, 2180–2192. [Google Scholar] [CrossRef] [PubMed]
  266. Raut, A.A.; Rege, N.N.; Tadvi, F.M.; Solanki, P.V.; Kene, K.R.; Shirolkar, S.G.; Pandey, S.N.; Vaidya, R.A.; Vaidya, A.B. Exploratory study to evaluate tolerability, safety, and activity of Ashwagandha (Withania somnifera) in healthy volunteers. J. Ayurveda Integr. Med. 2012, 3, 111–114. [Google Scholar] [CrossRef] [PubMed]
  267. Hu, J.; Webster, D.; Cao, J.; Shao, A. The safety of green tea and green tea extract consumption in adults—Results of a systematic review. Regul. Toxicol. Pharmacol. 2018, 95, 412–433. [Google Scholar] [CrossRef]
  268. Darweesh, R.S.; El-Elimat, T.; Zayed, A.; Khamis, T.N.; Babaresh, W.M.; Arafat, T.; Al Sharie, A.H. The effect of grape seed and green tea extracts on the pharmacokinetics of imatinib and its main metabolite, N-desmethyl imatinib, in rats. BMC Pharmacol. Toxicol. 2020, 21, 77. [Google Scholar] [CrossRef]
  269. Etheridge, A.S.; Black, S.R.; Patel, P.R.; So, J.; Mathews, J.M. An in vitro evaluation of cytochrome P450 inhibition and P-glycoprotein interaction with goldenseal, Ginkgo biloba, grape seed, milk thistle, and ginseng extracts and their constituents. Planta Med. 2007, 73, 731–741. [Google Scholar] [CrossRef]
  270. Malliou, F.; Andriopoulou, C.E.; Gonzalez, F.J.; Kofinas, A.; Skaltsounis, A.L.; Konstandi, M. Oleuropein-Induced Acceleration of Cytochrome P450-Catalyzed Drug Metabolism: Central Role for Nuclear Receptor Peroxisome Proliferator-Activated Receptor alpha. Drug Metab. Dispos. 2021, 49, 833–843. [Google Scholar] [CrossRef]
  271. Mooiman, K.D.; Maas-Bakker, R.F.; Hendrikx, J.J.; Bank, P.C.; Rosing, H.; Beijnen, J.H.; Schellens, J.H.; Meijerman, I. The effect of complementary and alternative medicines on CYP3A4-mediated metabolism of three different substrates: 7-benzyloxy-4-trifluoromethyl-coumarin, midazolam and docetaxel. J. Pharm. Pharmacol. 2014, 66, 865–874. [Google Scholar] [CrossRef]
  272. Ray, S.D.; Parikh, H.; Hickey, E.; Bagchi, M.; Bagchi, D. Differential effects of IH636 grape seed proanthocyanidin extract and a DNA repair modulator 4-aminobenzamide on liver microsomal cytochrome 4502E1-dependent aniline hydroxylation. Mol. Cell. Biochem. 2001, 218, 27–33. [Google Scholar] [CrossRef]
  273. Kumar, S.; Bouic, P.J.; Rosenkranz, B. Investigation of CYP2B6, 3A4 and beta-esterase interactions of Withania somnifera (L.) dunal in human liver microsomes and HepG2 cells. J. Ethnopharmacol. 2021, 270, 113766. [Google Scholar] [CrossRef]
  274. Savai, J.; Varghese, A.; Pandita, N.; Chintamaneni, M. Investigation of CYP3A4 and CYP2D6 Interactions of Withania somnifera and Centella asiatica in Human Liver Microsomes. Phytother. Res. 2015, 29, 785–790. [Google Scholar] [CrossRef]
  275. Savai, J.; Varghese, A.; Pandita, N.; Chintamaneni, M. In vitro assessment of CYP1A2 and 2C9 inhibition potential of Withania somnifera and Centella asiatica in human liver microsomes. Drug Metab. Pers. Ther. 2015, 30, 137–141. [Google Scholar] [CrossRef] [PubMed]
  276. Sak, K. The Val158Met polymorphism in COMT gene and cancer risk: Role of endogenous and exogenous catechols. Drug Metab. Rev. 2017, 49, 56–83. [Google Scholar] [CrossRef]
  277. Omar, S.H.; Scott, C.J.; Hamlin, A.S.; Obied, H.K. Biophenols: Enzymes (beta-secretase, Cholinesterases, histone deacetylase and tyrosinase) inhibitors from olive (Olea europaea L.). Fitoterapia 2018, 128, 118–129. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Representative picture of axillary (A) and lower belly/inguinal (B) skin lesions of HS patients.
Figure 1. Representative picture of axillary (A) and lower belly/inguinal (B) skin lesions of HS patients.
Nutrients 15 03797 g001
Figure 2. Botanical pictures of Olea europea (A), Withania somnifera (B), Vitis vinifera (C), and Camellia sinensis (D).
Figure 2. Botanical pictures of Olea europea (A), Withania somnifera (B), Vitis vinifera (C), and Camellia sinensis (D).
Nutrients 15 03797 g002
Figure 3. Schematic overview of metabolic alterations frequently observed in HS patients and their potential targeting by Olea europea, Withania somnifera, Vitis vinifera, and Camellia sinensis.
Figure 3. Schematic overview of metabolic alterations frequently observed in HS patients and their potential targeting by Olea europea, Withania somnifera, Vitis vinifera, and Camellia sinensis.
Nutrients 15 03797 g003
Table 1. Characteristics and main study outcomes of placebo-controlled, randomized clinical trials investigating the effects of Olea europea on metabolic parameters.
Table 1. Characteristics and main study outcomes of placebo-controlled, randomized clinical trials investigating the effects of Olea europea on metabolic parameters.
Study MedicationStudy TypeDose RegimenCohort Size
(n)
Study Cohort
Criteria
Main Study Results Verum vs. Control (Increased: ↑; Decreased: ↓; Unaffected: ≈)Ref.
500 mg OLEpc, db,
RCT
daily application
(8 weeks)
placebo: 38
verum: 39
overweight participants
age: 56 ± 10 years
BMI: 29 ± 2.7
≈ fasting glucose, insulin
≈ SBP, DBP
≈ lipid profile
[127]
250 mg OLEpc, db,
RCT
daily application
(12 weeks)
30/grouphypertension
participants
age: 23.4 ± 1.4 years
BMI: 22.7 ± 3.0
≈ fasting plasma glucose, insulin
≈ liver enzymes
↓ inflammatory cytokines (TNF-α, IL-8, IL-6)
[125]
oleuropein
(20 mg)
pc, db,
RCT, co
single applicationplacebo: 20
verum: 20
healthy participants
age: 33.9 ± 6.9
BMI: 20.7 ± 3.7
↓ postprandial plasma glucose
↑ postprandial plasma insulin
↓ postprandial oxidative stress
↑ GLP-1, ↓ DPP-4
[121]
20 mL OLE
(136.2 mg oleuropein; 6.4 mg hydroxytyrosol)
pc, db,
RCT, co
daily application
(6 weeks)
placebo: 60
verum: 60
PHT participants
age: 45.3 ± 12.7 years
BMI: 27.0 ± 3.4
↓ SBP, DBP (slight reduction)
↓ total CH, LDL-C, TG, IL-8
≈ vascular function, CRP, adiponectin
≈ fasting glucose, insulin, HOMA-IR, QUICKI, HDL-C
[126]
250 mg OLE
(oleuropein ≥ 100 mg)
pc, db,
RCT
daily application
(12 month)
placebo: 32
verum: 32
OST participants
verum/placebo:
age: 59.72/59.35 years
BMI: 25.90/27.52
↓ total CH, LDL-C, TG
≈ HDL-C
[129]
OLE (51.1 mg oleuropein; 9.7 mg hydroxytyrosol)pc, db,
RCT, co
daily application
(12 weeks)
placebo: 46
verum: 46
overweight participants
age: 46.4 ± 5.5 years
BMI: 28.0 ± 2.0
↓ postprandial plasma glucose, insulin
↑ insulin sensitivity (Matsuda index)
↑ pancreatic β-cell function (disposition index)
≈ lipid profile, body fat proportion, ABP
[122]
RCTs are listed according to the publication date, whereby 6 RCTs sorted for highest cohort size (n) of available studies are given. Only main metabolic and cardiovascular endpoint measures are presented. pc: placebo-controlled, db: double-blind, co: crossover design, RCT: randomized clinical trial, PHT: pre-hypertensive, OST: osteoporosis, SBP: systolic blood pressure, DBP: diastolic blood pressure, CH: total cholesterol, LDL: low-density lipoprotein, HDL: high-density lipoprotein, TGs: triglycerides, HOMA-IR: homoeostasis model assessment-estimated insulin resistance, QUICKI: quantitative insulin sensitivity check index, OLE: olive leaf extract.
Table 2. Characteristics and main study outcomes of placebo-controlled, randomized, clinical trials investigating the effects of Withania somnifera on metabolic parameters.
Table 2. Characteristics and main study outcomes of placebo-controlled, randomized, clinical trials investigating the effects of Withania somnifera on metabolic parameters.
Study MedicationStudy TypeDose RegimenCohort Size
(n)
Study Cohort CriteriaMain Study Results Verum vs. Control (Increased: ↑; Decreased:↓; Unaffected: ≈)Ref.
600 mg WSREpc, db,
RCT
daily application
(8 weeks)
placebo: 25
verum: 25
healthy athletes
age: 18–≤45 years
↑ cardiorespiratory endurance:
(↑ VO2 max outcome; ↑ TQR score; improved RESTQ score)
↑ anti-oxidative capacity
[153]
600 mg WSREpc, db,
RCT
daily application
(8 weeks)
placebo: 25
verum: 25
subclinical hypothyroid
participants
verum/placebo:
age: 35.6/35.1 years
↑ T3, T4
↓ TSH
[154]
600 mg WSRE
(5% withanolides)
pc, db,
RCT
daily application
(8 weeks)
placebo: 25
verum: 25
chronic stressed, overweight participants↓ perceived Stress Scale Score
↓ Food Cravings Questionnaire scores
↑ Oxford Happiness Questionnaire scores
↓ serum cortisol level
[151]
600 mg WSRE
(5% withanolides)
pc, db,
RCT
daily application
(8 weeks)
placebo: 25
verum: 25
healthy participants undergoing resistance training
verum/placebo:
age: 28 ± 8/29 ± 9 years
↑ muscle strength, muscle size (upper body)
↓ body fat percentage
[152]
RCT, listed according to publication date are given. Only main metabolic and cardiovascular endpoint measures are presented. pc: placebo-controlled, db: double-blind, co: crossover design, RCT: randomized clinical trial, T3: triiodothyronine, T4: thyroxine, TSH: thyroid stimulating hormone, WSRE: withania somnifera root extract.
Table 3. Characteristics and main study outcomes of randomized clinical trials investigating the effects of Vitis vinifera on metabolic parameters.
Table 3. Characteristics and main study outcomes of randomized clinical trials investigating the effects of Vitis vinifera on metabolic parameters.
Study MedicationStudy TypeDose RegimenCohort Size
(n)
Study Cohort CriteriaMain Study Results Verum vs. Control (Increased: ↑; Decreased: ↓; Unaffected: ≈)Ref.
300 mg GSEpc, db,
RCT
daily application
(16 weeks)
placebo: 38
verum:40
mild hypertension participants
verum/placebo:
age: 56.4/56.9 years
BMI: 25.2/26.1
↓ SBP, DBP (only in male participants)
↓ perceived stress score (PSQ)
[200]
200 mg GSEpc, db,
RCT, co
daily application
(8 weeks)
placebo: 45
verum: 45
mild hyperlipidemia participants
age: 48.22 ± 9.07 years
↓ CH, LDL, ox-LDL[193]
200 mg GSEpc, db,
RCT
daily application
(8 weeks)
placebo: 35
verum: 35
hyperlipidemia participants
verum/placebo
age: 46.6/47.3 years
↑ ApoA1, HDL
↑ PON activity
↓ CH, TG, LDL
[192]
300 mg GSEpc, db,
RCT
daily application
(8 weeks)
placebo: 35
verum: 34
pre- and stage-I hypertension participants
verum/placebo:
age: 62.9/64.5 years
BMI: 25.3/25.7
≈ SBP, DBP
≈ vasoactive markers
[204]
600 mg GSEpc, db,
RCT, co
daily application
(4 weeks)
placebo: 32
verum: 32
T2D participants
age: 61.8 ± 6.4 years
BMI: 30.2 ± 5.9
↓ fructosamine, CH, CRP
↑ GSH
≈ fasting glucose, HOMA-IR
≈ endothelial function
[189]
900 mg GSEpc, db,
RCT, co
daily application
(3 days)
placebo: 51
verum: 51
healthy participants
age: 48.7 ± 14.3 years
BMI: 25.6 ± 2.6
↓ 24 h energy intake (only in subjects
with ≥7.5 MJ/day)
[197]
RCTs are listed according to publication date, whereby 6 RCTs for each group, sorted for highest cohort size (n) of available studies, are provided. Only main metabolic and cardiovascular endpoint measures are presented. pc: placebo-controlled, db: double-blind, co: crossover design, RCT: randomized clinical trial, SBP: systolic blood pressure, DBP: diastolic blood pressure, CH: total cholesterol, LDL: low-density lipoprotein, ApoA1: apolipoprotein A1, PON: paraoxonase, HOMA-IR: homoeostasis model assessment-estimated insulin resistance, CRP: c-reactive protein, GSH: reduced glutathione, GSE: grape seed extract.
Table 4. Characteristics and main study outcomes of randomized clinical trials investigating the effects of Camellia sinensis on metabolic parameters.
Table 4. Characteristics and main study outcomes of randomized clinical trials investigating the effects of Camellia sinensis on metabolic parameters.
Study MedicationStudy TypeDose RegimenCohort Size
(n)
Study Cohort CriteriaMain Study Results Verum vs. Control (Increased: ↑; Decreased: ↓; Unaffected: ≈)Ref.
1500 mg GTE
(856.8 mg EGCG)
pc, db,
RCT, co
daily application
(6 weeks)
placebo: 73
verum: 73
overweight participants
age: 18–65 years
BMI: ≥27
↓ LDL-C
↑ Leptin
≈ CH, TG, HDL
[247]
500 mg EGCGpc, db,
RCT
daily application
(until birth)
placebo: 176
verum: 150
GDM participants
verum/placebo:
age: 29.6/28.7 years
BMI: 25.9/26.2
↓ fasting plasma glucose and insulin
↓ HOMA-IR/HOMA-ß scores
↑ QUICK-I index
[238]
green tea/GTE/EGCG) (200 mg EGCG each)pc,
RCT, co
single applicationplacebo: 50
verum: 50
healthy participants
age: 33.9 ± 7.6 years
BMI: 23.7 ± 2.5
↑ FMD (only in the green tea group)
≈ NMD
[256]
GTE
(843 mg EGCG; decaffeinated)
pc, db,
RCT
daily application
(12 month)
placebo: 473
verum: 463
healthy participants
verum/placebo:
age: 60.02/59.65 years
BMI: 25.16/25.01
↓ CH, LDL
↑ TG (mainly obese, statin users)
[248]
1500 mg GTE
(856.8 mg EGCG)
pc, db,
RCT
daily application
(12 weeks)
placebo: 38
verum: 39
obese participants
verum/placebo:
age: 44.1/44.9 years
BMI: 31/30
↓ CH, LDL[246]
1060 mg GTE
(431.5 mg EGCG)
pc, db,
RCT, co
daily application
(6 weeks)
placebo: 65
verum: 63
obese participants
verum/placebo:
age: 49.5/49.4 years
BMI: 31.7/31.4
≈ blood pressure
≈ body weight (only slight reduction during intervention period 1)
[251]
RCTs are listed according to the publication date, whereby 6 RCTs for each group, sorted for highest cohort size (n) in available studies, are provided. Only the main metabolic and cardiovascular endpoint measures are presented. pc: placebo-controlled, db: double-blind, co: crossover design, RCT: randomized clinical trial, GDM: gestational diabetes mellitus, CH: total cholesterol, LDL: low-density lipoprotein, TGs: triglycerides, HOMA-IR: homoeostasis model assessment-estimated insulin resistance, QUICKI: quantitative insulin sensitivity check index, FMD: flow-mediated dilation, NMD: nitro-mediated dilation, GTE: green tea extract, EGCG: epigallocatechin 3-gallate.
Table 5. Summary of main study outcomes of double-blind, placebo-controlled RCTs evaluating the effects of Olea europea, Withania somnifera, Vitis vinifera, and Camellia sinensis phytochemicals on metabolic syndrome parameters.
Table 5. Summary of main study outcomes of double-blind, placebo-controlled RCTs evaluating the effects of Olea europea, Withania somnifera, Vitis vinifera, and Camellia sinensis phytochemicals on metabolic syndrome parameters.
MetS ParameterOlea europeaWithania somniferaVitis viniferaCamellia sinensis
glucose metabolismimprovement of postprandial plasma glucoseonly preclinical data availableno clear impactimprovement of
glucose metabolism
dyslipidemiaimprovement of single
lipid parameters
only preclinical data availableimprovement of single
lipid parameters
improvement of single
lipid parameters
cardiovascular alterationsimprovement of
vascular function
improved cardiorespiratory enduranceimprovement of blood pressure parametersimprovement of cardiovascular
parameters
no clear impact on blood pressure
obesity/
weight management
no clear impactreduced perceived stress;
reduced food craving;
enhanced body weight reduction during reistance training
enhanced body weight reduction during caloric restrictionno clear impact
NAFLDonly preclinical data availableonly preclinical data availableonly preclinical data availableonly preclinical data available
RCT quantityn = 11n = 4n = 21n = 28
Nutrients 15 03797 i001 No RCTs or only 1 RCT available for this parameter. Nutrients 15 03797 i002 <50% of available RCTs show the effectivity of study medication on the respective parameter. Nutrients 15 03797 i003 =50% of available RCTs show the effectivity of study medication on the respective parameter. Nutrients 15 03797 i004 ≥50% of available RCTs show the effectivity of study medication on the respective parameter.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Witte, K.; Wolk, K.; Witte-Händel, E.; Krause, T.; Kokolakis, G.; Sabat, R. Targeting Metabolic Syndrome in Hidradenitis Suppurativa by Phytochemicals as a Potential Complementary Therapeutic Strategy. Nutrients 2023, 15, 3797. https://doi.org/10.3390/nu15173797

AMA Style

Witte K, Wolk K, Witte-Händel E, Krause T, Kokolakis G, Sabat R. Targeting Metabolic Syndrome in Hidradenitis Suppurativa by Phytochemicals as a Potential Complementary Therapeutic Strategy. Nutrients. 2023; 15(17):3797. https://doi.org/10.3390/nu15173797

Chicago/Turabian Style

Witte, Katrin, Kerstin Wolk, Ellen Witte-Händel, Torben Krause, Georgios Kokolakis, and Robert Sabat. 2023. "Targeting Metabolic Syndrome in Hidradenitis Suppurativa by Phytochemicals as a Potential Complementary Therapeutic Strategy" Nutrients 15, no. 17: 3797. https://doi.org/10.3390/nu15173797

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